1
|
Wang SW, Li P, Liu SY, Huang DL, Zhang SJ, Zeng XX, Lan T, Mao KL, Gao Y, Cheng YF, Shen Q, Ruan YP, Mao ZJ. Astragaloside IV inhibits retinal pigment epithelial cell senescence and reduces IL-1β mRNA stability by targeting FTO-mediated m 6A methylation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156408. [PMID: 39848020 DOI: 10.1016/j.phymed.2025.156408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Resistance to senescence in retinal pigment epithelial (RPE) cells can delay the progression of age-related macular degeneration (AMD). However, the mechanisms underlying RPE cell senescence remain inadequately understood, and effective therapeutic strategies are lacking. While astragaloside IV (Ast) has demonstrated anti-aging properties, its specific effects on RPE cell senescence and potential mechanisms are not yet fully clarified. PURPOSE This study aimed to explore the impacts of Ast on RPE cell senescence and to uncover the molecular mechanisms involved. METHODS The therapeutic efficacy of Ast was assessed using sodium iodate (NaIO3)-induced adult retinal pigment epithelial cell line 19 (ARPE-19) cell models and an AMD mouse model. To investigate the mechanisms by which Ast mitigated RPE cell senescence, RNA sequencing (RNA-seq), drug affinity responsive target stability-mass spectrometry (DARTS-MS), cellular thermal shift assay (CETSA), reverse transcription quantitative PCR (RT-qPCR), as well as western blotting were conducted. RESULTS Ast significantly inhibited NaIO3-treated ARPE-19 cell senescence and protected against NaIO3-induced AMD in mice. RNA-seq analysis revealed that Ast significantly attenuated inflammation-related signaling pathways and reduced the mRNA levels of interleukin-1 beta (IL-1β). Specifically, Ast decreased the stability of IL-1β mRNA while enhancing its N6-methyladenosine (m6A) methylation. Furthermore, Ast directly interacted with fat mass and obesity-associated protein (FTO). Knockdown or pharmacological inhibition of FTO mitigated the senescence and IL-1β expression in NaIO3-treated ARPE-19 cells. FTO was essential for Ast to inhibit cellular senescence and IL-1β expression. Additionally, inhibition or knockdown of FTO conferred also provided resistance to AMD in the murine model. CONCLUSION Our results indicated that Ast significantly attenuated RPE cell senescence and showed anti-AMD properties. FTO was demonstrated to be a promising therapeutic target for AMD treatment. These findings may provide a deeper understanding of the molecular mechanisms underlying RPE cell senescence in AMD and offer potential strategies for its prevention and management.
Collapse
Affiliation(s)
- Si-Wei Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| | - Ping Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shi-Yu Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - De-Lian Huang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Si-Jia Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xi-Xi Zeng
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Tian Lan
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Kai-Li Mao
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Yuan Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi-Fan Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qing Shen
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Ye-Ping Ruan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Zhu-Jun Mao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Ophthalmology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| |
Collapse
|
2
|
Xu W, Wang J, Cui L, Huang C, Xia N, Xie M, Liu D, Liao D. Il-1β Promotes Superficial Zone Cells Senescence in Articular Cartilage by Inhibiting Autophagy. Cartilage 2024; 15:428-439. [PMID: 37650417 PMCID: PMC11523166 DOI: 10.1177/19476035231194771] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/15/2023] [Accepted: 07/29/2023] [Indexed: 09/01/2023] Open
Abstract
OBJECTIVE The superficial zone cells in articular cartilage (SFZCs) have been identified as stem/progenitor chondrocytes and promoted cell self-renewal in the osteoarthritis (OA). Several studies emphasized the involvement of senescence and autophagy in OA. Interleukin-1β (IL-1β) is one of the main inflammatory mediators of OA, and whether it induces senescence and autophagy in SFZCs remains unclear. The present study aimed to investigate autophagy flux, mitochondrial function, and intracellular reactive oxygen species (ROS) that resulted in senescence in SFZCs induced by IL-1β. METHODS Using western blotting, reverse transcription-quantitative PCR, immunofluorescence, intracellular ROS detection, mitochondrial staining, and determination of mitochondrial membrane potential, we tested senescence and autophagy markers in SFZCs induced by IL-1β in vitro. The consequences of mitochondrial function and ROS were also studied with IL-1β-induced senescence. RESULTS IL-1β treatment decreased SFZC proliferation, induced SFZC senescence, and reduced SFZCs' chondrogenic differentiation capacity. Moreover, IL-1β impaired autophagy flux, and the autophagy activator, rapamycin, attenuated the senescence of SFZCs. IL-1β-induced autophagy defect resulted in mitochondrial dysfunction and overproduction of ROS, and autophagy activation notably protected against mitochondrial dysfunction and reduced the levels of ROS. Moreover, antioxidant N-acetylcysteine reversed the senescence of IL-1β in SFZCs. CONCLUSION IL-1β promotes autophagy impairment and subsequently results in dysfunctional mitochondria and overproduction of ROS, which finally causes SFZC senescence.
Collapse
Affiliation(s)
- Wei Xu
- Trauma Center, General Hospital of Western Theater Command, People’s Liberation Army, Chengdu, China
| | - Juan Wang
- Department of Pain Treatment, General Hospital of Western Theater Command, People’s Liberation Army, Chengdu, China
| | - Lin Cui
- Trauma Center, General Hospital of Western Theater Command, People’s Liberation Army, Chengdu, China
| | - Chen Huang
- Trauma Center, General Hospital of Western Theater Command, People’s Liberation Army, Chengdu, China
| | - Ning Xia
- Department of Orthopedics, General Hospital of Western Theater Command, People’s Liberation Army, Chengdu, China
| | - Meiming Xie
- Trauma Center, General Hospital of Western Theater Command, People’s Liberation Army, Chengdu, China
| | - Da Liu
- Department of Orthopedics, General Hospital of Western Theater Command, People’s Liberation Army, Chengdu, China
| | - Dongfa Liao
- Trauma Center, General Hospital of Western Theater Command, People’s Liberation Army, Chengdu, China
| |
Collapse
|
3
|
Boulestreau J, Maumus M, Bertolino Minani G, Jorgensen C, Noël D. Anti-aging effect of extracellular vesicles from mesenchymal stromal cells on senescence-induced chondrocytes in osteoarthritis. Aging (Albany NY) 2024; 16:13252-13270. [PMID: 39578049 PMCID: PMC11719114 DOI: 10.18632/aging.206158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Age is the most important risk factor for degenerative diseases such as osteoarthritis (OA). It is associated with the accumulation of senescent cells in joint tissues that contribute to the pathogenesis of OA, in particular through the release of senescence-associated secretory phenotype (SASP) factors. Mesenchymal stromal cells (MSCs) and their derived extracellular vesicles (EVs) are promising treatments for OA. However, the senoprotective effects of MSC-derived EVs in OA have been poorly investigated. Here, we used EVs from human adipose tissue-derived MSCs (ASC-EVs) in two models of inflammaging (IL1β)- and DNA damage (etoposide)-induced senescence in OA chondrocytes. We showed that the addition of ASC-EVs was effective in reducing senescence parameters, including the number of SA-β-Gal-positive cells, the accumulation of γH2AX foci in nuclei and the secretion of SASP factors. In addition, ASC-EVs demonstrated therapeutic efficacy when injected into a murine model of OA. Several markers of senescence, inflammation and oxidative stress were decreased shortly after injection likely explaining the therapeutic efficacy. In conclusion, ASC-EVs exert a senoprotective function both in vitro, in two models of induced senescence in OA chondrocytes and, in vivo, in the murine model of collagenase-induced OA.
Collapse
Affiliation(s)
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | | | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France
- Department of Rheumatology, Clinical Immunology and Osteoarticular Disease Therapeutic Unit, CHU de Montpellier, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France
- Department of Rheumatology, Clinical Immunology and Osteoarticular Disease Therapeutic Unit, CHU de Montpellier, Montpellier, France
| |
Collapse
|
4
|
Shen L, Zeng X, Zhang H. The protective effects of orexin-A in alleviating cell senescence against interleukin-1β (IL-1β) in chondrocytes. Aging (Albany NY) 2024; 16:9558-9568. [PMID: 38829778 PMCID: PMC11210258 DOI: 10.18632/aging.205884] [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: 09/22/2023] [Accepted: 03/03/2024] [Indexed: 06/05/2024]
Abstract
Osteoarthritis (OA) is one of the most important causes of global disability, and dysfunction of chondrocytes is an important risk factor. The treatment of OA is still a challenge. Orexin-A is a hypothalamic peptide, and its effects in OA are unknown. In this study, we found that exposure to interleukin-1β (IL-1β) reduced the expression of orexin-2R, the receptor of orexin-A in TC-28a2 chondrocytes. Importantly, the senescence-associated β-galactosidase (SA-β-gal) staining assay demonstrated that orexin-A treatment ameliorates IL-1β-induced cellular senescence. Importantly, the presence of IL-1β significantly reduced the telomerase activity of TC-28a2 chondrocytes, which was rescued by orexin-A. We also found that orexin-A prevented IL-1β-induced increase in the levels of Acetyl-p53 and the expression of p21. It is shown that orexin-A mitigates IL-1β-induced reduction of sirtuin 3 (SIRT3). Silencing of SIRT3 abolished the protective effects of orexin-A against IL-1β-induced cellular senescence. These results imply that orexin-A might serve as a promising therapeutic agent for OA.
Collapse
Affiliation(s)
- Lin Shen
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, China
| | - Xiantie Zeng
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, China
| | - Haiying Zhang
- Department of Orthopedics, Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100078, China
| |
Collapse
|
5
|
Liu Y, Yao J, Deng G, Zhong G, Zhao J, Lan Q, Meng J, Yu Y, Chen F. Microgel Encapsulated Nanoparticles for Intra-articular Disulfiram Delivery to Treat Osteoarthritis. Mol Pharm 2024; 21:87-101. [PMID: 38100656 DOI: 10.1021/acs.molpharmaceut.3c00462] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Osteoarthritis (OA) affects numerous patients worldwide, and there are no approved disease-modifying drugs. Repurposing FDA-approved small molecular drugs could be a promising alternative strategy to treat OA. Disulfiram (DSF), a clinically approved drug for treatment of alcoholism, inhibits inflammasome activation and exhibits a protective role in interleukin-1β-induced cardiac injury. However, its efficacy in treating OA remains to be explored due to its poor water solubility and stability, which limit its use in OA treatment. Here, the anti-inflammatory effect of DSF is evaluated in vitro, and a double-layer encapsulation approach is developed for intra-articular delivery of DSF for OA treatment in vivo. DSF is loaded into poly(lactic-co-glycolic acid)-based nanoparticles and encapsulated in gelatin methacrylate microgels through a microfluidic device. Results show that DSF effectively inhibits the expression of key inflammatory cytokines in OA chondrocytes, and the double-layer encapsulation approach reduces the burst release of DSF and prolongs its retention time in the in vitro study. Sustained release of DSF from microgels mitigates cartilage inflammation and subchondral bone erosion in a monoiodoacetate-induced rat OA model. This work demonstrates the potential of repurposing FDA-approved drugs for OA treatment and provides a promising platform for intra-articular delivery of small molecules for superior therapeutic effect.
Collapse
Affiliation(s)
- Yisi Liu
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jun Yao
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Guotao Deng
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Gang Zhong
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jianping Zhao
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qiumei Lan
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jinzhi Meng
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yin Yu
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fei Chen
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
6
|
Che J, Yang X, Jin Z, Xu C. Nrf2: A promising therapeutic target in bone-related diseases. Biomed Pharmacother 2023; 168:115748. [PMID: 37865995 DOI: 10.1016/j.biopha.2023.115748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
Nuclear factor erythroid-2-related factor 2 (Nrf2) plays an important role in maintaining cellular homeostasis, as it suppresses cell damage caused by external stimuli by regulating the transcription of intracellular defense-related genes. Accumulating evidence has highlighted the crucial role of reduction-oxidation (REDOX) imbalance in the development of bone-related diseases. Nrf2, a transcription factor linked to nuclear factor-erythrocyte 2, plays a pivotal role in the regulation of oxidative stress and induction of antioxidant defenses. Therefore, further investigation of the mechanism and function of Nrf2 in bone-related diseases is essential. Considerable evidence suggests that increased nuclear transcription of Nrf2 in response to external stimuli promotes the expression of intracellular antioxidant-related genes, which in turn leads to the inhibition of bone remodeling imbalance, improved fracture recovery, reduced occurrence of osteoarthritis, and greater tumor resistance. Certain natural extracts can selectively target Nrf2, potentially offering therapeutic benefits for osteogenic arthropathy. In this article, the biological characteristics of Nrf2 are reviewed, the intricate interplay between Nrf2-regulated REDOX imbalance and bone-related diseases is explored, and the potential preventive and protective effects of natural products targeting Nrf2 in these diseases are elucidated. A comprehensive understanding of the role of Nrf2 in the development of bone-related diseases provides valuable insights into clinical interventions and can facilitate the discovery of novel Nrf2-targeting drugs.
Collapse
Affiliation(s)
- Jingmin Che
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Xiaoli Yang
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Zhankui Jin
- Department of Orthopedics, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Cuixiang Xu
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
7
|
Marra PS, Nishizawa Y, Yamanashi T, Sullivan EJ, Comp KR, Crutchley KJ, Wahba NE, Shibata K, Nishiguchi T, Yamanishi K, Noiseux NO, Karam MD, Shinozaki G. NSAIDs use history: impact on the genome-wide DNA methylation profile and possible mechanisms of action. Clin Exp Med 2023; 23:3509-3516. [PMID: 37341931 DOI: 10.1007/s10238-023-01119-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/11/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND AND OBJECTIVE NSAIDs inhibit cyclooxygenase, but their role in aging and other diseases is not well understood. Our group previously showed the potential benefit of NSAIDs in decreasing the risk of delirium and mortality. Concurrently, epigenetics signals have also been associated with delirium. Therefore, we sought to find differentially methylated genes and biological pathways related to exposure with NSAIDs by comparing the genome-wide DNA methylation profiles of patients with and without a history of NSAIDs use. METHODS Whole blood samples were collected from 171 patients at the University of Iowa Hospital and Clinics from November 2017 to March 2020. History of NSAIDs use was assessed through a word-search function in the subjects' electronic medical records. DNA was extracted from the blood samples, processed with bisulfite conversion, and analyzed using Illumina's EPIC array. The analysis of top differentially methylated CpG sites and subsequent enrichment analysis were conducted using an established pipeline using R statistical software. RESULTS Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) showed several biological pathways relevant to NSAIDs' function. The identified GO terms included "arachidonic acid metabolic process," while KEGG results included "linoleic acid metabolism," "cellular senescence," and "circadian rhythm." Nonetheless, none of the top GO and KEGG pathways and the top differentially methylated CpG sites reached statistical significance. CONCLUSION Our results suggest a potential role of epigenetics in the mechanisms of the action of NSAIDs. However, the results should be viewed with caution as exploratory and hypothesis-generating given the lack of statistically significant findings.
Collapse
Affiliation(s)
- Pedro S Marra
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 3165 Porter Dr. Room 2175, Palo Alto, CA, 94304, USA
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Yoshitaka Nishizawa
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 3165 Porter Dr. Room 2175, Palo Alto, CA, 94304, USA
- Department of Neuropsychiatry, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Takehiko Yamanashi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 3165 Porter Dr. Room 2175, Palo Alto, CA, 94304, USA
- Department of Neuropsychiatry, Faculty of Medicine, Tottori University, Yonago-Shi, Tottori, Japan
| | - Eleanor J Sullivan
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Katie R Comp
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | - Nadia E Wahba
- Department of Psychiatry, Oregon Health and Science University School of Medicine, Portland, OR, USA
| | - Kazuki Shibata
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 3165 Porter Dr. Room 2175, Palo Alto, CA, 94304, USA
- Sumitomo Pharma Co. Ltd, Osaka, Osaka, Japan
| | - Tsuyoshi Nishiguchi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 3165 Porter Dr. Room 2175, Palo Alto, CA, 94304, USA
- Department of Neuropsychiatry, Faculty of Medicine, Tottori University, Yonago-Shi, Tottori, Japan
| | - Kyosuke Yamanishi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 3165 Porter Dr. Room 2175, Palo Alto, CA, 94304, USA
- Department of Neuropsychiatry, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Nicolas O Noiseux
- Department of Orthopedic Surgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Matthew D Karam
- Department of Orthopedic Surgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Gen Shinozaki
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 3165 Porter Dr. Room 2175, Palo Alto, CA, 94304, USA.
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
8
|
Arra M, Abu-Amer Y. Cross-talk of inflammation and chondrocyte intracellular metabolism in osteoarthritis. Osteoarthritis Cartilage 2023; 31:1012-1021. [PMID: 37094761 DOI: 10.1016/j.joca.2023.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
Osteoarthritis is a disease that impacts millions around the world, leading to significant financial and medical burden for patients and the healthcare system. However, no effective biomarkers or disease modifying therapeutics exist for the early identification and management of the disease. Inflammation drives chondrocytes to express extracellular matrix (ECM) degrading enzymes and interruption of this pathway is a viable target to prevent degradation of cartilage. It has been demonstrated that inflammation can alter the intracellular metabolism of chondrocytes, a process known as metabolic reprogramming. This metabolic reprogramming is critical for cartilage breakdown by shifting chondrocytes to an ECM-catabolic state and likely as a potential therapeutic target for osteoarthritis. Metabolic modulators hold the potential to reduce chondrocyte inflammatory responses and protect cartilage. In this narrative review, we explore some of the existing examples of interactions between metabolism and inflammatory pathways in chondrocytes. We summarize the impact of inflammatory stimulation on various metabolic pathways and describe several examples by which targeting of metabolism is able to modulate ECM-degrading activity of chondrocytes to protect against cartilage damage.
Collapse
Affiliation(s)
- M Arra
- Department of Orthopedic Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Y Abu-Amer
- Department of Orthopedic Surgery, Washington University School of Medicine, Saint Louis, MO, USA; Department of Cell Biology & Physiology, Washington University School of Medicine, Saint Louis, MO, USA; Shriners Hospital for Children, Saint Louis, MO, USA.
| |
Collapse
|
9
|
Briones-Suarez L, Cifuentes M, Bravo-Sagua R. Secretory Factors from Calcium-Sensing Receptor-Activated SW872 Pre-Adipocytes Induce Cellular Senescence and A Mitochondrial Fragmentation-Mediated Inflammatory Response in HepG2 Cells. Int J Mol Sci 2023; 24:ijms24065217. [PMID: 36982291 PMCID: PMC10049719 DOI: 10.3390/ijms24065217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 03/11/2023] Open
Abstract
Adipose tissue inflammation in obesity has a deleterious impact on organs such as the liver, ultimately leading to their dysfunction. We have previously shown that activation of the calcium-sensing receptor (CaSR) in pre-adipocytes induces TNF-α and IL-1β expression and secretion; however, it is unknown whether these factors promote hepatocyte alterations, particularly promoting cell senescence and/or mitochondrial dysfunction. We generated conditioned medium (CM) from the pre-adipocyte cell line SW872 treated with either vehicle (CMveh) or the CaSR activator cinacalcet 2 µM (CMcin), in the absence or presence of the CaSR inhibitor calhex 231 10 µM (CMcin+cal). HepG2 cells were cultured with these CM for 120 h and then assessed for cell senescence and mitochondrial dysfunction. CMcin-treated cells showed increased SA-β-GAL staining, which was absent in TNF-α- and IL-1β-depleted CM. Compared to CMveh, CMcin arrested cell cycle, increased IL-1β and CCL2 mRNA, and induced p16 and p53 senescence markers, which was prevented by CMcin+cal. Crucial proteins for mitochondrial function, PGC-1α and OPA1, were decreased with CMcin treatment, concomitant with fragmentation of the mitochondrial network and decreased mitochondrial transmembrane potential. We conclude that pro-inflammatory cytokines TNF-α and IL-1β secreted by SW872 cells after CaSR activation promote cell senescence and mitochondrial dysfunction, which is mediated by mitochondrial fragmentation in HepG2 cells and whose effects were reversed with Mdivi-1. This investigation provides new evidence about the deleterious CaSR-induced communication between pre-adipocytes and liver cells, incorporating the mechanisms involved in cellular senescence.
Collapse
Affiliation(s)
- Lautaro Briones-Suarez
- Laboratory of Obesity and Metabolism (OMEGA), Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
- Department of Nutrition and Public Health, Faculty of Health and Food Sciences, University of Bío-Bío, Chillán 3800708, Chile
| | - Mariana Cifuentes
- Laboratory of Obesity and Metabolism (OMEGA), Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
- Center for Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
- Correspondence: (M.C.); (R.B.-S.); Tel.: +56-229781428 (M.C.); +56-229781563 (R.B.-S.)
| | - Roberto Bravo-Sagua
- Laboratory of Obesity and Metabolism (OMEGA), Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
- Interuniversity Center for Healthy Aging (CIES), Consortium of Universities of the State of Chile (CUECH), Santiago 8320216, Chile
- Correspondence: (M.C.); (R.B.-S.); Tel.: +56-229781428 (M.C.); +56-229781563 (R.B.-S.)
| |
Collapse
|
10
|
Zhuang H, Ren X, Jiang F, Zhou P. Indole-3-propionic acid alleviates chondrocytes inflammation and osteoarthritis via the AhR/NF-κB axis. Mol Med 2023; 29:17. [PMID: 36721094 PMCID: PMC9890697 DOI: 10.1186/s10020-023-00614-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/23/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common chronic disease characterized by chronic inflammation and extracellular matrix degradation. Indole-3-propionic acid (IPA) is a tryptophan metabolite secreted by intestinal flora, which can exert anti-inflammatory effects in a variety of diseases. In this study, we further investigated the potential therapeutic role of IPA in OA and the underlying mechanism. METHODS IL-1β was utilized to induce chondrocyte inflammation. Then, the cytotoxicity of IPA on rat chondrocytes was assessed. Meanwhile, RT-qPCR, Griess reaction, ELISA, Western blot and immunofluorescence were performed to evaluate the expression of inflammatory factors and stromal proteins, and the NF-κB pathway in chondrocytes treated with IL-1β alone, with IPA or with aryl hydrocarbon receptor (AhR) knockdown. An OA rat model was established by anterior cruciate ligament transection, and hematoxylin-eosin staining, Safranin-O/Fast Green staining and immunochemistry were applied to estimate OA severity. RESULTS IPA did not affect cellular viability at concentrations up to 80 µM. IPA significantly inhibited the IL-1β-induced expression of inflammatory factors (Nitric oxide, PGE2, TNF-α, IL-6, iNOS and COX-2) and matrix-degrading enzymes (MMP-3, MMP-13 and ADAMTS-5), upregulated the expression of anabolic markers (aggrecan and collagen-II) and inactivated the NF-κB pathway. However, AhR knockdown could abolish the above protection capabilities and the suppression of the NF-κB pathway induced by IPA. Furthermore, IPA significantly reduced serum inflammatory cytokines expression, cartilage destruction and synovitis in vivo, demonstrating its protective role in OA progression. CONCLUSION IPA improved IL-1β-induced chondrocyte inflammation and extracellular matrix degradation through the AhR/NF-κB axis, which provides an innovative therapeutic strategy for OA.
Collapse
Affiliation(s)
- Huangming Zhuang
- grid.412632.00000 0004 1758 2270Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Xunshan Ren
- grid.412632.00000 0004 1758 2270Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Fuze Jiang
- grid.412632.00000 0004 1758 2270Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Panghu Zhou
- grid.412632.00000 0004 1758 2270Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| |
Collapse
|
11
|
Li L, Liu T, Zhang X, Hou X, Dong H, Li X, Ren W, Wang Y. Catalyst-free and atom-economical 1,3-dipolar cycloaddition of C,N-cyclic azomethine imines: Facile synthesis of isoquinoline-fused spirocycles. GREEN SYNTHESIS AND CATALYSIS 2021. [DOI: 10.1016/j.gresc.2021.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|