1
|
Zhevlakova I, Liu H, Dudiki T, Gao D, Yakubenko V, Tkachenko S, Cherepanova O, Podrez EA, Byzova TV. Mechanisms and consequences of myeloid adhesome dysfunction in atherogenesis. Cardiovasc Res 2025; 121:62-76. [PMID: 39393814 PMCID: PMC11999018 DOI: 10.1093/cvr/cvae223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/24/2024] [Accepted: 08/23/2024] [Indexed: 10/13/2024] Open
Abstract
AIMS In the context of atherosclerosis, macrophages exposed to oxidized low-density lipoproteins (oxLDLs) exhibit cellular abnormalities, specifically in adhesome functions, yet the mechanisms and implications of these adhesive dysfunctions remain largely unexplored. METHODS AND RESULTS This study reveals a significant depletion of Kindlin3 (K3) or Fermt3, an essential component of the adhesome regulating integrin functions, in macrophages located within atherosclerotic plaques in vivo and following oxLDL exposure in vitro. To examine the effects of K3 deficiency, the study utilized hyperlipidaemic bone marrow chimeras devoid of myeloid Kindlin3 expression. The absence of myeloid K3 increased atherosclerotic plaque burden in the aortas in vivo and enhanced lipid accumulation and lipoprotein uptake in macrophages from Kindlin3-null chimeric mice in vitro. Importantly, re-expression of K3 in macrophages ameliorated these abnormalities. RNA sequencing of bone marrow-derived macrophages (BMDM) from K3-deficient mice revealed extensive deregulation in adhesion-related pathways, echoing changes observed in wild-type cells treated with oxLDL. Notably, there was an increase in Olr1 expression [encoding the lectin-like oxidized LDL receptor-1 (LOX1)], a gene implicated in atherogenesis. The disrupted K3-integrin axis in macrophages led to a significant elevation in the LOX1 receptor, contributing to increased oxLDL uptake and foam cell formation. Inhibition of LOX1 normalized lipid uptake in Kindlin3-null macrophages. A similar proatherogenic phenotype, marked by increased macrophage LOX1 expression and foam cell formation, was observed in myeloid-specific Itgβ1-deficient mice but not in Itgβ2-deficient mice, underscoring the critical role of K3/Itgβ1 interaction. CONCLUSION This study shows that the loss of Kindlin3 in macrophages upon exposure to oxLDL leads to adhesome dysfunction in atherosclerosis and reveals the pivotal role of Kindlin3 in macrophage function and its contribution to the progression of atherosclerosis, providing valuable insights into the molecular mechanisms that could be targeted for therapeutic interventions.
Collapse
Affiliation(s)
- Irina Zhevlakova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Huan Liu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Tejasvi Dudiki
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Valentin Yakubenko
- Department of Biomedical Sciences, Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37684, USA
| | - Svyatoslav Tkachenko
- Department of Genetics and Genome Sciences, Case Western Reserve University, 2109 Adelbert Rd Building, Cleveland, OH 44106, USA
| | - Olga Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Eugene A Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
2
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
3
|
Davies B, Trelfa L, Rashbrook VS, Drydale E, Martin R, Bai B, Golebka J, Biggs DS, Channon KM, Bhattacharya S, Douglas G. Mutagenesis on a complex mouse genetic background by site-specific nucleases. Transgenic Res 2024; 33:415-426. [PMID: 39088185 PMCID: PMC11588839 DOI: 10.1007/s11248-024-00399-5] [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: 10/27/2023] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Mouse models with complex genetic backgrounds are increasingly used in preclinical research to accurately model human disease and to enable temporal and cell-specific evaluation of genetic manipulations. Backcrossing mice onto these complex genetic backgrounds takes time and leads to significant wastage of animals. In this study, we aimed to evaluate whether site-specific nucleases could be used to generate additional genetic mutations in a complex genetic background, using the REVERSA mouse model of atherosclerosis, a model harbouring four genetically altered alleles. The model is comprised of a functional null mutation in the Ldlr gene in combination with a ApoB100 allele, which, after high-fat diet, leads to the rapid development of atherosclerosis. The regression of the pathology is achieved by inducible knock-out of the Mttp gene. Here we report an investigation to establish if microinjection of site-specific nucleases directly into zygotes prepared from the REVERSA could be used to investigate the role of the ATP binding cassette transporter G1 (ABCG1) in atherosclerosis regression. We show that using this approach we could successfully generate two independent knockout lines on the REVERSA background, both of which exhibited the expected phenotype of a significant reduction in cholesterol efflux to HDL in bone marrow-derived macrophages. However, loss of Abcg1 did not impact atherosclerosis regression in either the aortic root or in aortic arch, demonstrating no important role for this transporter subtype. We have demonstrated that site-specific nucleases can be used to create genetic modifications directly onto complex disease backgrounds and can be used to explore gene function without the need for laborious backcrossing of independent strains, conveying a significant 3Rs advantage.
Collapse
Affiliation(s)
- Benjamin Davies
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
- Francis Crick Institute, 1 Midland Road, London, UK
| | - Lucy Trelfa
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Victoria S Rashbrook
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Edward Drydale
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Rachel Martin
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Boyan Bai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Jedrzej Golebka
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Daniel Stephen Biggs
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Shoumo Bhattacharya
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Gillian Douglas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK.
| |
Collapse
|
4
|
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S, Liu C. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:130. [PMID: 38816371 PMCID: PMC11139930 DOI: 10.1038/s41392-024-01840-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 06/01/2024] Open
Abstract
The immune response holds a pivotal role in cardiovascular disease development. As multifunctional cells of the innate immune system, macrophages play an essential role in initial inflammatory response that occurs following cardiovascular injury, thereby inducing subsequent damage while also facilitating recovery. Meanwhile, the diverse phenotypes and phenotypic alterations of macrophages strongly associate with distinct types and severity of cardiovascular diseases, including coronary heart disease, valvular disease, myocarditis, cardiomyopathy, heart failure, atherosclerosis and aneurysm, which underscores the importance of investigating macrophage regulatory mechanisms within the context of specific diseases. Besides, recent strides in single-cell sequencing technologies have revealed macrophage heterogeneity, cell-cell interactions, and downstream mechanisms of therapeutic targets at a higher resolution, which brings new perspectives into macrophage-mediated mechanisms and potential therapeutic targets in cardiovascular diseases. Remarkably, myocardial fibrosis, a prevalent characteristic in most cardiac diseases, remains a formidable clinical challenge, necessitating a profound investigation into the impact of macrophages on myocardial fibrosis within the context of cardiac diseases. In this review, we systematically summarize the diverse phenotypic and functional plasticity of macrophages in regulatory mechanisms of cardiovascular diseases and unprecedented insights introduced by single-cell sequencing technologies, with a focus on different causes and characteristics of diseases, especially the relationship between inflammation and fibrosis in cardiac diseases (myocardial infarction, pressure overload, myocarditis, dilated cardiomyopathy, diabetic cardiomyopathy and cardiac aging) and the relationship between inflammation and vascular injury in vascular diseases (atherosclerosis and aneurysm). Finally, we also highlight the preclinical/clinical macrophage targeting strategies and translational implications.
Collapse
Affiliation(s)
- Runkai Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Hongrui Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Botao Tang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yukun Luo
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yufei Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Xin Zhong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Sifei Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Shengkang Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Canzhao Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| |
Collapse
|
5
|
Zhou Y, Wu Q, Long X, He Y, Huang J. lncRNA HOTAIRM1 Activated by HOXA4 Drives HUVEC Proliferation Through Direct Interaction with Protein Partner HSPA5. Inflammation 2024; 47:421-437. [PMID: 37898994 PMCID: PMC10798933 DOI: 10.1007/s10753-023-01919-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/31/2023]
Abstract
Despite the substantial progress in deciphering the pathogenesis of atherosclerosis (AS), cardiovascular mortality is still increasing. Therefore, atherosclerotic cardiovascular disease remains a sweeping epidemic that jeopardizes human health. Disentangling the molecular underpinnings of AS is imperative in the molecular cardiology field. Overwhelming evidence has indicated that the recognition of a fascinating class of players, known as long non-coding RNAs (lncRNAs), provides causality for coordinating AS. However, the function and mechanism of HOTAIRM1 are still poorly understood in human umbilical vein endothelial cells (HUVECs) and AS. Herein, we primarily underscored that lncRNA HOTAIRM1 is potentially responsible for AS; as such, it was dramatically up-regulated in HUVECs upon ox-LDL stimulation. Functionally, HOTAIRM1 knockdown attenuated HUVEC proliferation and potentiated apoptosis in the absence and presence of ox-LDL. Furthermore, HOTAIRM1 was preferentially located in the nuclei of HUVECs. Mechanistically, HOXA4 is directly bound to the HOTAIRM1 promoter and activated its transcription. Of note, a positive feedback signaling between HOXA4 and HOTAIRM1 was determined. Intriguingly, the interplay between HOTAIRM1 and HSPA5 occurred in an RNA-binding protein pattern and a transcription-dependent regulatory manner. In addition, HSPA5 overexpression partially antagonized HUVEC proliferation inhibition of HOTAIRM1 depletion. Taken together, our findings delineate a pivotal functional interaction among HOXA4, HOTAIRM1, and HSPA5 as a novel regulatory circuit for modulating HUVEC proliferation. An in-depth investigation of the HOXA4-HOTAIRM1-HSPA5 axis promises to yield significant breakthroughs in identifying the molecular mechanisms governing AS and developing therapeutic avenues for AS.
Collapse
Affiliation(s)
- Yu Zhou
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Qiang Wu
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| | - Xiangshu Long
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Youfu He
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Jing Huang
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| |
Collapse
|
6
|
Akhter MS, Goodwin JE. Endothelial Dysfunction in Cardiorenal Conditions: Implications of Endothelial Glucocorticoid Receptor-Wnt Signaling. Int J Mol Sci 2023; 24:14261. [PMID: 37762564 PMCID: PMC10531724 DOI: 10.3390/ijms241814261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
The endothelium constitutes the innermost lining of the blood vessels and controls blood fluidity, vessel permeability, platelet aggregation, and vascular tone. Endothelial dysfunction plays a key role in initiating a vascular inflammatory cascade and is the pivotal cause of various devastating diseases in multiple organs including the heart, lung, kidney, and brain. Glucocorticoids have traditionally been used to combat vascular inflammation. Endothelial cells express glucocorticoid receptors (GRs), and recent studies have demonstrated that endothelial GR negatively regulates vascular inflammation in different pathological conditions such as sepsis, diabetes, and atherosclerosis. Mechanistically, the anti-inflammatory effects of GR are mediated, in part, through the suppression of Wnt signaling. Moreover, GR modulates the fatty acid oxidation (FAO) pathway in endothelial cells and hence can influence FAO-mediated fibrosis in several organs including the kidneys. This review summarizes the relationship between GR and Wnt signaling in endothelial cells and the effects of the Wnt pathway in different cardiac and renal diseases. Available data suggest that GR plays a significant role in restoring endothelial integrity, and research on endothelial GR-Wnt interactions could facilitate the development of novel therapies for many cardiorenal conditions.
Collapse
Affiliation(s)
- Mohammad Shohel Akhter
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06511, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Julie Elizabeth Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06511, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
7
|
Sun H, Feng J, Ma Y, Cai D, Luo Y, Wang Q, Li F, Zhang M, Hu Q. Down-regulation of microRNA-342-5p or Up-regulation of Wnt3a Inhibits Angiogenesis and Maintains Atherosclerotic Plaque Stability in Atherosclerosis Mice. NANOSCALE RESEARCH LETTERS 2021; 16:165. [PMID: 34807315 PMCID: PMC8609054 DOI: 10.1186/s11671-021-03608-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/21/2021] [Indexed: 05/12/2023]
Abstract
Evidence has demonstrated that microRNA-342-5p (miR-342-5p) is implicated in atherosclerosis (AS), but little is known regarding its intrinsic regulatory mechanisms. Here, we aimed to explore the effect of miR-342-5p targeting Wnt3a on formation of vulnerable plaques and angiogenesis of AS. ApoE-/- mice were fed with high-fat feed for 16 w to replicate the AS vulnerable plaque model. miR-342-5p and Wnt3a expression in aortic tissues of AS were detected. The target relationship between miR-342-5p and Wnt3a was verified. Moreover, ApoE-/- mice were injected with miR-342-5p antagomir and overexpression-Wnt3a vector to test their functions in serum lipid levels, inflammatory and oxidative stress-related cytokines, aortic plaque stability and angiogenesis in plaque of AS mice. miR-342-5p expression was enhanced and Wnt3a expression was degraded in aortic tissues of AS mice and miR-342-5p directly targeted Wnt3a. Up-regulating Wnt3a or down-regulating miR-342-5p reduced blood lipid content, inflammatory and oxidative stress levels, the vulnerability of aortic tissue plaque and inhibited angiogenesis in aortic plaque of AS mice. Functional studies show that depleting miR-342-5p can stabilize aortic tissue plaque and reduce angiogenesis in plaque in AS mice via restoring Wnt3a.
Collapse
Affiliation(s)
- Haixia Sun
- Department of Cardiac Ultrasound, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai Province, China
| | - Jinhua Feng
- Department of General Practitioner, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai, China
| | - Yan Ma
- Department of Cardiac Ultrasound, Haixi People's Hospital, Delingha, 817099, Qinghai, China
| | - Ding Cai
- Department of Neurology, Qinghai Provincial People's Hospital, No. 2 Gonghe Road, East District, Xining, 810007, Qinghai Province, China
| | - Yulu Luo
- Department of Cardiac Ultrasound, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai Province, China
| | - Qinggong Wang
- Department of Cardiac Ultrasound, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai Province, China
| | - Fang Li
- Department of Cardiac Ultrasound, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai Province, China
| | - Mingyue Zhang
- Department of Cardiac Ultrasound, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai Province, China
| | - Quanzhong Hu
- Department of Neurology, Qinghai Provincial People's Hospital, No. 2 Gonghe Road, East District, Xining, 810007, Qinghai Province, China.
| |
Collapse
|
8
|
Weinstock A, Rahman K, Yaacov O, Nishi H, Menon P, Nikain CA, Garabedian ML, Pena S, Akbar N, Sansbury BE, Heffron SP, Liu J, Marecki G, Fernandez D, Brown EJ, Ruggles KV, Ramsey SA, Giannarelli C, Spite M, Choudhury RP, Loke P, Fisher EA. Wnt signaling enhances macrophage responses to IL-4 and promotes resolution of atherosclerosis. eLife 2021; 10:e67932. [PMID: 33720008 PMCID: PMC7994001 DOI: 10.7554/elife.67932] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a disease of chronic inflammation. We investigated the roles of the cytokines IL-4 and IL-13, the classical activators of STAT6, in the resolution of atherosclerosis inflammation. Using Il4-/-Il13-/- mice, resolution was impaired, and in control mice, in both progressing and resolving plaques, levels of IL-4 were stably low and IL-13 was undetectable. This suggested that IL-4 is required for atherosclerosis resolution, but collaborates with other factors. We had observed increased Wnt signaling in macrophages in resolving plaques, and human genetic data from others showed that a loss-of-function Wnt mutation was associated with premature atherosclerosis. We now find an inverse association between activation of Wnt signaling and disease severity in mice and humans. Wnt enhanced the expression of inflammation resolving factors after treatment with plaque-relevant low concentrations of IL-4. Mechanistically, activation of the Wnt pathway following lipid lowering potentiates IL-4 responsiveness in macrophages via a PGE2/STAT3 axis.
Collapse
Affiliation(s)
- Ada Weinstock
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Karishma Rahman
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Or Yaacov
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Hitoo Nishi
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Prashanthi Menon
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Cyrus A Nikain
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Michela L Garabedian
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Stephanie Pena
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Brian E Sansbury
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Sean P Heffron
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
- NYU Center for the Prevention of Cardiovascular Disease, New York University Grossman School of MedicineNew YorkUnited States
| | - Jianhua Liu
- Department of Surgery, Mount Sinai School of MedicineNew YorkUnited States
| | - Gregory Marecki
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Dawn Fernandez
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Emily J Brown
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Kelly V Ruggles
- Division of Translational Medicine, Department of Medicine, New York University Langone Health, Institute for Systems Genetics, New York University Grossman School of MedicineNew YorkUnited States
| | - Stephen A Ramsey
- Department of Biomedical Sciences, School of Electrical Engineering and Computer Science, Oregon State UniversityCorvallisUnited States
| | - Chiara Giannarelli
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- The Precision Immunology Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Microbiology (Parasitology), New York University School of MedicineNew YorkUnited States
| | - Matthew Spite
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - P'ng Loke
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesdaUnited States
| | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
- NYU Center for the Prevention of Cardiovascular Disease, New York University Grossman School of MedicineNew YorkUnited States
- Departments of Cell Biology and Microbiology, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
9
|
Liu Y, Neogi A, Mani A. The role of Wnt signalling in development of coronary artery disease and its risk factors. Open Biol 2020; 10:200128. [PMID: 33081636 PMCID: PMC7653355 DOI: 10.1098/rsob.200128] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/23/2020] [Indexed: 02/05/2023] Open
Abstract
The Wnt signalling pathways are composed of a highly conserved cascade of events that govern cell differentiation, apoptosis and cell orientation. Three major and distinct Wnt signalling pathways have been characterized: the canonical Wnt pathway (or Wnt/β-catenin pathway), the non-canonical planar cell polarity pathway and the non-canonical Wnt/Ca2+ pathway. Altered Wnt signalling pathway has been associated with diverse diseases such as disorders of bone density, different malignancies, cardiac malformations and heart failure. Coronary artery disease is the most common type of heart disease in the United States. Atherosclerosis is a multi-step pathological process, which starts with lipid deposition and endothelial cell dysfunction, triggering inflammatory reactions, followed by recruitment and aggregation of monocytes. Subsequently, monocytes differentiate into tissue-resident macrophages and transform into foam cells by the uptake of modified low-density lipoprotein. Meanwhile, further accumulations of lipids, infiltration and proliferation of vascular smooth muscle cells, and deposition of the extracellular matrix occur under the intima. An atheromatous plaque or hyperplasia of the intima and media is eventually formed, resulting in luminal narrowing and reduced blood flow to the myocardium, leading to chest pain, angina and even myocardial infarction. The Wnt pathway participates in all different stages of this process, from endothelial dysfunction to lipid deposit, and from initial inflammation to plaque formation. Here, we focus on the role of Wnt cascade in pathophysiological mechanisms that take part in coronary artery disease from both clinical and experimental perspectives.
Collapse
Affiliation(s)
- Ya Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Arpita Neogi
- Yale Cardiovascular Genetics Program, Yale University, New Haven, CT, USA
| | - Arya Mani
- Yale Cardiovascular Genetics Program, Yale University, New Haven, CT, USA
- Yale Cardiovascular Research Center, Department of Medicine, Yale University, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
10
|
Cocco M, Care MA, Saadi A, Al-Maskari M, Doody G, Tooze R. A dichotomy of gene regulatory associations during the activated B-cell to plasmablast transition. Life Sci Alliance 2020; 3:e202000654. [PMID: 32843533 PMCID: PMC7471511 DOI: 10.26508/lsa.202000654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 01/22/2023] Open
Abstract
The activated B-cell (ABC) to plasmablast transition encompasses the cusp of antibody-secreting cell (ASC) differentiation. We explore this transition with integrated analysis in human cells, focusing on changes that follow removal from CD40-mediated signals. Within hours of input signal loss, cell growth programs shift toward enhanced proliferation, accompanied by ER-stress response, and up-regulation of ASC features. Clustering of genomic occupancy for IRF4, BLIMP1, XBP1, and CTCF with histone marks identifies a dichotomy: XBP1 and IRF4 link to induced but not repressed gene modules in plasmablasts, whereas BLIMP1 links to modules of ABC genes that are repressed, but not to activated genes. Between ABC and plasmablast states, IRF4 shifts away from AP1/IRF composite elements while maintaining occupancy at IRF and ETS/IRF elements. This parallels the loss of BATF expression, which is identified as a potential BLIMP1 target. In plasmablasts, IRF4 acquires an association with CTCF, a feature maintained in plasma cell myeloma lines. Thus, shifting occupancy links IRF4 to both ABC and ASC gene expression, whereas BLIMP1 occupancy links to repression of the activation state.
Collapse
Affiliation(s)
- Mario Cocco
- Division of Immunology and Haematology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Matthew A Care
- Division of Immunology and Haematology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Bioinformatics Group, Institute of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Amel Saadi
- Division of Immunology and Haematology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Muna Al-Maskari
- Division of Immunology and Haematology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Medicine, Sultan Qaboos University Hospital, Muscat, Oman
| | - Gina Doody
- Division of Immunology and Haematology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Reuben Tooze
- Division of Immunology and Haematology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| |
Collapse
|
11
|
Senatus L, López-Díez R, Egaña-Gorroño L, Liu J, Hu J, Daffu G, Li Q, Rahman K, Vengrenyuk Y, Barrett TJ, Dewan MZ, Guo L, Fuller D, Finn AV, Virmani R, Li H, Friedman RA, Fisher EA, Ramasamy R, Schmidt AM. RAGE impairs murine diabetic atherosclerosis regression and implicates IRF7 in macrophage inflammation and cholesterol metabolism. JCI Insight 2020; 5:137289. [PMID: 32641587 PMCID: PMC7406264 DOI: 10.1172/jci.insight.137289] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/21/2020] [Indexed: 12/27/2022] Open
Abstract
Despite advances in lipid-lowering therapies, people with diabetes continue to experience more limited cardiovascular benefits. In diabetes, hyperglycemia sustains inflammation and preempts vascular repair. We tested the hypothesis that the receptor for advanced glycation end-products (RAGE) contributes to these maladaptive processes. We report that transplantation of aortic arches from diabetic, Western diet-fed Ldlr-/- mice into diabetic Ager-/- (Ager, the gene encoding RAGE) versus WT diabetic recipient mice accelerated regression of atherosclerosis. RNA-sequencing experiments traced RAGE-dependent mechanisms principally to the recipient macrophages and linked RAGE to interferon signaling. Specifically, deletion of Ager in the regressing diabetic plaques downregulated interferon regulatory factor 7 (Irf7) in macrophages. Immunohistochemistry studies colocalized IRF7 and macrophages in both murine and human atherosclerotic plaques. In bone marrow-derived macrophages (BMDMs), RAGE ligands upregulated expression of Irf7, and in BMDMs immersed in a cholesterol-rich environment, knockdown of Irf7 triggered a switch from pro- to antiinflammatory gene expression and regulated a host of genes linked to cholesterol efflux and homeostasis. Collectively, this work adds a new dimension to the immunometabolic sphere of perturbations that impair regression of established diabetic atherosclerosis and suggests that targeting RAGE and IRF7 may facilitate vascular repair in diabetes.
Collapse
Affiliation(s)
- Laura Senatus
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Raquel López-Díez
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Lander Egaña-Gorroño
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Jianhua Liu
- Marc and Ruti Bell Program in Vascular Biology, Leon H. Charney Division of Cardiology, Department of Medicine
| | - Jiyuan Hu
- Division of Biostatistics, Department of Population Health, and Department of Environmental Medicine, and
| | - Gurdip Daffu
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Qing Li
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Karishma Rahman
- Marc and Ruti Bell Program in Vascular Biology, Leon H. Charney Division of Cardiology, Department of Medicine
| | - Yuliya Vengrenyuk
- Marc and Ruti Bell Program in Vascular Biology, Leon H. Charney Division of Cardiology, Department of Medicine
| | - Tessa J. Barrett
- Marc and Ruti Bell Program in Vascular Biology, Leon H. Charney Division of Cardiology, Department of Medicine
| | - M. Zahidunnabi Dewan
- Experimental Pathology Research Laboratory, Department of Pathology, New York University (NYU) Langone Medical Center, New York, New York, USA
| | - Liang Guo
- CVPath Institute, Gaithersburg, Maryland, USA
| | | | | | | | - Huilin Li
- Division of Biostatistics, Department of Population Health, and Department of Environmental Medicine, and
| | - Richard A. Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center, and Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, New York, USA
| | - Edward A. Fisher
- Marc and Ruti Bell Program in Vascular Biology, Leon H. Charney Division of Cardiology, Department of Medicine
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| |
Collapse
|
12
|
Guan X, Yang X, Wang C, Bi R. In silico analysis of the molecular regulatory networks in peripheral arterial occlusive disease. Medicine (Baltimore) 2020; 99:e20404. [PMID: 32481342 PMCID: PMC7250035 DOI: 10.1097/md.0000000000020404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Peripheral arterial occlusive disease (PAOD) is a global public health concern that decreases the quality of life of the patients and can lead to disabilities and death. The aim of this study was to identify the genes and pathways associated with PAOD pathogenesis, and the potential therapeutic targets. METHODS Differentially expressed genes (DEGs) and miRNAs related to PAOD were extracted from the GSE57691 dataset and through text mining. Additionally, bioinformatics analysis was applied to explore gene ontology, pathways and protein-protein interaction of those DEGs. The potential miRNAs targeting the DEGs and the transcription factors (TFs) regulating miRNAs were predicted by multiple different databases. RESULTS A total of 59 DEGs were identified, which were significantly enriched in the inflammatory response, immune response, chemokine-mediated signaling pathway and JAK-STAT signaling pathway. Thirteen genes including IL6, CXCL12, IL1B, and STAT3 were hub genes in protein-protein interaction network. In addition, 513 miRNA-target gene pairs were identified, of which CXCL12 and PTPN11 were the potential targets of miRNA-143, and IL1B of miRNA-21. STAT3 was differentially expressed and regulated 27 potential target miRNAs including miRNA-143 and miRNA-21 in TF-miRNA regulatory network. CONCLUSION In summary, inflammation, immune response and STAT3-mediated miRNA-target genes axis play an important role in PAOD development and progression.
Collapse
Affiliation(s)
| | - Xiaoyan Yang
- Geriatric Department, First People's Hospital of Jingmen City, Jingmen, Hubei Province
| | - Chunming Wang
- Department of Intervention, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | | |
Collapse
|
13
|
Babaev VR, Ding L, Zhang Y, May JM, Ramsey SA, Vickers KC, Linton MF. Loss of 2 Akt (Protein Kinase B) Isoforms in Hematopoietic Cells Diminished Monocyte and Macrophage Survival and Reduces Atherosclerosis in Ldl Receptor-Null Mice. Arterioscler Thromb Vasc Biol 2019; 39:156-169. [PMID: 30567482 DOI: 10.1161/atvbaha.118.312206] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective- Macrophages express 3 Akt (protein kinase B) isoforms, Akt1, Akt2, and Akt3, which display isoform-specific functions but may be redundant in terms of Akt survival signaling. We hypothesize that loss of 2 Akt isoforms in macrophages will suppress their ability to survive and modulate the development of atherosclerosis. Approach and Results- To test this hypothesis, we reconstituted male Ldlr-/- mice with double Akt2/Akt3 knockout hematopoietic cells expressing only the Akt1 isoform (Akt1only). There were no differences in body weight and plasma lipid levels between the groups after 8 weeks of the Western diet; however, Akt1only→ Ldlr-/- mice developed smaller (57.6% reduction) atherosclerotic lesions with more apoptotic macrophages than control mice transplanted with WT (wild type) cells. Next, male and female Ldlr-/- mice were reconstituted with double Akt1/Akt2 knockout hematopoietic cells expressing the Akt3 isoform (Akt3only). Female and male Akt3only→ Ldlr-/- recipients had significantly smaller (61% and 41%, respectively) lesions than the control WT→ Ldlr-/- mice. Loss of 2 Akt isoforms in hematopoietic cells resulted in markedly diminished levels of white blood cells, B cells, and monocytes and compromised viability of monocytes and peritoneal macrophages compared with WT cells. In response to lipopolysaccharides, macrophages with a single Akt isoform expressed low levels of inflammatory cytokines; however, Akt1only macrophages were distinct in expressing high levels of antiapoptotic Il10 compared with WT and Akt3only cells. Conclusions- Loss of 2 Akt isoforms in hematopoietic cells, preserving only a single Akt1 or Akt3 isoform, markedly compromises monocyte and macrophage viability and diminishes early atherosclerosis in Ldlr-/- mice.
Collapse
Affiliation(s)
- Vladimir R Babaev
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN
| | - Lei Ding
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN
| | - Youmin Zhang
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN
| | - James M May
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN.,Department of Molecular Physiology and Biophysics (J.M.M., K.C.V.), Vanderbilt University School of Medicine, Nashville, TN
| | - Stephen A Ramsey
- Department of Biomedical Sciences, Oregon State University, School of Electrical Engineering and Computer Science, Corvallis (S.A.R.)
| | - Kasey C Vickers
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN.,Department of Molecular Physiology and Biophysics (J.M.M., K.C.V.), Vanderbilt University School of Medicine, Nashville, TN
| | - MacRae F Linton
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN.,Department of Pharmacology (M.F.L.), Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
14
|
Abstract
Macrophages play a central role in the development of atherosclerotic cardiovascular disease (ASCVD), which encompasses coronary artery disease, peripheral artery disease, cerebrovascular disease, and aortic atherosclerosis. In each vascular bed, macrophages contribute to the maintenance of the local inflammatory response, propagate plaque development, and promote thrombosis. These central roles, coupled with their plasticity, makes macrophages attractive therapeutic targets in stemming the development of and stabilizing existing atherosclerosis. In the context of ASCVD, classically activated M1 macrophages initiate and sustain inflammation, and alternatively activated M2 macrophages resolve inflammation. However, this classification is now considered an oversimplification, and a greater understanding of plaque macrophage physiology in ASCVD is required to aid in the development of therapeutics to promote ASCVD regression. Reviewed herein are the macrophage phenotypes and molecular regulators characteristic of ASCVD regression, and the current murine models of ASCVD regression.
Collapse
Affiliation(s)
- Tessa J. Barrett
- From the Division of Cardiology, Department of Medicine, New York University
| |
Collapse
|
15
|
Zhang Q, Hu J, Wu Y, Luo H, Meng W, Xiao B, Xiao X, Zhou Z, Liu F. Rheb (Ras Homolog Enriched in Brain 1) Deficiency in Mature Macrophages Prevents Atherosclerosis by Repressing Macrophage Proliferation, Inflammation, and Lipid Uptake. Arterioscler Thromb Vasc Biol 2019; 39:1787-1801. [DOI: 10.1161/atvbaha.119.312870] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective:
Macrophage foam cell formation is an important process in atherosclerotic plaque development. The small GTPase Rheb (Ras homolog enriched in brain 1) regulates endocytic trafficking that is critical for foam cell formation. However, it is unclear whether and how macrophage Rheb regulates atherogenesis, which are the focuses of the current study.
Approach and Results:
Immunofluorescence study confirmed the colocalization of Rheb in F4/80 and Mac-2 (galectin-3)–labeled lesional macrophages. Western blot and fluorescence-activated cell sorting analysis showed that Rheb expression was significantly increased in atherosclerotic lesions of atherosclerosis-prone (apoE
−/−
[apolipoprotein E deficient]) mice fed with Western diet. Increased Rheb expression was also observed in oxidized LDL (low-density lipoprotein)–treated macrophages. To investigate the in vivo role of macrophage Rheb, we established mature Rheb
mKO
(macrophage-specific Rheb knockout) mice by crossing the Rheb floxed mice with
F4/80-cre
mice. Macrophage-specific knockout of Rheb in mice reduced Western diet–induced atherosclerotic lesion by 32%, accompanied with a decrease in macrophage content in plaque. Mechanistically, loss of Rheb in macrophages repressed oxidized LDL–induced lipid uptake, inflammation, and macrophage proliferation. On the contrary, lentivirus-mediated overexpression of Rheb in macrophages increased oxidized LDL–induced lipid uptake and inflammation, and the stimulatory effect of Rheb was suppressed by the mTOR (mammalian target of rapamycin) inhibitor rapamycin or the PKA (protein kinase A) activator forskolin.
Conclusions:
Macrophage Rheb plays important role in Western diet–induced atherosclerosis by promoting macrophage proliferation, inflammation, and lipid uptake. Inhibition of expression and function of Rheb in macrophages is beneficial to prevent diet-induced atherosclerosis.
Collapse
Affiliation(s)
- Qinghai Zhang
- From the Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Q.Z., J.H., Y.W., H.L., W.M., B.X., Z.Z., F.L.)
- Department of Metabolism and Endocrinology, First Affiliated Hospital of University of South China, Hengyang, Hunan (Q.Z.)
| | - Jie Hu
- From the Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Q.Z., J.H., Y.W., H.L., W.M., B.X., Z.Z., F.L.)
| | - Yan Wu
- From the Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Q.Z., J.H., Y.W., H.L., W.M., B.X., Z.Z., F.L.)
| | - Hairong Luo
- From the Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Q.Z., J.H., Y.W., H.L., W.M., B.X., Z.Z., F.L.)
| | - Wen Meng
- From the Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Q.Z., J.H., Y.W., H.L., W.M., B.X., Z.Z., F.L.)
| | - Bo Xiao
- From the Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Q.Z., J.H., Y.W., H.L., W.M., B.X., Z.Z., F.L.)
- Department of Biology, Southern University of Science and Technology, Shenzhen, China (B.X.)
| | - Xianzhong Xiao
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China (X.X.)
| | - Zhiguang Zhou
- From the Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Q.Z., J.H., Y.W., H.L., W.M., B.X., Z.Z., F.L.)
| | - Feng Liu
- From the Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Q.Z., J.H., Y.W., H.L., W.M., B.X., Z.Z., F.L.)
| |
Collapse
|
16
|
de Sousa JR, Da Costa Vasconcelos PF, Quaresma JAS. Functional aspects, phenotypic heterogeneity, and tissue immune response of macrophages in infectious diseases. Infect Drug Resist 2019; 12:2589-2611. [PMID: 31686866 PMCID: PMC6709804 DOI: 10.2147/idr.s208576] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/05/2019] [Indexed: 12/13/2022] Open
Abstract
Macrophages are a functionally heterogeneous group of cells with specialized functions depending not only on their subgroup but also on the function of the organ or tissue in which the cells are located. The concept of macrophage phenotypic heterogeneity has been investigated since the 1980s, and more recent studies have identified a diverse spectrum of phenotypic subpopulations. Several types of macrophages play a central role in the response to infectious agents and, along with other components of the immune system, determine the clinical outcome of major infectious diseases. Here, we review the functions of various macrophage phenotypic subpopulations, the concept of macrophage polarization, and the influence of these cells on the evolution of infections. In addition, we emphasize their role in the immune response in vivo and in situ, as well as the molecular effectors and signaling mechanisms used by these cells. Furthermore, we highlight the mechanisms of immune evasion triggered by infectious agents to counter the actions of macrophages and their consequences. Our aim here is to provide an overview of the role of macrophages in the pathogenesis of critical transmissible diseases and discuss how elucidation of this relationship could enhance our understanding of the host-pathogen association in organ-specific immune responses.
Collapse
Affiliation(s)
- Jorge Rodrigues de Sousa
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
| | - Pedro Fernando Da Costa Vasconcelos
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
| | - Juarez Antonio Simões Quaresma
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
- School of Medicine, São Paulo University, São Paulo, SP, Brazil
| |
Collapse
|
17
|
Asadipooya K, Weinstock A. Cardiovascular Outcomes of Romosozumab and Protective Role of Alendronate. Arterioscler Thromb Vasc Biol 2019; 39:1343-1350. [PMID: 31242037 DOI: 10.1161/atvbaha.119.312371] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Osteoporosis and cardiovascular diseases are major public health issues. Bone and cardiovascular remodeling share multiple biological markers and pathways. Medical intervention, such as using romosozumab, an antisclerostin antibody, improves the clinical outcome of osteoporosis. However, blocking sclerostin leads to Wnt (wingless/integrated) activation and participation in the cardiovascular remodeling process, which could potentially lead to adverse events. Based on the opposing roles of bisphosphonates and the Wnt pathway on endothelial dysfunction, lipid accumulation and calcification of the vessel walls, the combination of romosozumab and bisphosphonates could be a new therapeutic approach to reducing the risks of adverse cardiovascular events in romosozumab receivers. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Kamyar Asadipooya
- From the Division of Endocrinology and Molecular Medicine, Department of Medicine, University of Kentucky, Lexington (K.A.)
| | - Ada Weinstock
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York (A.W.)
| |
Collapse
|
18
|
HTRA1 synergizes with oxidized phospholipids in promoting inflammation and macrophage infiltration essential for ocular VEGF expression. PLoS One 2019; 14:e0216808. [PMID: 31100080 PMCID: PMC6524793 DOI: 10.1371/journal.pone.0216808] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/29/2019] [Indexed: 01/10/2023] Open
Abstract
Understanding oxidative stress and HTRA1 locus in abnormal angiogenesis resulting in wet AMD pathology is an important step in developing a novel therapeutic approach. Using subretinal injection of oxLDL into C57BL/6 mice, we observed a lesion resembling the features of choroidal neovascularization (CNV), including macrophage infiltration, increased VEGF expression, and neovascularization. However, incubating ARPE-19 cells with oxLDL–a carrier of oxidized phospholipids–resulted in increased expression of inflammatory cytokines and chemoattractant proteins that recruited monocytes, but no substantial increase in expression of VEGF. Furthermore, incubation of ARPE-19 with oxLDL induced higher expression of HTRA1, which we showed to synergize with oxLDL in elevating the expression of inflammatory cytokines and chemoattractant factors. To investigate the role of macrophage infiltration on these expression changes, we treated cultured J774 macrophages with oxLDL and applied the conditioned medium onto ARPE-19 cells. This treatment was found to greatly enhance the expression of VEGF in ARPE-19, indicating the necessity of macrophage secretory products to induce increased expression of VEGF in retinal pigment epithelium. Gene expression analysis revealed that oxLDL induced the expression of Wnt3A in macrophages, a key activator of canonical Wnt signaling pathways. In addition, western blot analysis showed that the macrophage conditioned media further enhanced the reduction of phosphorylated β-catenin induced by oxLDL. Lastly, we investigated HTRA1 as a potential target for AMD therapeutics. We demonstrated the ability of anti-HTRA1 antibody in vitro to neutralize the protease activity of HTRA1 and reduce the inflammatory and angiogenic response to oxidative stress. Finally, we validated the neutralizing effect of anti-HTRA1 antibody in vivo by evaluating lesion size and protein expression in a laser-photocoagulation murine model of CNV. We found that the combination of oxLDL and HTRA1 enhanced CNV size, which was reversed by the addition of anti-HTRA1 antibody. This study not only provides preliminary evidence that HTRA1 may be a viable target for AMD therapeutics but also elucidates the biochemical mechanisms by which this therapeutic effect may be mediated.
Collapse
|
19
|
Lin JD, Nishi H, Poles J, Niu X, Mccauley C, Rahman K, Brown EJ, Yeung ST, Vozhilla N, Weinstock A, Ramsey SA, Fisher EA, Loke P. Single-cell analysis of fate-mapped macrophages reveals heterogeneity, including stem-like properties, during atherosclerosis progression and regression. JCI Insight 2019; 4:124574. [PMID: 30830865 PMCID: PMC6478411 DOI: 10.1172/jci.insight.124574] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is a leading cause of death worldwide in industrialized countries. Disease progression and regression are associated with different activation states of macrophages derived from inflammatory monocytes entering the plaques. The features of monocyte-to-macrophage transition and the full spectrum of macrophage activation states during either plaque progression or regression, however, are incompletely established. Here, we use a combination of single-cell RNA sequencing and genetic fate mapping to profile, for the first time to our knowledge, plaque cells derived from CX3CR1+ precursors in mice during both progression and regression of atherosclerosis. The analyses revealed a spectrum of macrophage activation states with greater complexity than the traditional M1 and M2 polarization states, with progression associated with differentiation of CXC3R1+ monocytes into more distinct states than during regression. We also identified an unexpected cluster of proliferating monocytes with a stem cell-like signature, suggesting that monocytes may persist in a proliferating self-renewal state in inflamed tissue, rather than differentiating immediately into macrophages after entering the tissue.
Collapse
Affiliation(s)
| | - Hitoo Nishi
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| | | | - Xiang Niu
- Tri-Institutional Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, New York, USA
| | | | - Karishma Rahman
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Emily J. Brown
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| | | | | | - Ada Weinstock
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Stephen A. Ramsey
- Department of Biomedical Sciences, School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, USA
| | - Edward A. Fisher
- Department of Microbiology and
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| | | |
Collapse
|
20
|
Wang ZQ, Jing LL, Yan JC, Sun Z, Bao ZY, Shao C, Pang QW, Geng Y, Zhang LL, Li LH. Role of AGEs in the progression and regression of atherosclerotic plaques. Glycoconj J 2018; 35:443-450. [PMID: 29987432 DOI: 10.1007/s10719-018-9831-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 06/27/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023]
Abstract
The formation of advanced glycation end-products(AGEs) is an important cause of metabolic memory in diabetic patients and a key factor in the formation of atherosclerosis(AS) plaques in patients with diabetes mellitus. Related studies showed that AGEs could disrupt hemodynamic steady-state and destroy vascular wall integrity through the endothelial barrier damage, foam cell(FC) formation, apoptosis, calcium deposition and other aspects. At the same time, AGEs could initiate oxidative stress and inflammatory response cascade via receptor-depended and non-receptor-dependent pathways, promoting plaques to develop from a steady state to a vulnerable state and eventually tend to rupture and thrombosis. Numerous studies have confirmed that these pathological processes mentioned above could lead to acute coronary heart disease(CHD) and other acute cardiovascular and cerebrovascular events. However, the specific role of AGEs in the progression and regression of AS plaques has not yet been fully elucidated. In this paper, the formation, source, metabolism, physical and chemical properties of AGEs and their role in the migration of FCs and plaque calcification are briefly described, we hope to provide new ideas for the researchers that struggling in this field.
Collapse
Affiliation(s)
- Zhong-Qun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Le-le Jing
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Jin-Chuan Yan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zheng-Yang Bao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Qi-Wen Pang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yue Geng
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Li-Li Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Li-Hua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
21
|
Foulquier S, Daskalopoulos EP, Lluri G, Hermans KCM, Deb A, Blankesteijn WM. WNT Signaling in Cardiac and Vascular Disease. Pharmacol Rev 2018; 70:68-141. [PMID: 29247129 PMCID: PMC6040091 DOI: 10.1124/pr.117.013896] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
WNT signaling is an elaborate and complex collection of signal transduction pathways mediated by multiple signaling molecules. WNT signaling is critically important for developmental processes, including cell proliferation, differentiation and tissue patterning. Little WNT signaling activity is present in the cardiovascular system of healthy adults, but reactivation of the pathway is observed in many pathologies of heart and blood vessels. The high prevalence of these pathologies and their significant contribution to human disease burden has raised interest in WNT signaling as a potential target for therapeutic intervention. In this review, we first will focus on the constituents of the pathway and their regulation and the different signaling routes. Subsequently, the role of WNT signaling in cardiovascular development is addressed, followed by a detailed discussion of its involvement in vascular and cardiac disease. After highlighting the crosstalk between WNT, transforming growth factor-β and angiotensin II signaling, and the emerging role of WNT signaling in the regulation of stem cells, we provide an overview of drugs targeting the pathway at different levels. From the combined studies we conclude that, despite the sometimes conflicting experimental data, a general picture is emerging that excessive stimulation of WNT signaling adversely affects cardiovascular pathology. The rapidly increasing collection of drugs interfering at different levels of WNT signaling will allow the evaluation of therapeutic interventions in the pathway in relevant animal models of cardiovascular diseases and eventually in patients in the near future, translating the outcomes of the many preclinical studies into a clinically relevant context.
Collapse
Affiliation(s)
- Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Evangelos P Daskalopoulos
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Gentian Lluri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Arjun Deb
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| |
Collapse
|
22
|
Identifying novel transcription factors involved in the inflammatory response by using binding site motif scanning in genomic regions defined by histone acetylation. PLoS One 2017; 12:e0184850. [PMID: 28922390 PMCID: PMC5602638 DOI: 10.1371/journal.pone.0184850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/31/2017] [Indexed: 02/07/2023] Open
Abstract
The innate immune response to pathogenic challenge is a complex, multi-staged process involving thousands of genes. While numerous transcription factors that act as master regulators of this response have been identified, the temporal complexity of gene expression changes in response to pathogen-associated molecular pattern receptor stimulation strongly suggest that additional layers of regulation remain to be uncovered. The evolved pathogen response program in mammalian innate immune cells is understood to reflect a compromise between the probability of clearing the infection and the extent of tissue damage and inflammatory sequelae it causes. Because of that, a key challenge to delineating the regulators that control the temporal inflammatory response is that an innate immune regulator that may confer a selective advantage in the wild may be dispensable in the lab setting. In order to better understand the complete transcriptional response of primary macrophages to the bacterial endotoxin lipopolysaccharide (LPS), we designed a method that integrates temporally resolved gene expression and chromatin-accessibility measurements from mouse macrophages. By correlating changes in transcription factor binding site motif enrichment scores, calculated within regions of accessible chromatin, with the average temporal expression profile of a gene cluster, we screened for transcriptional factors that regulate the cluster. We have validated our predictions of LPS-stimulated transcriptional regulators using ChIP-seq data for three transcription factors with experimentally confirmed functions in innate immunity. In addition, we predict a role in the macrophage LPS response for several novel transcription factors that have not previously been implicated in immune responses. This method is applicable to any experimental situation where temporal gene expression and chromatin-accessibility data are available.
Collapse
|
23
|
Daugherty A, Tall AR, Daemen MJ, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Circ Res 2017; 121:e53-e79. [DOI: 10.1161/res.0000000000000169] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
24
|
Xu S. Transcriptome Profiling in Systems Vascular Medicine. Front Pharmacol 2017; 8:563. [PMID: 28970795 PMCID: PMC5609594 DOI: 10.3389/fphar.2017.00563] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/08/2017] [Indexed: 02/06/2023] Open
Abstract
In the post-genomic, big data era, our understanding of vascular diseases has been deepened by multiple state-of-the-art “–omics” approaches, including genomics, epigenomics, transcriptomics, proteomics, lipidomics and metabolomics. Genome-wide transcriptomic profiling, such as gene microarray and RNA-sequencing, emerges as powerful research tools in systems medicine and revolutionizes transcriptomic analysis of the pathological mechanisms and therapeutics of vascular diseases. In this article, I will highlight the workflow of transcriptomic profiling, outline basic bioinformatics analysis, and summarize recent gene profiling studies performed in vascular cells as well as in human and mice diseased samples. Further mining of these public repository datasets will shed new light on our understanding of the cellular basis of vascular diseases and offer novel potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Suowen Xu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, RochesterNY, United States
| |
Collapse
|
25
|
The transcription factor MafB promotes anti-inflammatory M2 polarization and cholesterol efflux in macrophages. Sci Rep 2017; 7:7591. [PMID: 28790455 PMCID: PMC5548719 DOI: 10.1038/s41598-017-07381-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 06/28/2017] [Indexed: 12/21/2022] Open
Abstract
Macrophages play pivotal roles in the progression and regression of atherosclerosis. Accumulating evidence suggests that macrophage polarization into an anti-inflammatory M2 state is a key characteristic of atherosclerotic plaques undergoing regression. However, the molecular mechanisms underlying this potential association of the M2 polarization with atherosclerosis regression remain poorly understood. Further, human genetic factors that facilitate these anti-atherogenic processes remain largely unknown. We report that the transcription factor MafB plays pivotal roles in promoting macrophage M2 polarization. Further, MafB promotes cholesterol efflux from macrophage foam cells by directly up-regulating its key cellular mediators. Notably, MafB expression is significantly up-regulated in response to various metabolic and immunological stimuli that promote macrophage M2 polarization or cholesterol efflux, and thereby MafB mediates their beneficial effects, in both liver x receptor (LXR)-dependent and independent manners. In contrast, MafB is strongly down-regulated upon elevated pro-inflammatory signaling or by pro-inflammatory and pro-atherogenic microRNAs, miR-155 and miR-33. Using an integrative systems biology approach, we also revealed that M2 polarization and cholesterol efflux do not necessarily represent inter-dependent events, but MafB is broadly involved in both the processes. These findings highlight physiological protective roles that MafB may play against atherosclerosis progression.
Collapse
|
26
|
Daugherty A, Tall AR, Daemen MJAP, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2017; 37:e131-e157. [PMID: 28729366 DOI: 10.1161/atv.0000000000000062] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
27
|
von Scheidt M, Zhao Y, Kurt Z, Pan C, Zeng L, Yang X, Schunkert H, Lusis AJ. Applications and Limitations of Mouse Models for Understanding Human Atherosclerosis. Cell Metab 2017; 25:248-261. [PMID: 27916529 PMCID: PMC5484632 DOI: 10.1016/j.cmet.2016.11.001] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/26/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
Abstract
Most of the biological understanding of mechanisms underlying coronary artery disease (CAD) derives from studies of mouse models. The identification of multiple CAD loci and strong candidate genes in large human genome-wide association studies (GWASs) presented an opportunity to examine the relevance of mouse models for the human disease. We comprehensively reviewed the mouse literature, including 827 literature-derived genes, and compared it to human data. First, we observed striking concordance of risk factors for atherosclerosis in mice and humans. Second, there was highly significant overlap of mouse genes with human genes identified by GWASs. In particular, of the 46 genes with strong association signals in CAD GWASs that were studied in mouse models, all but one exhibited consistent effects on atherosclerosis-related phenotypes. Third, we compared 178 CAD-associated pathways derived from human GWASs with 263 from mouse studies and observed that the majority were consistent between the species.
Collapse
Affiliation(s)
- Moritz von Scheidt
- Deutsches Herzzentrum München, Technische Universität München, 80333 Munich, Germany
| | - Yuqi Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zeyneb Kurt
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Calvin Pan
- Departments of Medicine, Microbiology, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lingyao Zeng
- Deutsches Herzzentrum München, Technische Universität München, 80333 Munich, Germany
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, 80333 Munich, Germany; Deutsches Zentrum für Herz- und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Aldons J Lusis
- Departments of Medicine, Microbiology, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
28
|
Differentially expressed genes and canonical pathway expression in human atherosclerotic plaques - Tampere Vascular Study. Sci Rep 2017; 7:41483. [PMID: 28128285 PMCID: PMC5270243 DOI: 10.1038/srep41483] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 12/21/2016] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases due to atherosclerosis are the leading cause of death globally. We aimed to investigate the potentially altered gene and pathway expression in advanced peripheral atherosclerotic plaques in comparison to healthy control arteries. Gene expression analysis was performed (Illumina HumanHT-12 version 3 Expression BeadChip) for 68 advanced atherosclerotic plaques (15 aortic, 29 carotid and 24 femoral plaques) and 28 controls (left internal thoracic artery (LITA)) from Tampere Vascular Study. Dysregulation of individual genes was compared to healthy controls and between plaques from different arterial beds and Ingenuity pathway analysis was conducted on genes with a fold change (FC) > ±1.5 and false discovery rate (FDR) < 0.05. 787 genes were significantly differentially expressed in atherosclerotic plaques. The most up-regulated genes were osteopontin and multiple MMPs, and the most down-regulated were cell death-inducing DFFA-like effector C and A (CIDEC, CIDEA) and apolipoprotein D (FC > 20). 156 pathways were differentially expressed in atherosclerotic plaques, mostly inflammation-related, especially related with leukocyte trafficking and signaling. In artery specific plaque analysis 50.4% of canonical pathways and 41.2% GO terms differentially expressed were in common for all three arterial beds. Our results confirm the inflammatory nature of advanced atherosclerosis and show novel pathway differences between different arterial beds.
Collapse
|
29
|
Ramsey SA. An Empirical Prior Improves Accuracy for Bayesian Estimation of Transcription Factor Binding Site Frequencies within Gene Promoters. Bioinform Biol Insights 2016; 9:59-69. [PMID: 27812284 PMCID: PMC5081247 DOI: 10.4137/bbi.s29330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/11/2016] [Accepted: 09/18/2016] [Indexed: 12/24/2022] Open
Abstract
A Bayesian method for sampling from the distribution of matches to a precompiled transcription factor binding site (TFBS) sequence pattern (conditioned on an observed nucleotide sequence and the sequence pattern) is described. The method takes a position frequency matrix as input for a set of representative binding sites for a transcription factor and two sets of noncoding, 5′ regulatory sequences for gene sets that are to be compared. An empirical prior on the frequency A (per base pair of gene-vicinal, noncoding DNA) of TFBSs is developed using data from the ENCODE project and incorporated into the method. In addition, a probabilistic model for binding site occurrences conditioned on λ is developed analytically, taking into account the finite-width effects of binding sites. The count of TFBS β (conditioned on the observed sequence) is sampled using Metropolis–Hastings with an information entropy-based move generator. The derivation of the method is presented in a step-by-step fashion, starting from specific conditional independence assumptions. Empirical results show that the newly proposed prior on β improves accuracy for estimating the number of TFBS within a set of promoter sequences.
Collapse
Affiliation(s)
- Stephen A Ramsey
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA.; School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
30
|
Escate R, Padro T, Borrell-Pages M, Suades R, Aledo R, Mata P, Badimon L. Macrophages of genetically characterized familial hypercholesterolaemia patients show up-regulation of LDL-receptor-related proteins. J Cell Mol Med 2016; 21:487-499. [PMID: 27680891 PMCID: PMC5323824 DOI: 10.1111/jcmm.12993] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/18/2016] [Indexed: 12/28/2022] Open
Abstract
Familial hypercholesterolaemia (FH) is a major risk for premature coronary heart disease due to severe long‐life exposure to high LDL levels. Accumulation of LDL in the vascular wall triggers atherosclerosis with activation of the innate immunity system. Here, we have investigated (i) gene expression of LDLR and LRPs in peripheral blood cells (PBLs) and in differentiated macrophages of young FH‐patients; and (ii) whether macrophage from FH patients have a differential response when exposed to high levels of atherogenic LDL. PBLs in young heterozygous genetically characterized FH patients have higher expression of LRP5 and LRP6 than age‐matched healthy controls or patients with secondary hypercholesterolaemia. LRP1 levels were similar among groups. In monocyte‐derived macrophages (MACs), LRP5 and LRP1 transcript levels did not differ between FHs and controls in resting conditions, but when exposed to agLDL, FH‐MAC showed a highly significant up‐regulation of LRP5, while LRP1 was unaffected. PBL and MAC cells from FH patients had significantly lower LDLR expression than control cells, independently of the lipid‐lowering therapy. Furthermore, exposure of FH‐MAC to agLDL resulted in a reduced expression of CD163, scavenger receptor with anti‐inflammatory and atheroprotective properties. In summary, our results show for first time that LRPs, active lipid‐internalizing receptors, are up‐regulated in innate immunity cells of young FH patients that have functional LDLR mutations. Additionally, their reduced CD163 expression indicates less atheroprotection. Both mechanisms may play a synergic effect on the onset of premature atherosclerosis in FH patients.
Collapse
Affiliation(s)
- Rafael Escate
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | | | - Rosa Suades
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Rosa Aledo
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | | | - Lina Badimon
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| |
Collapse
|
31
|
Patzig J, Erwig MS, Tenzer S, Kusch K, Dibaj P, Möbius W, Goebbels S, Schaeren-Wiemers N, Nave KA, Werner HB. Septin/anillin filaments scaffold central nervous system myelin to accelerate nerve conduction. eLife 2016; 5. [PMID: 27504968 PMCID: PMC4978525 DOI: 10.7554/elife.17119] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/13/2016] [Indexed: 12/15/2022] Open
Abstract
Myelination of axons facilitates rapid impulse propagation in the nervous system. The axon/myelin-unit becomes impaired in myelin-related disorders and upon normal aging. However, the molecular cause of many pathological features, including the frequently observed myelin outfoldings, remained unknown. Using label-free quantitative proteomics, we find that the presence of myelin outfoldings correlates with a loss of cytoskeletal septins in myelin. Regulated by phosphatidylinositol-(4,5)-bisphosphate (PI(4,5)P2)-levels, myelin septins (SEPT2/SEPT4/SEPT7/SEPT8) and the PI(4,5)P2-adaptor anillin form previously unrecognized filaments that extend longitudinally along myelinated axons. By confocal microscopy and immunogold-electron microscopy, these filaments are localized to the non-compacted adaxonal myelin compartment. Genetic disruption of these filaments in Sept8-mutant mice causes myelin outfoldings as a very specific neuropathology. Septin filaments thus serve an important function in scaffolding the axon/myelin-unit, evidently a late stage of myelin maturation. We propose that pathological or aging-associated diminishment of the septin/anillin-scaffold causes myelin outfoldings that impair the normal nerve conduction velocity. DOI:http://dx.doi.org/10.7554/eLife.17119.001 Normal communication within the brain or between the brain and other parts of the body requires information to flow quickly around the nervous system. This information travels along nerve cells in the form of electrical signals. To speed up the signals, a part of the nerve cell called the axon is frequently wrapped in an electrically insulating sheath made up of a membrane structure called myelin. The myelin sheath becomes impaired as a result of disease or ageing. In order to understand what might produce these changes, Patzig et al. have used biochemical and microscopy techniques to study mice that had similar defects in their myelin sheaths. The study reveals that forming a normal myelin sheath around an axon requires a newly identified ‘scaffold’ made of a group of proteins called the septins. Combining with another protein called anillin, septins assemble into filaments in the myelin sheath. These filaments then knit together into a scaffold that grows lengthways along the myelin-wrapped axon. Without this scaffold, the myelin sheath grew defects known as outfoldings. Axons transmitted electrical signals much more slowly than normal when the septin scaffold was missing from the myelin sheath. Future studies are needed to understand the factors that control how the septin scaffold forms. This could help to reveal ways of reversing the changes that alter the myelin sheath during ageing and disease. DOI:http://dx.doi.org/10.7554/eLife.17119.002
Collapse
Affiliation(s)
- Julia Patzig
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Michelle S Erwig
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Stefan Tenzer
- Institute of Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Payam Dibaj
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | | | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| |
Collapse
|
32
|
Dong X, Yambartsev A, Ramsey SA, Thomas LD, Shulzhenko N, Morgun A. Reverse enGENEering of Regulatory Networks from Big Data: A Roadmap for Biologists. Bioinform Biol Insights 2015; 9:61-74. [PMID: 25983554 PMCID: PMC4415676 DOI: 10.4137/bbi.s12467] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 12/29/2022] Open
Abstract
Omics technologies enable unbiased investigation of biological systems through massively parallel sequence acquisition or molecular measurements, bringing the life sciences into the era of Big Data. A central challenge posed by such omics datasets is how to transform these data into biological knowledge, for example, how to use these data to answer questions such as: Which functional pathways are involved in cell differentiation? Which genes should we target to stop cancer? Network analysis is a powerful and general approach to solve this problem consisting of two fundamental stages, network reconstruction, and network interrogation. Here we provide an overview of network analysis including a step-by-step guide on how to perform and use this approach to investigate a biological question. In this guide, we also include the software packages that we and others employ for each of the steps of a network analysis workflow.
Collapse
Affiliation(s)
- Xiaoxi Dong
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Anatoly Yambartsev
- Department of Statistics, Institute of Mathematics and Statistics, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Stephen A Ramsey
- School of Electrical Engineering and Computer Science, Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA. ; College of Veterinary Medicine, Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| | - Lina D Thomas
- Department of Statistics, Institute of Mathematics and Statistics, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Natalia Shulzhenko
- College of Veterinary Medicine, Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| |
Collapse
|