1
|
Chang C, Huang WZ, Cai RP, Mo LR, Wu Q, Su Q. Research Progress of Regulatory Cell Death in Coronary Microembolization. Int J Med Sci 2025; 22:132-139. [PMID: 39744170 PMCID: PMC11659838 DOI: 10.7150/ijms.105295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025] Open
Abstract
Coronary microembolization (CME) is defined as atherosclerotic plaque erosion, spontaneous rupture, or rupture of the plaque while undergoing interventional therapy resulting in the formation of tiny emboli that obstruct the coronary microcirculatory system. For percutaneous coronary intervention, CME is a major complication, with a periprocedural incidence of up to 25%. Recent studies have demonstrated that regulatory cell death (RCD) exerts a profound influence on CME through its modulation of inflammatory responses, oxidative stress, cell death, and angiogenesis. RCD, including apoptosis, autophagy, and pyroptosis, is a unique class of genetically highly regulated death patterns pervasive in instances of coronary microembolization. The aim of this review is to summarize the currently known molecular mechanisms underlying CME. Further investigations of the RCD mechanisms may unravel new avenues for the prevention and treatment of CME.
Collapse
Affiliation(s)
- Chen Chang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shanxi, People's Republic of China
| | - Wan-Zhong Huang
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, People's Republic of China
| | - Ru-Ping Cai
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University, Taian 271016, Shandong, People's Republic of China
| | - Li-Rong Mo
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, People's Republic of China
| | - Qiang Wu
- Senior Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Qiang Su
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, People's Republic of China
| |
Collapse
|
2
|
Kim H, Massett MP. Beneficial effects of rapamycin on endothelial function in systemic lupus erythematosus. Front Physiol 2024; 15:1446836. [PMID: 39234308 PMCID: PMC11372898 DOI: 10.3389/fphys.2024.1446836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Endothelial function is significantly impaired in patients with SLE compared to healthy controls. Elevated activation of the mammalian target of rapamycin complex 1 (mTORC1) is reported in humans and mice with SLE. However, it is unclear if elevated mTORC1 in SLE contributes to impaired mitophagy and endothelial dysfunction. Therefore, we tested the hypothesis that inhibiting mTORC1 with rapamycin would increase mitophagy and attenuate endothelial dysfunction and inflammatory responses in SLE. Methods Nine-week-old female lupus-prone (MRL/lpr) and healthy control (MRL/MpJ) mice were randomly assigned into rapamycin treatment (lpr_Rapamycin and MpJ_Rapamycin) or control (lpr_Control and MpJ_Control) groups. Rapamycin was injected i.p. 3 days per week for 8 weeks. After 8 weeks, endothelium-dependent vasorelaxation to acetylcholine (ACh) and endothelium-independent vasorelaxation to sodium nitroprusside (SNP) were measured in thoracic aortas using a wire myograph. Results MTORC1 activity was increased in aorta from lpr mice as demonstrated by increased phosphorylation of s6rp and p70s6k and significantly inhibited by rapamycin (s6rp, p < 0.0001, p70s6k, p = 0.04, respectively). Maximal responses to Ach were significantly impaired in lpr_Control (51.7% ± 6.6%) compared to MpJ_Control (86.7% ± 3.6%) (p < 0.0001). Rapamycin prevented endothelial dysfunction in the thoracic aorta from lupus mice (lpr_Rapamycin) (79.6% ± 4.2%) compared to lpr_Control (p = 0.002). Maximal responses to SNP were not different across groups. Phosphorylation of endothelial nitric oxide synthase also was 42% lower in lpr_Control than MpJ_Control and 46% higher in lpr_Rapamycin than lpr_Control. The inflammatory marker, vascular cell adhesion protein 1 (Vcam 1), was elevated in aorta from lupus mice compared with healthy mice (p = 0.001), and significantly reduced with Rapamycin treatment (p = 0.0021). Mitophagy markers were higher in lupus mice and reduced by rapamycin treatment, suggesting altered mitophagy in lpr mice. Conclusion Collectively, these results demonstrate the beneficial effects of inhibiting mTORC1 on endothelial function in SLE mice and suggest inflammation and altered mitophagy contribute to endothelial dysfunction in SLE.
Collapse
Affiliation(s)
- Hyoseon Kim
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| | - Michael P Massett
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
3
|
Feng W, Qiao J, Tan Y, Liu Q, Wang Q, Yang B, Yang S, Cui L. Interaction of antiphospholipid antibodies with endothelial cells in antiphospholipid syndrome. Front Immunol 2024; 15:1361519. [PMID: 39044818 PMCID: PMC11263079 DOI: 10.3389/fimmu.2024.1361519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease with arteriovenous thrombosis and recurrent miscarriages as the main clinical manifestations. Due to the complexity of its mechanisms and the diversity of its manifestations, its diagnosis and treatment remain challenging issues. Antiphospholipid antibodies (aPL) not only serve as crucial "biomarkers" in diagnosing APS but also act as the "culprits" of the disease. Endothelial cells (ECs), as one of the core target cells of aPL, bridge the gap between the molecular level of these antibodies and the tissue and organ level of pathological changes. A more in-depth exploration of the relationship between ECs and the pathogenesis of APS holds the potential for significant advancements in the precise diagnosis, classification, and therapy of APS. Many researchers have highlighted the vital involvement of ECs in APS and the underlying mechanisms governing their functionality. Through extensive in vitro and in vivo experiments, they have identified multiple aPL receptors on the EC membrane and various intracellular pathways. This article furnishes a comprehensive overview and summary of these receptors and signaling pathways, offering prospective targets for APS therapy.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| |
Collapse
|
4
|
Tan Y, Qiao J, Yang S, Liu H, Wang Q, Liu Q, Feng W, Cui L. H3K4me3-Mediated FOXJ2/SLAMF8 Axis Aggravates Thrombosis and Inflammation in β2GPI/Anti-β2GPI-Treated Monocytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309140. [PMID: 38639399 PMCID: PMC11199983 DOI: 10.1002/advs.202309140] [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: 11/28/2023] [Revised: 03/31/2024] [Indexed: 04/20/2024]
Abstract
Antiphospholipid syndrome (APS) is characterized by thrombus formation, poor pregnancy outcomes, and a proinflammatory response. H3K4me3-related monocytes activation are key regulators of APS pathogenesis. Therefore, H3K4me3 CUT&Tag and ATAC-seq are performed to examine the epigenetic profiles. The results indicate that the H3K4me3 signal and chromatin accessibility at the FOXJ2 promoter are enhanced in an in vitro monocyte model by stimulation with β2GPI/anti-β2GPI, which mimics APS, and decreases after OICR-9429 administration. Furthermore, FOXJ2 is highly expressed in patients with primary APS (PAPS) and is the highest in patients with triple-positive antiphospholipid antibodies (aPLs). Mechanistically, FOXJ2 directly binds to the SLAMF8 promoter and activates SLAMF8 transcription. SLAMF8 further interacts with TREM1 to stimulate TLR4/NF-κB signaling and prohibit autophagy. Knockdown of FOXJ2, SLAMF8, or TREM1 blocks TLR4/NF-κB and provokes autophagy, subsequently inhibiting the release of inflammatory and thrombotic indicators. A mouse model of vascular APS is established via β2GPI intraperitoneal injection, and the results suggest that OICR-9429 administration attenuates the inflammatory response and thrombus formation by inactivating FOXJ2/SLAMF8/TREM1 signaling. These findings highlight the overexpression of H3K4me3-mediated FOXJ2 in APS, which consequently accelerates APS pathogenesis by triggering inflammation and thrombosis via boosting the SLAMF8/TREM1 axis. Therefore, OICR-9429 is a promising candidate drug for APS therapy.
Collapse
Affiliation(s)
- Yuan Tan
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Jiao Qiao
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Shuo Yang
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Hongchao Liu
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Qingchen Wang
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Qi Liu
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Weimin Feng
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Liyan Cui
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| |
Collapse
|
5
|
Gao Y, Zhang L, Zhang F, Liu R, Liu L, Li X, Zhu X, Liang Y. Traditional Chinese medicine and its active substances reduce vascular injury in diabetes via regulating autophagic activity. Front Pharmacol 2024; 15:1355246. [PMID: 38505420 PMCID: PMC10949535 DOI: 10.3389/fphar.2024.1355246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/26/2024] [Indexed: 03/21/2024] Open
Abstract
Due to its high prevalence, poor prognosis, and heavy burden on healthcare costs, diabetic vascular complications have become a significant public health issue. Currently, the molecular and pathophysiological mechanisms underlying diabetes-induced vascular complications remain incompletely understood. Autophagy, a highly conserved process of lysosomal degradation, maintains intracellular homeostasis and energy balance via removing protein aggregates, damaged organelles, and exogenous pathogens. Increasing evidence suggests that dysregulated autophagy may contribute to vascular abnormalities in various types of blood vessels, including both microvessels and large vessels, under diabetic conditions. Traditional Chinese medicine (TCM) possesses the characteristics of "multiple components, multiple targets and multiple pathways," and its safety has been demonstrated, particularly with minimal toxicity in liver and kidney. Thus, TCM has gained increasing attention from researchers. Moreover, recent studies have indicated that Chinese herbal medicine and its active compounds can improve vascular damage in diabetes by regulating autophagy. Based on this background, this review summarizes the classification, occurrence process, and related molecular mechanisms of autophagy, with a focus on discussing the role of autophagy in diabetic vascular damage and the protective effects of TCM and its active compounds through the regulation of autophagy in diabetes. Moreover, we systematically elucidate the autophagic mechanisms by which TCM formulations, individual herbal extracts, and active compounds regulate diabetic vascular damage, thereby providing new candidate drugs for clinical treatment of vascular complications in diabetes. Therefore, further exploration of TCM and its active compounds with autophagy-regulating effects holds significant research value for achieving targeted therapeutic approaches for diabetic vascular complications.
Collapse
Affiliation(s)
- Yankui Gao
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Lei Zhang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Fei Zhang
- Department of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Lanzhou, China
| | - Rong Liu
- Department of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Lei Liu
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaoyan Li
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiangdong Zhu
- Department of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Yonglin Liang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
6
|
Ni D, Lei C, Liu M, Peng J, Yi G, Mo Z. Cell death in atherosclerosis. Cell Cycle 2024; 23:495-518. [PMID: 38678316 PMCID: PMC11135874 DOI: 10.1080/15384101.2024.2344943] [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: 08/10/2022] [Accepted: 04/14/2024] [Indexed: 04/29/2024] Open
Abstract
A complex and evolutionary process that involves the buildup of lipids in the arterial wall and the invasion of inflammatory cells results in atherosclerosis. Cell death is a fundamental biological process that is essential to the growth and dynamic equilibrium of all living things. Serious cell damage can cause a number of metabolic processes to stop, cell structure to be destroyed, or other irreversible changes that result in cell death. It is important to note that studies have shown that the two types of programmed cell death, apoptosis and autophagy, influence the onset and progression of atherosclerosis by controlling these cells. This could serve as a foundation for the creation of fresh atherosclerosis prevention and treatment strategies. Therefore, in this review, we summarized the molecular mechanisms of cell death, including apoptosis, pyroptosis, autophagy, necroptosis, ferroptosis and necrosis, and discussed their effects on endothelial cells, vascular smooth muscle cells and macrophages in the process of atherosclerosis, so as to provide reference for the next step to reveal the mechanism of atherosclerosis.
Collapse
Affiliation(s)
- Dan Ni
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
| | - Cai Lei
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Minqi Liu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Province Postgraduate Co-training Base for Cooperative Innovation in Basic Medicine (Guilin Medical University and Yueyang Women & Children’s Medical Center), Yueyang, China
| | - Jinfu Peng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Guanghui Yi
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Zhongcheng Mo
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Province Postgraduate Co-training Base for Cooperative Innovation in Basic Medicine (Guilin Medical University and Yueyang Women & Children’s Medical Center), Yueyang, China
| |
Collapse
|
7
|
Zhang Y, Jin S, Tian W, Shi D, Chen Y, Cui L, Zheng J. Proteomics of Serum Samples for the Exploration of the Pathological Mechanism of Obstetric Antiphospholipid Syndrome. J Proteome Res 2024; 23:289-300. [PMID: 38048430 PMCID: PMC10775856 DOI: 10.1021/acs.jproteome.3c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023]
Abstract
Obstetric antiphospholipid syndrome (OAPS) is a multisystem disorder characterized by thrombosis or recurrent fetal loss. In this study, we aim to explore the pathological mechanism of OAPS. Herein, we carried out data-independent acquisition (DIA) mass spectrometry quantitative proteomic analysis of serum samples from OAPS patients and healthy controls. A set of 93 differentially expressed proteins was identified, including 75 upregulated and 18 downregulated proteins compared with the levels in controls. Those proteins are enriched in KEGG pathways related to autoimmune diseases, allergic diseases, and pathogen infection. Interestingly, metabolic pathways such as fatty acid degradation and type I diabetes were enriched, indicating that OAPS is metabolic disease related. The significantly increased triglyceride also supported this idea. The differentially expressed proteins insulin-like growth factor-binding protein-1 (IGFBP-1), C-reactive protein (CRP), and ferritin light chain (FTL) were validated by ELISA. Our study presented a deep serum proteomics of OAPS and advanced our understanding of OAPS pathogenesis.
Collapse
Affiliation(s)
- Yinmei Zhang
- Department
of Laboratory Medicine, Peking University
Third Hospital, Beijing 100191, China
| | - Shangjia Jin
- Department
of Laboratory Medicine, Peking University
Third Hospital, Beijing 100191, China
| | - Wenmin Tian
- Department
of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics
Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Dongxue Shi
- Department
of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics
Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yang Chen
- Department
of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics
Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Liyan Cui
- Department
of Laboratory Medicine, Peking University
Third Hospital, Beijing 100191, China
| | - Jiajia Zheng
- Department
of Laboratory Medicine, Peking University
Third Hospital, Beijing 100191, China
| |
Collapse
|
8
|
He X, Sun Y, Lu X, Yang F, Li T, Deng C, Song J, Huang X. Assessment of the anti-inflammatory mechanism of quercetin 3,7-dirhamnoside using an integrated pharmacology strategy. Chem Biol Drug Des 2023; 102:1534-1552. [PMID: 37806949 DOI: 10.1111/cbdd.14346] [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: 05/21/2023] [Revised: 07/22/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023]
Abstract
Pouzolzia zeylanica (L.) Benn. is a Chinese herbal medicine widely used for its anti-inflammatory and pus-removal properties. To explore its potential anti-inflammatory mechanism, quercetin 3,7-dirhamnoside (QDR), the main flavonoid component of P. zeylanica (L.) Benn., was extracted and purified. The potential anti-inflammatory targets of QDR were predicted using network analysis. These potential targets were verified using molecular docking, molecular dynamics simulations, and in vitro experiments. Consequently, 342 potential anti-inflammatory QDR targets were identified. By analyzing the intersection between the protein-protein interaction and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, we identified several potential protein targets of QDR, including RAC-alpha serine/threonine-protein kinase (AKT1), Ras-related C3 botulinum toxin substrate 1 (RAC1), nitric oxide synthase 3 (NOS3), serine/threonine-protein kinase mTOR (mTOR), epidermal growth factor receptor (EGFR), growth factor receptor-bound protein 2 (GRB2), and endothelin-1 receptor (EDNRA). QDR has anti-inflammatory activity and regulates immune responses and apoptosis through chemokines, Phosphatidylinositol 3-kinase 3(PI3K)/AKT, cAMP, T-cell receptor, and Ras signaling pathways. Molecular docking analysis showed that QDR has good binding abilities with AKT1, mTOR, and NOS3. In addition, molecular dynamics simulations demonstrated that the protein-ligand complex systems formed between QDR and AKT1, mTOR, and NOS3 have high dynamic stability, and their protein-ligand complex systems possess strong binding ability. In RAW264.7 macrophages, QDR significantly inhibited lipopolysaccharides (LPS)-induced inducible nitric oxide synthase expression, nitric oxide (NO) release and the generation of proinflammatory cytokines IL-6, IL-1β, and TNF-α. QDR downregulated the expression of p-AKT1(Ser473)/AKT1 and p-mTOR (Ser2448)/mTOR, and upregulated the expression of NOS3, Rictor, and Raptor. This indicates that the anti-inflammatory mechanisms of QDR involve regulation of AKT1 and mTOR to prevent apoptosis and of NOS3 which leads to the release of endothelial NO. Thus, our study elucidated the potential anti-inflammatory mechanism of QDR, the main flavonoid found in P. zeylanica (L.) Benn.
Collapse
Affiliation(s)
- Xinqian He
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongzhi Sun
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaomeng Lu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fan Yang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Li
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changsheng Deng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianping Song
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin'an Huang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
9
|
Łanoszka K, Vlčková N. Natural Sirtuin1 Activators and Atherosclerosis: an Overview. Curr Atheroscler Rep 2023; 25:979-994. [PMID: 38038821 PMCID: PMC10770200 DOI: 10.1007/s11883-023-01165-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 12/02/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the most recent findings investigating the impact of several natural sirtuin (SIRT) activators, particularly SIRT1, on atherosclerosis. RECENT FINDINGS Sirtuins that belong to a family of class III histone deacetylases are believed to be novel therapeutic targets to treat age-related and chronic diseases. SIRT expression is regulated by small molecules called SIRT-activating compounds that can be found in natural food products. SIRT1 may exert protective effects in atherosclerosis, which is said to be a major cause of cardiovascular diseases. Most of the evidence supporting the beneficial effects of these natural compounds comes from in vitro or animal-based studies, while there have been particularly few or inconsistent human-based studies evaluating their long-term impact in recent years. SIRT1 activation has been demonstrated to mitigate or prevent atherosclerosis through various mechanisms. However, further research is required to determine the optimal SIRT activator dosage and to establish a stronger correlation between health effects and the administration of bioactive compounds. Additionally, conducting more human clinical trials is necessary to ensure the safety of these compounds for preventing atherosclerosis development.
Collapse
Affiliation(s)
- Karolina Łanoszka
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, University of Agriculture in Krakow, 122 Balicka Street, 30-149, Krakow, Poland
| | - Nimasha Vlčková
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, University of Agriculture in Krakow, 122 Balicka Street, 30-149, Krakow, Poland.
| |
Collapse
|
10
|
Tang D, Wang G, Liu Z, Wang B, Yao M, Wang Q, Hou X, Zheng Y, Sheng C, Zhou Z. Transcriptomic analysis of the effects of the HPV18 E6E7 gene on the cell death mode in esophageal squamous cell carcinoma. Oncol Lett 2023; 25:167. [PMID: 36960186 PMCID: PMC10028223 DOI: 10.3892/ol.2023.13753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 02/21/2023] [Indexed: 03/25/2023] Open
Abstract
Human papillomavirus (HPV) infection is one of the main causes of esophageal carcinoma (ESCA), and its carcinogenic mechanisms in ESCA require further investigation. E6 and E7 are HPV oncogenes, and their genomic integration is a crucial reason for the transformation of host cells into cancer cells. In order to reveal the role of oncogenes E6 and E7 in ESCA cells, the RNA-Seq raw data for HPV18-positive and -negative esophageal squamous cell carcinoma (ESCC) samples derived from the NCBI BioProject database were analyzed, and the differentially expressed genes were identified. Moreover, differentially expressed genes were enriched significantly in multiple cell death pathways, including apoptosis (cyclin-dependent kinase inhibitor 2A, plakophilin 1 and desmoglein 3), pyroptosis (gasdermin A, gasdermin C, NLR family pyrin domain containing 3, absent in melanoma 2, NLR family pyrin domain containing 1 and Toll like receptor 1) and autophagy (Unc-51 like autophagy activating kinase 1, adrenoceptor beta 2). Consequently, the effects of cisplatin-induced apoptosis and Hank's balanced salt solution-induced autophagy, and α-ketoglutarate-induced pyroptosis in the ESCC-expressing E6 and E7 cells were verified. Therefore, the expression of E6E7 may culminate in the inhibition of multiple cell death modes, which may also be one of the mechanisms of oncogene-induced carcinogenesis.
Collapse
Affiliation(s)
- Duo Tang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Guozhen Wang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Zijia Liu
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Biqi Wang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Mengfei Yao
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Qian Wang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Xiaonan Hou
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Yuchen Zheng
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Chao Sheng
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Zhixiang Zhou
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
- Correspondence to: Professor Zhixiang Zhou, Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, 100 Pingleyuan, Chaoyang, Beijing 100124, P.R. China, E-mail:
| |
Collapse
|
11
|
Maha MS, Panjaitan NSD. Response to Article “Autophagy Inhibits Inflammation via Down-Regulation of p38 MAPK/mTOR Signaling Cascade in Endothelial Cells” [Letter]. Clin Cosmet Investig Dermatol 2023; 16:749-750. [PMID: 37013152 PMCID: PMC10066714 DOI: 10.2147/ccid.s412865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023]
Affiliation(s)
- Masri Sembiring Maha
- Center for Biomedical Research, Research Organization for Health, National Research and Innovation Agency (BRIN), Cibinong Science Center, Cibinong, West Java, Indonesia
| | - Novaria Sari Dewi Panjaitan
- Center for Biomedical Research, Research Organization for Health, National Research and Innovation Agency (BRIN), Cibinong Science Center, Cibinong, West Java, Indonesia
- Correspondence: Novaria Sari Dewi Panjaitan, Center for Biomedical Research, Research Organization for Health, National Research and Innovation Agency (BRIN), Cibinong Science Center, Jl. Raya Bogor No. 490, Cibinong – Bogor Km. 46, Cibinong, West Java, Indonesia, Email
| |
Collapse
|
12
|
Wu Q, Lv Q, Liu X, Ye X, Cao L, Wang M, Li J, Yang Y, Li L, Wang S. Natural compounds from botanical drugs targeting mTOR signaling pathway as promising therapeutics for atherosclerosis: A review. Front Pharmacol 2023; 14:1083875. [PMID: 36744254 PMCID: PMC9894899 DOI: 10.3389/fphar.2023.1083875] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that is a major cause of cardiovascular diseases (CVDs), including coronary artery disease, hypertension, myocardial infarction, and heart failure. Hence, the mechanisms of AS are still being explored. A growing compendium of evidence supports that the activity of the mechanistic/mammalian target of rapamycin (mTOR) is highly correlated with the risk of AS. The mTOR signaling pathway contributes to AS progression by regulating autophagy, cell senescence, immune response, and lipid metabolism. Various botanical drugs and their functional compounds have been found to exert anti- AS effects by modulating the activity of the mTOR signaling pathway. In this review, we summarize the pathogenesis of AS based on the mTOR signaling pathway from the aspects of immune response, autophagy, cell senescence, and lipid metabolism, and comb the recent advances in natural compounds from botanical drugs to inhibit the mTOR signaling pathway and delay AS development. This review will provide a new perspective on the mechanisms and precision treatments of AS.
Collapse
Affiliation(s)
- Qian Wu
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Qianyu Lv
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao’an Liu
- Capital University of Medical, Beijing, China
| | - Xuejiao Ye
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Linlin Cao
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Manshi Wang
- Beijing Xicheng District Guangwai Hospital, Beijing, China
| | - Junjia Li
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yingtian Yang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Lanlan Li
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Wang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Lou D, Xing X, Liang Y. Dendrobine modulates autophagy to alleviate ox-LDL-induced oxidative stress and senescence in HUVECs. Drug Dev Res 2022; 83:1125-1137. [PMID: 35417048 DOI: 10.1002/ddr.21937] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/20/2022] [Accepted: 02/28/2022] [Indexed: 12/14/2022]
Abstract
Dendrobine has potential advantages in suppressing atherosclerosis (AS). FK506-binding protein 1A (FKBP1A) is implicated in the regulation of autophagy, inflammation, and apoptosis. To reveal the mechanism by which dendrobine inhibits AS by modulating autophagy, oxidative stress, apoptosis, and senescence. An in vitro AS cell model was induced by culturing human umbilical vein endothelial cells (HUVECs) with oxidized low-density lipoprotein (ox-LDL). The cells were treated with dendrobine alone or in combination with short hairpin RNA (shRNA) targeting FKBP1A or together with 3-methyladenine (3MA), an autophagy inhibitor. Inflammatory cytokines levels tumor necrosis factor-α, interleukin-6 (IL-6), and IL-1β were analyzed and oxidative stress levels were detected by the analysis of reactive oxygen species, malondialdehyde, and superoxide dismutase levels, followed by the analysis of apoptosis levels through terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Cell senescence was evaluated by senescence-associated β-galactosidase and light chain 3 (LC3) levels were detected by immunofluorescence (IF) staining. The targeting relationship of dendrobine and FKBP1A was predicted by SwissTarget, PyMol, Autodock, and Open Babel software. Dendrobine reduced the levels of proinflammation factors, oxidative stress levels, apoptosis levels, and senescence phenotype in ox-LDL-induced HUVECs. Besides, cell viability has an opposite change. Furthermore, there was an increase in LC3 IF tensity, and LC3-II/I and Beclin1 expressions, and a decrease in p62 expression. However, these effects of dendrobine could be markedly destroyed by shRNA silencing FKBP1A and 3MA. Dendrobine can suppress inflammatory responses, oxidative stress, apoptosis, and senescence via FKBP1A-involved autophagy ox-LDL-treated HUVECs.
Collapse
Affiliation(s)
- Danfei Lou
- Emergency Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyue Xing
- Emergency Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunyu Liang
- Geriatrics Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|