1
|
Zou X, Xu H, Qian W. The role and current research status of resveratrol in the treatment of osteoarthritis and its mechanisms: a narrative review. Drug Metab Rev 2024; 56:399-412. [PMID: 39376171 DOI: 10.1080/03602532.2024.2402751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Osteoarthritis (OA) is a chronic degenerative disease caused by various factors such as aging, obesity, trauma, and genetics. It is a challenging condition faced by orthopedic doctors in clinical practice and places a heavy burden on patients and their families. Currently, the treatment of OA primarily focuses on symptomatic relief and lacks ideal therapeutic methods. Resveratrol is a natural polyphenolic compound with anti-inflammatory and antioxidant properties, and in recent years, it has gained attention as a candidate drug for OA treatment. This article provides an overview of the research status on the role and mechanisms of resveratrol in treating OA. It has been found that resveratrol can prevent the development of OA by inhibiting inflammatory responses, protecting chondrocytes, maintaining cartilage homeostasis, promoting autophagy, and has shown certain therapeutic effects. This process may be related to the regulation of signaling pathways such as nuclear factor-kappa B (NF-κB), Toll-like receptor 4 (TLR4), and silent information regulator 1 (SIRT1). We summarize the current molecular mechanisms of resveratrol in treating OA, hoping to provide a reference for further research and application of resveratrol in OA treatment.
Collapse
Affiliation(s)
- Xiongfei Zou
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Hongjun Xu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Wenwei Qian
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
2
|
Zhang C, Pathrikar TV, Baby HM, Li J, Zhang H, Selvadoss A, Ovchinnikova A, Ionescu A, Chubinskaya S, Miller RE, Bajpayee AG. Charge-Reversed Exosomes for Targeted Gene Delivery to Cartilage for Osteoarthritis Treatment. SMALL METHODS 2024; 8:e2301443. [PMID: 38607953 PMCID: PMC11470115 DOI: 10.1002/smtd.202301443] [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: 10/19/2023] [Revised: 03/18/2024] [Indexed: 04/14/2024]
Abstract
Gene therapy has the potential to facilitate targeted expression of therapeutic proteins to promote cartilage regeneration in osteoarthritis (OA). The dense, avascular, aggrecan-glycosaminoglycan (GAG) rich negatively charged cartilage, however, hinders their transport to reach chondrocytes in effective doses. While viral vector mediated gene delivery has shown promise, concerns over immunogenicity and tumorigenic side-effects persist. To address these issues, this study develops surface-modified cartilage-targeting exosomes as non-viral carriers for gene therapy. Charge-reversed cationic exosomes are engineered for mRNA delivery by anchoring cartilage targeting optimally charged arginine-rich cationic motifs into the anionic exosome bilayer by using buffer pH as a charge-reversal switch. Cationic exosomes penetrated through the full-thickness of early-stage arthritic human cartilage owing to weak-reversible ionic binding with GAGs and efficiently delivered the encapsulated eGFP mRNA to chondrocytes residing in tissue deep layers, while unmodified anionic exosomes do not. When intra-articularly injected into destabilized medial meniscus mice knees with early-stage OA, mRNA loaded charge-reversed exosomes overcame joint clearance and rapidly penetrated into cartilage, creating an intra-tissue depot and efficiently expressing eGFP; native exosomes remained unsuccessful. Cationic exosomes thus hold strong translational potential as a platform technology for cartilage-targeted non-viral delivery of any relevant mRNA targets for OA treatment.
Collapse
Affiliation(s)
- Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Tanvi V. Pathrikar
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Helna M. Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Jun Li
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | | | - Andreia Ionescu
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical College, Chicago, IL 60612, USA
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
3
|
Liu Y, Shi Y, Zhang M, Han F, Liao W, Duan X. Natural polyphenols for drug delivery and tissue engineering construction: A review. Eur J Med Chem 2024; 266:116141. [PMID: 38237341 DOI: 10.1016/j.ejmech.2024.116141] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 02/05/2024]
Abstract
Polyphenols, natural compounds rich in phenolic structures, are gaining prominence due to their antioxidant, anti-inflammatory, antibacterial, and anticancer properties, making them valuable in biomedical applications. Through covalent and noncovalent interactions, polyphenols can bind to biomaterials, enhancing their performance and compensating for their shortcomings. Such polyphenol-based biomaterials not only increase the efficacy of polyphenols but also improve drug stability, control release kinetics, and boost the therapeutic effects of drugs. They offer the potential for targeted drug delivery, reducing off-target impacts and enhancing therapeutic outcomes. In tissue engineering, polyphenols promote cell adhesion, proliferation, and differentiation, thus aiding in the formation of functional tissues. Additionally, they offer excellent biocompatibility and mechanical strength, essential in designing scaffolds. This review explores the significant roles of polyphenols in tissue engineering and drug delivery, emphasizing their potential in advancing biomedical research and healthcare.
Collapse
Affiliation(s)
- Yu Liu
- Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiangxi, China; Jiujiang Clinical Precision Medicine Research Center, Jiangxi, China; Medical College of Jiujiang University, Jiangxi, China
| | - Yuying Shi
- Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiangxi, China; Jiujiang Clinical Precision Medicine Research Center, Jiangxi, China; Medical College of Jiujiang University, Jiangxi, China
| | - Mengqi Zhang
- Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiangxi, China; Jiujiang Clinical Precision Medicine Research Center, Jiangxi, China; Medical College of Jiujiang University, Jiangxi, China
| | - Feng Han
- Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiangxi, China; Jiujiang Clinical Precision Medicine Research Center, Jiangxi, China; Medical College of Jiujiang University, Jiangxi, China
| | - Weifang Liao
- Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiangxi, China; Jiujiang Clinical Precision Medicine Research Center, Jiangxi, China; Medical College of Jiujiang University, Jiangxi, China
| | - Xunxin Duan
- Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiangxi, China; Jiujiang Clinical Precision Medicine Research Center, Jiangxi, China; Medical College of Jiujiang University, Jiangxi, China.
| |
Collapse
|
4
|
Mehta S, Boyer TL, Akhtar S, He T, Zhang C, Vedadghavami A, Bajpayee AG. Sustained intra-cartilage delivery of interleukin-1 receptor antagonist using cationic peptide and protein-based carriers. Osteoarthritis Cartilage 2023; 31:780-792. [PMID: 36739939 PMCID: PMC10392024 DOI: 10.1016/j.joca.2023.01.573] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/20/2022] [Accepted: 01/17/2023] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Blocking the interleukin-1 (IL-1) catabolic cascade following joint trauma can be achieved using its receptor antagonist, IL-1Ra. However, its clinical translation for osteoarthritis therapy has been unsuccessful due to its rapid joint clearance and lack of targeting and penetration into deep cartilage layers at therapeutic concentrations. Here, we target the high negative charge of cartilage aggrecan-glycosaminoglycans (GAGs) by attaching cationic carriers to IL-1Ra. IL-1Ra was conjugated to the cartilage targeting glycoprotein, Avidin, and a short length optimally charged cationic peptide carrier (CPC+14). It is hypothesized that electro-diffusive transport and binding properties of IL-1Ra-Avidin and IL-1Ra-CPC+14 will create intra-cartilage depots of IL-1Ra, resulting in long-term suppression of IL-1 catabolism with only a single administration. DESIGN IL-1Ra was conjugated to Avidin or CPC+14 using site specific maleimide linkers, and confirmed using gel electrophoresis, high-performance liquid chromatography (HPLC), and mass spectrometry. Intra-cartilage transport and retention of conjugates was compared with native IL-1Ra. Attenuation of IL-1 catabolic signaling with one-time dose of IL-1Ra-CPC+14 and IL-1Ra-Avidin was assessed over 16 days using IL-1α challenged bovine cartilage and compared with unmodified IL-1Ra. RESULTS Positively charged IL-1Ra penetrated through the full-thickness of cartilage, creating a drug depot. A single dose of unmodified IL-1Ra was not sufficient to attenuate IL-1-induced cartilage deterioration over 16 days. However, when delivered using Avidin, and to a greater extent CPC+14, IL-1Ra significantly suppressed cytokine induced GAG loss and nitrite release while improving cell metabolism and viability. CONCLUSION Charge-based cartilage targeting drug delivery systems hold promise as they can enable long-term therapeutic benefit with only a single dose.
Collapse
Affiliation(s)
- S Mehta
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - T L Boyer
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - S Akhtar
- Department of Biochemistry, Northeastern University, Boston, MA, USA.
| | - T He
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - C Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - A Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - A G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Mechanical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
5
|
Zhang C, Vedadghavami A, He T, Charles JF, Bajpayee AG. Cationic Carrier Mediated Delivery of Anionic Contrast Agents in Low Doses Enable Enhanced Computed Tomography Imaging of Cartilage for Early Osteoarthritis Diagnosis. ACS NANO 2023; 17:6649-6663. [PMID: 36989423 PMCID: PMC10629240 DOI: 10.1021/acsnano.2c12376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/23/2023] [Indexed: 06/03/2023]
Abstract
Cartilage tissue exhibits early degenerative changes with onset of osteoarthritis (OA). Early diagnosis is critical as there is only a narrow time window during which therapeutic intervention can reverse disease progression. Computed tomography (CT) has been considered for cartilage imaging as a tool for early OA diagnosis by introducing radio-opaque contrast agents like ioxaglate (IOX) into the joint. IOX, however, is anionic and thus repelled by negatively charged cartilage glycosaminoglycans (GAGs) that hinders its intra-tissue penetration and partitioning, resulting in poor CT attenuation. This is further complicated by its short intra-tissue residence time owing to rapid clearance from joints, which necessitates high doses causing toxicity concerns. Here we engineer optimally charged cationic contrast agents based on cartilage negative fixed charge density by conjugating cartilage targeting a cationic peptide carrier (CPC) and multi-arm avidin nanoconstruct (mAv) to IOX, such that they can penetrate through the full thickness of cartilage within 6 h using electrostatic interactions and elicit similar CT signal with about 40× lower dose compared to anionic IOX. Their partitioning and distribution correlate strongly with spatial GAG distribution within healthy and early- to late-stage arthritic bovine cartilage tissues at 50-100× lower doses than other cationic contrast agents used in the current literature. The use of contrast agents at low concentrations also allowed for delineation of cartilage from subchondral bone as well as other soft tissues in rat tibial joints. These contrast agents are safe to use at current doses, making CT a viable imaging modality for early detection of OA and staging of its severity.
Collapse
Affiliation(s)
- Chenzhen Zhang
- Department
of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Armin Vedadghavami
- Department
of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Tengfei He
- Department
of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Julia F. Charles
- Department
of Orthopaedic Surgery, Brigham and Women’s
Hospital, 60 Fenwood Road, Boston, Massachusetts 02115, United States
| | - Ambika G. Bajpayee
- Department
of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
6
|
Ben Tahar S, Garnier J, Eller K, DiMauro N, Piet J, Mehta S, Bajpayee AG, Shefelbine SJ. Adolescent obesity incurs adult skeletal deficits in murine induced obesity model. J Orthop Res 2023; 41:386-395. [PMID: 35578981 DOI: 10.1002/jor.25378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/06/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023]
Abstract
Adolescent obesity has risen dramatically in the last few decades. While adult obesity may be osteoprotective, the effects of obesity during adolescence, which is a period of massive bone accrual, are not clear. We used a murine model of induced adolescent obesity to examine the structural, mechanical, and compositional differences between obese and healthy weight bone in 16-week-old female C57Bl6 mice. We also examined the effects of a return to normal weight after skeletal maturity (24 weeks old). We found obese adolescent bone exhibited decreased trabecular bone volume, increased cortical diameter, increased ultimate stress, and increased brittleness (decreased plastic energy to fracture), similar to an aging phenotype. The trabecular bone deficits remained after return to normal weight after skeletal maturity. However, after returning to normal diet, there was no difference in ultimate stress nor plastic energy to fracture between groups as the normal diet group increased ultimate stress and brittleness. Interestingly, compositional changes appeared in the former high-fat diet mice after skeletal maturity with a lower mineral to matrix ratio compared to normal diet mice. In addition there was a trend toward increased fluorescent advanced glycation endproducts in the former high-fat diet mice compared to normal diet mice but this did not reach significance (p < 0.05) due to the large variability. The skeletal consequences of adolescent obesity may have lasting implications for the adult skeleton even after return to normal weight. Given the rates of adolescent obesity, skeletal health should be a concern.
Collapse
Affiliation(s)
- Soha Ben Tahar
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Julien Garnier
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Kerry Eller
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Nicole DiMauro
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Judith Piet
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Shihkar Mehta
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA.,Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
7
|
He T, Zhang C, Colombani T, Bencherif SA, Porter RM, Bajpayee AG. Intra-articular kinetics of a cartilage targeting cationic PEGylated protein for applications in drug delivery. Osteoarthritis Cartilage 2023; 31:187-198. [PMID: 36241136 PMCID: PMC9892226 DOI: 10.1016/j.joca.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 08/22/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Cartilage targeting cationic glycoprotein Avidin was PEGylated to synthesize a multi-arm Avidin (mAv) nano-construct with high drug loading content. Here we investigate mAv biodistribution and kinetics over a 7-day period following intra-articular (IA) administration in rat knee joints. METHODS Labeled mAv was injected into healthy rat knees, and joint tissues (articular cartilage, menisci, ligaments, tendons, fat pad) were harvested following sacrifice at 6 h, 1, 4 and 7 days. Its IA biodistribution and retention were measured using fluorescence microscopy. Tissue localization was compared in young vs old rats by immunohistochemistry. mAv chondrotoxicity and immune response were evaluated to determine safe carrier dose limits. RESULTS mAv penetrated through the full thickness of rat cartilage and other joint tissues within 6 h, remaining detectable within most joint tissues over 7 days. Intra-tissue uptake correlated strongly with tissue GAG concentration, confirming the dominant role of electrostatic interactions between positively charged mAv and the negatively charged aggrecan proteoglycans. mAv was uptaken by chondrocytes and also penetrated the osteocyte lacuno-canalicular system of peri-articular bone in both young and old rats. mAv did not cause cytotoxicity at concentrations up to 300 μM but elicited a dose dependent immunogenic response. CONCLUSIONS mAv's ability to target a variety of joint tissues, chondrocytes, and peri-articular osteocytes without sequestration in synovial fluid makes it a versatile carrier for delivering a wide range of drugs for treating a broad class of musculoskeletal diseases. Drugs can be conjugated using simple aqueous based avidin-biotin reaction, supporting its clinical prospects.
Collapse
Affiliation(s)
- T He
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - C Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - T Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - S A Bencherif
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA; Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - R M Porter
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - A G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA; Department of Mechanical Engineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Sharma A, Chabloz S, Lapides RA, Roider E, Ewald CY. Potential Synergistic Supplementation of NAD+ Promoting Compounds as a Strategy for Increasing Healthspan. Nutrients 2023; 15:nu15020445. [PMID: 36678315 PMCID: PMC9861325 DOI: 10.3390/nu15020445] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Disrupted biological function, manifesting through the hallmarks of aging, poses one of the largest threats to healthspan and risk of disease development, such as metabolic disorders, cardiovascular ailments, and neurodegeneration. In recent years, numerous geroprotectors, senolytics, and other nutraceuticals have emerged as potential disruptors of aging and may be viable interventions in the immediate state of human longevity science. In this review, we focus on the decrease in nicotinamide adenine dinucleotide (NAD+) with age and the supplementation of NAD+ precursors, such as nicotinamide mononucleotide (NMN) or nicotinamide riboside (NR), in combination with other geroprotective compounds, to restore NAD+ levels present in youth. Furthermore, these geroprotectors may enhance the efficacy of NMN supplementation while concurrently providing their own numerous health benefits. By analyzing the prevention of NAD+ degradation through the inhibition of CD38 or supporting protective downstream agents of SIRT1, we provide a potential framework of the CD38/NAD+/SIRT1 axis through which geroprotectors may enhance the efficacy of NAD+ precursor supplementation and reduce the risk of age-related diseases, thereby potentiating healthspan in humans.
Collapse
Affiliation(s)
- Arastu Sharma
- Laboratory of Extracellular Matrix Regeneration, Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zürich, 8603 Schwerzenbach, Switzerland
- AVEA Life AG, Bahnhofplatz, 6300 Zug, Switzerland
| | | | - Rebecca A. Lapides
- Department of Dermatology, University Hospital of Basel, 4031 Basel, Switzerland
- Robert Larner, MD College of Medicine at the University of Vermont, Burlington, VT 05405, USA
| | - Elisabeth Roider
- Department of Dermatology, University Hospital of Basel, 4031 Basel, Switzerland
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Maximon AG, Bahnhofplatz, 6300 Zug, Switzerland
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zürich, 8603 Schwerzenbach, Switzerland
- Correspondence:
| |
Collapse
|
9
|
Zhou J, Wang Q. Daphnoretin relieves IL-1β-mediated chondrocytes apoptosis via repressing endoplasmic reticulum stress and NLRP3 inflammasome. J Orthop Surg Res 2022; 17:487. [PMID: 36384642 PMCID: PMC9670399 DOI: 10.1186/s13018-022-03316-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Background Osteoarthritis (OA), mainly caused by severe joint degeneration, is often accompanied by joint pain and dysfunction syndrome. Inflammatory mediators and apoptosis play key roles in the evolution of OA. It is reported that daphnoretin has significant antiviral and anti-tumor values. The present study aims at investigating the role of daphnoretin in OA. Methods The OA mouse model was constructed by performing the destabilization of the medial meniscus through surgery, and the OA cell model was induced in ATDC5 chondrocytes with IL-1β (10 ng/mL) in vitro. Chondrocyte viability and apoptosis were measured by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT), Caspase-3 activity, and flow cytometry. The levels of COX-2, iNOS, TNF-α, IL-6, Bax, Bcl2, cleaved-Caspase3, endoplasmic reticulum stress (ERS) proteins (GRP78, CHOP, ATF6, and Caspase-12), and NLRP3-ASC-Caspase1 inflammasome were determined by quantitative real-time PCR or western blot. The concentrations of TNF-α, IL-6, and PGE2 were tested by enzyme-linked immunosorbent assay. The content of nitrates was detected by the Griess method. In vivo, morphologic differences in knee joint sections and the thickness of the subchondral bone density plate in mice were observed by hematoxylin–eosin (H&E) staining and safranin O-fast green staining. Results Daphnoretin effectively choked IL-1β-induced chondrocyte apoptosis and facilitated cell viability. Daphnoretin dose-dependently abated ERS, inflammatory mediators, and the activation of NLRP3 inflammasomes in IL-1β-induced chondrocytes. What’s more, in vivo experiments confirmed that daphnoretin alleviated OA progression in a murine OA model by mitigating inflammation and ERS. Conclusion Daphnoretin alleviated IL-1β-induced chondrocyte apoptosis by hindering ERS and NLRP3 inflammasome. Graphical abstract ![]()
Collapse
|
10
|
Warren MR, Vedadghavami A, Bhagavatula S, Bajpayee AG. Effects of polycationic drug carriers on the electromechanical and swelling properties of cartilage. Biophys J 2022; 121:3542-3561. [PMID: 35765244 PMCID: PMC9515003 DOI: 10.1016/j.bpj.2022.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/15/2022] Open
Abstract
Cationic nanocarriers offer a promising solution to challenges in delivering drugs to negatively charged connective tissues, such as to articular cartilage for the treatment of osteoarthritis (OA). However, little is known about the effects that cationic macromolecules may have on the mechanical properties of cartilage at high interstitial concentrations. We utilized arginine-rich cationic peptide carriers (CPCs) with varying net charge (from +8 to +20) to investigate the biophysical mechanisms of nanocarrier-induced alterations to cartilage biomechanical properties. We observed that CPCs increased the compressive modulus of healthy bovine cartilage explants by up to 70% and decreased the stiffness of glycosaminoglycan-depleted tissues (modeling OA) by 69%; in both cases, the magnitude of the change in stiffness correlated with the uptake of CPC charge variants. Next, we directly measured CPC-induced osmotic deswelling in cartilage tissue due to shielding of charge repulsions between anionic extracellular matrix constituents, with magnitudes of reductions between 36 and 64 kPa. We then demonstrated that electrostatic interactions were required for CPC-induced stiffening to occur, evidenced by no observed increase in tissue stiffness when measured in hypertonic bathing salinity. We applied a non-ideal Donnan osmotic model (under triphasic theory) to separate bulk modulus measurements into Donnan and non-Donnan components, which further demonstrated the conflicting charge-shielding and matrix-stiffening effects of CPCs. These results show that cationic drug carriers can alter tissue mechanical properties via multiple mechanisms, including the expected charge shielding as well as a novel stiffening phenomenon mediated by physical linkages. We introduce a model for how the magnitudes of these mechanical changes depend on tunable physical properties of the drug carrier, including net charge, size, and spatial charge distribution. We envision that the results and theory presented herein will inform the design of future cationic drug-delivery systems intended to treat diseases in a wide range of connective tissues.
Collapse
Affiliation(s)
- Matthew R Warren
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Sanjana Bhagavatula
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts; Department of Mechanical Engineering, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
11
|
Wang Y, Zhao H, Jia S, Wang Q, Yao W, Yang Y, Bai L. Senomorphic agent pterostilbene ameliorates osteoarthritis through the PI3K/AKT/NF-κB axis: an in vitro and in vivo study. Am J Transl Res 2022; 14:5243-5262. [PMID: 36105068 PMCID: PMC9452324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Osteoarthritis (OA) is the most common joint disease in the world. Among the many risk factors for OA, aging is one of the most critical factors. The treatment with senop-associated secretory phenotype (SASP) is one of the important, promising anti-aging strategies at present. Pterostilbene (PTE) is a trans-stilbene compound with anti-tumor, anti-oxidation, anti-inflammatory, and anti-aging pharmacologic activities. The purpose of this study is to explore the therapeutic effects of PTE on articular chondrocyte senescence and OA and its related mechanisms. METHODS Male Sprague-Dawley rats were operated on with transection of the anterior cruciate ligament (ACLT) and a destabilized medial meniscus (DMM) surgery to establish the OA model and then injected intraperitoneally with PTE (20 mg/kg) for 5 weeks. Finally, rats were sacrificed and knee joints were collected for histologic analysis. Rat chondrocytes were stimulated with interleukin-1β (IL-1β) with or without PTE treatment. The therapeutic effects of PTE and related mechanisms were investigated by examining and analyzing relative markers through senescence-associated β-galactosidase (SA-β-Gal) assay, cell cycle, qRT-PCR, western blot, bioinformatic analysis, immunofluorescence, and molecular modeling. RESULTS With in vivo experiments, PTE can significantly reduce the Mankin scores and OARSI scores of the knee joint in ACLT+DMM OA model rats and reduce the interleukin-6 (IL-6) level in the knee lavage fluid. Immunohistochemical staining showed that compared to the OA group, the PTE treatment group had significantly increased expression of collagen type II in articular cartilage, and significantly decreased matrix metalloproteinase 13 (MMP-13) and IL-6, the main SASP proteins, and had expression of p16 and p21, markers of aging in chondrocytes. In vitro, PTE reduced the ratio of SA-β-Gal positive chondrocytes and G0-G1 phase chondrocytes in IL-1β-induced rat chondrocytes. PTE significantly inhibited the expression of MMP-13, IL-6, thrombospondin motif 5 (ADAMTS5), p16, and p21, and significantly increased the expression of collagen type II. Bioassay and subsequent western blot showed that PTE significantly inhibited the activation of PI3K/AKT and NF-κB signaling pathways. The results of molecular docking experiments showed that PTE could bind closely to the sites of PI3K protein, thereby inhibiting the phosphorylation of PI3K. CONCLUSIONS The experimental results indicate that PTE plays an anti-chondrocyte senescence role in the treatment of OA by inhibiting the PI3K/AKT/NF-κB signaling pathway and reducing expression of SASP.
Collapse
Affiliation(s)
- Yu Wang
- Department of Orthopedic Surgery, Shengjing Hospital Affiliated China Medical UniversityShenyang 110004, Liaoning, PR China
- Department of Orthopedics Surgery, Affiliated Hospital of Chengde Medical College36 Nanyingzi Street, Shuangqiao District, Chengde 067000, Hebei, PR China
| | - Huai Zhao
- Department of General Surgery, Shengjing Hospital Affiliated China Medical UniversityShenyang 110004, Liaoning PR China
| | - Shuangshuo Jia
- Department of Orthopedic Surgery, Shengjing Hospital Affiliated China Medical UniversityShenyang 110004, Liaoning, PR China
| | - Qian Wang
- Department of Orthopedics Surgery, Affiliated Hospital of Chengde Medical College36 Nanyingzi Street, Shuangqiao District, Chengde 067000, Hebei, PR China
| | - Wuyi Yao
- Department of Orthopedics Surgery, Affiliated Hospital of Chengde Medical College36 Nanyingzi Street, Shuangqiao District, Chengde 067000, Hebei, PR China
| | - Yue Yang
- Department of Orthopedic Surgery, Shengjing Hospital Affiliated China Medical UniversityShenyang 110004, Liaoning, PR China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital Affiliated China Medical UniversityShenyang 110004, Liaoning, PR China
| |
Collapse
|
12
|
Yang S, Sun M, Zhang X. Protective Effect of Resveratrol on Knee Osteoarthritis and its Molecular Mechanisms: A Recent Review in Preclinical and Clinical Trials. Front Pharmacol 2022; 13:921003. [PMID: 35959426 PMCID: PMC9357872 DOI: 10.3389/fphar.2022.921003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/21/2022] [Indexed: 12/04/2022] Open
Abstract
Osteoarthritis (OA) is one of the progressing chronic joint associated with by many complex factors such as age, obesity, and trauma. Knee osteoarthritis (KOA) is the most common type of OA. KOA is characterized by articular cartilage destruction and degeneration, synovial inflammation, and abnormal subchondral bone changes. To date, no practical clinical approach has been able to modify the pathological progression of KOA. Drug therapy is limited to pain control and may lead to serious side effects when taken for a long time. Therefore, searching for safer and more reliable treatments has become necessary. Interestingly, more and more research has focused on natural products, and monomeric compounds derived from natural products have received much attention as drug candidates for KOA treatment. Resveratrol (RES), a natural phenolic compound, has various pharmacological and biological activities, including anti-cancer, anti-apoptotic, and anti-decay. Recently, studies on the effects of RES on maintaining the normal homeostasis of chondrocytes in KOA have received increasing attention, which seems to be attributed to the multi-targeted effects of RES on chondrocyte function. This review summarizes preclinical trials, clinical trials, and emerging tissue engineering studies of RES for KOA and discusses the specific mechanisms by which RES alleviates KOA. A better understanding of the pharmacological role of RES in KOA could provide clinical implications for intervention in the development of KOA.
Collapse
Affiliation(s)
| | - Mingli Sun
- *Correspondence: Mingli Sun, ; Xinan Zhang,
| | | |
Collapse
|
13
|
Vedadghavami A, Hakim B, He T, Bajpayee AG. Cationic peptide carriers enable long-term delivery of insulin-like growth factor-1 to suppress osteoarthritis-induced matrix degradation. Arthritis Res Ther 2022; 24:172. [PMID: 35858920 PMCID: PMC9297664 DOI: 10.1186/s13075-022-02855-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Insulin-like growth factor-1 (IGF-1) has the potential to be used for osteoarthritis (OA) treatment but has not been evaluated in clinics yet owing to toxicity concerns. It suffers from short intra-joint residence time and a lack of cartilage targeting following its intra-articular administration. Here, we synthesize an electrically charged cationic formulation of IGF-1 by using a short-length arginine-rich, hydrophilic cationic peptide carrier (CPC) with a net charge of +14, designed for rapid and high uptake and retention in both healthy and arthritic cartilage. METHODS IGF-1 was conjugated to CPC by using a site-specific sulfhydryl reaction via a bifunctional linker. Intra-cartilage depth of penetration and retention of CPC-IGF-1 was compared with the unmodified IGF-1. The therapeutic effectiveness of a single dose of CPC-IGF-1 was compared with free IGF-1 in an IL-1α-challenged cartilage explant culture post-traumatic OA model. RESULTS CPC-IGF-1 rapidly penetrated through the full thickness of cartilage creating a drug depot owing to electrostatic interactions with negatively charged aggrecan-glycosaminoglycans (GAGs). CPC-IGF-1 remained bound within the tissue while unmodified IGF-1 cleared out. Treatment with a single dose of CPC-IGF-1 effectively suppressed IL-1α-induced GAG loss and nitrite release and rescued cell metabolism and viability throughout the 16-day culture period, while free IGF at the equivalent dose was not effective. CONCLUSIONS CPC-mediated depot delivery of IGF-1 protected cartilage by suppressing cytokine-induced catabolism with only a single dose. CPC is a versatile cationic motif that can be used for intra-cartilage delivery of other similar-sized drugs.
Collapse
Affiliation(s)
| | - Bill Hakim
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Tengfei He
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
- Departments of Mechanical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
14
|
Suki B, Bates JH, Bartolák-Suki E. Remodeling of the Aged and Emphysematous Lungs: Roles of Microenvironmental Cues. Compr Physiol 2022; 12:3559-3574. [PMID: 35766835 PMCID: PMC11470990 DOI: 10.1002/cphy.c210033] [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] [Indexed: 11/06/2022]
Abstract
Aging is a slow process that affects all organs, and the lung is no exception. At the alveolar level, aging increases the airspace size with thicker and stiffer septal walls and straighter and thickened collagen and elastic fibers. This creates a microenvironment that interferes with the ability of cells in the parenchyma to maintain normal homeostasis and respond to injury. These changes also make the lung more susceptible to disease such as emphysema. Emphysema is characterized by slow but progressive remodeling of the deep alveolar regions that leads to airspace enlargement and increased but disorganized elastin and collagen deposition. This remodeling has been attributed to ongoing inflammation that involves inflammatory cells and the cytokines they produce. Cellular senescence, another consequence of aging, weakens the ability of cells to properly respond to injury, something that also occurs in emphysema. These factors conspire to make alveolar walls more prone to mechanical failure, which can set emphysema in motion by driving inflammation through immune stimulation by protein fragments. Both aging and emphysema are influenced by microenvironmental conditions such as local inflammation, chemical makeup, tissue stiffness, and mechanical stresses. Although aging and emphysema are not equivalent, they have the potential to influence each other in synergistic ways; aging sets up the conditions for emphysema to develop, while emphysema may accelerate cellular senescence and thus aging itself. This article focuses on the similarities and differences between the remodeled microenvironment of the aging and emphysematous lung, with special emphasis on the alveolar septal wall. © 2022 American Physiological Society. Compr Physiol 12:3559-3574, 2022.
Collapse
Affiliation(s)
- Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Jason H.T. Bates
- Depatment of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | | |
Collapse
|
15
|
He T, Shaw I, Vedadghavami A, Bajpayee AG. Single-Dose Intra-Cartilage Delivery of Kartogenin Using a Cationic Multi-Arm Avidin Nanocarrier Suppresses Cytokine-Induced Osteoarthritis-Related Catabolism. Cartilage 2022; 13:19476035221093072. [PMID: 35491681 PMCID: PMC9251829 DOI: 10.1177/19476035221093072] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/01/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Kartogenin (KGN) has proven as a both chondrogenic and chondroprotective drug for osteoarthritis (OA) therapy. However, being a small hydrophobic molecule, KGN suffers from rapid joint clearance and inability to penetrate cartilage to reach chondrocytes following intra-articular administration. As such multiple high doses are needed that can lead to off-target effects including stimulation and tissue outgrowth. Here we design charge-based cartilage targeting formulation of KGN by using a multi-arm cationic nano-construct of Avidin (mAv) that can rapidly penetrate into cartilage in high concentrations owing to weak-reversible electrostatic binding interactions with negatively charged aggrecan-glycosaminoglycans (GAGs) and form an extended-release drug depot such that its therapeutic benefit can be reaped in just a single dose. DESIGN We synthesized 2 novel formulations, one with a releasable ester linker (mAv-OH-KGN, release half-life ~58 h) that enables sustained KGN release over 2 weeks and another with a non-releasable amide linker (mAv-NH-KGN) that relies on mAv's ability to be uptaken and endocytosed by chondrocytes for drug delivery. Their effectiveness in suppressing cytokine-induced catabolism was evaluated in vitro using cartilage explant culture model. RESULTS A single 100 μM dose of cartilage homing mAv-KGN was significantly more effective in suppressing cytokine-induced GAG loss, cell death, inflammatory response and in rescuing cell metabolism than a single dose of free KGN; multiple doses of free KGN were needed to match this therapeutic response. CONCLUSION mAv mediated delivery of KGN is promising and can facilitate clinical translation of KGN for OA treatment with only a single dose.
Collapse
Affiliation(s)
- Tengfei He
- Department of Bioengineering,
Northeastern University, Boston, MA, USA
| | - Irfhan Shaw
- Department of Bioengineering,
Northeastern University, Boston, MA, USA
| | | | - Ambika G. Bajpayee
- Department of Bioengineering,
Northeastern University, Boston, MA, USA
- Department of Mechanical Engineering,
Northeastern University, Boston, MA, USA
| |
Collapse
|
16
|
Huo Y, Yang D, Lai K, Tu J, Zhu Y, Ding W, Yang S. Antioxidant Effects of Resveratrol in Intervertebral Disk. J INVEST SURG 2021; 35:1135-1144. [PMID: 34670455 DOI: 10.1080/08941939.2021.1988771] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Intervertebral disk (IVD) degeneration (IVDD) can cause various spinal degenerative diseases. Cumulative evidence has indicated that IVDD can result from inflammation, apoptosis, autophagy, biomechanical changes and other factors. Currently, lack of conservative treatment for degenerative spinal diseases leads to an urgent demand for clinically applicable medication to ameliorate the progression of IVDD. Resveratrol (3,5,4'-trihydroxy-trans-stilbene), a polyphenol compound extracted from red wine or grapes, has shown protective effects on IVD, alleviating the progression of IVDD. Resveratrol has been demonstrated as a scavenger of free radicals both in vivo and in vitro. The antioxidant effects of resveratrol are likely attributed to its regulation on mitochondrial dysfunction or the elimination of reactive oxygen species. This review will summarize the mechanisms of the reactive oxygen species production and elaborate the mechanisms of resveratrol in retarding IVDD progression, providing a comprehensive understanding of the antioxidant effects of resveratrol in IVD.
Collapse
Affiliation(s)
- Yachong Huo
- Department of Spine Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, PR China.,Hebei Medical University, Shijiazhuang, PR China
| | - Dalong Yang
- Department of Spine Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Kaitao Lai
- ANZAC Research Institute, The University of Sydney, Sydney, Australia
| | - Ji Tu
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Yibo Zhu
- School of Chemical Engineering, The University of Queensland, Brisbane, Australia
| | - Wenyuan Ding
- Department of Spine Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Sidong Yang
- Department of Spine Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, PR China.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| |
Collapse
|
17
|
Partain BD, Zhang Q, Unni M, Aldrich J, Rinaldi-Ramos CM, Narayanan S, Allen KD. Spatially-resolved nanometer-scale measurement of cartilage extracellular matrix mobility. Osteoarthritis Cartilage 2021; 29:1351-1361. [PMID: 34052396 PMCID: PMC8543368 DOI: 10.1016/j.joca.2021.05.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/07/2021] [Accepted: 05/19/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Tissues have complex structures, comprised of solid and fluid phases. Improved understanding of interactions between joint fluid and extracellular matrix (ECM) is required in models of cartilage mechanics. X-ray photon correlation spectroscopy (XPCS) directly measures nanometer-scale dynamics and can provide insight into biofluid-biosolid interactions in cartilage. This study applies XPCS to evaluate dynamic interactions between intact cartilage and biofluids. DESIGN Cartilage biopsies were collected from bovine femoral condyles. During XPCS measurements, cartilage samples were exposed to different fluids: deionized water, PBS, synovial fluid, or sonicated synovial fluid. ECM-biofluid interactions were also assessed at different length scales and different depths from the cartilage surface. RESULTS Using XPCS, cartilage ECM mobility was detected at length scales from 50 to 207 nm. As length scale decreased, time scale for autocorrelation decay decreased, suggesting smaller ECM components are more mobile. ECM dynamics were slowed by dehydrating the sample, demonstrating XPCS assesses matrix mobility in hydrated environments. At all length scales, the matrix was more mobile in deionized water and slowest in synovial fluid. Using the 207 nm length scale assessment, ECM dynamics in synovial fluid were fastest at the cartilage surface and progressively slowed as depth into the sample increased, demonstrating XPCS can assess spatial distribution of ECM dynamics. Finally, ECM mobility increased for degraded synovial fluid. CONCLUSIONS This study demonstrates the potential of XPCS to provide unique insights into nanometer-scale cartilage ECM mobility in a spatially resolved manner and illustrates the importance of biosolid-biofluid interactions in dictating ECM dynamics.
Collapse
Affiliation(s)
- B D Partain
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Q Zhang
- X-ray Science Division, Argonne National Laboratory, Lemont, IL, USA
| | - M Unni
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - J Aldrich
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - C M Rinaldi-Ramos
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - S Narayanan
- X-ray Science Division, Argonne National Laboratory, Lemont, IL, USA
| | - K D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Orthopaedics and Rehabilitation, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
18
|
Li R, Fang H, Shen J, Jin Y, Zhao Y, Wang R, Fu Y, Tian Y, Yu H, Zhang J. Curcumin Alleviates LPS-Induced Oxidative Stress, Inflammation and Apoptosis in Bovine Mammary Epithelial Cells via the NFE2L2 Signaling Pathway. Toxins (Basel) 2021; 13:208. [PMID: 33809242 PMCID: PMC7999830 DOI: 10.3390/toxins13030208] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Lipopolysaccharide (LPS) is an endotoxin, which may cause immune response and inflammation of bovine mammary glands. Mastitis impairs animal health and results in economic loss. Curcumin (CUR) is a naturally occurring diketone compound, which has attracted widespread attention as a potential anti-inflammatory antioxidant. The purpose of this study is to investigate whether CUR can reduce the damage of bovine mammary epithelial cells (MAC-T) induced by LPS and its underlying molecular mechanism. The MAC-T cell line was treated with different concentrations of LPS and CUR for 24 h. The results showed that CUR rescued the decrease of MAC-T cell viability and cell damage induced by LPS. At the same time, 10 µM CUR and 100 µg/mL LPS were used to treat the cells in the follow-up study. The results showed CUR treatment reduced the accumulation of reactive oxygen species (ROS), the expression of inflammatory cytokines (tumor necrosis factor-a (TNF-α), interleukin-8 (IL-8), IL-6 and IL-1β) and the rate of apoptosis induced by LPS. These effects were associated with the activation of the nuclear factor E2-related factor 2 (NFE2L2)-antioxidant response element (ARE) pathway coupled with inactivation of the nuclear factor-κB (NF-κB) inflammatory and caspase/Bcl2 apoptotic pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hao Yu
- Key Laboratory of Zoonosis Research, Ministry of Education, Department of Animal Science, College of Animal Sciences, Jilin University, Changchun 130062, China; (R.L.); (H.F.); (J.S.); (Y.J.); (Y.Z.); (R.W.); (Y.F.); (Y.T.)
| | - Jing Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, Department of Animal Science, College of Animal Sciences, Jilin University, Changchun 130062, China; (R.L.); (H.F.); (J.S.); (Y.J.); (Y.Z.); (R.W.); (Y.F.); (Y.T.)
| |
Collapse
|