1
|
Jeradeh E, Frangie C, Bazzi S, Daher J. The in vitro effect of myeloperoxidase oxidized LDL on THP-1 derived macrophages. Innate Immun 2024; 30:82-89. [PMID: 39090856 PMCID: PMC11418607 DOI: 10.1177/17534259241269687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Cardiovascular diseases (CVDs) linked to atherosclerosis remains the leading cause of death worldwide. Atherosclerosis is primarily caused by the accumulation of oxidized forms of low density lipoprotein (LDL) in macrophages (MΦs) in the subendothelial layer of arteries leading to foam cell and fatty streak formation. Many studies suggest that LDL that is modified by myeloperoxidase (MPO) is a key player in the development of atherosclerosis. MΦs can adopt a variety of functional phenotypes that include mainly the proinflammatory M1 and the anti-inflammatory M2 MΦ phenotypes which are both implicated in the process of atherogenesis. In fact, MΦs that reside in atherosclerostic lesions were shown to express a variety of phenotypes ranging between the M1- and M2 MΦ types. Recently, we pointed out the involvement of MPO oxidized-LDL (Mox-LDL) in increasing inflammation in MΦs by reducing their secretion of IL-10. Since little is known about Mox-LDL-mediated pro-atherosclerostic responses in MΦs, our study aimed at analyzing the in vitro effects of Mox-LDL at this level through making use of the well-established model of human THP-1-derived Mφs. Our results demonstrate that Mox-LDL has no effect on apoptosis, reactive oxygen species (ROS) generation and cell death in our cell model; yet, interestingly, our results show that Mox-LDL is significantly engulfed at a higher rate in the different MΦ subtypes supporting its key role in foam cell formation during the progression of the disease as well as previous data that were generated using another primary MΦ cell model of atherosclerosis.
Collapse
Affiliation(s)
- Elias Jeradeh
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El-Koura, Lebanon
| | - Christian Frangie
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Gosselies Campus, Gosselies, Belgium
| | - Samer Bazzi
- Department of Biomedical Sciences, Faculty of Medicine and Medical Science, University of Balamand, El-Koura, Lebanon
| | - Jalil Daher
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El-Koura, Lebanon
| |
Collapse
|
2
|
Solanki K, Bezsonov E, Orekhov A, Parihar SP, Vaja S, White FA, Obukhov AG, Baig MS. Effect of reactive oxygen, nitrogen, and sulfur species on signaling pathways in atherosclerosis. Vascul Pharmacol 2024; 154:107282. [PMID: 38325566 DOI: 10.1016/j.vph.2024.107282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease in which fats, lipids, cholesterol, calcium, proliferating smooth muscle cells, and immune cells accumulate in the intima of the large arteries, forming atherosclerotic plaques. A complex interplay of various vascular and immune cells takes place during the initiation and progression of atherosclerosis. Multiple reports indicate that tight control of reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS) production is critical for maintaining vascular health. Unrestricted ROS and RNS generation may lead to activation of various inflammatory signaling pathways, facilitating atherosclerosis. Given these deleterious consequences, it is important to understand how ROS and RNS affect the signaling processes involved in atherogenesis. Conversely, RSS appears to exhibit an atheroprotective potential and can alleviate the deleterious effects of ROS and RNS. Herein, we review the literature describing the effects of ROS, RNS, and RSS on vascular smooth muscle cells, endothelial cells, and macrophages and focus on how changes in their production affect the initiation and progression of atherosclerosis. This review also discusses the contribution of ROS, RNS, and RSS in mediating various post-translational modifications, such as oxidation, nitrosylation, and sulfation, of the molecules involved in inflammatory signaling.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Evgeny Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia; Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia; Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; The Cell Physiology and Pathology Laboratory, Turgenev State University of Orel, Orel, Russia
| | - Alexander Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Suraj P Parihar
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Shivani Vaja
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Fletcher A White
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India.
| |
Collapse
|
3
|
Wang C, Zhao R, Wang Z, Xu T, Huang P. Synthetic ditempolphosphatidylcholine liposome-like nanoparticles for anti-oxidative therapy of atherosclerosis. RSC Adv 2023; 13:16211-16221. [PMID: 37266511 PMCID: PMC10230271 DOI: 10.1039/d3ra01822a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Atherosclerosis (AS), a chronic inflammatory disease, is the leading cause of death worldwide. Anti-oxidative therapy has been developed for AS therapy in light of the critical role of ROS in pathogenesis of AS, but current anti-oxidants have exhibited limited outcomes in the clinic. Herein, new ROS-eliminating liposome-like NPs (Tempol-Lips) were assembled from synthetic lipids that covalently conjugated two Tempol molecules with phosphatidylcholine by esterification reaction. The obtained Tempol-Lips can be efficiently internalized into inflammatory macrophages and attenuated inflammation via scavenging overproduced intracellular ROS. After i.v. administration, Tempol-Lips with nanoscale character accumulated in the plaques of ApoE-/- mice through passive targeting and significantly inhibited the pathogenesis of AS, compared with those treated with control drugs. The therapeutic benefits of Tempol-Lips primarily are ascribed to the reduced local and systematic oxidative stress and inflammation. Preliminary studies in vivo further demonstrated Tempol-Lips were safe and biocompatible after long-term i.v. injection. Conclusively, Tempol-Lips can be developed as a novel anti-AS nanotherapy with potential translation in the clinic.
Collapse
Affiliation(s)
- Chunxiao Wang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| | - Ruifu Zhao
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| | - Zhen Wang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| | - Tingting Xu
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| | - Peng Huang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| |
Collapse
|
4
|
Becker PH, Thérond P, Gaignard P. Targeting mitochondrial function in macrophages: A novel treatment strategy for atherosclerotic cardiovascular disease? Pharmacol Ther 2023; 247:108441. [PMID: 37201736 DOI: 10.1016/j.pharmthera.2023.108441] [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: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 05/20/2023]
Abstract
Atherosclerotic cardiovascular disease is a major cause of morbidity and mortality due to chronic arterial injury caused by hyperlipidemia, hypertension, inflammation and oxidative stress. Recent studies have shown that the progression of this disease is associated with mitochondrial dysfunction and with the accumulation of mitochondrial alterations within macrophages of atherosclerotic plaques. These alterations contribute to processes of inflammation and oxidative stress. Among the many players involved, macrophages play a pivotal role in atherogenesis as they can exert both beneficial and deleterious effects due to their anti- and pro-inflammatory properties. Their atheroprotective functions, such as cholesterol efflux and efferocytosis, as well as the maintenance of their polarization towards an anti-inflammatory state, are particularly dependent on mitochondrial metabolism. Moreover, in vitro studies have demonstrated deleterious effects of oxidized LDL on macrophage mitochondrial function, resulting in a switch to a pro-inflammatory state and to a potential loss of atheroprotective capacity. Therefore, preservation of mitochondrial function is now considered a legitimate therapeutic strategy. This review focuses on the potential therapeutic strategies that could improve the mitochondrial function of macrophages, enabling them to maintain their atheroprotective capacity. These emerging therapies could play a valuable role in counteracting the progression of atherosclerotic lesions and possibly inducing their regression.
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.
| | - 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
| | - 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
| |
Collapse
|
5
|
Ning Y, Jia Y, Yang Y, Wen W, Huang M, Liu S, Yang Y, Dong Y, Zhang M. Thyroid hormones inhibit apoptosis of macrophage induced by oxidized low-density lipoprotein. Biofactors 2022; 48:86-99. [PMID: 34882872 DOI: 10.1002/biof.1803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
Increasing evidence suggests that hypothyroidism aggravates atherosclerosis. Macrophage apoptosis plays a significant role in the development of atherosclerotic plaque. We aimed to explore the effect of thyroid hormones on macrophage apoptosis induced by oxidized low-density lipoprotein (oxLDL). Peripheral blood samples from 20 patients (normal group, hypothyroidism group, coronary artery disease [CAD] group, hypothyroidism + CAD group) were collected to perform messenger RNA microarray analysis. Bioinformatics analysis identified apoptosis and mitogen-activated protein kinase (MAPK) signaling as differentially expressed pathways between CAD and hypothyroidism + CAD group. In vitro, thyroid hormones concentration-dependently promoted cell survival and inhibited apoptosis in oxLDL-treated RAW264.7 macrophages, along with elevated extracellular signal-regulated kinases 1 and 2 (Erk1/2) phosphorylation. The STRING database showed an interaction of thyroid hormone receptor alpha1 (TRα1) and MAPK pathway. TRα1 knockdown increased cell apoptosis and decreased Erk1/2 phosphorylation. Erk1/2 inhibitor aggravated macrophage apoptosis. Moreover, thyroid hormones inhibited oxidative stress in oxLDL-treated macrophages. The study indicates that thyroid hormones concentration-dependently attenuate oxLDL-induced macrophage apoptosis through activating TRα1-Erk1/2 pathway and inhibiting oxidative stress, which implies a potential mechanism of hypothyroid-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Yu Ning
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yifan Jia
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yunxiao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wanwan Wen
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mengling Huang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Sheng Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yugang Dong
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Ginsenoside Rg1 can restore hematopoietic function by inhibiting Bax translocation-mediated mitochondrial apoptosis in aplastic anemia. Sci Rep 2021; 11:12742. [PMID: 34140535 PMCID: PMC8211841 DOI: 10.1038/s41598-021-91471-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
The present study investigated, the anti-apoptotic activity of Ginsenoside Rg1 (Rg1) via inhibition of Bax translocation and the subsequent recovery of hematopoietic function. Mitochondrial apoptosis in bone marrow mononuclear cells (BMNCs) was observed in aplastic anemia (AA) patients. To establish a mouse model of AA, BALB/c mice were transplanted with lymph node cells from DBA/2 donor mice via vein injection after treatment with Co60 γ-radiation. After treatment with Rg1 for 14 days, the peripheral blood and Lin–Sca-1 + c-Kit + (LSK) cell counts of the treated group were increased compared with those of the untreated model mice. In in vivo and in vitro tests of LSKs, Rg1 was found to increase mitochondrial number and the ratio of Bcl-2/Bax and to decrease damage to the mitochondrial inner and outer membranes, the mitochondrial Bax level and the protein levels of mitochondrial apoptosis-related proteins AIF and Cyt-C by decreasing the ROS level. Rg1 also improved the concentration–time curve of MAO and COX and levels of ATP, ADP and AMP in an in vitro test. In addition, high levels of Bax mitochondrial translocation could be corrected by Rg1 treatment. Levels of markers of mitochondrial apoptosis in the Rg1-treated group were significantly better than those in the AA model group, implying that Rg1 might improve hematopoietic stem cells and thereby restore hematopoietic function in AA by suppressing the mitochondrial apoptosis mediated by Bax translocation.
Collapse
|
7
|
Sun Y, Lu Y, Saredy J, Wang X, Drummer Iv C, Shao Y, Saaoud F, Xu K, Liu M, Yang WY, Jiang X, Wang H, Yang X. ROS systems are a new integrated network for sensing homeostasis and alarming stresses in organelle metabolic processes. Redox Biol 2020; 37:101696. [PMID: 32950427 PMCID: PMC7767745 DOI: 10.1016/j.redox.2020.101696] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are critical for the progression of cardiovascular diseases, inflammations and tumors. However, the mechanisms of how ROS sense metabolic stress, regulate metabolic pathways and initiate proliferation, inflammation and cell death responses remain poorly characterized. In this analytic review, we concluded that: 1) Based on different features and functions, eleven types of ROS can be classified into seven functional groups: metabolic stress-sensing, chemical connecting, organelle communication, stress branch-out, inflammasome-activating, dual functions and triple functions ROS. 2) Among the ROS generation systems, mitochondria consume the most amount of oxygen; and nine types of ROS are generated; thus, mitochondrial ROS systems serve as the central hub for connecting ROS with inflammasome activation, trained immunity and immunometabolic pathways. 3) Increased nuclear ROS production significantly promotes cell death in comparison to that in other organelles. Nuclear ROS systems serve as a convergent hub and decision-makers to connect unbearable and alarming metabolic stresses to inflammation and cell death. 4) Balanced ROS levels indicate physiological homeostasis of various metabolic processes in subcellular organelles and cytosol, while imbalanced ROS levels present alarms for pathological organelle stresses in metabolic processes. Based on these analyses, we propose a working model that ROS systems are a new integrated network for sensing homeostasis and alarming stress in metabolic processes in various subcellular organelles. Our model provides novel insights on the roles of the ROS systems in bridging metabolic stress to inflammation, cell death and tumorigenesis; and provide novel therapeutic targets for treating those diseases. (Word count: 246).
Collapse
Affiliation(s)
- Yu Sun
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Yifan Lu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Jason Saredy
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xianwei Wang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Charles Drummer Iv
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Ying Shao
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Fatma Saaoud
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Keman Xu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Ming Liu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - William Y Yang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaohua Jiang
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA; Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA; Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
8
|
Helal MG, Samra YA. Irbesartan mitigates acute liver injury, oxidative stress, and apoptosis induced by acetaminophen in mice. J Biochem Mol Toxicol 2020; 34:e22447. [DOI: 10.1002/jbt.22447] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/20/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Manar G. Helal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy Mansoura University Mansoura Egypt
| | - Yara A. Samra
- Department of Biochemistry, Faculty of Pharmacy Mansoura University Mansoura Egypt
| |
Collapse
|
9
|
Tao J, Zhang D, Man Y, Wang W, Bi Y. RETRACTED ARTICLE: Sodium ferulate inhibits high-fat diet-induced inflammatory factors expression in human umbilical vein endothelial cells. CYTOL GENET+ 2019. [DOI: 10.3103/s0095452717030124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
10
|
Gao LN, Zhou X, Lu YR, Li K, Gao S, Yu CQ, Cui YL. Dan-Lou Prescription Inhibits Foam Cell Formation Induced by ox-LDL via the TLR4/NF-κB and PPARγ Signaling Pathways. Front Physiol 2018; 9:590. [PMID: 29896109 PMCID: PMC5987004 DOI: 10.3389/fphys.2018.00590] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 05/02/2018] [Indexed: 01/01/2023] Open
Abstract
Atherosclerosis is the major worldwide cause of mortality for patients with coronary heart disease. Many traditional Chinese medicine compound prescriptions for atherosclerosis treatment have been tried in patients. Dan-Lou prescription, which is improved from Gualou-Xiebai-Banxia decoction, has been used to treat chest discomfort (coronary atherosclerosis) for approximately 2,000 years in China. Although the anti-inflammatory activities of Dan-Lou prescription have been proposed previously, the mechanism remains to be explored. Based on the interaction between inflammation and atherosclerosis, we further investigated the effect of Dan-Lou prescription on macrophage-derived foam cell formation and disclosed the underlying mechanisms. In the oxidative low-density lipoprotein (ox-LDL) induced foam cells model using murine macrophage RAW 264.7 cells, the ethanol extract from Dan-Lou prescription (EEDL) reduced ox-LDL uptake and lipid deposition by inhibiting the protein and mRNA expression of Toll-like receptor (TLR)4 and scavenger receptor (SR)B1. After stimulation with ox-LDL, the metabolic profile of macrophages was also changed, while the intervention of the EEDL mainly regulated the metabolism of isovalerylcarnitine, arachidonic acid, cholesterol, aspartic acid, arginine, lysine, L-glutamine and phosphatidylethanolamine (36:3), which participated in the regulation of the inflammatory response, lipid accumulation and cell apoptosis. In total, 27 inflammation-related gene targets were screened, and the biological mechanisms, pathways and biological functions of the EEDL on macrophage-derived foam cells were systemically analyzed by Ingenuity Pathway Analysis system (IPA). After verification, we found that EEDL alleviated ox-LDL induced macrophage foam cell formation by antagonizing the mRNA and protein over-expression of PPARγ, blocking the phosphorylation of IKKα/β, IκBα and NF-κB p65 and maintaining the expression balance between Bax and Bcl-2. In conclusion, we provided evidences that Dan-Lou prescription effectively attenuated macrophage foam cell formation via the TLR4/NF-κB and PPARγ signaling pathways.
Collapse
Affiliation(s)
- Li-Na Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,College of Pharmacy, Jining Medical University, Rizhao, China.,Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Research Laboratory of Prescription Compatibility among Components, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Zhou
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Research Laboratory of Prescription Compatibility among Components, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu-Ren Lu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Research Laboratory of Prescription Compatibility among Components, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kefeng Li
- Tianjin Sunnypeak Biotech Co., Ltd., Tianjin, China
| | - Shan Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chun-Quan Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuan-Lu Cui
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Research Laboratory of Prescription Compatibility among Components, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
11
|
Nowak M, Tardivel S, Nguyen-Khoa T, Abreu S, Allaoui F, Fournier N, Chaminade P, Paul JL, Lacour B. Mycophenolate Mofetil and Rapamycin Induce Apoptosis in the Human Monocytic U937 Cell Line Through Two Different Pathways. J Cell Biochem 2017; 118:3480-3487. [PMID: 28345768 DOI: 10.1002/jcb.26007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
Transplant vasculopathy may be considered as an accelerated form of atherosclerosis resulting in chronic rejection of vascularized allografts. After organ transplantation, a diffuse intimal thickening is observed, leading to the development of an atherosclerosis plaque due to a significant monocyte infiltration. This results from a chronic inflammatory process induced by the immune response. In this study, we investigated the impact of two immunosuppressive drugs used in therapy initiated after organ transplantation, mycophenolate mofetil, and rapamycin, on the apoptotic response of monocytes induced or not by oxidized LDL. Here we show the pro-apoptotic effect of these two drugs through two distinct signaling pathways and we highlight a synergistic effect of rapamycin on apoptosis induced by oxidized LDL. In conclusion, since immunosuppressive therapy using mycophenolate mofetil or rapamycin can increase the cell death in a monocyte cell line, this treatment could exert similar effects on human monocytes in transplant patients, and thus, prevent transplant vasculopathy, atherosclerosis development, and chronic allograft rejection. J. Cell. Biochem. 118: 3480-3487, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Maxime Nowak
- Lip(Sys)2-Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Sylviane Tardivel
- Lip(Sys)2-Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France.,Ecole Pratique des Hautes Etudes, Laboratoire nutrition lipidique et apoptose dans le système vasculaire-Faculté de Pharmacie, 92290 Châtenay-Malabry, France
| | - Thao Nguyen-Khoa
- Laboratoire de Biochimie générale-AP-HP (Assistance publique-Hôpitaux de Paris)-Hôpital Necker Enfants Malades, 75015 Paris, France
| | - Sonia Abreu
- Lip(Sys)2-Chimie Analytique Pharmaceutique, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Fatima Allaoui
- Lip(Sys)2-Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Natalie Fournier
- Lip(Sys)2-Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France.,Laboratoire de Biochimie-AP-HP (Assistance publique-Hôpitaux de Paris)-Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Pierre Chaminade
- Lip(Sys)2-Chimie Analytique Pharmaceutique, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Jean-Louis Paul
- Lip(Sys)2-Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France.,Laboratoire de Biochimie-AP-HP (Assistance publique-Hôpitaux de Paris)-Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Bernard Lacour
- Lip(Sys)2-Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France.,Ecole Pratique des Hautes Etudes, Laboratoire nutrition lipidique et apoptose dans le système vasculaire-Faculté de Pharmacie, 92290 Châtenay-Malabry, France.,Laboratoire de Biochimie générale-AP-HP (Assistance publique-Hôpitaux de Paris)-Hôpital Necker Enfants Malades, 75015 Paris, France
| |
Collapse
|
12
|
Chen HM, Luo H, Zeng WB, Liu B, Huang JC, Liu M, Zeng YJ, Zheng Q, Li JQ, Sun XG, Zhou YC. Salvianolic acid B attenuates oxidized low-density lipoprotein-induced endothelial cell apoptosis through inhibition of oxidative stress, p53, and caspase-3 pathways. Chin J Integr Med 2017:10.1007/s11655-016-2645-4. [PMID: 28116660 DOI: 10.1007/s11655-016-2645-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the effect of salvianolic acid B (Sal B) on oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) apoptosis and the possible mechanism. METHODS HUVECs were divided into 6 groups, including control group, ox-LDL group, vitamin C group (positive control), and 5, 10 and 20 μg/mL Sal B groups. Cell viability of HUVECs was determined by 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The anti-apoptotic effect of Sal B was tested by Hoechst 33258 staining and Annexin V/propidium iodide flflow cytometry analysis. Apoptosis-related genes (p53, Bcl-2 and Bax) expression and caspase-3 activity were also determined. Oxidative stress markers malondialdehyde (MDA) and superoxide dismutase (SOD) were determined by the corresponding kits. RESULTS In HUVECs, ox-LDL signifificantly reduced cell viability and induced apoptosis (P<0.05 or P<0.01), however, Sal B diminished the effects of ox-LDL in a dose-dependent manner (P<0.05). Moreover, 10 and 20 μg/mL Sal B reduced the expression levels of p53, increased the Bcl-2/Bax ratio and inhibited the caspase-3 activity in ox-LDL-treated HUVECs (P<0.05). In addition, 5, 10 and 20 μg/mL Sal B signifificantly enhanced the activity of SOD, while decreased the level of MDA in the HUVECs which treated with ox-LDL (P<0.05). CONCLUSION Sal B exhibited anti-apoptotic effects in ox-LDL-induced endothelial cell injury by suppressing oxidative stress, p53, and caspase-3.
Collapse
Affiliation(s)
- Hong-Mei Chen
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hao Luo
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Bi Zeng
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Bin Liu
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Cheng Huang
- The Fifth Department of Internal Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong Province, 523000, China
| | - Min Liu
- Department of Parasitology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yan-Jin Zeng
- The Fifth Department of Internal Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong Province, 523000, China
| | - Qiang Zheng
- Department of Traditional Chinese Medicine, Shantou Central Hospital, Shantou, Guangdong Province, 515031, China
| | - Ji-Qiang Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xue-Gang Sun
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Ying-Chun Zhou
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
13
|
Tao J, Zhang D, Man Y, Wang W, Bi Y. Sodium ferulate inhibits high-fat diet-induced inflammatory factors expression in human umbilical vein endothelial cells. Mol Cell Toxicol 2016. [DOI: 10.1007/s13273-016-0015-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
14
|
Shao Q, Han F, Peng S, He B. Nur77 inhibits oxLDL induced apoptosis of macrophages via the p38 MAPK signaling pathway. Biochem Biophys Res Commun 2016; 471:633-8. [DOI: 10.1016/j.bbrc.2016.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 01/02/2016] [Indexed: 11/29/2022]
|
15
|
Mahmoudi MJ, Saboor-Yaraghi AA, Zabetian-Targhi F, Siassi F, Zarnani AH, Eshraghian MR, Shokri F, Rezaei N, Kalikias Y, Mahmoudi M. Vitamin A Decreases Cytotoxicity of Oxidized Low-Density Lipoprotein in Patients with Atherosclerosis. Immunol Invest 2015; 45:52-62. [PMID: 26700065 DOI: 10.3109/08820139.2015.1095208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Oxidized low-density lipoprotein (ox-LDL) is implicated in initiation and progression of atherosclerosis. Previously, we found that ox-LDL increases vulnerability of peripheral blood mononuclear cells (PBMCs) in atherosclerotic patients compared to controls. Vitamin A induces proliferation of PBMCs. The aim of this study was to determine the effect of vitamin A supplementation on PBMC survival against LDL and different doses of ox-LDL. METHOD In this double-blind placebo-controlled trial, we recruited 35 atherosclerotic patients and 38 healthy controls and randomly allocated them into placebo and vitamin A groups, which received either placebo or 25,000 IU/day of vitamin A for 3 months. PBMCs were isolated, cultured, and stimulated by 1 µg/mL LDL as well as 1 µg/mL and 50 µg/mL ox-LDL. The stimulation indexes (SIs) of PBMCs were calculated to identify cell viability. Additionally, the circulating ox-LDL levels were measured by ELISA. RESULTS Viability of PBMCs stimulated by 50 µg/mL ox-LDL significantly increased following vitamin A supplementation in patients (p < 0.01). The levels of circulating ox-LDL were not changed by vitamin A treatment. Ox-LDL levels were strongly and positively correlated to SI of PBMCs stimulated by 1 µg/mL LDL and1 µg/mL ox-LDL in all groups. CONCLUSION Vitamin A decreases cytotoxicity of high-dose ox-LDL and improves PBMC viability. The protective effect of vitamin A is not mediated by an antioxidative mechanism, but may instead have been due to intracellular protection of the apoptotic machinery or induction of proliferation of the cells. Higher levels of ox-LDL increase PBMC irritability in all participants.
Collapse
Affiliation(s)
- Mohammad Jafar Mahmoudi
- a Division of Cardiology, Department of Internal Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Ali-Akbar Saboor-Yaraghi
- b Department of Cellular Molecular Nutrition , School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences , Tehran , Iran
| | - Fateme Zabetian-Targhi
- b Department of Cellular Molecular Nutrition , School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences , Tehran , Iran
| | - Fereydoon Siassi
- c Department of Community Nutrition , School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences , Tehran , Iran
| | - Amir Hassan Zarnani
- d Nanobiotechnology Research Center , Avicenna Research Institute, ACECR , Tehran , Iran.,e Immunology Research Center, Faculty of Medicine , Iran University of Medical Sciences , Tehran , Iran
| | - Mohammad Reza Eshraghian
- f Department of Biostatistics , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Fazel Shokri
- g Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Nima Rezaei
- h Molecular Immunology Research Center, Department of Immunology , School of Medicine, Tehran University of Medical Sciences , Tehran , Iran.,i Research Group for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran
| | - Yas Kalikias
- b Department of Cellular Molecular Nutrition , School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences , Tehran , Iran
| | - Maryam Mahmoudi
- b Department of Cellular Molecular Nutrition , School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
16
|
Wang J, Chen L, Li H, Yang J, Gong Z, Wang B, Zhao X. Clopidogrel reduces apoptosis and promotes proliferation of human vascular endothelial cells induced by palmitic acid via suppression of the long non-coding RNA HIF1A-AS1 in vitro. Mol Cell Biochem 2015; 404:203-10. [PMID: 25761653 DOI: 10.1007/s11010-015-2379-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/05/2015] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is recognized as a major and increasing health problem affected older subjects in China, and clopidogrel has been widely used for treatment of CVD patients such as atherosclerosis, myocardial infarction, and myocardial ischaemia-reperfusion damage. However, the molecular mechanisms of clopidogrel for treatment of CVD are only partially understood. This study investigated the effects of clopidogrel on palmitic acid-induced damage of human vascular endothelial cells (HUVECs), and the molecular mechanisms of LncRNA HIF1A-AS1 in regulating the proliferation and apoptosis of HUVECs in vitro. We firstly established a damage model of HUVECs through palmitic acid (PA) treatment. And the effect of clopidogrel reducing PA-induced apoptosis of HUVECs was observed by the flow cytometric measurement. To further understand the molecular mechanism of clopidogrel rescues PA-induced apoptosis, we used human LncRNA PCR array to compare the LncRNA expression profile difference between clopidogrel-treated cells and control cells. The expression of LncRNA HIF 1 alpha-antisense RNA 1 (HIF1A-AS1) was significantly altered in clopidogrel-treated cells. We further proved that suppression of HIF1A-AS1 by siRNA reduce PA-induced apoptosis and promote proliferation of HUVECs. Furthermore, we also demonstrated inhibition apoptosis effect by HIF1A-AS1 is related to mitochondrial apoptosis pathway. Hence, our results suggest that clopidogrel rescues apoptosis and promotes proliferation of PA-induced damage model of HUVECs through inhibiting the mediator LncRNA HIF1A-AS1. These findings indicate that LncRNA HIF1A-AS1 may play an important role in the pathogenesis of CVD, and provide a novel molecular mechanism of clopidogrel for treatment of CVD.
Collapse
Affiliation(s)
- Jing Wang
- Department of Rheumatology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Vlaminck B, Calay D, Genin M, Sauvage A, Ninane N, Zouaoui Boudjeltia K, Raes M, Michiels C. Effects of copper sulfate-oxidized or myeloperoxidase-modified LDL on lipid loading and programmed cell death in macrophages under hypoxia. HYPOXIA (AUCKLAND, N.Z.) 2014; 2:153-169. [PMID: 27774474 PMCID: PMC5045064 DOI: 10.2147/hp.s65242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Atheromatous plaques contain heavily lipid-loaded macrophages that die, hence generating the necrotic core of these plaques. Since plaque instability and rupture is often correlated with a large necrotic core, it is important to understand the mechanisms underlying foam cell death. Furthermore, macrophages within the plaque are associated with hypoxic areas but little is known about the effect of low oxygen partial pressure on macrophage death. The aim of this work was to unravel macrophage death mechanisms induced by oxidized low-density lipoproteins (LDL) both under normoxia and hypoxia. Differentiated macrophages were incubated in the presence of native, copper sulfate-oxidized, or myeloperoxidase-modified LDL. The unfolded protein response, apoptosis, and autophagy were then investigated. The unfolded protein response and autophagy were triggered by myeloperoxidase-modified LDL and, to a larger extent, by copper sulfate-oxidized LDL. Electron microscopy observations showed that oxidized LDL induced excessive autophagy and apoptosis under normoxia, which were less marked under hypoxia. Myeloperoxidase-modified LDL were more toxic and induced a higher level of apoptosis. Hypoxia markedly decreased apoptosis and cell death, as marked by caspase activation. In conclusion, the cell death pathways induced by copper sulfate-oxidized and myeloperoxidase-modified LDL are different and are differentially modulated by hypoxia.
Collapse
Affiliation(s)
- Benoit Vlaminck
- Laboratory of Biochemistry and Cellular Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Damien Calay
- Laboratory of Biochemistry and Cellular Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Marie Genin
- Laboratory of Biochemistry and Cellular Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Aude Sauvage
- Laboratory of Biochemistry and Cellular Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Noelle Ninane
- Laboratory of Biochemistry and Cellular Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Karim Zouaoui Boudjeltia
- Laboratory of Experimental Medicine (ULB 222 Unit), Universite Libre de Bruxelles, CHU de Charleroi, Charleroi, Belgium
| | - Martine Raes
- Laboratory of Biochemistry and Cellular Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Carine Michiels
- Laboratory of Biochemistry and Cellular Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| |
Collapse
|
18
|
Orekhov AN, Bobryshev YV, Sobenin IA, Melnichenko AA, Chistiakov DA. Modified low density lipoprotein and lipoprotein-containing circulating immune complexes as diagnostic and prognostic biomarkers of atherosclerosis and type 1 diabetes macrovascular disease. Int J Mol Sci 2014; 15:12807-41. [PMID: 25050779 PMCID: PMC4139876 DOI: 10.3390/ijms150712807] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 06/29/2014] [Accepted: 07/03/2014] [Indexed: 12/14/2022] Open
Abstract
In atherosclerosis; blood low-density lipoproteins (LDL) are subjected to multiple enzymatic and non-enzymatic modifications that increase their atherogenicity and induce immunogenicity. Modified LDL are capable of inducing vascular inflammation through activation of innate immunity; thus, contributing to the progression of atherogenesis. The immunogenicity of modified LDL results in induction of self-antibodies specific to a certain type of modified LDL. The antibodies react with modified LDL forming circulating immune complexes. Circulating immune complexes exhibit prominent immunomodulatory properties that influence atherosclerotic inflammation. Compared to freely circulating modified LDL; modified LDL associated with the immune complexes have a more robust atherogenic and proinflammatory potential. Various lipid components of the immune complexes may serve not only as diagnostic but also as essential predictive markers of cardiovascular events in atherosclerosis. Accumulating evidence indicates that LDL-containing immune complexes can also serve as biomarker for macrovascular disease in type 1 diabetes.
Collapse
Affiliation(s)
- Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia.
| | - Yuri V Bobryshev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia.
| | - Igor A Sobenin
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia.
| | - Alexandra A Melnichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia.
| | - Dimitry A Chistiakov
- Department of Medical Nanobiotechnology, Pirogov Russian State Medical University, Moscow 117997, Russia.
| |
Collapse
|
19
|
Suppressive effects of irbesartan on inflammation and apoptosis in atherosclerotic plaques of apoE-/- mice: molecular imaging with 14C-FDG and 99mTc-annexin A5. PLoS One 2014; 9:e89338. [PMID: 24586699 PMCID: PMC3929710 DOI: 10.1371/journal.pone.0089338] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/20/2014] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES To investigate the effects of irbesartan on inflammation and apoptosis in atherosclerotic plaques by histochemical examination and molecular imaging using (14)C-FDG and (99m)Tc-annexin A5. BACKGROUND Irbesartan has a peroxisome proliferator-activated receptor gamma (PPARγ) activation property in addition to its ability to block the AT1 receptor. Accordingly, irbesartan may exert further anti-inflammatory and anti-apoptotic effects in atherosclerotic plaques. However, such effects of irbesartan have not been fully investigated. Molecular imaging using (18)F-FDG and (99m)Tc-annexin A5 is useful for evaluating inflammation and apoptosis in atherosclerotic plaques. METHODS Female apoE(-/-) mice were treated with irbesartan-mixed (50 mg/kg/day) or irbesartan-free (control) diet for 12 weeks (n = 11/group). One week after the treatment, the mice were co-injected with (14)C-FDG and (99m)Tc-annexin A5, and cryostat sections of the aortic root were prepared. Histochemical examination with Movat's pentachrome (plaque size), Oil Red O (lipid deposition), Mac-2 (macrophage infiltration), and TUNEL (apoptosis) stainings were performed. Dual-tracer autoradiography was carried out to evaluate the levels of (14)C-FDG and (99m)Tc-annexin A5 in plaques (%ID×kg). In vitro experiments were performed to investigate the mechanism underlying the effects. RESULTS Histological examination indicated that irbesartan treatment significantly reduced plaque size (to 56.4%±11.1% of control), intra-plaque lipid deposition (53.6%±20.2%) and macrophage infiltration (61.9%±20.8%) levels, and the number of apoptotic cells (14.5%±16.6%). (14)C-FDG (43.0%±18.6%) and (99m)Tc-annexin A5 levels (45.9%±16.8%) were also significantly reduced by irbesartan treatment. Irbesartan significantly suppressed MCP-1 mRNA expression in TNF-α stimulated THP-1 monocytes (64.8%±8.4% of un-treated cells). PPARγ activation was observed in cells treated with irbesartan (134%±36% at 3 µM to 3329%±218% at 81 µM) by a PPARγ reporter assay system. CONCLUSIONS Remissions of inflammation and apoptosis as potential therapeutic effects of irbesartan on atherosclerosis were observed. The usefulness of molecular imaging using (18)F-FDG and (99m)Tc-annexin A5 for evaluating the therapeutic effects of irbesartan on atherosclerosis was also suggested.
Collapse
|
20
|
Afman L, Milenkovic D, Roche HM. Nutritional aspects of metabolic inflammation in relation to health--insights from transcriptomic biomarkers in PBMC of fatty acids and polyphenols. Mol Nutr Food Res 2014; 58:1708-20. [PMID: 24449395 DOI: 10.1002/mnfr.201300559] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/27/2013] [Accepted: 12/03/2013] [Indexed: 11/08/2022]
Abstract
Recent research has highlighted potential important interaction between metabolism and inflammation, within the context of metabolic health and nutrition, with a view to preventing diet-related disease. In addition to this, there is a paucity of evidence in relation to accurate biomarkers that are capable of reflecting this important biological interplay or relationship between metabolism and inflammation, particularly in relation to diet and health. Therefore the objective of this review is to highlight the potential role of transcriptomic approaches as a tool to capture the mechanistic basis of metabolic inflammation. Within this context, this review has focused on the potential of peripheral blood mononuclear cells transcriptomic biomarkers, because they are an accessible tissue that may reflect metabolism and subacute chronic inflammation. Also these pathways are often dysregulated in the common diet-related diseases obesity, type 2 diabetes, and cardiovascular disease, thus may be used as markers of systemic health. The review focuses on fatty acids and polyphenols, two classes of nutrients/nonnutrient food components that modulate metabolism/inflammation, which we have used as an example of a proof-of-concept with a view to understanding the extent to which transcriptomic biomarkers are related to nutritional status and/or sensitive to dietary interventions. We show that both nutritional components modulate inflammatory markers at the transcriptomic level with the capability of profiling pro- and anti-inflammatory mechanisms in a bidirectional fashion; to this end transcriptomic biomarkers may have potential within the context of metabolic inflammation. This transcriptomic biomarker approach may be a sensitive indicator of nutritional status and metabolic health.
Collapse
Affiliation(s)
- Lydia Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands
| | | | | |
Collapse
|
21
|
Zhao Y, Zhao S, Kuge Y, Strauss HW, Blankenberg FG, Tamaki N. Attenuation of Apoptosis by Telmisartan in Atherosclerotic Plaques of Apolipoprotein E
−/−
Mice: Evaluation Using Technetium 99m–Annexin A5. Mol Imaging 2013. [DOI: 10.2310/7290.2012.00045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Yan Zhao
- From the Departments of Tracer Kinetics and Bioanalysis and Nuclear Medicine, Graduate School of Medicine, and Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan; Department of Nuclear Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; and Department of Pediatric Radiology, Stanford University School of Medicine, Palo Alto, CA
| | - Songji Zhao
- From the Departments of Tracer Kinetics and Bioanalysis and Nuclear Medicine, Graduate School of Medicine, and Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan; Department of Nuclear Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; and Department of Pediatric Radiology, Stanford University School of Medicine, Palo Alto, CA
| | - Yuji Kuge
- From the Departments of Tracer Kinetics and Bioanalysis and Nuclear Medicine, Graduate School of Medicine, and Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan; Department of Nuclear Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; and Department of Pediatric Radiology, Stanford University School of Medicine, Palo Alto, CA
| | - H. William Strauss
- From the Departments of Tracer Kinetics and Bioanalysis and Nuclear Medicine, Graduate School of Medicine, and Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan; Department of Nuclear Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; and Department of Pediatric Radiology, Stanford University School of Medicine, Palo Alto, CA
| | - Francis G. Blankenberg
- From the Departments of Tracer Kinetics and Bioanalysis and Nuclear Medicine, Graduate School of Medicine, and Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan; Department of Nuclear Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; and Department of Pediatric Radiology, Stanford University School of Medicine, Palo Alto, CA
| | - Nagara Tamaki
- From the Departments of Tracer Kinetics and Bioanalysis and Nuclear Medicine, Graduate School of Medicine, and Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan; Department of Nuclear Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; and Department of Pediatric Radiology, Stanford University School of Medicine, Palo Alto, CA
| |
Collapse
|
22
|
Lopez S, Jaramillo S, Varela LM, Ortega A, Bermudez B, Abia R, Muriana FJG. p38 MAPK protects human monocytes from postprandial triglyceride-rich lipoprotein-induced toxicity. J Nutr 2013; 143:620-6. [PMID: 23486980 DOI: 10.3945/jn.113.174656] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Postprandial triglyceride (TG)-rich lipoproteins (TRLs) transport dietary fatty acids through the circulatory system to satisfy the energy and structural needs of the tissues. However, fatty acids are also able to modulate gene expression and/or induce cell death. We investigated the underlying mechanism by which postprandial TRLs of different fatty acid compositions can induce cell death in human monocytes. Three types of dietary fat [refined olive oil (ROO), high-palmitic sunflower oil (HPSO), and butter] with progressively increasing SFA:MUFA ratios (0.18, 0.41, and 2.08, respectively) were used as a source of postprandial TRLs (TRL-ROO, TRL-HPSO, and TRL-BUTTER) from healthy men. The monocytic cell line THP-1 was used as a model for this study. We demonstrated that postprandial TRLs increased intracellular lipid accumulation (31-106%), reactive oxygen species production (268-349%), DNA damage (133-1467%), poly(ADP-ribose) polymerase 1 (800-1710%) and caspase-3 (696-1244%) activities, and phosphorylation of c-Jun NH2-terminal kinase (JNK) (54 kDa, 141-288%) and p38 (24-92%). These effects were significantly greater with TRL-BUTTER, and TRL-ROO did not induce DNA damage, DNA fragmentation, or p38 phosphorylation. In addition, blockade of p38, but not of JNK, significantly decreased intracellular lipid accumulation and increased cell death in postprandial TRL-treated cells. These results suggest that in human monocytes, p38 is involved in survival signaling pathways that protect against the lipid-mediated cytotoxicity induced by postprandial TRLs that are abundant in saturated fatty acids.
Collapse
Affiliation(s)
- Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council, Seville, Spain.
| | | | | | | | | | | | | |
Collapse
|
23
|
Nussbaum C, Klinke A, Adam M, Baldus S, Sperandio M. Myeloperoxidase: a leukocyte-derived protagonist of inflammation and cardiovascular disease. Antioxid Redox Signal 2013; 18:692-713. [PMID: 22823200 DOI: 10.1089/ars.2012.4783] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE The heme-enzyme myeloperoxidase (MPO) is one of the major neutrophil bactericidal proteins and is stored in large amounts inside azurophilic granules of neutrophils. Upon cell activation, MPO is released and extracellular MPO has been detected in a wide range of acute and chronic inflammatory conditions. Recent ADVANCES AND CRITICAL ISSUES: Apart from its role during infection, MPO has emerged as a critical modulator of inflammation throughout the last decade and is currently discussed in the initiation and propagation of cardiovascular diseases. MPO-derived oxidants (e.g., hypochlorous acid) interfere with various cell functions and contribute to tissue injury. Recent data also suggest that MPO itself exerts proinflammatory properties independent of its catalytic activity. Despite advances in unraveling the complex action of MPO and MPO-derived oxidants, further research is warranted to determine the precise nature and biological role of MPO in inflammation. FUTURE DIRECTIONS The identification of MPO as a central player in inflammation renders this enzyme an attractive prognostic biomarker and a potential target for therapeutic interventions. A better understanding of the (patho-) physiology of MPO is essential for the development of successful treatment strategies in acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Claudia Nussbaum
- Walter Brendel Centre for Experimental Medicine, Ludwig-Maximilians-University Munich, Munich, Germany.
| | | | | | | | | |
Collapse
|
24
|
Degradation of heparan sulfate proteoglycans enhances oxidized-LDL-mediated autophagy and apoptosis in human endothelial cells. Biochem Biophys Res Commun 2012; 426:106-11. [DOI: 10.1016/j.bbrc.2012.08.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 08/09/2012] [Indexed: 11/19/2022]
|
25
|
Hulsmans M, Van Dooren E, Holvoet P. Mitochondrial Reactive Oxygen Species and Risk of Atherosclerosis. Curr Atheroscler Rep 2012; 14:264-76. [DOI: 10.1007/s11883-012-0237-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
26
|
Human monocytes are severely impaired in base and DNA double-strand break repair that renders them vulnerable to oxidative stress. Proc Natl Acad Sci U S A 2011; 108:21105-10. [PMID: 22160723 DOI: 10.1073/pnas.1111919109] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Monocytes are key players in the immune system. Crossing the blood barrier, they infiltrate tissues and differentiate into (i) macrophages that fight off pathogens and (ii) dendritic cells (DCs) that activate the immune response. A hallmark of monocyte/macrophage activation is the generation of reactive oxygen species (ROS) as a defense against invading microorganisms. How monocytes, macrophages, and DCs in particular respond to ROS is largely unknown. Here we studied the sensitivity of primary human monocytes isolated from peripheral blood and compared them with macrophages and DCs derived from them by cytokine maturation following DNA damage induced by ROS. We show that monocytes are hypersensitive to ROS, undergoing excessive apoptosis. These cells exhibited a high yield of ROS-induced DNA single- and double-strand breaks and activation of the ATR-Chk1-ATM-Chk2-p53 pathway that led to Fas and caspase-8, -3, and -7 activation, whereas macrophages and DCs derived from them were protected. Monocytes are also hypersensitive to ionizing radiation and oxidized low-density lipoprotein. The remarkable sensitivity of monocytes to oxidative stress is caused by a lack of expression of the DNA repair proteins XRCC1, ligase IIIα, poly(ADP-ribose) polymerase-1, and catalytic subunit of DNA-dependent protein kinase (DNA-PK(cs)), causing a severe DNA repair defect that impacts base excision repair and double-strand break repair by nonhomologous end-joining. During maturation of monocytes into macrophages and DCs triggered by the cytokines GM-CSF and IL-4, these proteins become up-regulated, making macrophages and DCs repair-competent and ROS-resistant. We propose that impaired DNA repair in monocytes plays a role in the regulation of the monocyte/macrophage/DC system following ROS exposure.
Collapse
|
27
|
Orsó E, Grandl M, Schmitz G. Oxidized LDL-induced endolysosomal phospholipidosis and enzymatically modified LDL-induced foam cell formation determine specific lipid species modulation in human macrophages. Chem Phys Lipids 2011; 164:479-87. [DOI: 10.1016/j.chemphyslip.2011.06.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 05/31/2011] [Accepted: 06/01/2011] [Indexed: 01/01/2023]
|
28
|
Wu CY, Tang ZH, Jiang L, Li XF, Jiang ZS, Liu LS. PCSK9 siRNA inhibits HUVEC apoptosis induced by ox-LDL via Bcl/Bax-caspase9-caspase3 pathway. Mol Cell Biochem 2011; 359:347-58. [PMID: 21847580 DOI: 10.1007/s11010-011-1028-6] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 08/05/2011] [Indexed: 12/21/2022]
Abstract
This paper investigated the effects of ox-LDL on PCSK9, and the molecular mechanisms of PCSK9 siRNA-inhibited apoptosis induced by ox-LDL in human umbilical vein endothelial cells (HUVECs), to clarify the role of PCSK9 in atherosclerogenesis. HUVECs were incubated with ox-LDL for 24 h. The apoptosis was observed by Hoechst 33258 staining. The expression of PCSK9, LOX-1 mRNAs and proteins was detected by RT-PCR, western blot, respectively. The PCSK9 siRNAs labeled with fluorescence were transfected into HUVECs by Lipofectamine 2000. After transfection for 24 h, cells were treated with ox-LDL for 24 h, HUVECs apoptosis transfected siRNA was detected by Hoechst 33258 staining and flow cytometer. The expression of Bcl-2, Bax, caspase3, 8, 9 was detected by western blot. The activity of caspase3, 9 was detected by kits. Our results showed that apoptosis of HUVECs and the expressions of PCSK9 and LOX-1 were upregulated secondary to induction by ox-LDL in a concentration-dependent manner. However, ox-LDL-induced HUVEC apoptosis and PCSK9 expression, but not LOX-1 expression, were significantly reduced by PCSK9 siRNA. These results demonstrate a linkage between HUVEC apoptosis and PCSK9 expression. Furthermore, we detected the possible pathway involved in apoptotic regulation by PCSK9 siRNA; our results showed that the expression of Bcl-2 decreased, whereas that of Bax increased. In addition, ox-LDL enhanced the activity of caspase9 and then caspase3. Pretreatment of HUVECs with PCSK9 siRNA blocked these effects of ox-LDL. These findings suggest that ox-LDL-induced HUVECs apoptosis could be inhibited by PCSK9 siRNA, in which Bcl/Bax-caspase9-caspase3 pathway maybe was involved through reducing the Bcl-2/Bax ratio and inhibited the activation of both caspase9 and 3.
Collapse
Affiliation(s)
- Chun-Yan Wu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, Hunan, China
| | | | | | | | | | | |
Collapse
|
29
|
Resch U, Semlitsch M, Hammer A, Susani-Etzerodt H, Walczak H, Sattler W, Malle E. Hypochlorite-modified low-density lipoprotein induces the apoptotic machinery in Jurkat T-cell lines. Biochem Biophys Res Commun 2011; 410:895-900. [PMID: 21708126 PMCID: PMC3144388 DOI: 10.1016/j.bbrc.2011.06.089] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 06/12/2011] [Indexed: 11/01/2022]
Abstract
Myeloperoxidase is abundantly present in inflammatory diseases where activation of monocytes/macrophages and T-cell-mediated immune response occurs. The potent oxidant hypochlorous acid (HOCl), generated by the myeloperoxidase-H(2)O(2)-chloride system of activated phagocytes, converts low-density lipoprotein (LDL) into a proinflammatory lipoprotein particle. Here, we investigated the apoptotic effect of HOCl-LDL, an in vivo occurring LDL modification, on human T-cell lymphoblast-like Jurkat cells. Experiments revealed that HOCl-LDL, depending on the oxidant:lipoprotein molar ratio, induces apoptosis via activation of caspase-3, PARP cleavage and accumulation of reactive oxygen species. The absence of Fas-associated protein with death domain or caspase-8 in mutant cells did not prevent HOCl-LDL induced apoptosis. In contrast, overexpression of the anti-apoptotic Bcl-2 protein protects Jurkat cells against HOCl-LDL-induced apoptosis and prevents accumulation of reactive oxygen species. We conclude that HOCl-LDL-mediated apoptosis in Jurkat cells follows predominantly the intrinsic, mitochondrial pathway. Insitu experiments revealed that an antibody raised against HOCl-LDL recognized epitopes that colocalize both with myeloperoxidase and CD3-positive T-cells in human decidual tissue where local stimulation of the immune system occurs. We provide convincing evidence that formation of HOCl-modified (lipo)proteins generated by the myeloperoxidase-H(2)O(2)-chloride system contributes to apoptosis in T-cells.
Collapse
Affiliation(s)
- Ulrike Resch
- Institute of Molecular Biology and Biochemistry, Center for Molecular Medicine, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | |
Collapse
|
30
|
Poitz DM, Augstein A, Weinert S, Braun-Dullaeus RC, Strasser RH, Schmeisser A. OxLDL and macrophage survival: essential and oxygen-independent involvement of the Hif-pathway. Basic Res Cardiol 2011; 106:761-72. [DOI: 10.1007/s00395-011-0186-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Revised: 04/17/2011] [Accepted: 04/26/2011] [Indexed: 01/11/2023]
|
31
|
Wei YF, Zhong ZG, Huang RB, Peng Y, Xie HY, Duan XL, Zhao TJ. Effects of plumbagin on apoptosis and expression of apoptosis-related proteins in human hepatic stellate cells. Shijie Huaren Xiaohua Zazhi 2011; 19:349-354. [DOI: 10.11569/wcjd.v19.i4.349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of plumbagin on leptin-induced apoptosis and expression of apoptosis-related protein in human hepatic satellite cells (HSC-LX2) and to explore the anti-fibrotic mechanism of plumbagin.
METHODS: After HSC-LX2 cells were cultured in vitro, stimulated with leptin for 24 h, and treated with different concentrations of plumbagin for 24 h, cell apoptosis was detected by flow cytometry; cell ultrastructure was observed by transmission electron microscopy; and the protein expression of P53, Bax, and Bcl-2 was determined by immunocytochemistry.
RESULTS: HSC-LX2 cells were divided into 6 groups: untreated cells (blank control group), those treated with 100 μg/L leptin (leptin control group), those treated with both leptin and colchicin (colchicin group), those treated with both leptin and 2, 8 or 16 μmol/L plumbagin (2, 8, 16 μmol/L plumbagin group). The apoptosis rate of HSC-LX2 cells was significantly increased in plumbagin groups. The apoptosis rates of cells treated with 8 or 16 µmol/L plumbagin or colchicine were significantly higher than those of the blank control group and leptin group (5.21% ± 0.41%, 8.10% ± 0.63%, 10.1% ± 1.08% vs 1.40% ± 0.13%, 2.85% ± 0.21%, all P < 0.01). Transmission electron microscopy revealed varying degrees of apoptosis in the leptin group or plumbagin groups. Immunocytochemistry analysis showed that the protein expression levels of P53 and Bax were higher and that of Bcl-2 was lower in plumbagin groups than in the leptin group (Bax: 85.24 ± 1.08, 86.35 ± 1.12, 91.13 ± 1.13 vs 56.63 ± 0.94; P53: 25.32 ± 0.6, 38.14 ± 0.71, 41.19 ± 0.72 vs 19.25 ± 0.46; Bcl-2: 32.12 ± 0.43, 27.71 ± 0.38, 21.46 ± 0.46 vs 44.51 ± 0.56, all P < 0.05 or 0.01).
CONCLUSION: Plumbagin can significantly accelerate leptin-induced apoptosis of HSC-LX2 cells possibly by up-regulating P53 and Bax expression and down-regulating Bcl-2 expression.
Collapse
|
32
|
Nowak M, Tardivel S, Sayegrih K, Robert V, Abreu S, Chaminade P, Vicca S, Grynberg A, Lacour B. Impact of Polyunsaturated Fatty Acids on Oxidized Low Density Lipoprotein-Induced U937 Cell Apoptosis. J Atheroscler Thromb 2011; 18:494-503. [DOI: 10.5551/jat.7062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
33
|
The CD14(+/low)CD16(+) monocyte subset is more susceptible to spontaneous and oxidant-induced apoptosis than the CD14(+)CD16(-) subset. Cell Death Dis 2010; 1:e95. [PMID: 21368871 PMCID: PMC3032320 DOI: 10.1038/cddis.2010.69] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human monocytes can be classified into two subsets with distinctive characteristics. In this study, we report a difference in apoptotic potential between these two subsets with CD14+/lowCD16+ monocytes being more susceptible than CD14+CD16− monocytes to undergo spontaneous apoptosis and apoptosis induced by reactive oxygen species (ROS). By global transcriptomic and proteomic approaches, we observed that CD14+/lowCD16+ monocytes expressed higher levels of pro-apoptotic genes and proteins such as TNFα, caspase 3, Bax and cytochrome c and showed more caspases 3 and 7 activities. They also exhibited greater aerobic respiration resulting in a higher production of ROS from the mitochondria. CD14+CD16− monocytes, in contrast, showed higher expression of glutathione (GSH)-metabolizing genes such as GSH peroxidase and microsomal GSH S-transferase and were more resistant to oxidative stress than CD14+/lowCD16+ monocytes. The apoptosis of CD14+/lowCD16+ monocytes was ROS dependent as reducing ROS levels significantly reduced cell death. This is the first report of a differential apoptotic propensity of human monocyte subsets, and gaining a better understanding of this process may help to provide a better understanding of the roles of these subsets during homeostasis and under pathological conditions, particularly in situations in which high levels of oxidants are present.
Collapse
|
34
|
Wu JH, Wang YR, Huang WY, Tan RX. Anti-proliferative and pro-apoptotic effects of tectorigenin on hepatic stellate cells. World J Gastroenterol 2010; 16:3911-8. [PMID: 20712052 PMCID: PMC2923765 DOI: 10.3748/wjg.v16.i31.3911] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2010] [Revised: 04/20/2010] [Accepted: 04/27/2010] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effect of tectorigenin on proliferation and apoptosis of hepatic stellate cells (HSC)-T6 cells. METHODS HSC-T6 cells were incubated with tectorigenin at different concentrations, and their proliferation was assessed by bromodeoxyuridine incorporation assay. Apoptosis was detected by flow cytometry assay with Hoechst 33342 staining. Also, generation of reactive oxygen species (ROS), intracellular [Ca(2+)](i), potential of mitochondrial membrane, activities of cytochrome c and caspase-9 and -3 were investigated to explore a conceivable apoptotic pathway. RESULTS Tectorigenin suppressed the proliferation of HSC-T6 cells and induced apoptosis of HSC-T6 cells in a time- and dose-dependent manner. Tectorigenin at the concentration of 100 microg/mL greatly inhibited the viability of HSC-T6 cells and induced the condensation of chromatin and fragmentation of nuclei. When treated for 48 h, the percentage of cell growth and apoptosis reached 46.3% +/- 2.37% (P = 0.004) and 50.67% +/- 3.24% (P = 0.003), respectively. Furthermore, tectorigenin-induced apoptosis of HSC-T6 cells was associated with the generation of ROS, increased intracellular [Ca(2+)](i), loss of mitochondrial membrane potential, translocation of cytochrome c, and activation of caspase-9 and -3. CONCLUSION Tectorigenin inhibits proliferation of HSC-T6 cells and induces apoptosis of HSC-T6 cells.
Collapse
|
35
|
Bachar AR, Scheffer L, Schroeder AS, Nakamura HK, Cobb LJ, Oh YK, Lerman LO, Pagano RE, Cohen P, Lerman A. Humanin is expressed in human vascular walls and has a cytoprotective effect against oxidized LDL-induced oxidative stress. Cardiovasc Res 2010; 88:360-6. [PMID: 20562421 DOI: 10.1093/cvr/cvq191] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Humanin (HN) is a 24-amino acid peptide that has been shown to have an anti-apoptotic function against neuronal cell death caused by Alzheimer's disease. Increased oxidative stress, one of the major factors contributing to this cell death, also plays an important role in the inflammatory process of atherosclerosis. The current study was designed to test the hypothesis that HN is expressed in the human vascular wall and may protect against oxidative stress. METHODS AND RESULTS HN expression in the vascular wall was detected by immunostaining in the endothelial cell layer of human internal mammary arteries (n = 5), atherosclerotic coronary arteries (n = 17), and sections of the greater saphenous vein (n = 3). HN mRNA was expressed in the human aortic endothelial cells (HAECs). Cytoprotective effects of HN against oxidative stress were tested in vitro in HAECs. Pre-treatment with 0.1 µM HN reduced oxidized LDL (Ox-LDL)-induced (i) formation of reactive oxygen species by 50%, (ii) apoptosis by ∼50% as determined by TUNEL staining, and (iii) formation of ceramide, a lipid second messenger involved in the apoptosis signalling cascade, by ∼20%. CONCLUSION The current study demonstrates for the first time the expression of HN in the endothelial cell layer of human blood vessels. Exogenous addition of HN to endothelial cell cultures was shown to be effective against Ox-LDL-induced apoptosis. These findings suggest that HN may play a role and may have a protective effect in early atherosclerosis in humans.
Collapse
Affiliation(s)
- Adi R Bachar
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Quercetin supplementation and its effect on human monocyte gene expression profiles in vivo. Br J Nutr 2010; 104:336-45. [DOI: 10.1017/s0007114510000711] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Quercetin has been described as having a wide range of beneficial effects in humans, ranging from anti-carcinogenic properties to reducing the risk of CVD. Nevertheless, underlying molecular mechanisms have been mostly investigated in vitro. Here, we tested whether a daily supplementation of quercetin leads to reproducible changes in human monocyte gene expression profiles. In study I, quercetin in varying dosages was given to healthy subjects for 2 weeks. RNA from monocytes isolated at the beginning and end of the study from subjects receiving 150 mg quercetin per d was subjected to transcriptome-wide microarray analysis. In study II, a double-blind cross-over study, twenty subjects exhibiting a ‘cardiovascular risk phenotype’ received 150 mg quercetin or placebo daily for 6 weeks each and served as the verification group. Microarray analysis revealed a number of differentially expressed genes. The most significantly represented functional groups were those of the immune system, nucleic acid metabolism, apoptosis and O-glycan biosynthesis. Twenty-four genes were chosen for technical replication and independent verification by quantitative real-time PCR. When comparing placebo and quercetin treatment, four genes showed significantly different expression changes (C1GALT1, O-glycan biosynthesis; GM2A, glycolipid catabolism; HDGF, cell proliferation; SERPINB9, apoptosis). However, these were minimal in respect to magnitude of fold change. In conclusion, although microarray analysis revealed extensive effects of quercetin on gene expression, the employment of a placebo-controlled study design showed no comparable results for twenty-four verification targets. This emphasises the need for stringent designs in nutritional intervention studies with the aim to identify relevant changes in gene expression.
Collapse
|
37
|
Ermak N, Lacour B, Goirand F, Drüeke TB, Vicca S. Differential apoptotic pathways activated in response to Cu-induced or HOCl-induced LDL oxidation in U937 monocytic cell line. Biochem Biophys Res Commun 2010; 393:783-7. [DOI: 10.1016/j.bbrc.2010.02.082] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 02/12/2010] [Indexed: 10/19/2022]
|
38
|
Zhang D, Bi Z, Li Y, Zheng H, Li L, Ouyang J, Wang B, Bi Y. Sodium Ferulate Modified Gene Expression Profile of Oxidized Low-Density Lipoprotein-Stimulated Human Umbilical Vein Endothelial Cells. J Cardiovasc Pharmacol Ther 2009; 14:302-13. [PMID: 19837969 DOI: 10.1177/1074248409347986] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Oxidized low-density lipoprotein (ox-LDL) is known to trigger vascular injury in atherosclerosis development. Sodium ferulate is an effective component from Chinese medicines with various beneficial cardiovascular pharmacological activities. Here, we investigated the effects of sodium ferulate on the gene expression profile of ox-LDL-stimulated endothelial cells. Cultured human umbilical vein endothelial cells (HUVECs) were treated with ox-LDL (50 μg/mL) in the absence or presence of sodium ferulate (5 μmol/L). Sodium ferulate significantly reduced ox-LDL-induced endothelial cell death as evaluated by cell viability assay. Human oligonucleotide microarray analysis demonstrated that a total of 32 ox-LDL-induced genes were significantly downregulated to control levels by sodium ferulate. These genes included members from families of chemokine, inflammatory factor, growth factor, and nuclear receptor. These data provided an overview of the gene expression profile of endothelial cells in response to ox-LDL and sodium ferulate, and demonstrated that sodium ferulate could regulate the expression of inflammation-related genes in endothelial cells and has the potential to benefit endothelial function in the setting of atherosclerosis.
Collapse
Affiliation(s)
- Dongxian Zhang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, PR China, Nanyang Medical College, Henan, PR China
| | - Zhuoyue Bi
- School of Pharmacy, Wuhan University, Wuhan, PR China
| | - Yang Li
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, PR China
| | - Hong Zheng
- School of Basic Medical Science, Yunyang Medical College, Hubei, PR China
| | - Li Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, PR China
| | - Jingping Ouyang
- Department of Pathology and Pathophysiology, School of Medicine, Wuhan University, Wuhan, PR China
| | - Baohua Wang
- Department of Pathology and Pathophysiology, School of Medicine, Wuhan University, Wuhan, PR China
| | - Yongyi Bi
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, PR China
| |
Collapse
|
39
|
Diabetes mellitus and apoptosis: inflammatory cells. Apoptosis 2009; 14:1435-50. [PMID: 19360474 DOI: 10.1007/s10495-009-0340-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 03/17/2009] [Indexed: 12/24/2022]
Abstract
Since the early observation that similarities between thyroiditis and insulitis existed, the important role played by inflammation in the development of diabetes has been appreciated. More recently, experiments have shown that inflammation also plays a prominent role in the development of target organ damage arising as complications, with both elements of the innate and the adaptive immune system being involved, and that cytokines contributing to local tissue damage may arise from both infiltrating and resident cells. This review will discuss the experimental evidence that shows that inflammatory cell-mediated apoptosis contributes to target organ damage, from beta cell destruction to both micro- and macro-vascular disease complications, and also how alterations in leukocyte turnover affects immune function.
Collapse
|
40
|
|
41
|
Scazzocchio B, Varì R, D'Archivio M, Santangelo C, Filesi C, Giovannini C, Masella R. Oxidized LDL impair adipocyte response to insulin by activating serine/threonine kinases. J Lipid Res 2009; 50:832-45. [PMID: 19136667 DOI: 10.1194/jlr.m800402-jlr200] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oxidized LDL (oxLDL) increase in patients affected by type-2 diabetes, obesity, and metabolic syndrome. Likewise, insulin resistance, an impaired responsiveness of target tissues to insulin, is associated with those pathological conditions. To investigate a possible causal relationship between oxLDL and the onset of insulin resistance, we evaluated the response to insulin of 3T3-L1 adipocytes treated with oxLDL. We observed that oxLDL inhibited glucose uptake (-40%) through reduced glucose transporter 4 (GLUT4) recruitment to the plasma membrane (-70%), without affecting GLUT4 gene expression. These findings were associated to the impairment of insulin signaling. Specifically, in oxLDL-treated cells insulin receptor (IR) substrate-1 (IRS-1) was highly degraded likely because of the enhanced Ser(307)phosphorylation. This process was largely mediated by the activation of the inhibitor of kappaB-kinase beta (IKKbeta) and the c-Jun NH(2)-terminal kinase (JNK). Moreover, the activation of IKKbeta positively regulated the nuclear content of nuclear factor kappaB (NF-kappaB), by inactivating the inhibitor of NF-kappaB (IkappaBalpha). The activated NF-kappaB further impaired per se GLUT4 functionality. Specific inhibitors of IKKbeta, JNK, and NF-kappaB restored insulin sensitivity in adipocytes treated with oxLDL. These data provide the first evidence that oxLDL, by activating serine/threonine kinases, impaired adipocyte response to insulin affecting pathways involved in the recruitment of GLUT4 to plasma membranes (PM). This suggests that oxLDL might participate in the development of insulin resistance.
Collapse
Affiliation(s)
- Beatrice Scazzocchio
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Effects of increasing docosahexaenoic acid intake in human healthy volunteers on lymphocyte activation and monocyte apoptosis. Br J Nutr 2008; 101:852-8. [PMID: 18710607 DOI: 10.1017/s0007114508051520] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dietary intake of long-chain n-3 PUFA has been reported to decrease several markers of lymphocyte activation and modulate monocyte susceptibility to apoptosis. However, most human studies examined the combined effect of DHA and EPA using relatively high daily amounts of n-3 PUFA. The present study investigated the effects of increasing doses of DHA added to the regular diet of human healthy volunteers on lymphocyte response to tetradecanoylphorbol acetate plus ionomycin activation, and on monocyte apoptosis induced by oxidized LDL. Eight subjects were supplemented with increasing daily doses of DHA (200, 400, 800, 1600 mg) in a TAG form containing DHA as the only PUFA, for 2 weeks each dose. DHA intake dose-dependently increased the proportion of DHA in mononuclear cell phospholipids, the augmentation being significant after 400 mg DHA/d. The tetradecanoylphorbol acetate plus ionomycin-stimulated IL-2 mRNA level started to increase after ingestion of 400 mg DHA/d, with a maximum after 800 mg intake, and was positively correlated (P < 0.003) with DHA enrichment in cell phospholipids. The treatment of monocytes by oxidized LDL before DHA supplementation drastically reduced mitochondrial membrane potential as compared with native LDL treatment. Oxidized LDL apoptotic effect was significantly attenuated after 400 mg DHA/d and the protective effect was maintained throughout the experiment, although to a lesser extent at higher doses. The present results show that supplementation of the human diet with low DHA dosages improves lymphocyte activability. It also increases monocyte resistance to oxidized LDL-induced apoptosis, which may be beneficial in the prevention of atherosclerosis.
Collapse
|