1
|
Dai Y, Yang L, Cao G, Mo L, Yang C, Zhu Y, Guo Y, Hong Y, Xu H, Lu S, Du S, He J. Combination therapy and drug co-delivery systems for atherosclerosis. J Control Release 2025; 381:113543. [PMID: 39986476 DOI: 10.1016/j.jconrel.2025.02.039] [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: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of plaque within the arteries. Despite advances in therapeutic strategies including anti-inflammatory, antioxidant, and lipid metabolism modulation treatments over the past two decades, the treatment of atherosclerosis remains challenging, as arterial damage is the result of interconnected pathological factors. Therefore, current monotherapies often fail to address the complex nature of this disease, leading to insufficient therapeutic outcomes. This review addressed this paucity of effective treatment options by comprehensively exploring the potential for combination therapies and advanced drug co-delivery systems for the treatment of atherosclerosis. We investigated the pathological features of and risk factors for atherosclerosis, underscoring the importance of drug combination therapies for the treatment of atherosclerotic diseases. We discuss herein mathematical models for quantifying the efficacy of the combination therapies and provide a systematic summary of drug combinations for the treatment of atherosclerosis. We also provide a detailed review of the latest advances in nanoparticle-based drug co-delivery systems for the treatment of atherosclerosis, focusing on the design of carriers with high biocompatibility and efficacy. By exploring the possibilities and challenges inherent to this approach, we aim to highlight cutting-edge technologies that can foster the development of innovative strategies, optimize drug co-administration, improve treatment outcomes, and reduce the burden of atherosclerosis-related morbidity and mortality on the healthcare system.
Collapse
Affiliation(s)
- Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Guosheng Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Hanlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| |
Collapse
|
2
|
Yu S, Zhu X, Zhao X, Li Y, Niu X, Chen Y, Ying J. Improvement of chronic metabolic inflammation and regulation of gut homeostasis: Tea as a potential therapy. Pharmacol Ther 2025; 269:108828. [PMID: 40020787 DOI: 10.1016/j.pharmthera.2025.108828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 12/27/2024] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
Chronic metabolic inflammation is a common mechanism linked to the development of metabolic disorders such as obesity, diabetes, and cardiovascular disease (CVD). Chronic metabolic inflammation often related to alterations in gut homeostasis, and pathological processes involve the activation of endotoxin receptors, metabolic reprogramming, mitochondrial dysfunction, and disruption of intestinal nuclear receptor activity. Recent investigations into homeostasis and chronic metabolic inflammation have revealed a novel mechanism which is characterized by a timing interaction involving multiple components and targets. This article explores the positive impact of tea consumption on metabolic health of populations, with a special focus on the improvement of inflammatory indicators and the regulation of gut microbiota. Studies showed that tea consumption is related to the enrichment of gut microbiota. The relative proportion of Firmicutes/Bacteroidetes (F/B) is altered, while the abundance of Lactobacillus, Bifidobacterium, and A. muciniphila increased significantly in most of the studies. Thus, tea consumption could provide potential protection from the development of chronic diseases by improving gut homeostasis and reducing chronic metabolic inflammation. The direct impact of tea on intestinal homeostasis primarily targets lipopolysaccharide (LPS)-related pathways. This includes reducing the synthesis of intestinal LPS, inhibiting LPS translocation, and preventing the binding of LPS to TLR4 receptors to block downstream inflammatory pathways. The TLR4/MyD88/NF-κB p65 pathway is crucial for anti-metaflammatory responses. The antioxidant properties of tea are linked to enhancing mitochondrial function and mitigating mitochondria-related inflammation by eliminating free radicals, inhibiting NLRP3 inflammasomes, and modulating Nrf2/ARE activity. Tea also contributes to safeguarding the intestinal barrier through various mechanisms, such as promoting the synthesis of short-chain fatty acids in the intestine, activating intestinal aryl hydrocarbon receptor (AhR) and farnesoid X receptor (FXR), and improving enteritis. Functional components that improve chronic metabolic inflammation include tea polyphenols, tea pigments, TPS, etc. Tea metabolites such as 4-Hydroxyphenylacetic acid and 3,4-Dihydroxyflavan derivatives, etc., also contribute to anti-chronic metabolic inflammation effects of tea consumption. The raw materials and processing technologies affect the functional component compositions of tea; therefore, consuming different types of tea may result in varying action characteristics and mechanisms. However, there is currently limited elaboration on this aspect. Future research should conduct in-depth studies on the mechanism of tea and its functional components in improving chronic metabolic inflammation. Researchers should pay attention to whether there are interactions between tea and other foods or drugs, explore safe and effective usage and dosage, and investigate whether there are individual differences in the tea-drinking population leading to different effects of tea intervention. Ultimately, the application of tea drinking could be a universal therapy for regulating intestinal homeostasis, anti-chronic metabolic inflammatory responses, and promoting metabolic health.
Collapse
Affiliation(s)
- Shiyi Yu
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China
| | - Xuan Zhu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Xiayu Zhao
- National Institute of Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yan Li
- National Institute of Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xinghe Niu
- Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; COFCO Nutrition and Health Research Institute, Beijing 102209, China
| | - Yinghua Chen
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China
| | - Jian Ying
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China.
| |
Collapse
|
3
|
Zhang R, Jiang Y, Zhang G, Zeng W, Suo Y, Zhang F, Jiang X. Mitochondrial DNA in atherosclerosis: Mechanisms, biomarker potential, and therapeutic perspectives. Int Immunopharmacol 2025; 152:114449. [PMID: 40073813 DOI: 10.1016/j.intimp.2025.114449] [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: 01/15/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease in which mitochondrial DNA (mtDNA) has emerged as a key contributor to its pathogenesis. We synthesized evidence from experimental and clinical studies showing that mtDNA damage, release, and mutation profoundly affect endothelial cells, macrophages, and vascular smooth muscle cells, thereby driving plaque initiation and progression. By activating immune signaling pathways-including cGAS-STING, NLRP3 inflammasome, and TLR9-mtDNA amplifies inflammation and oxidative stress, exacerbating atherosclerotic lesion development. We further highlight that mtDNA copy number variations and specific mtDNA mutations may serve as biomarkers for early atherosclerosis detection and risk stratification. In reviewing these data, we also discuss promising therapeutic interventions aimed at mitigating mtDNA damage, such as mitochondria-targeted antioxidants and enhanced mitophagy, which have shown preliminary efficacy in delaying plaque progression. Overall, this review underscores mtDNA's dual role as both a driver of atherosclerosis and a potential diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yifang Jiang
- School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenyun Zeng
- Department of Oncology, Ganzhou People 's Hospital, Jiangxi, China
| | - Yanrong Suo
- Department of Traditional Chinese Medicine, Ganzhou People's Hospital, Jiangxi, China
| | - Fayan Zhang
- Department of Rheumatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
4
|
Bonacina F, Zhang X, Manel N, Yvan-Charvet L, Razani B, Norata GD. Lysosomes in the immunometabolic reprogramming of immune cells in atherosclerosis. Nat Rev Cardiol 2025; 22:149-164. [PMID: 39304748 PMCID: PMC11835540 DOI: 10.1038/s41569-024-01072-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Lysosomes have a central role in the disposal of extracellular and intracellular cargo and also function as metabolic sensors and signalling platforms in the immunometabolic reprogramming of macrophages and other immune cells in atherosclerosis. Lysosomes can rapidly sense the presence of nutrients within immune cells, thereby switching from catabolism of extracellular material to the recycling of intracellular cargo. Such a fine-tuned degradative response supports the generation of metabolic building blocks through effectors such as mTORC1 or TFEB. By coupling nutrients to downstream signalling and metabolism, lysosomes serve as a crucial hub for cellular function in innate and adaptive immune cells. Lysosomal dysfunction is now recognized to be a hallmark of atherogenesis. Perturbations in nutrient-sensing and signalling have profound effects on the capacity of immune cells to handle cholesterol, perform phagocytosis and efferocytosis, and limit the activation of the inflammasome and other inflammatory pathways. Strategies to improve lysosomal function hold promise as novel modulators of the immunoinflammatory response associated with atherosclerosis. In this Review, we describe the crosstalk between lysosomal biology and immune cell function and polarization, with a particular focus on cellular immunometabolic reprogramming in the context of atherosclerosis.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Nicolas Manel
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU), Oncoage, Nice, France
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Giuseppe D Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
5
|
Yan J, Zhang L, Zeng Q, Qian Y, Li K, Liu X, Wu Y, Yan Y, Zhang H, Cheung S, Liu J, Sroka R, Wang X, Shi L. Mechanistic insights into 5-aminolevulinic acid photodynamic therapy for acne vulgaris: targeting lipogenesis via the OLR1-Wnt/β-catenin pathway. Mol Med 2025; 31:41. [PMID: 39905294 PMCID: PMC11792739 DOI: 10.1186/s10020-025-01104-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025] Open
Abstract
Acne vulgaris, a prevalent chronic inflammatory skin disorder, is often characterized by hyperactive sebaceous glands and excessive sebum production, presenting a significant therapeutic challenge. While 5-aminolevulinic acid photodynamic therapy (ALA-PDT) is clinically effective in treating moderate to severe acne, the molecular mechanisms underlying its therapeutic effects remain largely unexplored. In this study, we investigated the impact of ALA-PDT on lipid metabolism in an acne-like mouse model and in immortalized human sebocytes (XL-i-20), focusing on the role of the OLR1-Wnt/β-catenin pathway. We employed transcriptomic analysis, lipid staining, and gene silencing techniques to dissect the molecular interactions induced by ALA-PDT. Our findings revealed that ALA-PDT significantly reduces lipogenesis by upregulating OLR1, which in turn suppresses the SREBP1-FAS axis, thereby decreasing lipid accumulation in sebocytes. Furthermore, activation of the OLR1-Wnt/β-catenin pathway was essential for these lipogenic effects, as silencing OLR1 or activating Wnt/β-catenin signaling reversed lipogenesis inhibition. This study elucidates a novel mechanistic pathway in ALA-PDT-mediated acne treatment, highlighting OLR1 as a promising target for future therapeutic strategies.
Collapse
Affiliation(s)
- Jia Yan
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China
| | - Linglin Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China
- Laser-Forschungslabor, LIFE Center, University Hospital, Ludwig-Maximilian University, 82152, Planegg, Germany
| | - Qingyu Zeng
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China
| | - Yitao Qian
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Ke Li
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xiaojing Liu
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China
| | - Yun Wu
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China
| | - Yu Yan
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China
| | - Haiyan Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China
| | - Szeman Cheung
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jia Liu
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China
| | - Ronald Sroka
- Laser-Forschungslabor, LIFE Center, University Hospital, Ludwig-Maximilian University, 82152, Planegg, Germany.
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092,, China.
| | - Lei Shi
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai, 200040, P. R. China.
| |
Collapse
|
6
|
Guan X, Li H, Zhang L, Zhi H. Mechanisms of mitochondrial damage-associated molecular patterns associated with inflammatory response in cardiovascular diseases. Inflamm Res 2025; 74:18. [PMID: 39806203 DOI: 10.1007/s00011-025-01993-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: 10/20/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Cardiovascular diseases (CVDs) continue to be a substantial global healthcare burden despite considerable progress in therapies. The inflammatory response during the progression of CVD has attracted considerable attention. Mitochondria serve as the principal energy source for the heart. In cardiovascular illnesses, mitochondrial homeostasis is disrupted, accompanied by structural and functional impairments. During mitochondrial stress or injury, mitochondrial damage-associated molecular patterns (mtDAMPs), such as mitochondrial DNA, cardiolipin, N-formyl peptide, and adenosine triphosphate, are released to activate pattern recognition receptors and trigger immunological responses. Inflammatory responses mediated by mtDAMPs substantially contribute to the pathophysiology of cardiovascular illnesses. In this review, we discuss the molecular mechanisms by which different mtDAMPs control the inflammatory response, address the pathological consequences of mtDAMPs in inducing or exacerbating the inflammatory response in CVDs, and summarize potential therapeutic targets in relevant experimental studies. Preventing or reducing mtDAMP release may play a role in CVD progression by alleviating the inflammatory response.
Collapse
Affiliation(s)
- Xiuju Guan
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Haitao Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China
| | - Lijuan Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China.
| | - Hongwei Zhi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
7
|
Hu Y, Li Y, Luo Y, Wang N, Zheng Y. Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1 (LOX-1): A Potential Therapeutic Target in Ischemic Stroke. Transl Stroke Res 2024. [DOI: 10.1007/s12975-024-01307-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 01/03/2025]
|
8
|
Zhu L, Liao Y, Jiang B. Role of ROS and autophagy in the pathological process of atherosclerosis. J Physiol Biochem 2024; 80:743-756. [PMID: 39110405 DOI: 10.1007/s13105-024-01039-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/25/2024] [Indexed: 12/29/2024]
Abstract
Activation of autophagy and production of reactive oxygen species occur at various stages of atherosclerosis. To clarify the role and mechanism of autophagy and reactive oxygen species in atherosclerosis is of great significance to the prevention and treatment of atherosclerosis. Recent studies have shown that basal autophagy plays an important role in protecting cells from oxidative stress, reducing apoptosis and enhancing atherosclerotic plaque stability. Autophagy deficiency and excessive accumulation of reactive oxygen species can impair the function of endothelial cells, macrophages and smooth muscle cells, trigger autophagic cell death, and lead to instability and even rupture of plaques. However, the main signaling pathways regulating autophagy, the molecular mechanisms of autophagy and reactive oxygen species interaction, how they are initiated and distributed in plaques, and how they affect atherosclerosis progression, remain to be clarified. At present, there is no autophagy inducer used to treat atherosclerosis clinically. Therefore, it is urgent to clarify the mechanism of autophagy and find new targets for autophagy. Antioxidant agents generally have defects such as low reactive oxygen species scavenging efficiency and high cytotoxicity. Highly potent autophagy inducers and reactive oxygen species scavengers still need to be further developed and validated to provide more possibilities for innovative treatments for atherosclerosis.
Collapse
Affiliation(s)
- Liyuan Zhu
- Center of Clinical Pharmacology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yingnan Liao
- Department of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Bo Jiang
- Center of Clinical Pharmacology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Kaur P, Nazeer N, Gurjar V, Tiwari R, Mishra PK. Nanophotonic waveguide-based sensing of circulating cell-free mitochondrial DNA: implications for personalized medicine. Drug Discov Today 2024; 29:104086. [PMID: 38960132 DOI: 10.1016/j.drudis.2024.104086] [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: 11/30/2023] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Circulating cell-free mitochondrial DNA (ccf-mtDNA) has emerged as a promising biomarker, with potential implications for disease diagnosis. Changes in mtDNA, such as deletions, mutations or variations in the number of copies, have been associated with mitochondrial disorders, heart diseases, cancer and age-related non-communicable diseases. Previous methods, such as polymerase chain reaction-based approaches, next-generation sequencing and imaging-based techniques, have shown improved accuracy in identifying rare mtDNA variants or mutations, but they have limitations. This article explains the basic principles and benefits of using planar optical waveguide-based detection devices, which represent an advanced approach in the field of sensing.
Collapse
Affiliation(s)
- Prasan Kaur
- Division of Environmental Biotechnology, Genetics & Molecular Biology, ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Nazim Nazeer
- Division of Environmental Biotechnology, Genetics & Molecular Biology, ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Vikas Gurjar
- Division of Environmental Biotechnology, Genetics & Molecular Biology, ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Rajnarayan Tiwari
- Division of Environmental Biotechnology, Genetics & Molecular Biology, ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Pradyumna Kumar Mishra
- Division of Environmental Biotechnology, Genetics & Molecular Biology, ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India.
| |
Collapse
|
10
|
Gąssowska-Dobrowolska M, Olech-Kochańczyk G, Culmsee C, Adamczyk A. Novel Insights into Parkin-Mediated Mitochondrial Dysfunction and "Mito-Inflammation" in α-Synuclein Toxicity. The Role of the cGAS-STING Signalling Pathway. J Inflamm Res 2024; 17:4549-4574. [PMID: 39011416 PMCID: PMC11249072 DOI: 10.2147/jir.s468609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/22/2024] [Indexed: 07/17/2024] Open
Abstract
The prevalence of age-related neurodegenerative diseases, such as Parkinson's disease (PD) and related disorders continues to grow worldwide. Increasing evidence links intracellular inclusions of misfolded alpha-synuclein (α-syn) aggregates, so-called Lewy bodies (LB) and Lewy neuritis, to the progressive pathology of PD and other synucleinopathies. Our previous findings established that α-syn oligomers induce S-nitrosylation and deregulation of the E3-ubiquitin ligase Parkin, leading to mitochondrial disturbances in neuronal cells. The accumulation of damaged mitochondria as a consequence, together with the release of mitochondrial-derived damage-associated molecular patterns (mtDAMPs) could activate the innate immune response and induce neuroinflammation ("mito-inflammation"), eventually accelerating neurodegeneration. However, the molecular pathways that transmit pro-inflammatory signals from damaged mitochondria are not well understood. One of the proposed pathways could be the cyclic GMP-AMP synthase (cGAS) - stimulator of interferon genes (STING) (cGAS-STING) pathway, which plays a pivotal role in modulating the innate immune response. It has recently been suggested that cGAS-STING deregulation may contribute to the development of various pathological conditions. Especially, its excessive engagement may lead to neuroinflammation and appear to be essential for the development of neurodegenerative brain diseases, including PD. However, the precise molecular mechanisms underlying cGAS-STING pathway activation in PD and other synucleinopathies are not fully understood. This review focuses on linking mitochondrial dysfunction to neuroinflammation in these disorders, particularly emphasizing the role of the cGAS-STING signaling. We propose the cGAS-STING pathway as a critical driver of inflammation in α-syn-dependent neurodegeneration and hypothesize that cGAS-STING-driven "mito-inflammation" may be one of the key mechanisms promoting the neurodegeneration in PD. Understanding the molecular mechanisms of α-syn-induced cGAS-STING-associated "mito-inflammation" in PD and related synucleinopathies may contribute to the identification of new targets for the treatment of these disorders.
Collapse
Affiliation(s)
| | - Gabriela Olech-Kochańczyk
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
- Center for Mind Brain and Behavior - CMBB, University of Marburg, Marburg, Germany
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
11
|
Liu Y, Zhang B, Duan R, Liu Y. Mitochondrial DNA Leakage and cGas/STING Pathway in Microglia: Crosstalk Between Neuroinflammation and Neurodegeneration. Neuroscience 2024; 548:1-8. [PMID: 38685462 DOI: 10.1016/j.neuroscience.2024.04.009] [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: 02/03/2024] [Revised: 04/04/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
Neurodegenerative diseases, characterized by abnormal deposition of misfolded proteins, often present with progressive loss of neurons. Chronic neuroinflammation is a striking hallmark of neurodegeneration. Microglia, as the primary immune cells in the brain, is the main type of cells that participate in the formation of inflammatory microenvironment. Cytoplasmic free mitochondrial DNA (mtDNA), a common component of damage-associated molecular patterns (DAMPs), can activate the cGas/stimulator of interferon genes (STING) signalling, which subsequently produces type I interferon and proinflammatory cytokines. There are various sources of free mtDNA in microglial cytoplasm, but mitochondrial oxidative stress accumulation plays the vital role. The upregulation of cGas/STING pathway in microglia contributes to the abnormal and persistent microglial activation, accompanied by excessive secretion of neurotoxic inflammatory mediators such as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), which exacerbates the damage of neurons and promotes the development of neurodegeneration. Currently, novel therapeutic approaches need to be found to delay the progression of neurodegenerative disorders, and regulation of the cGas/STING signaling in microglia may be a potential target.
Collapse
Affiliation(s)
- Yuqian Liu
- Qilu Hospital of Shandong University, Jinan, China
| | - Bohan Zhang
- Qilu Hospital of Shandong University, Jinan, China
| | - Ruonan Duan
- Qilu Hospital of Shandong University, Jinan, China.
| | - Yiming Liu
- Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
12
|
Gurubaran IS. Mitochondrial damage and clearance in retinal pigment epithelial cells. Acta Ophthalmol 2024; 102 Suppl 282:3-53. [PMID: 38467968 DOI: 10.1111/aos.16661] [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: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
Age-related macular degeneration (AMD) is a devastating eye disease that causes permanent vision loss in the central part of the retina, known as the macula. Patients with such severe visual loss face a reduced quality of life and are at a 1.5 times greater risk of death compared to the general population. Currently, there is no cure for or effective treatment for dry AMD. There are several mechanisms thought to underlie the disease, for example, ageing-associated chronic oxidative stress, mitochondrial damage, harmful protein aggregation and inflammation. As a way of gaining a better understanding of the molecular mechanisms behind AMD and thus developing new therapies, we have created a peroxisome proliferator-activated receptor gamma coactivator 1-alpha and nuclear factor erythroid 2-related factor 2 (PGC1α/NFE2L2) double-knockout (dKO) mouse model that mimics many of the clinical features of dry AMD, including elevated levels of oxidative stress markers, damaged mitochondria, accumulating lysosomal lipofuscin and extracellular drusen-like structures in retinal pigment epithelial cells (RPE). In addition, a human RPE cell-based model was established to examine the impact of non-functional intracellular clearance systems on inflammasome activation. In this study, we found that there was a disturbance in the autolysosomal machinery responsible for clearing mitochondria in the RPE cells of one-year-old PGC1α/NFE2L2-deficient mice. The confocal immunohistochemical analysis revealed an increase in autophagosome marker microtubule-associated proteins 1A/1B light chain 3B (LC3B) as well as multiple mitophagy markers such as PTE-induced putative kinase 1 (PINK1) and E3 ubiquitin ligase (PARKIN), along with signs of damaged mitochondria. However, no increase in autolysosome formation was detected, nor was there a colocalization of the lysosomal marker LAMP2 or the mitochondrial marker, ATP synthase β. There was an upregulation of late autolysosomal fusion Ras-related protein (Rab7) in the perinuclear space of RPE cells, together with autofluorescent aggregates. Additionally, we observed an increase in the numbers of Toll-like receptors 3 and 9, while those of NOD-like receptor 3 were decreased in PGC1α/NFE2L2 dKO retinal specimens compared to wild-type animals. There was a trend towards increased complement component C5a and increased involvement of the serine protease enzyme, thrombin, in enhancing the terminal pathway producing C5a, independent of C3. The levels of primary acute phase C-reactive protein and receptor for advanced glycation end products were also increased in the PGC1α/NFE2L2 dKO retina. Furthermore, selective proteasome inhibition with epoxomicin promoted both nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondrial-mediated oxidative stress, leading to the release of mitochondrial DNA to the cytosol, resulting in potassium efflux-dependent activation of the absent in melanoma 2 (AIM2) inflammasome and the subsequent secretion of interleukin-1β in ARPE-19 cells. In conclusion, the data suggest that there is at least a relative decrease in mitophagy, increases in the amounts of C5 and thrombin and decreased C3 levels in this dry AMD-like model. Moreover, selective proteasome inhibition evoked mitochondrial damage and AIM2 inflammasome activation in ARPE-19 cells.
Collapse
Affiliation(s)
- Iswariyaraja Sridevi Gurubaran
- Department of Medicine, Clinical Medicine Unit, University of Eastern Finland Institute of Clinical Medicine, Kuopio, Northern Savonia, Finland
| |
Collapse
|
13
|
Bagheri B, Khatibiyan Feyzabadi Z, Nouri A, Azadfallah A, Mahdizade Ari M, Hemmati M, Darban M, Alavi Toosi P, Banihashemian SZ. Atherosclerosis and Toll-Like Receptor4 (TLR4), Lectin-Like Oxidized Low-Density Lipoprotein-1 (LOX-1), and Proprotein Convertase Subtilisin/Kexin Type9 (PCSK9). Mediators Inflamm 2024; 2024:5830491. [PMID: 38445291 PMCID: PMC10914434 DOI: 10.1155/2024/5830491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/31/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Atherosclerosis is a leading cause of death in the world. A significant body of evidence suggests that inflammation and various players are implicated and have pivotal roles in the formation of atherosclerotic plaques. Toll-like receptor 4 (TLR4) is linked with different stages of atherosclerosis. This receptor is highly expressed in the endothelial cells (ECs) and atherosclerotic plaques. TLR4 activation can lead to the production of inflammatory cytokines and related responses. Lectin-like oxidized low-density lipoprotein-1 (LOX-1), an integral membrane glycoprotein with widespread expression on the ECs, is involved in atherosclerosis and has some common pathways with TLR4 in atherosclerotic lesions. In addition, proprotein convertase subtilisin/kexin type9 (PCSK9), which is a regulatory enzyme with different roles in cholesterol uptake, is implicated in atherosclerosis. At present, TLR4, PCSK9, and LOX-1 are increasingly acknowledged as key players in the pathogenesis of atherosclerotic cardiovascular diseases. Herein, we presented the current evidence on the structure, functions, and roles of TLR4, PCSK9, and LOX-1 in atherosclerosis.
Collapse
Affiliation(s)
- Bahador Bagheri
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Ahmad Nouri
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Azadfallah
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahyar Mahdizade Ari
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maral Hemmati
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahboubeh Darban
- Department of Internal Medicine, Kowsar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Parisa Alavi Toosi
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | |
Collapse
|
14
|
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y, Dou X. Orientin alleviates ox-LDL-induced oxidative stress, inflammation and apoptosis in human vascular endothelial cells by regulating Sestrin 1 (SESN1)-mediated autophagy. J Mol Histol 2024; 55:109-120. [PMID: 38165567 DOI: 10.1007/s10735-023-10176-z] [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: 07/21/2023] [Accepted: 11/04/2023] [Indexed: 01/04/2024]
Abstract
Endothelial cells are a crucial component of the vessel-tissue wall and exert an important role in atherosclerosis (AS). To explore the role of Orientin in AS, human vascular endothelial cells (HUVECs) were induced by oxidized low-density lipoprotein (ox-LDL) to simulate the vascular endothelial injury during AS. Cell viability was detected by CCK-8 assay. Oxidative stress and inflammation related markers were measured using kits, RT-qPCR or western blot. Besides, cell apoptosis was assessed with TUNEL staining and cell autophagy was evaluated by LC3 immunofluorescent staining. Additionally, western blot was utilized to evaluate the expression of Sestrin 1 (SESN1) and proteins in AMPK/mTOR signaling. Afterwards, SESN1 was silenced to determine the expression of autophagy-related proteins. The further application of autophagy inhibitor 3-methyladenine (3-MA) was used to clarify the regulatory mechanism of Orientin on autophagy. Results showed that the decreased viability of HUVECs caused by ox-LDL induction was elevated by Orientin. Oxidative stress and inflammation were also attenuated after Orientin addition in HUVECs under ox-LDL condition. Moreover, Orientin suppressed apoptosis and induced autophagy of HUVECs stimulated by ox-LDL, accompanied by enhanced level of phospho (p)-AMPK and declined level of p-mTOR. Interestingly, SESN1 level was elevated by Orientin, and SESN1 depletion alleviated autophagy and reduced p-AMPK expression but enhanced p-mTOR expression. The further experiments indicated that SESN1 silencing or 3-MA addition reversed the inhibitory effects of Orientin on the oxidative stress, inflammation and apoptosis of HUVECs. Collectively, Orientin could induce autophagy by activating SESN1 expression, thereby regulating AMPK/mTOR signaling in ox-LDL-induced HUVECs.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China.
| | - Yongcheng Zhao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Chunwei Xiao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Zhanfa Sun
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Yuan Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Xueyong Dou
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| |
Collapse
|
15
|
Velpuri P, Rai V, Agrawal DK. Role of sirtuins in attenuating plaque vulnerability in atherosclerosis. Mol Cell Biochem 2024; 479:51-62. [PMID: 36952068 PMCID: PMC10034899 DOI: 10.1007/s11010-023-04714-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023]
Abstract
Atherosclerosis is characterized by the development of intimal plaque, thrombosis, and stenosis of the vessel lumen causing decreased blood flow and hypoxia precipitating angina. Chronic inflammation in the stable plaque renders it unstable and rupture of unstable plaques results in the formation of emboli leading to hypoxia/ischemia to the organs by occluding the terminal branches and precipitate myocardial infarction and stroke. Such delibitating events could be controlled by the strategies that prevent plaque development or plaque stabilization. Despite the use of statins to stabilize plaques, there is a need for novel targets due to continuously increasing cases of cardiovascular events. Sirtuins (SIRTs), a family of signaling proteins, are involved in sustaining genome integrity, DNA damage response and repair, modulating oxidative stress, aging, inflammation, and energy metabolism. SIRTs play a critical role in modulating inflammation and involves in the development and progression of atherosclerosis. The role of SIRTs in relation to atherosclerosis and plaque vulnerability is scarcely discussed in the literature. Since SIRTs regulate oxidative stress, inflammation, and aging, they may also regulate plaque progression and vulnerability as these molecular mechanisms underlie the pathogenesis of plaque development, progression, and vulnerability. This review critically discusses the role of SIRTs in plaque progression and vulnerability and the possibility of targeting SIRTs to attenuate plaque rupture, focusing on the highlights in genomics, molecular pathways, and cell types involved in the underlying pathophysiology.
Collapse
Affiliation(s)
- Prathosh Velpuri
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
16
|
Abstract
According to the endosymbiotic theory, most of the DNA of the original bacterial endosymbiont has been lost or transferred to the nucleus, leaving a much smaller (∼16 kb in mammals), circular molecule that is the present-day mitochondrial DNA (mtDNA). The ability of mtDNA to escape mitochondria and integrate into the nuclear genome was discovered in budding yeast, along with genes that regulate this process. Mitochondria have emerged as key regulators of innate immunity, and it is now recognized that mtDNA released into the cytoplasm, outside of the cell, or into circulation activates multiple innate immune signaling pathways. Here, we first review the mechanisms through which mtDNA is released into the cytoplasm, including several inducible mitochondrial pores and defective mitophagy or autophagy. Next, we cover how the different forms of released mtDNA activate specific innate immune nucleic acid sensors and inflammasomes. Finally, we discuss how intracellular and extracellular mtDNA release, including circulating cell-free mtDNA that promotes systemic inflammation, are implicated in human diseases, bacterial and viral infections, senescence and aging.
Collapse
Affiliation(s)
- Laura E Newman
- Salk Institute for Biological Studies, La Jolla, California, USA;
| | - Gerald S Shadel
- Salk Institute for Biological Studies, La Jolla, California, USA;
| |
Collapse
|
17
|
Chen S, Liao Z, Xu P. Mitochondrial control of innate immune responses. Front Immunol 2023; 14:1166214. [PMID: 37325622 PMCID: PMC10267745 DOI: 10.3389/fimmu.2023.1166214] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023] Open
Abstract
Mitochondria are versatile organelles and essential components of numerous biological processes such as energy metabolism, signal transduction, and cell fate determination. In recent years, their critical roles in innate immunity have come to the forefront, highlighting impacts on pathogenic defense, tissue homeostasis, and degenerative diseases. This review offers an in-depth and comprehensive examination of the multifaceted mechanisms underlying the interactions between mitochondria and innate immune responses. We will delve into the roles of healthy mitochondria as platforms for signalosome assembly, the release of mitochondrial components as signaling messengers, and the regulation of signaling via mitophagy, particularly to cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling and inflammasomes. Furthermore, the review will explore the impacts of mitochondrial proteins and metabolites on modulating innate immune responses, the polarization of innate immune cells, and their implications on infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Shasha Chen
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Zhiyong Liao
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Pinglong Xu
- Institute of Intelligent Medicine, Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University (HIC-ZJU), Hangzhou, China
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Borah S, Mishra R, Dey S, Suchanti S, Bhowmick NA, Giri B, Haldar S. Prognostic Value of Circulating Mitochondrial DNA in Prostate Cancer and Underlying Mechanism. Mitochondrion 2023; 71:40-49. [PMID: 37211294 DOI: 10.1016/j.mito.2023.05.005] [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: 01/09/2023] [Revised: 04/03/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
Circulating DNAs are considered as degraded DNA fragments of approximately 50-200 bp, found in blood plasma, consisting of cell-free mitochondrial and nuclear DNA. Such cell-free DNAs in the blood are found to be altered in different pathological conditions including lupus, heart disease, and malignancies. While nuclear DNAs are being used and being developed as a powerful clinical biomarker in liquid biopsies, mitochondrial DNAs (mtDNAs) are associated with inflammatory conditions including cancer progression. Patients with cancer including prostate cancer are found to have measurable concentrations of mitochondrial DNA in circulation in comparison with healthy controls. The plasma content of mitochondrial DNA is dramatically elevated in both prostate cancer patients and mouse models treated with the chemotherapeutic drug. Cell-free mtDNA, in its oxidized form, induced a pro-inflammatory condition and activates NLRP3-mediated inflammasome formation which causes IL-1β-mediated activation of growth factors. On the other hand, interacting with TLR9, mtDNAs trigger NF-κB-mediated complement C3a positive feedback paracrine loop and activate pro-proliferating signaling through upregulating AKT, ERK, and Bcl2 in the prostate tumor microenvironment. In this review, we discuss the growing evidence supporting cell-free mitochondrial DNA copy number, size, and mutations in mtDNA genes as potential prognostic biomarkers in different cancers and targetable prostate cancer therapeutic candidates impacting stromal-epithelial interactions essential for chemotherapy response.
Collapse
Affiliation(s)
- Supriya Borah
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
| | - Rajeev Mishra
- Department of Life Sciences, CSJM University, Kanpur, Uttar Pradesh 208012, India
| | - Sananda Dey
- Department of Research, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Surabhi Suchanti
- Department of Biosciences, Manipal University Jaipur, Rajasthan 303007, India
| | - Neil A Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA; Department of Research, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, Malda 732103, India.
| | - Subhash Haldar
- Department of Biochemistry, Bose Institute, Kolkata 700091, India.
| |
Collapse
|
19
|
Xiao S, Song X, Zheng M, Cao X, Ai G, Li B, Zhao G, Yuan H. Interleukin-37 ameliorates atherosclerosis by regulating autophagy-mediated endothelial cell apoptosis and inflammation. Int Immunopharmacol 2023; 118:110098. [PMID: 37023695 DOI: 10.1016/j.intimp.2023.110098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/18/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease. Endothelial dysfunction is the initiating factor of atherosclerosis. Although much work has been done on the antiatherosclerotic effects of interleukin-37 (IL-37), the exact mechanism is still not fully understood. The aim of this study was to investigate whether IL-37 attenuates atherosclerosis by protecting endothelial cells and to confirm whether autophagy plays a role in this effect. In apolipoprotein E knockout (ApoE-/-) mice fed with a high fat diet, IL-37 treatment significantly attenuated progression of atherosclerotic plaques, reduced endothelial cell apoptosis and inflammasome activation. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to establish an endothelial dysfunction model. We observed that IL-37 alleviated ox-LDL-induced endothelial cell inflammation and dysfunction, as evidenced by decreased nod-like receptor pyrin domain-containing 3 (NLRP3) inflammasome activation, ROS production, apoptosis rate and secretion of inflammatory cytokines IL-1β and TNF-α. Furthermore, IL-37 could activate autophagy in endothelial cells, which is characterized by the upregulation of LC3II/LC3I, the downregulation of p62 and an increase in autophagosomes. The autophagy inhibitor 3-Methyladenine (3-MA) dramatically reversed the promotion of autophagy and the protective effect of IL-37 against endothelial injury. Our data illustrate that IL-37 alleviated inflammation and apoptosis of atherosclerotic endothelial cells by enhancing autophagy. The current study provides new insights and promising therapeutic strategies for atherosclerosis.
Collapse
Affiliation(s)
- Shengyang Xiao
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoning Song
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Man Zheng
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xinran Cao
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Guo Ai
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Baona Li
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Gang Zhao
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
20
|
Ma DW, Ha J, Yoon KS, Kang I, Choi TG, Kim SS. Innate Immune System in the Pathogenesis of Non-Alcoholic Fatty Liver Disease. Nutrients 2023; 15:2068. [PMID: 37432213 DOI: 10.3390/nu15092068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 07/12/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent condition characterized by lipid accumulation in hepatocytes with low alcohol consumption. The development of sterile inflammation, which occurs in response to a range of cellular stressors or injuries, has been identified as a major contributor to the pathogenesis of NAFLD. Recent studies of the pathogenesis of NAFLD reported the newly developed roles of damage-associated molecular patterns (DAMPs). These molecules activate pattern recognition receptors (PRRs), which are placed in the infiltrated neutrophils, dendritic cells, monocytes, or Kupffer cells. DAMPs cause the activation of PRRs, which triggers a number of immunological responses, including the generation of cytokines that promote inflammation and the localization of immune cells to the site of the damage. This review provides a comprehensive overview of the impact of DAMPs and PRRs on the development of NAFLD.
Collapse
Affiliation(s)
- Dae Won Ma
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
21
|
García-Beltrán O, Urrutia PJ, Núñez MT. On the Chemical and Biological Characteristics of Multifunctional Compounds for the Treatment of Parkinson's Disease. Antioxidants (Basel) 2023; 12:214. [PMID: 36829773 PMCID: PMC9952574 DOI: 10.3390/antiox12020214] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Protein aggregation, mitochondrial dysfunction, iron dyshomeostasis, increased oxidative damage and inflammation are pathognomonic features of Parkinson's disease (PD) and other neurodegenerative disorders characterized by abnormal iron accumulation. Moreover, the existence of positive feed-back loops between these pathological components, which accelerate, and sometimes make irreversible, the neurodegenerative process, is apparent. At present, the available treatments for PD aim to relieve the symptoms, thus improving quality of life, but no treatments to stop the progression of the disease are available. Recently, the use of multifunctional compounds with the capacity to attack several of the key components of neurodegenerative processes has been proposed as a strategy to slow down the progression of neurodegenerative processes. For the treatment of PD specifically, the necessary properties of new-generation drugs should include mitochondrial destination, the center of iron-reactive oxygen species interaction, iron chelation capacity to decrease iron-mediated oxidative damage, the capacity to quench free radicals to decrease the risk of ferroptotic neuronal death, the capacity to disrupt α-synuclein aggregates and the capacity to decrease inflammatory conditions. Desirable additional characteristics are dopaminergic neurons to lessen unwanted secondary effects during long-term treatment, and the inhibition of the MAO-B and COMPT activities to increase intraneuronal dopamine content. On the basis of the published evidence, in this work, we review the molecular basis underlying the pathological events associated with PD and the clinical trials that have used single-target drugs to stop the progress of the disease. We also review the current information on multifunctional compounds that may be used for the treatment of PD and discuss the chemical characteristics that underlie their functionality. As a projection, some of these compounds or modifications could be used to treat diseases that share common pathology features with PD, such as Friedreich's ataxia, Multiple sclerosis, Huntington disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Olimpo García-Beltrán
- Facultad de Ciencias Naturales y Matemáticas, Universidad de Ibagué, Carrera 22 Calle 67, Ibagué 730002, Colombia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, General Gana 1702, Santiago 8370854, Chile
| | - Pamela J. Urrutia
- Faculty of Medicine and Science, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
| | - Marco T. Núñez
- Faculty of Sciences, Universidad de Chile, Las Palmeras 3425, Santiago 7800024, Chile
| |
Collapse
|
22
|
Fathieh S, Grieve SM, Negishi K, Figtree GA. Potential Biological Mediators of Myocardial and Vascular Complications of Air Pollution-A State-of-the-Art Review. Heart Lung Circ 2023; 32:26-42. [PMID: 36585310 DOI: 10.1016/j.hlc.2022.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 12/29/2022]
Abstract
Ambient air pollution is recognised globally as a significant contributor to the burden of cardiovascular diseases. The evidence from both human and animal studies supporting the cardiovascular impact of exposure to air pollution has grown substantially, implicating numerous pathophysiological pathways and related signalling mediators. In this review, we summarise the list of activated mediators for each pathway that lead to myocardial and vascular injury in response to air pollutants. We performed a systematic search of multiple databases, including articles between 1990 and Jan 2022, summarising the evidence for activated pathways in response to each significant air pollutant. Particulate matter <2.5 μm (PM2.5) was the most studied pollutant, followed by particulate matter between 2.5 μm-10 μm (PM10), nitrogen dioxide (NO2) and ozone (O3). Key pathogenic pathways that emerged included activation of systemic and local inflammation, oxidative stress, endothelial dysfunction, and autonomic dysfunction. We looked at how potential mediators of each of these pathways were linked to both cardiovascular disease and air pollution and included the overlapping mediators. This review illustrates the complex relationship between air pollution and cardiovascular diseases, and discusses challenges in moving beyond associations, towards understanding causal contributions of specific pathways and markers that may inform us regarding an individual's exposure, response, and likely risk.
Collapse
Affiliation(s)
- Sina Fathieh
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Stuart M Grieve
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Radiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Kazuaki Negishi
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia; Department of Cardiology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan; Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia.
| |
Collapse
|
23
|
Bikomeye JC, Terwoord JD, Santos JH, Beyer AM. Emerging mitochondrial signaling mechanisms in cardio-oncology: beyond oxidative stress. Am J Physiol Heart Circ Physiol 2022; 323:H702-H720. [PMID: 35930448 PMCID: PMC9529263 DOI: 10.1152/ajpheart.00231.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 12/27/2022]
Abstract
Many anticancer therapies (CTx) have cardiotoxic side effects that limit their therapeutic potential and cause long-term cardiovascular complications in cancer survivors. This has given rise to the field of cardio-oncology, which recognizes the need for basic, translational, and clinical research focused on understanding the complex signaling events that drive CTx-induced cardiovascular toxicity. Several CTx agents cause mitochondrial damage in the form of mitochondrial DNA deletions, mutations, and suppression of respiratory function and ATP production. In this review, we provide a brief overview of the cardiovascular complications of clinically used CTx agents and discuss current knowledge of local and systemic secondary signaling events that arise in response to mitochondrial stress/damage. Mitochondrial oxidative stress has long been recognized as a contributor to CTx-induced cardiotoxicity; thus, we focus on emerging roles for mitochondria in epigenetic regulation, innate immunity, and signaling via noncoding RNAs and mitochondrial hormones. Because data exploring mitochondrial secondary signaling in the context of cardio-oncology are limited, we also draw upon clinical and preclinical studies, which have examined these pathways in other relevant pathologies.
Collapse
Affiliation(s)
- Jean C Bikomeye
- Doctorate Program in Public and Community Health, Division of Epidemiology and Social Sciences, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Janée D Terwoord
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Biomedical Sciences Department, Rocky Vista University, Ivins, Utah
| | - Janine H Santos
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Andreas M Beyer
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
24
|
Yu W, Ilyas I, Aktar N, Xu S. A review on therapeutical potential of paeonol in atherosclerosis. Front Pharmacol 2022; 13:950337. [PMID: 35991897 PMCID: PMC9385965 DOI: 10.3389/fphar.2022.950337] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
The morbidity and mortality of atherosclerotic cardiovascular disease (ASCVD) is increasing year by year. Cortex Moutan is a traditional Chinese medicinal herb that has been widely used for thousands of years to treat a wide variety of diseases in Eastern countries due to its heat-clearing and detoxifying effects. Paeonol is a bioactive monomer extracted from Cortex Moutan, which has anti-atherosclerotic effects. In this article, we reviewed the pharmacological effects of paeonol against experimental atherosclerosis, as well as its protective effects on vascular endothelial cells, smooth muscle cells, macrophages, platelets, and other important cell types. The pleiotropic effects of paeonol in atherosclerosis suggest that it can be a promising therapeutic agent for atherosclerosis and its complications. Large-scale randomized clinical trials are warranted to elucidate whether paeonol are effective in patients with ASCVD.
Collapse
Affiliation(s)
- Wei Yu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, Anhui, China
- Anhui Renovo Pharmaceutical Co., Ltd., Hefei, Anhui, China
- *Correspondence: Wei Yu, ; Suowen Xu,
| | - Iqra Ilyas
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Nasrin Aktar
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- *Correspondence: Wei Yu, ; Suowen Xu,
| |
Collapse
|
25
|
Becker PH, Le Guillou E, Duque M, Blondel A, Gons C, Ben Souna H, Imbard A, Fournier N, Gaignard P, Thérond P. Cholesterol accumulation induced by acetylated LDL exposure modifies the enzymatic activities of the TCA cycle without impairing the respiratory chain functionality in macrophages. Biochimie 2022; 200:87-98. [PMID: 35618159 DOI: 10.1016/j.biochi.2022.05.011] [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: 11/13/2021] [Revised: 03/31/2022] [Accepted: 05/19/2022] [Indexed: 11/27/2022]
Abstract
The unregulated uptake of modified low-density lipoproteins (LDL) by macrophages leads to foam cell formation, promoting atherosclerotic plaque progression. The cholesterol efflux capacity of macrophages by the ATP-Binding Cassette transporters depends on the ATP mitochondrial production. Therefore, the mitochondrial function maintenance is crucial in limiting foam cell formation. Thus, we aimed to investigate the mechanisms involved in the mitochondrial dysfunction that may occur in cholesterol-laden macrophages. We incubated THP-1 macrophages with acetylated LDL (acLDL) to obtain cholesterol-laden cells or with mildly oxidized LDL (oxLDL) to generate cholesterol- and oxidized lipids-laden cells. Cellular cholesterol content was measured in each condition. Mitochondrial function was evaluated by measurement of several markers of energetic metabolism, oxidative phosphorylation, oxidative stress, mitochondrial biogenesis and dynamics. OxLDL-exposed macrophages exhibited a significantly reduced mitochondrial respiration and complexes I and III activities, associated to an oxidative stress state and a reduced mitochondrial DNA copy number. Meanwhile, acLDL-exposed macrophages featured an efficient oxidative phosphorylation despite the decreased activities of aconitase, isocitrate dehydrogenase and α-ketoglutarate dehydrogenase. Our study revealed that mitochondrial function was differently impacted according to the nature of modified LDL. Exposure to cholesterol and oxidized lipids carried by oxLDL leads to a mitochondrial dysfunction in macrophages, affecting the mitochondrial respiratory chain functional capacity, whereas the cellular cholesterol enrichment induced by acLDL exposure results in a tricarboxylic acid cycle shunt while maintaining mitochondrial energetic production, reflecting a metabolic adaptation to cholesterol intake. These new mechanistic insights are of direct relevance to the understanding of the mitochondrial dysfunction in foam cells.
Collapse
Affiliation(s)
- Pierre-Hadrien Becker
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France.
| | - Edouard Le Guillou
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Mathilde Duque
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Amélie Blondel
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Camille Gons
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Hajar Ben Souna
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Apolline Imbard
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Necker-Enfants Malades, AP-HP, Laboratoire de Biochimie Métabolique, Paris, 75015, France
| | - Natalie Fournier
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Européen Georges Pompidou, AP-HP, Laboratoire de Biochimie, Paris, 75015, France
| | - Pauline Gaignard
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Patrice Thérond
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| |
Collapse
|
26
|
Shaker ME. The contribution of sterile inflammation to the fatty liver disease and the potential therapies. Biomed Pharmacother 2022; 148:112789. [PMID: 35272137 DOI: 10.1016/j.biopha.2022.112789] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/02/2022] Open
Abstract
Hepatic inflammation is prevalent in several metabolic liver diseases. Recent scientific advances about the pathogenesis of metabolic liver diseases showed an emerging role of several damage-associated molecular patterns (DAMPs), including DNA, high-mobility group box 1 (HMGB1), ATP and uric acid. For these DAMPs to induce inflammation, they should stimulate pattern recognition receptors (PRRs), which are located in the hepatic immune cells like resident Kupffer cells, infiltrated neutrophils, monocytes or dendritic cells. As a consequence, proinflammatory cytokines like interleukins (ILs)-1β and 18 alongside tumor necrosis factor (TNF)-α are overproduced and released, leading to pronounced hepatic inflammation and cellular death. This review highlights the contribution of these DAMPs and PRRs in the settings of alcoholic and nonalcoholic steatohepatitis. The review also summarizes the therapeutic usefulness of targeting NLR family pyrin domain containing 3 (NLRP3)-inflammasome, Toll-like receptors (TLRs) 4 and 9, IL-1 receptor (IL-1R), caspase 1, uric acid and GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) in these hepatic inflammatory disorders.
Collapse
Affiliation(s)
- Mohamed E Shaker
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Aljouf, Saudi Arabia.
| |
Collapse
|
27
|
Guo Y, Tsai HI, Zhang L, Zhu H. Mitochondrial DNA on Tumor-Associated Macrophages Polarization and Immunity. Cancers (Basel) 2022; 14:1452. [PMID: 35326602 PMCID: PMC8946090 DOI: 10.3390/cancers14061452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/26/2022] [Accepted: 03/09/2022] [Indexed: 12/04/2022] Open
Abstract
As the richest immune cells in most tumor microenvironments (TMEs), tumor-associated macrophages (TAMs) play an important role in tumor development and treatment sensitivity. The phenotypes and functions of TAMs vary according to their sources and tumor progression. Different TAM phenotypes display distinct behaviors in terms of tumor immunity and are regulated by intracellular and exogenous molecules. Additionally, dysfunctional and oxidatively stressed mitochondrial-derived mitochondrial DNA (mtDNA) plays an important role in remodeling the phenotypes and functions of TAMs. This article reviews the interactions between mtDNA and TAMs in the TME and further discusses the influence of their performance on tumor genesis and development.
Collapse
Affiliation(s)
- Yaxin Guo
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Hsiang-i Tsai
- Laboratory of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Lirong Zhang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Haitao Zhu
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
- Laboratory of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| |
Collapse
|
28
|
Lin L, Zhang MX, Zhang L, Zhang D, Li C, Li YL. Autophagy, Pyroptosis, and Ferroptosis: New Regulatory Mechanisms for Atherosclerosis. Front Cell Dev Biol 2022; 9:809955. [PMID: 35096837 PMCID: PMC8793783 DOI: 10.3389/fcell.2021.809955] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by the gradual buildup of plaques within the vessel wall of middle-sized and large arteries. The occurrence and development of atherosclerosis and the rupture of plaques are related to the injury of vascular cells, including endothelial cells, smooth muscle cells, and macrophages. Autophagy is a subcellular process that plays an important role in the degradation of proteins and damaged organelles, and the autophagy disorder of vascular cells is closely related to atherosclerosis. Pyroptosis is a proinflammatory form of regulated cell death, while ferroptosis is a form of regulated nonapoptotic cell death involving overwhelming iron-dependent lipid peroxidation. Both of them exhibit distinct features from apoptosis, necrosis, and autophagy in morphology, biochemistry, and genetics. However, a growing body of evidence suggests that pyroptosis and ferroptosis interact with autophagy and participate in the development of cancers, degenerative brain diseases and cardiovascular diseases. This review updated the current understanding of autophagy, pyroptosis, and ferroptosis, finding potential links and their effects on atherogenesis and plaque stability, thus providing ways to develop new pharmacological strategies to address atherosclerosis and stabilize vulnerable, ruptured plaques.
Collapse
Affiliation(s)
- Lin Lin
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mu-Xin Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dan Zhang
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun-Lun Li
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
29
|
Bradshaw JL, Cushen SC, Phillips NR, Goulopoulou S. Circulating cell-free mitochondrial DNA in pregnancy. Physiology (Bethesda) 2022; 37:0. [PMID: 35001655 DOI: 10.1152/physiol.00037.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Circulating cell-free mitochondrial DNA (ccf-mtDNA) released upon cell injury or death stimulates diverse pattern recognition receptors to activate innate immune responses and initiate systemic inflammation. In this review, we discuss the temporal changes of ccf-mtDNA during pregnancy and its potential contribution to adverse pregnancy outcomes in pregnancy complications.
Collapse
Affiliation(s)
- Jessica L Bradshaw
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Spencer C Cushen
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States.,Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, United States
| | - Nicole R Phillips
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
30
|
Wei Q, Ren H, Zhang J, Yao W, Zhao B, Miao J. An Inhibitor of Grp94 Inhibits OxLDL-Induced Autophagy and Apoptosis in VECs and Stabilized Atherosclerotic Plaques. Front Cardiovasc Med 2021; 8:757591. [PMID: 34938782 PMCID: PMC8687133 DOI: 10.3389/fcvm.2021.757591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/15/2021] [Indexed: 01/18/2023] Open
Abstract
Background: Oxidized low-density lipoprotein (oxLDL) induces vascular endothelial cell (VEC) injury and atherosclerosis through activating endoplasmic reticulum stress. Expression of glucose-regulated protein 94 (Grp94) is induced by endoplasmic reticulum stress and Grp94 is involved in cardiovascular diseases. This study aimed to determine the role of Grp94 in oxLDL-induced vascular endothelial cell injury and atherosclerosis. Methods and Results: An inhibitor of Grp94, HCP1, was used to investigate the role of Grp94 in oxLDL-induced VEC injury in human umbilical vein endothelial cells and atherosclerosis in apolipoprotein E−/− mice. Results showed that HCP1 inhibited autophagy and apoptosis induced by oxLDL in VECs. And we found that Grp94 might interact with adenosine monophosphate-activated protein kinase (AMPK) and activate its activity. HCP1 inhibited AMPK activity and overexpression of Grp94 blocked the effect of HCP1. Besides, HCP1 activated the activity of mechanistic target of rapamycin complex 1 (mTORC1), co-treatment with AMPK activator acadesine eliminated the effect of HCP1 on mTORC1 activity as well as autophagy. In apolipoprotein E−/− mice, HCP1 suppressed autophagy and apoptosis of atherosclerotic plaque endothelium. In addition, HCP1 increased the content of collagen, smooth muscle cells, and anti-inflammatory macrophages while reducing the activity of MMP-2/9 and pro-inflammatory macrophages in the atherosclerotic lesion. Conclusion: HCP1 inhibited oxLDL-induced VEC injury and promoted the stabilization of atherosclerotic plaque in apoE−/− mice. Grp94 might be a potential therapeutic target in the clinical treatment of atherosclerosis.
Collapse
Affiliation(s)
- Qun Wei
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China.,NHC Key Laboratory of Otorhinolaryngology (Shandong University), Department of Otorhinolaryngology, Qilu Hospital, Shandong University, Jinan, China
| | - Hui Ren
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| | - Jun Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| | - Wen Yao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| | - Baoxiang Zhao
- School of Chemistry and Chemical Engineering, Institute of Organic Chemistry, Shandong University, Jinan, China
| | - Junying Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| |
Collapse
|
31
|
Ma C, Wu H, Yang G, Xiang J, Feng K, Zhang J, Hua Y, Kang L, Fan G, Yang S. Calycosin ameliorates atherosclerosis by enhancing autophagy via regulating the interaction between KLF2 and MLKL in apoE -/- mice. Br J Pharmacol 2021; 179:252-269. [PMID: 34713437 DOI: 10.1111/bph.15720] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/23/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis is one of the underlying causes of cardiovascular disease. Formation of foam cells and necrotic core in the plaque is a hallmark of atherosclerosis, which results from lipid deposition, apoptosis, and inflammation in macrophage. Macrophage autophagy is a critical anti-atherogenic process and defective autophagy aggravates atherosclerosis by enhancing foam cell formation, apoptosis, and inflammation. Hence, enhancing autophagy can be a strategy for atherosclerosis treatment. Calycosin, a flavonoid from Astragali Radix, displays antioxidant and anti-inflammatory activities, and therefore is potential to reduce the risk of cardiovascular disease. However, the antiatherogenic effect of calycosin and the involved mechanism remains unclear. In this study, we assessed the potential benefits of calycosin on autophagy and atherosclerosis, and revealed the underlying mechanism. EXPERIMENTAL APPROACH In this study, apoE-/- mice were fed high-fat diet for 16 weeks in presence of calycosin and/or autophagy inhibitor chloroquine, which was followed by determination of atherosclerosis development, autophagy activity, and the involved mechanisms. KEY RESULTS Calycosin protected against atherosclerosis and enhanced plaque stability via promoting autophagy. Calycosin inhibited foam cells formation, inflammation, and apoptosis by enhancing autophagy. MLKL was demonstrated as a new autophagy regulator, which can be negatively regulated by KLF2. Mechanistically, inhibitory effects of calycosin on atherogenesis were via improving autophagy through modulating KLF2-MLKL signaling pathway. CONCLUSIONS AND IMPLICATIONS This study demonstrated the atheroprotective effect of calycosin was through upregulating KLF2-MLKL-mediated autophagy, which not only proposed novel mechanistic insights into the atherogenesis but also identified calycosin as a potential drug candidate for atherosclerosis treatment.
Collapse
Affiliation(s)
- Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion
| | - Han Wu
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Guangyan Yang
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jiaqing Xiang
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Ke Feng
- College of Life Sciences, Nankai University, Tianjin, China
| | - Jing Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion
| | - Lin Kang
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China.,The Biobank of National Innovation Center for Advanced Medical Devices, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion
| | - Shu Yang
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| |
Collapse
|
32
|
Fang S, Sun S, Cai H, Zou X, Wang S, Hao X, Wan X, Tian J, Li Z, He Z, Huang W, Liang C, Zhang Z, Yang L, Tian J, Yu B, Sun B. IRGM/Irgm1 facilitates macrophage apoptosis through ROS generation and MAPK signal transduction: Irgm1 +/- mice display increases atherosclerotic plaque stability. Theranostics 2021; 11:9358-9375. [PMID: 34646375 PMCID: PMC8490524 DOI: 10.7150/thno.62797] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/02/2021] [Indexed: 02/04/2023] Open
Abstract
Rationale: Atherosclerosis plaque rupture (PR) is the pathological basis and chief culprit of most acute cardiovascular events and death. Given the complex and important role of macrophage apoptosis and autophagy in affecting plaque stability, an important unanswered question include is whether, and how, immunity-related GTPase family M protein (IRGM) and its mouse orthologue IRGM1 affect macrophage survival and atherosclerotic plaque stability. Methods: To investigate whether serum IRGM of ST-segment elevation myocardial infarction (STEMI) patients is related to plaque morphology, we divided 85 STEMI patients into those with and without plaque rupture (PR and non-PR, respectively) based on OCT image analysis, and quantified the patients' serum IRGM levels. Next, we engineered Irgm1 deficient mice (Irgm1+/-) and chimera mice with Irgm1 deficiency in the bone marrow on an ApoE-/- background, which were then fed a high-fat diet for 16 weeks. Pathological staining was used to detect necrotic plaque cores, ratios of neutral lipids and cholesterol crystal, as well as collagen fiber contents in these mice to characterize plaque stability. In addition, immunofluorescence, immunohistochemical staining and western blot were used to detect the apoptosis of macrophages in the plaques. In vitro, THP-1 and RAW264.7 cells were stimulated with ox-LDL to mimic the in vivo environment, and IRGM/IRGM1 expression were modified by specific siRNA (knockdown) or IRGM plasmid (knocked-in). The effect of IRGM/Irgm1 on autophagy and apoptosis of macrophages induced by ox-LDL was then evaluated. In addition, we introduced inhibitors of the JNK/p38/ERK signaling pathway to verify the specific mechanism by which Irgm1 regulates RAW264.7 cell apoptosis. Results: The serum IRGM levels of PR patients is significantly higher than that of non-PR patients and healthy volunteers, which may be an effective predictor of PR. On a high-fat diet, Irgm1-deficient mice exhibit reduced necrotic plaque cores, as well as neutral lipid and cholesterol crystal ratios, with increased collagen fiber content. Additionally, macrophage apoptosis is inhibited in the plaques of Irgm1-deficient mice. In vitro, IRGM/Irgm1 deficiency rapidly inhibits ox-LDL-induced macrophage autophagy while inhibiting ox-LDL-induced macrophage apoptosis in late stages. Additionally, IRGM/Irgm1 deficiency suppresses reactive oxygen species (ROS) production in macrophages, while removal of ROS effectively inhibits macrophage apoptosis induced by IRGM overexpression. We further show that Irgm1 can affect macrophage apoptosis by regulating JNK/p38/ERK phosphorylation in the MAPK signaling pathway. Conclusions: Serum IRGM may be related to the process of PR in STEMI patients, and IRGM/Irgm1 deficiency increases plaque stability. In addition, IRGM/Irgm1 deficiency suppresses macrophage apoptosis by inhibiting ROS generation and MAPK signaling transduction. Cumulatively, these results suggest that targeting IRGM may represent a new treatment strategy for the prevention and treatment of acute cardiovascular deaths caused by PR.
Collapse
|
33
|
Kim SK, Park KY, Choe JY. Toll-Like Receptor 9 Is Involved in NLRP3 Inflammasome Activation and IL-1β Production Through Monosodium Urate-Induced Mitochondrial DNA. Inflammation 2021; 43:2301-2311. [PMID: 32700178 DOI: 10.1007/s10753-020-01299-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The NLR family pyrin domain-containing 3 (NLRP3) inflammasome is a cytoplasmic multimolecular complex that generates interleukin (IL)-1β and is considered a main pathogenic mechanism for uric acid-induced inflammation. Whether toll-like receptor 9 (TLR9) is responsible for uric acid-induced NLRP3 inflammasome activation remains unclear. Thus, the aim of this study was to identify the role of TLR9 in NLRP3 inflammasome activation through monosodium urate (MSU) crystal-induced mitochondrial DNA. RAW 264.7 cells treated with MSU crystals, CpG oligonucleotides (ODNs), or a combination of both were used to assess nuclear factor (NF)-κB signaling, NLRP3 inflammasome components such as NLRP3, ASC, and caspase-1, and IL-1β. Real-time polymerase chain reaction (RT-PCR), Western blotting, DNA fragmentation assay, mitochondrial DNA copy number assay, and immunofluorescence were used in the in vitro study. RAW 264.7 cells treated with CpG-ODN stimulated the activation of NF-κB signaling, the NLRP3 inflammasome components NLRP3, ASC, and caspase-1, and IL-1β gene and protein expression. DNA fragmentation assay showed that MSU crystals induced cellular apoptosis. Fragmented DNA prompted by MSU crystals induced TLR9 expression. RAW 264.7 cells treated with CpG-ODN or MSU crystals and both increased expression of mitochondrial DNA relative to nuclear DNA. CpG-ODN and MSU crystals augmented the activation of NLRP3 inflammasome components and IL-1β expression, which was significantly suppressed in RAW 264.7 cells transfected with TLR9 siRNA. This study suggests that TLR9 activated by MSU crystal-mediated mitochondrial DNA contributes to the activation of NLRP3 inflammasomes and IL-1β production.
Collapse
Affiliation(s)
- Seong-Kyu Kim
- Department of Internal Medicine, Division of Rheumatology, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea. .,Arthritis and Autoimmunity Research Center, Catholic University of Daegu, Daegu, Republic of Korea.
| | - Ki-Yeun Park
- Arthritis and Autoimmunity Research Center, Catholic University of Daegu, Daegu, Republic of Korea
| | - Jung-Yoon Choe
- Department of Internal Medicine, Division of Rheumatology, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea.,Arthritis and Autoimmunity Research Center, Catholic University of Daegu, Daegu, Republic of Korea
| |
Collapse
|
34
|
Hao C, Zhang J, Zhang F, Wu J, Cao H, Wang W. Mitochondrial DNA may act as a biomarker to predict donor-kidney quality. Clin Transplant 2021; 35:e14469. [PMID: 34448256 DOI: 10.1111/ctr.14469] [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: 02/02/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 11/29/2022]
Abstract
Kidney transplantation is the best therapy for end-stage renal disease. Demand for kidney transplantation rises year-on-year, and the gap between kidney supply and demand remains large. To meet this clinical need, a gradual expansion in the supply of donors is required. However, clinics lack appropriate tools capable of quickly and accurately predicting post-transplant renal allograft function, and thus assess donor-kidney quality before transplantation. Mitochondrial DNA (mtDNA) is a key component of damage-associated molecular patterns (DAMPs) and plays an important part in ischemia-reperfusion injury (IRI), accelerating the progression of IRI by inducing inflammation and type I interferon responses. mtDNA is known to be closely involved in delayed graft function (DGF) and acute kidney injury (AKI) after transplantation. Thus, mtDNA is a potential biomarker able to predict post-transplant renal allograft function. This review summarizes mtDNA biology, the role mtDNA plays in renal transplantation, outlines advances in detecting mtDNA, and details mtDNA's able to predict post-transplant renal allograft function. We aim to elucidate the potential value of mtDNA as a biomarker in the prediction of IRI, and eventually provide help for predicting donor-kidney quality.
Collapse
Affiliation(s)
- Changzhen Hao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Feilong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Huawei Cao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
Vellasamy S, Murugan D, Abas R, Alias A, Seng WY, Woon CK. Biological Activities of Paeonol in Cardiovascular Diseases: A Review. Molecules 2021; 26:4976. [PMID: 34443563 PMCID: PMC8400614 DOI: 10.3390/molecules26164976] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 11/16/2022] Open
Abstract
Paeonol is a naturally existing bioactive compound found in the root bark of Paeonia suffruticosa and it is traditionally used in Chinese medicine for the prevention and management of cardiovascular diseases. To date, a great deal of studies has been reported on the pharmacological effects of paeonol and its mechanisms of action in various diseases and conditions. In this review, the underlying mechanism of action of paeonol in cardiovascular disease has been elucidated. Recent studies have revealed that paeonol treatment improved endothelium injury, demoted inflammation, ameliorated oxidative stress, suppressed vascular smooth muscle cell proliferation, and repressed platelet activation. Paeonol has been reported to effectively protect the cardiovascular system either employed alone or in combination with other traditional medicines, thus, signifying it could be a hypothetically alternative or complementary atherosclerosis treatment. This review summarizes the biological and pharmacological activities of paeonol in the treatment of cardiovascular diseases and its associated underlying mechanisms for a better insight for future clinical practices.
Collapse
Affiliation(s)
- Shalini Vellasamy
- Department of Microbiology and Parasitology, School of Medicine, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarum 42610, Selangor, Malaysia;
| | - Dharmani Murugan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Razif Abas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri Kembangan 43400, Selangor, Malaysia;
| | - Aspalilah Alias
- Department of Basic Sciences and Oral Biology, Faculty of Dentistry, Universiti Sains Islam Malaysia, Kuala Lumpur 55100, Malaysia;
- Fakultas Kedokteran Gigi, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Wu Yuan Seng
- Centre for Virus and Vaccine Research, Sunway University, Bandar Sunway 47500, Selangor, Malaysia;
- Department of Biological Sciences, Sunway University, Bandar Sunway 47500, Selangor, Malaysia
| | - Choy Ker Woon
- Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh 47000, Selangor, Malaysia
| |
Collapse
|
36
|
Pham PT, Fukuda D, Nishimoto S, Kim-Kaneyama JR, Lei XF, Takahashi Y, Sato T, Tanaka K, Suto K, Kawabata Y, Yamaguchi K, Yagi S, Kusunose K, Yamada H, Soeki T, Wakatsuki T, Shimada K, Kanematsu Y, Takagi Y, Shimabukuro M, Setou M, Barber GN, Sata M. STING, a cytosolic DNA sensor, plays a critical role in atherogenesis: a link between innate immunity and chronic inflammation caused by lifestyle-related diseases. Eur Heart J 2021; 42:4336-4348. [PMID: 34226923 DOI: 10.1093/eurheartj/ehab249] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/02/2021] [Accepted: 04/15/2021] [Indexed: 12/23/2022] Open
Abstract
AIMS Lifestyle-related diseases promote atherosclerosis, a chronic inflammatory disease; however, the molecular mechanism remains largely unknown. Endogenous DNA fragments released under over-nutrient condition provoke sterile inflammation through the recognition by DNA sensors. Here, we investigated the role of stimulator of interferon genes (STING), a cytosolic DNA sensor, in atherogenesis. METHODS AND RESULTS Apolipoprotein E-deficient (Apoe-/-) mice fed a western-type diet (WTD), a hypercholesterolaemic mouse model, showed higher STING expression and markers for DNA damage such as γH2AX, p53, and single-stranded DNA (ssDNA) accumulation in macrophages in the aorta compared with wild-type (WT) mice. The level of cGAMP, a STING agonist, in the aorta was higher in Apoe-/- mice. Genetic deletion of Sting in Apoe-/- mice reduced atherosclerotic lesions in the aortic arch, lipid, and macrophage accumulation in plaques, and inflammatory molecule expression in the aorta compared with the control. Pharmacological blockade of STING using a specific inhibitor, C-176, ameliorated atherogenesis in Apoe-/- mice. In contrast, bone marrow-specific STING expression in Apoe-/- mice stimulated atherogenesis. Expression or deletion of STING did not affect metabolic parameters and blood pressure. In vitro studies revealed that STING activation by cGAMP or mitochondrial DNA accelerated inflammatory molecule expression (e.g. TNF-α or IFN-β) in mouse and human macrophages. Activation of nuclear factor-κB and TANK binding kinase 1 was involved in STING-associated vascular inflammation and macrophage activation. Furthermore, human atherosclerotic lesions in the carotid arteries expressed STING and cGAMP. CONCLUSION Stimulator of interferon genes stimulates pro-inflammatory activation of macrophages, leading to the development of atherosclerosis. Stimulator of interferon genes signalling may serve as a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Phuong Tran Pham
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan.,Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Sachiko Nishimoto
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan.,Faculty of Clinical Nutrition and Dietetics, Konan Women's University, 6-2-23, Morikita-machi, Higashinada-ku, Kobe 658-0001, Japan
| | - Joo-Ri Kim-Kaneyama
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Xiao-Feng Lei
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan.,Preppers, Co., Ltd, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Tokyo 140-001, Japan
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kimie Tanaka
- Division for Health Service Promotion, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kumiko Suto
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Yutaka Kawabata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Shusuke Yagi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Kenya Kusunose
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Hirotsugu Yamada
- Department of Community Medicine for Cardiology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima, 770-8503, Japan
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Tetsuzo Wakatsuki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Kenji Shimada
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Yasuhisa Kanematsu
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Yasushi Takagi
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Michio Shimabukuro
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan.,Department of Diabetes, Endocrinology and Metabolism, School of Medicine, 1 Hikariga-oka, Fukushima 960-1295, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Glen N Barber
- Department of Cell Biology, University of Miami Miller School of Medicine, 1550 NW 10th Avenue, PAP 5th floor Miami, Florida 33136, USA
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| |
Collapse
|
37
|
Oxidative Stress and Mitochondrial Damage in Dry Age-Related Macular Degeneration Like NFE2L2/PGC-1α -/- Mouse Model Evoke Complement Component C5a Independent of C3. BIOLOGY 2021; 10:biology10070622. [PMID: 34356477 PMCID: PMC8301195 DOI: 10.3390/biology10070622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022]
Abstract
Aging-associated chronic oxidative stress and inflammation are known to be involved in various diseases, e.g., age-related macular degeneration (AMD). Previously, we reported the presence of dry AMD-like signs, such as elevated oxidative stress, dysfunctional mitophagy and the accumulation of detrimental oxidized materials in the retinal pigment epithelial (RPE) cells of nuclear factor erythroid 2-related factor 2, and a peroxisome proliferator-activated receptor gamma coactivator 1-alpha (NFE2L2/PGC1α) double knockout (dKO) mouse model. Here, we investigated the dynamics of inflammatory markers in one-year-old NFE2L2/PGC1α dKO mice. Immunohistochemical analysis revealed an increase in levels of Toll-like receptors 3 and 9, while those of NOD-like receptor 3 were decreased in NFE2L2/PGC1α dKO retinal specimens as compared to wild type animals. Further analysis showed a trend towards an increase in complement component C5a independent of component C3, observed to be tightly regulated by complement factor H. Interestingly, we found that thrombin, a serine protease enzyme, was involved in enhancing the terminal pathway producing C5a, independent of C3. We also detected an increase in primary acute phase C-reactive protein and receptor for advanced glycation end products in NFE2L2/PGC1α dKO retina. Our main data show C5 and thrombin upregulation together with decreased C3 levels in this dry AMD-like model. In general, the retina strives to mount an orchestrated inflammatory response while attempting to maintain tissue homeostasis and resolve inflammation.
Collapse
|
38
|
Chen Z, Chao Y, Li W, Wallace GG, Bussell T, Ding J, Wang C. Abuse-Tolerant Electrolytes for Lithium-Ion Batteries. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2003694. [PMID: 34105300 PMCID: PMC8188208 DOI: 10.1002/advs.202003694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/31/2021] [Indexed: 05/22/2023]
Abstract
Safety issues currently limit the development of advanced lithium-ion batteries (LIBs) and this is exacerbated when they are misused or abused. The addition of small amounts of fillers or additives into common liquid electrolytes can greatly improve resistance to abuse without impairing electrochemical performance. This review discusses the recent progress in such abuse-tolerant electrolytes. It covers electrolytes with shear thickening properties for tolerating mechanical abuse, electrolytes with redox shuttle additives for suppressing electrochemical abuse, and electrolytes with flame-retardant additives for resisting thermal abuse. It aims to provide insights into the functioning of such electrolytes and the understanding of electrolyte composition-property relationship. Future perspectives, challenges, and opportunities towards practical applications are also presented.
Collapse
Affiliation(s)
- Zhiqi Chen
- ARC Centre of Excellence for Electromaterials ScienceIntelligent Polymer Research InstituteAIIM FacilityInnovation CampusUniversity of WollongongWollongongNSW2500Australia
| | - Yunfeng Chao
- ARC Centre of Excellence for Electromaterials ScienceIntelligent Polymer Research InstituteAIIM FacilityInnovation CampusUniversity of WollongongWollongongNSW2500Australia
| | - Weihua Li
- School of Mechanical, Materials, Mechatronic and Biomedical EngineeringUniversity of WollongongWollongongNSW2522Australia
| | - Gordon G. Wallace
- ARC Centre of Excellence for Electromaterials ScienceIntelligent Polymer Research InstituteAIIM FacilityInnovation CampusUniversity of WollongongWollongongNSW2500Australia
| | - Tim Bussell
- Defence Science and Technology GroupDepartment of DefenceMelbourneVIC3207Australia
| | - Jie Ding
- Defence Science and Technology GroupDepartment of DefenceMelbourneVIC3207Australia
| | - Caiyun Wang
- ARC Centre of Excellence for Electromaterials ScienceIntelligent Polymer Research InstituteAIIM FacilityInnovation CampusUniversity of WollongongWollongongNSW2500Australia
| |
Collapse
|
39
|
Kyaw T, Loveland P, Kanellakis P, Cao A, Kallies A, Huang AL, Peter K, Toh BH, Bobik A. Alarmin-activated B cells accelerate murine atherosclerosis after myocardial infarction via plasma cell-immunoglobulin-dependent mechanisms. Eur Heart J 2021; 42:938-947. [PMID: 33338208 DOI: 10.1093/eurheartj/ehaa995] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/30/2020] [Accepted: 11/29/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Myocardial infarction (MI) accelerates atherosclerosis and greatly increases the risk of recurrent cardiovascular events for many years, in particular, strokes and MIs. Because B cell-derived autoantibodies produced in response to MI also persist for years, we investigated the role of B cells in adaptive immune responses to MI. METHODS AND RESULTS We used an apolipoprotein-E-deficient (ApoE-/-) mouse model of MI-accelerated atherosclerosis to assess the importance of B cells. One week after inducing MI in atherosclerotic mice, we depleted B cells using an anti-CD20 antibody. This treatment prevented subsequent immunoglobulin G accumulation in plaques and MI-induced accelerated atherosclerosis. In gain of function experiments, we purified spleen B cells from mice 1 week after inducing MI and transferred these cells into atherosclerotic ApoE-/- mice, which greatly increased immunoglobulin G (IgG) accumulation in plaque and accelerated atherosclerosis. These B cells expressed many cytokines that promote humoural immunity and in addition, they formed germinal centres within the spleen where they differentiated into antibody-producing plasma cells. Specifically deleting Blimp-1 in B cells, the transcriptional regulator that drives their terminal differentiation into antibody-producing plasma cells prevented MI-accelerated atherosclerosis. Alarmins released from infarcted hearts were responsible for activating B cells via toll-like receptors and deleting MyD88, the canonical adaptor protein for inflammatory signalling downstream of toll-like receptors, prevented B-cell activation and MI-accelerated atherosclerosis. CONCLUSION Our data implicate early B-cell activation and autoantibodies as a central cause for accelerated atherosclerosis post-MI and identifies novel therapeutic strategies towards preventing recurrent cardiovascular events such as MI and stroke.
Collapse
Affiliation(s)
- Tin Kyaw
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Paula Loveland
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Peter Kanellakis
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Anh Cao
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, University of Melbourne, 792 Elizabeth Street, Melbourne, Vic 3000, Australia
| | - Alex L Huang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Cardiology, Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Cardiology, Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia.,Department of Immunology, Central Clinical School, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia.,Department of Immunology, Central Clinical School, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| |
Collapse
|
40
|
Matsui H, Ito J, Matsui N, Uechi T, Onodera O, Kakita A. Cytosolic dsDNA of mitochondrial origin induces cytotoxicity and neurodegeneration in cellular and zebrafish models of Parkinson's disease. Nat Commun 2021; 12:3101. [PMID: 34035300 PMCID: PMC8149644 DOI: 10.1038/s41467-021-23452-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/28/2021] [Indexed: 01/18/2023] Open
Abstract
Mitochondrial dysfunction and lysosomal dysfunction have been implicated in Parkinson's disease (PD), but the links between these dysfunctions in PD pathogenesis are still largely unknown. Here we report that cytosolic dsDNA of mitochondrial origin escaping from lysosomal degradation was shown to induce cytotoxicity in cultured cells and PD phenotypes in vivo. The depletion of PINK1, GBA and/or ATP13A2 causes increases in cytosolic dsDNA of mitochondrial origin and induces type I interferon (IFN) responses and cell death in cultured cell lines. These phenotypes are rescued by the overexpression of DNase II, a lysosomal DNase that degrades discarded mitochondrial DNA, or the depletion of IFI16, which acts as a sensor for cytosolic dsDNA of mitochondrial origin. Reducing the abundance of cytosolic dsDNA by overexpressing human DNase II ameliorates movement disorders and dopaminergic cell loss in gba mutant PD model zebrafish. Furthermore, IFI16 and cytosolic dsDNA puncta of mitochondrial origin accumulate in the brain of patients with PD. These results support a common causative role for the cytosolic leakage of mitochondrial DNA in PD pathogenesis.
Collapse
Affiliation(s)
- Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan. .,Department of Neuroscience of Disease, Center for Transdisciplinary Research, Niigata University, Niigata, Japan.
| | - Junko Ito
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Noriko Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tamayo Uechi
- Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
41
|
Role of macrophage autophagy in atherosclerosis: modulation by bioactive compounds. Biochem J 2021; 478:1359-1375. [PMID: 33861844 DOI: 10.1042/bcj20200894] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/28/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease associated with lipid metabolism disorder. Autophagy is a catabolic process and contributes to maintaining cellular homeostasis. Substantial evidence suggests that defective autophagy is implicated in several diseases, including atherosclerosis, while increased autophagy mitigates atherosclerosis development. Thus, understanding the mechanisms of autophagy regulation and its association with atherosclerosis is vital to develop new therapies against atherosclerosis. Dietary bioactive compounds are non-nutrient natural compounds that include phenolics, flavonoids, and carotenoids. Importantly, these bioactive compounds possess anti-inflammatory, antioxidant, and antibacterial properties that may alleviate various chronic diseases. Recently, examining the effects of bioactive compounds on autophagy activity in atherogenesis has drawn considerable attention. The current review discusses the role of macrophage autophagy in the development and progression of atherosclerosis. We also summarize our current knowledge of the therapeutic potential of bioactive compounds on atherosclerosis and autophagy.
Collapse
|
42
|
Tao H, Yancey PG, Blakemore JL, Zhang Y, Ding L, Jerome WG, Brown JD, Vickers KC, Linton MF. Macrophage SR-BI modulates autophagy via VPS34 complex and PPARα transcription of Tfeb in atherosclerosis. J Clin Invest 2021; 131:94229. [PMID: 33661763 PMCID: PMC8011903 DOI: 10.1172/jci94229] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Autophagy modulates lipid turnover, cell survival, inflammation, and atherogenesis. Scavenger receptor class B type I (SR-BI) plays a crucial role in lysosome function. Here, we demonstrate that SR-BI regulates autophagy in atherosclerosis. SR-BI deletion attenuated lipid-induced expression of autophagy mediators in macrophages and atherosclerotic aortas. Consequently, SR-BI deletion resulted in 1.8- and 2.5-fold increases in foam cell formation and apoptosis, respectively, and increased oxidized LDL-induced inflammatory cytokine expression. Pharmacological activation of autophagy failed to reduce lipid content or apoptosis in Sr-b1-/- macrophages. SR-BI deletion reduced both basal and inducible levels of transcription factor EB (TFEB), a master regulator of autophagy, causing decreased expression of autophagy genes encoding VPS34 and Beclin-1. Notably, SR-BI regulated Tfeb expression by enhancing PPARα activation. Moreover, intracellular macrophage SR-BI localized to autophagosomes, where it formed cholesterol domains resulting in enhanced association of Barkor and recruitment of the VPS34-Beclin-1 complex. Thus, SR-BI deficiency led to lower VPS34 activity in macrophages and in atherosclerotic aortic tissues. Overexpression of Tfeb or Vps34 rescued the defective autophagy in Sr-b1-/- macrophages. Taken together, our results show that macrophage SR-BI regulates autophagy via Tfeb expression and recruitment of the VPS34-Beclin-1 complex, thus identifying previously unrecognized roles for SR-BI and potentially novel targets for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Huan Tao
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - Patricia G. Yancey
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - John L. Blakemore
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - Youmin Zhang
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - Lei Ding
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - W. Gray Jerome
- Department of Pathology, Microbiology and Immunology, and
| | - Jonathan D. Brown
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - Kasey C. Vickers
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - MacRae F. Linton
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
43
|
Carresi C, Mollace R, Macrì R, Scicchitano M, Bosco F, Scarano F, Coppoletta AR, Guarnieri L, Ruga S, Zito MC, Nucera S, Gliozzi M, Musolino V, Maiuolo J, Palma E, Mollace V. Oxidative Stress Triggers Defective Autophagy in Endothelial Cells: Role in Atherothrombosis Development. Antioxidants (Basel) 2021; 10:antiox10030387. [PMID: 33807637 PMCID: PMC8001288 DOI: 10.3390/antiox10030387] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/18/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Atherothrombosis, a multifactorial and multistep artery disorder, represents one of the main causes of morbidity and mortality worldwide. The development and progression of atherothrombosis is closely associated with age, gender and a complex relationship between unhealthy lifestyle habits and several genetic risk factors. The imbalance between oxidative stress and antioxidant defenses is the main biological event leading to the development of a pro-oxidant phenotype, triggering cellular and molecular mechanisms associated with the atherothrombotic process. The pathogenesis of atherosclerosis and its late thrombotic complications involve multiple cellular events such as inflammation, endothelial dysfunction, proliferation of vascular smooth muscle cells (SMCs), extracellular matrix (ECM) alterations, and platelet activation, contributing to chronic pathological remodeling of the vascular wall, atheromatous plague formation, vascular stenosis, and eventually, thrombus growth and propagation. Emerging studies suggest that clotting activation and endothelial cell (EC) dysfunction play key roles in the pathogenesis of atherothrombosis. Furthermore, a growing body of evidence indicates that defective autophagy is closely linked to the overproduction of reactive oxygen species (ROS) which, in turn, are involved in the development and progression of atherosclerotic disease. This topic represents a large field of study aimed at identifying new potential therapeutic targets. In this review, we focus on the major role played by the autophagic pathway induced by oxidative stress in the modulation of EC dysfunction as a background to understand its potential role in the development of atherothrombosis.
Collapse
Affiliation(s)
- Cristina Carresi
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Correspondence: ; Tel.: +39-09613694128; Fax: +39-09613695737
| | - Rocco Mollace
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Roberta Macrì
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Miriam Scicchitano
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Francesca Bosco
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Federica Scarano
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Anna Rita Coppoletta
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Lorenza Guarnieri
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Stefano Ruga
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Maria Caterina Zito
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Saverio Nucera
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Micaela Gliozzi
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Vincenzo Musolino
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Jessica Maiuolo
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Ernesto Palma
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| | - Vincenzo Mollace
- Research for Food Safety & Health IRC-FSH, Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (R.M.); (M.S.); (F.B.); (F.S.); (A.R.C.); (L.G.); (S.R.); (M.C.Z.); (S.N.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| |
Collapse
|
44
|
Picca A, Calvani R, Coelho-Junior HJ, Marzetti E. Cell Death and Inflammation: The Role of Mitochondria in Health and Disease. Cells 2021; 10:cells10030537. [PMID: 33802550 PMCID: PMC7998762 DOI: 10.3390/cells10030537] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/14/2022] Open
Abstract
Mitochondria serve as a hub for a multitude of vital cellular processes. To ensure an efficient deployment of mitochondrial tasks, organelle homeostasis needs to be preserved. Mitochondrial quality control (MQC) mechanisms (i.e., mitochondrial dynamics, biogenesis, proteostasis, and autophagy) are in place to safeguard organelle integrity and functionality. Defective MQC has been reported in several conditions characterized by chronic low-grade inflammation. In this context, the displacement of mitochondrial components, including mitochondrial DNA (mtDNA), into the extracellular compartment is a possible factor eliciting an innate immune response. The presence of bacterial-like CpG islands in mtDNA makes this molecule recognized as a damaged-associated molecular pattern by the innate immune system. Following cell death-triggering stressors, mtDNA can be released from the cell and ignite inflammation via several pathways. Crosstalk between autophagy and apoptosis has emerged as a pivotal factor for the regulation of mtDNA release, cell’s fate, and inflammation. The repression of mtDNA-mediated interferon production, a powerful driver of immunological cell death, is also regulated by autophagy–apoptosis crosstalk. Interferon production during mtDNA-mediated inflammation may be exploited for the elimination of dying cells and their conversion into elements driving anti-tumor immunity.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (E.M.)
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, 17165 Stockholm, Sweden
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (E.M.)
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, 17165 Stockholm, Sweden
- Correspondence: ; Tel.: +39-(06)-3015-5559; Fax: +39-(06)-3051-911
| | - Hélio José Coelho-Junior
- Università Cattolica del Sacro Cuore, Institute of Internal Medicine and Geriatrics, 00168 Rome, Italy;
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (E.M.)
- Università Cattolica del Sacro Cuore, Institute of Internal Medicine and Geriatrics, 00168 Rome, Italy;
| |
Collapse
|
45
|
Guo Y, Yang JH, Cao SD, Gao CX, He Y, Wang Y, Wan HT, Jin B. Effect of main ingredients of Danhong Injection against oxidative stress induced autophagy injury via miR-19a/SIRT1 pathway in endothelial cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 83:153480. [PMID: 33548866 DOI: 10.1016/j.phymed.2021.153480] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 12/25/2020] [Accepted: 01/21/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Autophagy plays an important role in cellular homeostasis. Oxidative stress stimulated endothelial excessive autophagy has been proposed as a major risk factor for cardiovascular diseases (CVD). Danhong injection (DHI), the most prescribed traditional Chinese medicine for the treatment of CVD, has been shown to elicit vascular protective effects. However, its underlying mechanisms remain poorly defined. This study aimed to uncover the protective effects of DHI and its main bioactive components on autophagy injury of human umbilical vein endothelial cells (HUVECs) induced by H2O2 and reveal the possible mechanisms. METHODS HUVECs were treated with different concentrations of DHI or its components, after exposed to H2O2. The protective effects of DHI and its components in H2O2-induced HUVECs were examined via a cytotoxicity assay and western blot. Apoptosis was evaluated with flow cytometry. Autophagy flux was assessed by transmission electron microscopy and LC3 plasmid transfection. Besides, the role miR-19a and SIRT1 in DHI and components-mediated anti-autophagy responses were validated with inhibitors transfection. RESULTS Our results showed that DHI and its components do have different effects on different aspects. In terms of HUVECs survival rate, Salvianolic acid B (Sal B) and danshensu (DSS) performed better than DHI, Hydroxysafflor yellow A (HSYA) and Tanshinone IIA (DST-IIA). As for the proliferation effect on HUVECs, only Sal B has the most obvious performance as same as 3MA. Besides, DHI and its components are sensitive and superior in regulating and balancing ROS concentration. Among the GSH/GSSG indicators, DSS and HSYA performed better. In terms of SOD content and apoptotic rate, the SOD level showed the opposite trend compared with H2O2 group. For the expression of LC3, Beclin-1 and P62, DHI and its components all had significant effects. When miR-19a or SIRT1 was inhibited, Sal B (0.5 μg/ml) can not decrease autophagy-related protein effectively. CONCLUSION DHI and its components all had anti-autophagy effects. And Sal B (0.5 μg/ml) inhibited HUVECs autophagy via miR-19a/SIRT1 pathway.
Collapse
Affiliation(s)
- Yan Guo
- College of Basic Medicine &Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Jie-Hong Yang
- College of Basic Medicine &Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Shi-Dong Cao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Cheng-Xian Gao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yu He
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yu Wang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Hai-Tong Wan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Bo Jin
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
46
|
Bérat CM, Montealegre S, Wiedemann A, Nuzum MLC, Blondel A, Debruge H, Cano A, Chabrol B, Hoebeke C, Polak M, Stoupa A, Feillet F, Torre S, Boddaert N, Bruel H, Barth M, Damaj L, Abi-Wardé MT, Afenjar A, Benoist JF, Madrange M, Caccavelli L, Renard P, Hubas A, Nusbaum P, Pontoizeau C, Gobin S, van Endert P, Ottolenghi C, Maltret A, de Lonlay P. Clinical and biological characterization of 20 patients with TANGO2 deficiency indicates novel triggers of metabolic crises and no primary energetic defect. J Inherit Metab Dis 2021; 44:415-425. [PMID: 32929747 DOI: 10.1002/jimd.12314] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/07/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
Abstract
TANGO2 disease is a severe inherited disorder associating multiple symptoms such as metabolic crises, encephalopathy, cardiac arrhythmias, and hypothyroidism. The mechanism of action of TANGO2 is currently unknown. Here, we describe a cohort of 20 French patients bearing mutations in the TANGO2 gene. We found that the main clinical presentation was the association of neurodevelopmental delay (n = 17), acute metabolic crises (n = 17) and hypothyroidism (n = 12), with a large intrafamilial clinical variability. Metabolic crises included rhabdomyolysis (15/17), neurological symptoms (14/17), and cardiac features (12/17; long QT (n = 10), Brugada pattern (n = 2), cardiac arrhythmia (n = 6)) that required intensive care. We show previously uncharacterized triggers of metabolic crises in TANGO2 patients, such as some anesthetics and possibly l-carnitine. Unexpectedly, plasma acylcarnitines, plasma FGF-21, muscle histology, and mitochondrial spectrometry were mostly normal. Moreover, in patients' primary myoblasts, palmitate and glutamine oxidation rates, and the mitochondrial network were also normal. Finally, we found variable mitochondrial respiration and defective clearance of oxidized DNA upon cycles of starvation and refeeding. We conclude that TANGO2 disease is a life-threatening disease that needs specific cardiac management and anesthesia protocol. Mechanistically, TANGO2 disease is unlikely to originate from a primary mitochondrial defect. Rather, we suggest that mitochondrial defects are secondary to strong extrinsic triggers in TANGO2 deficient patients.
Collapse
Affiliation(s)
- Claire-Marine Bérat
- Inserm U1151, Institut Necker Enfants-Malades, Paris, France
- Université de Paris, Paris, France
- Reference Center of inherited Metabolic Diseases, Necker-Enfants-Malades University hospital, APHP, Imagine Institute, Filière G2M, Paris, France
| | - Sebastian Montealegre
- Inserm U1151, Institut Necker Enfants-Malades, Paris, France
- Reference Center of inherited Metabolic Diseases, Necker-Enfants-Malades University hospital, APHP, Imagine Institute, Filière G2M, Paris, France
| | - Arnaud Wiedemann
- Department of Pediatric Intensive Care, Reference Center of Inherited Metabolic Disorders, INSERM U1256, Nancy Hospital, Nancy, France
| | | | - Amélie Blondel
- Department of Biochemistry, Necker-Enfants-Malades University Hospital, APHP, Filière G2M, Paris, France
| | - Hugo Debruge
- Inserm U1151, Institut Necker Enfants-Malades, Paris, France
| | - Aline Cano
- Reference Center of Inherited Metabolic Disorders, La Timone Hospital, Filière G2M, Marseille, France
| | - Brigitte Chabrol
- Reference Center of Inherited Metabolic Disorders, La Timone Hospital, Filière G2M, Marseille, France
| | - Célia Hoebeke
- Reference Center of Inherited Metabolic Disorders, La Timone Hospital, Filière G2M, Marseille, France
| | - Michel Polak
- Université de Paris, Paris, France
- Endocrinology Unit, Reference Center of Rare Endocrine Diseases of Growth and Development, Necker-Enfants-Malades, University hospital, APHP, Imagine Institute, Paris, France
| | - Athanasia Stoupa
- Université de Paris, Paris, France
- Endocrinology Unit, Reference Center of Rare Endocrine Diseases of Growth and Development, Necker-Enfants-Malades, University hospital, APHP, Imagine Institute, Paris, France
| | - François Feillet
- Department of Pediatric Intensive Care, Reference Center of Inherited Metabolic Disorders, INSERM U1256, Nancy Hospital, Nancy, France
| | - Stéphanie Torre
- Competence Center of Inherited Metabolic Disorders, Rouen Hospital, Filière G2M, Rouen, France
| | - Nathalie Boddaert
- Université de Paris, Paris, France
- Paediatric Radiology Department, Necker-Enfants-Malades University hospital, APHP and INSERM U1163, Imagine Institute, Paris, France
| | - Henri Bruel
- Pediatrics Department, Le Havre Hospital, Le Havre, France
| | - Magalie Barth
- Pediatrics Department, Angers Hospital, Angers, France
| | - Lena Damaj
- Pediatrics Department, Rennes Hospital, Rennes, France
| | | | - Alexandra Afenjar
- Reference Center of Cerebellar Malformations and Congenital Diseases, Trousseau Hospital, APHP, Paris, France
| | - Jean-François Benoist
- Department of Biochemistry, Necker-Enfants-Malades University Hospital, APHP, Filière G2M, Paris, France
| | - Marine Madrange
- Inserm U1151, Institut Necker Enfants-Malades, Paris, France
- Reference Center of inherited Metabolic Diseases, Necker-Enfants-Malades University hospital, APHP, Imagine Institute, Filière G2M, Paris, France
| | - Laure Caccavelli
- Inserm U1151, Institut Necker Enfants-Malades, Paris, France
- Reference Center of inherited Metabolic Diseases, Necker-Enfants-Malades University hospital, APHP, Imagine Institute, Filière G2M, Paris, France
| | - Perrine Renard
- Inserm U1151, Institut Necker Enfants-Malades, Paris, France
| | - Arnaud Hubas
- Genetics and Molecular Biology, Laboratoire de culture cellulaire, Hôpital Cochin, Paris, France
| | - Patrick Nusbaum
- Genetics and Molecular Biology, Laboratoire de culture cellulaire, Hôpital Cochin, Paris, France
| | - Clément Pontoizeau
- Department of Biochemistry, Necker-Enfants-Malades University Hospital, APHP, Filière G2M, Paris, France
| | - Stéphanie Gobin
- Genetics Department, Necker-Enfants-Malades University Hospital, APHP, Paris, France
| | - Peter van Endert
- Inserm U1151, Institut Necker Enfants-Malades, Paris, France
- Université de Paris, Paris, France
| | - Chris Ottolenghi
- Department of Biochemistry, Necker-Enfants-Malades University Hospital, APHP, Filière G2M, Paris, France
| | - Alice Maltret
- Cardiology Unit, Necker-Enfants-Malades University Hospital, APHP, Paris, France
| | - Pascale de Lonlay
- Inserm U1151, Institut Necker Enfants-Malades, Paris, France
- Université de Paris, Paris, France
- Reference Center of inherited Metabolic Diseases, Necker-Enfants-Malades University hospital, APHP, Imagine Institute, Filière G2M, Paris, France
| |
Collapse
|
47
|
Meng Q, Li Y, Ji T, Chao Y, Li J, Fu Y, Wang S, Chen Q, Chen W, Huang F, Wang Y, Zhang Q, Wang X, Bian H. Estrogen prevent atherosclerosis by attenuating endothelial cell pyroptosis via activation of estrogen receptor α-mediated autophagy. J Adv Res 2021; 28:149-164. [PMID: 33364052 PMCID: PMC7753237 DOI: 10.1016/j.jare.2020.08.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/22/2020] [Accepted: 08/18/2020] [Indexed: 12/21/2022] Open
Abstract
Excessive inflammation and the pyroptosis of vascular endothelial cells caused by estrogen deficiency is one cause of atherosclerosis in post-menopausal women. Because autophagy is highly regulated by estrogen, we hypothesized that estrogen can reduce vascular endothelial cell pyroptosis through estrogen receptor alpha (ERα)-mediated activation of autophagy to improve atherosclerosis in post-menopausal stage. Aortic samples from pro-menopausal and post-menopausal women with ascending aortic arteriosclerosis were analyzed, and bilateral ovariectomized (OVX) female ApoE-/- mice and homocysteine (Hcy)-treated HUVECs were used to analyze the effect of estrogen supplementation therapy. The aortic endothelium showed a decrease in ERα expression and autophagy, but presented an increase in inflammation and pyroptosis in female post-menopausal patients. Estrogen treatment accelerated autophagy and ameliorated cell pyroptosis in the cardiac aortas of OVX ApoE-/- mice and Hcy-treated HUVECs. Estrogen had therapeutic effect on atherosclerosis and improved the symptoms associated with lipid metabolism disorders in OVX ApoE-/- mice. Inhibition and silencing of ERα led to a reduction in the autophagy promoting ability of estrogen and aggravated pyroptosis. Moreover, the inhibition of autophagy promoted pyroptosis and abolished the protective effect of estrogen, but had no influence on ERα expression. Thus, the results of the present study demonstrated that post-menopausal women present decreased autophagy and ERα expression and excessive damage to the ascending aorta. In addition, in vitro and in vivo assay results demonstrated that estrogen prevents atherosclerosis by upregulating ERα expression and subsequently induces autophagy to reduce inflammation and pyroptosis.
Collapse
Affiliation(s)
- Qinghai Meng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tingting Ji
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Chao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jun Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Fu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Suyun Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qi Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210023, China
| | - Fuhua Huang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210023, China
| | - Youran Wang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210023, China
| | - Qichun Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoliang Wang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210023, China
| | - Huimin Bian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
48
|
Mitochondrial DAMPs and altered mitochondrial dynamics in OxLDL burden in atherosclerosis. Mol Cell Biochem 2021; 476:1915-1928. [PMID: 33492610 DOI: 10.1007/s11010-021-04061-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
Atherosclerosis results in life-threatening cardiovascular pathologies, including ischemic heart disease, stroke, myocardial infarction, and peripheral arterial disease. The role of increased serum low-density lipoprotein (LDL) and resultant accumulation of oxidized-LDL (oxLDL) in atheroma formation is well established. Recent findings elucidate the significance of mitochondrial damage-associated molecular patterns (mtDAMPs) in triggering sterile inflammation in concert with oxLDL. The mtDAMPs including mitochondrial DNA (mtDNA), cytochrome C, cardiolipin, heat shock protein 60 (HSP60), mitochondrial transcription factor A (TFAM), and N-formyl peptides, are expected to possess proatherogenic roles. However, limited data are available in the literature. The mtDAMPs initiate sterile inflammation in atherosclerotic lesions via numerous signaling pathways, most of which converge to the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome. Priming the activation of the NLRP3 inflammasome, mtDAMPs promote secretion of proinflammatory cytokines, including interleukin-1β (IL-1β), implicated in atherosclerotic lesions through vascular smooth muscle and fibroblast proliferation, arterial wall thickening, and plaque formation. In this article we critically reviewed and discussed the central role of the NLRP3 inflammasome in mtDAMP-induced sterile inflammation in atherosclerosis with specific components including caspase-1, pregnane X receptor (PXR), adenosine monophosphate activated protein kinase (AMPK), protein phosphatase 2A (PP2A), thioredoxin-interacting protein (TXNIP), and downstream cytokines including IL-1β and IL-18 as potential mediators of atherosclerosis. Better understanding of the proinflammatory effects of mtDAMPs and its pathological association with oxLDL possess immense translational significance for novel therapeutic intervention.
Collapse
|
49
|
Shen ZQ, Huang YL, Teng YC, Wang TW, Kao CH, Yeh CH, Tsai TF. CISD2 maintains cellular homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118954. [PMID: 33422617 DOI: 10.1016/j.bbamcr.2021.118954] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023]
Abstract
CDGSH Iron Sulfur Domain 2 (CISD2) is the causative gene for the disease Wolfram syndrome 2 (WFS2; MIM 604928), which is an autosomal recessive disorder showing metabolic and neurodegenerative manifestations. CISD2 protein can be localized on the endoplasmic reticulum (ER), outer mitochondrial membrane (OMM) and mitochondria-associated membrane (MAM). CISD2 plays a crucial role in the regulation of cytosolic Ca2+ homeostasis, ER integrity and mitochondrial function. Here we summarize the most updated publications and discuss the central role of CISD2 in maintaining cellular homeostasis. This review mainly focuses on the following topics. Firstly, that CISD2 has been recognized as a prolongevity gene and the level of CISD2 is a key determinant of lifespan and healthspan. In mice, Cisd2 deficiency shortens lifespan and accelerates aging. Conversely, a persistently high level of Cisd2 promotes longevity. Intriguingly, exercise stimulates Cisd2 gene expression and thus, the beneficial effects offered by exercise may be partly related to Cisd2 activation. Secondly, that Cisd2 is down-regulated in a variety of tissues and organs during natural aging. Three potential mechanisms that may mediate the age-dependent decrease of Cisd2, via regulating at different levels of gene expression, are discussed. Thirdly, the relationship between CISD2 and cell survival, as well as the potential mechanisms underlying the cell death control, are discussed. Finally we discuss that, in cancers, CISD2 may functions as a double-edged sword, either suppressing or promoting cancer development. This review highlights the importance of the CISD2 in aging and age-related diseases and identifies the urgent need for the translation of available genetic evidence into pharmaceutic interventions in order to alleviate age-related disorders and extend a healthy lifespan in humans.
Collapse
Affiliation(s)
- Zhao-Qing Shen
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Long Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan; Aging and Health Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yuan-Chi Teng
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Tai-Wen Wang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Hsiao Yeh
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Linko, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung, Taiwan.
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan; Aging and Health Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
50
|
Liu Y, Song A, Wu H, Sun Y, Dai M. Paeonol inhibits apoptosis of vascular smooth muscle cells via up-regulation of autophagy by activating class III PI3K/Beclin-1 signaling pathway. Life Sci 2021; 264:118714. [PMID: 33157088 DOI: 10.1016/j.lfs.2020.118714] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022]
Abstract
AIMS The cross talk between autophagy and apoptosis of vascular smooth muscle cells (VSMCs) plays a vital role in the development of atherosclerosis (AS). Paeonol is isolated from the radix of Cortex Moutan with anti-atherosclerotic and anti-apoptosis effects. However, the mechanisms of paeonol on VSMCs apoptosis are still not fully understood. In this study, we aimed to explore whether paeonol could inhibit VSMCs apoptosis though modulating VSMCs autophagy. MATERIALS AND METHODS The proteins expressions were detected by western blotting. Autophagosomes and apoptoticbody formation in VSMCs was observed by transmission electron microscopy (TEM). VSMCs autophagy was detected by monodansylcadaverine (MDC) staining using fluorescence microscopy, while VSMCs apoptosis was determined by 4',6-diamidino-2-phenylindole (DAPI) and flow cytometry. KEY FINDINGS We found that paeonol could significantly increase LC3II protein level, decrease p62 and cleaved caspase-3 proteins levels in aorta of AS mice and ox-LDL-injured VSMCs. Paeonol could augment the number of autophagosomes and reduce the amount of apoptotic bodies in ox-LDL-injured VSMCs. Moreover, paeonol obviously induced VSMCs autophagy compared to ox-LDL group and remarkably suppressed VSMCs apoptosis. However, the effects of paeonol on VSMCs apoptosis could be reversed obviously by 3-MA, the autophagy inhibitor. Furthermore, paeonol could activate class III PI3K-Beclin-1 pathway significantly. Gene silencing of either class III PI3K or Beclin-1 could reverse the effects of paeonol on VSMCs autophagy and apoptosis. SIGNIFICANCE Based on our results, paeonol could induce VSMCs autophagy by activating class III PI3K/Beclin-1 signaling pathway, thus ultimately inhibiting VSMCs apoptosis.
Collapse
Affiliation(s)
- Yarong Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui 230038, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, China
| | - Aiwei Song
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Hongfei Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui 230038, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, China
| | - Yin Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Min Dai
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui 230038, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|