1
|
Liu R, Dai L, Jia S, Geng S, Niu Y, Chen J, Dong C, Li C, Shi Y, Wang X, Zhang J, Zhao N, Gao Z, Yang X, Gao S. Fut8 regulated Unc5b hyperfucosylation reduces macrophage emigration and accelerates atherosclerosis development via the ferroptosis pathway. Free Radic Biol Med 2025; 235:1-14. [PMID: 40262667 DOI: 10.1016/j.freeradbiomed.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
The accumulation of foam cells in the arterial walls is a defining characteristic of atherosclerosis. Enhancing their migration from plaques may represent a key strategy for slowing disease progression. Recent studies suggest that fucosyltransferase 8 (Fut8) impairs macrophage migration from the intima by modifying the Unc5b membrane receptor, thereby influencing the development of atherosclerosis. This study investigated the roles of Fut8 and Unc5b in foam cell migration using ApoE-/- mouse and foam cell models, employing techniques such as western blotting, mitochondrial function assays, wound healing experiments, and immunofluorescence staining. The findings indicate that Fut8 upregulation increases P53 expression and reduces SLC7A11 and GPX4 levels, leading to altered intracellular concentrations of GSH and Fe2+, impaired mitochondrial function, and reduced migration capacity, all of which promote atherosclerosis. These mechanisms are closely associated with ferroptosis. Intervention with N-acetylcysteine (NAC) and buthionine sulfoximine (BSO) demonstrated that NAC mitigates oxidative stress and migration inhibition, induced by oxidized low-density lipoprotein (ox-LDL). Additionally, inhibiting ferroptosis slowed the progression of atherosclerosis in ApoE-/- mice. Together, these results highlight that Fut8 exacerbates atherosclerosis through a P53/SLC7A11-mediated enhancement of ferroptosis in foam cells, offering a novel perspective on the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Rujin Liu
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Lina Dai
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Sihui Jia
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010110, PR China; Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, PR China.
| | - Shijia Geng
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Yan Niu
- Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Jie Chen
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Chongyang Dong
- Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Chenlei Li
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Yuanjia Shi
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Xiaomeng Wang
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Jing Zhang
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Ningxia Zhao
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Zhanfeng Gao
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Xi Yang
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Shang Gao
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| |
Collapse
|
2
|
Wang X, Xie Z, Zhang J, Chen Y, Li Q, Yang Q, Chen X, Liu B, Xu S, Dong Y. Interaction between lipid metabolism and macrophage polarization in atherosclerosis. iScience 2025; 28:112168. [PMID: 40201117 PMCID: PMC11978336 DOI: 10.1016/j.isci.2025.112168] [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] [Indexed: 04/10/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory condition associated with lipid deposition. The interaction between abnormal lipid metabolism and the inflammatory response has been identified as the underlying cause of AS. Lipid metabolism disorders are considered the basis of atherosclerotic lesion formation and macrophages are involved in the entire process of AS formation. Macrophages have a high degree of plasticity, and the change of their polarization direction can determine the progress or regression of AS. The disturbances in bioactive lipid metabolism affect the polarization of different phenotypes of macrophages, thus, affecting lipid metabolism and the expression of key signal factors. Therefore, understanding the interaction between lipid metabolism and macrophages as well as their key targets is important for preventing and treating AS and developing new drugs. Recent studies have shown that traditional Chinese medicines play a positive role in the prevention and treatment of AS, providing a basis for clinical individualized treatment.
Collapse
Affiliation(s)
- Xinge Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zheng Xie
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jing Zhang
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Chen
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qi Li
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xu Chen
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Bing Liu
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Dong
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
3
|
Wacker BK, Bi L, Saenz-Pipaon G, Sanford N, Regan AZ, Lim NS, Liu L, Kim F, Dichek DA. Overexpression of ABCA1 in Carotid Endothelium of Hyperlipidemic Rabbits Modulates Vascular Inflammation. Hum Gene Ther 2025; 36:750-764. [PMID: 40111153 DOI: 10.1089/hum.2024.166] [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] [Indexed: 03/22/2025] Open
Abstract
Endothelial activation and dysfunction are key early steps in atherogenesis. Vascular gene therapy targeting endothelial inflammation and cholesterol accumulation could decrease atherosclerosis progression. ATP-binding cassette subfamily A member 1 (ABCA1) exhibits anti-inflammatory properties and promotes cholesterol efflux. A mouse model showed that systemic endothelial overexpression of ABCA1 decreased diet-induced atherosclerosis. To test if local ABCA1 endothelial overexpression protects against atherosclerosis, we used helper-dependent adenoviral vectors (HDAd) to express ABCA1 or a "Null" control in the carotid endothelium of hyperlipidemic rabbits. Both ABCA1 mRNA and endothelial protein were increased 3 days after vector infusion. After 24 weeks on a high-fat diet, laser-microdissected endothelium showed increased ABCA1 mRNA expression, but whole-vessel ABCA1 mRNA was decreased with HDAdABCA1. Endothelial ABCA1 protein could not be measured at 24 weeks, so its overexpression may be transient. CD68 expression was decreased (-23%, p < 0.001), but ITGAM (-15%, p = 0.3) was unchanged. Macrophage markers for both M1-like macrophages (IL1B: -44% [p = 0.02]; IL6: -40% [p = 0.02]; CCL2: -25% [p = 0.02]) and M2-like macrophages (ARG1: -27% [p = 0.03]; IL10: -23% [p = 0.09]; TGFB1: -13% [p < 0.001]) were also decreased. The inflammatory cytokines IL6 (-100%; p < 0.001) and TNF (p < 0.05) were significantly decreased in the laser-microdissected endothelium, but VCAM1 (+5%, p = 1.0) was unchanged and ICAM1 (+101%; p = 0.03) increased. Lesion size, intimal lipid, and intimal macrophage content were all unchanged (p > 0.5 for all), and vascular cholesterol measured by mass spectrometry (-11%; p = 0.9) also showed no difference. There was a small decrease in the intimal/medial ratio. scRNAseq revealed that vector transcripts were not restricted to endothelial cells after 24+ weeks but were detected in most cell types. The exception was modulated smooth muscle cells, which were found in substantial numbers in larger lesions. Overall, transient overexpression of ABCA1 in the vascular endothelium subtly alters the expression of inflammatory markers, providing only a modest atheroprotection.
Collapse
Affiliation(s)
- Bradley K Wacker
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Lianxiang Bi
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Goren Saenz-Pipaon
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Nicole Sanford
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Abigail Z Regan
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Natalie S Lim
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Li Liu
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Francis Kim
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - David A Dichek
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
4
|
Zhen Y, Li D, Meng Y, Xing Z, Zheng J. Unveiling the roles of HIPK2 in atherosclerosis: Insights into the β-catenin/STAT1 signaling cascade and the involvement of SENP1. Biochem Pharmacol 2025; 237:116911. [PMID: 40164339 DOI: 10.1016/j.bcp.2025.116911] [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: 11/29/2024] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Atherosclerosis is a disorder of lipid metabolism, but its pathogenesis has not yet been fully elucidated. This study aimed to clarify the roles of homeodomain interacting protein kinase 2 (HIPK2) in atherosclerosis. Atherosclerotic model was constructed by feeding apolipoprotein E knockout (ApoE-/-) mice with a high-fat diet. Human THP-1 macrophages and mouse RAW 264.7 macrophages were stimulated with IFN-γ to establish an in vitro model. We showed an upregulation of HIPK2 in the aorta of atherosclerotic mice. HIPK2 knockdown reduced macrophage infiltration, M1 polarization, and attenuates atherosclerosis development. Downregulation of HIPK2 in macrophages led to a significant suppression in the expression of pro-inflammatory factors, which was accompanied by an enhancement in the phosphorylation and degradation of β-catenin, as well as the activation of the signal transducer and activator of transcription 1 (STAT1) signaling pathway. Silencing of HIPK2 alone in THP-1 macrophages resulted in anti-inflammatory effects and suppression of M1 macrophage polarization. However, simultaneous silencing of HIPK2 and β-catenin (CTNNB1) reversed these effects, counteracting the outcomes observed with HIPK2 silencing alone. We validated that small ubiquitin-like modifier (SUMO)-specific peptidase 1 (SENP1) regulated HIPK2 function by affecting the SUMOylation of HIPK2 at the K32 site. SENP1 knockdown promoted HIPK2 SUMOylation, impairing its protein stability. In the rescue experiments, IFN-γ-induced inflammation and M1 polarization were resumed upon restoration of HIPK2 expression in SENP1-silenced macrophages. Our work demonstrated that HIPK2 accelerated the progression of atherosclerosis by regulating β-catenin/STAT1 signaling cascade to promote macrophage infiltration and M1 polarization. HIPK2 was regulated by SENP1-mediated de-SUMOylation.
Collapse
Affiliation(s)
- Yanhua Zhen
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Dongdong Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Yulu Meng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Zeyu Xing
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Jiahe Zheng
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China.
| |
Collapse
|
5
|
Ji X, Dong Q, Li W, Luo W, Zhou N, Li H, Yang X. The role of Morin in attenuating atherosclerosis via STAT1 pathway inhibition. Biochem Biophys Res Commun 2025; 754:151537. [PMID: 40022814 DOI: 10.1016/j.bbrc.2025.151537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Atherosclerotic cardiovascular diseases can lead to myocardial infarction and stroke, which are linked to elevated rates of mortality. Morin is a flavonoid compound that can be extracted from mulberries and possesses anti-inflammatory and antioxidant properties. The objective of this research is to elucidate Morin's impact on atherosclerosis. The ApoE-/- mice were divided into three groups: control group, HFD group and HFD + Morin group. The mice in control group received a normal diet (ND). To create an atherosclerosis model, ApoE-/- mice were subjected to a high-fat diet (HFD) for 8 weeks. The mice were assigned to two distinct categories at random based on whether Morin intervention was administered: one serving as the HFD group and the other as the HFD + Morin group. The mice received Morin for 4 weeks at a dosage of 50 mg/kg orally in the model + Morin group. Subsequently, ORO staining assay was performed to evaluate the formation of aortic plaques. ELISA was used to measure IFN-γ and TNF-α levels in plasma of the mice. In vitro, mouse macrophages RAW264.7 were cultured and treated with IFN-γ for 24 h, followed by Morin treatment for another 24 h. Western blotting was conducted to analyze changes in macrophage polarization markers CD86 and CD206, as well as P-STAT1 levels. DCFH-DA was used to detect changes in intracellular ROS levels. Subsequently, RAW264.7 cells were treated with the STAT1 inhibitor Lenvatinib to further investigate changes in CD86 and CD206, as well as ROS levels. In vivo data showed that Morin markedly diminished the size of aortic plaques and suppressed the secretion of IFN-γ and TNF-α. In vitro data indicated that Morin reduced M1 polarization and intracellular ROS levels through inhibiting the STAT1 pathway activation in RAW264.7 cells, ultimately suppressing inflammation.
Collapse
Affiliation(s)
- Xin Ji
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Collaborative Innovation Center for Eco-Environment, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China; Department of Clinical Laboratory, Southern University of Science and Technology Hospital, Shenzhen, 518000, Guangdong Province, China
| | - Qianqian Dong
- Department of Clincal Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, China
| | - Wanqiu Li
- Department of Clincal Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, China
| | - Wei Luo
- Department of Clinical Laboratory, Southern University of Science and Technology Hospital, Shenzhen, 518000, Guangdong Province, China
| | - Ning Zhou
- Department of Clinical Laboratory, Southern University of Science and Technology Hospital, Shenzhen, 518000, Guangdong Province, China
| | - Hanzhao Li
- Department of Clinical Laboratory, Southern University of Science and Technology Hospital, Shenzhen, 518000, Guangdong Province, China
| | - Xiaolong Yang
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Collaborative Innovation Center for Eco-Environment, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China.
| |
Collapse
|
6
|
Zhang X, Liu S, Wu K, Shu L, Li Y, Li L, Wang D. Structural characteization and anti-colorectal cancer activity of a fucogalactan purified from Ganoderma tsugae. Carbohydr Polym 2025; 352:123203. [PMID: 39843104 DOI: 10.1016/j.carbpol.2024.123203] [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: 11/07/2024] [Revised: 12/25/2024] [Accepted: 12/29/2024] [Indexed: 01/24/2025]
Abstract
Ganoderma tsugae, a traditional medicinal mushroom, exhibits anti-tumor properties; however, the effects of its polysaccharide on anti-colorectal cancer remain undetermined. Herein, a fucogalactan of Ganoderma tsugae (GTP-a2) was isolated and purified from its fruiting body. The molecular weight of GTP-a2 is 7.056 kDa, consisting of →6)-α-D-Galp-(1→ backbone with branches of α-L-Fucp-(1→, which is attached at C2. Subsequently, the anti-colorectal cancer activity and potential mechanism of GTP-a2 were investigated in azoxymethane/dextran sulfate sodium (AOM/DSS)-induced colitis-associated colorectal cancer (CAC) mice. GTP-a2 reduced colorectal tumor numbers and suppressed tumor development. Metabolite analysis of the colon revealed that GTP-a2 altered cancer-related metabolites, notably increasing ophiobolin A level. Combined with proteomics and biochemical detection data revealed that GTP-a2 regulated the levels of Aldh1a3 through the mammalian target of rapamycin (mTOR)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway in vivo and in vitro. Additionally, GTP-a2 regulated immune function by inhibiting macrophage polarization to M1-like phenotype. These results suggest the potential application of GTP-a2 as a therapeutic agent for CAC.
Collapse
Affiliation(s)
- Xin Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China.
| | - Shuai Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| | - Keyi Wu
- College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| | - Lili Shu
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China.
| | - Yu Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China; College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
7
|
Chen Y, Xu B, Lin Q, Zhu X, Lv Y, Bai X, Weng X, Du J, Li M, Zhu Y, Mou J, Wang M, Wang Y, Luo X, Xu C. Spermine delivered by ZIF90 nanoparticles alleviates atherosclerosis by targeted inhibition of macrophage ferroptosis in plaque. J Nanobiotechnology 2025; 23:165. [PMID: 40038689 DOI: 10.1186/s12951-025-03271-8] [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: 08/25/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Nowadays, emerging evidence have suggested that the ferroptosis of macrophages could contribute to the progression of atherosclerosis (AS). Meanwhile, Spermine (Sp) could serve as an endogenous small molecule exhibiting a wide range of cardiovascular protective effects. METHODS Zeolitic imidazolate framework-90 (ZIF90) nanoparticles were synthesized and utilized to create a novel delivery nanosystem encapsulated with Sp (CD16/32-ZIF90@Sp). The efficacy of CD16/32-ZIF90@Sp in protecting against AS and ferroptosis was evaluated in ApoE-/- mice and macrophages, with a focus on assessing potential adverse effects in vivo. RESULTS CD16/32-ZIF90@Sp exhibited reliable and stable delivery of Sp within acidic environments and ATP sensitivity. CD16/32-ZIF90@Sp effectively reduced the cytotoxicity of Sp. As is evidenced by in vitro and vivo experiments, CD16/32-ZIF90@Sp showed precise targeting of macrophages within atherosclerotic plaques and ox-LDL-activated macrophages. Furthermore, treatment with CD16/32-ZIF90@Sp effectively attenuated the progression of AS and the ferroptosis of macrophage within plaque in ApoE-/- mice without causing significant side effects. Mechanistically, we found that CD16/32-ZIF90@Sp inhibited ferroptosis via improving mitochondrial function and upregulating the expression level of GPX4/xCT. CONCLUSION Our study demonstrated that CD16/32-modified ZIF90 nanoparticles could effectively target macrophages within atherosclerotic plaques, leading to the inhibition of atherosclerotic plaque progression in ApoE-/- mice. These effects were attributed to the enhancement of mitochondrial function and the inhibition of macrophage ferroptosis, with limited side effects.
Collapse
Affiliation(s)
- Yuwu Chen
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Biyi Xu
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Quan Lin
- Department of Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Xinxin Zhu
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Ying Lv
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Xiaoxuan Bai
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Xiuzhu Weng
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Jie Du
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Man Li
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Yuxiao Zhu
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Junke Mou
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Mengyang Wang
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Yuehong Wang
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China.
- Department of Pathophysiology, Harbin Medical University, Harbin, 150001, China.
| | - Xing Luo
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China.
- Department of Pathophysiology, Harbin Medical University, Harbin, 150001, China.
| | - Changqing Xu
- Department of Pathophysiology, Harbin Medical University, Harbin, 150001, China
| |
Collapse
|
8
|
Yu W, Fu L, Lei G, Luo F, Yu P, Shen W, Wu Q, Yang P. Chemokine Ligands and Receptors Regulate Macrophage Polarization in Atherosclerosis: A Comprehensive Database Mining Study. CJC Open 2025; 7:310-324. [PMID: 40182401 PMCID: PMC11963153 DOI: 10.1016/j.cjco.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 11/18/2024] [Indexed: 04/05/2025] Open
Abstract
Background Atherosclerosis is a systemic disease involving multiple blood vessels and a major cause of cardiovascular disease. Current treatment methods (eg, statins) for atherosclerosis can reduce the risk of cardiovascular diseases effectively, but they are insufficient to completely reverse existing atherosclerosis. Macrophages play a central role in development of atherosclerosis. Chemokines, the main mediators of macrophage chemotaxis, are important in immune and inflammatory responses. The effects of chemokines on mechanisms involved in atherosclerosis are unknown. This study preliminarily investigated these effects and mechanisms via bioinformatics methods. Methods In this study, data on chemokine ligands and receptors were obtained by mining public databases (the National Center of Biotechnology Information-Gene Expression Omnibus [NCBI-GEO] database, ArrayExpress database, and single-cell RNA sequencing [scRNA-seq] database), and an extensive literature search was performed. The expression levels of chemokines in mouse tissues were analyzed via Metascape software for signalling pathway enrichment, scRNA-seq data for chemokine expression in atherosclerotic plaque progression and regression, and GEO2R data for chemokine expression during macrophage polarization. Ingenuity Pathway Analysis (IPA) software was used to analyze regulatory factors such as transcription factors and microRNAs that are significantly differentially expressed upstream of chemokines in macrophage polarization. Finally, a model of the chemokine regulation of atherosclerosis was established on the basis of these results. Results There are 5 main findings: (1) In atherosclerosis, chemokines are regulated by transcription factors and microRNAs. (2) The transcription factor STAT1 promotes the polarization of dormant (M0) macrophages into classically activated (M1) macrophages and alternative activated (M2) macrophages by regulating chemokines. The transcription factors STAT1, IRF7 and IRF1 regulate the polarization of M0 macrophages into M2a and M2b macrophages via different chemokines. For example, some transcription factors promote M1 polarization of M0 macrophages through CCL4, but M2 macrophage polarization is regulated via CCL19, CCL5 and CCR7. (3) Transcription factors can promote and inhibit, whereas miRNAs can only inhibit atherosclerosis. (4) CCL4 existed in all 5 different chemokine-regulated macrophage models, whereas CXCL3 only existed in the M2b macrophage transcriptional regulation model, indicating that CXCL3 may promote the M2b type macrophages polarization of M0 macrophages. (5) CCL5 and CCR7 can promote the M2a macrophages and M2b macrophages polarization of M0 macrophages. Conclusions Atherosclerosis can be treated by regulating chemokines and regulating the polarization of macrophages. The chemokines CCL4, CCL5, CCL8, CCL19, CXCL3, CXCL10, CXCL13, and CCR7 may play key roles in the progression and regression of atherosclerosis.
Collapse
Affiliation(s)
- Wanqian Yu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Linghua Fu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Guangtao Lei
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fan Luo
- Department of Gastroenterology, Jiangxi Provincial Hospital of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wen Shen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Cheng Y, Zhao A, Li Y, Li C, Miao X, Yang W, Wang Y. Roles of SIRT3 in cardiovascular and neurodegenerative diseases. Ageing Res Rev 2025; 104:102654. [PMID: 39755174 DOI: 10.1016/j.arr.2024.102654] [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: 11/20/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
Sirtuin-3 (SIRT3) in mitochondria has nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase activity. As such, SIRT3 is crucial in cardiovascular and neurodegenerative diseases. Advanced proteomics and transcriptomics studies have revealed that SIRT3 expression becomes altered when the heart or brain is affected by external stimuli or disease, such as diabetic cardiomyopathy, atherosclerosis, myocardial infarction, Alzheimer's disease, Huntington's disease, and Parkinson's disease. More specifically, SIRT3 participates in the development of these disorders through its deacetylase activity and in combination with downstream signaling pathways. The paper reviews SIRT3's expression changes, roles, and mechanisms associated with the development of cardiovascular and neurodegenerative diseases. Additionally, strategies targeting SIRT3 to treat or regulate cardiovascular and neurodegenerative disease development are discussed.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China; Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Anqi Zhao
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Ying Li
- Department of Medical Clinic, Jilin Women and Children Health Hospital, Changchun, Jilin, China
| | - Cheng Li
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiao Miao
- The Second Hosptial of Jilin University, Changchun, Jilin, China.
| | - Wanshan Yang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China.
| | - Yonggang Wang
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
10
|
Zhang X, Liu S, Kong F, Shu L, Li Y, Wang D, Li L. Acidic polysaccharide from Ganoderma tsugae: Structural characterization and antiatherosclerotic related to macrophage polarization. Food Res Int 2025; 203:115913. [PMID: 40022418 DOI: 10.1016/j.foodres.2025.115913] [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: 10/24/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 03/03/2025]
Abstract
Herein, a water-soluble Ganoderma tsugae acidic polysaccharide (GTP-2) was isolated and purified from the fruiting bodies of G. tsugae. GTP-2 has a molecular weight of 13.059 kDa, composed of →3)-β-d-Glcp-(1 → 3)-β-d-Glcp-(1 → 4)-GlcpUA-(1 → 4)-β-d-Glcp-1→ glucan backbone and branches ending with β-d-Glcp-(1 → 6)-β-d-Glcp-(1→, which is attached at C6 of →3,6)-β-d-Glcp-(1→. Subsequently, the antiatherosclerotic activity of GTP-2 was examined in apolipoprotein E deficient (ApoE-/-) mice fed with high-fat diet, and its potential mechanism of action was investigated. GTP-2 ameliorated blood lipid levels (total cholesterol, triglycerides, and low-density lipoprotein), while improving the serum levels of high-density lipoprotein. Furthermore, GTP-2 alleviated the atherosclerotic lesions and reduced levels of inflammatory cytokines. Analysis of the gut microbiota revealed that GTP-2 enhanced the abundance of beneficial bacteria (Lactobacillu and Akkermansia). The serum metabolite composition was further altered, with a significant reduction in octadecanoic acid level. GTP-2 regulated the nuclear factor kappa-B signaling pathway by inhibiting macrophage polarization to M1 phenotype. Collectively, these findings support the potential use of GTP-2 as an antiatherosclerotic therapy.
Collapse
Affiliation(s)
- Xin Zhang
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866 China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China.
| | - Shuai Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China; College of Plant Protection, Jilin Agricultural University, Changchun 130118 China.
| | - Fange Kong
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China; College of Plant Protection, Jilin Agricultural University, Changchun 130118 China.
| | - Lili Shu
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866 China.
| | - Yu Li
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866 China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China.
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China; College of Plant Protection, Jilin Agricultural University, Changchun 130118 China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China; College of Plant Protection, Jilin Agricultural University, Changchun 130118 China.
| |
Collapse
|
11
|
Wang Q, Cao Y, Jia L. Lipidomics-based investigation of its impact on the pathogenesis of coronary atherosclerosis: a Mendelian randomization study. Hereditas 2025; 162:13. [PMID: 39893493 PMCID: PMC11787735 DOI: 10.1186/s41065-025-00367-x] [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: 12/02/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Considerable attention has been devoted to investigating the association between lipid metabolites and cardiovascular diseases, particularly coronary atherosclerosis. METHODS A two-sample MR framework was used to investigate the relationship between lipid metabolites and the risk of coronary atherosclerosis. Two GWAS datasets were examined to take intersections of SNPs from 51,589 cases and 343,079 controls, and 14,334 cases and 346,860 controls to determine genetic susceptibility to coronary atherosclerosis. Random-effects inverse variance weighted (IVW) MR analyses were performed by a series of sensitivity assessments to measure the robustness of our findings and to detect any violations of MR assumptions. RESULTS Through IVW, MR-Egger and weighted median regression methods, we inferred that these six lipid metabolites: cholesterol levels, sterol ester (27:1/18:2) levels, triacylglycerol (52:4) levels, triacylglycerol (52:5) levels, diacylglycerol (18:1_18.2) levels, triacylglycerol (53:4), could directly impact the development of atherosclerosis. CONCLUSION In conclusion, our study comprehensively illustrates a causal relationship between lipid metabolites and the risk of coronary atherosclerosis. Furthermore, cholesterol levels, sterol ester (27:1/18:2) levels, triacylglycerol (52:4) levels, triacylglycerol (52:5) levels, diacylglycerol (18:1_18.2) levels, and triacylglycerol (53:4) levels are positively correlated with the risk of coronary atherosclerosis. These six lipid metabolites have the potential as new predictors of the risk of atherosclerosis, providing new insights into the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Qun Wang
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Ministry of Education of China, Liaoning University of Traditional Chinese Medicine, 79 Chongshan Road, Huanggu District, Shenyang, 110847, Liaoning Province, China
| | - Yuan Cao
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Ministry of Education of China, Liaoning University of Traditional Chinese Medicine, 79 Chongshan Road, Huanggu District, Shenyang, 110847, Liaoning Province, China
| | - Lianqun Jia
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Ministry of Education of China, Liaoning University of Traditional Chinese Medicine, 79 Chongshan Road, Huanggu District, Shenyang, 110847, Liaoning Province, China.
| |
Collapse
|
12
|
Liu WB, Dai J, Chen X, Du N, Hu J. Integrated Network Pharmacology and In-silico Approaches to Decipher the Pharmacological Mechanism of Dioscorea septemloba Thunb in Treating Gout and Its Complications. Comb Chem High Throughput Screen 2025; 28:74-88. [PMID: 37957901 DOI: 10.2174/0113862073258523231025095117] [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: 05/19/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Dioscorea septemloba Thunb. (DST) has demonstrated therapeutic potential in the treatment of gout and its associated complications. However, the underlying mechanisms of DST's pharmacological activity remain unclear. This study aims to investigate the pharmacological substances and network regulatory mechanisms of DST in treating gout and its complications using network pharmacology. METHODS According to ultra-high performance liquid chromatography coupled with hybrid quadrupole-Orbitrap mass spectrometry (UPLC-Q-Exactive Orbitrap-MS) data and Lipinski's rule of five, 24 bioactive phytochemicals from DST were identified. The targets of gout were retrieved from Gene Expression Omnibus (GEO), GeneCards, and DisGeNET databases, followed by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG pathway) enrichment analysis. The Cytoscape network analysis was used to identify the primary pathological pathways and key targets. Finally, LeDock was used for molecular docking to verify the active components of DST and their core target proteins. RESULTS DST contains several core active ingredients, such as tetrahydroimidazo[1,2-a]pyridine- 2,5-dione, diosgenin, beta-sitosterol, dioscorol B, montroumarin and 9,10-dihydro-5,7- dimethoxy-3,4-phenanthrenediol. Moreover, these active components were found to strongly bind to the key targets for treating gout and its complications, including HSP90AA1, STAT3, PTGS2, PPARG, MTOR, HIF1A, MMP9, ESR1, and TLR4. As a result, DST alleviates gout and its complications by inhibiting xanthine dehydrogenase (XDH) to reduce uric acid levels and regulating the HIF-1α, EZH2/STAT3, and COX-2/PPAR-γ pathways to reduce inflammation. Additionally, it also plays an analgesic role by regulating the neuroactive ligand-receptor interaction pathway and calcium ion signaling pathway. CONCLUSION This study has provided insights into the underlying mechanisms of DST in the treatment of gout and its complications, which could serve as a scientific foundation for its clinical translation.
Collapse
Affiliation(s)
- Wen-Bin Liu
- School of Pharmaceutical Engineering, Jiangsu Food & Pharmaceutical Science College, Jiangsu, Huai'an, 223003, China
| | - Jie Dai
- School of Pharmaceutical Engineering, Jiangsu Food & Pharmaceutical Science College, Jiangsu, Huai'an, 223003, China
| | - Xuan Chen
- School of Pharmaceutical Engineering, Jiangsu Food & Pharmaceutical Science College, Jiangsu, Huai'an, 223003, China
| | - Ning Du
- School of Pharmaceutical Engineering, Jiangsu Food & Pharmaceutical Science College, Jiangsu, Huai'an, 223003, China
| | - Jian Hu
- School of Pharmaceutical Engineering, Jiangsu Food & Pharmaceutical Science College, Jiangsu, Huai'an, 223003, China
| |
Collapse
|
13
|
Wang A, Wang C, Xuan B, Sun Y, Li B, Zhao Q, Yu R, Wang X, Zhu M, Wei J. The role of splicing events in the inflammatory response of atherosclerosis: molecular mechanisms and modulation. Front Immunol 2024; 15:1507420. [PMID: 39742258 PMCID: PMC11685076 DOI: 10.3389/fimmu.2024.1507420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by persistent inflammatory responses throughout all stages of its progression. Modulating these inflammatory responses is a promising avenue for the development of cardiovascular disease therapies. Splicing events modulate gene expression and diversify protein functionality, exerting pivotal roles in the inflammatory mechanisms underlying atherosclerosis. These insights may provide novel opportunities for developing anti-inflammatory therapies for this disease. This article systematically discusses the diverse splice variants and how splicing events impact the inflammatory response in atherosclerosis via endothelial cells, macrophages, and vascular smooth muscle cells, highlighting their underlying molecular mechanisms and implications. Furthermore, this study summarizes clinical evidence supporting splicing-related molecules as diagnostic biomarkers and therapeutic targets in atherosclerosis. Lastly, we outline the current challenges and future research directions concerning splicing events and inflammatory responses in atherosclerosis. This offers a novel perspective and evidence for formulating new therapeutic strategies aimed at lowering the risk of atherosclerosis.
Collapse
Affiliation(s)
- Aolong Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Chengzhi Wang
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Bihan Xuan
- College of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yanqin Sun
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Bin Li
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Henan Evidence-Based Medicine Center of Chinese Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qifei Zhao
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Rui Yu
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinlu Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Mingjun Zhu
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jingjing Wei
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
14
|
Gao H, Li J, Huang J, Jiang X. Screening and regulatory mechanism exploration of M1 macrophage polarization and efferocytosis-related biomarkers in coronary heart disease. Front Cardiovasc Med 2024; 11:1478827. [PMID: 39723414 PMCID: PMC11669322 DOI: 10.3389/fcvm.2024.1478827] [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: 08/11/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
Background Macrophage polarization and efferocytosis have been implicated in CHD. However, the underlying mechanisms remain elusive. This study aimed to identify CHD-associated biomarkers using transcriptomic data. Methods This study examined 74 efferocytosis-related genes (ERGs) and 17 M1 macrophage polarization-related genes (MRGs) across two CHD-relevant datasets, GSE113079 and GSE42148. Differential expression analysis was performed separately on each dataset to identify differentially expressed genes (DEGs1 and DEGs2). The intersection of upregulated and downregulated genes from both sets was then used to define the final DEGs. Subsequently, MRG and ERG scores were calculated within the GSE113079 dataset, followed by weighted gene co-expression network analysis (WGCNA) to identify key module genes. The overlap between these module genes and the DEGs yielded candidate biomarkers, which were further evaluated through machine learning, receiver operating characteristic (ROC) curve analysis, and expression profiling. These biomarkers were subsequently leveraged to explore immune infiltration patterns and to construct a molecular regulatory network. To further validate their expression, quantitative reverse transcriptase PCR (qRT-PCR) was performed on clinical CHD samples, confirming the relevance and expression patterns of these biomarkers in the disease. Results A total of 93 DEGs were identified by intersecting the upregulated and downregulated genes from DEGs1 and DEGs2. WGCNA of the MRG and ERG scores identified 15,936 key module genes in the GSE113079 dataset. Machine learning and ROC analysis highlighted four biomarkers: C5orf58, CTAG1A, ZNF180, and IL13RA1. Among these, C5orf58, and ZNF180 were downregulated in CHD cases, while CTAG1A and IL13RA1 was upregulated. qRT-PCR results validated these findings for C5orf58, CTAG1A, ZNF180, and IL13RA1 showed inconsistent expression trends. Immune infiltration analysis indicated IL13RA1 all had a positive correlation with M0 macrophage, while had a negative correlation with. NK cells activated. The molecular regulatory network displayed that GATA2 and YY1 could regulate CTAG1A and ZNF180. Conclusions These results suggest that C5orf58, CTAG1A, ZNF180, and IL13RA1 serve as biomarkers linking M1 macrophage polarization and efferocytosis to CHD, providing valuable insights for CHD diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Xiaojie Jiang
- Department of Cardiology, The First Hospital of Nanchang, Nanchang, China
| |
Collapse
|
15
|
Chen H, Wu H, Wang Q, Zhang H. IFIT2 mediates iron retention and cholesterol efflux in atherosclerosis. Int Immunopharmacol 2024; 142:113131. [PMID: 39276454 DOI: 10.1016/j.intimp.2024.113131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Abnormalities in iron and lipid metabolism are recognized as key contributors to atherosclerosis (AS). Therefore, this study proposes to characterize the biomarker related to iron and lipid metabolism in AS using bioinformatics, animal, and cell experiments. METHODS The limma package was utilized to identify differentially expressed genes (DEGs) in GSE70126 and GSE70619 datasets, and biomarkers were screened using enrichment analysis and PPI networks. IFIT2 was knocked down using shRNA lentivirus in a high fat diet (HFD)-induced APOE-/- AS model to investigate its effects of IFIT2 on the pathology, iron retention, and lipid accumulation. Iron storage-related and cholesterol efflux-related proteins were evaluated following exogenous modulation of IFIT2 expression in ox-LDL-induced foamy macrophages. RESULTS Compared to non-foamy macrophages from the aorta, 189 and 4152 DEGs were identified in foamy macrophages within the GSE70126 and GSE70619 datasets, respectively. Moreover, intersecting DEGs may modulate immune responses, cell adhesion, vascular permeability, and oxidative stress through NF-kappa B, Wnt, TNF and HIF-1 signaling pathways. Notably, IFIT2 was significantly upregulated in foamy macrophages and AS models. In vivo, IFIT2 co-localized with foamy macrophages, and its knockdown led to reductions in plasma lipid levels, plaque area, immune infiltration, iron retention, and lipid accumulation. In vitro, IFIT2 knockdown alleviated the ox-LDL-induced increase in iron storage-related proteins (Ferritin-L and Ferritin-H) and iron (Fe2+ and Fe3+) in foamy macrophages. Furthermore, IFIT2 knockdown reduced lipid accumulation and upregulated cholesterol efflux-related proteins (PPARγ, LXRα, ABCA1, and ABCG1) in foamy macrophages. CONCLUSION IFIT2 knockdown attenuates iron retention and lipid accumulation in AS plaques, and facilitated cholesterol efflux from foamy macrophages via the PPARγ/LXRα/ABCA1-ABCG1 pathway.
Collapse
Affiliation(s)
- Haoqiang Chen
- Department of Cardiovascular Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, Yunnan 650032, P.R.China
| | - Haiyan Wu
- Department of Cardiovascular Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, Yunnan 650032, P.R.China; Faculty of Life Science and Technology, Kunming University of Science and Technology, No.727 Jingming South Road, Kunming 650500, P.R.China
| | - Qian Wang
- Department of Cardiovascular Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, Yunnan 650032, P.R.China.
| | - Hong Zhang
- Department of Cardiovascular Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, Yunnan 650032, P.R.China.
| |
Collapse
|
16
|
Liang Y, Wang Y, Tan J, Shu J, Xu Y, Wang M, Yang S, Ma L. Homocysteine affects macrophage polarization by altering m6A methylation of scavenger receptors CD209 and CD163L1. Epigenetics 2024; 19:2437272. [PMID: 39627020 PMCID: PMC11622620 DOI: 10.1080/15592294.2024.2437272] [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: 07/02/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/08/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by fatty plaque deposits on artery walls. Elevated plasma homocysteine (Hcy) levels are an independent risk factor for atherosclerosis. Research on the mechanism by which Hcy promotes atherosclerosis has gradually turned to epigenetic inheritance, but the correlation between Hcy and m6A (N6-methyladenosine) modification has not been reported. In this study, MeRIP-seq was performed on macrophages and Hcy-treated macrophages. GO and KEGG analyses were used to perform functional analysis of differentially methylated genes. qRT-PCR and western blot were taken to determine the expression of CD209, CD163L1, proinflammatory, and anti-inflammatory factors. Flow cytometry was used to detect the proportion of M2 macrophages. The results showed that after Hcy treatment, the overall m6A methylation of macrophages was down-regulated, and 856 differential methylation peaks were annotated to 781 genes. These included CD209 and CD163L1, whose m6A methylation was inhibited after treatment with Hcy. In addition, mRNA and protein expressions of CD209 and CD163L1 were also inhibited after Hcy treatment. Overexpression of CD209 or CD163L1 prevents the Hcy-induced decrease in the proportion of M2 macrophages. This article identified changes in the modification level of m6A in macrophages by Hcy and revealed the possible mechanism by which Hcy induces macrophage polarization.
Collapse
Affiliation(s)
- Yu Liang
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Yongbo Wang
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Jia Tan
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Jingxuan Shu
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Ya Xu
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Mingyuan Wang
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Shengjun Yang
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Linna Ma
- Department of Medical Laboratory Technique, Kunming Medical University Haiyuan College, Kunming, Yunnan, P.R. China
| |
Collapse
|
17
|
Luo W, Meng J, Yu XH, Zhang ZZ, Wang G, He J. Indole-3-Carboxaldehyde Inhibits Inflammatory Response and Lipid Accumulation in Macrophages Through the miR-1271-5p/HDAC9 Pathway. J Cell Mol Med 2024; 28:e70263. [PMID: 39698913 DOI: 10.1111/jcmm.70263] [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: 08/10/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/20/2024] Open
Abstract
Indole-3-carboxaldehyde (ICA), a microbiota-derived tryptophan metabolite, has been reported to protect against atherosclerosis. However, the molecular mechanisms for its atheroprotective effect remain largely unknown. This study aimed to explore the influence of ICA on lipid accumulation and inflammatory response in THP-1 macrophage-derived foam cells. Our results showed that administration of ICA upregulated the expression of miR-1271-5p, ATP binding cassette transporter A1 (ABCA1) and ABCG1, downregulated histone deacetylase 9 (HDAC9) expression and inhibited macrophage lipid accumulation. ICA treatment also facilitated macrophage polarisation to the M2 phenotype and alleviated inflammatory response, as evidenced by decreased IL-6 levels and increased IL-10 levels. HDAC9 was identified as a direct target of miR-1271-5p. HDAC9 overexpression or miR-1271-5p knockdown decreased the effect of ICA on ABCA1 and ABCG1 expression as well as inflammatory response. Taken together, these results suggest that ICA can suppress lipid accumulation and mitigate inflammatory response in macrophages by activating the miR-1271-5p/HDAC9 signalling cascade, thereby providing new explanations for how ICA reduces atherosclerosis.
Collapse
Affiliation(s)
- Wei Luo
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jun Meng
- The First Affiliated Hospital, Department of Function, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zi-Zhen Zhang
- School of Medical and Pharmacological Technology, Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, China
| | - Gang Wang
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jin He
- The First Affiliated Hospital, Department of Function, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
18
|
Visarnta S, Ratisoontorn C, Panichuttra A, Sinpitaksakul P, Chantarangsu S, Dhanuthai K. Macrophage polarization in human periapical lesions in relation to histopathological diagnosis, clinical features and lesion volume: An ex vivo study. Int Endod J 2024; 57:1829-1847. [PMID: 39222032 DOI: 10.1111/iej.14138] [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: 12/19/2023] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
AIM To evaluate M1 and M2 macrophage polarization in radicular cysts and periapical granulomas through an immunohistochemical analysis and the correlation between macrophage polarization and histopathological diagnosis, clinical characteristics and lesion volume using cone-beam computed tomography. METHODOLOGY Periapical biopsies diagnosed as radicular cysts (n = 52) and periapical granulomas (n = 51) were analysed by immunohistochemical method. Teeth with periapical lesion with no history of root canal treatment (primary lesion) and lesions persistent to root canal treatment (persistent lesions) were included. Pathological diagnosis, patients' age, gender and clinical characteristics were obtained from treatment records. A cone-beam computed tomographic periapical volume index (CBCTPAVI) score was assigned to each periapical lesion based on the volume of the lesion. Immuno-expressions of CD68 and CD163 were quantified. The CD68/CD163 ratio was adopted to represent M1 or M2 macrophage polarization. Mann-Whitney U test was used to determine the different CD68/CD163 ratio between groups of radicular cyst and periapical granuloma. Spearman's correlation test was performed to assess the correlation between the CD68/CD163 ratio and lesion volume and CBCTPAVI score. RESULTS Radicular cysts and periapical granulomas had CD68/CD163 median of 2.05 (IQR = 1.33) and 1.26 (IQR = 0.81), respectively. A significantly higher CD68/CD163 ratio was observed in radicular cysts (p < .001). In contrast, periapical granulomas had significantly lower median of CD68/CD163 ratio. Larger lesions had a higher median of CD68/CD163 ratio, while smaller lesions had lower median of CD68/CD163 ratio (p = .007, rs = .262). CD68/CD163 ratio was significantly correlated with the CBCTPAVI score in the overall periapical lesions (p = .002, rs = .306). The higher CD68/CD163 ratio in larger lesions indicated a higher degree of M1 polarization compared to smaller lesions. Regarding the pathological diagnosis, there was a significant positive correlation between CBCTPAVI score and CD68/CD163 ratio in periapical granulomas (p < .001, rs = .453), whereas the negative correlation was observed for radicular cysts (p < .001, rs = -.471). CONCLUSIONS Periapical granulomas are characterized by a M2-dominant macrophage polarization, while radicular cysts have significantly higher M1 macrophages. The higher degree of M1 macrophage polarization was significantly correlated with larger volume and higher CBCTPAVI scores of overall periapical lesion and periapical granuloma.
Collapse
Affiliation(s)
- Supanant Visarnta
- Section of Endodontics, Department of Operative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chootima Ratisoontorn
- Section of Endodontics, Department of Operative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Anchana Panichuttra
- Section of Endodontics, Department of Operative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Phonkit Sinpitaksakul
- Department of Radiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Soranun Chantarangsu
- Department of Oral Pathology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Kittipong Dhanuthai
- Department of Oral Pathology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
19
|
Tian W, Qiu H, He Y, Zhang M, Pan X, Wang Y, Shi X, Wen C, Chen J. Qinghao-Biejia Herb Pair attenuates SLE atherosclerosis by regulating macrophage polarization via ABCA1/G1-mediated cholesterol efflux. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118545. [PMID: 39002826 DOI: 10.1016/j.jep.2024.118545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/29/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qinghao-Biejia herb pair (QB) is the core herb pair of "Jieduquyuziyin prescription" and is one of the commonly used herb pairs for the clinical treatment of systemic lupus erythematosus (SLE). Previous studies have shown that QB reduces the expression of inflammatory cytokines like IL-6 and TNF-α in the serum and kidney of MRL/lpr mice. Additionally, it inhibits the expression of TLR4 and MyD88 in the kidney and aorta and reduces the deposition of renal complement C3 and aortic plaque after treatment. These findings suggest that QB has a preventive and therapeutic effect on lupus rats. AIM OF THE STUDY This study sought to investigate the mechanisms underlying the anti-SLE combined with atherosclerosis activity of the Qinghao-Biejia herb pair. MATERIALS AND METHODS Drug targets for QB were identified using the HERB database, while targets associated with SLE and atherosclerosis were retrieved from the GeneCards database. The intersection of these drug and disease targets was then analyzed using a protein-protein interaction (PPI) network with GO and KEGG pathway enrichment analysis. In vivo, apolipoprotein E-deficient (ApoE-/-) mice were induced to develop SLE-AS by intraperitoneal injection of pristane and continued feeding of a high-fat diet. The changes in relevant indexes were observed after 12 weeks of gavage treatment with hydroxychloroquine, QB, Q (Qinghao alone), and B (Biejia alone). Bone marrow-derived macrophages from ApoE-/- mice and Raw 264.7 macrophages were used to explore the mechanisms of QB treatment. RESULTS The levels of inflammatory cytokines in serum and pathological liver changes in mice were improved to varying degrees in the treatment groups. Additionally, there was a reduction in aortic atheromatous plaque formation and some improvement in cholesterol efflux. Furthermore, QB suppressed the expression of inflammatory cytokines in M1 macrophages, suggesting a role in regulating macrophage polarization. CONCLUSION QB demonstrates clear efficacy for treating SLE-AS, and its therapeutic mechanism may involve the regulation of macrophage phenotypes by promoting cholesterol efflux.
Collapse
Affiliation(s)
- Weiyu Tian
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China
| | - Haonan Qiu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China
| | - Yuanfang He
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China
| | - Miao Zhang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China
| | - Xinyu Pan
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China
| | - Yiqi Wang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China
| | - Xiaowei Shi
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China
| | - Chengping Wen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China.
| | - Juan Chen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China; Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, China.
| |
Collapse
|
20
|
Ouyang X, Liu Z. Regulatory T cells and macrophages in atherosclerosis: from mechanisms to clinical significance. Front Immunol 2024; 15:1435021. [PMID: 39582868 PMCID: PMC11581946 DOI: 10.3389/fimmu.2024.1435021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024] Open
Abstract
Atherosclerosis is a complex pathological process, which causes diseases that threaten the health of an increasing number of people. Studies have found that the original view of lipid accumulation is not comprehensive because the use of lipid-lowering drugs alone cannot effectively treat atherosclerosis. As the study of the pathogenesis of atherosclerosis develops in-depth, the impact of immune-inflammatory response on atherosclerosis has garnered a great deal of attention. Some new advances have been made in the role of regulatory T cells (Tregs) and macrophages with unique immunomodulatory functions in atherosclerosis. Herein, the role of Tregs, macrophages, the mechanisms of Tregs-regulated macrophages, and the effects of potential factors on Tregs and macrophages in atherosclerosis are overviewed. Targeting Tregs and macrophages may provide new research strategies for the treatment of atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Xin Ouyang
- Clinical Medical College, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zhongyong Liu
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
21
|
Zhang L, Li J, Kou Y, Shen L, Wang H, Wang Y, Ma R, Wu T, Yang X, Gu Y, Yi L. Mechanisms and treatment of atherosclerosis: focus on macrophages. Front Immunol 2024; 15:1490387. [PMID: 39569201 PMCID: PMC11576186 DOI: 10.3389/fimmu.2024.1490387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024] Open
Abstract
Macrophages are the basic mediators and coordinators of various types of chronic inflammation and play a crucial role in the formation and development of atherosclerosis (AS). In the complex microenvironment of atherosclerotic plaques, macrophages of different sources are exposed to different signal stimuli and thus polarized into various subpopulations. Various types of macrophages with predominantly M1 and M2 phenotypes also play different regulatory roles in the initiation and progression of AS. Lipid-lowering drugs, mainly statins, are widely used in clinical practice, but the adverse reactions are obvious and there is a lack of personalized treatment. Emerging targeted macrophage and Traditional Chinese medicine (TCM)-related therapies can regulate the cellular microenvironment, inhibit the polarization of M1 macrophages, and promote the activation of M2 macrophages, providing new ideas for the prevention and treatment of AS.
Collapse
Affiliation(s)
- LingNa Zhang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - JiaWei Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YuShun Kou
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - LuFan Shen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Hong Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YiYuan Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ruiling Ma
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Tao Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xin Yang
- First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YuanHui Gu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Lin Yi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Chronic Disease Laboratory, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
22
|
Ding Y, Sun Y, Wang H, Zhao H, Yin R, Zhang M, Pan X, Zhu X. Atherosis-associated lnc_000048 activates PKR to enhance STAT1-mediated polarization of THP-1 macrophages to M1 phenotype. Neural Regen Res 2024; 19:2488-2498. [PMID: 38526285 PMCID: PMC11090429 DOI: 10.4103/nrr.nrr-d-23-01355] [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: 08/13/2023] [Revised: 12/12/2023] [Accepted: 01/20/2024] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00029/figure1/v/2024-03-08T184507Z/r/image-tiff Our previous study has demonstrated that lnc_000048 is upregulated in large-artery atherosclerotic stroke and promotes atherosclerosis in ApoE-/- mice. However, little is known about the role of lnc_000048 in classically activated macrophage (M1) polarization. In this study, we established THP-1-derived testing state macrophages (M0), M1 macrophages, and alternately activated macrophages (M2). Real-time fluorescence quantitative PCR was used to verify the expression of marker genes and the expression of lnc_000048 in macrophages. Flow cytometry was used to detect phenotypic proteins (CD11b, CD38, CD80). We generated cell lines with lentivirus-mediated upregulation or downregulation of lnc_000048. Flow cytometry, western blot, and real-time fluorescence quantitative PCR results showed that down-regulation of lnc_000048 reduced M1 macrophage polarization and the inflammation response, while over-expression of lnc_000048 led to the opposite effect. Western blot results indicated that lnc_000048 enhanced the activation of the STAT1 pathway and mediated the M1 macrophage polarization. Moreover, catRAPID prediction, RNA-pull down, and mass spectrometry were used to identify and screen the protein kinase RNA-activated (PKR), then catRAPID and RPIseq were used to predict the binding ability of lnc_000048 to PKR. Immunofluorescence (IF)-RNA fluorescence in situ hybridization (FISH) double labeling was performed to verify the subcellular colocalization of lnc_000048 and PKR in the cytoplasm of M1 macrophage. We speculate that lnc_000048 may form stem-loop structure-specific binding and activate PKR by inducing its phosphorylation, leading to activation of STAT1 phosphorylation and thereby enhancing STAT1 pathway-mediated polarization of THP-1 macrophages to M1 and inflammatory factor expression. Taken together, these results reveal that the lnc_000048/PKR/STAT1 axis plays a crucial role in the polarization of M1 macrophages and may be a novel therapeutic target for atherosclerosis alleviation in stroke.
Collapse
Affiliation(s)
- Yuanyuan Ding
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hongyan Wang
- Qingdao Cadre Health Care Service Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hongqin Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Ruihua Yin
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
23
|
Xu M, Wei S, Su T, Ma D, Wang Z, Zhu D, Weng L, Ding X. Visualizing Macrophage Polarization through Fluorescent mRNA Profiling. BIOSENSORS 2024; 14:475. [PMID: 39451688 PMCID: PMC11506351 DOI: 10.3390/bios14100475] [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: 09/13/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024]
Abstract
Macrophages, known for their phenotypic plasticity, play a critical role in maintaining homeostasis and inflammation-related pathogenesis. Although identifying diverse macrophage phenotypes holds promise for enhancing diagnoses and treatments of diseases mediated by macrophages, existing methodologies for differentiating macrophages often lack precision. They are limited by the cumbersome procedures that require large-scale equipment, such as flow cytometry and transcriptomic analysis. In this context, we have engineered fluorescent polyadenine (polyA)-mediated sticky flares that enable practical visualization of macrophages. This technology facilitates the highly sensitive detection of macrophage phenotypes through the specific recognition of intracellular mRNAs, permitting in situ imaging. Our approach demonstrates the potential for determining macrophage polarization status at the single-cell level within dynamic immune microenvironments, thereby providing crucial diagnostic and prognostic information that could guide the development of tailored treatments for macrophage-related diseases in personalized medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lixing Weng
- State Key Laboratory of Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; (M.X.); (S.W.); (T.S.); (D.M.); (Z.W.)
| | - Xianguang Ding
- State Key Laboratory of Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; (M.X.); (S.W.); (T.S.); (D.M.); (Z.W.)
| |
Collapse
|
24
|
Qu K, Zhong Y, Zhu L, Mou N, Cao Y, Liu J, Wu S, Yan M, Yan F, Li J, Zhang C, Wu G, Zhang K, Qin X, Wu W. A Macrophage Membrane-Functionalized, Reactive Oxygen Species-Activatable Nanoprodrug to Alleviate Inflammation and Improve the Lipid Metabolism for Atherosclerosis Management. Adv Healthc Mater 2024; 13:e2401113. [PMID: 38686849 DOI: 10.1002/adhm.202401113] [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: 03/24/2024] [Revised: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Atherosclerosis (AS) management typically relies on therapeutic drug interventions, but these strategies typically have drawbacks, including poor site specificity, high systemic intake, and undesired side effects. The field of cell membrane camouflaged biomimetic nanomedicine offers the potential to address these challenges thanks to its ability to mimic the natural properties of cell membranes that enable enhanced biocompatibility, prolonged blood circulation, targeted drug delivery, and evasion of immune recognition, ultimately leading to improved therapeutic outcomes and reduced side effects. In this study, a novel biomimetic approach is developed to construct the M1 macrophage membrane-coated nanoprodrug (MM@CD-PBA-RVT) for AS management. The advanced MM@CD-PBA-RVT nanotherapeutics are proved to be effective in inhibiting macrophage phagocytosis and facilitating the cargo delivery to the activated endothelial cells of AS lesion both in vitro and in vivo. Over the 30-day period of nanotherapy, MM@CD-PBA-RVT is capable of significantly inhibiting the progression of AS, while also maintaining a favorable safety profile. In conclusion, the biomimetic MM@CD-PBA-RVT shows promise as feasible drug delivery systems for safe and effective anti-AS applications.
Collapse
Affiliation(s)
- Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Nianlian Mou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yu Cao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Jie Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Shuai Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Meng Yan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Fei Yan
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Jiawei Li
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Cheng Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Guicheng Wu
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
- Jin Feng Laboratory Chongqing, Chongqing, 401329, China
| |
Collapse
|
25
|
Rui H, Yu H, Chi K, Han Z, Zhu W, Zhang J, Guo H, Zou W, Wang F, Xu P, Zou D, Song X, Liu L, Wu X, Wu W, Qin D, Cao Y, Xu F, Xue L, Chen Y. ALDH2 deficiency augments atherosclerosis through the USP14-cGAS-dependent polarization of proinflammatory macrophages. Redox Biol 2024; 76:103318. [PMID: 39178733 PMCID: PMC11388276 DOI: 10.1016/j.redox.2024.103318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
The aldehyde dehydrogenase 2 (ALDH2) rs671 polymorphism commonly exists in the East Asian populations and is associated with high risks of cardiovascular disease (CVD). However, the cellular and molecular mechanisms that underlie the ALDH2 rs671 mutant-linked high CVD remain elusive. Here, we show that macrophages derived from human ALDH2 rs671 carriers and ALDH2 knockout mice exhibited an enhanced pro-inflammatory macrophage phenotype and an impaired anti-inflammatory macrophage phenotype. Transplanting bone marrow from ALDH2-/-ApoE-/- to ApoE-/- mice significantly increased atherosclerotic plaque growth and pro-inflammatory macrophage polarization in vivo. Mechanistically, ALDH2 inhibited activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway in macrophages. Pharmacological inhibition of cGAS by RU.521 completely neutralized ALDH2-deficiency-induced macrophage polarization. In-depth mechanistic investigation showed that ALDH2 accelerated cGAS K48-linked polyubiquitination degradation at lysine 282 in macrophages by reducing the interaction between ubiquitin-specific protease 14 (USP14) and cGAS, mainly through its enzymatic role in mitigating 4-hydroxy-2-nonenal (4-HNE) accumulation. Consistently, USP14 knockdown in bone marrow cells alleviated proinflammatory responses in macrophages and protected against atherosclerosis. Our findings provide new mechanistic insights of ALDH2 deficiency-associated proinflammation and atherosclerosis and new therapeutic and preventive paradigms for treatment of atherosclerosis-associated CVD.
Collapse
Affiliation(s)
- Haiying Rui
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Huaxiang Yu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Kai Chi
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Ziqi Han
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Wenyong Zhu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Qingdao, 266035, China
| | - Jian Zhang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Haipeng Guo
- Department of Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenyi Zou
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Fengxin Wang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Ping Xu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Dan Zou
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiaoshuai Song
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Lulu Liu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xuting Wu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Wenxiao Wu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Dandan Qin
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, 17165, Sweden
| | - Feng Xu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Li Xue
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Yuguo Chen
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China; Shandong Key Laboratory, Magnetic Field-free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, 250012, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
26
|
Ya X, Ma L, Li H, Ge P, Zheng Z, Mou S, Liu C, Zhang Y, Wang R, Zhang Q, Ye X, Zhang D, Zhao J. Exploring the relationship between hemodynamics and the immune microenvironment in carotid atherosclerosis: Insights from CFD and CyTOF technologies. J Cereb Blood Flow Metab 2024; 44:1733-1744. [PMID: 38833561 PMCID: PMC11494853 DOI: 10.1177/0271678x241251976] [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: 10/13/2023] [Revised: 02/24/2024] [Accepted: 03/28/2024] [Indexed: 06/06/2024]
Abstract
Carotid atherosclerosis is a major cause of stroke. Hemodynamic forces, such as shear stress and oscillatory shear, play an important role in the initiation and progression of atherosclerosis. The alteration of the immune microenvironment is the fundamental pathological mechanism by which diverse external environmental factors impact the formation and progression of plaques. However, Current research on the relationship between hemodynamics and immunity in atherosclerosis still lack of comprehensive understanding. In this study, we combined computational fluid dynamics (CFD) and Mass cytometry (CyTOF) technologies to explore the changes in the immune microenvironment within plaques under different hemodynamic conditions. Our results indicated that neutrophils were enriched in adverse flow environments. M2-like CD163+CD86+ macrophages were predominantly enriched in high WSS and low OSI environments, while CD163-CD14+ macrophages were enriched in low WSS and high OSI environments. Functional analysis further revealed T cell pro-inflammatory activation and dysregulation in modulation, along with an imbalance in M1-like/M2-like macrophages, suggesting their potential involvement in the progression of atherosclerotic lesions mediated by adverse flow patterns. Our study elucidated the potential mechanisms by which hemodynamics regulated the immune microenvironment within plaques, providing intervention targets for future precision therapies.
Collapse
Affiliation(s)
- Xiaolong Ya
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Long Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hao Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhiyao Zheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Siqi Mou
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xun Ye
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
27
|
Chuang HJ, Chen YY, Chung YD, Huang E, Huang CY, Lung J, Chen CY, Liao HF. The Immunosuppressive Receptor CD32b Regulation of Macrophage Polarization and Its Implications in Tumor Progression. Int J Mol Sci 2024; 25:9737. [PMID: 39273683 PMCID: PMC11395990 DOI: 10.3390/ijms25179737] [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: 07/31/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Macrophages, pivotal components of the immune system, orchestrate host defense mechanisms in humans and mammals. Their polarization into classically activated macrophages (CAMs or M1) and alternatively activated macrophages (AAMs or M2) dictates distinct functional roles in immunity and tissue homeostasis. While the negative regulatory role of CD32b within the FC gamma receptor (FCγR) family is recognized across various immune cell types, its influence on macrophage polarization remains elusive. This study aimed to elucidate the regulatory role of CD32b in macrophage polarization and discern the differential expression markers between the M1 and M2 phenotypes following CD32b siRNA transfection. The results revealed a decrease in the CD32b levels in lipopolysaccharide (LPS)-treated M1 and an increase in interleukin-4 (IL-4)-treated M2 macrophages, as observed in macrophage Raw264.7 cells. Furthermore, CD32b siRNA transfection significantly downregulated the M2 markers (IL-10, VEGF, Arg-1, and STAT6), while upregulating the M1 markers (IL-6, NF-κB, NOS2, and STAT1) in the Raw264.7 cells. Similar findings were recapitulated in macrophage-rich adherent cells isolated from mouse spleens. Additionally, the cytopathological analysis of pleural effusions and ascitic fluids from patients with cancer revealed a positive correlation between advanced tumor stages, metastasis, and elevated CD32b levels. In conclusion, this study highlights the regulatory influence of CD32b in suppressing M1 expression and promoting M2 polarization. Moreover, heightened M2 activation and CD32b levels appear to correlate with tumor progression. A targeted CD32b blockade may serve as a novel therapeutic strategy to inhibit M2 macrophage polarization and is promising for anti-tumor intervention.
Collapse
Affiliation(s)
- Hong-Jing Chuang
- Department of Pathology, National Taiwan University Hospital Yunlin Branch, Yunlin 640, Taiwan
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600, Taiwan
| | - Ying-Yin Chen
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin 640, Taiwan
| | - Yi-Da Chung
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600, Taiwan
| | - Evelyn Huang
- Cell and Molecular Biology, Northeastern University, Boston, MA 02115, USA
| | | | - Jrhau Lung
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi 613, Taiwan
| | - Chung-Yu Chen
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin 640, Taiwan
| | - Hui-Fen Liao
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600, Taiwan
| |
Collapse
|
28
|
Hu X, Yan Y, Liu W, Liu J, Fan T, Deng H, Cai Y. Advances and perspectives on pharmacological activities and mechanisms of the monoterpene borneol. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155848. [PMID: 38964157 DOI: 10.1016/j.phymed.2024.155848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/31/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Borneol, a highly lipid-soluble bicyclic terpene mainly extracted from plants, is representative of monoterpenoids. Modern medicine has established that borneol exhibits a range of pharmacological activities and used in the treatment of many diseases, particularly Cardio-cerebrovascular diseases (CVDs). The crucial role in enhancing drug delivery and improving bioavailability has attracted much attention. In addition, borneol is also widely utilized in food, daily chemicals, fragrances, and flavors industries. PURPOSE This review systematically summarized the sources, pharmacological activities and mechanisms, clinical trial, pharmacokinetics, toxicity, and application of borneol. In addition, this review describes the pharmacological effects of borneol ester and the combination of borneol with nanomaterial. This review will provide a valuable resource for those pursuing researches on borneol inspiring the pharmacological applications in the medicine, food and daily chemical products, and developing of new drugs containing borneol or its derivatives. METHODS This review searched the keywords ("borneol" or "bornyl esters") and ("pharmacology" or "Traditional Chinese medicine" or "Cardio-cerebrovascular diseases" or "blood-brain barrier" or "ischemic stroke" or "nanomaterials" or "neurodegenerative diseases" or "diabetes" or "toxicity") in Web of Science, PubMed, Google Scholar and China National Knowledge Infrastructure (CNKI) from January 1990 to May 2024. The search was limited to articles published in English and Chinese. RESULTS Borneol exhibits extensive pharmacological activities including anti-inflammatory effects, analgesia, antioxidation, and has the property of crossing biological barriers and treating CVDs. The intrinsic molecular mechanisms are involved in multiple components, such as regulation of various key factors (including Tumor necrosis factor-α, Nuclear factor kappa-B, Interleukin-1β, Malondialdehyde), inhibiting transporter protein function, regulating biochemical levels, and altering physical structural changes. In addition, this review describes the pharmacological effects of borneol ester and the combination of borneol with nanomaterial. CONCLUSION The pharmacological properties and applications of borneol are promising, including anti-inflammatory, analgesic, antimicrobial, and antioxidant properties, as well as enhancing drug delivery and treating CVDs. However, its clinical application is hindered by the limited research on safety, efficacy, and pharmacokinetics. Therefore, this review systemically summarized the advances on pharmacological activities and mechanisms of the borneol. Standardized clinical trials and exploration of synergistic effects with other drugs were also are outlined.
Collapse
Affiliation(s)
- Xiaoxiang Hu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, PR China
| | - Yi Yan
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, PR China
| | - Wenjing Liu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, PR China
| | - Jie Liu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, PR China
| | - Taipin Fan
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1T, UK
| | - Huaxiang Deng
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, PR China.
| | - Yujie Cai
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
29
|
Mackay CDA, Meechem MB, Patel VB. Macrophages in vascular disease: Roles of mitochondria and metabolic mechanisms. Vascul Pharmacol 2024; 156:107419. [PMID: 39181483 DOI: 10.1016/j.vph.2024.107419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Macrophages are a dynamic cell type of the immune system implicated in the pathophysiology of vascular diseases and are a major contributor to pathological inflammation. Excessive macrophage accumulation, activation, and polarization is observed in aortic aneurysm (AA), atherosclerosis, and pulmonary arterial hypertension. In general, macrophages become activated and polarized to a pro-inflammatory phenotype, which dramatically changes cell behavior to become pro-inflammatory and infiltrative. These cell types become cumbersome and fail to be cleared by normal mechanisms such as autophagy. The result is a hyper-inflammatory environment causing the recruitment of adjacent cells and circulating immune cells to further augment the inflammatory response. In AA, this leads to excessive ECM degradation and chemokine secretion, ultimately causing macrophages to dominate the immune cell landscape in the aortic wall. In atherosclerosis, monocytes are recruited to the vascular wall, where they polarize to the pro-inflammatory phenotype and induce inflammatory pathway activation. This leads to the development of foam cells, which significantly contribute to neointima and necrotic core formation in atherosclerotic plaques. Pro-inflammatory macrophages, which affect other vascular diseases, present with fragmented mitochondria and corresponding metabolic dysfunction. Targeting macrophage mitochondrial dynamics has proved to be an exciting potential therapeutic approach to combat vascular disease. This review will summarize mitochondrial and metabolic mechanisms of macrophage activation, polarization, and accumulation in vascular diseases.
Collapse
Affiliation(s)
- Cameron D A Mackay
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Megan B Meechem
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Vaibhav B Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
30
|
Ma Y, Yang X, Ning K, Guo H. M1/M2 macrophage-targeted nanotechnology and PROTAC for the treatment of atherosclerosis. Life Sci 2024; 352:122811. [PMID: 38862062 DOI: 10.1016/j.lfs.2024.122811] [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: 01/05/2024] [Revised: 03/17/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
Macrophages play key roles in atherosclerosis progression, and an imbalance in M1/M2 macrophages leads to unstable plaques; therefore, M1/M2 macrophage polarization-targeted treatments may serve as a new approach in the treatment of atherosclerosis. At present, there is little research on M1/M2 macrophage polarization-targeted nanotechnology. Proteolysis-targeting chimera (PROTAC) technology, a targeted protein degradation technology, mediates the degradation of target proteins and has been widely promoted in preclinical and clinical applications as a novel therapeutic modality. This review summarizes the recent studies on M1/M2 macrophage polarization-targeted nanotechnology, focusing on the mechanism and advantages of PROTACs in M1/M2 macrophage polarization as a new approach for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yupeng Ma
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Xiaofan Yang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| | - Haidong Guo
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
31
|
Liu D, Qin H, Gao Y, Sun M, Wang M. Cardiovascular disease: Mitochondrial dynamics and mitophagy crosstalk mechanisms with novel programmed cell death and macrophage polarisation. Pharmacol Res 2024; 206:107258. [PMID: 38909638 DOI: 10.1016/j.phrs.2024.107258] [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: 02/20/2024] [Revised: 06/08/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024]
Abstract
Several cardiovascular illnesses are associated with aberrant activation of cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis, and macrophage polarisation as hallmarks contributing to vascular damage and abnormal cardiac function. Meanwhile, these three novel forms of cellular dysfunction are closely related to mitochondrial homeostasis. Mitochondria are the main organelles that supply energy and maintain cellular homeostasis. Mitochondrial stability is maintained through a series of regulatory pathways, such as mitochondrial fission, mitochondrial fusion and mitophagy. Studies have shown that mitochondrial dysfunction (e.g., impaired mitochondrial dynamics and mitophagy) promotes ROS production, leading to oxidative stress, which induces cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis and macrophage M1 phenotypic polarisation. Therefore, an in-depth knowledge of the dynamic regulation of mitochondria during cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis and macrophage polarisation is necessary to understand cardiovascular disease development. This paper systematically summarises the impact of changes in mitochondrial dynamics and mitophagy on regulating novel cellular dysfunctions and macrophage polarisation to promote an in-depth understanding of the pathogenesis of cardiovascular diseases and provide corresponding theoretical references for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Dandan Liu
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Hewei Qin
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China; Department of Rehabilitation Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450002, China.
| | - Yang Gao
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Mengyan Sun
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Mengnan Wang
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
32
|
Wang D, Sun Z, Yin Y, Xiang J, Wei Y, Ma Y, Wang L, Liu G. Vitamin D and Atherosclerosis: Unraveling the Impact on Macrophage Function. Mol Nutr Food Res 2024; 68:e2300867. [PMID: 38864846 DOI: 10.1002/mnfr.202300867] [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: 12/05/2023] [Revised: 04/02/2024] [Indexed: 06/13/2024]
Abstract
Vitamin D plays a crucial role in preventing atherosclerosis and in the regulation of macrophage function. This review aims to provide a comprehensive summary of the clinical evidence regarding the impact of vitamin D on atherosclerotic cardiovascular disease, atherosclerotic cerebrovascular disease, peripheral arterial disease, and associated risk factors. Additionally, it explores the mechanistic studies investigating the influence of vitamin D on macrophage function in atherosclerosis. Numerous findings indicate that vitamin D inhibits monocyte or macrophage recruitment, macrophage cholesterol uptake, and esterification. Moreover, it induces autophagy of lipid droplets in macrophages, promotes cholesterol efflux from macrophages, and regulates macrophage polarization. This review particularly focuses on analyzing the molecular mechanisms and signaling pathways through which vitamin D modulates macrophage function in atherosclerosis. It claims that vitamin D has a direct inhibitory effect on the formation, adhesion, and migration of lipid-loaded monocytes, thus exerting anti-atherosclerotic effects. Therefore, this review emphasizes the crucial role of vitamin D in regulating macrophage function and preventing the development of atherosclerosis.
Collapse
Affiliation(s)
- Dongxia Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhen Sun
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yajuan Yin
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Jingyi Xiang
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yuzhe Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Le Wang
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Gang Liu
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| |
Collapse
|
33
|
Fliri A, Kajiji S. Effects of vitamin D signaling in cardiovascular disease: centrality of macrophage polarization. Front Cardiovasc Med 2024; 11:1388025. [PMID: 38984353 PMCID: PMC11232491 DOI: 10.3389/fcvm.2024.1388025] [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: 02/19/2024] [Accepted: 05/24/2024] [Indexed: 07/11/2024] Open
Abstract
Among the leading causes of natural death are cardiovascular diseases, cancer, and respiratory diseases. Factors causing illness include genetic predisposition, aging, stress, chronic inflammation, environmental factors, declining autophagy, and endocrine abnormalities including insufficient vitamin D levels. Inconclusive clinical outcomes of vitamin D supplements in cardiovascular diseases demonstrate the need to identify cause-effect relationships without bias. We employed a spectral clustering methodology capable of analyzing large diverse datasets for examining the role of vitamin D's genomic and non-genomic signaling in disease in this study. The results of this investigation showed the following: (1) vitamin D regulates multiple reciprocal feedback loops including p53, macrophage autophagy, nitric oxide, and redox-signaling; (2) these regulatory schemes are involved in over 2,000 diseases. Furthermore, the balance between genomic and non-genomic signaling by vitamin D affects autophagy regulation of macrophage polarization in tissue homeostasis. These findings provide a deeper understanding of how interactions between genomic and non-genomic signaling affect vitamin D pharmacology and offer opportunities for increasing the efficacy of vitamin D-centered treatment of cardiovascular disease and healthy lifespans.
Collapse
Affiliation(s)
- Anton Fliri
- Emergent System Analytics LLC, Clinton, CT, United States
| | - Shama Kajiji
- Emergent System Analytics LLC, Clinton, CT, United States
| |
Collapse
|
34
|
Peng D, Zhuge F, Wang M, Zhang B, Zhuang Z, Zhou R, Zhang Y, Li J, Yu Z, Shi J. Morus alba L. (Sangzhi) alkaloids mitigate atherosclerosis by regulating M1/M2 macrophage polarization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155526. [PMID: 38564921 DOI: 10.1016/j.phymed.2024.155526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/01/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Atherosclerosis (AS) is an important cause of cardiovascular disease, posing a substantial health risk. Recognized as a chronic inflammatory disorder, AS hinges on the pivotal involvement of macrophages in arterial inflammation, participating in its formation and progression. Sangzhi alkaloid (SZ-A) is a novel natural alkaloid extracted from the mulberry branches, has extensive pharmacological effects and stable pharmacokinetic characteristics. However, the effects and mechanisms of SZ-A on AS remain unclear. PURPOSE To explore the effect and underlying mechanisms of SZ-A on inflammation mediated by macrophages and its role in AS development. METHODS Atherosclerosis was induced in vivo in apolipoprotein E-deficient mice through a high-fat and high-choline diet. We utilized macrophages and vascular endothelial cells to investigate the effects of SZ-A on macrophage polarization and its anti-inflammatory properties on endothelial cells in vitro. The transcriptomic analyses were used to investigate the major molecule that mediates cell-cell interactions and the antiatherogenic mechanisms of SZ-A based on AS, subsequently validated in vivo and in vitro. RESULTS SZ-A demonstrated a significant inhibition in vascular inflammation and alleviation of AS severity by mitigating macrophage infiltration and modulating M1/M2 macrophage polarization in vitro and in vivo. Moreover, SZ-A effectively reduced the release of the proinflammatory mediator C-X-C motif chemokine ligand (CXCL)-10, predominantly secreted by M1 macrophages. This reduction in CXCL-10 contributed to improved endothelial cell function, reduced recruitment of additional macrophages, and inhibited the inflammatory amplification effect. This ultimately led to the suppression of atherogenesis. CONCLUSION SZ-A exhibited potent anti-inflammatory effects by inhibiting macrophage-mediated inflammation, providing a new therapeutic avenue against AS. This is the first study demonstrating the efficacy of SZ-A in alleviating AS severity and offers novel insights into its anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Dandan Peng
- Department of Endocrinology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China; Guizhou Medical University, Guiyang, Guizhou, China
| | - Fen Zhuge
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Mingwei Wang
- Department of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Binbin Zhang
- Department of Infectious Diseases and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zhenjie Zhuang
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Run Zhou
- College of Nursing, Hangzhou Normal University, Zhejiang, China
| | - Yuanyuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Jie Li
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Zhenqiu Yu
- Guizhou Medical University, Guiyang, Guizhou, China; The Department of Hypertension, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.
| | - Junping Shi
- Department of Infectious Diseases and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Medical Epigenetics, Hangzhou, Zhejiang, China; Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, Zhejiang, China.
| |
Collapse
|
35
|
Dong Y, Lau HX, Suaini NHA, Kee MZL, Ooi DSQ, Shek LPC, Lee BW, Godfrey KM, Tham EH, Ong MEH, Liu N, Wong L, Tan KH, Chan JKY, Yap FKP, Chong YS, Eriksson JG, Feng M, Loo EXL. A machine-learning exploration of the exposome from preconception in early childhood atopic eczema, rhinitis and wheeze development. ENVIRONMENTAL RESEARCH 2024; 250:118523. [PMID: 38382664 DOI: 10.1016/j.envres.2024.118523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/19/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Most previous research on the environmental epidemiology of childhood atopic eczema, rhinitis and wheeze is limited in the scope of risk factors studied. Our study adopted a machine learning approach to explore the role of the exposome starting already in the preconception phase. METHODS We performed a combined analysis of two multi-ethnic Asian birth cohorts, the Growing Up in Singapore Towards healthy Outcomes (GUSTO) and the Singapore PREconception Study of long Term maternal and child Outcomes (S-PRESTO) cohorts. Interviewer-administered questionnaires were used to collect information on demography, lifestyle and childhood atopic eczema, rhinitis and wheeze development. Data training was performed using XGBoost, genetic algorithm and logistic regression models, and the top variables with the highest importance were identified. Additive explanation values were identified and inputted into a final multiple logistic regression model. Generalised structural equation modelling with maternal and child blood micronutrients, metabolites and cytokines was performed to explain possible mechanisms. RESULTS The final study population included 1151 mother-child pairs. Our findings suggest that these childhood diseases are likely programmed in utero by the preconception and pregnancy exposomes through inflammatory pathways. We identified preconception alcohol consumption and maternal depressive symptoms during pregnancy as key modifiable maternal environmental exposures that increased eczema and rhinitis risk. Our mechanistic model suggested that higher maternal blood neopterin and child blood dimethylglycine protected against early childhood wheeze. After birth, early infection was a key driver of atopic eczema and rhinitis development. CONCLUSION Preconception and antenatal exposomes can programme atopic eczema, rhinitis and wheeze development in utero. Reducing maternal alcohol consumption during preconception and supporting maternal mental health during pregnancy may prevent atopic eczema and rhinitis by promoting an optimal antenatal environment. Our findings suggest a need to include preconception environmental exposures in future research to counter the earliest precursors of disease development in children.
Collapse
Affiliation(s)
- Yizhi Dong
- Saw Swee Hock School of Public Health, National University Health System, National University of Singapore, Singapore.
| | - Hui Xing Lau
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore.
| | - Noor Hidayatul Aini Suaini
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore.
| | - Michelle Zhi Ling Kee
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore.
| | - Delicia Shu Qin Ooi
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore.
| | - Lynette Pei-Chi Shek
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Bee Wah Lee
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Keith M Godfrey
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom; MRC Lifecourse Epidemiology Centre, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.
| | - Elizabeth Huiwen Tham
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore; Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Marcus Eng Hock Ong
- Department of Emergency Medicine, Singapore General Hospital, Singapore, Singapore; Health Services and Systems Research, Duke-NUS Graduate Medical School, Singapore, Singapore.
| | - Nan Liu
- Duke-NUS Medical School, National University of Singapore, Singapore; Health Services Research Centre, Singapore Health Services, Singapore, Singapore; Institute of Data Science, National University of Singapore, Singapore.
| | - Limsoon Wong
- School of Computing, National University of Singapore, 13 Computing Drive, Singapore 117417, Singapore.
| | - Kok Hian Tan
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital (KKH), Singapore.
| | - Jerry Kok Yen Chan
- Duke-NUS Medical School, National University of Singapore, Singapore; Department of Reproductive Medicine, KK Women's and Children's Hospital (KKH), Singapore.
| | - Fabian Kok Peng Yap
- Duke-NUS Medical School, National University of Singapore, Singapore; Department of Paediatrics, KK Women's and Children's Hospital (KKH), Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore; Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore.
| | - Johan Gunnar Eriksson
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore; Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore; Folkhälsan Research Center, Helsinki, Finland; Department of General Practice and Primary Health Care, University of Helsinki, Finland.
| | - Mengling Feng
- Saw Swee Hock School of Public Health, National University Health System, National University of Singapore, Singapore.
| | - Evelyn Xiu Ling Loo
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Dean's Office, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
36
|
Zhang Y, Kong X, Liang L, Xu D. Regulation of vascular remodeling by immune microenvironment after the establishment of autologous arteriovenous fistula in ESRD patients. Front Immunol 2024; 15:1365422. [PMID: 38807593 PMCID: PMC11130379 DOI: 10.3389/fimmu.2024.1365422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
Autogenous arteriovenous fistula (AVF) is the preferred dialysis access for receiving hemodialysis treatment in end-stage renal disease patients. After AVF is established, vascular remodeling occurs in order to adapt to hemodynamic changes. Uremia toxins, surgical injury, blood flow changes and other factors can induce inflammatory response, immune microenvironment changes, and play an important role in the maintenance of AVF vascular remodeling. This process involves the infiltration of pro-inflammatory and anti-inflammatory immune cells and the secretion of cytokines. Pro-inflammatory and anti-inflammatory immune cells include neutrophil (NEUT), dendritic cell (DC), T lymphocyte, macrophage (Mφ), etc. This article reviews the latest research progress and focuses on the role of immune microenvironment changes in vascular remodeling of AVF, in order to provide a new theoretical basis for the prevention and treatment of AVF failure.
Collapse
Affiliation(s)
| | | | - Liming Liang
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, Shandong, China
| | | |
Collapse
|
37
|
Mao J, Chen Y, Zong Q, Liu C, Xie J, Wang Y, Fisher D, Hien NTT, Pronyuk K, Musabaev E, Li Y, Zhao L, Dang Y. Corilagin alleviates atherosclerosis by inhibiting NLRP3 inflammasome activation via the Olfr2 signaling pathway in vitro and in vivo. Front Immunol 2024; 15:1364161. [PMID: 38803504 PMCID: PMC11128681 DOI: 10.3389/fimmu.2024.1364161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 05/01/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Atherosclerosis, a leading cause of global cardiovascular mortality, is characterized by chronic inflammation. Central to this process is the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, which significantly influences atherosclerotic progression. Recent research has identified that the olfactory receptor 2 (Olfr2) in vascular macrophages is instrumental in driving atherosclerosis through NLRP3- dependent IL-1 production. Methods To investigate the effects of Corilagin, noted for its anti-inflammatory attributes, on atherosclerotic development and the Olfr2 signaling pathway, our study employed an atherosclerosis model in ApoE-/- mice, fed a high-fat, high-cholesterol diet, alongside cellular models in Ana-1 cells and mouse bone marrow-derived macrophages, stimulated with lipopolysaccharides and oxidized low-density lipoprotein. Results The vivo and vitro experiments indicated that Corilagin could effectively reduce serum lipid levels, alleviate aortic pathological changes, and decrease intimal lipid deposition. Additionally, as results showed, Corilagin was able to cut down expressions of molecules associated with the Olfr2 signaling pathway. Discussion Our findings indicated that Corilagin effectively inhibited NLRP3 inflammasome activation, consequently diminishing inflammation, macrophage polarization, and pyroptosis in the mouse aorta and cellular models via the Olfr2 pathway. This suggests a novel therapeutic mechanism of Corilagin in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jinqian Mao
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiushuo Zong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cuiling Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Xie
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yujie Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of The Western Cape, Cape Town, South Africa
| | | | - Khrystyna Pronyuk
- Department of Infectious Diseases, O.Bogomolets National Medical University, Kyiv, Ukraine
| | - Erkin Musabaev
- Research Institute of Virology, Ministry of Health, Tashkent, Uzbekistan
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
38
|
Chen S, Zeng J, Li R, Zhang Y, Tao Y, Hou Y, Yang L, Zhang Y, Wu J, Meng X. Traditional Chinese medicine in regulating macrophage polarization in immune response of inflammatory diseases. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117838. [PMID: 38310986 DOI: 10.1016/j.jep.2024.117838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Numerous studies have demonstrated that various traditional Chinese medicines (TCMs) exhibit potent anti-inflammatory effects against inflammatory diseases mediated through macrophage polarization and metabolic reprogramming. AIM OF THE STUDY The objective of this review was to assess and consolidate the current understanding regarding the pathogenic mechanisms governing macrophage polarization in the context of regulating inflammatory diseases. We also summarize the mechanism action of various TCMs on the regulation of macrophage polarization, which may contribute to facilitate the development of natural anti-inflammatory drugs based on reshaping macrophage polarization. MATERIALS AND METHODS We conducted a comprehensive review of recently published articles, utilizing keywords such as "macrophage polarization" and "traditional Chinese medicines" in combination with "inflammation," as well as "macrophage polarization" and "inflammation" in conjunction with "natural products," and similar combinations, to search within PubMed and Google Scholar databases. RESULTS A total of 113 kinds of TCMs (including 62 components of TCMs, 27 TCMs as well as various types of extracts of TCMs and 24 Chinese prescriptions) was reported to exert anti-inflammatory effects through the regulation of key pathways of macrophage polarization and metabolic reprogramming. CONCLUSIONS In this review, we have analyzed studies concerning the involvement of macrophage polarization and metabolic reprogramming in inflammation therapy. TCMs has great advantages in regulating macrophage polarization in treating inflammatory diseases due to its multi-pathway and multi-target pharmacological action. This review may contribute to facilitate the development of natural anti-inflammatory drugs based on reshaping macrophage polarization.
Collapse
Affiliation(s)
- Shiyu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Rui Li
- The Affiliated Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, PR China
| | - Yingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yiwen Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Ya Hou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Lu Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yating Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiasi Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
39
|
He P, Dai M, Li Z, Wang X, Liu H, He Y, Jiang H. Effect of connexin 43 in LPS/IL-4-induced macrophage M1/M2 polarization: An observational study. Medicine (Baltimore) 2024; 103:e37811. [PMID: 38608055 PMCID: PMC11018209 DOI: 10.1097/md.0000000000037811] [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: 02/06/2024] [Accepted: 03/15/2024] [Indexed: 04/14/2024] Open
Abstract
Lipopolysaccharide (LPS) and interleukin-4 (IL-4) play important roles in inducing M1 and M2 macrophage polarization. Studies have shown that LPS can promote the polarization of macrophages to M1-type and produce many pro-inflammatory cytokines, while IL-4 can promote the polarization of macrophages to M2-type and produce many anti-inflammatory cytokines. Moreover, Connexin 43 (Cx43) is widely expressed in macrophages and has various regulatory functions. However, whether Cx43 is involved in the regulation of macrophage M1/M2 polarization has not been fully studied. This study examined the role of Cx43 and M2 polarization markers using Western blot, immunofluorescence, flow cytometry. Cx43 overexpression was induced using Cx43 overexpressing lentivirus. The statistical software SPSS 20.0 (IBM Corp.) and GraphPad Prism 8.0 (GraphPad Software, La Jolla, CA, United States) were used to analyze the results. P values < .05 were considered to indicate statistically significant differences. Our results showed that LPS promotes the polarization of macrophages to M1-type, which is accompanied by an increase in Cx43 expression from 0 to 24 hours. Moreover, the application of the Cx43-specific blockers Gap19 and Gap26 reduces the expression of macrophage M1-type polarization markers. Thus, the expression of Cx43 increases first, and then, due to the initiation of intracellular autophagy during LPS-induced macrophage M1 polarization. Cx43 is degraded and the expression of Cx43 decreases from 24 hours to 48 hours. IL-4 decreases the expression of Cx43 from 24 hours to 48 hours and promotes the transformation of macrophages to M2-type. The application of Cx43 overexpression lentivirus leads to a reduction in the expression of M2 polarization markers. IL-4-induced M2 polarization of macrophages inhibits cell autophagy, reducing Cx43 degradation and leading to an increase in Cx43 from 24 hours to 48 hours. Thus, Cx43 expression in M2-type polarization experiences a reduction at first and then an increase from 24 hours to 48 hours. The direction of macrophage polarization can be controlled by regulating the expression of Cx43, thus providing a theoretical basis for treating atherosclerosis, tumors, and other diseases associated with macrophage polarization.
Collapse
Affiliation(s)
- Pengchen He
- Department of Neurosurgery, Mianyang Central Hospital Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Mingxing Dai
- Department of Rehabilitation Therapy, Mianyang Central Hospital Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Zongpin Li
- Department of Neurosurgery, Mianyang Central Hospital Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Xiaoyi Wang
- Department of Neurosurgery, Mianyang Central Hospital Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Hongyuan Liu
- Department of Neurosurgery, Mianyang Central Hospital Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Yixiao He
- Department of Pathology, Mianyang Central Hospital Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Han Jiang
- Department of Rehabilitation Therapy, Mianyang Central Hospital Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
40
|
Luo M, Zhao F, Cheng H, Su M, Wang Y. Macrophage polarization: an important role in inflammatory diseases. Front Immunol 2024; 15:1352946. [PMID: 38660308 PMCID: PMC11039887 DOI: 10.3389/fimmu.2024.1352946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Macrophages are crucial cells in the human body's innate immunity and are engaged in a variety of non-inflammatory reactions. Macrophages can develop into two kinds when stimulated by distinct internal environments: pro-inflammatory M1-like macrophages and anti-inflammatory M2-type macrophages. During inflammation, the two kinds of macrophages are activated alternatively, and maintaining a reasonably steady ratio is critical for maintaining homeostasis in vivo. M1 macrophages can induce inflammation, but M2 macrophages suppress it. The imbalance between the two kinds of macrophages will have a significant impact on the illness process. As a result, there are an increasing number of research being conducted on relieving or curing illnesses by altering the amount of macrophages. This review summarizes the role of macrophage polarization in various inflammatory diseases, including autoimmune diseases (RA, EAE, MS, AIH, IBD, CD), allergic diseases (allergic rhinitis, allergic dermatitis, allergic asthma), atherosclerosis, obesity and type 2 diabetes, metabolic homeostasis, and the compounds or drugs that have been discovered or applied to the treatment of these diseases by targeting macrophage polarization.
Collapse
Affiliation(s)
| | | | | | | | - Yuanmin Wang
- The Third Affiliated Hospital of Zunyi Medical University, The First People’s Hospital of Zunyi, Zunyi, Guizhou, China
| |
Collapse
|
41
|
Quan YZ, Ma A, Ren CQ, An YP, Qiao PS, Gao C, Zhang YK, Li XW, Lin SM, Li NN, Chen DL, Pan Y, Zhou H, Lin DM, Lin SQ, Li M, Yang BX. Ganoderic acids alleviate atherosclerosis by inhibiting macrophage M1 polarization via TLR4/MyD88/NF-κB signaling pathway. Atherosclerosis 2024; 391:117478. [PMID: 38417185 DOI: 10.1016/j.atherosclerosis.2024.117478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS) is a chronic inflammatory disease characterized by lipid infiltration and plaque formation in blood vessel walls. Ganoderic acids (GA), a class of major bioactive compounds isolated from the Chinese traditional medicine Ganoderma lucidum, have multiple pharmacological activities. This study aimed to determine the anti-atherosclerotic effect of GA and reveal the pharmacological mechanism. METHODS ApoE-/- mice were fed a high-cholesterol diet and treated with GA for 16 weeks to induce AS and identify the effect of GA. Network pharmacological analysis was performed to predict the anti-atherosclerotic mechanisms. An invitro cell model was used to explore the effect of GA on macrophage polarization and the possible mechanism involved in bone marrow dereived macrophages (BMDMs) and RAW264.7 cells stimulated with lipopolysaccharide or oxidized low-density lipoprotein. RESULTS It was found that GA at 5 and 25 mg/kg/d significantly inhibited the development of AS and increased plaque stability, as evidenced by decreased plaque in the aorta, reduced necrotic core size and increased collagen/lipid ratio in lesions. GA reduced the proportion of M1 macrophages in plaques, but had no effect on M2 macrophages. In vitro experiments showed that GA (1, 5, 25 μg/mL) significantly decreased the proportion of CD86+ macrophages and the mRNA levels of IL-6, IL-1β, and MCP-1 in macrophages. Experimental results showed that GA inhibited M1 macrophage polarization by regulating TLR4/MyD88/NF-κB signaling pathway. CONCLUSIONS This study demonstrated that GA play an important role in plaque stability and macrophage polarization. GA exert the anti-atherosclerotic effect partly by regulating TLR4/MyD88/NF-κB signaling pathways to inhibit M1 polarization of macrophages. Our study provides theoretical basis and experimental data for the pharmacological activity and mechanisms of GA against AS.
Collapse
Affiliation(s)
- Ya-Zhu Quan
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Ang Ma
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100007, China
| | - Chao-Qun Ren
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yong-Pan An
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Pan-Shuang Qiao
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Cai Gao
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yu-Kun Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, Chongqing, 404020, China
| | - Xiao-Wei Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; China Resources Pharmaceutical Group Limited, Beijing, 100000, China
| | - Si-Mei Lin
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Nan-Nan Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Di-Long Chen
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, Chongqing, 404020, China
| | - Yan Pan
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hong Zhou
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Dong-Mei Lin
- China National Engineering Research Center on JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Shu-Qian Lin
- China National Engineering Research Center on JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Min Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Bao-Xue Yang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
42
|
Fagbohun OF, Thilakarathna WPDW, Zhou J, Lehmann C, Jiao G, Rupasinghe HPV. Sea Cucumber and Blueberry Extracts Suppress Inflammation and Reduce Acute Lung Injury through the Regulation of NF-κB/MAPK/JNK Signaling Pathway in Lipopolysaccharide-Treated C57BL/6 Mice. Molecules 2024; 29:1511. [PMID: 38611791 PMCID: PMC11013731 DOI: 10.3390/molecules29071511] [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: 02/11/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Acute lung injury (ALI) represents a life-threatening condition with high morbidity and mortality despite modern mechanical ventilators and multiple pharmacological strategies. Therefore, there is a need to develop efficacious interventions with minimal side effects. The anti-inflammatory activities of sea cucumber (Cucumaria frondosa) and wild blueberry (Vaccinium angustifolium) extracts have been reported recently. However, their anti-inflammatory activities and the mechanism of action against ALI are not fully elucidated. Thus, the present study aims to understand the mechanism of the anti-inflammatory activity of sea cucumber and wild blueberry extracts in the context of ALI. Experimental ALI was induced via intranasal lipopolysaccharide (LPS) instillation in C57BL/6 mice and the anti-inflammatory properties were determined by cytokine analysis, histological examination, western blot, and qRT-PCR. The results showed that oral supplementation of sea cucumber extracts repressed nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, thereby downregulating the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF) in the lung tissue and in the plasma. Wild blueberry extracts also suppressed the expression of IL-4. Furthermore, the combination of sea cucumber and wild blueberry extracts restrained MAPK signaling pathways by prominent attenuation of phosphorylation of NF-κB, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) while the levels of pro-inflammatory cytokines were significantly suppressed. Moreover, there was a significant and synergistic reduction in varying degrees of ALI lesions such as distorted parenchyma, increased alveoli thickness, lymphocyte and neutrophil infiltrations, fibrin deposition, pulmonary emphysema, pneumonia, intra-alveolar hemorrhage, and edema. The anti-inflammatory effect of the combination of sea cucumber and wild blueberry extracts is associated with suppressing MAPK and NF-κB signaling pathways, thereby significantly reducing cytokine storm in LPS-induced experimental ALI.
Collapse
Affiliation(s)
- Oladapo F. Fagbohun
- Department Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada; (O.F.F.); (W.P.D.W.T.)
- Department of Biology, Center for Agriculture and Sciences, Wilmington College, 1870 Quaker Way, Wilmington, OH 45177, USA
| | - Wasitha P. D. W. Thilakarathna
- Department Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada; (O.F.F.); (W.P.D.W.T.)
| | - Juan Zhou
- Departments of Anaesthesia, Pain Management and Perioperative Medicine, Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.Z.); (C.L.)
| | - Christian Lehmann
- Departments of Anaesthesia, Pain Management and Perioperative Medicine, Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.Z.); (C.L.)
| | - Guangling Jiao
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - H. P. Vasantha Rupasinghe
- Department Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada; (O.F.F.); (W.P.D.W.T.)
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
43
|
Liang W, Wei R, Zhu X, Li J, Lin A, Chen J, Wu W, Jie Q. Downregulation of HMGB1 carried by macrophage-derived extracellular vesicles delays atherosclerotic plaque formation through Caspase-11-dependent macrophage pyroptosis. Mol Med 2024; 30:38. [PMID: 38493291 PMCID: PMC10943908 DOI: 10.1186/s10020-023-00753-z] [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: 04/02/2023] [Accepted: 11/02/2023] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Macrophage-derived extracellular vesicle (macrophage-EV) is highly studied for its regulatory role in atherosclerosis (AS). Our current study tried to elucidate the possible role of macrophage-EV loaded with small interfering RNA against high-mobility group box 1 (siHMGB1) affecting atherosclerotic plaque formation. METHODS In silico analysis was performed to find critical factors in mouse atherosclerotic plaque formation. EVs secreted by RAW 264.7 cells were collected by ultracentrifugation and characterized, followed by the preparation of macrophage-EV-loaded siHMGB1 (macrophage-EV/siHMGB1). ApoE-/- mice were used to construct an AS mouse model by a high-fat diet, followed by injection of macrophage-EV/siHMGB1 to assess the in vivo effect of macrophage-EV/siHMGB1 on AS mice. RAW264.7 cells were subjected to ox-LDL, LPS or macrophage-EV/siHMGB1 for analyzing the in vitro effect of macrophage-EV/siHMGB1 on macrophage pyrophosis and inflammation. RESULTS In silico analysis found that HMGB1 was closely related to the development of AS. Macrophage-EV/siHMGB could inhibit the release of HMGB1 from macrophages to outside cells, and the reduced HMGB1 release could inhibit foam cell formation. Besides, macrophage-EV/siHMGB also inhibited the LPS-induced Caspase-11 activation, thus inhibiting macrophage pyroptosis and preventing atherosclerotic plaque formation. CONCLUSION Our results proved that macrophage-EV/siHMGB could inhibit foam cell formation and suppress macrophage pyroptosis, finally preventing atherosclerotic plaque formation in AS mice.
Collapse
Affiliation(s)
- Weijie Liang
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Ruibin Wei
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Xingxing Zhu
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Jinliang Li
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Aiwen Lin
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Jun Chen
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Wen Wu
- Department of Endocrinology, Guangdong Geriatrics Institute, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, No. 106, Zhongshan Second Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Qiang Jie
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China.
| |
Collapse
|
44
|
Zhou S, Yang X, Mo K, Ning Z. Pyroptosis and polarization of macrophages in septic acute lung injury induced by lipopolysaccharide in mice. Immun Inflamm Dis 2024; 12:e1197. [PMID: 38501547 PMCID: PMC10949386 DOI: 10.1002/iid3.1197] [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: 09/27/2023] [Revised: 01/13/2024] [Accepted: 02/07/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Pyroptosis and polarization are significant contributors to the onset and development of many diseases. At present, the relationship between pyroptosis and polarization in acute lung injury (ALI) caused by sepsis remains unclear. METHODS The ALI model for sepsis was created in mice and categorized into the blank control, lipopolysaccharide (LPS) group, LPS + low-dose Belnacasan group, LPS + high-dose Belnacasan group, LPS + low-dose Wedelolactone group, LPS + high-dose Wedelolactone group, and positive control group. The wet-dry specific gravity was evaluated to compare pulmonary edema. Hematoxylin-eosin, Masson, and terminal deoxynucleotidyl transferase dUTP nick end labeling staining techniques were conducted to observe and contrast the pathological changes in lung tissue. ELISA was utilized to identify M1 and M2 macrophages and correlated inflammatory factors. Immunohistochemical staining and flow cytometry were employed to identify markers of M1 and M2 macrophages in lung tissue. Propidium iodide staining, together with flow cytometry, was utilized to observe the degree and positive rate of pyroptosis of alveolar macrophages. Western blot analysis was conducted to detect the expression levels of Caspase 1, Caspase 11, GSDMD, and IL-18 in the lung tissues of each group. The real-time quantitative polymerase chain reaction method was used to ascertain relative expression levels of NLRP3, Caspase 1, Caspase 11, GSDMD, IL-18, iNOS, and Arg-1 in lung tissues of all groups. RESULTS In mice with sepsis-induced ALI, both classical and nonclassical pathways of pyroptosis are observed. Inhibiting pyroptosis has been found to ameliorate lung injury, pulmonary edema, and inflammation induced by LPS. Notably, the expression of NLRP3, Caspase 1, Caspase 11, GSDMD, IL-1β, IL-18, TGF-β, CD86, CD206, iNOS, and Arg-1 were all altered in this process. Additionally, alveolar macrophages were polarized along with pyroptosis in mice with ALI caused by sepsis. CONCLUSION Pyroptosis of alveolar macrophages in the context of ALI in mice infected with sepsis has been linked to the polarization of alveolar macrophages toward type M1.
Collapse
Affiliation(s)
- Sijiang Zhou
- Department of Emergencythe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Xia Yang
- Department of General Medicinethe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Kanglin Mo
- Department of Respiratory Medicinethe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Zong Ning
- Department of General Medicinethe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| |
Collapse
|
45
|
Ji M, Mao L, Wei Y, Zhu B, Zhai Y, Zhou X, Tao W, Wang W, Wu H. The Anti-Atherosclerotic Effects of Buyang Huanwu Decoction through M1 and M2 Macrophage Polarization in an ApoE Knockout Mouse Model. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:79-87. [PMID: 38780292 DOI: 10.4103/ejpi.ejpi-d-23-00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/02/2024] [Indexed: 05/25/2024]
Abstract
ABSTRACT Arteriosclerosis (AS) is a chronic inflammatory disease and Buyang Huanwu decoction (BHD) has been identified as an anti-atherosclerosis effect, and the study is aimed to investigate the underlying mechanism. The E4 allele of Apolipoprotein E (ApoE) is associated with both metabolic dysfunction and an enhanced pro-inflammatory response, ApoE-knockout (ApoE-/-) mice were fed with a high-fat diet to establish an arteriosclerosis model and treated with BHD or atorvastatin (as a positive control). The atherosclerotic plaque in each mouse was evaluated using Oil red O Staining. Elisa kits were used to evaluate blood lipid, interleukin-6 (IL-6), IL-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), IL-4, IL-10, and tumor growth factor beta (TGF-β) contents, while Western blot was applicated to measure inducible nitric oxide synthase (iNOS), arginase I (Arg-1) expression. Meanwhile, pyruvate kinase M2 (PKM2), hypoxia-inducible factor-1 alpha (HIF-1α) and its target genes glucose transporter type 1 (GLUT1), lactate dehydrogenase A (LDHA), and 3-phosphoinositide-dependent kinase 1 (PDK1), as well as IL-6, IL-1β, TNF-α, IL-4, IL-10, and TGF-β were evaluated by the quantitative reverse transcription-polymerase chain reaction. BHD treatment significantly reduced body weight and arteriosclerosis plaque area and blood lipid levels including total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C). Meanwhile, BHD demonstrated a significant suppression of M1 polarization, by decreased secretion of iNOS and pro-inflammatory factors (IL-6, IL-1β, and TNF-α) in ApoE-/- mice. The present study also revealed that BHD promotes the activation of M2 polarization, characterized by the expression of Arg-1 and anti-inflammatory factors (IL-4 and IL-10). In addition, PKM2/HIF-1α signaling was improved by M1/M2 macrophages polarization induced by BHD. The downstream target genes (GLUT1, LDHA, and PDK1) expression was significantly increased in high fat feeding ApoE-/- mice, and those of which were recused by BHD and Atorvastatin. These results suggested that M1/M2 macrophages polarization produce the inflammatory response against AS progress after BHD exposure.
Collapse
Affiliation(s)
- Mengjiao Ji
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Mao
- Experimental Center for Science and Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanan Wei
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Boran Zhu
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Zhai
- Experimental Center for Science and Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Zhou
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haoxin Wu
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
46
|
He Y, Cai Y, Wei D, Cao L, He Q, Zhang Y. Elucidating the mechanisms of formononetin in modulating atherosclerotic plaque formation in ApoE-/- mice. BMC Cardiovasc Disord 2024; 24:121. [PMID: 38388385 PMCID: PMC10882812 DOI: 10.1186/s12872-024-03774-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Atherosclerosis(AS) poses a pressing challenge in contemporary medicine. Formononetin (FMN) plays a crucial role in its prevention and treatment. However, the detailed impact of FMN on the stability of atherosclerotic plaques and its underlying mechanisms remain to be elucidated. METHODS An intervention consisting of FMN was given along with a high-fat food regimen in the ApoE-/- mouse model. The investigation included the evaluation of the degree of atherosclerotic lesion, the main components of the plaque, lipid profiles, particular markers indicating M1/M2 macrophage phenotypes, the quantities of factors related to inflammation, the infiltration of macrophages, and the identification of markers linked to the α7nAChR/JAK2/STAT3 axis effect molecules. RESULTS The evaluation of aortic morphology in ApoE-/-mice revealed that FMN significantly improved the plaque area, fibrous cap protrusion, lipid deposition, and structural alterations on the aortic surface, among other markers of atherosclerosis,and there is concentration dependence. Furthermore, the lipid content of mouse serum was assessed, and the results showed that the low-, medium-, and high-dosage FMN groups had significantly lower levels of LDL-C, ox-LDL, TC, and TG. The results of immunohistochemical staining indicated that the low-, medium-, and high-dose FMN therapy groups had enhanced CD206 expression and decreased expression of CD68 and iNOS. According to RT-qPCR data, FMN intervention has the potential to suppress the expression of iNOS, COX-2, miR-155-5p, IL-6, and IL-1β mRNA, while promoting the expression of IL-10, SHIP1, and Arg-1 mRNA levels. However, the degree of inhibition varied among dosage groups. Western blot investigation of JAK/STAT signaling pathway proteins and cholinergic α7nAChR protein showed that p-JAK2 and p-STAT3 protein expression was suppressed at all dosages, whereas α7nAChR protein expression was enhanced. CONCLUSIONS According to the aforementioned findings, FMN can reduce inflammation and atherosclerosis by influencing macrophage polarization, blocking the JAK/STAT signaling pathway, and increasing α7nAChR expression.
Collapse
Affiliation(s)
- Ying He
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Youde Cai
- Jinyang Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, 550081, China
| | - Dingling Wei
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Liping Cao
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Qiansong He
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China.
| | - Yazhou Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
47
|
Yuan D, Zheng Z, Shen C, Ye J, Zhu L. Cytoprotective effects of C1s enzyme in macrophages in atherosclerosis mediated through the LRP5 and Wnt/β-catenin pathway. Mol Immunol 2024; 166:29-38. [PMID: 38218080 DOI: 10.1016/j.molimm.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/05/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
C1s enzyme (active C1s) is a subunit of the complement C1 complex that cleaves low-density lipoprotein receptor-related proteins 5 and 6, leading to Wnt/β-catenin pathway activation in some cell lines. Macrophages have two major functional polarization states (the classically activated M1 state and the alternatively activated M2 state) and play an essential role in atherosclerosis. An increasing amount of evidence suggests that canonical Wnt signaling is related to macrophage polarization. In this study, we explored the cytoprotective effects of C1s enzyme in macrophages. The results show that C1s enzyme activates canonical Wnt signaling in macrophages, exacerbates macrophage M2 polarization, and inhibits M1 polarization. Moreover, C1s enzyme reduces foam cell formation and simultaneously enhances efferocytosis. This study reveals a novel function of C1s enzyme in macrophages in the context of atherosclerosis.
Collapse
Affiliation(s)
- Dong Yuan
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | | | - Cheng Shen
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Ye
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China.
| | - Li Zhu
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China.
| |
Collapse
|
48
|
Apeku E, Tantuoyir MM, Zheng R, Tanye N. Exploring the polarization of M1 and M2 macrophages in the context of skin diseases. Mol Biol Rep 2024; 51:269. [PMID: 38302766 DOI: 10.1007/s11033-023-09014-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/30/2023] [Indexed: 02/03/2024]
Abstract
Macrophages are critical components of the immune system and play vital roles in pathogen defense, immune regulation, and tissue repair. These cells exhibit different polarization states depending on environmental signals, and the M1/M2 paradigm is a useful tool for comprehending these states. This review article comprehensively presents the underlying mechanisms of M1 and M2 macrophage polarization and examines their polarization in various skin diseases. Additionally, this paper discusses therapeutic strategies that target M1 and M2 macrophage polarization in skin diseases. A more profound understanding of macrophage polarization in skin diseases could provide valuable insights for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Ernestina Apeku
- Department of Dermatology, The 1st Hospital of Shanxi Medical University; Graduate Department of Shanxi Medical University, Taiyuan, Shanxi, China
| | | | - Rui Zheng
- Department of Dermatology, The 1st Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Nestor Tanye
- School of Automation Science and Engineering, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
49
|
Gu Y, Cui M, Wang W, Zhang J, Wang H, Zheng C, Lei L, Ji M, Chen W, Xu Y, Wang P. Visualization of the Ferroptosis in Atherosclerotic Plaques with Nanoprobe Engineered by Macrophage Cell Membranes. Anal Chem 2024; 96:281-291. [PMID: 38153251 DOI: 10.1021/acs.analchem.3c03999] [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: 12/29/2023]
Abstract
Atherosclerosis (AS) is the root cause of cardiovascular diseases. Ferroptosis is characterized by highly iron-dependent lipid peroxidation and has been reported to play an important role in the pathogenesis of AS. Visualization of the ferroptosis process in atherosclerotic plaques is of great importance for diagnosing and treating AS. In this work, the rationally designed fluorescent probe FAS1 exhibited excellent advantages including large Stokes shift, sensitivity to environmental viscosity, good photostability, and improved water solubility. It also could co-locate with commercial lipid droplets (LDs) probes (BODIPY 493/503) well in RAW264.7 cells treated by the ferroptosis inducer. After self-assembly into nanoparticles and then encapsulation with macrophage membranes, the engineered FAS1@MM NPs could successfully target the atherosclerotic plaques in Western diet-induced apolipoprotein E knockout (ApoE-/-) mice and reveal the association of ferroptosis with AS through fluorescence imaging in vivo. This study may provide additional insights into the roles of ferroptosis in the diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Yinhui Gu
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Mengyuan Cui
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Weizhi Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Jiaqi Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Huizhe Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Zheng
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Lijuan Lei
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Wei Chen
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Peng Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
50
|
Wang Z, Zhou S, Hao Y, Xu T, An P, Luo Y, Luo J. Nicotinamide mononucleotide protects against high-fat-diet-induced atherosclerosis in mice and dampens aortic inflammation and oxidative stress. J Funct Foods 2024; 112:105985. [DOI: 10.1016/j.jff.2023.105985] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
|