1
|
Harber KJ, Neele AE, van Roomen CP, Gijbels MJ, Beckers L, Toom MD, Schomakers BV, Heister DA, Willemsen L, Griffith GR, de Goede KE, van Dierendonck XA, Reiche ME, Poli A, L-H Mogensen F, Michelucci A, Verberk SG, de Vries H, van Weeghel M, Van den Bossche J, de Winther MP. Targeting the ACOD1-itaconate axis stabilizes atherosclerotic plaques. Redox Biol 2024; 70:103054. [PMID: 38309122 PMCID: PMC10848031 DOI: 10.1016/j.redox.2024.103054] [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: 10/30/2023] [Revised: 01/16/2024] [Accepted: 01/20/2024] [Indexed: 02/05/2024] Open
Abstract
Inflammatory macrophages are key drivers of atherosclerosis that can induce rupture-prone vulnerable plaques. Skewing the plaque macrophage population towards a more protective phenotype and reducing the occurrence of clinical events is thought to be a promising method of treating atherosclerotic patients. In the current study, we investigate the immunomodulatory properties of itaconate, an immunometabolite derived from the TCA cycle intermediate cis-aconitate and synthesised by the enzyme Aconitate Decarboxylase 1 (ACOD1, also known as IRG1), in the context of atherosclerosis. Ldlr-/- atherogenic mice transplanted with Acod1-/- bone marrow displayed a more stable plaque phenotype with smaller necrotic cores and showed increased recruitment of monocytes to the vessel intima. Macrophages from Acod1-/- mice contained more lipids whilst also displaying reduced induction of apoptosis. Using multi-omics approaches, we identify a metabolic shift towards purine metabolism, in addition to an altered glycolytic flux towards production of glycerol for triglyceride synthesis. Overall, our data highlight the potential of therapeutically blocking ACOD1 with the aim of stabilizing atherosclerotic plaques.
Collapse
Affiliation(s)
- Karl J Harber
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands
| | - Annette E Neele
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands
| | - Cindy Paa van Roomen
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Marion Jj Gijbels
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Department of Pathology, CARIM, Cardiovascular Research Institute Maastricht, GROW-School for Oncology and Developmental Biology, Maastricht UMC, University of Maastricht, 6229 HX, Maastricht, the Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Bauke V Schomakers
- Department of Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Daan Af Heister
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands
| | - Lisa Willemsen
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands
| | - Guillermo R Griffith
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Kyra E de Goede
- Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, the Netherlands
| | - Xanthe Amh van Dierendonck
- Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, the Netherlands
| | - Myrthe E Reiche
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Department of Medical Cell Biology, Uppsala University, 75236, Uppsala, Sweden
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Frida L-H Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Sanne Gs Verberk
- Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands
| | - Helga de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Michel van Weeghel
- Department of Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Jan Van den Bossche
- Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, the Netherlands.
| | - Menno Pj de Winther
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands.
| |
Collapse
|
2
|
Al-Hawary SIS, Jasim SA, Romero-Parra RM, Bustani GS, Hjazi A, Alghamdi MI, Kareem AK, Alwaily ER, Zabibah RS, Gupta J, Mahmoudi R, Hosseini-Fard S. NLRP3 inflammasome pathway in atherosclerosis: Focusing on the therapeutic potential of non-coding RNAs. Pathol Res Pract 2023; 246:154490. [PMID: 37141699 DOI: 10.1016/j.prp.2023.154490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome pathway has a critical role in the pathogenesis of atherosclerosis. Activation of this pathway is implicated in the subendothelial inflammation and atherosclerosis progression. The NLRP3 inflammasome are cytoplasmic sensors with the distinct capacity to identify a wide range of inflammation-related signals, which enhance NLRP3 inflammasome assembly and allow it to trigger inflammation. This pathway is triggered by a variety of intrinsic signals which exist in atherosclerotic plaques, like cholesterol crystals and oxidized LDL. Further pharmacological findings indicated that NLRP3 inflammasome enhanced caspase-1-mediated secretion of pro-inflammatory mediators like interleukin (IL)- 1β/18. Newly published cutting-edge studies suggested that non-coding RNAs (ncRNAs) including microRNAs (miRNAs, miRs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs) are major modulators of NLRP3 inflammasome in atherosclerosis. Therefore, in this review, we aimed to discuss the NLRP3 inflammasome pathway, biogenesis of ncRNAs as well as the modulatory role of ncRNAs in regulating the various mediators of NLRP3 inflammasome pathway including TLR4, NF-kB, NLRP3, and caspase 1. We also discussed the importance of NLRP3 inflammasome pathway-related ncRNAs as a diagnostic biomarker in atherosclerosis and current therapeutics in the modulation of NLRP3 inflammasome in atherosclerosis. Finally, we speak about the limitations and future prospects of ncRNAs in regulating inflammatory atherosclerosis via the NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Al-anbar-Ramadi, Iraq
| | | | | | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Kingdom of Saudi Arabia
| | | | - Ali Kamil Kareem
- Biomedical Engineering Department, Al-Mustaqbal University College, Hillah 51001, Iraq
| | - Enas R Alwaily
- Microbiology Research Group, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India
| | - Reza Mahmoudi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyedreza Hosseini-Fard
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Kazemian S, Ahmadi R, Ferns GA, Rafiei A, Azadegan-Dehkordi F, Khaledifar A, Mohammad-Rezaei M, Bagheri N. Correlation of miR-24-3p and miR-595 expression with CCL3, CCL4, IL1-beta, TNFalphaIP3, and NF-kappaBIalpha genes in PBMCs of patients with coronary artery disease. EXCLI JOURNAL 2022; 21:1184-1195. [PMID: 36381642 PMCID: PMC9650696 DOI: 10.17179/excli2022-5266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/05/2022] [Indexed: 01/25/2023]
Abstract
Inflammation has been well recognized to play an important role in developing coronary artery disease (CAD). By regulating essential genes in this pathway post-transcriptionally, MicroRNAs (miRNAs) may help or hinder the development of atherosclerotic lesions. The aim of this study was to investigate the expression of miR-24-3p, miR-595, CCL3, CCL4, IL-1β, TNFαIP3, and NF-κBIα in the peripheral blood mononuclear cells (PBMCs) of CAD and control groups and to examine whether any correlation exists between the expression of miRs and genes in CAD group. A total of 168 subjects (84 CAD subjects and 84 control subjects) were examined in this research. Expression levels of miR-24-3p, miR-595, CCL3, CCL4, IL-1β, TNFαIP3, and NF-κBIα in PBMCs were measured using the real-time PCR technique. A comparison of the CAD group with the control group indicated significantly increased expression levels of CCL3, CCL4, and IL-1β (Fold Change (FC)=4, P=0.009; FC=2.9, P=0.01; FC=1.8, P=0.019, respectively) and remarkably reduced expression levels of TNFαIP3 and NF-κBIα (FC=-1.4, P=0.03 and FC=-5.9, P=0.001, respectively). Moreover, the expression levels of miR-24-3p downregulated (FC=-2.5, P=0.005) and miR-595 upregulated (FC=1.9, P=0.009) in the CAD group. There was a statistical correlation between the number of clogged arteries with expression levels of miR-24-3p, miR-595, CCL3, CCL4, IL-1β, TNFαIP3, and NF-κBIα in the CAD group. Also, there was a statistical correlation between expression levels of miR-24-3p and miR-595 with CCL3, CCL4, IL-1β, TNFαIP3, and NF-κBIα gene expression in the CAD group. In CAD patients, decreased expression of miR-24-3p and increased expression of miR-595 may aid the progression of atherosclerotic plaques by regulating CCL3, CCL4, IL-1β, TNFαIP3, and NF-κBIα gene expression.
Collapse
Affiliation(s)
- Shakiba Kazemian
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Ali Rafiei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Azadegan-Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Arsalan Khaledifar
- Department of Cardiology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mina Mohammad-Rezaei
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nader Bagheri
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran,*To whom correspondence should be addressed: Nader Bagheri, Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran; Tel: +989181731073, Fax: +98-3813330709, E-mail:
| |
Collapse
|
4
|
Meng Z, Hernandez R, Liu J, Gwag T, Lu W, Hsiai TK, Kaul M, Zhou T, Zhou C. HIV Protein Tat Induces Macrophage Dysfunction and Atherosclerosis Development in Low-Density Lipoprotein Receptor-Deficient Mice. Cardiovasc Drugs Ther 2022; 36:201-215. [PMID: 33459922 PMCID: PMC8286272 DOI: 10.1007/s10557-021-07141-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE HIV infection is consistently associated with an increased risk of atherosclerotic cardiovascular disease, but the underlying mechanisms remain elusive. HIV protein Tat, a transcriptional activator of HIV, has been shown to activate NF-κB signaling and promote inflammation in vitro. However, the atherogenic effects of HIV Tat have not been investigated in vivo. Macrophages are one of the major cell types involved in the initiation and progression of atherosclerosis. We and others have previously revealed the important role of IκB kinase β (IKKβ), a central inflammatory coordinator through activating NF-κB, in the regulation of macrophage functions and atherogenesis. This study investigated the impact of HIV Tat exposure on macrophage functions and atherogenesis. METHODS To investigate the effects of Tat on macrophage IKKβ activation and atherosclerosis development in vivo, myeloid-specific IKKβ-deficient LDLR-deficient (IKKβΔMyeLDLR-/-) mice and their control littermates (IKKβF/FLDLR-/-) were exposed to recombinant HIV protein Tat. RESULTS Exposure to Tat significantly increased atherosclerotic lesion size and plaque vulnerability in IKKβF/FLDLR-/- but not IKKβΔMyeLDLR-/- mice. Deficiency of myeloid IKKβ attenuated Tat-elicited macrophage inflammatory responses and atherosclerotic lesional inflammation in IKKβΔMyeLDLR-/- mice. Further, RNAseq analysis demonstrated that HIV protein Tat affects the expression of many atherosclerosis-related genes in vitro in an IKKβ-dependent manner. CONCLUSIONS Our findings reveal atherogenic effects of HIV protein Tat in vivo and demonstrate a pivotal role of myeloid IKKβ in Tat-driven atherogenesis.
Collapse
Affiliation(s)
- Zhaojie Meng
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jingwei Liu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Weiwei Lu
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Tzung K Hsiai
- Departments of Medicine and Bioengineering, David Geffen School of Medicine, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA, USA
| | - Marcus Kaul
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA.
| |
Collapse
|
5
|
Hernandez R, Zhou C. Recent Advances in Understanding the Role of IKKβ in Cardiometabolic Diseases. Front Cardiovasc Med 2021; 8:752337. [PMID: 34957242 PMCID: PMC8692734 DOI: 10.3389/fcvm.2021.752337] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/12/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiometabolic diseases, including cardiovascular disease, obesity, and diabetes, are the leading cause of mortality and morbidity worldwide. Cardiometabolic diseases are associated with many overlapping metabolic syndromes such as hypertension, hyperlipidemia, insulin resistance, and central adiposity. However, the underlying causes of cardiometabolic diseases and associated syndromes remain poorly understood. Within the past couple of decades, considerable progresses have been made to understand the role of inflammatory signaling in the pathogenesis of cardiometabolic diseases. The transcription factor, NF-κB, a master regulator of the innate and adaptive immune responses, is highly active in cardiometabolic diseases. IκB kinase β (IKKβ), the predominant catalytic subunit of the IKK complex, is required for canonical activation of NF-κB, and has been implicated as the critical molecular link between inflammation and cardiometabolic diseases. Recent studies have revealed that IKKβ has diverse and unexpected roles in mediating adiposity, insulin sensitivity, glucose homeostasis, vascular function, and atherogenesis through complex mechanisms. IKKβ has been demonstrated as a critical player in the development of cardiometabolic diseases and is implicated as a promising therapeutic target. This review summarizes current knowledge of the functions of IKKβ in mediating the development and progression of cardiometabolic diseases.
Collapse
Affiliation(s)
- Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | | |
Collapse
|
6
|
Zhang T, Ma C, Zhang Z, Zhang H, Hu H. NF-κB signaling in inflammation and cancer. MedComm (Beijing) 2021; 2:618-653. [PMID: 34977871 PMCID: PMC8706767 DOI: 10.1002/mco2.104] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Since nuclear factor of κ-light chain of enhancer-activated B cells (NF-κB) was discovered in 1986, extraordinary efforts have been made to understand the function and regulating mechanism of NF-κB for 35 years, which lead to significant progress. Meanwhile, the molecular mechanisms regulating NF-κB activation have also been illuminated, the cascades of signaling events leading to NF-κB activity and key components of the NF-κB pathway are also identified. It has been suggested NF-κB plays an important role in human diseases, especially inflammation-related diseases. These studies make the NF-κB an attractive target for disease treatment. This review aims to summarize the knowledge of the family members of NF-κB, as well as the basic mechanisms of NF-κB signaling pathway activation. We will also review the effects of dysregulated NF-κB on inflammation, tumorigenesis, and tumor microenvironment. The progression of the translational study and drug development targeting NF-κB for inflammatory diseases and cancer treatment and the potential obstacles will be discussed. Further investigations on the precise functions of NF-κB in the physiological and pathological settings and underlying mechanisms are in the urgent need to develop drugs targeting NF-κB for inflammatory diseases and cancer treatment, with minimal side effects.
Collapse
Affiliation(s)
- Tao Zhang
- Cancer Center and Center for Immunology and HematologyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Chao Ma
- Cancer Center and Center for Immunology and HematologyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Zhiqiang Zhang
- Immunobiology and Transplant Science CenterHouston Methodist HospitalHoustonTexasUSA
| | - Huiyuan Zhang
- Cancer Center and Center for Immunology and HematologyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Hongbo Hu
- Cancer Center and Center for Immunology and HematologyWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
7
|
Lu W, Park SH, Meng Z, Wang F, Zhou C. Deficiency of Adipocyte IKKβ Affects Atherosclerotic Plaque Vulnerability in Obese LDLR Deficient Mice. J Am Heart Assoc 2019; 8:e012009. [PMID: 31203708 PMCID: PMC6645619 DOI: 10.1161/jaha.119.012009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Obesity‐associated chronic inflammation has been known to contribute to atherosclerosis development, but the underlying mechanisms remain elusive. Recent studies have revealed novel functions of IKKβ (inhibitor of NF‐κB [nuclear factor κB] kinase β), a key coordinator of inflammation through activation of NF‐κB, in atherosclerosis and adipose tissue development. However, it is not clear whether IKKβ signaling in adipocytes can also affect atherogenesis. This study aims to investigate the impact of adipocyte IKKβ expression on atherosclerosis development in lean and obese LDLR (low‐density lipoprotein receptor)–deficient (LDLR−/−) mice. Methods and Results To define the role of adipocyte IKKβ in atherogenesis, we generated adipocyte‐specific IKKβ‐deficient LDLR−/− (IKKβΔAdLDLR−/−) mice. Targeted deletion of IKKβ in adipocytes did not affect adiposity and atherosclerosis in lean LDLR−/− mice when fed a low‐fat diet. In response to high‐fat feeding, however, IKKβΔAdLDLR−/− mice had defective adipose remodeling and increased adipose tissue and systemic inflammation. Deficiency of adipocyte IKKβ did not affect atherosclerotic lesion sizes but resulted in enhanced lesional inflammation and increased plaque vulnerability in obese IKKβΔAdLDLR−/− mice. Conclusions These data demonstrate that adipocyte IKKβ signaling affects the evolution of atherosclerosis plaque vulnerability in obese LDLR−/− mice. This study suggests that the functions of IKKβ signaling in atherogenesis are complex, and IKKβ in different cell types or tissues may have different effects on atherosclerosis development.
Collapse
Affiliation(s)
- Weiwei Lu
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY
| | - Se-Hyung Park
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY
| | - Zhaojie Meng
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY
| | - Fang Wang
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY
| | - Changcheng Zhou
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY.,2 Saha Cardiovascular Research Center University of Kentucky Lexington KY
| |
Collapse
|
8
|
Yang C, Lu M, Chen W, He Z, Hou X, Feng M, Zhang H, Bo T, Zhou X, Yu Y, Zhang H, Zhao M, Wang L, Yu C, Gao L, Jiang W, Zhang Q, Zhao J. Thyrotropin aggravates atherosclerosis by promoting macrophage inflammation in plaques. J Exp Med 2019; 216:1182-1198. [PMID: 30940720 PMCID: PMC6504213 DOI: 10.1084/jem.20181473] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/07/2019] [Accepted: 02/11/2019] [Indexed: 12/31/2022] Open
Abstract
The increased cardiovascular risk in subclinical hypothyroidism has traditionally been attributed to the associated metabolic disorders. This paper, however, revealed that TSH can aggravate atherosclerosis by promoting macrophage inflammation in the plaque, which deepens our understanding of the significance of TSH elevation in subclinical hypothyroidism. Subclinical hypothyroidism is associated with cardiovascular diseases, yet the underlying mechanism remains largely unknown. Herein, in a common population (n = 1,103), TSH level was found to be independently correlated with both carotid plaque prevalence and intima-media thickness. Consistently, TSH receptor ablation in ApoE−/− mice attenuated atherogenesis, accompanied by decreased vascular inflammation and macrophage burden in atherosclerotic plaques. These results were also observed in myeloid-specific Tshr-deficient ApoE−/− mice, which indicated macrophages to be a critical target of the proinflammatory and atherogenic effects of TSH. In vitro experiments further revealed that TSH activated MAPKs (ERK1/2, p38α, and JNK) and IκB/p65 pathways in macrophages and increased inflammatory cytokine production and their recruitment of monocytes. Thus, the present study has elucidated the new mechanisms by which TSH, as an independent risk factor of atherosclerosis, aggravates vascular inflammation and contributes to atherogenesis.
Collapse
Affiliation(s)
- Chongbo Yang
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Ming Lu
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Wenbin Chen
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Zhao He
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China.,School of Medicine, Shandong University, Jinan, Shandong, China
| | - Xu Hou
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Mei Feng
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Laboratory for Cardiovascular Precision Medicine, Beijing, China
| | - Tao Bo
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Xiaoming Zhou
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Yong Yu
- Department of Sonography, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Haiqing Zhang
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Meng Zhao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Laicheng Wang
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Chunxiao Yu
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Ling Gao
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Laboratory for Cardiovascular Precision Medicine, Beijing, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Ministry of Public Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| |
Collapse
|
9
|
Mussbacher M, Salzmann M, Brostjan C, Hoesel B, Schoergenhofer C, Datler H, Hohensinner P, Basílio J, Petzelbauer P, Assinger A, Schmid JA. Cell Type-Specific Roles of NF-κB Linking Inflammation and Thrombosis. Front Immunol 2019; 10:85. [PMID: 30778349 PMCID: PMC6369217 DOI: 10.3389/fimmu.2019.00085] [Citation(s) in RCA: 375] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/11/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor NF-κB is a central mediator of inflammation with multiple links to thrombotic processes. In this review, we focus on the role of NF-κB signaling in cell types within the vasculature and the circulation that are involved in thrombo-inflammatory processes. All these cells express NF-κB, which mediates important functions in cellular interactions, cell survival and differentiation, as well as expression of cytokines, chemokines, and coagulation factors. Even platelets, as anucleated cells, contain NF-κB family members and their corresponding signaling molecules, which are involved in platelet activation, as well as secondary feedback circuits. The response of endothelial cells to inflammation and NF-κB activation is characterized by the induction of adhesion molecules promoting binding and transmigration of leukocytes, while simultaneously increasing their thrombogenic potential. Paracrine signaling from endothelial cells activates NF-κB in vascular smooth muscle cells and causes a phenotypic switch to a “synthetic” state associated with a decrease in contractile proteins. Monocytes react to inflammatory situations with enforced expression of tissue factor and after differentiation to macrophages with altered polarization. Neutrophils respond with an extension of their life span—and upon full activation they can expel their DNA thereby forming so-called neutrophil extracellular traps (NETs), which exert antibacterial functions, but also induce a strong coagulatory response. This may cause formation of microthrombi that are important for the immobilization of pathogens, a process designated as immunothrombosis. However, deregulation of the complex cellular links between inflammation and thrombosis by unrestrained NET formation or the loss of the endothelial layer due to mechanical rupture or erosion can result in rapid activation and aggregation of platelets and the manifestation of thrombo-inflammatory diseases. Sepsis is an important example of such a disorder caused by a dysregulated host response to infection finally leading to severe coagulopathies. NF-κB is critically involved in these pathophysiological processes as it induces both inflammatory and thrombotic responses.
Collapse
Affiliation(s)
- Marion Mussbacher
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Manuel Salzmann
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Christine Brostjan
- Department of Surgery, General Hospital, Medical University of Vienna, Vienna, Austria
| | - Bastian Hoesel
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | | | - Hannes Datler
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Philipp Hohensinner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - José Basílio
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelial Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Johannes A Schmid
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
The CD44-HA axis and inflammation in atherosclerosis: A temporal perspective. Matrix Biol 2018; 78-79:201-218. [PMID: 29792915 DOI: 10.1016/j.matbio.2018.05.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/17/2018] [Accepted: 05/19/2018] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease (CVD) due to atherosclerosis is a disease of chronic inflammation at both the systemic and the tissue level. CD44 has previously been implicated in atherosclerosis in both humans and mice. This multi-faceted receptor plays a critical part in the inflammatory response during the onset of CVD, though little is known of CD44's role during the latter stages of the disease. This review focuses on the role of CD44-dependent HA-dependent effects on inflammatory cells in several key processes, from disease initiation throughout the progression of atherosclerosis. Understanding how CD44 and HA regulate inflammation in atherogenesis is key in determining the utility of the CD44-HA axis as a therapeutic target to halt disease and potentially promote disease regression.
Collapse
|
11
|
Song D, Fang G, Mao SZ, Ye X, Liu G, Miller EJ, Greenberg H, Liu SF. Selective inhibition of endothelial NF-κB signaling attenuates chronic intermittent hypoxia-induced atherosclerosis in mice. Atherosclerosis 2018; 270:68-75. [PMID: 29407890 DOI: 10.1016/j.atherosclerosis.2018.01.027] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 01/07/2018] [Accepted: 01/18/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Chronic intermittent hypoxia (CIH) exposure causes atherosclerosis, although the underlying mechanisms are poorly understood. This study defines the role of endothelial intrinsic NF-κB signaling in the atherogenic response to CIH. METHODS We created ApoE-ECI-κBmt mice that are deficient in the apolipoprotein E gene (ApoE-/-) and overexpress an I-κBα mutant (I-κBmt) selectively in endothelial cells. ApoE-/- and ApoE-ECI-κBmt mice were fed a normal chow diet (NCD) or high cholesterol diet (HCD) and exposed to sham or CIH, and atherosclerotic lesions were quantified. RESULTS CIH exposure activated NF-κB in aortas, and induced the expression of endothelial-specific and NF-κB-dependent genes, E-selectin and vascular cell adhesion molecule (VCAM)-1, in the aortas and hearts. Endothelial I-κBmt overexpression in ApoE-ECI-κBmt mice significantly inhibited CIH-induced NF-κB activity, and suppressed E-selectin and VCAM-1 expressions, confirming endothelial NF-κB inhibition in ApoE-ECI-κBmt mice. ApoE-/- mice, on NCD, developed mild atherosclerotic lesions spontaneously, and developed advanced and larger areas of atherosclerotic plaques when exposed to CIH. ApoE-/- mice also developed advanced atherosclerotic lesions when fed an HCD alone. The HCD-induced atherosclerotic plaques became more advanced, and plaque area was doubled in mice exposed to HCD + CIH. Endothelial I-κBmt overexpression in ApoE-ECI-κBmt mice attenuated spontaneously developed atherosclerotic lesions, abrogated CIH-induced atherosclerosis and mitigated CIH-mediated facilitation of HCD-induced atherosclerosis. CONCLUSIONS These results suggest that endothelial intrinsic NF-kB signaling may play a pivotal role in CIH-induced atherosclerosis.
Collapse
Affiliation(s)
- Dongmei Song
- The First Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Center for Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| | - Guoqiang Fang
- Center for Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Sun-Zhong Mao
- Center for Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Xiaobing Ye
- Center for Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Gang Liu
- Center for Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Edmund J Miller
- Center for Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Harly Greenberg
- Center for Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Shu Fang Liu
- The First Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Center for Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| |
Collapse
|
12
|
Abstract
The transcription factor NF-κB regulates multiple aspects of innate and adaptive immune functions and serves as a pivotal mediator of inflammatory responses. NF-κB induces the expression of various pro-inflammatory genes, including those encoding cytokines and chemokines, and also participates in inflammasome regulation. In addition, NF-κB plays a critical role in regulating the survival, activation and differentiation of innate immune cells and inflammatory T cells. Consequently, deregulated NF-κB activation contributes to the pathogenic processes of various inflammatory diseases. In this review, we will discuss the activation and function of NF-κB in association with inflammatory diseases and highlight the development of therapeutic strategies based on NF-κB inhibition.
Collapse
|
13
|
Jeurissen MLJ, Walenbergh SMA, Houben T, Gijbels MJJ, Li J, Hendrikx T, Oligschlaeger Y, van Gorp PJ, Binder CJ, Donners MMPC, Shiri-Sverdlov R. Prevention of oxLDL uptake leads to decreased atherosclerosis in hematopoietic NPC1-deficient Ldlr -/- mice. Atherosclerosis 2016; 255:59-65. [PMID: 27816810 DOI: 10.1016/j.atherosclerosis.2016.10.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/03/2016] [Accepted: 10/19/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis is a chronic inflammatory disease of medium and large vessels and is typically characterized by the predominant accumulation of low-density lipoprotein (LDL)-cholesterol inside macrophages that reside in the vessel walls. Previous studies clearly demonstrated an association specifically between the oxidized type of LDL (oxLDL) and atherosclerotic lesion formation. Further observations revealed that these atherosclerotic lesions displayed enlarged, lipid-loaded lysosomes. By increasing natural antibodies against oxLDL, pneumococcal vaccination has been shown to reduce atherosclerosis in LDL receptor knockout (Ldlr-/-) mice. Relevantly, loss of the lysosomal membrane protein Niemann-Pick Type C1 (NPC1) led to lysosomal accumulation of various lipids and promoted atherosclerosis. Yet, the importance of lysosomal oxLDL accumulation inside macrophages, compared to non-modified LDL, in atherosclerosis has never been established. METHODS By transplanting NPC1 bone marrow into lethally irradiated Ldlr-/- mice, a hematopoietic mouse model for lysosomal cholesterol accumulation was created. Through injections with heat-inactivated pneumococci, we aimed to demonstrate the specific contribution of lysosomal oxLDL accumulation inside macrophages in atherosclerosis development. RESULTS While there were no differences in plaque morphology, a reduction in plaque size and plaque inflammation was found in immunized NPC1mut-transplanted mice, compared to non-immunized NPC1mut-transplanted mice. CONCLUSIONS Lysosomal oxLDL accumulation within macrophages contributes to murine atherosclerosis. Future intervention strategies should focus specifically on preventing oxLDL, unlike non-modified LDL, from being internalized into lysosomes. Such an intervention can have an additive effect to current existing treatments against atherosclerosis.
Collapse
Affiliation(s)
- Mike L J Jeurissen
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sofie M A Walenbergh
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Tom Houben
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marion J J Gijbels
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands; Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jieyi Li
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Tim Hendrikx
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Yvonne Oligschlaeger
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Patrick J van Gorp
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; Center for Molecular Medicine (CeMM), Austrian Academy of Sciences, Vienna, Austria
| | - Marjo M P C Donners
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ronit Shiri-Sverdlov
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands.
| |
Collapse
|
14
|
Song D, Fang G, Greenberg H, Liu SF. Chronic intermittent hypoxia exposure-induced atherosclerosis: a brief review. Immunol Res 2016; 63:121-30. [PMID: 26407987 DOI: 10.1007/s12026-015-8703-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Obstructive sleep apnea (OSA) is highly prevalent in the USA and is recognized as an independent risk factor for atherosclerotic cardiovascular disease. Identification of atherosclerosis risk factor attributable to OSA may provide opportunity to develop preventive measures for cardiovascular risk reduction. Chronic intermittent hypoxia (CIH) is a prominent feature of OSA pathophysiology and may be a major mechanism linking OSA to arteriosclerosis. Animal studies demonstrated that CIH exposure facilitated high-cholesterol diet (HCD)-induced atherosclerosis, accelerated the progression of existing atherosclerosis, and induced atherosclerotic lesions in the absence of other atherosclerosis risk factors, demonstrating that CIH is an independent causal factor of atherosclerosis. Comparative studies revealed major differences between CIH-induced and the classic HCD-induced atherosclerosis. Systemically, CIH was a much weaker inducer of atherosclerosis. CIH and HCD differentially activated inflammatory pathways. Histologically, CIH-induced atherosclerotic plaques had no clear necrotic core, contained a large number of CD31+ endothelial cells, and had mainly elastin deposition, whereas HCD-induced plaques had typical necrotic cores and fibrous caps, contained few endothelial cells, and had mainly collagen deposition. Metabolically, CIH caused mild, but HCD caused more severe dyslipidemia. Mechanistically, CIH did not, but HCD did, cause macrophage foam cell formation. NF-κB p50 gene deletion augmented CIH-induced, but not HCD-induced atherosclerosis. These differences reflect the intrinsic differences between the two types of atherosclerosis in terms of pathological nature and underlying mechanisms and support the notion that CIH-induced atherosclerosis is a new paradigm that differs from the classic HCD-induced atherosclerosis.
Collapse
Affiliation(s)
- Dongmei Song
- Centers for Heart and Lung Research, and Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Hofstra North Shore-LIJ School of Medicine, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Guoqiang Fang
- Centers for Heart and Lung Research, and Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Hofstra North Shore-LIJ School of Medicine, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Harly Greenberg
- Centers for Heart and Lung Research, and Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Hofstra North Shore-LIJ School of Medicine, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Shu Fang Liu
- Centers for Heart and Lung Research, and Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Hofstra North Shore-LIJ School of Medicine, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
15
|
van der Valk FM, Schulte DM, Meiler S, Tang J, Zheng KH, Van den Bossche J, Seijkens T, Laudes M, de Winther M, Lutgens E, Alaarg A, Metselaar JM, Dallinga-Thie GM, Mulder WJ, Stroes ES, Hamers AA. Liposomal prednisolone promotes macrophage lipotoxicity in experimental atherosclerosis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1463-70. [DOI: 10.1016/j.nano.2016.02.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/17/2016] [Accepted: 02/25/2016] [Indexed: 01/09/2023]
|
16
|
Tian Y, Guo S, Guo Y, Jian L. Anesthetic Propofol Attenuates Apoptosis, Aβ Accumulation, and Inflammation Induced by Sevoflurane Through NF-κB Pathway in Human Neuroglioma Cells. Cell Mol Neurobiol 2015; 35:891-8. [PMID: 25809614 DOI: 10.1007/s10571-015-0184-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/17/2015] [Indexed: 01/22/2023]
Abstract
Anesthetics have been reported to promote Alzheimer's disease neuropathogenesis by inducing amyloid beta (Aβ) protein accumulation and apoptosis. The aim of this study was to evaluate the effect of propofol on the apoptosis, Aβ accumulation, and inflammation induced by sevoflurane in human neuroglioma cells. Human neuroglioma cells were treated with or without sevoflurane and then co-incubated with or without propofol. Cell apoptosis was evaluated by fluorescence-activated cell sorting analysis (FACS) using AV-PI kits, and data showed that apoptosis induced by sevoflurane was significantly attenuated by propofol treatment. In addition, with the reactive oxygen species (ROS) production measured by FACS after staining with dichloro-dihydrofluorescein diacetate, propofol could significantly reduce the production of ROS as well as the accumulation of Aβ induced by sevoflurane assessed by enzyme-linked immuno sorbent assay (ELISA) analysis. On the other hand, the same treatment decreased the inflammation factor production of interleukin-6. Moreover, the level of nuclear factor-kappa B (NF-κB) was tested by Western blot and immunofluorescence assay. We found that the activation of NF-κB pathway was suppressed by propofol. The results suggest that propofol can effectively attenuate the apoptosis, Aβ accumulation, and inflammation induced by sevoflurane in human neuroglioma cells through NF-κB signal pathway.
Collapse
Affiliation(s)
- Yue Tian
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | | | | | | |
Collapse
|
17
|
Buckley ML, Ramji DP. The influence of dysfunctional signaling and lipid homeostasis in mediating the inflammatory responses during atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1498-510. [PMID: 25887161 DOI: 10.1016/j.bbadis.2015.04.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/25/2015] [Accepted: 04/08/2015] [Indexed: 12/14/2022]
Abstract
Atherosclerosis, the underlying cause of myocardial infarction and thrombotic cerebrovascular events, is responsible for the majority of deaths in westernized societies. Mortality from this disease is also increasing at a marked rate in developing countries due to the acquisition of a westernized lifestyle accompanied with elevated rates of obesity and diabetes. Atherosclerosis is recognized as a chronic inflammatory disorder associated with lipid accumulation and the development of fibrotic plaques within the walls of medium and large arteries. A range of immune cells, such as macrophages and T-lymphocytes, through the action of various cytokines, such as interleukins-1 and -33, transforming growth factor-β and interferon-γ, orchestrates the inflammatory response in this disease. The disease is also characterized by marked dysfunction in lipid homeostasis and signaling pathways that control the inflammatory response. This review will discuss the molecular basis of atherosclerosis with particular emphasis on the roles of the immune cells and cytokines along with the dysfunctional lipid homeostasis and cell signaling associated with this disease.
Collapse
Affiliation(s)
- Melanie L Buckley
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK.
| |
Collapse
|
18
|
Cao Q, Jiang Y, Shi J, Xu C, Liu X, Yang T, Fu P, Niu T. Artemisinin inhibits the proliferation, migration, and inflammatory reaction induced by tumor necrosis factor-α in vascular smooth muscle cells through nuclear factor kappa B pathway. J Surg Res 2015; 194:667-678. [DOI: 10.1016/j.jss.2014.12.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/28/2014] [Accepted: 12/04/2014] [Indexed: 01/28/2023]
|
19
|
Elliott KJ, Eguchi S. Phosphorylation Regulation by Kinases and Phosphatases in Atherosclerosis. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
20
|
Sui Y, Park SH, Xu J, Monette S, Helsley RN, Han SS, Zhou C. IKKβ links vascular inflammation to obesity and atherosclerosis. ACTA ACUST UNITED AC 2014; 211:869-86. [PMID: 24799533 PMCID: PMC4010900 DOI: 10.1084/jem.20131281] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
IKKβ functions in smooth muscle cells to regulate vascular inflammatory responses and atherosclerosis development. IκB kinase β (IKKβ), a central coordinator of inflammatory responses through activation of NF-κB, has been implicated in vascular pathologies, but its role in atherogenesis remains elusive. Here, we demonstrate that IKKβ functions in smooth muscle cells (SMCs) to regulate vascular inflammatory responses and atherosclerosis development. IKKβ deficiency in SMCs driven by a SM22Cre-IKKβ-flox system rendered low density lipoprotein receptor-null mice resistant to vascular inflammation and atherosclerosis induced by high-fat feeding. Unexpectedly, IKKβ-deficient mice were also resistant to diet-induced obesity and metabolic disorders. Cell lineage analysis revealed that SM22Cre is active in primary adipose stromal vascular cells and deficiency of IKKβ diminished the ability of these cells to differentiate, leading to accumulation of adipocyte precursor cells in adipose tissue. Mechanistically, reduction of IKKβ expression or pharmacological inhibition of IKKβ inhibited proteasome-mediated β-catenin ubiquitination and degradation in murine preadipocytes, resulting in elevated β-catenin levels and impaired adipocyte differentiation. Further, chronic treatment of mice with a potent IKKβ inhibitor decreased adipogenesis and ameliorated diet-induced obesity. Our findings demonstrate a pivotal role of IKKβ in linking vascular inflammation to atherosclerosis and adipose tissue development, and provide evidence for using appropriate IKKβ inhibitors in the treatment of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Yipeng Sui
- Graduate Center for Nutritional Sciences, 2 Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536
| | | | | | | | | | | | | |
Collapse
|
21
|
Wang Y, Wang GZ, Rabinovitch PS, Tabas I. Macrophage mitochondrial oxidative stress promotes atherosclerosis and nuclear factor-κB-mediated inflammation in macrophages. Circ Res 2013; 114:421-33. [PMID: 24297735 DOI: 10.1161/circresaha.114.302153] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE Mitochondrial oxidative stress (mitoOS) has been shown to correlate with the progression of human atherosclerosis. However, definitive cell type-specific causation studies in vivo are lacking, and the molecular mechanisms of potential proatherogenic effects remain to be determined. OBJECTIVE Our aims were to assess the importance of macrophage mitoOS in atherogenesis and to explore the underlying molecular mechanisms. METHODS AND RESULTS We first validated Western diet-fed Ldlr(-/-) mice as a model of human mitoOS-atherosclerosis association by showing that non-nuclear oxidative DNA damage, a marker of mitoOS in lesional macrophages, correlates with aortic root lesion development. To investigate the importance of macrophage mitoOS, we used a genetic engineering strategy in which the OS suppressor catalase was ectopically expressed in mitochondria (mCAT) in macrophages. MitoOS in lesional macrophages was successfully suppressed in these mice, and this led to a significant reduction in aortic root lesional area. The mCAT lesions had less monocyte-derived cells, less Ly6c(hi) monocyte infiltration into lesions, and lower levels of monocyte chemotactic protein-1. The decrease in lesional monocyte chemotactic protein-1 was associated with the suppression of other markers of inflammation and with decreased phosphorylation of RelA (NF-κB p65), indicating decreased activation of the proinflammatory NF-κB pathway. Using models of mitoOS in cultured macrophages, we showed that mCAT suppressed monocyte chemotactic protein-1 expression by decreasing the activation of the IκB-kinase β-RelA NF-κB pathway. CONCLUSIONS MitoOS in lesional macrophages amplifies atherosclerotic lesion development by promoting NF-κB-mediated entry of monocytes and other inflammatory processes. In view of the mitoOS-atherosclerosis link in human atheromata, these findings reveal a potentially new therapeutic target to prevent the progression of atherosclerosis.
Collapse
Affiliation(s)
- Ying Wang
- From the Departments of Medicine, Physiology, and Cellular Biophysics, and Pathology and Cell Biology (Y.W., I.T.), and Medical Scientist Training Program (G.Z.W.), Columbia University, New York, NY; and Department of Pathology, University of Washington, Seattle, WA (P.S.R.)
| | | | | | | |
Collapse
|
22
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
23
|
Sun X, He S, Wara AKM, Icli B, Shvartz E, Tesmenitsky Y, Belkin N, Li D, Blackwell TS, Sukhova GK, Croce K, Feinberg MW. Systemic delivery of microRNA-181b inhibits nuclear factor-κB activation, vascular inflammation, and atherosclerosis in apolipoprotein E-deficient mice. Circ Res 2013; 114:32-40. [PMID: 24084690 DOI: 10.1161/circresaha.113.302089] [Citation(s) in RCA: 234] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE Activated nuclear factor (NF)-κB signaling in the vascular endothelium promotes the initiation and progression of atherosclerosis. Targeting endothelial NF-κB may provide a novel strategy to limit chronic inflammation. OBJECTIVE To examine the role of microRNA-181b (miR-181b) in endothelial NF-κB signaling and effects on atherosclerosis. METHODS AND RESULTS MiR-181b expression was reduced in the aortic intima and plasma in apolipoprotein E-deficient mice fed a high-fat diet. Correspondingly, circulating miR-181b in the plasma was markedly reduced in human subjects with coronary artery disease. Systemic delivery of miR-181b resulted in a 2.3-fold overexpression of miR-181b in the aortic intima of apolipoprotein E-deficient mice and suppressed NF-κB signaling revealed by bioluminescence imaging and reduced target gene expression in the aortic arch in apolipoprotein E-deficient/NF-κB-luciferase transgenic mice. MiR-181b significantly inhibited atherosclerotic lesion formation, proinflammatory gene expression and the influx of lesional macrophages and CD4+ T cells in the vessel wall. Mechanistically, miR-181b inhibited the expression of the target gene importin-α3, an effect that reduced NF-κB nuclear translocation specifically in the vascular endothelium of lesions, whereas surprisingly leukocyte NF-κB signaling was unaffected despite a 7-fold overexpression of miR-181b. Our findings uncover that NF-κB nuclear translocation in leukocytes does not involve importin-α3, but rather importin-α5, which miR-181b does not target, highlighting that inhibition of NF-κB signaling in the endothelium is sufficient to mediate miR-181b's protective effects. CONCLUSIONS Systemic delivery of miR-181b inhibits the activation of NF-κB and atherosclerosis through cell-specific mechanisms in the vascular endothelium. These findings support the rationale that delivery of miR-181b may provide a novel therapeutic approach to treat chronic inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Xinghui Sun
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Shaolin He
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - A K M Wara
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Basak Icli
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Eugenia Shvartz
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Yevgenia Tesmenitsky
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Nathan Belkin
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Dazhu Li
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Timothy S Blackwell
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Galina K Sukhova
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Kevin Croce
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.S., S.H., A.K.M.W., B.I., E.S., Y.T., N.B., G.K.S., K.C., M.W.F.); Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (S.H., D.L.); and Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN (T.S.B.)
| |
Collapse
|
24
|
Wolfs IMJ, Stöger JL, Goossens P, Pöttgens C, Gijbels MJJ, Wijnands E, Vorst EPC, Gorp P, Beckers L, Engel D, Biessen EAL, Kraal G, Die I, Donners MMPC, Winther MPJ. Reprogramming macrophages to an anti‐inflammatory phenotype by helminth antigens reduces murine atherosclerosis. FASEB J 2013; 28:288-99. [DOI: 10.1096/fj.13-235911] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Ine M. J. Wolfs
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
- Department of PathologyMaastricht UniversityMaastrichtThe Netherlands
| | - J. Lauran Stöger
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
- Department of Medical BiochemistryAcademic Medical Center (AMC)University of AmsterdamAmsterdamThe Netherlands
| | - Pieter Goossens
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
- Centre d'Immunologie de Marseille‐Luminy (CIML)Aix‐Marseille UniversityMarseilleFrance
| | - Chantal Pöttgens
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
- Department of PhysiologyCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Marion J. J. Gijbels
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
- Department of PathologyMaastricht UniversityMaastrichtThe Netherlands
- Department of Medical BiochemistryAcademic Medical Center (AMC)University of AmsterdamAmsterdamThe Netherlands
| | - Erwin Wijnands
- Department of PathologyMaastricht UniversityMaastrichtThe Netherlands
| | - Emiel P. C. Vorst
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
| | - Patrick Gorp
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
| | - Linda Beckers
- Department of PathologyMaastricht UniversityMaastrichtThe Netherlands
- Department of Medical BiochemistryAcademic Medical Center (AMC)University of AmsterdamAmsterdamThe Netherlands
| | - David Engel
- Department of PathologyMaastricht UniversityMaastrichtThe Netherlands
| | | | - Georg Kraal
- Department of Molecular Cell Biology and ImmunologyVrije Universiteit (VU) Medical Center AmsterdamAmsterdamThe Netherlands
| | - Irma Die
- Department of Molecular Cell Biology and ImmunologyVrije Universiteit (VU) Medical Center AmsterdamAmsterdamThe Netherlands
| | - Marjo M. P. C. Donners
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
- Department of PathologyMaastricht UniversityMaastrichtThe Netherlands
| | - Menno P. J. Winther
- Department of Molecular GeneticsMaastricht UniversityMaastrichtThe Netherlands
- Department of Medical BiochemistryAcademic Medical Center (AMC)University of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
25
|
Atkins GB, Simon DI. Interplay between NF-κB and Kruppel-like factors in vascular inflammation and atherosclerosis: location, location, location. J Am Heart Assoc 2013; 2:e000290. [PMID: 23757395 PMCID: PMC3698797 DOI: 10.1161/jaha.113.000290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- G. Brandon Atkins
- Harrington Heart & Vascular Institute, Case Cardiovascular Research Institute, Department of Medicine, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH (B.A., D.I.S.)
- Correspondence to: G. Brandon Atkins, MD, PhD, Case Cardiovascular Research Institute, Iris S. & Bert L. Wolstein Research Building, 2103 Cornell Road, Room 4‐542, Cleveland, OH 44106. E‐mail:
| | - Daniel I. Simon
- Harrington Heart & Vascular Institute, Case Cardiovascular Research Institute, Department of Medicine, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH (B.A., D.I.S.)
| |
Collapse
|
26
|
Yoshida T, Yamashita M, Horimai C, Hayashi M. Smooth muscle-selective inhibition of nuclear factor-κB attenuates smooth muscle phenotypic switching and neointima formation following vascular injury. J Am Heart Assoc 2013; 2:e000230. [PMID: 23702880 PMCID: PMC3698790 DOI: 10.1161/jaha.113.000230] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Vascular proliferative diseases such as atherosclerosis are inflammatory disorders involving multiple cell types including macrophages, lymphocytes, endothelial cells, and smooth muscle cells (SMCs). Although activation of the nuclear factor-κB (NF-κB) pathway in vessels has been shown to be critical for the progression of vascular diseases, the cell-autonomous role of NF-κB within SMCs has not been fully understood. METHODS AND RESULTS We generated SMC-selective truncated IκB expressing (SM22α-Cre/IκBΔN) mice, in which NF-κB was inhibited selectively in SMCs, and analyzed their phenotype following carotid injury. Results showed that neointima formation was markedly reduced in SM22α-Cre/IκBΔN mice after injury. Although vascular injury induced downregulation of expression of SMC differentiation markers and myocardin, a potent activator of SMC differentiation markers, repression of these markers and myocardin was attenuated in SM22α-Cre/IκBΔN mice. Consistent with these findings, NF-κB activation by interleukin-1β (IL-1β) decreased expression of SMC differentiation markers as well as myocardin in cultured SMCs. Inhibition of NF-κB signaling by BAY 11-7082 attenuated repressive effects of IL-1β. Of interest, Krüppel-like factor 4 (Klf4), a transcription factor critical for regulating SMC differentiation and proliferation, was also involved in IL-1β-mediated myocardin repression. Promoter analyses and chromatin immunoprecipitation assays revealed that NF-κB repressed myocardin by binding to the myocardin promoter region in concert with Klf4. CONCLUSIONS These results provide novel evidence that activation of the NF-κB pathway cell-autonomously mediates SMC phenotypic switching and contributes to neointima formation following vascular injury.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan.
| | | | | | | |
Collapse
|
27
|
Peptide inhibitor of NF-κB translocation ameliorates experimental atherosclerosis. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1910-21. [PMID: 23597852 DOI: 10.1016/j.ajpath.2013.01.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 12/17/2012] [Accepted: 01/10/2013] [Indexed: 11/22/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall. NF-κB is a major regulator of inflammation that controls the expression of many genes involved in atherogenesis. Activated NF-κB was detected in human atherosclerotic plaques, and modulation of NF-κB inflammatory activity limits disease progression in mice. Herein, we investigate the anti-inflammatory and atheroprotective effects of a cell-permeable peptide containing the NF-κB nuclear localization sequence (NLS). In vascular smooth muscle cells and macrophages, NLS peptide specifically blocked the importin α-mediated nuclear import of NF-κB and prevented lipopolysaccharide-induced pro-inflammatory gene expression, cell migration, and oxidative stress. In experimental atherosclerosis (apolipoprotein E-knockout mice fed a high-fat diet), i.p., 0.13 μmol/day NLS peptide administration for 5 weeks attenuated NF-κB activation in atherosclerotic plaques. NLS peptide significantly inhibited lesion development at both early (age 10 weeks) and advanced (age 28 weeks) stages of atherosclerosis in mice, without affecting serum lipid levels. Plaques from NLS-treated mice contained fewer macrophages of pro-inflammatory M1 subtype than those from respective untreated controls. By contrast, the relative smooth muscle cell and collagen content was increased, indicating a more stable plaque phenotype. NLS peptide also attenuated pro-inflammatory gene expression and oxidative stress in aortic lesions. Our study demonstrates that targeting NF-κB nuclear translocation hampers inflammation and atherosclerosis development and identifies cell-permeable NLS peptide as a potential anti-atherosclerotic agent.
Collapse
|
28
|
Park SH, Sui Y, Gizard F, Xu J, Rios-Pilier J, Helsley RN, Han SS, Zhou C. Myeloid-specific IκB kinase β deficiency decreases atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol 2012; 32:2869-76. [PMID: 23023371 DOI: 10.1161/atvbaha.112.254573] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Inflammatory responses are the driving force of atherosclerosis development. IκB kinase β (IKKβ), a central coordinator in inflammation through regulation of nuclear factor-κB, has been implicated in the pathogenesis of atherosclerosis. Macrophages play an essential role in the initiation and progression of atherosclerosis, yet the role of macrophage IKKβ in atherosclerosis remains elusive and controversial. This study aims to investigate the impact of IKKβ expression on macrophage functions and to assess the effect of myeloid-specific IKKβ deletion on atherosclerosis development. METHODS AND RESULTS To explore the issue of macrophage IKKβ involvement of atherogenesis, we generated myeloid-specific IKKβ-deficient low-density lipoprotein receptor-deficient mice (IKKβ(ΔMye)LDLR(-/-)). Deficiency of IKKβ in myeloid cells did not affect plasma lipid levels but significantly decreased diet-induced atherosclerotic lesion areas in the aortic root, brachiocephalic artery, and aortic arch of low-density lipoprotein receptor-deficient mice. Ablation of myeloid IKKβ attenuated macrophage inflammatory responses and decreased atherosclerotic lesional inflammation. Furthermore, deficiency of IKKβ decreased adhesion, migration, and lipid uptake in macrophages. CONCLUSIONS The present study demonstrates a pivotal role for myeloid IKKβ expression in atherosclerosis by modulating macrophage functions involved in atherogenesis. These results suggest that inhibiting nuclear factor-κB activation in macrophages may represent a feasible approach to combat atherosclerosis.
Collapse
Affiliation(s)
- Se-Hyung Park
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Coban D, Milenkovic D, Chanet A, Khallou-Laschet J, Sabbe L, Palagani A, Vanden Berghe W, Mazur A, Morand C. Dietary curcumin inhibits atherosclerosis by affecting the expression of genes involved in leukocyte adhesion and transendothelial migration. Mol Nutr Food Res 2012; 56:1270-81. [PMID: 22753158 DOI: 10.1002/mnfr.201100818] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 05/10/2012] [Accepted: 05/12/2012] [Indexed: 02/06/2023]
Abstract
SCOPE The aim of the study was to examine the atheroprotective effect of dietary curcumin in a mouse model of atherosclerosis and to identify its cellular and molecular targets at the vascular level. METHODS AND RESULTS ApoE(-/-) mice were fed with curcumin at 0.2% (wt/wt) in diet for 4 months. This supplementation reduced the extent of atherosclerotic lesion by 26% and induced changes in expression of genes implicated in cell adhesion and transendothelial migration or cytoskeleton organization, as revealed by a transcriptomic analysis in the aorta. Expression profile of these genes suggests reduction in both leukocyte adhesion and transendothelial migration. In agreement with this hypothesis, we observed a reduction (-37%) in macrophage infiltration in the plaque, as measured by immunohistochemistry, and, in vitro, a lower adhesion of monocytes to TNF-α-stimulated endothelial cells (-32%) after exposure to a nutritionally achievable concentration of curcumin. These changes in gene expression could be related to the observed increased expression of IκB protein and decrease of TNF-α-induced NF-κB/DNA binding and NF-κB-transcriptional activity upon exposure to curcumin. CONCLUSION Our findings pointed out that the antiatherogenic effect of curcumin could be linked to its effect on gene networks and cell functions related to leukocyte adhesion and transendothelial migration via NF-κB-dependent pathways.
Collapse
Affiliation(s)
- Dilek Coban
- INRA, UMR 1019, UNH, CRNH Auvergne, Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hematopoietic miR155 deficiency enhances atherosclerosis and decreases plaque stability in hyperlipidemic mice. PLoS One 2012; 7:e35877. [PMID: 22558252 PMCID: PMC3338496 DOI: 10.1371/journal.pone.0035877] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 03/23/2012] [Indexed: 11/19/2022] Open
Abstract
microRNA-155 (miR155) is a central regulator of immune responses that is induced by inflammatory mediators. Although miR155 is considered to be a pro-inflammatory microRNA, in vitro reports show anti-inflammatory effects in lipid-loaded cells. In this study we examined the role of miR155 in atherosclerosis in vivo using bone marrow transplantation from miR155 deficient or wildtype mice to hyperlipidemic mice. Hematopoietic deficiency of miR155 enhanced atherosclerotic plaque development and decreased plaque stability, as evidenced by increased myeloid inflammatory cell recruitment to the plaque. The increased inflammatory state was mirrored by a decrease in circulating CD4+CD25+FoxP3+ regulatory T cells, and an increase in granulocytes (CD11b+Ly6G+) in blood of miR155−/− transplanted mice. Moreover, we show for the first time a crucial role of miR155 in monocyte subset differentiation, since hematopoietic deficiency of miR155 increases the ‘inflammatory’ monocyte subset (CD11b+Ly6G−Ly6Chi) and reduces ‘resident’ monocytes (CD11b+Ly6G−Ly6Clow) in the circulation. Furthermore, cytokine production by resident peritoneal macrophages of miR155−/− transplanted hyperlipidemic mice was skewed towards a more pro-inflammatory state since anti-inflammatory IL-10 production was reduced. In conclusion, in this hyperlipidemic mouse model miR155 acts as an anti-inflammatory, atheroprotective microRNA. Additionally, besides a known role in lymphoid cell development, we show a crucial role of miR155 in myeloid lineage differentiation.
Collapse
|
31
|
Aparicio-Vergara M, Shiri-Sverdlov R, Koonen DPY, Hofker MH. Bone marrow transplantation as an established approach for understanding the role of macrophages in atherosclerosis and the metabolic syndrome. Curr Opin Lipidol 2012; 23:111-21. [PMID: 22274753 DOI: 10.1097/mol.0b013e3283508c4f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Bone marrow transplantation (BMT) technology is a firmly established tool for studying atherosclerosis. Only recently it is helping us to understand the inflammatory mechanisms leading to the development of obesity, insulin resistance and type 2 diabetes. Here we review the use of BMT as a tool for studying the metabolic syndrome. RECENT FINDINGS Bone marrow-derived cells, and particularly monocytes and macrophages, have been a major subject in the study of atherogenesis, and they are highly amenable for research purposes because of their application in bone marrow transplantations. For example, the many pathways studied using BMT have helped unmask ABC transporters as the genes controlling reverse cholesterol transport and foam cell formation, as well as other genes like CCR2 and IκBα controlling leukocyte development, migration and activation. The invasion of leukocytes, not only in the vessel wall, but also in adipose tissue and liver, shares many common mechanisms relevant to atherosclerosis and metabolic diseases. SUMMARY BMT is an efficient and versatile tool for assessing the roles of specific genes that are restricted to hematopoietic cells, and especially the monocytes and macrophages in metabolic syndrome and its related pathologies.
Collapse
Affiliation(s)
- Marcela Aparicio-Vergara
- Molecular Genetics, Medical Biology Section, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|