1
|
Lin M, Wang Y, Zhao Y, Zhai X, Hu Y, Luan Q, Lv L, Wang Z, Yao J. Protective effects of albiflorin on acetaminophen-induced hepatotoxicity: Regulation of blood-biliary barrier integrity by ATF3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156722. [PMID: 40250002 DOI: 10.1016/j.phymed.2025.156722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/19/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Acetaminophen-induced hepatotoxicity remains a clinical challenge with limited targeted therapeutic options. While recent advances have identified blood‒biliary barrier disruption as a critical pathogenic mechanism, effective interventions preserving this barrier are notably lacking. Albiflorin, a key bioactive constituent of Paeonia lactiflora Pall., exhibits unique bile acid modulation along with antioxidant and anti-inflammatory activities, suggesting its potential efficacy in preventing acetaminophen-induced liver injury. However, its specific role and underlying mechanisms in alleviating acetaminophen-induced hepatotoxicity remain unclear. OBJECTIVE This study's objective was to examine the pharmacological effects and primary molecular mechanisms of albiflorin in alleviating acetaminophen-induced liver injury. METHODS An acetaminophen-induced liver injury mouse model was created using a 300 mg/kg dose of acetaminophen. The hepatoprotective effects of albiflorin were assessed through histological and biochemical analyses. Blood‒biliary barrier integrity was evaluated via Evans blue dye tests, immunofluorescence, and bile acid assays. Transcriptomic analysis, gene overexpression and interference techniques, and ChIP‒qPCR were employed to explore the molecular mechanisms underlying the protective effects of albiflorin. RESULTS Albiflorin significantly reduced acetaminophen-induced liver injury, as evidenced by improved biochemical profiles and hepatocyte morphology. It also prevented increases in blood‒biliary barrier permeability and bile acid levels. RNA sequencing identified 3,184 differentially expressed genes, revealing critical pathways involved in maintaining blood‒biliary barrier integrity. AF reversed the acetaminophen-induced changes in the expression of genes related to the blood‒biliary barrier, particularly occludin, claudin 5, ABCG5, and ABCG8. Albiflorin protected the blood‒biliary barrier and mitigated acetaminophen-induced liver injury by enhancing ATF3 protein stability, with ATF3 identified as a critical mediator of these protective effects. CONCLUSION This study provides pioneering evidence that albiflorin protects against acetaminophen-induced liver injury by interacting with ATF3 to regulate blood-biliary barrier proteins and maintain the integrity of the blood-biliary barrier. These findings deepen our understanding of the role of the blood‒biliary barrier in liver diseases and suggest a therapeutic strategy for drug overdose-induced liver injury.
Collapse
Affiliation(s)
- Musen Lin
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China; Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Xiaohan Zhai
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yan Hu
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Qinrong Luan
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Li Lv
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China.
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China.
| |
Collapse
|
2
|
Lv S, Luo C. Ferroptosis in schizophrenia: Mechanisms and therapeutic potentials (Review). Mol Med Rep 2025; 31:37. [PMID: 39611491 PMCID: PMC11613623 DOI: 10.3892/mmr.2024.13402] [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: 09/07/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Schizophrenia, a complex psychiatric disorder, presents with multifaceted symptoms and important challenges in treatment, primarily due to its pathophysiological complexity, which involves oxidative stress and aberrant iron metabolism. Recent insights into ferroptosis, a unique form of iron‑dependent cell death characterized by lipid peroxidation and antioxidant system failures, open new avenues for understanding the neurobiological foundation of schizophrenia. The present review explores the interplay between ferroptosis and schizophrenia, emphasizing the potential contributions of disrupted iron homeostasis and oxidative mechanisms to the pathology and progression of this disease. The emerging evidence linking ferroptosis with the oxidative stress observed in schizophrenia provides a compelling narrative for re‑evaluating current therapeutic strategies and exploring novel interventions targeting these molecular pathways, such as the glutathione peroxidase 4 pathway and the ferroptosis suppressor protein 1 pathway. By integrating recent advances in ferroptosis research, the current review highlights innovative therapeutic potentials, including N‑acetylcysteine, selenium, omega‑3 fatty acids and iron chelation therapy, which could address the limitations of existing treatments and improve clinical outcomes for individuals with schizophrenia.
Collapse
Affiliation(s)
- Shuang Lv
- Department of Psychiatry, Guangzhou Kangning Hospital (The Psychiatric Hospital of Guangzhou Civil Administration Bureau), Guangzhou, Guangdong 510430, P.R. China
| | - Chunxia Luo
- Department of Psychiatry, Guangzhou Kangning Hospital (The Psychiatric Hospital of Guangzhou Civil Administration Bureau), Guangzhou, Guangdong 510430, P.R. China
| |
Collapse
|
3
|
Lu H, Zheng Y, Wang D. ATF3 affects osteogenic differentiation in inflammatory hPDLSCs by mediating ferroptosis via regulating the Nrf2/HO-1 signaling pathway. Tissue Cell 2024; 89:102447. [PMID: 38991270 DOI: 10.1016/j.tice.2024.102447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024]
Abstract
Activating transcription factor 3 (ATF3) has been identified as a regulator associated with osteoblast differentiation. However, the effects of ATF3 on the osteogenic differentiation and proliferation of human periodontal stem cells (hPDLSCs) in periodontitis have not been reported. With the purpose of establishing an in vitro model of periodontitis, hPDLSCs were challenged with lipopolysaccharide (LPS). The Cell Counting Kit-8 assay was applied to assess cell viability, while reverse transcription-quantitative PCR and western blotting were employed to detect ATF3 expression. Inflammatory release was assessed using ELISA, together with western blotting. Lipid peroxidation was explored using the C11 BODIPY 581/591 probe, biochemical kits, thiobarbituric acid reactive substances (TBARS) assay and DCFH-DA staining. Iron and Fe2+ levels, and the expression levels of ferroptosis-related proteins were measured using corresponding kits and western blotting. Osteogenic differentiative capability was evaluated using alkaline phosphatase staining, Alizarin red staining and western blotting. The expression levels of proteins associated with Nrf2/HO-1 signaling were identified using western blotting. The results indicated that ATF3 expression was upregulated in LPS-induced hPDLSCs. The knockdown of ATF3 alleviated the LPS-induced inflammatory response in hPDLSCs, together with increased levels of TNF-α, IL-6, IL-1β, Cox-2 and iNOS, and decreased levels of IL-10. ATF3 silencing also led to lower TBARS production rate, and reduced levels of reactive oxygen species, iron, Fe2+, ACSL4 and TFR1, whereas it elevated the levels of SLC7A11 and GPX4. In addition, ATF3 silencing promoted hPDLSC mineralization and cell differentiation, and elevated the levels of OCN2, RUNX2 and BMP2. Additionally, ATF3 depletion upregulated the expression levels of proteins related with Nrf2/HO-1 signaling. The Nrf2 inhibitor ML385 partially counteracted the effects of ATF3 interference on the LPS-challenged inflammatory response, lipid peroxidation, ferroptosis as well as osteogenic differentiative capability in hPDLSCs. In summary, the results revealed that ATF3 silencing suppressed inflammation and ferroptosis, while it improved osteogenic differentiation in LPS-induced hPDLSCs by regulating Nrf2/HO-1 signaling, which may provide promising therapeutic targets for the treatment of periodontitis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pediatric Dentistry, Stomatological Hospital of Guizhou Medical University, Guiyang 550004, China.
| | - Yuemei Zheng
- School of Stomatology, Guizhou Medical University, Guiyang 550004, China
| | - Dan Wang
- School of Stomatology, Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
4
|
Alqasoumi A, Alsharidah M, Mahmood A, Elsafadi M, Al Rugaie O, Mohany KM, Al-Regaiey KA, Alyahya KI, Alanteet AA, Algarzae NK, AlGhibiwi HK, AlHomaidi A, Abumaree M. Mesenchymal Stem Cell-Conditioned Media Modulate HUVEC Response to H 2O 2: Impact on Gene Expression and Potential for Atherosclerosis Intervention. BIOMED RESEARCH INTERNATIONAL 2024; 2024:7726493. [PMID: 39050921 PMCID: PMC11268959 DOI: 10.1155/2024/7726493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
Background: We studied the potential of human bone marrow-derived mesenchymal stem cell conditioned media (hBMSC CM) in protecting endothelial cell properties (viability, proliferation, and migrations) from the deleterious effects produced by the inflammatory environment of H2O2. Additionally, we investigated their impact on the endothelial cells' gene expression of some inflammatory-related genes, namely, TGF-β1, FOS, ATF3, RAF-1, and SMAD3. Methods: Human umbilical vein endothelial cells (HUVECs) were cultured individually under three conditions: alone, with varying concentrations of H2O2, or with varying concentrations of H2O2 and hBMSC CM. HUVEC adhesion, proliferation, and migration were evaluated using the xCELLigence system. The HUVECs' gene expressions were evaluated by real-time polymerase chain reaction (RT-PCR). Results: Generally, we observed enhanced HUVEC viability, proliferation, and migration when cultured in media supplemented with H2O2 and hBMSC CM. Furthermore, the CM modulated the expressions of the studied inflammatory-related genes in HUVECs, promoting a more robust cellular response. Conclusion: This study has illuminated the protective role of hBMSC CM in mitigating the damaging effects of H2O2 on endothelial cell function. Our data demonstrate that hBMSC CM enhances the viability, proliferation, and migration of HUVECs even under oxidative stress conditions. Additionally, the conditioned medium was found to modulate the gene expression of pivotal markers related to inflammation, suggesting a favorable influence on cellular response mechanisms.
Collapse
Affiliation(s)
- Abdulmajeed Alqasoumi
- Department of Pharmacy PracticeCollege of PharmacyQassim University, Qassim, Saudi Arabia
| | - Mansour Alsharidah
- Department of PhysiologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Amer Mahmood
- Stem Cell Unit Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Mona Elsafadi
- Stem Cell Unit Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Osamah Al Rugaie
- Department of Biology and ImmunologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Khalid M. Mohany
- Department of Medical BiochemistryFaculty of MedicineAssiut University, El Gamma Street, Assiut City 71515, Egypt
| | - Khalid A. Al-Regaiey
- Department of PhysiologyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Khaleel I. Alyahya
- Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Alaa A. Alanteet
- Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud University, Riyadh, Saudi Arabia
| | - Norah K. Algarzae
- Department of PhysiologyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Hanan K. AlGhibiwi
- Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud University, Riyadh, Saudi Arabia
| | - Adel AlHomaidi
- Department of PathologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Mohammad Abumaree
- Stem Cells and Regenerative MedicineCell Therapy and Cancer Research (CTCR)King Abdullah International Medical Research Center (KAIMRC)King Saud Bin Abdulaziz University for Health Sciences (KSAU)King Abdulaziz Medical CityMinistry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Wen Y, Liu Y, Li Q, Tan J, Fu X, Liang Y, Tuo Y, Liu L, Zhou X, LiuFu D, Fan X, Chen C, Chen Z, Wang Z, Fan S, Liu R, Pan L, Zhang Y, Tang WH. Spatiotemporal ATF3 Expression Determines VSMC Fate in Abdominal Aortic Aneurysm. Circ Res 2024; 134:1495-1511. [PMID: 38686580 DOI: 10.1161/circresaha.124.324323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a catastrophic disease with little effective therapy, likely due to the limited understanding of the mechanisms underlying AAA development and progression. ATF3 (activating transcription factor 3) has been increasingly recognized as a key regulator of cardiovascular diseases. However, the role of ATF3 in AAA development and progression remains elusive. METHODS Genome-wide RNA sequencing analysis was performed on the aorta isolated from saline or Ang II (angiotensin II)-induced AAA mice, and ATF3 was identified as the potential key gene for AAA development. To examine the role of ATF3 in AAA development, vascular smooth muscle cell-specific ATF3 knockdown or overexpressed mice by recombinant adeno-associated virus serotype 9 vectors carrying ATF3, or shRNA-ATF3 with SM22α (smooth muscle protein 22-α) promoter were used in Ang II-induced AAA mice. In human and murine vascular smooth muscle cells, gain or loss of function experiments were performed to investigate the role of ATF3 in vascular smooth muscle cell proliferation and apoptosis. RESULTS In both Ang II-induced AAA mice and patients with AAA, the expression of ATF3 was reduced in aneurysm tissues but increased in aortic lesion tissues. The deficiency of ATF3 in vascular smooth muscle cell promoted AAA formation in Ang II-induced AAA mice. PDGFRB (platelet-derived growth factor receptor β) was identified as the target of ATF3, which mediated vascular smooth muscle cell proliferation in response to TNF-alpha (tumor necrosis factor-α) at the early stage of AAA. ATF3 suppressed the mitochondria-dependent apoptosis at the advanced stage by upregulating its direct target BCL2. Our chromatin immunoprecipitation results also demonstrated that the recruitment of NFκB1 and P300/BAF/H3K27ac complex to the ATF3 promoter induces ATF3 transcription via enhancer activation. NFKB1 inhibitor (andrographolide) inhibits the expression of ATF3 by blocking the recruiters NFKB1 and ATF3-enhancer to the ATF3-promoter region, ultimately leading to AAA development. CONCLUSIONS Our results demonstrate a previously unrecognized role of ATF3 in AAA development and progression, and ATF3 may serve as a novel therapeutic and prognostic marker for AAA.
Collapse
MESH Headings
- Activating Transcription Factor 3/genetics
- Activating Transcription Factor 3/metabolism
- Animals
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/chemically induced
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice
- Male
- Mice, Inbred C57BL
- Apoptosis
- Cells, Cultured
- Angiotensin II
- Cell Proliferation
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Disease Models, Animal
Collapse
Affiliation(s)
- Ying Wen
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Yingying Liu
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Qiang Li
- Department of Vascular Surgery (Q.L.), the Second Affiliated Hospital of Guangzhou Medical University, China
| | - Jinlin Tan
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Xing Fu
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Yiwen Liang
- Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, China (Y. Liang)
| | - Yonghua Tuo
- Department of Neurosurgery (Y.T.), the Second Affiliated Hospital of Guangzhou Medical University, China
| | - Luhao Liu
- Department of Organ Transplantation (L.L., Z.C.), the Second Affiliated Hospital of Guangzhou Medical University, China
| | - Xueqiong Zhou
- Department of Occupational Health and Medicine, School of Public Health, Southern Medical University, China (X.Z.)
| | - Dongkai LiuFu
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Xuejiao Fan
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Chaofei Chen
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Zheng Chen
- Department of Organ Transplantation (L.L., Z.C.), the Second Affiliated Hospital of Guangzhou Medical University, China
| | - Zhouping Wang
- Department of Cardiology (Z.W.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Shunyang Fan
- Heart Center, The Third Affiliated Hospital of Zhengzhou University, China (S.F., W.H.T.)
| | - Renjing Liu
- Victor Chang Cardiac Research Institute, Sydney, Australia (R.L.)
| | - Lei Pan
- The Center for Microbes, Development, and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, China (L.P.)
| | - Yuan Zhang
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Wai Ho Tang
- Institute of Pediatrics (Y.W., Y. Liu, J.T., X.F., D.L., X.F., C.C., Y.Z., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
- Heart Center, The Third Affiliated Hospital of Zhengzhou University, China (S.F., W.H.T.)
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Kowloon, Hong Kong SAR, China (W.H.T.)
| |
Collapse
|
6
|
Punziano C, Trombetti S, Cesaro E, Grosso M, Faraonio R. Antioxidant Systems as Modulators of Ferroptosis: Focus on Transcription Factors. Antioxidants (Basel) 2024; 13:298. [PMID: 38539832 PMCID: PMC10967371 DOI: 10.3390/antiox13030298] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 10/28/2024] Open
Abstract
Ferroptosis is a type of programmed cell death that differs from apoptosis, autophagy, and necrosis and is related to several physio-pathological processes, including tumorigenesis, neurodegeneration, senescence, blood diseases, kidney disorders, and ischemia-reperfusion injuries. Ferroptosis is linked to iron accumulation, eliciting dysfunction of antioxidant systems, which favor the production of lipid peroxides, cell membrane damage, and ultimately, cell death. Thus, signaling pathways evoking ferroptosis are strongly associated with those protecting cells against iron excess and/or lipid-derived ROS. Here, we discuss the interaction between the metabolic pathways of ferroptosis and antioxidant systems, with a particular focus on transcription factors implicated in the regulation of ferroptosis, either as triggers of lipid peroxidation or as ferroptosis antioxidant defense pathways.
Collapse
Affiliation(s)
- Carolina Punziano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Silvia Trombetti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy
| | - Elena Cesaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| |
Collapse
|
7
|
Lou Y, Song F, Kang Y, Xu Y. Periodic Mechanical Stress Inhibits the Development of Osteoarthritis via Regulating ATF3-Akt Axis. J Inflamm Res 2023; 16:5613-5628. [PMID: 38046403 PMCID: PMC10693248 DOI: 10.2147/jir.s419186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/25/2023] [Indexed: 12/05/2023] Open
Abstract
Purpose The development of osteoarthritis (OA) has been linked to mechanical factors. Studies suggest that periodic mechanical stress (PMS) may be a factor contributing to cartilage repair and the onset of OA. Therefore, this study was designed to explore the effects and underlying mechanisms of PMS on OA development. Patients and Methods Firstly, surgery and interleukin (IL)-1β were used for the establishment of rat/cell models of OA, respectively. Subsequently, activating transcription factor (ATF) 3 expression was knocked down in OA rats, and OA chondrocytes were treated with different heights (0, 1, 2, 4, 8 cm) of PMS or si-ATF. Safranin O staining was used to observe the histological changes in the rat knee joint, and enzyme-linked immunosorbent assay (ELISA) was performed to detect levels of tumor necrosis factor (TNF)-α, IL-6, and IL-8 in vivo and in vitro. Further, the expression of extracellular matrix (ECM) proteins in the rat knee joint was assessed immunohistochemistry. Flow cytometry was used to evaluate chondrocyte apoptosis. Lastly, Western blot was performed to detect the expression of related proteins of the protein kinase B (Akt) signaling pathway and ECM. Results The OA rat model was successfully constructed. Further experiments indicated that the knockdown of ATF3 not only alleviated joint swelling, pain, inflammatory response and pathological damage, but also promoted ECM synthesis and the phosphorylation of Akt in OA rats. In vitro experiments showed that PMS (4 cm) effectively inhibited cell apoptosis, decreased the levels of TNF-α, IL-6 and IL-8, promoted ECM synthesis, and activated the Akt signaling pathway in osteoarthritic chondrocytes. However, ATF3 overexpression reversed the positive effects of PMS on osteoarthritic chondrocytes. Conclusion PMS can effectively inhibit the development of OA, and its protective effects may be attributed to the down-regulation of ATF3 expression and activation of the Akt signaling pathway.
Collapse
Affiliation(s)
- Yi Lou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
- Department of Orthopaedics, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai, 201805, People’s Republic of China
| | - Fanglong Song
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Yifan Kang
- Department of Orthopaedics, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai, 201805, People’s Republic of China
| | - Yaozeng Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| |
Collapse
|
8
|
Ke H, Chen Z, Zhao X, Yang C, Luo T, Ou W, Wang L, Liu H. Research progress on activation transcription factor 3: A promising cardioprotective molecule. Life Sci 2023:121869. [PMID: 37355225 DOI: 10.1016/j.lfs.2023.121869] [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: 03/31/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
Activation transcription factor 3 (ATF3), a member of the ATF/cyclic adenosine monophosphate response element binding family, can be induced by a variety of stresses. Numerous studies have indicated that ATF3 plays multiple roles in the development and progression of cardiovascular diseases, including atherosclerosis, hypertrophy, fibrosis, myocardial ischemia-reperfusion, cardiomyopathy, and other cardiac dysfunctions. In past decades, ATF3 has been demonstrated to be detrimental to some cardiac diseases. Current studies have indicated that ATF3 can function as a cardioprotective molecule in antioxidative stress, lipid metabolic metabolism, energy metabolic regulation, and cell death modulation. To unveil the potential therapeutic role of ATF3 in cardiovascular diseases, we organized this review to explore the protective effects and mechanisms of ATF3 on cardiac dysfunction, which might provide rational evidence for the prevention and cure of cardiovascular diseases.
Collapse
Affiliation(s)
- Haoteng Ke
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zexing Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xuanbin Zhao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chaobo Yang
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Tao Luo
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Wen Ou
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Lizi Wang
- Department of Health Management, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haiqiong Liu
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Health Management, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
9
|
Kotlyarov S. Effects of Atherogenic Factors on Endothelial Cells: Bioinformatics Analysis of Differentially Expressed Genes and Signaling Pathways. Biomedicines 2023; 11:1216. [PMID: 37189834 PMCID: PMC10135807 DOI: 10.3390/biomedicines11041216] [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: 03/11/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
(1) Background: Atherosclerosis is a serious medical condition associated with high morbidity and mortality rates. It develops over many years as a complex chain of events in the vascular wall involving various cells and is influenced by many factors of clinical interest. (2) Methods: In this study, we performed a bioinformatic analysis of Gene Expression Omnibus (GEO) datasets to investigate the gene ontology of differentially expressed genes (DEGs) in endothelial cells exposed to atherogenic factors such as tobacco smoking, oscillatory shear, and oxidized low-density lipoproteins (oxLDL). DEGs were identified using the limma R package, and gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and protein-protein interaction (PPI) network analysis were performed. (3) Results: We studied biological processes and signaling pathways involving DEGs in endothelial cells under the influence of atherogenic factors. GO enrichment analysis demonstrated that the DEGs were mainly involved in cytokine-mediated signaling pathway, innate immune response, lipid biosynthetic process, 5-lipoxygenase activity, and nitric-oxide synthase activity. KEGG pathway enrichment analysis showed that common pathways included tumor necrosis factor signaling pathway, NF-κB signaling pathway, NOD-like receptor signaling pathway, lipid and atherosclerosis, lipoprotein particle binding, and apoptosis. (4) Conclusions: Atherogenic factors such as smoking, impaired flow, and oxLDL contribute to impaired innate immune response, metabolism, and apoptosis in endothelial cells, potentially leading to the development of atherosclerosis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|