1
|
Wang J, Shen Y, Chen H, Guan J, Li Z, Liu X, Guo S, Wang L, Yan B, Jin C, Li H, Guo T, Sun Y, Zhang W, Zhang Z, Tian Y, Tian Z. Non-lethal sonodynamic therapy inhibits high glucose and palmitate-induced macrophage inflammasome activation through mtROS-DRP1-mitophagy pathway. FASEB J 2024; 38:e70178. [PMID: 39556373 DOI: 10.1096/fj.202402008r] [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: 08/28/2024] [Revised: 10/18/2024] [Accepted: 11/01/2024] [Indexed: 11/19/2024]
Abstract
Obesity plays a crucial role in the development and progression of type 2 diabetes mellitus (T2DM) by causing excessive release of free fatty acid from adipose tissue, which in turn leads to systemic infiltration of macrophages. In individuals with T2DM, the infiltration of macrophages into pancreatic islets results in islet inflammation that impairs beta cell function, as evidenced by increased apoptosis and decreased glucose-stimulated insulin secretion. The present study aimed to investigate the effects of non-lethal sonodynamic therapy (NL-SDT) on bone marrow-derived macrophages (BMDMs) exposed to high glucose and palmitic acid (HG/PA). These findings indicate that NL-SDT facilitates the expression of DRP1 through the transient production of mitochondrial ROS, which subsequently promotes mitophagy. This mitophagy was shown to limit the activation of the NLRP3 inflammasome and the secretion of IL-1β in BMDMs exposed to HG/PA. In co-culture experiments, beta cells exhibited significant dysfunction when interacting with HG/PA-treated BMDMs. However, this dysfunction was markedly alleviated when the BMDMs had undergone NL-SDT treatment. Moreover, NL-SDT was found to lower blood glucose levels and elevate serum insulin concentrations in db/db mice. Furthermore, NL-SDT effectively reduced the infiltration of F4/80-positive macrophages and the expression of CASP1 within islets. These findings provide fundamental insights into the mechanisms through which NL-SDT may serve as a promising approach for the treatment of T2DM.
Collapse
Affiliation(s)
- Jiayu Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Yicheng Shen
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Heyu Chen
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Jinwei Guan
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Zhitao Li
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Xianna Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Shuyuan Guo
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
- Department of Cardiology, 1st Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
| | - Linxin Wang
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
- Department of Cardiology, 1st Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
| | - Baoyue Yan
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Chenrun Jin
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - He Li
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Tian Guo
- Medical College of Jining Medical University, Jining, P. R. China
| | - Yun Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| | - Zhiguo Zhang
- School of Physics, Harbin Institute of Technology, Harbin, P. R. China
- School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin, P. R. China
| | - Ye Tian
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
- Department of Cardiology, 1st Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
| | - Zhen Tian
- Department of Pathophysiology, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Acoustic, Optical, Electrical and Magnetic Diagnostics and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, P. R. China
| |
Collapse
|
2
|
Pang Y, Li Q, Wang J, Wang S, Sharma A, Xu Y, Hu H, Li J, Liu S, Sun Y. An Ultrasound-Activated Supramolecular Modulator Enhancing Autophagy to Prevent Ventricular Arrhythmias Post-Myocardial Infarction. Angew Chem Int Ed Engl 2024:e202415802. [PMID: 39292161 DOI: 10.1002/anie.202415802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/19/2024]
Abstract
Ventricular arrhythmias (VAs) triggered by myocardial infarction (MI) are the leading cause of sudden cardiac mortality worldwide. Current therapeutic strategies for managing MI-induced VAs, such as left stellate ganglion resection and ablation, are suboptimal, highlighting the need to explore safer and more effective intervention strategies. Herein, we rationally designed two supramolecular sonosensitizers RuA and RuB, engineered through acceptor modification to generate moderate reactive oxygen species (ROS) to modulate VAs. Both RuA and RuB demonstrated high ultrasound (US)-activated ROS production efficiency, with singlet oxygen (1O2) quantum yield (ΦΔ) of 0.70 and 0.88, respectively, surpassing ligand IR1105 and the conventional sonosensitizer ICG (ΦΔ=0.40). In vitro, RuB, at a modest concentration and under US intensity notably boosts pro-survival autophagy in microglia BV2 cell. To improve in vivo stability and biocompatibility, RuB was further encapsulated into DSPE-PEG5000 to prepare RuB nanoparticles (RuB NPs). In vivo studies after microinjection of RuB NPs into the paraventricular nucleus (PVN) and subsequent US exposure, demonstrated that RuB NPs-mediated US modulation effectively suppresses sympathetic nervous activity (SNA) and inflammatory responses, thereby preventing VAs. Importantly, no tissue injury was observed post RuB NPs-mediated US modulation. This work pioneers the design of long-wave emission supramolecular sonosensitizers, offering new insights into regulating cardiovascular diseases.
Collapse
Affiliation(s)
- Yida Pang
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Qian Li
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Jiale Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan Univesity, Cardiovascular Research Institute, Wuhan University, Wuhan, 430072, China
| | - Songyun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan Univesity, Cardiovascular Research Institute, Wuhan University, Wuhan, 430072, China
| | - Amit Sharma
- Amity School of Chemical Sciences, Amity University Punjab Sector 82 A, Mohali, Punjab, 140306, India
| | - Yuling Xu
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Haoyuan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan Univesity, Cardiovascular Research Institute, Wuhan University, Wuhan, 430072, China
| | - Junrong Li
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Shuang Liu
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Yao Sun
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| |
Collapse
|
3
|
Huang J, Hong X, Chen S, He Y, Xie L, Gao F, Zhu C, Jin X, Yan H, Ye Y, Shao M, Du X, Feng G. Biomimetic Metal-Organic Framework Gated Nanoplatform for Sonodynamic Therapy against Extensively Drug Resistant Bacterial Lung Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402473. [PMID: 38962911 PMCID: PMC11434100 DOI: 10.1002/advs.202402473] [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: 03/08/2024] [Revised: 06/03/2024] [Indexed: 07/05/2024]
Abstract
Novel antimicrobial strategies are urgently needed to treat extensively drug-resistant (XDR) bacterial infections due to the high mortality rate and lack of effective therapeutic agents. Herein, nanoengineered human umbilical cord mesenchymal stem cells (hUC-MSCs), named PMZMU, are designed as a sonosensitizer for synergistic sonodynamic-nano-antimicrobial therapy against gram-negative XDR bacteria. PMZMU is composed of a bacterial targeting peptide (UBI29-41) modified hUC-MSCs membrane (MSCm), a sonosensitizer meso-tetra(4-car-boxyphenyl) porphine doped mesoporous organo-silica nanoparticle and an acidity-responsive metal-organic framework ZIF-8. This innovative formulation enables efficient loading of polymyxin B, reduces off-target drug release, increases circulation and targeting efficacy, and generates reactive oxygen species upon ultrasound irradiation. PMZMU exhibits remarkable in vitro inhibitory activity against four XDR bacteria: Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa (PA), and Escherichia coli. Taking advantage of the bacterial targeting ability of UBI29-41 and the inflammatory chemotaxis of hUC-MSC, PMZMU can be precisely delivered to lung infection sites thereby augmenting polymyxin B concentration. PMZMU-mediated sonodynamic therapy significantly reduces bacterial burden, relieves inflammatory damage by promoting the polarization of macrophages toward M2 phenotype, and improves survival rates without introducing adverse events. Overall, this study offers promising strategies for treating deep-tissue XDR bacterial infections, and guides the design and optimization of biomimetic nanomedicine.
Collapse
Affiliation(s)
- Jianling Huang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiuwen Hong
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Sixi Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yucong He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Lixu Xie
- Department of Pulmonary and Critical Care Medicine, Qi Lu Hospital of Shandong University, Wen hua xi Road 107#, Jinan, 250012, China
| | - Fenglin Gao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Chenghua Zhu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiao Jin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Haihao Yan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yongxia Ye
- Department of Radiology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, 210009, China
| | - Mingyue Shao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xingran Du
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China
| | - Ganzhu Feng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| |
Collapse
|
4
|
Hu H, Li Q, Wang J, Cheng Y, Zhao J, Hu C, Yin X, Wu Y, Sang R, Jiang H, Sun Y, Wang S. Mitochondria-targeted sonodynamic modulation of neuroinflammation to protect against myocardial ischemia‒reperfusion injury. Acta Biomater 2024:S1742-7061(24)00445-8. [PMID: 39122136 DOI: 10.1016/j.actbio.2024.08.003] [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: 03/22/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Sympathetic hyperactivation and inflammatory responses are the main causes of myocardial ischemia‒reperfusion (I/R) injury and myocardial I/R-related ventricular arrhythmias (VAs). Previous studies have demonstrated that light-emitting diodes (LEDs) could modulate post-I/R neuroinflammation, thus providing protection against myocardial I/R injury. Nevertheless, further applications of LEDs are constrained due to the low penetration depth (<1 cm) and potential phototoxicity. Low-intensity focused ultrasound (LIFU), an emerging noninvasive neuromodulation strategy with deeper penetration depth (∼10 cm), has been confirmed to modulate sympathetic nerve activity and inflammatory responses. Sonodynamic therapy (SDT), which combines LIFU with sonosensitizers, confers additional advantages, including superior therapeutic efficacy, precise localization of neuronal modulation and negligible side effects. Herein, LIFU and SDT were introduced to modulate post-myocardial I/R neuroinflammation to protect against myocardial I/R injury. The results indicated that LIFU and SDT inhibited sympathetic neural activity, suppressed the activation of astrocytes and microglia, and promoted microglial polarization towards the M2 phenotype, thereby attenuating myocardial I/R injury and preventing I/R-related malignant VAs. These insights suggest that LIFU and SDT inspire a noninvasive and efficient neuroinflammatory modulation strategy with great clinical translation potential thus benefiting more patients with myocardial I/R in the future. STATEMENT OF SIGNIFICANCE: Myocardial ischemia-reperfusion (I/R) may cause I/R injury and I/R-induced ventricular arrhythmias. Sympathetic hyperactivation and inflammatory response play an adverse effect in myocardial I/R injury. Previous studies have shown that light emitting diode (LED) can regulate I/R-induced neuroinflammation, thus playing a myocardial protective role. However, due to the low penetration depth and potential phototoxicity of LED, it is difficult to achieve clinical translation. Herein, we introduced sonodynamic modulation of neuroinflammation to protect against myocardial I/R injury, based on mitochondria-targeted nanosonosensitizers (CCNU980 NPs). We demonstrated that sonodynamic modulation could promote microglial autophagy, thereby preventing myocardial I/R injury and I/R-induced ventricular arrhythmias. This is the first example of sonodynamic modulation of myocardial I/R-induced neuroinflammation, providing a novel strategy for clinical translation.
Collapse
Affiliation(s)
- Haoyuan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qian Li
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Jiale Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ye Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jiahui Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Changhao Hu
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xinyue Yin
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuzhe Wu
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ruiqi Sang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Yao Sun
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China.
| | - Songyun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
5
|
Yan L, Wang J, Cai X, Liou Y, Shen H, Hao J, Huang C, Luo G, He W. Macrophage plasticity: signaling pathways, tissue repair, and regeneration. MedComm (Beijing) 2024; 5:e658. [PMID: 39092292 PMCID: PMC11292402 DOI: 10.1002/mco2.658] [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: 03/03/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Macrophages are versatile immune cells with remarkable plasticity, enabling them to adapt to diverse tissue microenvironments and perform various functions. Traditionally categorized into classically activated (M1) and alternatively activated (M2) phenotypes, recent advances have revealed a spectrum of macrophage activation states that extend beyond this dichotomy. The complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications orchestrates macrophage polarization, allowing them to respond to various stimuli dynamically. Here, we provide a comprehensive overview of the signaling cascades governing macrophage plasticity, focusing on the roles of Toll-like receptors, signal transducer and activator of transcription proteins, nuclear receptors, and microRNAs. We also discuss the emerging concepts of macrophage metabolic reprogramming and trained immunity, contributing to their functional adaptability. Macrophage plasticity plays a pivotal role in tissue repair and regeneration, with macrophages coordinating inflammation, angiogenesis, and matrix remodeling to restore tissue homeostasis. By harnessing the potential of macrophage plasticity, novel therapeutic strategies targeting macrophage polarization could be developed for various diseases, including chronic wounds, fibrotic disorders, and inflammatory conditions. Ultimately, a deeper understanding of the molecular mechanisms underpinning macrophage plasticity will pave the way for innovative regenerative medicine and tissue engineering approaches.
Collapse
Affiliation(s)
- Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Jue Wang
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Xin Cai
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Han‐Ming Shen
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences and Forensic MedicineSichuan University, and Collaborative Innovation Center for BiotherapyChengduChina
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| |
Collapse
|
6
|
Singh B, Cui K, Eisa-Beygi S, Zhu B, Cowan DB, Shi J, Wang DZ, Liu Z, Bischoff J, Chen H. Elucidating the crosstalk between endothelial-to-mesenchymal transition (EndoMT) and endothelial autophagy in the pathogenesis of atherosclerosis. Vascul Pharmacol 2024; 155:107368. [PMID: 38548093 PMCID: PMC11303600 DOI: 10.1016/j.vph.2024.107368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis, a chronic systemic inflammatory condition, is implicated in most cardiovascular ischemic events. The pathophysiology of atherosclerosis involves various cell types and associated processes, including endothelial cell activation, monocyte recruitment, smooth muscle cell migration, involvement of macrophages and foam cells, and instability of the extracellular matrix. The process of endothelial-to-mesenchymal transition (EndoMT) has recently emerged as a pivotal process in mediating vascular inflammation associated with atherosclerosis. This transition occurs gradually, with a significant portion of endothelial cells adopting an intermediate state, characterized by a partial loss of endothelial-specific gene expression and the acquisition of "mesenchymal" traits. Consequently, this shift disrupts endothelial cell junctions, increases vascular permeability, and exacerbates inflammation, creating a self-perpetuating cycle that drives atherosclerotic progression. While endothelial cell dysfunction initiates the development of atherosclerosis, autophagy, a cellular catabolic process designed to safeguard cells by recycling intracellular molecules, is believed to exert a significant role in plaque development. Identifying the pathological mechanisms and molecular mediators of EndoMT underpinning endothelial autophagy, may be of clinical relevance. Here, we offer new insights into the underlying biology of atherosclerosis and present potential molecular mechanisms of atherosclerotic resistance and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Bandana Singh
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Bo Zhu
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Douglas B Cowan
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jinjun Shi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Da-Zhi Wang
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zhenguo Liu
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
7
|
Zhang Y, Yang Y, Feng Y, Gao X, Pei L, Li X, Gao B, Liu L, Wang C, Gao S. Sonodynamic therapy for the treatment of atherosclerosis. J Pharm Anal 2024; 14:100909. [PMID: 38799235 PMCID: PMC11127226 DOI: 10.1016/j.jpha.2023.11.016] [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: 08/11/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 05/29/2024] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of large and medium-sized arteries that leads to ischemic heart disease, stroke, and peripheral vascular disease. Despite the current treatments, mortality and disability still remain high. Sonodynamic therapy (SDT), a non-invasive and localized methodology, has been developed as a promising new treatment for inhibiting atherosclerotic progression and stabilizing plaques. Promising progress has been made through cell and animal assays, as well as clinical trials. For example, the effect of SDT on apoptosis and autophagy of cells in AS, especially macrophages, and the concept of non-lethal SDT has also been proposed. In this review, we summarize the ultrasonic parameters and known sonosensitizers utilized in SDT for AS; we elaborate on SDT's therapeutic effects and mechanisms in terms of macrophages, T lymphocytes, neovascularization, smooth muscle cells, lipid, extracellular matrix and efferocytosis within plaques; additionally, we discuss the safety of SDT. A comprehensive summary of the confirmed effects of SDT on AS is conducted to establish a framework for future researchers.
Collapse
Affiliation(s)
- Yan Zhang
- The Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ying Yang
- The Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yudi Feng
- The Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xueyan Gao
- The Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Liping Pei
- The Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaopan Li
- The Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Bingxin Gao
- The Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Lin Liu
- The Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chengzeng Wang
- The Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shuochen Gao
- The Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
8
|
Chen C, Cao J, Gao Y, Xie X, Zhao Z, Yuan Q, He Y, Zu X, Liu J. High glucose promotes macrophage M1 polarization through miR-32/Mef2d/cAMP signaling pathway. Genes Dis 2024; 11:539-541. [PMID: 37692480 PMCID: PMC10491914 DOI: 10.1016/j.gendis.2023.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 03/07/2023] [Accepted: 03/29/2023] [Indexed: 09/12/2023] Open
Affiliation(s)
- Cong Chen
- The First Affiliated Hospital, Department of Laboratory Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jingsong Cao
- The First Affiliated Hospital, Institute of Endocrinology and Metabolism; Center for Clinical Research in Diabetes, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yan Gao
- The First Affiliated Hospital, Institute of Endocrinology and Metabolism; Center for Clinical Research in Diabetes, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoping Xie
- The First Affiliated Hospital, Department of Laboratory Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhibo Zhao
- The First Affiliated Hospital, Institute of Endocrinology and Metabolism; Center for Clinical Research in Diabetes, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qing Yuan
- The First Affiliated Hospital, Institute of Endocrinology and Metabolism; Center for Clinical Research in Diabetes, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yuqi He
- The First Affiliated Hospital, Department of Laboratory Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xuyu Zu
- The First Affiliated Hospital, Institute of Oncology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jianghua Liu
- The First Affiliated Hospital, Institute of Endocrinology and Metabolism; Center for Clinical Research in Diabetes, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
9
|
Solanki K, Bezsonov E, Orekhov A, Parihar SP, Vaja S, White FA, Obukhov AG, Baig MS. Effect of reactive oxygen, nitrogen, and sulfur species on signaling pathways in atherosclerosis. Vascul Pharmacol 2024; 154:107282. [PMID: 38325566 DOI: 10.1016/j.vph.2024.107282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease in which fats, lipids, cholesterol, calcium, proliferating smooth muscle cells, and immune cells accumulate in the intima of the large arteries, forming atherosclerotic plaques. A complex interplay of various vascular and immune cells takes place during the initiation and progression of atherosclerosis. Multiple reports indicate that tight control of reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS) production is critical for maintaining vascular health. Unrestricted ROS and RNS generation may lead to activation of various inflammatory signaling pathways, facilitating atherosclerosis. Given these deleterious consequences, it is important to understand how ROS and RNS affect the signaling processes involved in atherogenesis. Conversely, RSS appears to exhibit an atheroprotective potential and can alleviate the deleterious effects of ROS and RNS. Herein, we review the literature describing the effects of ROS, RNS, and RSS on vascular smooth muscle cells, endothelial cells, and macrophages and focus on how changes in their production affect the initiation and progression of atherosclerosis. This review also discusses the contribution of ROS, RNS, and RSS in mediating various post-translational modifications, such as oxidation, nitrosylation, and sulfation, of the molecules involved in inflammatory signaling.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Evgeny Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia; Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia; Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; The Cell Physiology and Pathology Laboratory, Turgenev State University of Orel, Orel, Russia
| | - Alexander Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Suraj P Parihar
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Shivani Vaja
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Fletcher A White
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India.
| |
Collapse
|
10
|
Pan W, Zhang J, Zhang L, Zhang Y, Song Y, Han L, Tan M, Yin Y, Yang T, Jiang T, Li H. Comprehensive view of macrophage autophagy and its application in cardiovascular diseases. Cell Prolif 2024; 57:e13525. [PMID: 37434325 PMCID: PMC10771119 DOI: 10.1111/cpr.13525] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the primary drivers of the growing public health epidemic and the leading cause of premature mortality and economic burden worldwide. With decades of research, CVDs have been proven to be associated with the dysregulation of the inflammatory response, with macrophages playing imperative roles in influencing the prognosis of CVDs. Autophagy is a conserved pathway that maintains cellular functions. Emerging evidence has revealed an intrinsic connection between autophagy and macrophage functions. This review focuses on the role and underlying mechanisms of autophagy-mediated regulation of macrophage plasticity in polarization, inflammasome activation, cytokine secretion, metabolism, phagocytosis, and the number of macrophages. In addition, autophagy has been shown to connect macrophages and heart cells. It is attributed to specific substrate degradation or signalling pathway activation by autophagy-related proteins. Referring to the latest reports, applications targeting macrophage autophagy have been discussed in CVDs, such as atherosclerosis, myocardial infarction, heart failure, and myocarditis. This review describes a novel approach for future CVD therapies.
Collapse
Affiliation(s)
- Wanqian Pan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jun Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lei Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yiyi Song
- Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Lianhua Han
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Mingyue Tan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yunfei Yin
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Tianke Yang
- Department of Ophthalmology, Eye Institute, Eye & ENT HospitalFudan UniversityShanghaiChina
- Department of OphthalmologyThe First Affiliated Hospital of USTC, University of Science and Technology of ChinaHefeiChina
| | - Tingbo Jiang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hongxia Li
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
11
|
Cui Y, Chen J, Zhang Z, Shi H, Sun W, Yi Q. The role of AMPK in macrophage metabolism, function and polarisation. J Transl Med 2023; 21:892. [PMID: 38066566 PMCID: PMC10709986 DOI: 10.1186/s12967-023-04772-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is a ubiquitous sensor of energy and nutritional status in eukaryotic cells. It plays a key role in regulating cellular energy homeostasis and multiple aspects of cell metabolism. During macrophage polarisation, AMPK not only guides the metabolic programming of macrophages, but also counter-regulates the inflammatory function of macrophages and promotes their polarisation toward the anti-inflammatory phenotype. AMPK is located at the intersection of macrophage metabolism and inflammation. The metabolic characteristics of macrophages are closely related to immune-related diseases, infectious diseases, cancer progression and immunotherapy. This review discusses the structure of AMPK and its role in the metabolism, function and polarisation of macrophages. In addition, it summarises the important role of the AMPK pathway and AMPK activators in the development of macrophage-related diseases.
Collapse
Affiliation(s)
- Yinxing Cui
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Junhua Chen
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Zhao Zhang
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Houyin Shi
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Weichao Sun
- Department of Bone Joint and Bone Oncology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China.
- The Central Laboratory, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China.
| | - Qian Yi
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
12
|
Chen F, Xue Q, He N, Zhang X, Li S, Zhao C. The association and application of sonodynamic therapy and autophagy in diseases. Life Sci 2023; 334:122215. [PMID: 37907152 DOI: 10.1016/j.lfs.2023.122215] [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: 08/30/2023] [Revised: 10/09/2023] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
Sonodynamic therapy (SDT) is a new non-invasive treatment method proposed based on photodynamic therapy (PDT). It has advantages such as high precision, strong tissue penetration, minimal side effects, and good patient compliance. With the maturation of nanomedicine, the application of nanosonosensitizers has further propelled the development of SDT. In recent years, people have developed many new types of sonosensitizers and explored the mechanisms of SDT. Among them, the studies about the relationship between autophagy and SDT have attracted increasing attention. After the SDT, cells usually undergo autophagy as a self-protective mechanism to resist external stimuli and reduce cell damage, which is beneficial for the treatment of atherosclerosis (AS), diabetes, and myocardial infarction but counterproductive in cancer treatment. However, under certain treatment conditions, excessive upregulation of autophagy can also promote cell death, which is beneficial for cancer treatment. This article reviews the latest research progress on the relationship between SDT and autophagy in cancers, AS, diabetes, and myocardial infarction. We also discuss and propose the challenges and prospects in enhancing SDT efficacy by regulating autophagy, with the hope of promoting the development of this promising therapeutic approach.
Collapse
Affiliation(s)
- Fang Chen
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qingwen Xue
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xuehui Zhang
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
| | - Cheng Zhao
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
13
|
Li D, Zhao B, Zhuang P, Mei X. Development of nanozymes for promising alleviation of COVID-19-associated arthritis. Biomater Sci 2023; 11:5781-5796. [PMID: 37475700 DOI: 10.1039/d3bm00095h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has been identified as a culprit in the development of a variety of disorders, including arthritis. Although the emergence of arthritis following SARS-CoV-2 infection may not be immediately discernible, its underlying pathogenesis is likely to involve a complex interplay of infections, oxidative stress, immune responses, abnormal production of inflammatory factors, cellular destruction, etc. Fortunately, recent advancements in nanozymes with enzyme-like activities have shown potent antiviral effects and the ability to inhibit oxidative stress and cytokines and provide immunotherapeutic effects while also safeguarding diverse cell populations. These adaptable nanozymes have already exhibited efficacy in treating common types of arthritis, and their distinctive synergistic therapeutic effects offer great potential in the fight against arthritis associated with COVID-19. In this comprehensive review, we explore the potential of nanozymes in alleviating arthritis following SARS-CoV-2 infection by neutralizing the underlying factors associated with the disease. We also provide a detailed analysis of the common therapeutic pathways employed by these nanozymes and offer insights into how they can be further optimized to effectively address COVID-19-associated arthritis.
Collapse
Affiliation(s)
- Dan Li
- Department of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, China.
| | - Baofeng Zhao
- Liaoning Provincial Key Laboratory of Medical Testing, Jinzhou Medical University, Jinzhou, 121001, China.
| | - Pengfei Zhuang
- Department of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, China.
| | - Xifan Mei
- Liaoning Provincial Key Laboratory of Medical Testing, Jinzhou Medical University, Jinzhou, 121001, China.
| |
Collapse
|
14
|
Wu G, Yu G, Zheng M, Peng W, Li L. Recent Advances for Dynamic-Based Therapy of Atherosclerosis. Int J Nanomedicine 2023; 18:3851-3878. [PMID: 37469455 PMCID: PMC10352141 DOI: 10.2147/ijn.s402678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 05/06/2023] [Indexed: 07/21/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease, which may lead to high morbidity and mortality. Currently, the clinical treatment strategy for AS is administering drugs and performing surgery. However, advanced therapy strategies are urgently required because of the deficient therapeutic effects of current managements. Increased number of energy conversion-based organic or inorganic materials has been used in cancer and other major disease treatments, bringing hope to patients with the development of nanomedicine and materials. These treatment strategies employ specific nanomaterials with specific own physiochemical properties (external stimuli: light or ultrasound) to promote foam cell apoptosis and cholesterol efflux. Based on the pathological characteristics of vulnerable plaques, energy conversion-based nano-therapy has attracted increasing attention in the field of anti-atherosclerosis. Therefore, this review focuses on recent advances in energy conversion-based treatments. In addition to summarizing the therapeutic effects of various techniques, the regulated pathological processes are highlighted. Finally, the challenges and prospects for further development of dynamic treatment for AS are discussed.
Collapse
Affiliation(s)
- Guanghao Wu
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, People’s Republic of China
| | - Guanye Yu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, 200072, People’s Republic of China
| | - Meiling Zheng
- Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 101121, People’s Republic of China
| | - Wenhui Peng
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, 200072, People’s Republic of China
| | - Lei Li
- National Clinical Research Center for Obstetric & Gynecologic Diseases, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, People’s Republic of China
| |
Collapse
|
15
|
Chen J, Ma H, Meng Y, Liu Q, Wang Y, Lin Y, Yang D, Yao W, Wang Y, He X, Li P. Analysis of the mechanism underlying diabetic wound healing acceleration by Calycosin-7-glycoside using network pharmacology and molecular docking. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154773. [PMID: 36990011 DOI: 10.1016/j.phymed.2023.154773] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/27/2023] [Accepted: 03/14/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Diabetic wounds represent a severe clinical challenge in which impaired M2 macrophage polarization and continuous macrophage glycolysis play crucial roles. Calycosin-7-glucoside (CG) is an isoflavone component in Astragali Radix (AR), which has become a research focus for treating diabetic wounds following reports indicating that it has anti-inflammatory effects. However, the mechanism through which CG can treat diabetic wounds is yet to be deciphered. PURPOSE This study aimed to evaluate the therapeutic effect of CG on diabetic wounds and its underlying mechanism. METHODS The potential mechanism underlying the treatment of diabetic wounds by CG was screened using bioinformatics. The therapeutic effects of CG were then investigated using a db/db diabetic wound model. Moreover, an LPS- and IFN-γ-induced RAW264.7 cell inflammation model was used to elucidate the mechanism underlying the therapeutic effects of CG against diabetic wounds. RESULTS Network pharmacology predicted that the AMPK pathway could be the main target through which CG treats diabetic wounds. In db/db diabetic mice, CG could accelerate wound healing and promote granulation tissue regeneration. Protein chip technology revealed that CG increased the production of M-CSF, G-CSF, GM-CSF, IL-10, IL-13, and IL-4 but not that of MCP-1, IL-1β, IL-1α, TNF-α, and TNF-RII. Moreover, CG elevated the proportion of Ly6CLo/- anti-inflammatory monocytes in peripheral blood and M2 macrophages in the wound. The ELISA and flow cytometry analyses revealed that CG enhanced the levels of IL-10, VEGF, CD206, and Arg-1 expression whereas it considerably reduced the levels of IL-1, IL-6, IL-12, TNF-α, CD86, and iNOS expression. Meanwhile, CG increased the macrophage mitochondrial membrane potential and decreased the mitochondrial ADP/ATP ratio and glycolysis rate of M1 macrophages through the ROS/AMPK/STAT6 pathway. CONCLUSIONS The network pharmacology and molecular dockin identified the AMPK pathway as a critical pathway for treating diabetic wounds using topical CG application. CG was found to promote anti-inflammatory monocyte recruitment and decrease the mitochondrial glycolysis rate to induce M2 macrophage polarization via the ROS/AMPK/STAT6 pathway. These results suggest that CG might be a promising therapeutic agent for diabetic wounds.
Collapse
Affiliation(s)
- Jia Chen
- Beijing University of Chinese Medicine, Beijing 100105, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Huike Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Yujiao Meng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Qingwu Liu
- Beijing University of Chinese Medicine, Beijing 100105, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Yan Lin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Danyang Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Wentao Yao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Yazhuo Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Xiujuan He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China.
| | - Ping Li
- Beijing University of Chinese Medicine, Beijing 100105, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China.
| |
Collapse
|
16
|
Theofilis P, Oikonomou E, Tsioufis K, Tousoulis D. The Role of Macrophages in Atherosclerosis: Pathophysiologic Mechanisms and Treatment Considerations. Int J Mol Sci 2023; 24:ijms24119568. [PMID: 37298518 DOI: 10.3390/ijms24119568] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Atherosclerotic diseases are a leading cause of morbidity and mortality worldwide, despite the recent diagnostic and therapeutic advances. A thorough understanding of the pathophysiologic mechanisms is thus essential to improve the care of affected individuals. Macrophages are crucial mediators of the atherosclerotic cascade, but their role has not been fully elucidated. The two main subtypes, tissue-resident and monocyte-derived macrophages, have distinct functions that contribute to atherosclerosis development or regression. Since polarization of macrophages to an M2 phenotype and induction of macrophage autophagy have been demonstrated to be atheroprotective, targeting these pathways could represent an appealing approach. Interestingly, macrophage receptors could act as drug targets, as seen in recent experimental studies. Last but not least, macrophage-membrane-coated carriers have been investigated with encouraging results.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- First Department of Cardiology, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Evangelos Oikonomou
- Third Department of Cardiology, Thoracic Diseases General Hospital "Sotiria", University of Athens Medical School, 11527 Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Dimitris Tousoulis
- First Department of Cardiology, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece
| |
Collapse
|
17
|
Yang Y, Sheng J, Sheng Y, Wang J, Zhou X, Li W, Kong Y. Lapachol treats non-alcoholic fatty liver disease by modulating the M1 polarization of Kupffer cells via PKM2. Int Immunopharmacol 2023; 120:110380. [PMID: 37244116 DOI: 10.1016/j.intimp.2023.110380] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/08/2023] [Accepted: 05/21/2023] [Indexed: 05/29/2023]
Abstract
AIM This study investigated the mechanism of action of lapachol (LAP) against non-alcoholic fatty liver disease (NAFLD). METHODS Primary Kupffer cells (KCs) of rats were used for in-vitro experiments. The proportion of M1 cells was assayed by flow cytometry, the levels of M1 inflammatory markers were determined by enzyme-linked immunosorbent assay (ELISA) combined with real-time quantitative fluorescence PCR (RT-qPCR), the expression of p-PKM2 was detected by Western-Blotting. A SD rat model of NAFLD was established with high-fat diet. Following LAP intervention, the changes in blood glucose/lipid, insulin resistance and liver function were detected, and the hepatic histopathologic changes were examined by histological staining. RESULTS The results showed that LAP could inhibit the M1 polarization of KCs, lower the levels of inflammatory cytokines, and suppress the activation of PKM2. The effect of LAP could be counteracted after using PKM2 inhibitor PKM2-IN-1 or knocking out PKM2. Small molecule docking revealed that LAP could inhibit the phosphorylation process of PKM2 by binding to ARG-246, the phosphorylation site of PKM2. In rat experiments, LAP could ameliorate the liver function and lipid metabolism of NAFLD rats, and inhibit the hepatic histopathologic changes. CONCLUSION Our study found that LAP can inhibit the phosphorylation of PKM2 by binding to PKM2-ARG-246, thereby regulating the M1 polarization of KCs and inhibiting the inflammatory response of liver tissues to treat NAFLD. LAP has potential as a novel pharmaceutical for treating NAFLD.
Collapse
Affiliation(s)
- Yi Yang
- The Second Affiliated Hospital of Jiaxing University, 314001, China
| | - Jian Sheng
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| | - Yongjia Sheng
- The Second Affiliated Hospital of Jiaxing University, 314001, China
| | - Jin Wang
- The Second Affiliated Hospital of Jiaxing University, 314001, China
| | - Xiaohong Zhou
- The Second Affiliated Hospital of Jiaxing University, 314001, China
| | - Wenyan Li
- The Second Affiliated Hospital of Jiaxing University, 314001, China
| | - Yun Kong
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| |
Collapse
|
18
|
Lyons N, Giri R, Begun J, Clark DA, Proud D, He Y, Hooper J, Kryza T. Reactive oxygen species as mediators of disease progression and therapeutic response in colorectal cancer. Antioxid Redox Signal 2023. [PMID: 36792932 DOI: 10.1089/ars.2022.0127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Significance Reactive oxygen species (ROS) are critical to normal cellular function with redox homeostasis achieved by balancing ROS production with removal through detoxification mechanisms. Many of the conventional chemotherapies used to treat colorectal cancer (CRC) derive a proportion of their cytotoxicity from ROS generation and resistance to chemotherapy is associated with elevated detoxification mechanisms. Furthermore, cancer stem cells demonstrate elevated detoxification mechanisms making definitive treatment with existing chemotherapy challenging. In this article we review the roles of ROS in normal and malignant colonic cell biology and how existing and emerging therapies might harness ROS for therapeutic benefit. Recent advances Recent publications have elucidated the contribution of ROS to the cytotoxicity of conventional chemotherapy alongside the emerging approaches of photodynamic therapy (PDT), sonodynamic therapy (SDT) and radiodynamic therapy (RDT) in which ROS are generated in response to excitatory light, sound or X-ray stimuli to promote cancer cell apoptosis. Critical issues The majority of patients with metastatic CRC have a very poor prognosis with 5-year survival of approximately 13% making the need for new or more effective treatments an imperative. Future Directions Modulation of ROS through a combination of new and emerging therapies may improve the efficacy of current chemotherapy providing novel approaches to treat otherwise resistant disease.
Collapse
Affiliation(s)
- Nicholas Lyons
- The University of Queensland, 1974, Mater Research, Woolloongabba, Queensland, Australia;
| | - Rabina Giri
- The University of Queensland, 1974, Mater Research, Woolloongabba, Queensland, Australia;
| | - Jakob Begun
- The University of Queensland, 1974, Mater Research, Woolloongabba, Queensland, Australia;
| | - David A Clark
- Royal Brisbane and Women's Hospital, 3883, Surgery, Herston, Queensland, Australia;
| | - David Proud
- Austin Hospital, 96043, Surgery, Heidelberg, Victoria, Australia;
| | - Yaowu He
- The University of Queensland, 1974, Mater Research, Woolloongabba, Queensland, Australia;
| | - John Hooper
- The University of Queensland, 1974, Mater Research, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, Australia, 4102;
| | - Thomas Kryza
- The University of Queensland, 1974, Mater Research, Woolloongabba, Queensland, Australia;
| |
Collapse
|
19
|
Xu Y, Dong Z, Zhang R, Wang Z, Shi Y, Liu M, Yang J, Yang T, Zhang R, Wang T, Zhang J, Zhang Y, Xiang F, Han Y, Wu J, Miao Z, Chen Q, Li Q, Wang Z, Tian Y, Guo Y. Sonodynamic therapy reduces cardiomyocyte apoptosis through autophagy activated by reactive oxygen species in myocardial infarction. Free Radic Biol Med 2023; 195:36-46. [PMID: 36529292 DOI: 10.1016/j.freeradbiomed.2022.12.080] [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: 10/19/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Myocardial infarction (MI) is lethal to patients because of acute ischemia and hypoxia leading to cardiac tissue apoptosis. Autophagy played a key role in MI through affecting the survival of cardiomyocytes. LncRNA-MHRT (myosin heavy-chain-associated RNA transcripts) was specific to the heart and cardiomyocytes, and inhibition of lncRNA-MHRT transcription under pathological stimuli could cause cardiac hypertrophy and even heart failure (HF). Sonodynamic therapy (SDT) is a new and developing medical technique that utilizes low-intensity ultrasound to locally activate a preloaded sonosensitizer. Our group previously reported that SDT could regulate autophagy. In this study, we investigated whether SDT could reduce MI-induced cardiomyocyte apoptosis via activating autophagy pathway. SDT improved cardiac function and suppresses MI-induced cardiomyocyte apoptosis. SDT alleviated MI-induced cardiomyocyte apoptosis by improving autophagy. MHRT mediated the inhibiting effect of SDT on cardiomyocyte apoptosis via activating autophagy pathway. Our data reveal a novel effect that SDT protects against MI and confirm that SDT reduces MI-induced cardiomyocyte apoptosis via activating MHRT-mediated-autophagy. Thus, our findings also indicate that SDT may be used as a potential method for treatment of post-myocardial infarction heart failure.
Collapse
Affiliation(s)
- Yingjie Xu
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zengxiang Dong
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Rongzhen Zhang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zeng Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuanqi Shi
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Mingyu Liu
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiemei Yang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tao Yang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | | | - Tengyu Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jingyu Zhang
- Department of Geriatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Shenzhen Qianhai Henkou Free Trade Zone Hospital, Shenzhen, China
| | - Yu Zhang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Fei Xiang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yingjun Han
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiawen Wu
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhihan Miao
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Qiuyu Chen
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Qi Li
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zeyao Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China; The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China; Department of Pathophysiology and Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China.
| | - Yuanyuan Guo
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China; Department of Geriatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
20
|
Shen H, He Z, Pei H, Zhai L, Guan Q, Wang G. Aurantiamide promotes M2 polarization of microglial cells to improve the cognitive ability of mice with Alzheimer's disease. Phytother Res 2023; 37:101-110. [PMID: 36062448 DOI: 10.1002/ptr.7597] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 01/19/2023]
Abstract
This work aimed to investigate the effect of aurantiamide (Aur) in promoting the M2 polarization of microglial cells to improve the cognitive ability of mice with Alzheimer's disease (AD). The M2 polarization of BV2 cells was induced by interleukin-4 (IL-4) treatment.Aur promoted the M2 polarization of BV2 cells, and up-regulated the expression of CD206 and SOCS3. In the meantime, it increased TGF-β1, Arg-1 and IL-10 levels, and promoted the polarization of JAK1-STAT6. Treatment with STAT6 inhibitor antagonized the effect of Aur. Besides, the cognitive ability of AD mice was improved after Aur treatment, meanwhile, the expression of CD206 was up-regulated, while that of IBA-1 was down-regulated. Aur promotes the M2 polarization of microglial cells to improve the cognitive ability of AD mice, and such effect is related to the STAT6 signal.
Collapse
Affiliation(s)
- Heping Shen
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhongmei He
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Hongyan Pei
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Liping Zhai
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qiaobing Guan
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Genghuan Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
21
|
Tian Y, Sheng S, Gao W, Yao J, Tian Y. Sonodynamic therapy suppresses matrix collagen degradation in vulnerable atherosclerotic plaque by modulating caspase 3 - PEDF/HIF-1α - MMP-2/MMP-9 signaling in macrophages. PLoS One 2022; 17:e0279191. [PMID: 36574366 PMCID: PMC9794047 DOI: 10.1371/journal.pone.0279191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 12/01/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The rupture of vulnerable atherosclerotic plaque is the main cause of acute ischemic vascular events, and is characterized by pathological degradation of matrix collagen in the fibrous cap. In a previous study, we reported that 5-aminolevulinic acid-mediated sonodynamic therapy suppressed collagen degradation in rabbit plaque. However, the underlying molecular mechanism has yet to be fully elucidated. METHODS We applied sinoporphyrin sodium-mediated sonodynamic therapy (DVDMS-SDT) to balloon-denuded rabbit and apolipoprotein E-deficient (ApoE-/-) mouse models to observe collagen content in plaque. Cultured human THP-1 and mouse peritoneal macrophage-derived foam cells were used for in vitro mechanistic studies. RESULTS We observed that DVDMS-SDT decreased plaque area and increased the percentages of collagen and smooth muscle cells and reduced the percentage of macrophages in rabbit and ApoE-/- mouse advanced plaques. In vitro, DVDMS-SDT modulated the caspase 3-pigment epithelium-derived factor/hypoxia-inducible factor-1α (PEDF/HIF-1α)-matrix metalloprotease-2/9 (MMP-2/MMP-9) signaling in macrophage foam cells. CONCLUSIONS Our findings show that DVDMS-SDT effectively inhibits matrix collagen degradation in advanced atherosclerotic plaque by modulating caspase 3-PEDF/HIF-1α-MMP-2/MMP-9 signaling in macrophage foam cells and therefore represents a suitable and promising clinical regimen to stabilize vulnerable plaques.
Collapse
Affiliation(s)
- Yanfeng Tian
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, China
| | - Siqi Sheng
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, China
| | - Weiwei Gao
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, China
| | - Jianting Yao
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, China
- * E-mail: , (YT); (JY)
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, China
- The Second Affiliated Hospital of Southern University of Science and Technology (The Third People’s Hospital of Shenzhen), Southern University of Science and Technology, Shenzhen, China
- * E-mail: , (YT); (JY)
| |
Collapse
|
22
|
Fang F, Xiao C, Li C, Liu X, Li S. Tuning macrophages for atherosclerosis treatment. Regen Biomater 2022; 10:rbac103. [PMID: 36683743 PMCID: PMC9845526 DOI: 10.1093/rb/rbac103] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease and a leading cause of death worldwide. Macrophages play an important role in inflammatory responses, cell-cell communications, plaque growth and plaque rupture in atherosclerotic lesions. Here, we review the sources, functions and complex phenotypes of macrophages in the progression of atherosclerosis, and discuss the recent approaches in modulating macrophage phenotype and autophagy for atherosclerosis treatment. We then focus on the drug delivery strategies that target macrophages or use macrophage membrane-coated particles to deliver therapeutics to the lesion sites. These biomaterial-based approaches that target, modulate or engineer macrophages have broad applications for disease therapies and tissue regeneration.
Collapse
Affiliation(s)
- Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Crystal Xiao
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Chunli Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
23
|
Wang S, Gao Z, Yang Y, Zhang Q, Huang J, Wang B, Lei S, Tan Q, Liu D, Guo L, Song Y, Liu J, Ma T, Tian Y. Sonodynamic Therapy With Concentric Ultrasound Imaging Array for Precision Theranostics for Atherosclerotic Plaque. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:3270-3283. [PMID: 36269912 DOI: 10.1109/tuffc.2022.3215436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Atherosclerotic cardiovascular disease is a major cause of human disability and mortality. Our previous study demonstrated the safety and efficacy of sonodynamic therapy (SDT) on atherosclerotic plaques. However, traditional single-element therapeutic transducer has single acoustic field, and positioning therapeutic and imaging transducers in the same position is difficult during ultrasound imaging-guided SDT. Continuously changing the position of transducers to intervene lesions in different positions is required, increasing the difficulty of treatment. Thus, an SDT device with precise theranostics is required. Therefore, we designed and fabricated a "concentric ultrasound transducer for theranostics" (CUST-T), comprising a central 8-MHz linear array transducer for ultrasound imaging, and a peripheral 1-MHz hollow two-dimensional (2-D) planar array transducer for generating phased-array focused ultrasound (PAFUS). The CUST-T exhibited high imaging resolution at a distance of up to 20 mm from the transducer and could generate a personalized complex PAFUS acoustic field to match various lesions. In vitro biomedical results showed that PAFUS-SDT induced RAW264.7-derived foam cell apoptosis leading to a targeting field apoptotic rate 4.36-6.24 times that of the nontargeting field and the significant apoptotic region was consistent with the PAFUS acoustic field. In vivo, PAFUS-SDT guided by ultrasound imaging significantly increased the lumen area ( ) and collagen level ( ), whereas the wall thickness ( ) and lipid content ( ) of rabbit femoral artery were reduced. In conclusion, CUST-T provided image guidance sufficient for accurate SDT for atherosclerotic plaques in peripheral arteries and could be applied in clinical practice.
Collapse
|
24
|
Lu L, Wang T, Fang C, Song L, Qian C, Lv Z, Fang Y, Liu X, Yu X, Xu X, Su C, Chen F, Zhang K. Oncolytic Impediment/Promotion Balance Disruption by Sonosensitizer-Free Nanoplatforms Unfreezes Autophagy-Induced Resistance to Sonocatalytic Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:36462-36472. [PMID: 35939287 DOI: 10.1021/acsami.2c09443] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Autophagy as a double-edged sword features an oncolytic impediment/promotion balance, which manipulates tumor progression. From this perspective, a sonosensitizer-free targeting oncolytic nanoplatform (SFTON) consisting of chloroquine (CQ) and porphyrin-structured metal centers (PMCS) was engineered to break this balance for enhancing antitumor activity. Porphyrin structure retention in a ZIF-8-derived hydrophobic carbon skeleton retained high stability and high sonocatalytic activity, and the hydrophobic carbon skeleton capable of adsorbing air provided cavitation nuclei for further elevating sonocatalytic activity. More significantly, the encapsulated CQ as the autophagy inhibitor reprogrammed autophagy, terminated the autophagy-induced self-protection or self-detoxification, and unfroze the resistances to reactive oxygen species (ROS) therapy associated with ROS accumulation and ROS activity. Systematic experiments reveal the action principles and validate that the induced apoptosis and blockaded autophagosome escalation into the autolysosome were two activated pathways to magnify the antitumor sonocatalytic therapy. Contributed by these actions, the SFTON-unlocked oncolytic impediment/promotion balance disruption strategy acquired considerable antitumor outcomes in vivo and in vitro against liver tumor progression, especially after combining with AS1411-mediated active targeting. This impediment/promotion balance disruption enabled by the SFTON can serve as a general method to elevate ROS-based antitumor activity.
Collapse
Affiliation(s)
- Lu Lu
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
- Department of Medical Ultrasound, Affiliated Hospital of Guangdong Medical University, No. 57 Peoples Avenue, Zhanjiang 524000, Guangdong Province, P. R. China
| | - Taixia Wang
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Chao Fang
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Li Song
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Cheng Qian
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Zheng Lv
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Yujia Fang
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Xinyu Liu
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Xin Yu
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Xiaohong Xu
- Department of Medical Ultrasound, Affiliated Hospital of Guangdong Medical University, No. 57 Peoples Avenue, Zhanjiang 524000, Guangdong Province, P. R. China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Fubo Chen
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Kun Zhang
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| |
Collapse
|
25
|
Kim SW, Oh SA, Seol SI, Davaanyam D, Lee JK. Cytosolic HMGB1 Mediates LPS-Induced Autophagy in Microglia by Interacting with NOD2 and Suppresses Its Proinflammatory Function. Cells 2022; 11:cells11152410. [PMID: 35954253 PMCID: PMC9368039 DOI: 10.3390/cells11152410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/29/2022] Open
Abstract
The high mobility group box 1 (HMGB1), a well-known danger-associated molecule pattern (DAMP) molecule, is a non-histone chromosomal protein localized in the nucleus under normal physiological conditions. HMGB1 exhibits diverse functions depending on its subcellular location. In the present study, we investigated the role of HMGB1-induced autophagy in the lipopolysaccharide (LPS)-treated BV2 microglial cell line in mediating the transition between the inflammatory and autophagic function of the nucleotide-binding oligomerization domain-containing 2 (NOD2), a cytoplasmic pattern-recognition receptor. The induction of the microtubule-associated protein 1 light chain 3 (LC3), an autophagy biomarker, was detected slowly in BV2 cells after the LPS treatment, and peak induction was detected at 12 h. Under these conditions, NOD2 level was significantly increased and the binding between HMGB1 and NOD2 and between HMGB1 and ATG16L1 was markedly enhanced and the temporal profiles of the LC3II induction and HMGB1-NOD2 and HMGB1-ATG16L1 complex formation coincided with the cytosolic accumulation of HMGB1. The LPS-mediated autophagy induction was significantly suppressed in BV2 cells after HMGB1 or NOD2 knock-down (KD), indicating that HMGB1 contributes to NOD2-mediated autophagy induction in microglia. Moreover, NOD2-RIP2 interaction-mediated pro-inflammatory cytokine induction and NF-κB activity were significantly enhanced in BV2 cells after HMGB1 KD, indicating that HMGB1 plays a critical role in the modulation of NOD2 function between pro-inflammation and pro-autophagy in microglia. The effects of the cell-autonomous pro-autophagic pathway operated by cytoplasmic HMGB1 may be beneficial, whereas those from the paracrine pro-inflammatory pathway executed by extracellularly secreted HMGB1 can be detrimental. Thus, the overall functional significance of HMGB1-induced autophagy is different, depending on its temporal activity.
Collapse
Affiliation(s)
- Seung-Woo Kim
- Department of Biomedical Sciences, Inha University School of Medicine, Inchon 22212, Korea
| | - Sang-A Oh
- Department of Anatomy, Inha University School of Medicine, Incheon 22212, Korea
| | - Song-I Seol
- Department of Anatomy, Inha University School of Medicine, Incheon 22212, Korea
| | - Dashdulam Davaanyam
- Department of Anatomy, Inha University School of Medicine, Incheon 22212, Korea
| | - Ja-Kyeong Lee
- Department of Anatomy, Inha University School of Medicine, Incheon 22212, Korea
- Correspondence: ; Tel.: +82-32-860-9893
| |
Collapse
|
26
|
Su Y, Sun X, Liu X, Qu Q, Yang L, Chen Q, Liu F, Li Y, Wang Q, Huang B, Huang XH, Zhang XJ. hUC-EVs-ATO reduce the severity of acute GVHD by resetting inflammatory macrophages toward the M2 phenotype. J Hematol Oncol 2022; 15:99. [PMID: 35864538 PMCID: PMC9306027 DOI: 10.1186/s13045-022-01315-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/06/2022] [Indexed: 01/22/2023] Open
Abstract
Background Both extracellular vesicles from mesenchymal stromal cell-derived human umbilical cords (hUC-EVs) and arsenic trioxides (ATOs) have been demonstrated to treat acute graft-versus-host disease (aGVHD) via immunomodulation. Apart from immunomodulation, hUC-EVs have a unique function of drug delivery, which has been proposed to enhance their efficacy. In this study, we first prepared ATO-loaded hUC-EVs (hUC-EVs-ATO) to investigate the therapeutic effect and potential mechanisms of hUC-EVs-ATO in a mouse model of aGVHD after allogeneic hematopoietic stem cell transplantation (HSCT). Methods An aGVHD model was established to observe the therapeutic effects of hUC-EVs-ATO on aGVHD. Target organs were harvested for histopathological analysis on day 14 after transplantation. The effects of hUC-EVs-ATO on alloreactive CD4+ were evaluated by flow cytometry in vivo and in vitro. Flow cytometry, RT-PCR, immunofluorescence colocalization analysis and Western blot (Wb) analysis were performed to examine macrophage polarization after hUC-EV-ATO treatment. The cytokines in serum were measured by a cytometric bead array (CBA). TEM, confocal microscopy and Wb were performed to observe the level of autophagy in macrophages. A graft-versus-lymphoma (GVL) mouse model was established to observe the role of hUC-EVs-ATO in the GVL effect. Results The clinical manifestations and histological scores of aGVHD in the hUC-EVs-ATO group were significantly reduced compared with those in the ATO and hUC-EVs groups. The mice receiving hUC-EVs-ATO lived longer than the control mice. Notably, hUC-EVs-ATO interfering with alloreactive CD4+ T cells differentiation were observed in aGVHD mice but not in an in vitro culture system. Additional studies showed that depletion of macrophages blocked the therapeutic effects of hUC-EVs-ATO on aGVHD. Mechanistically, hUC-EVs-ATO induced autophagic flux by inhibiting mammalian target of rapamycin (mTOR) activity to repolarize M1 to M2 macrophages. Additionally, using a murine model of GVL effects, hUC-EVs-ATO were found not only to reduce the severity of aGVHD but also to preserve the GVL effects. Taken together, hUC-EVs-ATO may be promising candidates for aGVHD treatment. Conclusions hUC-EVs-ATO enhanced the alleviation of aGVHD severity in mice compared with ATO and hUC-EVs without weakening GVL activity. hUC-EVs-ATO promoted M1 to M2 polarization via the mTOR-autophagy pathway. hUC-EVs-ATO could be a potential therapeutic approach in aGVHD after allo-HSCT. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01315-2.
Collapse
Affiliation(s)
- Yan Su
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xueyan Sun
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiao Liu
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qingyuan Qu
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Liping Yang
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qi Chen
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Fengqi Liu
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Yueying Li
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
| | - Qianfei Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
| | - Bo Huang
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Hui Huang
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China. .,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China. .,Collaborative Innovation Center of Hematology, Peking University, Beijing, China. .,National Clinical Research Center for Hematologic Disease, Beijing, China.
| | - Xiao-Jun Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, China. .,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China. .,Collaborative Innovation Center of Hematology, Peking University, Beijing, China. .,National Clinical Research Center for Hematologic Disease, Beijing, China.
| |
Collapse
|
27
|
Chen W, Xiao W, Liu X, Yuan P, Zhang S, Wang Y, Wu W. Pharmacological manipulation of macrophage autophagy effectively rejuvenates the regenerative potential of biodegrading vascular graft in aging body. Bioact Mater 2022; 11:283-299. [PMID: 34977432 PMCID: PMC8668428 DOI: 10.1016/j.bioactmat.2021.09.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/28/2021] [Accepted: 09/22/2021] [Indexed: 12/25/2022] Open
Abstract
Declined regenerative potential and aggravated inflammation upon aging create an inappropriate environment for arterial regeneration. Macrophages are one of vital effector cells in the immune microenvironment, especially during biomaterials mediated repairing process. Here, we revealed that the macrophage autophagy decreased with aging, which led to aggravated inflammation, thereby causing poor vascular remodeling of artificial grafts in aging body. Through loading the autophagy-targeted drugs, rapamycin and 3-MA (3-methyladenine), in PCL (polycaprolactone) sheath of the PGS (poly glycerol sebacate) - PCL vascular graft, the essential role of macrophage autophagy was confirmed in regulating macrophage polarization and biomaterial degradation. Moreover, the utilization of rapamycin promoted anti-inflammatory polarization of macrophage by activating autophagy, which further promoted myogenic differentiation of vascular progenitor cells and accelerated endothelialization. Our study elucidated the contribution of pharmacological manipulation of macrophage autophagy in promoting regeneration of small caliber artery, which may pave a new avenue for clinical translation of vascular grafts in aging body.
Collapse
Affiliation(s)
- Wanli Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 399 Middle Yanchang Road, Shanghai 200072, China
| | - Weiwei Xiao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xuzheng Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pingping Yuan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Siqian Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yinggang Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wei Wu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
28
|
Hua Y, Zhang J, Liu Q, Su J, Zhao Y, Zheng G, Yang Z, Zhuo D, Ma C, Fan G. The Induction of Endothelial Autophagy and Its Role in the Development of Atherosclerosis. Front Cardiovasc Med 2022; 9:831847. [PMID: 35402552 PMCID: PMC8983858 DOI: 10.3389/fcvm.2022.831847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/08/2022] [Indexed: 12/29/2022] Open
Abstract
Increasing attention is now being paid to the important role played by autophagic flux in maintaining normal blood vessel walls. Endothelial cell dysfunction initiates the development of atherosclerosis. In the endothelium, a variety of critical triggers ranging from shear stress to circulating blood lipids promote autophagy. Furthermore, emerging evidence links autophagy to a range of important physiological functions such as redox homeostasis, lipid metabolism, and the secretion of vasomodulatory substances that determine the life and death of endothelial cells. Thus, the promotion of autophagy in endothelial cells may have the potential for treating atherosclerosis. This paper reviews the role of endothelial cells in the pathogenesis of atherosclerosis and explores the molecular mechanisms involved in atherosclerosis development.
Collapse
Affiliation(s)
- Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Zhang
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianqian Liu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Su
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guobin Zheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhihui Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Danping Zhuo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
29
|
miRNA-29a inhibits atherosclerotic plaque formation by mediating macrophage autophagy via PI3K/AKT/mTOR pathway. Aging (Albany NY) 2022; 14:2418-2431. [PMID: 35288486 PMCID: PMC8954956 DOI: 10.18632/aging.203951] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 02/28/2022] [Indexed: 11/25/2022]
Abstract
Background: miR-29a plays a vital role in AS, but the relationship between the miR-29a-targeted PI3K signaling pathway and AS remains unclear. Therefore, this study was carried out. Methods: Gene expression profiles from the GEO database containing AS samples were analyzed. ApoE−/− mice and RAW264.7 cells were treated with miR-29a negative control (NC), miR-29a mimic and miR-29a inhibitor to establish the AS model. Then MOVAT staining, TEM, Western blotting, and immunofluorescence staining were adopted for testing target proteins. Results: DEGs were identified from GSE137578, GSE132651, GSE113969, GSE43292, and GSE97210 datasets. It was found that there were targeted binding sites between miR-29a and PIK3CA. Besides, GO and KEGG analysis demonstrated that autophagy was an enriched pathway in AS. Later, PPI network was depicted, and hub genes were then determined. The results revealed that miR-29a suppressed the areas of plaques and lesional macrophages, but had no impact on VSMCs. TEM results showed the organelles pyknosis of lesional macrophages damaged morphological changes. Furthermore, miR-29a amplified the M2-like macrophages but suppressed the polarization of M1-like macrophages in atherosclerotic plaques. According to mouse and RAW 264.7 cell experiments, miR-29a significantly inhibited the protein expressions of PI3K, p-PI3K, p-AKT, and p-mTOR, which were consistent with the increased expressions of autophagy-related proteins, Beclin 1 and LC3II. However, the miR-29a suppression exhibited the contrary results. Conclusion: MiR-29a elevation induces the increase of autophagy by down-regulating the PI3K/AKT/mTOR pathway in the progression of AS, indicating that miR-29a is a novel therapeutic strategy for AS.
Collapse
|
30
|
A novel therapeutic strategy for atherosclerosis: autophagy-dependent cholesterol efflux. J Physiol Biochem 2022; 78:557-572. [DOI: 10.1007/s13105-021-00870-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/25/2021] [Indexed: 10/19/2022]
|
31
|
Zhu Y, Ruan S, Shen H, Guan Q, Zhai L, Yang Y. Oridonin regulates the polarized state of Kupffer cells to alleviate nonalcoholic fatty liver disease through ROS-NF-κB. Int Immunopharmacol 2021; 101:108290. [PMID: 34717194 DOI: 10.1016/j.intimp.2021.108290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 10/20/2022]
Abstract
Oridonin (Ori) is a kind of diterpenoid small molecule, but its role in nonalcoholic fatty liver disease (NAFLD) has not been reported yet. This study aimed to explore the pharmacological function of Ori in liver protection through the reactive oxygen species (ROS)-mediated polarization of Kupffer cells (KCs). In the present work, KCs were adopted for study in vitro. To be specific, LPS and IFN-γ were utilized to induce M1 polarization, then the influence of Ori intervention on the expression of inflammatory factors IL-1β, IL-6 and TNF-α was detected by enzyme-linked immunosorbent assay (ELISA), that of CD86 and P65 was measured through fluorescence staining, that of p-P65 and p-P50 was detected by Western blotting (WB) assay, and ROS expression was measured by using the DCFH-DA probe. The C57BL/6J mice were fed with the high fat diet (HFD) to construct the NAFLD model, and intervened with Ori. The blood glucose (BG), body weight (BW), food intake and water intake of mice were monitored; meanwhile, glucose and insulin tolerance tests were conducted. The liver tissues of mice were subjected to H&E staining and oil red O staining. Moreover, the serum ALT, AST and TG levels in mice were monitored, the CD86 and CD206 levels were measured through histochemical staining, the expression of inflammatory factors was detected by ELISA, and the p-P65 and p-P50 protein levels were detected by WB assay. Ori suppressed the M1 polarization of KCs, reduced the levels of inflammatory factors, and decreased the expression of ROS, p-P65 and p-P50. In animal experiments, Ori improved lipid deposition and liver injury in the liver tissues of NAFLD mice, increased the proportion of M2 cells (up-regulated CD206 expression), reduced that of M1 cells (down-regulated CD86 expression), and decreased the serum ALT, AST and TG levels. This study discovered that Ori suppressed ROS production and regulated the M1 polarization of KCs, thus protecting the liver in NAFLD.
Collapse
Affiliation(s)
- Yu Zhu
- Department of critical medicine, The Second Affiliated Hospital of Jiaxing University, China
| | - Shuiliang Ruan
- Department of Gastroenterology, The Second Affiliated Hospital of Jiaxing University, China
| | - Heping Shen
- Department of pharmacy, The Second Affiliated Hospital of Jiaxing University, China
| | - Qiaobing Guan
- Department of pharmacy, The Second Affiliated Hospital of Jiaxing University, China
| | - Liping Zhai
- Department of pharmacy, The Second Affiliated Hospital of Jiaxing University, China.
| | - Yi Yang
- Department of pharmacy, The Second Affiliated Hospital of Jiaxing University, China.
| |
Collapse
|
32
|
Abstract
Low-intensity ultrasound-triggered sonodynamic therapy (SDT) is a promising noninvasive therapeutic modality due to its strong tissue penetration ability. In recent years, with the development of nanotechnology, nanoparticle-based sonosensitizer-mediated SDT has been widely investigated. With the increasing demand for precise and personalized treatment, abundant novel sonosensitizers with imaging capabilities have been developed for determining the optimal therapeutic window, thus significantly enhancing treatment efficacy. In this review, we focus on the molecular imaging-guided SDT. The prevalent mechanisms of SDT are discussed, including ultrasonic cavitation, sonoluminescence, reactive oxygen species, and mechanical damage. In addition, we introduce the major molecular imaging techniques and the design principles based on nanoparticles to achieve efficient imaging. Furthermore, the imaging-guided SDT for the treatment of cancer, bacterial infections, and vascular diseases is summarized. The ultimate goal is to design more effective imaging-guided SDT modalities and provide novel ideas for clinical translation of SDT.
Collapse
Affiliation(s)
- Juan Guo
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Xueting Pan
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Chaohui Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| |
Collapse
|
33
|
Song L, Zhang J, Lai R, Li Q, Ju J, Xu H. Chinese Herbal Medicines and Active Metabolites: Potential Antioxidant Treatments for Atherosclerosis. Front Pharmacol 2021; 12:675999. [PMID: 34054550 PMCID: PMC8155674 DOI: 10.3389/fphar.2021.675999] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a complex chronic disease that occurs in the arterial wall. Oxidative stress plays a crucial role in the occurrence and progression of atherosclerotic plaques. The dominance of oxidative stress over antioxidative capacity generates excess reactive oxygen species, leading to dysfunctions of the endothelium and accelerating atherosclerotic plaque progression. Studies showed that Chinese herbal medicines and traditional Chinese medicine (TCM) might regulate oxidative stress; they have already been used to treat diseases related to atherosclerosis, including stroke and myocardial infarction. This review will summarize the mechanisms of oxidative stress in atherosclerosis and discuss studies of Chinese herbal medicines and TCM preparations treating atherosclerosis, aiming to increase understanding of TCM and stimulate research for new drugs to treat diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Luxia Song
- Graduate School, Beijing University of Chinese Medicine, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Runmin Lai
- Graduate School, Beijing University of Chinese Medicine, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiuyi Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianqing Ju
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
34
|
Wei Y, Liang M, Xiong L, Su N, Gao X, Jiang Z. PD-L1 induces macrophage polarization toward the M2 phenotype via Erk/Akt/mTOR. Exp Cell Res 2021; 402:112575. [PMID: 33771483 DOI: 10.1016/j.yexcr.2021.112575] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/04/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
PD-L1 (programmed death-ligand 1) is the ligand of PD-1 (programmed cell death protein 1) and regulates inhibitory immune responses. It is well known that PD-L1 suppresses T cell function via binding to PD-1. However, little is known about the role of the PD-1/PD-L1 axis in macrophage polarization. According to previous studies, the function of the PD-1/PD-L1 axis in macrophage polarization is controversial, and the underlying mechanism has not been fully elucidated. Thus, we treated THP-1-derived macrophages with human PD-L1 Fc to determine the role of the PD-1/PD-L1 axis in macrophage polarization. To further explore the mechanism, we performed RNA sequencing and used specific inhibitors to identify the implicated signalling pathways. In this study, we found that PD-L1 induces the upregulation of CD206 expression, which is inhibited by nivolumab, LY294002, U0126, and rapamycin. Evaluation of differentially expressed genes (DEGs) and bioinformatics analysis indicated that PD-L1 also induces the upregulation of the expression of genes that maintain mitochondrial function and mediate metabolic switching. In addition, we did not detect PD-L1-induced CD86 alterations, indicating that PD-L1 treatment has no significant influence on M1 polarization. Taken together, these results suggest that PD-L1 binds to PD-1 and promotes M2 polarization accompanied by mitochondrial function enhancement and metabolic reprogramming via Erk/Akt/mTOR. This study elucidates the role of PD-L1 in macrophage polarization and verifies the underlying mechanisms for the first time. Considering that aberrantly upregulated PD-L1 expression contributes to a wide variety of diseases, targeting PD-L1-mediated macrophage polarization is a prospective therapeutic strategy for both neoplastic and nonneoplastic diseases.
Collapse
Affiliation(s)
- Yi Wei
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mengjun Liang
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liping Xiong
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ning Su
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Gao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zongpei Jiang
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
35
|
Zhang X, Qin Y, Wan X, Liu H, Lv C, Ruan W, He L, Lu L, Guo X. Rosuvastatin exerts anti-atherosclerotic effects by improving macrophage-related foam cell formation and polarization conversion via mediating autophagic activities. J Transl Med 2021; 19:62. [PMID: 33568202 PMCID: PMC7877030 DOI: 10.1186/s12967-021-02727-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/28/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Atherosclerosis is a chronic vascular disease posing a great threat to public health. We investigated whether rosuvastatin (RVS) enhanced autophagic activities to inhibit lipid accumulation and polarization conversion of macrophages and then attenuate atherosclerotic lesions. METHODS All male Apolipoprotein E-deficient (ApoE-/-) mice were fed high-fat diet supplemented with RVS (10 mg/kg/day) or the same volume of normal saline gavage for 20 weeks. The burden of plaques in mice were determined by histopathological staining. Biochemical kits were used to examine the levels of lipid profiles and inflammatory cytokines. The potential mechanisms by which RVS mediated atherosclerosis were explored by western blot, real-time PCR assay, and immunofluorescence staining in mice and RAW264.7 macrophages. RESULTS Our data showed that RVS treatment reduced plaque areas in the aorta inner surface and the aortic sinus of ApoE-/- mice with high-fat diet. RVS markedly improved lipid profiles and reduced contents of inflammatory cytokines in the circulation. Then, results of Western blot showed that RVS increased the ratio LC3II/I and level of Beclin 1 and decreased the expression of p62 in aortic tissues, which might be attributed to suppression of PI3K/Akt/mTOR pathway, hinting that autophagy cascades were activated by RVS. Moreover, RVS raised the contents of ABCA1, ABCG1, Arg-1, CD206 and reduced iNOS expression of arterial wall, indicating that RVS promoted cholesterol efflux and M2 macrophage polarization. Similarly, we observed that RVS decreased lipids contents and inflammatory factors expressions in RAW264.7 cells stimulated by ox-LDL, accompanied by levels elevation of ABCA1, ABCG1, Arg-1, CD206 and content reduction of iNOS. These anti-atherosclerotic effects of RVS were abolished by 3-methyladenine intervention. Moreover, RVS could reverse the impaired autophagy flux in macrophages insulted by chloroquine. We further found that PI3K inhibitor LY294002 enhanced and agonist 740 Y-P weakened the autophagy-promoting roles of RVS, respectively. CONCLUSIONS Our study indicated that RVS exhibits atheroprotective effects involving regulation lipid accumulation and polarization conversion by improving autophagy initiation and development via suppressing PI3K/Akt/mTOR axis and enhancing autophagic flux in macrophages.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yating Qin
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoning Wan
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Liu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chao Lv
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weibin Ruan
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin He
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiaotong University, Shanghai, China.
| | - Li Lu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xiaomei Guo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
36
|
Wang L, Liu S, Pan B, Cai H, Zhou H, Yang P, Wang W. The role of autophagy in abdominal aortic aneurysm: protective but dysfunctional. Cell Cycle 2020; 19:2749-2759. [PMID: 32960711 PMCID: PMC7714418 DOI: 10.1080/15384101.2020.1823731] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/17/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Autophagy, an evolutionarily conserved mechanism that promotes cell survival by recycling nutrients and degrading long-lived proteins and dysfunctional organelles, is an important defense mechanism, and its attenuation has been well documented in senescence and aging-related diseases. Abdominal aortic aneurysm (AAA), a well-known aging-related disease, has been defined as a chronic degenerative process in the abdominal aortic wall; however, the complete mechanism is unknown, and a clinical treatment is lacking. Accumulating evidence has recently revealed that numerous drugs that can induce autophagy are effective in the treatment of AAA. The purpose of this systematic review was to focus on the cross-talk between autophagy and high-risk factors and the potential pathogenesis of AAA to understand not only the host defense and pathogenesis but also potential treatments.
Collapse
Affiliation(s)
- Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Baihong Pan
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huoying Cai
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pu Yang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
37
|
Shan R, Liu N, Yan Y, Liu B. Apoptosis, autophagy and atherosclerosis: Relationships and the role of Hsp27. Pharmacol Res 2020; 166:105169. [PMID: 33053445 DOI: 10.1016/j.phrs.2020.105169] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a multifactorial chronic inflammatory disease of the arterial wall, and an important pathological basis of coronary heart disease. Endothelial cells, vascular smooth muscle cells, and macrophages play important roles in the development of atherosclerosis. Of note, apoptosis and autophagy, two types of programmed cell death, influence the development and progression of atherosclerosis via the modulation of such cells. The small heat shock protein Hsp27 is a multifunctional protein induced by various stress factors and has a protective effect on cells. A large number of studies have demonstrated that Hsp27 plays an important role in regulating apoptosis. Recently, some studies have suggested that Hsp27 also participates in the autophagic process. Moreover, Hsp27 is closely related to the occurrence and development of atherosclerosis. Here, we summarize the molecular mechanisms of apoptosis and autophagy and discuss their effects on endothelial cells, vascular smooth muscle cells, and macrophages in the context of atherosclerotic procession. We further explore the involvement of Hsp27 in apoptosis, autophagy, and atherosclerosis. We speculate that Hsp27 may exert its anti-atherosclerotic role via the regulation of apoptosis and autophagy; this may provide the basis for the development of new approaches for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Ruiting Shan
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Ning Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Youyou Yan
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|