1
|
Elsaid KA, Zhang LX, Zhao T, Marks A, Jenkins D, Schmidt TA, Jay GD. Proteoglycan 4 (Lubricin) and Regulation of Xanthine Oxidase in Synovial Macrophage as A Mechanism of Controlling Synovitis. RESEARCH SQUARE 2024:rs.3.rs-4934175. [PMID: 39372933 PMCID: PMC11451733 DOI: 10.21203/rs.3.rs-4934175/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background Synovial macrophages (SMs) are important effectors of joint health and disease. A novel Cx3CR1 + TREM2 + SM population expressing the tight junction protein claudin-5, was recently discovered in synovial lining. Ablation of these SMs was associated with onset of arthritis. Proteoglycan 4 (PRG4) is a mucinous glycoprotein that fulfills lubricating and homeostatic roles in the joint. The aim of this work is to study the role of PRG4 in modulating synovitis in the context of SM homeostasis and assess the contribution of xanthine oxidase (XO)-hypoxia inducible factor alpha (HIF-1α) axis to this regulation. Methods We used Prg4 FrlioxP/FrtloxP ;R26 FlPoER/+ , a novel transgenic mouse, where the Prg4 Frt allele normally expresses the PRG4 protein and was designed to flank the first two exons of Prg4 with a flippase recognition target and "LOXP" sites. Inducing flippase activity with tamoxifen (TAM) inactivates the Frt allele and thus creates a conditional knockout state. We studied anti-inflammatory SMs and XO by quantitative immunohistochemistry, isolated RNA and studied immune pathway activations by multiplexed assays and isolated SMs and studied PRG4 signaling dysfunction in relation to glycolytic switching due to pro-inflammatory activation. Prg4 inactivated mice were treated with oral febuxostat, a specific XO inhibitor, and quantification of Cx3CR1 + TREM2 + SMs, XO immunostaining and synovitis assessment were conducted. Results Prg4 inactivation induced Cx3CR1 + TREM2 + SM loss (p < 0.001) and upregulated glycolysis and innate immune pathways in the synovium. In isolated SMs, Xdh (p < 0.01) and Hif1a (p < 0.05) were upregulated. Pro-inflammatory activation of SMs was evident by enhanced glycolytic flux and XO-generated reactive oxygen species (ROS). Febuxostat reduced glycolytic flux (p < 0.001) and HIF-1α levels (p < 0.0001) in SMs. Febuxostat also reduced systemic inflammation (p < 0.001), synovial hyperplasia (p < 0.001) and preserved Cx3CR1 + TREM2 + SMs (p < 0.0001) in synovia of Prg4 inactivated mice. Conclusions PRG4 is a biologically significant modulator of synovial homeostasis via inhibition of XO expression and downstream HIF-1a activation. PRG4 signaling is anti-inflammatory and promotes synovial homeostasis in chronic synovitis, where direct XO inhibition is potentially therapeutic in chronic synovitis.
Collapse
|
2
|
Zhou W, Wang N, Dong S, Huan Z, Sui L, Ge X. PRG4 mitigates hemorrhagic shock-induced cardiac injury by inhibiting mitochondrial dysregulation, oxidative stress and NLRP3-mediated pyroptosis. Int Immunopharmacol 2024; 137:112507. [PMID: 38897120 DOI: 10.1016/j.intimp.2024.112507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
Hemorrhagic shock (HS) is one of the main causes of morbidity and death in patients with trauma or major surgery. Cardiac dysfunction is a well-known complication of HS. PRG4, also known as lubricin, is a mucin-like glycoprotein that plays anti-inflammatory and anti-apoptotic roles in a variety of diseases. In this study, we aimed to explore the cardioprotective efficacy of PRG4 in HS-induced cardiac injury. Employing the HS model and RNA-seq, we found that PRG4 was increased in the myocardial tissue of rats after HS. In vivo studies suggested that HS led to abnormal hemodynamic parameters and increased cTnI levels, and PRG4 overexpression effectively reversed these changes. PRG4 also suppressed HS-induced mitochondrial disorders, as reflected by increased mitochondrial membrane potential (MMP), ATP and mitochondria cytochrome c, COXIV and TOM20, as well as decreased BNIP3L and cytoplasmic cytochrome c. Furthermore, HS led to enhanced oxidative stress, as evidenced by upregulated ROS and MDA contents, and downregulated SOD and CAT activities, and these alterations were negated by PRG4 overexpression. Notably, PRG4 repressed the NLRP3-mediated pyroptosis pathway, as illustrated by decreased NLRP3 levels, caspase-1 activity and GSDMD-NT levels. In summary, these observations indicate that PRG4 overexpression protects against HS-induced cardiac dysfunction by inhibiting mitochondrial dysregulation, oxidative stress and NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Wuming Zhou
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Nan Wang
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Sheng Dong
- Department of Emergency, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Zhirong Huan
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Lijun Sui
- Department of Cardiology, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China.
| | - Xin Ge
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China; Department of Emergency, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China; Orthopedic Institution of Wuxi City, Wuxi, Jiangsu 214000, People's Republic of China.
| |
Collapse
|
3
|
Raftery RM, Pregizer SK, Kocher S, Craft AM. Regenerative capacity of human pluripotent stem cell-derived articular chondrocytes in vitro. J Orthop Res 2024; 42:1841-1851. [PMID: 38433390 PMCID: PMC11222035 DOI: 10.1002/jor.25823] [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: 12/02/2023] [Revised: 02/06/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
The ideal cell source for articular cartilage repair remains elusive. Using developmentally inspired differentiation protocols, we induced human pluripotent stem cells (hPSCs) toward articular chondrocytes capable of joint cartilage repair in rodent models, which were distinct from growth plate chondrocytes, fated to be replaced by bone in vivo. Working toward clinical translation, we demonstrated controlled differentiation into chondrocytes by comprehensive gene expression analysis at each step of the differentiation. Articular chondrocytes derived from hPSCs could be expanded several passages in vitro without losing chondrogenic potential. Furthermore, chondrocytes isolated from these articular cartilage tissues had the potential to serially regenerate new articular and growth plate cartilage tissues. Finally, the ability to cryopreserve articular chondrocytes with the desired phenotype is critical for clinical translation and here we report no loss in cell viability or regenerative potential following cryopreservation. These results support the immense potential of hPSC-derived articular chondrocytes as a cell-based therapy for cartilage repair.
Collapse
Affiliation(s)
- Rosanne M. Raftery
- Department of Orthopedic Research, Boston Children’s Hospital, Boston, MA USA
- Department of Orthopedic Surgery, Harvard Medical School, Boston, MA USA
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Ireland
| | - Steven K. Pregizer
- Department of Orthopedic Research, Boston Children’s Hospital, Boston, MA USA
- Department of Orthopedic Surgery, Harvard Medical School, Boston, MA USA
| | - Sophia Kocher
- Department of Orthopedic Research, Boston Children’s Hospital, Boston, MA USA
| | - April M. Craft
- Department of Orthopedic Research, Boston Children’s Hospital, Boston, MA USA
- Department of Orthopedic Surgery, Harvard Medical School, Boston, MA USA
- Harvard Stem Cell Institute, Cambridge, MA USA
| |
Collapse
|
4
|
Vilardo B, De Marchi F, Raineri D, Manfredi M, De Giorgis V, Bebeti A, Scotti L, Kustrimovic N, Cappellano G, Mazzini L, Chiocchetti A. Shotgun Proteomics Links Proteoglycan-4 + Extracellular Vesicles to Cognitive Protection in Amyotrophic Lateral Sclerosis. Biomolecules 2024; 14:727. [PMID: 38927130 PMCID: PMC11202157 DOI: 10.3390/biom14060727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder lacking reliable biomarkers for early diagnosis and disease progression monitoring. This study aimed to identify the novel biomarkers in plasmatic extracellular vesicles (EVs) isolated from ALS patients and healthy controls (HCs). A total of 61 ALS patients and 30 age-matched HCs were enrolled in the study and the protein content of circulating EVs was analyzed by shotgun proteomics. The study was divided into a discovery phase (involving 12 ALS and 12 HC patients) and a validation one (involving 49 ALS and 20 HC patients). In the discovery phase, more than 300 proteins were identified, with 32 proteins showing differential regulation in ALS patients compared to HCs. In the validation phase, over 400 proteins were identified, with 20 demonstrating differential regulation in ALS patients compared to HCs. Notably, seven proteins were found to be common to both phases, all of which were significantly upregulated in EVs from ALS patients. Most of them have previously been linked to ALS since they have been detected in the serum or cerebrospinal fluid of ALS patients. Among them, proteoglycan (PRG)-4, also known as lubricin, was of particular interest since it was significantly increased in ALS patients with normal cognitive and motor functions. This study highlights the significance of EVs as a promising avenue for biomarker discovery in ALS. Moreover, it sheds light on the unexpected role of PRG-4 in relation to cognitive status in ALS patients.
Collapse
Affiliation(s)
- Beatrice Vilardo
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy; (B.V.); (D.R.); (N.K.); (G.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont, 28100 Novara, Italy; (M.M.); (V.D.G.)
| | - Fabiola De Marchi
- Department of Neurology and ALS Center, University Hospital “Maggiore della Carità”, 28100 Novara, Italy; (F.D.M.); (A.B.); (L.M.)
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy; (B.V.); (D.R.); (N.K.); (G.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont, 28100 Novara, Italy; (M.M.); (V.D.G.)
| | - Marcello Manfredi
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont, 28100 Novara, Italy; (M.M.); (V.D.G.)
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Veronica De Giorgis
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont, 28100 Novara, Italy; (M.M.); (V.D.G.)
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Alen Bebeti
- Department of Neurology and ALS Center, University Hospital “Maggiore della Carità”, 28100 Novara, Italy; (F.D.M.); (A.B.); (L.M.)
| | - Lorenza Scotti
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Natasa Kustrimovic
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy; (B.V.); (D.R.); (N.K.); (G.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont, 28100 Novara, Italy; (M.M.); (V.D.G.)
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy; (B.V.); (D.R.); (N.K.); (G.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont, 28100 Novara, Italy; (M.M.); (V.D.G.)
| | - Letizia Mazzini
- Department of Neurology and ALS Center, University Hospital “Maggiore della Carità”, 28100 Novara, Italy; (F.D.M.); (A.B.); (L.M.)
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy; (B.V.); (D.R.); (N.K.); (G.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont, 28100 Novara, Italy; (M.M.); (V.D.G.)
| |
Collapse
|
5
|
Cui L, Pi J, Qin B, Cui T, Liu Z, Lei L, Wu S. Advanced application of carbohydrate-based micro/nanoparticles for rheumatoid arthritis. Int J Biol Macromol 2024; 269:131809. [PMID: 38677672 DOI: 10.1016/j.ijbiomac.2024.131809] [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: 12/31/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024]
Abstract
Rheumatoid arthritis (RA) is a kind of synovitis and progressive joint destruction disease. Dysregulated immune cell activation, inflammatory cytokine overproduction, and subsequent reactive oxidative species (ROS) production contribute to the RA process. Carbohydrates, including cellulose, chitosan, alginate and dextran, are among the most abundant and important biomolecules in nature and are widely used in biomedicine. Carbohydrate-based micro/nanoparticles(M/NPs) as functional excipients have the ability to improve the bioavailability, solubility and stability of numerous drugs used in RA therapy. For on-demand therapy, smart reactive M/NPs have been developed to respond to a variety of chemical and physical stimuli, including light, temperature, enzymes, pH and ROS, alternating their physical and macroscopic properties, resulting in innovative new drug delivery systems. In particular, advanced products with targeted dextran or hyaluronic acid are exploiting multiple beneficial properties at the same time. In addition to those that respond, there are promising new derivatives in development with microenvironment and chronotherapy effects. In this review, we provide an overview of these recent developments and an outlook on how this class of agents will further shape the landscape of drug delivery for RA treatment.
Collapse
Affiliation(s)
- Linxian Cui
- Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Department of Cardiology, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan 611130, PR China
| | - Jinkui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Boquan Qin
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ting Cui
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Zhenfei Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lei Lei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Shizhou Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
6
|
Wu WT, Wang CC, Lu DH, Lu KJ, Chang YC, Yang KC. Lovastatin impairs cellular proliferation and enhances hyaluronic acid production in fibroblast-like synoviocytes. Toxicol In Vitro 2024; 97:105806. [PMID: 38432573 DOI: 10.1016/j.tiv.2024.105806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Statins have demonstrated chondroprotective effects by reducing inflammation and mitigating extracellular matrix degradation. However, statins are also reported to be cytotoxic to several types of cells. Early-onset osteoarthritis (OA) is characterized by synovial inflammation, which adversely affects hyaluronan (HA) production in fibroblast-like synoviocytes (FLSs). Nevertheless, the precise effects of statins on the synovium remain unclear. METHODS This study investigated the impact of lovastatin on human FLSs, and HA secretion-related genes, signaling pathways, and production were evaluated. RESULTS The findings revealed that high doses of lovastatin (20 or 40 μM) decreased FLS viability and increased cell death. FLS proliferation ceased when cultured in a medium containing 5 or 10 μM lovastatin. mRNA expression analysis demonstrated that lovastatin (5 and 10 μM) upregulated the gene level of hyaluronan synthase 1 (HAS1), HAS2, and proteoglycan 4 (PRG4), but not HAS3. While the expression of multidrug resistance-associated protein 5 transporter gene remained unaffected, both inward-rectifying potassium channel and acid-sensing ion channel 3 were upregulated. Western blot further confirmed that lovastatin increased the production of HAS1 and PRG4, and activated the PKC-α, ERK1/2, and p38-MAPK signaling pathways. Additionally, lovastatin elevated intracellular cAMP levels and HA production in FLSs. CONCLUSION Lovastatin impairs cellular proliferation but enhances HA production in human FLSs.
Collapse
Affiliation(s)
- Wen-Tien Wu
- Department of Orthopedic Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan; Department of Orthopedics, School of Medicine, Tzu Chi University, Hualien 970473, Taiwan
| | - Chen-Chie Wang
- Department of Orthopedics, School of Medicine, Tzu Chi University, Hualien 970473, Taiwan; Department of Orthopedic Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan
| | - Dai-Hua Lu
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Kuan-Jung Lu
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Yu-Chia Chang
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Kai-Chiang Yang
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan; Orthopedics Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan.
| |
Collapse
|
7
|
Hoekstra M, Snip OSC, Janusz P, Bernabé Kleijn MNA, Truitt ER, Sullivan BD, Schmidt TA, Jay GD, Van Eck M. Recombinant human proteoglycan 4 lowers inflammation and atherosclerosis susceptibility in female low-density lipoprotein receptor knockout mice. J Physiol 2024; 602:1939-1951. [PMID: 38606903 DOI: 10.1113/jp286354] [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: 01/29/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Recombinant human proteoglycan 4 (rhPRG4) is a macromolecular mucin-like glycoprotein that is classically studied as a lubricant within eyes and joints. Given that endogenously produced PRG4 is present within atherosclerotic lesions and genetic PRG4 deficiency increases atherosclerosis susceptibility in mice, in the current study we investigated the anti-atherogenic potential of chronic rhPRG4 treatment. Female low-density lipoprotein receptor knockout mice were fed an atherogenic Western-type diet for 6 weeks and injected three times per week intraperitoneally with 0.5 mg rhPRG4 or PBS as control. Treatment with rhPRG4 was associated with a small decrease in plasma-free cholesterol levels, without a change in cholesteryl ester levels. A marked increase in the number of peritoneal foam cells was detected in response to the peritoneal rhPRG4 administration, which could be attributed to elevated peritoneal leukocyte MSR1 expression levels. However, rhPRG4-treated mice exhibited significantly smaller aortic root lesions of 278 ± 21 × 103 μm2 compared with 339 ± 15 × 103 μm2 in the aortic root of control mice. The overall decreased atherosclerosis susceptibility coincided with a shift in the monocyte and macrophage polarization states towards the patrolling and anti-inflammatory M2-like phenotypes, respectively. Furthermore, rhPRG4 treatment significantly reduced macrophage gene expression levels as well as plasma protein levels of the pro-inflammatory/pro-atherogenic cytokine TNF-alpha. In conclusion, we have shown that peritoneal administration and subsequent systemic exposure to rhPRG4 beneficially impacts the inflammatory state and reduces atherosclerosis susceptibility in mice. Our findings highlight that PRG4 is not only a lubricant but also acts as an anti-inflammatory agent. KEY POINTS: Endogenously produced proteoglycan 4 is found in atherosclerotic lesions and its genetic deficiency in mice is associated with enhanced atherosclerosis susceptibility. In this study we investigated the anti-atherogenic potential of chronic treatment with recombinant human PRG4 in hypercholesterolaemic female low-density lipoprotein receptor knockout mice. We show that recombinant human PRG4 stimulates macrophage foam cell formation, but also dampens the pro-inflammatory state of monocyte/macrophages, eventually leading to a significant reduction in plasma TNF-alpha levels and a lowered atherosclerosis susceptibility. Our findings highlight that peritoneal recombinant human PRG4 treatment can execute effects both locally and systemically and suggest that it will be of interest to study whether rhPRG4 treatment is also able to inhibit the progression and/or induce regression of previously established atherosclerotic lesions.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| | - Olga S C Snip
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Philip Janusz
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mireia N A Bernabé Kleijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | | | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Gregory D Jay
- Department of Emergency Medicine, Warren Alpert Medical School and Division of Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| |
Collapse
|
8
|
Martin-Alarcon L, Govedarica A, Ewoldt RH, Bryant SL, Jay GD, Schmidt TA, Trifkovic M. Scale-Dependent Rheology of Synovial Fluid Lubricating Macromolecules. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306207. [PMID: 38161247 DOI: 10.1002/smll.202306207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 11/20/2023] [Indexed: 01/03/2024]
Abstract
Synovial fluid (SF) is the complex biofluid that facilitates the exceptional lubrication of articular cartilage in joints. Its primary lubricating macromolecules, the linear polysaccharide hyaluronic acid (HA) and the mucin-like glycoprotein proteoglycan 4 (PRG4 or lubricin), interact synergistically to reduce boundary friction. However, the precise manner in which these molecules influence the rheological properties of SF remains unclear. This study aimed to elucidate this by employing confocal microscopy and multiscale rheometry to examine the microstructure and rheology of solutions containing recombinant human PRG4 (rhPRG4) and HA. Contrary to previous assumptions of an extensive HA-rhPRG4 network, it is discovered that rhPRG4 primarily forms stiff, gel-like aggregates. The properties of these aggregates, including their size and stiffness, are found to be influenced by the viscoelastic characteristics of the surrounding HA matrix. Consequently, the rheology of this system is not governed by a single length scale, but instead responds as a disordered, hierarchical network with solid-like rhPRG4 aggregates distributed throughout the continuous HA phase. These findings provide new insights into the biomechanical function of PRG4 in cartilage lubrication and may have implications in the development of HA-based therapies for joint diseases like osteoarthritis.
Collapse
Affiliation(s)
- Leonardo Martin-Alarcon
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Aleksandra Govedarica
- Department of Chemical and Petroleum Engineering, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Randy H Ewoldt
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Steven L Bryant
- Department of Chemical and Petroleum Engineering, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Gregory D Jay
- Department of Emergency Medicine - Warren Alpert Medical School & School of Engineering, Brown University, Providence, RI, 02912, USA
| | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Milana Trifkovic
- Department of Chemical and Petroleum Engineering, University of Calgary, Calgary, AB, T2N 1N4, Canada
| |
Collapse
|
9
|
Ninkovic N, Sparks HD, Ponjevic D, Muench G, Biernaskie JA, Krawetz RJ. Proteoglycan 4 (PRG4) treatment improves skin wound healing in a porcine model. FASEB J 2024; 38:e23547. [PMID: 38498368 DOI: 10.1096/fj.202301289rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 02/08/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
Proteoglycan 4 (PRG4) is a boundary lubricant originally identified in articular cartilage and has been since shown to have immunomodulation and antifibrotic properties. Previously, we have demonstrated that recombinant human (rh)PRG4 treatment accelerates auricular cartilage injury closure through an inhibition of the fibrotic response, and promotion of tissue regeneration in mice. The purpose of the current study was to examine the effects of rhPRG4 treatment (vs. a DMSO carried control) on full-thickness skin wound healing in a preclinical porcine model. Our findings suggest that while rhPRG4 did not significantly accelerate nor impede full-thickness skin wound closure, it did improve repair quality by decreasing molecular markers of fibrosis and increasing re-vascularization. We also demonstrated that rhPRG4 treatment increased dermal adipose tissue during the healing process specifically by retaining adipocytes in the wound area but did not inhibit lipolysis. Overall, the results of the current study have demonstrated that rhPRG4 acts as antifibrotic agent and regulates dermal adipose tissue during the healing processes resulting in a tissue with a trajectory that more resembles the native skin vs. a fibrotic patch. This study provides strong rationale to examine if rhPRG4 can improve regeneration in human wounds.
Collapse
Affiliation(s)
- Nicoletta Ninkovic
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Holly D Sparks
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Dragana Ponjevic
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Greg Muench
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeff A Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Roman J Krawetz
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, Canada
- Department Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Fu C, Wang Z, Zhou X, Hu B, Li C, Yang P. Protein-based bioactive coatings: from nanoarchitectonics to applications. Chem Soc Rev 2024; 53:1514-1551. [PMID: 38167899 DOI: 10.1039/d3cs00786c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Protein-based bioactive coatings have emerged as a versatile and promising strategy for enhancing the performance and biocompatibility of diverse biomedical materials and devices. Through surface modification, these coatings confer novel biofunctional attributes, rendering the material highly bioactive. Their widespread adoption across various domains in recent years underscores their importance. This review systematically elucidates the behavior of protein-based bioactive coatings in organisms and expounds on their underlying mechanisms. Furthermore, it highlights notable advancements in artificial synthesis methodologies and their functional applications in vitro. A focal point is the delineation of assembly strategies employed in crafting protein-based bioactive coatings, which provides a guide for their expansion and sustained implementation. Finally, the current trends, challenges, and future directions of protein-based bioactive coatings are discussed.
Collapse
Affiliation(s)
- Chengyu Fu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Zhengge Wang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Xingyu Zhou
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Bowen Hu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Chen Li
- School of Chemistry and Chemical Engineering, Henan Institute of Science and Technology, Eastern HuaLan Avenue, Xinxiang, Henan 453003, China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
11
|
Bonnevie ED, Scanzello CR, Mauck RL. Modulating mechanobiology as a therapeutic target for synovial fibrosis to restore joint lubrication. Osteoarthritis Cartilage 2024; 32:41-51. [PMID: 37866546 PMCID: PMC10880438 DOI: 10.1016/j.joca.2023.09.012] [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: 12/21/2022] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVES Fibroses are disorders linked to persistence of myofibroblasts due to biochemical (e.g., Transforming growth factor-β) and biophysical cues (e.g., a stiff microenvironment). In the context of osteoarthritis, fibrotic changes in the joint-lining synovium have been linked with disease progression. The objective of this study was to probe synovial fibroblast mechanobiology and how essential functions (i.e., lubrication) are altered in fibrotic environments. DESIGN Both ex vivo and in vitro synovium models were assessed for fibrotic and lubrication biomarkers to better understand the role of mechanobiology and lubrication. Additionally, in vitro, work on small molecules targeting mechanobiology was assessed. RESULTS Our results indicated that modulating mechanobiology could rescue the fibrotic phenotype instigated by stiffening microenvironment that resulted in altered lubricant expression. A small molecule therapeutic, fasudil, blocked ROCK-mediated contractility and this inhibition of the fibrotic mechano-response of synovial fibroblasts restored proper lubrication function, providing insight into mechanisms of disease progression as well as a new avenue for therapeutic development. CONCLUSION This study identifies synovial fibrosis as a condition that potentially has joint-wide deficits through inhibiting lubrication. Additionally, modulating mechanobiology (i.e., ROCK-mediated contractility) may pose a potential target for small molecule therapies that can be delivered to the joint space. CLASSIFICATION Applied Biological Sciences.
Collapse
Affiliation(s)
- Edward D Bonnevie
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, United States.
| | - Carla R Scanzello
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; Division of Rheumatology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Robert L Mauck
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, United States; Department of Bioengineering, University of Pennsylvania, United States.
| |
Collapse
|
12
|
Yin Y, Zhang Y, Guo L, Li P, Wang D, Huang L, Zhao X, Wu G, Li L, Wei X. Effect of Moderate Exercise on the Superficial Zone of Articular Cartilage in Age-Related Osteoarthritis. Diagnostics (Basel) 2023; 13:3193. [PMID: 37892013 PMCID: PMC10605492 DOI: 10.3390/diagnostics13203193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
This study aimed to evaluate the effect of exercise on the superficial zone of the osteoarticular cartilage during osteoarthritis progression. Three-month-old, nine-month-old, and eighteen-month-old Sprague Dawley rats were randomly divided into two groups, moderate exercise and no exercise, for 10 weeks. Histological staining, immunostaining, and nanoindentation measurements were conducted to detect changes in the superficial zone. X-ray and micro-CT were quantitated to detect alterations in the microarchitecture of the tibial subchondral bone. Cells were extracted from the superficial zone of the cartilage under fluid-flow shear stress conditions to further verify changes in vitro. The number of cells and proteoglycan content in the superficial zone increased more in the exercise group than in the control group. Exercise can change the content and distribution of collagen types I and III in the superficial layer. In addition, TGFβ/pSmad2/3 and Prg4 expression levels increased under the intervention of exercise on the superficial zone. Exercise can improve the Young's modulus of the cartilage and reduce the abnormal subchondral bone remodeling which occurs after superficial zone changes. Moderate exercise delays the degeneration of the articular cartilage by its effect on the superficial zone, and the TGFβ/pSmad2/3 signaling pathways and Prg4 play an important role.
Collapse
Affiliation(s)
- Yukun Yin
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
| | - Yuanyu Zhang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
| | - Li Guo
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
| | - Pengcui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
| | - Dongming Wang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
| | - Lingan Huang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
- Beijing Key Laboratory of Sports Injuries, Department of Sports Medicine, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xiaoqin Zhao
- College of Physical Education, Taiyuan University of Technology, Taiyuan 030024, China;
| | - Gaige Wu
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
| | - Lu Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; (Y.Y.); (Y.Z.); (L.G.); (P.L.); (D.W.); (L.H.); (G.W.)
| |
Collapse
|
13
|
Qadri MM. Targeting CD44 Receptor Pathways in Degenerative Joint Diseases: Involvement of Proteoglycan-4 (PRG4). Pharmaceuticals (Basel) 2023; 16:1425. [PMID: 37895896 PMCID: PMC10609794 DOI: 10.3390/ph16101425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Rheumatoid arthritis (RA), osteoarthritis (OA), and gout are the most prevalent degenerative joint diseases (DJDs). The pathogenesis underlying joint disease in DJDs remains unclear. Considering the severe toxicities reported with anti-inflammatory and disease-modifying agents, there is a clear need to develop new treatments that are specific in their effect while not being associated with significant toxicities. A key feature in the development of joint disease is the overexpression of adhesion molecules, e.g., CD44. Expression of CD44 and its variants in the synovial tissues of patients with DJDs is strongly associated with cartilage damage and appears to be a predicting factor of synovial inflammation in DJDs. Targeting CD44 and its downstream signaling proteins has emerged as a promising therapeutic strategy. PRG4 is a mucinous glycoprotein that binds to the CD44 receptor and is physiologically involved in joint lubrication. PRG4-CD44 is a pivotal regulator of synovial lining cell hemostasis in the joint, where lack of PRG4 expression triggers chronic inflammation and fibrosis, driven by persistent activation of synovial cells. In view of the significance of CD44 in DJD pathogenesis and the potential biological role for PRG4, this review aims to summarize the involvement of PRG4-CD44 signaling in controlling synovitis, synovial hypertrophy, and tissue fibrosis in DJDs.
Collapse
Affiliation(s)
- Marwa M. Qadri
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Inflammation Pharmacology and Drug Discovery Unit, Medical Research Center (MRC), Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
14
|
Menon NG, Tanguay AP, Zhou L, Zhang LX, Bobst CE, Han M, Ghosh M, Greene GW, Deymier A, Sullivan BD, Chen Y, Jay GD, Schmidt TA. A structural and functional comparison between two recombinant human lubricin proteins: Recombinant human proteoglycan-4 (rhPRG4) vs ECF843. Exp Eye Res 2023; 235:109643. [PMID: 37678729 PMCID: PMC10691279 DOI: 10.1016/j.exer.2023.109643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/22/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Proteoglycan 4 (PRG4, lubricin) is a mucin-like glycoprotein present on the ocular surface that has both boundary lubricating and anti-inflammatory properties. Full-length recombinant human PRG4 (rhPRG4) has been shown to be clinically effective in improving signs and symptoms of dry eye disease (DED). In vitro, rhPRG4 has been shown to reduce inflammation-induced cytokine production and NFκB activity in corneal epithelial cells, as well as to bind to and inhibit MMP-9 activity. A different form of recombinant human lubricin (ECF843), produced from the same cell line as rhPRG4 but manufactured using a different process, was recently assessed in a DED clinical trial. However, ECF843 did not significantly improve signs or symptoms of DED compared to vehicle. Initial published characterization of ECF843 showed it had a smaller hydrodynamic diameter and was less negatively charged than native PRG4. Further examination of the structural and functional properties of ECF843 and rhPRG4 could contribute to the understanding of what led to their disparate clinical efficacy. Therefore, the objective of this study was to characterize and compare rhPRG4 and ECF843 in vitro, both biophysically and functionally. Hydrodynamic diameter and charge were measured by dynamic light scattering (DLS) and zeta potential, respectively. Size and molecular weight was determined for individual species by size exclusion chromatography (SEC) with in-line DLS and multi-angle light scattering (MALS). Bond structure was measured by Raman spectroscopy, and sedimentation properties were measured by analytical ultracentrifugation (AUC). Functionally, MMP-9 inhibition was measured using a commercial MMP-9 activity kit, coefficient of friction was measured using an established boundary lubrication test at a latex-glass interface, and collagen 1-binding ability was measured by quart crystal microbalance with dissipation (QCMD). Additionally, the ability of rhPRG4 and ECF843 to inhibit urate acid crystal formation and cell adhesion was assessed. ECF843 had a significantly smaller hydrodynamic diameter and was less negatively charged than rhPRG4, as assessed by DLS and zeta potential. Size was further explored with SEC-DLS-MALS, which indicated that while rhPRG4 had 3 main peaks, corresponding to monomer, dimer, and multimer as expected, ECF843 had 2 peaks that were similar in size and molecular weight compared to rhPRG4's monomer peak and a third peak that was significantly smaller in both size and molar mass than the corresponding peak of rhPRG4. Raman spectroscopy demonstrated that ECF843 had significantly more disulfide bonds, which are functionally determinant structures, relative to the carbon-carbon backbone compared to rhPRG4, and AUC indicated that ECF843 was more compact than rhPRG4. Functionally, ECF843 was significantly less effective at inhibiting MMP-9 activity and functioning as a boundary lubricant compared to rhPRG4, as well as being slower to bind to collagen 1. Additionally, ECF843 was significantly less effective at inhibiting urate acid crystal formation and at preventing cell adhesion. Collectively, these data demonstrate ECF843 and rhPRG4 are significantly different in both structure and function. Given that a protein's structure sets the foundation for its interactions with other molecules and tissues in vivo, which ultimately determine its function, these differences most likely contributed to the disparate DED clinical trial results.
Collapse
Affiliation(s)
- Nikhil G Menon
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Adam P Tanguay
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Libo Zhou
- Biomedical Engineering Department, University of Connecticut, Storrs, CT, USA
| | - Ling X Zhang
- Emergency Medicine, Brown University, Providence, RI, USA
| | - Cedric E Bobst
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA
| | - Mingyu Han
- Institute for Frontier Materials and ARC Centre of Excellence for Electromaterials Science, Deakin University, Melbourne, Victoria, Australia; Commonwealth Scientific and Industrial Research Organization (CSIRO), Agriculture and Food, Werribee, Victoria, Australia
| | - Mallika Ghosh
- Department of Cell Biology, School of Medicine, UConn Health, Farmington, CT, USA
| | - George W Greene
- Institute for Frontier Materials and ARC Centre of Excellence for Electromaterials Science, Deakin University, Melbourne, Victoria, Australia; Department of Chemistry and Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Alix Deymier
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | | | - Yupeng Chen
- Biomedical Engineering Department, University of Connecticut, Storrs, CT, USA
| | - Gregory D Jay
- Emergency Medicine, Brown University, Providence, RI, USA
| | - Tannin A Schmidt
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA.
| |
Collapse
|
15
|
Tarafder S, Ghataure J, Langford D, Brooke R, Kim R, Eyen SL, Bensadoun J, Felix JT, Cook JL, Lee CH. Advanced bioactive glue tethering Lubricin/PRG4 to promote integrated healing of avascular meniscus tears. Bioact Mater 2023; 28:61-73. [PMID: 37214259 PMCID: PMC10199165 DOI: 10.1016/j.bioactmat.2023.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/24/2023] Open
Abstract
Meniscus injuries are extremely common with approximately one million patients undergoing surgical treatment annually in the U.S. alone, but no regenerative therapy exist. Previously, we showed that controlled applications of connective tissue growth factor (CTGF) and transforming growth factor beta 3 (TGFβ3) via fibrin-based bio-glue facilitate meniscus healing by inducing recruitment and stepwise differentiation of synovial mesenchymal stem/progenitor cells. Here, we first explored the potential of genipin, a natural crosslinker, to enhance fibrin-based glue's mechanical and degradation properties. In parallel, we identified the harmful effects of lubricin on meniscus healing and investigated the mechanism of lubricin deposition on the injured meniscus surface. We found that the pre-deposition of hyaluronic acid (HA) on the torn meniscus surface mediates lubricin deposition. Then we implemented chemical modifications with heparin conjugation and CD44 on our bioactive glue to achieve strong initial bonding and integration of lubricin pre-coated meniscal tissues. Our data suggested that heparin conjugation significantly enhances lubricin-coated meniscal tissues. Similarly, CD44, exhibiting a strong binding affinity to lubricin and hyaluronic acid (HA), further improved the integrated healing of HA/lubricin pre-coated meniscus injuries. These findings may represent an important foundation for developing a translational bio-active glue guiding the regenerative healing of meniscus injuries.
Collapse
Affiliation(s)
- Solaiman Tarafder
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| | - Jaskirti Ghataure
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| | - David Langford
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| | - Rachel Brooke
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| | - Ryunhyung Kim
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| | - Samantha Lewis Eyen
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| | - Julian Bensadoun
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| | - Jeffrey T. Felix
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| | - James L. Cook
- Thompson Laboratory for Regenerative Orthopaedics, Missouri Orthopedic Institute, University of Missouri, 1100 Virginia Avenue, Columbia, MO, 65212, USA
| | - Chang H. Lee
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168 St. – VC12-212, New York, NY, 10032, USA
| |
Collapse
|
16
|
Sprott H, Fleck C. Hyaluronic Acid in Rheumatology. Pharmaceutics 2023; 15:2247. [PMID: 37765216 PMCID: PMC10537104 DOI: 10.3390/pharmaceutics15092247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Hyaluronic acid (HA), also known as hyaluronan, is an anionic glycosaminoglycan widely distributed throughout various tissues of the human body. It stands out from other glycosaminoglycans as it lacks sulfation and can attain considerable size: the average human synovial HA molecule weighs about 7 million Dalton (Da), equivalent to roughly 20,000 disaccharide monomers; although some sources report a lower range of 3-4 million Da. In recent years, HA has garnered significant attention in the field of rheumatology due to its involvement in joint lubrication, cartilage maintenance, and modulation of inflammatory and/or immune responses. This review aims to provide a comprehensive overview of HA's involvement in rheumatology, covering its physiology, pharmacology, therapeutic applications, and potential future directions for enhancing patient outcomes. Nevertheless, the use of HA therapy in rheumatology remains controversial with conflicting evidence regarding its efficacy and safety. In conclusion, HA represents a promising therapeutic option to improve joint function and alleviate inflammation and pain.
Collapse
Affiliation(s)
- Haiko Sprott
- Medical Faculty, University of Zurich (UZH), CH-8006 Zurich, Switzerland
- Arztpraxis Hottingen, CH-8032 Zurich, Switzerland
| | | |
Collapse
|
17
|
Wei Q, Zhu X, Wang L, Zhang W, Yang X, Wei W. Extracellular matrix in synovium development, homeostasis and arthritis disease. Int Immunopharmacol 2023; 121:110453. [PMID: 37331300 DOI: 10.1016/j.intimp.2023.110453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/20/2023]
Abstract
Extracellular matrix (ECM) is a three-dimensional network entity composed of extracellular macromolecules. ECM in synovium not only supports the structural integrity of synovium, but also plays a crucial role in regulating homeostasis and damage repair response in synovium. Obvious disorders in the composition, behavior and function of synovial ECM will lead to the occurrence and development of arthritis diseases such as rheumatoid arthritis (RA), osteoarthritis (OA) and psoriatic arthritis (PsA). Based on the importance of synovial ECM, targeted regulation of the composition and structure of ECM is considered to be an effective measure for the treatment of arthritis disease. This paper reviews the current research status of synovial ECM biology, discusses the role and mechanism of synovial ECM in physiological status and arthritis disease, and summarizes the current strategies for targeting synovial ECM to provide information for the pathogenesis, diagnosis and treatment of arthritis disease.
Collapse
Affiliation(s)
- Qi Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xuemin Zhu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Luping Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wankang Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xuezhi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
18
|
Kim SS, Shin H, Ahn KG, Park YM, Kwon MC, Lim JM, Oh EK, Kim Y, Han SM, Noh DY. Quantifiable peptide library bridges the gap for proteomics based biomarker discovery and validation on breast cancer. Sci Rep 2023; 13:8991. [PMID: 37268731 DOI: 10.1038/s41598-023-36159-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023] Open
Abstract
Mass spectrometry (MS) based proteomics is widely used for biomarker discovery. However, often, most biomarker candidates from discovery are discarded during the validation processes. Such discrepancies between biomarker discovery and validation are caused by several factors, mainly due to the differences in analytical methodology and experimental conditions. Here, we generated a peptide library which allows discovery of biomarkers in the equal settings as the validation process, thereby making the transition from discovery to validation more robust and efficient. The peptide library initiated with a list of 3393 proteins detectable in the blood from public databases. For each protein, surrogate peptides favorable for detection in mass spectrometry was selected and synthesized. A total of 4683 synthesized peptides were spiked into neat serum and plasma samples to check their quantifiability in a 10 min liquid chromatography-MS/MS run time. This led to the PepQuant library, which is composed of 852 quantifiable peptides that cover 452 human blood proteins. Using the PepQuant library, we discovered 30 candidate biomarkers for breast cancer. Among the 30 candidates, nine biomarkers, FN1, VWF, PRG4, MMP9, CLU, PRDX6, PPBP, APOC1, and CHL1 were validated. By combining the quantification values of these markers, we generated a machine learning model predicting breast cancer, showing an average area under the curve of 0.9105 for the receiver operating characteristic curve.
Collapse
Affiliation(s)
- Sung-Soo Kim
- Manufacturing and Technology Division, Bertis Inc., Hungdeok 1-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16954, Republic of Korea
- Bio Convergence Research Institute, Bertis Inc., Heungdeok 1-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16954, Republic of Korea
| | - HyeonSeok Shin
- Bio Convergence Research Institute, Bertis Inc., Heungdeok 1-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16954, Republic of Korea
| | - Kyung-Geun Ahn
- Manufacturing and Technology Division, Bertis Inc., Hungdeok 1-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16954, Republic of Korea
| | - Young-Min Park
- Manufacturing and Technology Division, Bertis Inc., Hungdeok 1-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16954, Republic of Korea
| | - Min-Chul Kwon
- Manufacturing and Technology Division, Bertis Inc., Hungdeok 1-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16954, Republic of Korea
| | - Jae-Min Lim
- Manufacturing and Technology Division, Bertis Inc., Hungdeok 1-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16954, Republic of Korea
| | - Eun-Kyung Oh
- Manufacturing and Technology Division, Bertis Inc., Hungdeok 1-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16954, Republic of Korea
| | - Yumi Kim
- Department of Surgery, CHA Gangnam Medical Center, CHA University School of Medicine, 566, Nonhyeon-ro, Gangnam-gu, Seoul, 06135, Republic of Korea
| | - Seung-Man Han
- Bertis Inc., 172, Dolma-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13605, Republic of Korea
| | - Dong-Young Noh
- Department of Surgery, CHA Gangnam Medical Center, CHA University School of Medicine, 566, Nonhyeon-ro, Gangnam-gu, Seoul, 06135, Republic of Korea.
- Bertis Inc., 172, Dolma-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13605, Republic of Korea.
- Seoul National University College of Medicine, 103 Daehak-Ro, Seoul, 03080, Republic of Korea.
| |
Collapse
|
19
|
Kim SJ, Kim JE, Choe G, Song DH, Kim SJ, Kim TH, Yoo J, Kim SH, Jung Y. Self-assembled peptide-substance P hydrogels alleviate inflammation and ameliorate the cartilage regeneration in knee osteoarthritis. Biomater Res 2023; 27:40. [PMID: 37143133 PMCID: PMC10161637 DOI: 10.1186/s40824-023-00387-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Self-assembled peptide (SAP)-substance P (SP) hydrogels can be retained in the joint cavity longer than SP alone, and they can alleviate inflammation and ameliorate cartilage regeneration in knee osteoarthritis (OA). We conducted a preclinical study using diverse animal models of OA and an in vitro study using human synoviocytes and patient-derived synovial fluids to demonstrate the effect of SAP-SP complex on the inflammation and cartilage regeneration. METHODS Surgical induction OA model was prepared with New Zealand white female rabbits and chemical induction, and naturally occurring OA models were prepared using Dunkin Hartely female guinea pigs. The SAP-SP complex or control (SAP, SP, or saline) was injected into the joint cavities in each model. We performed micro-computed tomography (Micro-CT) analysis, histological evaluation, immunofluorescent analysis, and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling (TUNEL) assay and analyzed the recruitment of intrinsic mesenchymal stem cells (MSCs), macrophage activity, and inflammatory cytokine in each OA model. Human synoviocytes were cultured in synovial fluid extracted from human OA knee joints injected with SAP-SP complexes or other controls. Proliferative capacity and inflammatory cytokine levels were analyzed. RESULTS Alleviation of inflammation, inhibition of apoptosis, and enhancement of intrinsic MSCs have been established in the SAP-SP group in diverse animal models. Furthermore, the inflammatory effects on human samples were examined in synoviocytes and synovial fluid from patients with OA. In this study, we observed that SAP-SP showed anti-inflammatory action in OA conditions and increased cartilage regeneration by recruiting intrinsic MSCs, inhibiting progression of OA. CONCLUSIONS These therapeutic effects have been validated in diverse OA models, including rabbits, Dunkin Hartley guinea pigs, and human synoviocytes. Therefore, we propose that SAP-SP may be an effective injectable therapeutic agent for treating OA. In this manuscript, we report a preclinical study of novel self-assembled peptide (SAP)-substance P (SP) hydrogels with diverse animal models and human synoviocytes and it displays anti-inflammatory effects, apoptosis inhibition, intrinsic mesenchymal stem cells recruitments and cartilage regeneration.
Collapse
Affiliation(s)
- Sang Jun Kim
- Department of Physical and Rehabilitation Medicine, Seoul Jun Rehabilitation Clinic and Research Center, Seoul, Republic of Korea
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji Eun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Goeun Choe
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Da Hyun Song
- Department of Physical and Rehabilitation Medicine, Seoul Jun Rehabilitation Clinic and Research Center, Seoul, Republic of Korea
| | - Sun Jeong Kim
- Stem Cell Institute, ENCell Co. Ltd, Seoul, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Tae Hee Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jin Yoo
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Soo Hyun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Youngmee Jung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- School of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Das N, de Almeida LGN, Derakhshani A, Young D, Mehdinejadiani K, Salo P, Rezansoff A, Jay GD, Sommerhoff CP, Schmidt TA, Krawetz R, Dufour A. Tryptase β regulation of joint lubrication and inflammation via proteoglycan-4 in osteoarthritis. Nat Commun 2023; 14:1910. [PMID: 37024468 PMCID: PMC10079686 DOI: 10.1038/s41467-023-37598-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
PRG4 is an extracellular matrix protein that maintains homeostasis through its boundary lubricating and anti-inflammatory properties. Altered expression and function of PRG4 have been associated with joint inflammatory diseases, including osteoarthritis. Here we show that mast cell tryptase β cleaves PRG4 in a dose- and time-dependent manner, which was confirmed by silver stain gel electrophoresis and mass spectrometry. Tryptase-treated PRG4 results in a reduction of lubrication. Compared to full-length, cleaved PRG4 further activates NF-κB expression in cells overexpressing TLR2, -4, and -5. In the destabilization of the medial meniscus model of osteoarthritis in rat, tryptase β and PRG4 colocalize at the site of injury in knee cartilage and is associated with disease severity. When human primary synovial fibroblasts from male osteoarthritis patients or male healthy subjects treated with tryptase β and/or PRG4 are subjected to a quantitative shotgun proteomics and proteome changes are characterized, it further supports the role of NF-κB activation. Here we show that tryptase β as a modulator of joint lubrication in osteoarthritis via the cleavage of PRG4.
Collapse
Affiliation(s)
- Nabangshu Das
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Luiz G N de Almeida
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Afshin Derakhshani
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Daniel Young
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kobra Mehdinejadiani
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul Salo
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Alexander Rezansoff
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gregory D Jay
- Department of Emergency Medicine, Warren Alpert Medical School & School of Engineering, Brown University, Providence, RI, USA
| | - Christian P Sommerhoff
- Institute of Medical Education and Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Tannin A Schmidt
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, CT, USA
| | - Roman Krawetz
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Antoine Dufour
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
21
|
Tang H, Ma Y, Li J, Zhang Z, Li W, Cai C, Zhang L, Li Z, Tian Y, Zhang Y, Ji J, Han L, Kang X, Jiang R, Han R. Identification and genetic analysis of major gene ST3GAL4 related to serum alkaline phosphatase in chicken. Res Vet Sci 2023; 155:115-123. [PMID: 36680949 DOI: 10.1016/j.rvsc.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
Alkaline phosphatase (ALP) is a marker of osteoblast maturation and an important indicator of bone metabolism. The activity of ALP can reflect the bone metabolism and growth traits of animals, so the polymorphism affecting ALP expression deserves further study. In this study, we identified an SNP site in ST3GAL4 found by genome-wide association studies (GWAS) in previous studies, 8 SNPs were also identified by DNA sequencing. Interestingly, there were 4 SNPs (rs475471G > A, rs475533C > T, rs475621A > G, rs475647C > A) completely linked by haplotype analysis. Therefore, we selected a tag SNP rs475471G > A to further analyze the ALP level of different genotypes in Hubbard leg disease population and an F2 chicken resource population produced by Anka and Gushi chickens and carried out population genetic analysis in 18 chicken breeds. Association analysis showed that this QTL within ST3GAL4 was highly correlated with ALP level. The mutant individuals with genotype AA had the highest ALP level, followed by GA and GG carriers. The mutant individual carriers of AA and GA genotype had higher values for body weight (BW), chest width (CW), body slanting length (BSL), pelvis width (PW) at 4-week, the semi-evisceration weight (SEW), evisceration weight (EW) and Leg weight (LW) than GG genotypes. The amplification and typing of 4852 DNA samples from 18 different breeds showed GG genotype mainly existed in egg-type chickens and dual-type chickens, while the AA genotype was mainly distributed in commercial broilers and F2 resource population. The individual carriers of the AA genotype had the highest ALP and showed better growth performance. Besides, tissue expression analysis used Cobb broiler showed significant differences between different genotypes in the spleen and duodenum. Taken together, this was the first time to determine 9 SNPs within ST3GAL4 related to ALP in chickens, 4 of them were complete linkage with each other, which provides useful information on the mutation of ST3GAL4 and could predict the serum ALP level of chicken early and as an effective potential molecular breeding marker for chickens.
Collapse
Affiliation(s)
- Hehe Tang
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Yanchao Ma
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Jianzeng Li
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Zhenzhen Zhang
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Wenting Li
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Chunxia Cai
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Lujie Zhang
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Zhuanjian Li
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Yadong Tian
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Yanhua Zhang
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China
| | - Jinqing Ji
- Henan Husbandry Breau, Zhengzhou 450008, China
| | - Lu Han
- Henan Husbandry Breau, Zhengzhou 450008, China
| | - Xiangtao Kang
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China.
| | - Ruirui Jiang
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China.
| | - Ruili Han
- College of animal science and technology, Henan Agricultural University, Zhengzhou, Henan Province 4500046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, Henan Province 450046, China.
| |
Collapse
|
22
|
Manasa CS, Silva SM, Caballero-Aguilar LM, Quigley AF, Kapsa RMI, Greene GW, Moulton SE. Active and passive drug release by self-assembled lubricin (PRG4) anti-fouling coatings. J Control Release 2022; 352:35-46. [PMID: 36228955 DOI: 10.1016/j.jconrel.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 11/08/2022]
Abstract
Electroactive polymers (EAPs) have been investigated as materials for use in a range of biomedical applications, ranging from cell culture, electrical stimulation of cultured cells as well as controlled delivery of growth factors and drugs. Despite their excellent drug delivery ability, EAPs are susceptible to biofouling thus they often require surface functionalisation with antifouling coatings to limit unwanted non-specific protein adsorption. Here we demonstrate the surface modification of para toluene sulfonate (pTS) doped polypyrrole with the glycoprotein lubricin (LUB) to produce a self-assembled coating that both prevents surface biofouling while also serving as a high-capacity reservoir for cationic drugs which can then be released passively via diffusion or actively via an applied electrical potential. We carried out our investigation in two parts where we initially assessed the antifouling and cationic drug delivery ability of LUB tethered on a gold surface using quartz crystal microbalance with dissipation monitoring (QCM) to monitor molecular interactions occurring on a gold sensor surface. After confirming the ability of tethered LUB nano brush layers on a gold surface, we introduced an electrochemically grown EAP layer to act as the immobilisation surface for LUB before subsequently introducing the cationic drug doxorubicin hydrochloride (DOX). The release of cationic drug was then investigated under passive and electrochemically stimulated conditions. High-performance liquid chromatography (HPLC) was then carried out to quantify the amount of DOX released. It was shown that the amount of DOX released from nano brush layers of LUB tethered on gold and EAP surfaces could be increased by up to 30% per minute by applying a positive electrochemically stimulating pulse at 0.8 V for one minute. Using bovine serum albumin (BSA), we show that DOX loaded LUB tethered on para toluene sulfonic acid (pTS) doped polypyrrole retained antifouling ability of up to 75% when compared to unloaded tethered LUB. This work demonstrates the unique, novel ability of tethered LUB to actively participate in the delivery of cationic therapeutics on different substrate surfaces. This study could lead to the development of versatile multifunctional biomaterials for use in wide range of biomedical applications, such as dual drug delivery and lubricating coatings, dual drug delivery and antifouling coatings, cellular recording and stimulation.
Collapse
Affiliation(s)
- Clayton S Manasa
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia; The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Saimon M Silva
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia; ARC Centre of Excellence for Electromaterials Science, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia; The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia; Iverson Health Innovation Research Institute, Swinburne University of Technology, Victoria 3122, Australia
| | - Lilith M Caballero-Aguilar
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia; The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Anita F Quigley
- School of Electrical and Biomedical Engineering, RMIT University, Melbourne, Victoria 3001, Australia; The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia; Department of Medicine, St Vincent's Hospital Melbourne, Fitzroy 3065, Melbourne, Australia
| | - Robert M I Kapsa
- School of Electrical and Biomedical Engineering, RMIT University, Melbourne, Victoria 3001, Australia; The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia; Department of Medicine, St Vincent's Hospital Melbourne, Fitzroy 3065, Melbourne, Australia
| | - George W Greene
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria 3216, Australia; ARC Centre of Excellence for Electromaterials Science, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| | - Simon E Moulton
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia; ARC Centre of Excellence for Electromaterials Science, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia; The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia; Iverson Health Innovation Research Institute, Swinburne University of Technology, Victoria 3122, Australia.
| |
Collapse
|
23
|
Satake T, Komura S, Aoki H, Hirakawa A, Imai Y, Akiyama H. Induction of iPSC-derived Prg4-positive cells with characteristics of superficial zone chondrocytes and fibroblast-like synovial cells. BMC Mol Cell Biol 2022; 23:30. [PMID: 35870887 PMCID: PMC9308249 DOI: 10.1186/s12860-022-00431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022] Open
Abstract
Background Lubricin, a proteoglycan encoded by the PRG4 gene, is synthesised by superficial zone (SFZ) chondrocytes and synovial cells. It reduces friction between joints and allows smooth sliding of tendons. Although lubricin has been shown to be effective against osteoarthritis and synovitis in animals, its clinical application remains untested. In this study, we aimed to induce lubricin-expressing cells from pluripotent stem cells (iPSCs) and applied them locally via cell transplantation. Methods To generate iPSCs, OCT3/4, SOX2, KLF4, and L-MYC were transduced into fibroblasts derived from Prg4-mRFP1 transgenic mice. We established a protocol for the differentiation of iPSC-derived Prg4-mRFP1-positive cells and characterised their mRNA expression profile. Finally, we injected Prg4-mRFP1-positive cells into the paratenon, surrounding the Achilles tendons and knee joints of severe combined immunodeficient mice and assessed lubricin expression. Result Wnt3a, activin A, TGF-β1, and bFGF were applied to induce the differentiation of iPSC-derived Prg4-mRFP1-positive cells. Markers related to SFZ chondrocytes and fibroblast-like synovial cells (FLSs) were expressed during differentiation. RNA-sequencing indicated that iPSC-derived Prg4-mRFP1-positive cells manifested expression profiles typical of SFZ chondrocytes and FLSs. Transplanted iPSC-derived Prg4-mRFP1-positive cells survived around the Achilles tendons and in knee joints. Conclusions The present study describes a protocol for the differentiation of iPSC-derived Prg4-positive cells with characteristics of SFZ chondrocytes and FLSs. Transplantation of lubricin-expressing cells offers promise as a therapy against arthritis and synovitis.
Collapse
|
24
|
Guo Y, Hu HT, Xu SJ, Xia WL, Zhao Y, Zhao XH, Zhu WB, Li FT, Li HL. Proteoglycan-4 predicts good prognosis in patients with hepatocellular carcinoma receiving transcatheter arterial chemoembolization and inhibits cancer cell migration in vitro. Front Oncol 2022; 12:1023801. [PMID: 36439456 PMCID: PMC9691762 DOI: 10.3389/fonc.2022.1023801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/24/2022] [Indexed: 08/26/2023] Open
Abstract
PURPOSE To search for adaptive response molecules that affect the efficacy of transcatheter arterial chemoembolization (TACE), analyze their clinical correlation with and prognostic value for hepatocellular carcinoma (HCC), and explore their impact on cell biological behavior and their mechanisms of action. METHODS HCC tissue gene sequencing was used to identify differentially expressed genes. The expression of proteoglycan 4 (PRG4) in the serum of 117 patients with HCC who received TACE was detected by enzyme-linked immunosorbent assay. Serum-free medium mimicked TACE-induced nutrient deprivation. Cells with stable knockdown of PRG4 (shPRG4) were constructed to verify the effect and mechanism of PRG4 on the biological behavior of HCC cells in vitro. RESULTS The expression of PRG4 was significantly elevated under TACE-induced starvation conditions. Low PRG4 expression was associated with worse response to TACE treatment, shorter survival time, and stronger HCC migration ability. Furthermore, in vitro experiments showed that knockdown of PRG4 promoted HCC cell migration by enhancing epithelial-mesenchymal transition (EMT) while did not affect proliferation. When PRG4 expression was low, starvation treatment impaired the migratory ability of HCC cells and reduced the chemosensitivity of HCC cells to epirubicin. CONCLUSIONS PRG4 expression predicts survival and TACE treatment response in patients with HCC. Furthermore, knockdown of PRG4 enhanced EMT, leading to HCC cell migration. PRG4 may serve as a biomarker for HCC patients receiving TACE.
Collapse
Affiliation(s)
- Yuan Guo
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hong Tao Hu
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Shi Jun Xu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wei Li Xia
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yan Zhao
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xiao Hui Zhao
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wen Bo Zhu
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Fang Ting Li
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hai Liang Li
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
25
|
ST3GAL3 Promotes the Inflammatory Response of Fibroblast-Like Synoviocytes in Rheumatoid Arthritis by Activating the TLR9/MyD88 Pathway. Mediators Inflamm 2022; 2022:4258742. [DOI: 10.1155/2022/4258742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/12/2022] Open
Abstract
This study is aimed at investigating the role of β-galactoside-α2,3-sialyltransferase III (ST3GAL3) in fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA), as well as its potential mechanism of action. The Gene Expression Omnibus (GEO) database and gene set enrichment analysis (GSEA) were used to analyse the expression of ST3GAL3 and the enrichment signalling pathways associated with ST3GAL3 in RA. The effects of ST3GAL3 on tumour necrosis factor- (TNF-) α and interleukin- (IL-) 1β-treated MH7A cells were determined using methyl thiazolyl tetrazolium (MTT), transwell, and enzyme-linked immunosorbent assays (ELISA). The expression of proliferation-associated proteins and Toll-like receptor (TLR) pathway-enriched proteins was analysed using western blotting. As a main result, ST3GAL3 was screened as an overlapping upregulated gene from GSE101193 and GSE94519 datasets. ST3GAL3 expression in MH7A cells significantly increased with increasing treatment time with TNF-α or IL-1β. TLR9/myeloid differentiation primary response protein 88 (MyD88) is a downstream activation pathway of ST3GAL3. ST3GAL3 overexpression promoted MH7A cell proliferation and migration. Additionally, ST3GAL3 overexpression upregulated the expression of proliferation-associated proteins (cyclinD, cyclinE, and proliferating cell nuclear antigen) and TLR pathway enrichment factors (TLR9 and MyD88) and increased the production of matrix metallopeptidase (MMP) 1, MMP3, interleukin- (IL-) 6, and IL-8, whereas si-ST3GAL3 had the opposite effect. The addition of TLR9 agonists (CpG 2216 and CpG 2006) reversed the effects of si-ST3GAL3 on MH7A cell proliferation, migration, and inflammation. TLR9-specific siRNA reversed the effects of ST3GAL3 overexpression on MH7A cell proliferation, migration, and inflammation. In conclusion, ST3GAL3 is likely involved in RA pathogenesis by activating the TLR9/MyD88 pathway.
Collapse
|
26
|
Recombinant Human Proteoglycan 4 (rhPRG4) Downregulates TNFα-Stimulated NFκB Activity and FAT10 Expression in Human Corneal Epithelial Cells. Int J Mol Sci 2022; 23:ijms232112711. [DOI: 10.3390/ijms232112711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
Dry Eye Disease (DED) is a complex pathology affecting millions of people with significant impact on quality of life. Corneal inflammation, including via the nuclear factor kappa B (NFκB) pathway, plays a key etiological role in DED. Recombinant human proteoglycan 4 (rhPRG4) has been shown to be a clinically effective treatment for DED that has anti-inflammatory effects in corneal epithelial cells, but the underlying mechanism is still not understood. Our goal was to understand if rhPRG4 affects tumor necrosis factor α (TNFα)-stimulated inflammatory activity in corneal epithelial cells. We treated hTERT-immortalized corneal epithelial (hTCEpi) cells ± TNFα ± rhPRG4 and performed Western blotting on cell lysate and RNA sequencing. Bioinformatics analysis revealed that rhPRG4 had a significant effect on TNFα-mediated inflammation with potential effects on matricellular homeostasis. rhPRG4 reduced activation of key inflammatory pathways and decreased expression of transcripts for key inflammatory cytokines, interferons, interleukins, and transcription factors. TNFα treatment significantly increased phosphorylation and nuclear translocation of p65, and rhPRG4 significantly reduced both these effects. RNA sequencing identified human leukocyte antigen (HLA)-F adjacent transcript 10 (FAT10), a ubiquitin-like modifier protein which has not been studied in the context of DED, as a key pro-inflammatory transcript increased by TNFα and decreased by rhPRG4. These results were confirmed at the protein level. In summary, rhPRG4 is able to downregulate NFκB activity in hTCEpi cells, suggesting a potential biological mechanism by which it may act as a therapeutic for DED.
Collapse
|
27
|
Sin YJA, MacLeod R, Tanguay AP, Wang A, Braender-Carr O, Vitelli TM, Jay GD, Schmidt TA, Cowman MK. Noncovalent hyaluronan crosslinking by TSG-6: Modulation by heparin, heparan sulfate, and PRG4. Front Mol Biosci 2022; 9:990861. [PMID: 36275631 PMCID: PMC9579337 DOI: 10.3389/fmolb.2022.990861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
The size, conformation, and organization of the glycosaminoglycan hyaluronan (HA) affect its interactions with soluble and cell surface-bound proteins. HA that is induced to form stable networks has unique biological properties relative to unmodified soluble HA. AlphaLISA assay technology offers a facile and general experimental approach to assay protein-mediated networking of HA in solution. Connections formed between two end-biotinylated 50 kDa HA (bHA) chains can be detected by signal arising from streptavidin-coated donor and acceptor beads being brought into close proximity when the bHA chains are bridged by proteins. We observed that incubation of bHA with the protein TSG-6 (tumor necrosis factor alpha stimulated gene/protein 6, TNFAIP/TSG-6) leads to dimerization or higher order multimerization of HA chains in solution. We compared two different heparin (HP) samples and two heparan sulfate (HS) samples for the ability to disrupt HA crosslinking by TSG-6. Both HP samples had approximately three sulfates per disaccharide, and both were effective in inhibiting HA crosslinking by TSG-6. HS with a relatively high degree of sulfation (1.75 per disaccharide) also inhibited TSG-6 mediated HA networking, while HS with a lower degree of sulfation (0.75 per disaccharide) was less effective. We further identified Proteoglycan 4 (PRG4, lubricin) as a TSG-6 ligand, and found it to inhibit TSG-6-mediated HA crosslinking. The effects of HP, HS, and PRG4 on HA crosslinking by TSG-6 were shown to be due to HP/HS/PRG4 inhibition of HA binding to the Link domain of TSG-6. Using the AlphaLISA platform, we also tested other HA-binding proteins for ability to create HA networks. The G1 domain of versican (VG1) effectively networked bHA in solution but required a higher concentration than TSG-6. Cartilage link protein (HAPLN1) and the HA binding protein segment of aggrecan (HABP, G1-IGD-G2) showed only low and variable magnitude HA networking effects. This study unambiguously demonstrates HA crosslinking in solution by TSG-6 and VG1 proteins, and establishes PRG4, HP and highly sulfated HS as modulators of TSG-6 mediated HA crosslinking.
Collapse
Affiliation(s)
- Yun Jin Ashley Sin
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, United States
| | - Rebecca MacLeod
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, United States
| | - Adam P. Tanguay
- Department of Biomedical Engineering, School of Dental Medicine, UConn Health, Farmington, CT, United States
| | - Andrew Wang
- New York Medical College, Valhalla, NY, United States
| | - Olivia Braender-Carr
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, United States
| | - Teraesa M. Vitelli
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, United States
| | - Gregory D. Jay
- Department of Emergency Medicine, Warren Alpert Medical School and School of Engineering, Brown University, Providence, RI, United States
| | - Tannin A. Schmidt
- Department of Biomedical Engineering, School of Dental Medicine, UConn Health, Farmington, CT, United States
- *Correspondence: Mary K. Cowman, ; Tannin A. Schmidt,
| | - Mary K. Cowman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, United States
- Department of Orthopedic Surgery, Grossman School of Medicine, New York University, New York, NY, United States
- *Correspondence: Mary K. Cowman, ; Tannin A. Schmidt,
| |
Collapse
|
28
|
Huang S, Thomsson KA, Jin C, Ryberg H, Das N, Struglics A, Rolfson O, Björkman LI, Eisler T, Schmidt TA, Jay GD, Krawetz R, Karlsson NG. Truncated lubricin glycans in osteoarthritis stimulate the synoviocyte secretion of VEGFA, IL-8, and MIP-1 α: Interplay between O-linked glycosylation and inflammatory cytokines. Front Mol Biosci 2022; 9:942406. [PMID: 36213120 PMCID: PMC9532613 DOI: 10.3389/fmolb.2022.942406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
The primary aim of the study was to identify inflammatory markers relevant for osteoarthritis (OA)-related systemic (plasma) and local (synovial fluid, SF) inflammation. From this, we looked for inflammatory markers that coincided with the increased amount of O-linked Tn antigen (GalNAcα1-Ser/Thr) glycan on SF lubricin. Inflammatory markers in plasma and SF in OA patients and controls were measured using a 44-multiplex immunoassay. We found consistently 29 markers detected in both plasma and SF. The difference in their concentration and the low correlation when comparing SF and plasma suggests an independent inflammatory environment in the two biofluids. Only plasma MCP-4 and TARC increased in our patient cohort compared to control plasma. To address the second task, we concluded that plasma markers were irrelevant for a direct connection with SF glycosylation. Hence, we correlated the SF-inflammatory marker concentrations with the level of altered glycosylation of SF-lubricin. We found that the level of SF-IL-8 and SF-MIP-1α and SF-VEGFA in OA patients displayed a positive correlation with the altered lubricin glycosylation. Furthermore, when exposing fibroblast-like synoviocytes from both controls and OA patients to glycovariants of recombinant lubricin, the secretion of IL-8 and MIP-1α and VEGFA were elevated using lubricin with Tn antigens, while lubricin with sialylated and nonsialylated T antigens had less or no measurable effect. These data suggest that truncated glycans of lubricin, as found in OA, promote synovial proinflammatory cytokine production and exacerbate local synovial inflammation.
Collapse
Affiliation(s)
- Shan Huang
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina A. Thomsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Chunsheng Jin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Ryberg
- Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Nabangshu Das
- Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - André Struglics
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Ola Rolfson
- Department of Orthopaedics, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lena I. Björkman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Eisler
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Tannin A. Schmidt
- Biomedical Engineering Department, University of Connecticut Health Centre, Farmington, CT, United States
| | - Gregory D. Jay
- Department of Emergency Medicine, Warren Alpert Medical School and Division of Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Roman Krawetz
- Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Niclas G. Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Pharmacy, Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
29
|
Liao C, Wang Q, An J, Chen J, Li X, Long Q, Xiao L, Guan X, Liu J. CD44 Glycosylation as a Therapeutic Target in Oncology. Front Oncol 2022; 12:883831. [PMID: 35936713 PMCID: PMC9351704 DOI: 10.3389/fonc.2022.883831] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/10/2022] [Indexed: 11/27/2022] Open
Abstract
The interaction of non-kinase transmembrane glycoprotein CD44 with ligands including hyaluronic acid (HA) is closely related to the occurrence and development of tumors. Changes in CD44 glycosylation can regulate its binding to HA, Siglec-15, fibronectin, TM4SF5, PRG4, FGF2, collagen and podoplanin and activate or inhibit c-Src/STAT3/Twist1/Bmi1, PI3K/AKT/mTOR, ERK/NF-κB/NANOG and other signaling pathways, thereby having a profound impact on the tumor microenvironment and tumor cell fate. However, the glycosylation of CD44 is complex and largely unknown, and the current understanding of how CD44 glycosylation affects tumors is limited. These issues must be addressed before targeted CD44 glycosylation can be applied to treat human cancers.
Collapse
Affiliation(s)
- Chengcheng Liao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Qian Wang
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Jiaxing An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jie Chen
- Department of Urology, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaolan Li
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Qian Long
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Linlin Xiao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Linlin Xiao, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Xiaoyan Guan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Linlin Xiao, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Jianguo Liu
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Linlin Xiao, ; Xiaoyan Guan, ; Jianguo Liu,
| |
Collapse
|
30
|
Proteoglycan 4 (PRG4) treatment enhances wound closure and tissue regeneration. NPJ Regen Med 2022; 7:32. [PMID: 35750773 PMCID: PMC9232611 DOI: 10.1038/s41536-022-00228-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 05/20/2022] [Indexed: 01/13/2023] Open
Abstract
The wound healing response is one of most primitive and conserved physiological responses in the animal kingdom, as restoring tissue integrity/homeostasis can be the difference between life and death. Wound healing in mammals is mediated by immune cells and inflammatory signaling molecules that regulate tissue resident cells, including local progenitor cells, to mediate closure of the wound through formation of a scar. Proteoglycan 4 (PRG4), a protein found throughout the animal kingdom from fish to elephants, is best known as a glycoprotein that reduces friction between articulating surfaces (e.g. cartilage). Previously, PRG4 was also shown to regulate the inflammatory and fibrotic response. Based on this, we asked whether PRG4 plays a role in the wound healing response. Using an ear wound model, topical application of exogenous recombinant human (rh)PRG4 hastened wound closure and enhanced tissue regeneration. Our results also suggest that rhPRG4 may impact the fibrotic response, angiogenesis/blood flow to the injury site, macrophage inflammatory dynamics, recruitment of immune and increased proliferation of adult mesenchymal progenitor cells (MPCs) and promoting chondrogenic differentiation of MPCs to form the auricular cartilage scaffold of the injured ear. These results suggest that PRG4 has the potential to suppress scar formation while enhancing connective tissue regeneration post-injury by modulating aspects of each wound healing stage (blood clotting, inflammation, tissue generation and tissue remodeling). Therefore, we propose that rhPRG4 may represent a potential therapy to mitigate scar and improve wound healing.
Collapse
|
31
|
Abstract
The superficial zone of articular cartilage contributes to smooth joint motion through the production of proteoglycan 4 (PRG4), also known as lubricin. Recent studies indicate novel effects of PRG4 as a signaling molecule, other than a simple extracellular matrix protein. Additionally, the accumulating evidence displays that various molecules and signaling pathways are involved in regulating the superficial zone and PRG4 expression. In addition, Prg4-expressing cells include a progenitor population of articular chondrocytes. Several non-clinical and clinical studies have shown that PRG4 and related molecules are promising candidates for disease-modifying drugs for treating osteoarthritis. Since PRG4 is also expressed in the synovium, tendons, and ligaments, further studies of PRG4-related pathways and PRG4-positive cells may elucidate the mechanisms underlying joint homeostasis.
Collapse
|
32
|
Yang DS, Dickerson EE, Zhang LX, Richendrfer H, Karamchedu PN, Badger GJ, Schmidt TA, Fredericks AM, Elsaid KA, Jay GD. Quadruped Gait and Regulation of Apoptotic Factors in Tibiofemoral Joints following Intra-Articular rhPRG4 Injection in Prg4 Null Mice. Int J Mol Sci 2022; 23:ijms23084245. [PMID: 35457064 PMCID: PMC9025840 DOI: 10.3390/ijms23084245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 12/03/2022] Open
Abstract
Camptodactyly-arthropathy-coxa vara-pericarditis (CACP) syndrome leads to diarthrodial joint arthropathy and is caused by the absence of lubricin (proteoglycan 4—PRG4), a surface-active mucinous glycoprotein responsible for lubricating articular cartilage. In this study, mice lacking the orthologous gene Prg4 served as a model that recapitulates the destructive arthrosis that involves biofouling of cartilage by serum proteins in lieu of Prg4. This study hypothesized that Prg4-deficient mice would demonstrate a quadruped gait change and decreased markers of mitochondrial dyscrasia, following intra-articular injection of both hindlimbs with recombinant human PRG4 (rhPRG4). Prg4−/− (N = 44) mice of both sexes were injected with rhPRG4 and gait alterations were studied at post-injection day 3 and 6, before joints were harvested for immunohistochemistry for caspase-3 activation. Increased stance and propulsion was shown at 3 days post-injection in male mice. There were significantly fewer caspase-3-positive chondrocytes in tibiofemoral cartilage from rhPRG4-injected mice. The mitochondrial gene Mt-tn, and myosin heavy (Myh7) and light chains (Myl2 and Myl3), known to play a cytoskeletal stabilizing role, were significantly upregulated in both sexes (RNA-Seq) following IA rhPRG4. Chondrocyte mitochondrial dyscrasias attributable to the arthrosis in CACP may be mitigated by IA rhPRG4. In a supporting in vitro crystal microbalance experiment, molecular fouling by albumin did not block the surface activity of rhPRG4.
Collapse
Affiliation(s)
- Daniel S. Yang
- School of Engineering, Brown University, Providence, RI 02912, USA; (D.S.Y.); (G.D.J.)
- Department of Emergency Medicine, Alpert School of Medicine, Brown University, Providence, RI 02903, USA; (L.X.Z.); (H.R.)
| | - Edward E. Dickerson
- North Carolina Agricultural Technical State University, Greensboro, NC 27411, USA;
| | - Ling X. Zhang
- Department of Emergency Medicine, Alpert School of Medicine, Brown University, Providence, RI 02903, USA; (L.X.Z.); (H.R.)
| | - Holly Richendrfer
- Department of Emergency Medicine, Alpert School of Medicine, Brown University, Providence, RI 02903, USA; (L.X.Z.); (H.R.)
| | - Padmini N. Karamchedu
- Department of Orthopedics, Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| | - Gary J. Badger
- Department of Medical Biostatistics, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Tannin A. Schmidt
- Department of Biomedical Engineering, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, USA;
| | - Alger M. Fredericks
- Department of Surgery, Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| | - Khaled A. Elsaid
- School of Pharmacy, Chapman University, Irvine, CA 92618, USA
- Correspondence:
| | - Gregory D. Jay
- School of Engineering, Brown University, Providence, RI 02912, USA; (D.S.Y.); (G.D.J.)
- Department of Emergency Medicine, Alpert School of Medicine, Brown University, Providence, RI 02903, USA; (L.X.Z.); (H.R.)
- Department of Orthopedics, Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| |
Collapse
|
33
|
Dituri F, Gigante G, Scialpi R, Mancarella S, Fabregat I, Giannelli G. Proteoglycans in Cancer: Friends or Enemies? A Special Focus on Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14081902. [PMID: 35454809 PMCID: PMC9024587 DOI: 10.3390/cancers14081902] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Proteoglycans affect multiple molecular and cellular processes during the progression of solid tumors with a highly desmoplastic stroma, such as HCC. Due to their role in enhancing or limiting the traits of cancer cells underlying their aggressiveness, such as proliferation, angiogenesis, epithelial to mesenchymal transition (EMT), and stemness, these macromolecules could be exploited as molecular targets or therapeutic agents. Proteoglycans, such as biglycan, versican, syndecan-1, glypican-3, and agrin, promote HCC cell proliferation, EMT, and angiogenesis, while endostatin and proteoglycan 4 were shown to impair cancer neovascularization or to enhance the sensitivity of HCC cells to drugs, such as sorafenib and regorafenib. Based on this evidence, interventional strategies involving the use of humanized monoclonal antibodies, T cells engineered with chimeric antigen receptors, or recombinant proteins mimicking potentially curative proteoglycans, are being employed or may be adopted in the near future for the treatment of HCC. Abstract Proteoglycans are a class of highly glycosylated proteins expressed in virtually all tissues, which are localized within membranes, but more often in the pericellular space and extracellular matrix (ECM), and are involved in tissue homeostasis and remodeling of the stromal microenvironment during physiological and pathological processes, such as tissue regeneration, angiogenesis, and cancer. In general, proteoglycans can perform signaling activities and influence a range of physical, chemical, and biological tissue properties, including the diffusivity of small electrolytes and nutrients and the bioavailability of growth factors. While the dysregulated expression of some proteoglycans is observed in many cancers, whether they act as supporters or limiters of neoplastic progression is still a matter of controversy, as the tumor promoting or suppressive function of some proteoglycans is context dependent. The participation of multiple proteoglycans in organ regeneration (as demonstrated for the liver in hepatectomy mouse models) and in cancer suggests that these molecules actively influence cell growth and motility, thus contributing to key events that characterize neoplastic progression. In this review, we outline the main roles of proteoglycans in the physiology and pathology of cancers, with a special mention to hepatocellular carcinoma (HCC), highlighting the translational potential of proteoglycans as targets or therapeutic agents for the treatment of this disease.
Collapse
Affiliation(s)
- Francesco Dituri
- National Institute of Gastroenterology Saverio de Bellis, IRCCS Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.G.); (R.S.); (S.M.); (G.G.)
- Correspondence:
| | - Gianluigi Gigante
- National Institute of Gastroenterology Saverio de Bellis, IRCCS Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.G.); (R.S.); (S.M.); (G.G.)
| | - Rosanna Scialpi
- National Institute of Gastroenterology Saverio de Bellis, IRCCS Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.G.); (R.S.); (S.M.); (G.G.)
| | - Serena Mancarella
- National Institute of Gastroenterology Saverio de Bellis, IRCCS Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.G.); (R.S.); (S.M.); (G.G.)
| | - Isabel Fabregat
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), CIBEREHD and University of Barcelona, L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
| | - Gianluigi Giannelli
- National Institute of Gastroenterology Saverio de Bellis, IRCCS Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.G.); (R.S.); (S.M.); (G.G.)
| |
Collapse
|
34
|
ElSayed S, Jay GD, Cabezas R, Qadri M, Schmidt TA, Elsaid KA. Recombinant Human Proteoglycan 4 Regulates Phagocytic Activation of Monocytes and Reduces IL-1β Secretion by Urate Crystal Stimulated Gout PBMCs. Front Immunol 2022; 12:771677. [PMID: 34992596 PMCID: PMC8725049 DOI: 10.3389/fimmu.2021.771677] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Objectives To compare phagocytic activities of monocytes in peripheral blood mononuclear cells (PBMCs) from acute gout patients and normal subjects, examine monosodium urate monohydrate (MSU) crystal-induced IL-1β secretion ± recombinant human proteoglycan 4 (rhPRG4) or interleukin-1 receptor antagonist (IL-1RA), and study the anti-inflammatory mechanism of rhPRG4 in MSU stimulated monocytes. Methods Acute gout PBMCs were collected from patients in the Emergency Department and normal PBMCs were obtained from a commercial source. Monocytes in PBMCs were identified by flow cytometry. PBMCs were primed with Pam3CSK4 (1μg/mL) for 24h and phagocytic activation of monocytes was determined using fluorescently labeled latex beads. MSU (200μg/mL) stimulated IL-1β secretion was determined by ELISA. Reactive oxygen species (ROS) generation in monocytes was determined fluorometrically. PBMCs were incubated with IL-1RA (250ng/mL) or rhPRG4 (200μg/mL) and bead phagocytosis by monocytes was determined. THP-1 monocytes were treated with MSU crystals ± rhPRG4 and cellular levels of NLRP3 protein, pro-IL-1β, secreted IL-1β, and activities of caspase-1 and protein phosphatase-2A (PP2A) were quantified. The peritoneal influx of inflammatory and anti-inflammatory monocytes and neutrophils in Prg4 deficient mice was studied and the impact of rhPRG4 on immune cell trafficking was assessed. Results Enhanced phagocytic activation of gout monocytes under basal conditions (p<0.001) was associated with ROS generation and MSU stimulated IL-1β secretion (p<0.05). rhPRG4 reduced bead phagocytosis by normal and gout monocytes compared to IL-1RA and both treatments were efficacious in reducing IL-1β secretion (p<0.05). rhPRG4 reduced pro-IL-1β content, caspase-1 activity, conversion of pro-IL-1β to mature IL-1β and restored PP2A activity in monocytes (p<0.05). PP2A inhibition reversed rhPRG4’s effects on pro-IL-1β and mature IL-1β in MSU stimulated monocytes. Neutrophils accumulated in peritoneal cavities of Prg4 deficient mice (p<0.01) and rhPRG4 treatment reduced neutrophil accumulation and enhanced anti-inflammatory monocyte influx (p<0.05). Conclusions MSU phagocytosis was higher in gout monocytes resulting in higher ROS and IL-1β secretion. rhPRG4 reduced monocyte phagocytic activation to a greater extent than IL-1RA and reduced IL-1β secretion. The anti-inflammatory activity of rhPRG4 in monocytes is partially mediated by PP2A, and in vivo, PRG4 plays a role in regulating the trafficking of immune cells into the site of a gout flare.
Collapse
Affiliation(s)
- Sandy ElSayed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, United States
| | - Gregory D Jay
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, United States
| | - Ralph Cabezas
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, United States
| | - Marwa Qadri
- Department of Pharmacology, School of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, CT, United States
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, United States
| |
Collapse
|
35
|
Seol D, Choe HH, Zheng H, Brouillette MJ, Fredericks DC, Petersen EB, Song I, Jaidev LR, Salem A, Martin JA. Intra-Articular Adeno-Associated Virus-Mediated Proteoglycan 4 Gene Therapy for Preventing Post-Traumatic Osteoarthritis. Hum Gene Ther 2021; 33:529-540. [PMID: 34610749 PMCID: PMC9142765 DOI: 10.1089/hum.2021.177] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Lubricin, a glycoprotein encoded by the proteoglycan 4 (PRG4) gene, is an essential boundary lubricant that reduces friction between articular cartilage surfaces. The loss of lubricin subsequent to joint injury plays a role in the pathogenesis of post-traumatic osteoarthritis (PTOA). Here we describe the development and evaluation of an adeno-associated virus (AAV)-based PRG4 gene therapy intended to restore lubricin in injured joints. The green fluorescent protein (GFP) gene was inserted the PRG4 gene to facilitate tracing the distribution of the transgene product (AAV-PRG4-GFP) in vivo. METHODS Transduction efficiency of AAV-PRG4-GFP was evaluated in joint cells, and the conditioned medium containing secreted PRG4-GFP was used for shear loading/friction and viability tests. In vivo transduction of joint tissues following intra-articular injection of AAV-PRG4-GFP was confirmed in the mouse stifle joint in a surgical model of destabilization of the medial meniscus (DMM), and chondroprotective activity was tested in a rabbit anterior cruciate ligament transection (ACLT) model. RESULTS In vitro studies showed that PRG4-GFP has lubricin-like cartilage binding and anti-friction properties. Significant cytoprotective effects were seen when cartilage was soaked in PRG4-GFP prior to cyclic shear loading (n = 3). Polymerase chain reaction and confocal microscopy confirmed the presence of PRG4-GFP DNA and protein, respectively, in a mouse DMM (n = 3 per group). In the rabbit ACLT model, AAV-PRG4-GFP gene therapy enhanced lubricin expression (p = 0.001 versus AAV-GFP: n = 7-14) and protected the cartilage from degeneration (p = 0.014 versus AAV-GFP: n = 9-10) when treatments were administered immediately post-operation, but efficacy was lost when treatment was delayed for 2 weeks. CONCLUSION AAV-PRG4-GFP gene therapy protected cartilage from degeneration in a rabbit ACLT model; however, data from the ACLT model suggest that early intervention is essential for efficacy.
Collapse
Affiliation(s)
- Dongrim Seol
- The University of Iowa, 4083, Orthopedics and Rehabilitation, Iowa City, Iowa, United States.,The University of Iowa, 4083, Orthodontics, Iowa City, Iowa, United States;
| | - Hyeong Hun Choe
- The University of Iowa, 4083, Orthopedics and Rehabilitation, Iowa City, Iowa, United States;
| | - Hongjun Zheng
- Washington University School of Medicine in Saint Louis, 12275, Orthopaedic Surgery, St Louis, Missouri, United States;
| | - Marc J Brouillette
- The University of Iowa, 4083, Orthopedics and Rehabilitation, Iowa City, Iowa, United States;
| | - Douglas C Fredericks
- The University of Iowa, 4083, Orthopedics and Rehabilitation, Iowa City, Iowa, United States;
| | - Emily B Petersen
- The University of Iowa, 4083, Orthopedics and Rehabilitation, Iowa City, Iowa, United States;
| | - Ino Song
- The University of Iowa, 4083, Orthopedics and Rehabilitation, Iowa City, Iowa, United States.,The University of Iowa, 4083, Roy J. Carver Biomedical Engineering, Iowa City, Iowa, United States;
| | - L R Jaidev
- The University of Iowa, 4083, Pharmaceutical Sciences and Experimental Therapeutics, Iowa City, Iowa, United States;
| | - Aliasger Salem
- The University of Iowa, 4083, Pharmaceutical Sciences and Experimental Therapeutics, Iowa City, Iowa, United States;
| | - James A Martin
- The University of Iowa, 4083, Orthopedics and Rehabilitation, Iowa City, Iowa, United States.,The University of Iowa, 4083, Roy J. Carver Biomedical Engineering, Iowa City, Iowa, United States.,The University of Iowa, 4083, Pharmaceutical Sciences and Experimental Therapeutics, Iowa City, Iowa, United States;
| |
Collapse
|
36
|
Qadri M, Jay GD, Zhang LX, Schmidt TA, Totonchy J, Elsaid KA. Proteoglycan-4 is an essential regulator of synovial macrophage polarization and inflammatory macrophage joint infiltration. Arthritis Res Ther 2021; 23:241. [PMID: 34521469 PMCID: PMC8439011 DOI: 10.1186/s13075-021-02621-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/05/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Synovial macrophages perform a multitude of functions that include clearance of cell debris and foreign bodies, tissue immune surveillance, and resolution of inflammation. The functional diversity of macrophages is enabled by distinct subpopulations that express unique surface markers. Proteoglycan-4 (PRG4) is an important regulator of synovial hyperplasia and fibrotic remodeling, and the involvement of macrophages in PRG4's synovial role is yet to be defined. Our objectives were to study the PRG4's importance to macrophage homeostatic regulation in the synovium and infiltration of pro-inflammatory macrophages in acute synovitis and investigate whether macrophages mediated synovial fibrosis in Prg4 gene-trap (Prg4GT/GT) murine knee joints. METHODS Macrophage phenotyping in Prg4GT/GT and Prg4+/+ joints was performed by flow cytometry using pan-macrophage markers, e.g., CD11b, F4/80, and surface markers of M1 macrophages (CD86) and M2 macrophages (CD206). Characterizations of the various macrophage subpopulations were performed in 2- and 6-month-old animals. The expression of inflammatory markers, IL-6, and iNOS in macrophages that are CD86+ and/or CD206+ was studied. The impact of Prg4 recombination on synovial macrophage populations of 2- and 6-month-old animals and infiltration of pro-inflammatory macrophages in response to a TLR2 agonist challenge was determined. Macrophages were depleted using liposomal clodronate and synovial membrane thickness, and the expression of fibrotic markers α-SMA, PLOD2, and collagen type I (COL-I) was assessed using immunohistochemistry. RESULTS Total macrophages in Prg4GT/GT joints were higher than Prg4+/+ joints (p<0.0001) at 2 and 6 months, and the percentages of CD86+/CD206- and CD86+/CD206+ macrophages increased in Prg4GT/GT joints at 6 months (p<0.0001), whereas the percentage of CD86-/CD206+ macrophages decreased (p<0.001). CD86+/CD206- and CD86+/CD206+ macrophages expressed iNOS and IL-6 compared to CD86-/CD206+ macrophages (p<0.0001). Prg4 re-expression limited the accumulation of CD86+ macrophages (p<0.05) and increased CD86-/CD206+ macrophages (p<0.001) at 6 months. Prg4 recombination attenuated synovial recruitment of pro-inflammatory macrophages in 2-month-old animals (p<0.001). Clodronate-mediated macrophage depletion reduced synovial hyperplasia, α-SMA, PLOD2, and COL-I expressions in the synovium (p<0.0001). CONCLUSIONS PRG4 regulates the accumulation and homeostatic balance of macrophages in the synovium. In its absence, the synovium becomes populated with M1 macrophages. Furthermore, macrophages exert an effector role in synovial fibrosis in Prg4GT/GT animals.
Collapse
Affiliation(s)
- Marwa Qadri
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, 82826 Kingdom of Saudi Arabia
| | - Gregory D. Jay
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI USA
| | - Ling X. Zhang
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI USA
| | - Tannin A. Schmidt
- Biomedical Engineering Department, School of Dental Medicine, University of Connecticut, Farmington, CT USA
| | - Jennifer Totonchy
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, 9401 Jeronimo Road, Irvine, CA 92618 USA
| | - Khaled A. Elsaid
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, 9401 Jeronimo Road, Irvine, CA 92618 USA
| |
Collapse
|
37
|
Solakyildirim K, Li Y, Bayer AS, Sullam PM, Xiong YQ, Lebrilla CB, Bensing BA. Proteoglycan 4 (lubricin) is a highly sialylated glycoprotein associated with cardiac valve damage in animal models of infective endocarditis. Glycobiology 2021; 31:1582-1595. [PMID: 34459483 DOI: 10.1093/glycob/cwab095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/30/2021] [Accepted: 08/16/2021] [Indexed: 11/12/2022] Open
Abstract
S. gordonii and S. sanguinis are primary colonizers of tooth surfaces, and are generally associated with oral health, but can also cause infective endocarditis (IE). These species express "Siglec-like" adhesins that bind sialylated glycans on host glycoproteins, which can aid the formation of infected platelet-fibrin thrombi (vegetations) on cardiac valve surfaces. We previously determined that the ability of S. gordonii to bind sialyl T-antigen (sTa) increased pathogenicity, relative to recognition of sialylated core 2 O-glycan structures, in an animal model of IE. However, it is unclear when and where the sTa structure is displayed, and which sTa-modified host factors promote valve colonization. In this study, we identified sialylated glycoproteins in the aortic valve vegetations and plasma of rat and rabbit models of this disease. Glycoproteins that display sTa versus core 2 O-glycan structures were identified by using recombinant forms of the streptococcal Siglec-like adhesins for lectin blotting and affinity capture, and the O-linked glycans were profiled by mass spectrometry. Proteoglycan 4 (PRG4), also known as lubricin, was a major carrier of sTa in the infected vegetations. Moreover, plasma PRG4 levels were significantly higher in animals with damaged or infected valves, as compared with healthy animals. The combined results demonstrate that, in addition to platelet GPIbα, PRG4 is a highly sialylated mucin-like glycoprotein found in aortic valve vegetations and may contribute to the persistence of oral streptococci in this protected endovascular niche. Moreover, plasma PRG4 could serve as a biomarker for endocardial injury and infection.
Collapse
Affiliation(s)
- Kemal Solakyildirim
- Department of Chemistry, University of California, Davis, California, United States of America.,Department of Chemistry, Erzincan Binali Yildirim University, Erzincan, 24100, Turkey
| | - Yi Li
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Arnold S Bayer
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America.,David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Paul M Sullam
- Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, California, United States of America
| | - Yan Q Xiong
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America.,David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, California, United States of America
| | - Barbara A Bensing
- Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, California, United States of America
| |
Collapse
|
38
|
Nakamoto H, Katanosaka Y, Chijimatsu R, Mori D, Xuan F, Yano F, Omata Y, Maenohara Y, Murahashi Y, Kawaguchi K, Yamagami R, Inui H, Taketomi S, Taniguchi Y, Kanagawa M, Naruse K, Tanaka S, Saito T. Involvement of Transient Receptor Potential Vanilloid Channel 2 in the Induction of Lubricin and Suppression of Ectopic Endochondral Ossification in Mouse Articular Cartilage. Arthritis Rheumatol 2021; 73:1441-1450. [PMID: 33586252 DOI: 10.1002/art.41684] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 02/04/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Transient receptor potential vanilloid channel 2 (TRPV2) is a Ca2+ -permeable channel and plays a role in mediating intracellular Ca2+ current via mechanical stimuli. This study was undertaken to examine the expression and role of TRPV2 in adult articular cartilage and the development of osteoarthritis (OA). METHODS We examined TRPV2 expression in mouse and human articular cartilage. We analyzed the development of OA in Col2a1-CreERt2 ;Trpv2fl/fl mice and Trpv2fl/fl littermates in the resection of the medial meniscus and medial collateral ligament model (n = 5 each), the destabilization of the medial meniscus model (n = 5 each), and the aging mouse model (n = 8-9 each). We examined marker protein expression in these joints, Ca2+ influx by mechanical stimuli, and downstream pathways in vitro. RESULTS TRPV2 was expressed in mouse and human articular cartilage and ectopic ossification lesions. In all mouse models of OA examined, Col2a1-CreERt2 ;Trpv2fl/fl mice were observed to have enhanced degradation of articular cartilage accompanied by decreased expression of lubricin/Prg4, and marked formation of periarticular ectopic ossification. Mechanical stress-induced Ca2+ influx was decreased by Trpv2 knockout (KO). Prg4 induction by fluid-flow shear stress was diminished in Trpv2-KO mouse chondrocytes, and this was mediated by the Ca2+ /calmodulin-dependent protein kinase kinase-cyclic AMP response element binding protein axis. Hypertrophic differentiation was enhanced in Trpv2-KO mouse chondrocytes. Increased activity of calcineurin and nuclear translocation of nuclear factor in activated T cells 1 induced by fluid-flow shear stress or TRP agonist treatment was reversed by Trpv2 knockout. CONCLUSION Our findings demonstrate regulation of articular cartilage by TRPV2 through Prg4 induction and suppression of ectopic ossification.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Motoi Kanagawa
- Kobe University Graduate School of Medicine, Kobe, Japan, and Ehime University School of Medicine, Toon, Japan
| | | | | | | |
Collapse
|
39
|
Menon NG, Goyal R, Lema C, Woods PS, Tanguay AP, Morin AA, Das N, Jay GD, Krawetz RJ, Dufour A, Shapiro LH, Redfern RL, Ghosh M, Schmidt TA. Proteoglycan 4 (PRG4) expression and function in dry eye associated inflammation. Exp Eye Res 2021; 208:108628. [PMID: 34048779 DOI: 10.1016/j.exer.2021.108628] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/30/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022]
Abstract
Dry eye disease (DED) affects hundreds of millions of people worldwide. It is characterized by the production of inflammatory cytokines and chemokines as well as damaging matrix metalloproteinases (MMPs) at the ocular surface. While proteoglycan 4 (PRG4), a mucin-like glycoprotein present at the ocular surface, is most well known as a boundary lubricant that contributes to ocular surface integrity, it has been shown to blunt inflammation in various cell types, suggesting a dual mechanism of action. Recently, full-length recombinant human PRG4 (rhPRG4) has been shown to improve signs and symptoms of DED in humans. However, there remains a significant need for basic science research on rhPRG4's biological properties and its potential therapeutic mechanisms of action in treating DED. Therefore, the objectives of this study were to characterize endogenous PRG4 expression by telomerase-immortalized human corneal epithelial (hTCEpi) cells, examine whether exogenous rhPRG4 modulates cytokine and chemokine secretion in response to dry eye associated inflammation (TNFα and IL-1β), explore interactions between rhPRG4 and MMP-9, and understand how experimental dry eye (EDE) in mice affects PRG4 expression. PRG4 secretion from hTCEpi cells was quantified by Western blot and expression visualized by immunocytochemistry. Cytokine/chemokine production was measured by ELISA and Luminex, while rhPRG4's effect on MMP-9 activity, binding, and expression was quantified using an MMP-9 inhibitor kit, surface plasmon resonance, and reverse transcription polymerase chain reaction (RT-PCR), respectively. Finally, EDE was induced in mice, and PRG4 was visualized by immunohistochemistry in the cornea and by Western blot in lacrimal gland lysate. In vitro results demonstrate that hTCEpi cells synthesize and secrete PRG4, and PRG4 secretion is inhibited by TNFα and IL-1β. In response to these pro-inflammatory stresses, exogenous rhPRG4 significantly reduced the stimulated production of IP-10, RANTES, ENA-78, GROα, MIP-3α, and MIG, and trended towards a reduction of MIP-1α and MIP-1β. The hTCEpi cells were also able to internalize fluorescently-labelled rhPRG4, consistent with a mechanism of action that includes downstream biological signaling pathways. rhPRG4 was not digested by MMP-9, and it did not modulate MMP-9 gene expression in hTCEpi cells, but it was able to bind to MMP-9 and inhibited in vitro activity of exogenous MMP-9 in the presence of human tears. Finally, in vivo results demonstrate that EDE significantly decreased immunolocalization of PRG4 on the corneal epithelium and trended towards a reduction of PRG4 in lacrimal gland lysate. Collectively these results demonstrate rhPRG4 has anti-inflammatory properties on corneal epithelial cells, particularly as it relates to mitigating chemokine production, and is an inhibitor of MMP-9 activity, as well as that in vivo expression of PRG4 can be altered in preclinical models of DED. In conclusion, these findings contribute to our understanding of PRG4's immunomodulatory properties in the context of DED inflammation and provide the foundation and motivation for further mechanistic research of PRG4's properties on the ocular surface as well as expanding clinical evaluation of its ability as a multifunctional therapeutic agent to effectively provide relief to those who suffer from DED.
Collapse
Affiliation(s)
- Nikhil G Menon
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Ruchi Goyal
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Carolina Lema
- The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX, USA
| | - Paige S Woods
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Adam P Tanguay
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Alyssa A Morin
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Nabangshu Das
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Gregory D Jay
- Department of Emergency Medicine, Warren Alpert Medical School & School of Engineering, Brown University, Providence, RI, USA
| | - Roman J Krawetz
- Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada; Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Linda H Shapiro
- Department of Cell Biology, School of Medicine, UConn Health, Farmington, CT, USA
| | - Rachel L Redfern
- The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX, USA
| | - Mallika Ghosh
- Department of Cell Biology, School of Medicine, UConn Health, Farmington, CT, USA
| | - Tannin A Schmidt
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, CT, USA.
| |
Collapse
|
40
|
Seime T, Akbulut AC, Liljeqvist ML, Siika A, Jin H, Winski G, van Gorp RH, Karlöf E, Lengquist M, Buckler AJ, Kronqvist M, Waring OJ, Lindeman JHN, Biessen EAL, Maegdefessel L, Razuvaev A, Schurgers LJ, Hedin U, Matic L. Proteoglycan 4 Modulates Osteogenic Smooth Muscle Cell Differentiation during Vascular Remodeling and Intimal Calcification. Cells 2021; 10:1276. [PMID: 34063989 PMCID: PMC8224064 DOI: 10.3390/cells10061276] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 01/02/2023] Open
Abstract
Calcification is a prominent feature of late-stage atherosclerosis, but the mechanisms driving this process are unclear. Using a biobank of carotid endarterectomies, we recently showed that Proteoglycan 4 (PRG4) is a key molecular signature of calcified plaques, expressed in smooth muscle cell (SMC) rich regions. Here, we aimed to unravel the PRG4 role in vascular remodeling and intimal calcification. PRG4 expression in human carotid endarterectomies correlated with calcification assessed by preoperative computed tomographies. PRG4 localized to SMCs in early intimal thickening, while in advanced lesions it was found in the extracellular matrix, surrounding macro-calcifications. In experimental models, Prg4 was upregulated in SMCs from partially ligated ApoE-/- mice and rat carotid intimal hyperplasia, correlating with osteogenic markers and TGFb1. Furthermore, PRG4 was enriched in cells positive for chondrogenic marker SOX9 and around plaque calcifications in ApoE-/- mice on warfarin. In vitro, PRG4 was induced in SMCs by IFNg, TGFb1 and calcifying medium, while SMC markers were repressed under calcifying conditions. Silencing experiments showed that PRG4 expression was driven by transcription factors SMAD3 and SOX9. Functionally, the addition of recombinant human PRG4 increased ectopic SMC calcification, while arresting cell migration and proliferation. Mechanistically, it suppressed endogenous PRG4, SMAD3 and SOX9, and restored SMC markers' expression. PRG4 modulates SMC function and osteogenic phenotype during intimal remodeling and macro-calcification in response to TGFb1 signaling, SMAD3 and SOX9 activation. The effects of PRG4 on SMC phenotype and calcification suggest its role in atherosclerotic plaque stability, warranting further investigations.
Collapse
Affiliation(s)
- Till Seime
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Asim Cengiz Akbulut
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands; (A.C.A.); (R.H.v.G.); (L.J.S.)
| | - Moritz Lindquist Liljeqvist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Antti Siika
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Hong Jin
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
- Department of Medicine, Karolinska Institutet, 17164 Stockholm, Sweden; (G.W.); (L.M.)
| | - Greg Winski
- Department of Medicine, Karolinska Institutet, 17164 Stockholm, Sweden; (G.W.); (L.M.)
| | - Rick H. van Gorp
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands; (A.C.A.); (R.H.v.G.); (L.J.S.)
| | - Eva Karlöf
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Mariette Lengquist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Andrew J. Buckler
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Malin Kronqvist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Olivia J. Waring
- Department of Pathology, CARIM, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands; (O.J.W.); (E.A.L.B.)
| | - Jan H. N. Lindeman
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Erik A. L. Biessen
- Department of Pathology, CARIM, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands; (O.J.W.); (E.A.L.B.)
| | - Lars Maegdefessel
- Department of Medicine, Karolinska Institutet, 17164 Stockholm, Sweden; (G.W.); (L.M.)
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technische Universität München, 81679 Munich, Germany
| | - Anton Razuvaev
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Leon J. Schurgers
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands; (A.C.A.); (R.H.v.G.); (L.J.S.)
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, 52062 Aachen, Germany
| | - Ulf Hedin
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Ljubica Matic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| |
Collapse
|
41
|
Maenohara Y, Chijimatsu R, Tachibana N, Uehara K, Xuan F, Mori D, Murahashi Y, Nakamoto H, Oichi T, Chang SH, Matsumoto T, Omata Y, Yano F, Tanaka S, Saito T. Lubricin Contributes to Homeostasis of Articular Cartilage by Modulating Differentiation of Superficial Zone Cells. J Bone Miner Res 2021; 36:792-802. [PMID: 33617044 DOI: 10.1002/jbmr.4226] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/19/2020] [Accepted: 11/27/2020] [Indexed: 11/10/2022]
Abstract
Lubricin encoded by the proteoglycan 4 (Prg4) gene is produced from superficial zone (SFZ) cells of articular cartilage and synoviocytes, which is indispensable for lubrication of joint surfaces. Loss-of-function of human and mouse Prg4 results in early-onset arthropathy accompanied by lost SFZ cells and hyperplastic synovium. Here, we focused on increases in the thickness of articular cartilage in Prg4-knockout joints and analyzed the underlying mechanisms. In the late stage of articular cartilage development, the articular cartilage was thickened at 2 to 4 weeks and the SFZ disappeared at 8 weeks in Prg4-knockout mice. Similar changes were observed in cultured Prg4-knockout femoral heads. Cell tracking showed that Prg4-knockout SFZ cells at 1 week of age expanded to deep layers after 1 week. In in vitro experiments, overexpression of Prg4 lacking a mucin-like domain suppressed differentiation of ATDC5 cells markedly, whereas pellets of Prg4-knockout SFZ cells showed enhanced differentiation. RNA sequencing identified matrix metalloproteinase 9 (Mmp9) as the top upregulated gene by Prg4 knockout. Mmp9 expressed in the SFZ was further induced in Prg4-knockout mice. The increased expression of Mmp9 by Prg4 knockout was canceled by IκB kinase (IKK) inhibitor treatment. Phosphorylation of Smad2 was also enhanced in Prg4-knockout cell pellets, which was canceled by the IKK inhibitor. Expression of Mmp9 and phosphorylated Smad2 during articular cartilage development was enhanced in Prg4-knockout joints. Lubricin contributes to homeostasis of articular cartilage by suppressing differentiation of SFZ cells, and the nuclear factor-kappa B-Mmp9-TGF-β pathway is probably responsible for the downstream action of lubricin. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Yuji Maenohara
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naohiro Tachibana
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kosuke Uehara
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fengjun Xuan
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Mori
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasutaka Murahashi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideki Nakamoto
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Oichi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Song Ho Chang
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takumi Matsumoto
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasunori Omata
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fumiko Yano
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
42
|
Lubricin as a tool for controlling adhesion in vivo and ex vivo. Biointerphases 2021; 16:020802. [PMID: 33736436 DOI: 10.1116/6.0000779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The ability to prevent or minimize the accumulation of unwanted biological materials on implantable medical devices is important in maintaining the long-term function of implants. To address this issue, there has been a focus on materials, both biological and synthetic, that have the potential to prevent device fouling. In this review, we introduce a glycoprotein called lubricin and report on its emergence as an effective antifouling coating material. We outline the versatility of lubricin coatings on different surfaces, describe the physical properties of its monolayer structures, and highlight its antifouling properties in improving implant compatibility as well as its use in treatment of ocular diseases and arthritis. This review further describes synthetic polymers mimicking the lubricin structure and function. We also discuss the potential future use of lubricin and its synthetic mimetics as antiadhesive biomaterials for therapeutic applications.
Collapse
|
43
|
Bennett M, Chin A, Lee HJ, Morales Cestero E, Strazielle N, Ghersi-Egea JF, Threlkeld SW, Schmidt TA, Richendrfer HA, Szmydynger-Chodobska J, Jay GD, Chodobski A. Proteoglycan 4 Reduces Neuroinflammation and Protects the Blood-Brain Barrier after Traumatic Brain Injury. J Neurotrauma 2021; 38:385-398. [PMID: 32940130 PMCID: PMC7875610 DOI: 10.1089/neu.2020.7229] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation and dysfunction of the blood-brain barrier (BBB) are two prominent mechanisms of secondary injury in neurotrauma. It has been suggested that Toll-like receptors (TLRs) play important roles in initiating and propagating neuroinflammation resulting from traumatic brain injury (TBI), but potential beneficial effects of targeting these receptors in TBI have not been broadly studied. Here, we investigated the effect of targeting TLRs with proteoglycan 4 (PRG4) on post-traumatic neuroinflammation and BBB function. PRG4 is a mucinous glycoprotein with strong anti-inflammatory properties, exerting its biological effects by interfering with TLR2/4 signaling. In addition, PRG4 has the ability to inhibit activation of cluster of differentiation 44 (CD44), a cell-surface glycoprotein playing an important role in inflammation. Using the controlled cortical impact model of TBI in rats, we showed a rapid and prolonged upregulation of message for TLR2/4 and CD44 in the injured cortex. In the in vitro model of the BBB, recombinant human PRG4 (rhPRG4) crossed the endothelial monolayers through a high-capacity, saturable transport system. In rats sustaining TBI, PRG4 delivery to the brain was enhanced by post-traumatic increase in BBB permeability. rhPRG4 injected intravenously at 1 h post-TBI potently inhibited post-traumatic activation of nuclear factor kappa B and extracellular signal-regulated kinases 1/2, the two major signal transduction pathways associated with TLR2/4 and CD44, and curtailed the post-traumatic influx of monocytes. In addition, PRG4 restored normal BBB function after TBI by preventing the post-traumatic loss of tight junction protein claudin 5 and reduced neuronal death. Our observations provide support for therapeutic strategies targeting TLRs in TBI.
Collapse
Affiliation(s)
- Marissa Bennett
- Neurotrauma and Brain Barriers Research Laboratory, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Andrea Chin
- Neurotrauma and Brain Barriers Research Laboratory, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Hyung Jin Lee
- Neurotrauma and Brain Barriers Research Laboratory, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | | | | | - Jean-François Ghersi-Egea
- FLUID Team and BIP Facility, Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR5292, Lyon 1 University, Lyon, France
| | | | - Tannin A. Schmidt
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Holly A. Richendrfer
- Department of Emergency Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Joanna Szmydynger-Chodobska
- Neurotrauma and Brain Barriers Research Laboratory, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Gregory D. Jay
- Department of Emergency Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Adam Chodobski
- Neurotrauma and Brain Barriers Research Laboratory, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
44
|
Kisla Ekinci RM, Balci S, Dogan H, Ceylaner S, Varan C, Erdem S, Coban F, Bisgin A. Camptodactyly-Arthropathy-Coxa Vara-Pericarditis Syndrome Resembling Juvenile Idiopathic Arthritis: A Single-Center Experience from Southern Turkey. Mol Syndromol 2021; 12:112-117. [PMID: 34012381 DOI: 10.1159/000513111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 11/17/2020] [Indexed: 11/19/2022] Open
Abstract
Camptodactyly-arthropathy-coxa vara-pericarditis (CACP) syndrome, caused by biallelic pathogenic mutations in the PRG4 gene, is characterized by early-onset camptodactyly, noninflammatory arthropathy, coxa vara deformity, and rarely, pericardial effusion. Herein, we report 3 patients with CACP syndrome from 2 unrelated families. All patients are female, born to consanguineous parents, and had camptodactyly since the first years of their lives. Two patients had a prior diagnosis of juvenile idiopathic arthritis. Hip changes were present in 2 patients, and 2 of 3 patients had undergone surgery for camptodactyly. Routine echocardiographic evaluations were normal during the 2-year follow-up. This paper represents the third study including CACP patients from Turkey. Clinically, all 3 patients resembled juvenile idiopathic arthritis cases and received unnecessary medication. There is also an ongoing need for improving awareness of CACP and an effective treatment focusing on the lubrication of the joint space in CACP patients.
Collapse
Affiliation(s)
| | - Sibel Balci
- Department of Pediatric Rheumatology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Haldun Dogan
- INTERGEN Genetic Diagnosis and Research Center, Ankara, Turkey
| | - Serdar Ceylaner
- INTERGEN Genetic Diagnosis and Research Center, Ankara, Turkey
| | - Celal Varan
- Department of Pediatric Cardiology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Sevcan Erdem
- Department of Pediatric Cardiology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Fatma Coban
- Department of Medical Genetics, AGENTEM, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Atil Bisgin
- Department of Medical Genetics, AGENTEM, Cukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
45
|
Dituri F, Scialpi R, Schmidt TA, Frusciante M, Mancarella S, Lupo LG, Villa E, Giannelli G. Proteoglycan-4 is correlated with longer survival in HCC patients and enhances sorafenib and regorafenib effectiveness via CD44 in vitro. Cell Death Dis 2020; 11:984. [PMID: 33199679 PMCID: PMC7669886 DOI: 10.1038/s41419-020-03180-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
Sorafenib and regorafenib administration is among the preferential approaches to treat hepatocellular carcinoma (HCC), but does not provide satisfactory benefits. Intensive crosstalk occurring between cancer cells and other multiple non-cancerous cell subsets present in the surrounding microenvironment is assumed to affect tumor progression. This interplay is mediated by a number of soluble and structural extracellular matrix (ECM) proteins enriching the stromal milieu. Here we assess the HCC tumor expression of the ECM protein proteoglycan 4 (PRG4) and its potential pharmacologic activity either alone, or in combination with sorafenib and regorafenib. PRG4 mRNA levels resulted strongly correlated with increased survival rate of HCC patients (p = 0.000) in a prospective study involving 78 HCC subjects. We next showed that transforming growth factor beta stimulates PRG4 expression and secretion by primary human HCC cancer-associated fibroblasts, non-invasive HCC cell lines, and ex vivo specimens. By functional tests we found that recombinant human PRG4 (rhPRG4) impairs HCC cell migration. More importantly, the treatment of HCC cells expressing CD44 (the main PRG4 receptor) with rhPRG4 dramatically enhances the growth-limiting capacity of sorafenib and regorafenib, whereas not significantly affecting cell proliferation per se. Conversely, rhPRG4 only poorly potentiates drug effectiveness on low CD44-expressing or stably CD44-silenced HCC cells. Overall, these data suggest that the physiologically-produced compound PRG4 may function as a novel tumor-suppressive agent by strengthening sorafenib and regorafenib effects in the treatment of HCC.
Collapse
Affiliation(s)
- Francesco Dituri
- National Institute of Gastroenterology "S. De Bellis" Research Hospital, 70013, Castellana Grotte, Italy.
| | - Rosanna Scialpi
- National Institute of Gastroenterology "S. De Bellis" Research Hospital, 70013, Castellana Grotte, Italy
| | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Centre, Farmington, CT, USA
| | - Martina Frusciante
- National Institute of Gastroenterology "S. De Bellis" Research Hospital, 70013, Castellana Grotte, Italy
| | - Serena Mancarella
- National Institute of Gastroenterology "S. De Bellis" Research Hospital, 70013, Castellana Grotte, Italy
| | - Luigi Giovanni Lupo
- University of Bari, Department of General Surgery and Liver Transplantation, Policlinico - piazza Giulio Cesare 14, 70125, Bari, Italy
| | - Erica Villa
- Gastroenterology Unit, Department of Internal Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "S. De Bellis" Research Hospital, 70013, Castellana Grotte, Italy
| |
Collapse
|
46
|
McQuitty CE, Williams R, Chokshi S, Urbani L. Immunomodulatory Role of the Extracellular Matrix Within the Liver Disease Microenvironment. Front Immunol 2020; 11:574276. [PMID: 33262757 PMCID: PMC7686550 DOI: 10.3389/fimmu.2020.574276] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease when accompanied by underlying fibrosis, is characterized by an accumulation of extracellular matrix (ECM) proteins and chronic inflammation. Although traditionally considered as a passive and largely architectural structure, the ECM is now being recognized as a source of potent damage-associated molecular pattern (DAMP)s with immune-active peptides and domains. In parallel, the ECM anchors a range of cytokines, chemokines and growth factors, all of which are capable of modulating immune responses. A growing body of evidence shows that ECM proteins themselves are capable of modulating immunity either directly via ligation with immune cell receptors including integrins and TLRs, or indirectly through release of immunoactive molecules such as cytokines which are stored within the ECM structure. Notably, ECM deposition and remodeling during injury and fibrosis can result in release or formation of ECM-DAMPs within the tissue, which can promote local inflammatory immune response and chemotactic immune cell recruitment and inflammation. It is well described that the ECM and immune response are interlinked and mutually participate in driving fibrosis, although their precise interactions in the context of chronic liver disease are poorly understood. This review aims to describe the known pro-/anti-inflammatory and fibrogenic properties of ECM proteins and DAMPs, with particular reference to the immunomodulatory properties of the ECM in the context of chronic liver disease. Finally, we discuss the importance of developing novel biotechnological platforms based on decellularized ECM-scaffolds, which provide opportunities to directly explore liver ECM-immune cell interactions in greater detail.
Collapse
Affiliation(s)
- Claire E. McQuitty
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Roger Williams
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Shilpa Chokshi
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Luca Urbani
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
47
|
|
48
|
Wang T, Hao Z, Liu C, Yuan L, Li L, Yin M, Li Q, Qi Z, Wang Z. MiR-193b modulates osteoarthritis progression through targeting ST3GAL4 via sialylation of CD44 and NF-кB pathway. Cell Signal 2020; 76:109814. [PMID: 33080315 DOI: 10.1016/j.cellsig.2020.109814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/30/2020] [Accepted: 10/16/2020] [Indexed: 01/08/2023]
Abstract
Osteoarthritis (OA) is a worldwide epidemic and debilitating disease. It is urgent to explore the potential molecular mechanisms of OA which has crucial roles in the treatment strategy. As a post-translational modification, sialylation mediates the progression of OA. In current study, differential expression of sialyltransferases (STs) in normal and OA cartilage tissues is detected. The ST3GAL4 expression is significantly increased and positively associated with modified Mankin's score in OA tissue. Alteration of ST3GAL4 respectively mediates the degradation of extracellular mechanisms (ECM), apoptosis and proliferation in chondrocytes. Additionally, miR-193b is identified as a direct regulatory target of ST3GAL4. Functional analysis shows that modulation of ST3GAL4 could be reversed by miR-193b. Over-expression ST3GAL4 modifies CD44 sialylation. Finally, sialylated CD44 reduces the binding capacity to lubricin and mediates the activity of the NF-кB pathway. Collectively, these researches indicate that miR-193b/ST3GAL4 axis impacts OA progression by regulating CD44 sialylation via NF-кB pathway. Our researches propose a precise molecular mechanism and provide a prospective therapeutic target in OA.
Collapse
Affiliation(s)
- Tianfu Wang
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian 116033, Liaoning Province, China; Department of Spinal Surgery, The Second Hospital of Dalian Medical University, Dalian 116033, Liaoning Province, China
| | - Zhiyu Hao
- Department of Imaging and Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian 116033, Liaoning Province, China
| | - Changcheng Liu
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian 116033, Liaoning Province, China
| | - Lebin Yuan
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian 116033, Liaoning Province, China
| | - Li Li
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian 116033, Liaoning Province, China
| | - Menghong Yin
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian 116033, Liaoning Province, China
| | - Qing Li
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian 116033, Liaoning Province, China
| | - Zhiming Qi
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian 116033, Liaoning Province, China
| | - Zi Wang
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian 116033, Liaoning Province, China.
| |
Collapse
|
49
|
Wang Y, Gludish DW, Hayashi K, Todhunter RJ, Krotscheck U, Johnson PJ, Cummings BP, Su J, Reesink HL. Synovial fluid lubricin increases in spontaneous canine cruciate ligament rupture. Sci Rep 2020; 10:16725. [PMID: 33028842 PMCID: PMC7542452 DOI: 10.1038/s41598-020-73270-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
Lubricin is an important boundary lubricant and chondroprotective glycoprotein in synovial fluid. Both increased and decreased synovial fluid lubricin concentrations have been reported in experimental post-traumatic osteoarthritis (PTOA) animal models and in naturally occurring joint injuries in humans and animals, with no consensus about how lubricin is altered in different species or injury types. Increased synovial fluid lubricin has been observed following intra-articular fracture in humans and horses and in human late-stage osteoarthritis; however, it is unknown how synovial lubricin is affected by knee-destabilizing injuries in large animals. Spontaneous rupture of cranial cruciate ligament (RCCL), the anterior cruciate ligament equivalent in quadrupeds, is a common injury in dogs often accompanied by OA. Here, clinical records, radiographs, and synovial fluid samples from 30 dogs that sustained RCCL and 9 clinically healthy dogs were analyzed. Synovial fluid lubricin concentrations were nearly 16-fold greater in RCCL joints as compared to control joints, while IL-2, IL-6, IL-8, and TNF-α concentrations did not differ between groups. Synovial fluid lubricin concentrations were correlated with the presence of radiographic OA and were elevated in three animals sustaining RCCL injury prior to the radiographic manifestation of OA, indicating that lubricin may be a potential biomarker for early joint injury.
Collapse
Affiliation(s)
- Yuyan Wang
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - David W Gludish
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Kei Hayashi
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Rory J Todhunter
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Ursula Krotscheck
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Philippa J Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | | | - Jin Su
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Heidi L Reesink
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
50
|
Watkins AR, Reesink HL. Lubricin in experimental and naturally occurring osteoarthritis: a systematic review. Osteoarthritis Cartilage 2020; 28:1303-1315. [PMID: 32504786 PMCID: PMC8043104 DOI: 10.1016/j.joca.2020.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/28/2020] [Accepted: 05/13/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Lubricin is increasingly being evaluated as an outcome measure in studies investigating post-traumatic and naturally occurring osteoarthritis. However, there are discrepancies in results, making it unclear as to whether lubricin is increased, decreased or unchanged in osteoarthritis. The purpose of this study was to review all papers that measured lubricin in joint injury or osteoarthritis in order to draw conclusions about lubricin regulation in joint disease. DESIGN A systematic search of the Pubmed, Web of Knowledge, and EBSCOhost databases for papers was performed. Inclusion criteria were in vivo studies that measured lubricin in humans or animals with joint injury, that investigated lubricin supplementation in osteoarthritic joints, or that described the phenotype of a lubricin knock-out model. A methodological assessment was performed. RESULTS Sixty-two studies were included, of which thirty-eight measured endogenous lubricin in joint injury or osteoarthritis. Nineteen papers found an increase or no change in lubricin and nineteen reported a decrease. Papers that reported a decrease in lubricin were cited four times more often than those that reported an increase. Fifteen papers described lubricin supplementation, and all reported a beneficial effect. Eleven papers described lubricin knock-out models. CONCLUSIONS The human literature reveals similar distributions of papers reporting increased lubricin as compared to decreased lubricin in osteoarthritis. The animal literature is dominated by reports of decreased lubricin in the rat anterior cruciate ligament transection model, whereas studies in large animal models report increased lubricin. Intra-articular lubricin supplementation may be beneficial regardless of whether lubricin increases or decreases in OA.
Collapse
Affiliation(s)
- A R Watkins
- Department of Clinical Sciences, University of Pennsylvania School of Veterinary Medicine, New Bolton Center, Kennett Square, PA, USA
| | - H L Reesink
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|