101
|
Biological profile of monocyte-derived macrophages in coronary heart disease patients: implications for plaque morphology. Sci Rep 2019; 9:8680. [PMID: 31213640 PMCID: PMC6581961 DOI: 10.1038/s41598-019-44847-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 05/24/2019] [Indexed: 02/06/2023] Open
Abstract
The prevalence of a macrophage phenotype in atherosclerotic plaque may drive its progression and/or instability. Macrophages from coronary plaques are not available, and monocyte-derived macrophages (MDMs) are usually considered as a surrogate. We compared the MDM profile obtained from coronary artery disease (CAD) patients and healthy subjects, and we evaluated the association between CAD MDM profile and in vivo coronary plaque characteristics assessed by optical coherence tomography (OCT). At morphological analysis, MDMs of CAD patients had a higher prevalence of round than spindle cells, whereas in healthy subjects the prevalence of the two morphotypes was similar. Compared to healthy subjects, MDMs of CAD patients had reduced efferocytosis, lower transglutaminase-2, CD206 and CD163 receptor levels, and higher tissue factor (TF) levels. At OCT, patients with a higher prevalence of round MDMs showed more frequently a lipid-rich plaque, a thin-cap fibroatheroma, a greater intra-plaque macrophage accumulation, and a ruptured plaque. The MDM efferocytosis correlated with minimal lumen area, and TF levels in MDMs correlated with the presence of ruptured plaque. MDMs obtained from CAD patients are characterized by a morpho-phenotypic heterogeneity with a prevalence of round cells, showing pro-inflammatory and pro-thrombotic properties. The MDM profile allows identifying CAD patients at high risk.
Collapse
|
102
|
Petrescu GED, Sabo AA, Torsin LI, Calin GA, Dragomir MP. MicroRNA based theranostics for brain cancer: basic principles. J Exp Clin Cancer Res 2019; 38:231. [PMID: 31142339 PMCID: PMC6542029 DOI: 10.1186/s13046-019-1180-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Because of the complexity of the blood-brain barrier (BBB), brain tumors, especially the most common and aggressive primary malignant tumor type arising from the central nervous system (CNS), glioblastoma, remain an essential challenge regarding diagnostic and treatment. There are no approved circulating diagnostic or prognostic biomarkers, nor novel therapies like immune checkpoint inhibitors for glioblastoma, and chemotherapy brings only minimal survival benefits. The development of molecular biology led to the discovery of new potential diagnostic tools and therapeutic targets, offering the premise to detect patients at earlier stages and overcome the current poor prognosis. MAIN BODY One potential diagnostic and therapeutic breakthrough might come from microRNAs (miRNAs). It is well-known that miRNAs play a role in the initiation and development of various types of cancer, including glioblastoma. The review aims to answer the following questions concerning the role of RNA theranostics for brain tumors: (1) which miRNAs are the best candidates to become early diagnostic and prognostic circulating biomarkers?; (2) how to deliver the therapeutic agents in the CNS to overcome the BBB?; (3) which are the best methods to restore/inhibit miRNAs? CONCLUSIONS Because of the proven roles played by miRNAs in gliomagenesis and of their capacity to pass from the CNS tissue into the blood or cerebrospinal fluid (CSF), we propose miRNAs as ideal diagnostic and prognostic biomarkers. Moreover, recent advances in direct miRNA restoration (miRNA mimics) and miRNA inhibition therapy (antisense oligonucleotides, antagomirs, locked nucleic acid anti-miRNA, small molecule miRNA inhibitors) make miRNAs perfect candidates for entering clinical trials for glioblastoma treatment.
Collapse
Affiliation(s)
- George E. D. Petrescu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Bagdasar-Arseni Clinical Emergency Hospital, Department of Neurosurgery, Bucharest, Romania
| | - Alexandru A. Sabo
- Marie Curie Emergency Clinical Hospital for Children, Bucharest, Romania
| | - Ligia I. Torsin
- Elias Clinical Emergency Hospital, Anaesthesiology and Critical Care Department, Bucharest, Romania
| | - George A. Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Mihnea P. Dragomir
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
103
|
Perspectives on the role of brain cellular players in cancer-associated brain metastasis: translational approach to understand molecular mechanism of tumor progression. Cancer Metastasis Rev 2019; 37:791-804. [PMID: 30284650 DOI: 10.1007/s10555-018-9766-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Brain metastasis is one of the leading causes of death among cancer patients. Cancer cells migrate to various sites and harbor different niche in the body which help cancer cells in their survival. The brain is one of the safest place where cancer cells are protected from immune cells. Breast, lung, and melanoma cancer cells have high propensity to migrate towards the brain. To enter the brain, cancer cells have to cross the blood brain barrier. Survival and finding new niche in the brain are directed by several mechanisms in which different cellular players take part such as astrocytes, microglia, Schwann cells, satellite cells, oligodendrocytes, and ependymal cells. Usually, cancer cells highjack the machinery of brain cellular players to survive in the brain environment. It has been shown that co-culture of M2 macrophage with cancer cells leads to increased proliferation and survival of cancer cells. One of the challenges of understanding brain metastasis is appropriate model system to understand dynamic interaction of cancer cells and brain cellular players. To meet this challenge, microfluidic-based devices are employed which can mimic the dynamic conditions as well as can be used for culturing human cells for personalized therapy. In this review, we have systematically reviewed the current status of the role of cellular players in brain metastasis along with explaining how translational approach of microfluidics can be employed for finding new drug target as well as biomarker for brain metastasis. Finally, we have also commented on the mechanism of action of drugs against brain metastasis.
Collapse
|
104
|
Roth Flach RJ, Su C, Bollinger E, Cortes C, Robertson AW, Opsahl AC, Coskran TM, Maresca KP, Keliher EJ, Yates PD, Kim AM, Kalgutkar AS, Buckbinder L. Myeloperoxidase inhibition in mice alters atherosclerotic lesion composition. PLoS One 2019; 14:e0214150. [PMID: 30889221 PMCID: PMC6424399 DOI: 10.1371/journal.pone.0214150] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/07/2019] [Indexed: 01/09/2023] Open
Abstract
Myeloperoxidase (MPO) is a highly abundant protein within the neutrophil that is associated with lipoprotein oxidation, and increased plasma MPO levels are correlated with poor prognosis after myocardial infarct. Thus, MPO inhibitors have been developed for the treatment of heart failure and acute coronary syndrome in humans. 2-(6-(5-Chloro-2-methoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2H)-yl)acetamide PF-06282999 is a recently described selective small molecule mechanism-based inactivator of MPO. Here, utilizing PF-06282999, we investigated the role of MPO to regulate atherosclerotic lesion formation and composition in the Ldlr-/- mouse model of atherosclerosis. Though MPO inhibition did not affect lesion area in Ldlr-/- mice fed a Western diet, reduced necrotic core area was observed in aortic root sections after MPO inhibitor treatment. MPO inhibition did not alter macrophage content in and leukocyte homing to atherosclerotic plaques. To assess non-invasive monitoring of plaque inflammation, [18F]-Fluoro-deoxy-glucose (FDG) was administered to Ldlr-/- mice with established atherosclerosis that had been treated with clinically relevant doses of PF-06282999, and reduced FDG signal was observed in animals treated with a dose of PF-06282999 that corresponded with reduced necrotic core area. These data suggest that MPO inhibition does not alter atherosclerotic plaque area or leukocyte homing, but rather alters the inflammatory tone of atherosclerotic lesions; thus, MPO inhibition could have utility to promote atherosclerotic lesion stabilization and prevent atherosclerotic plaque rupture.
Collapse
Affiliation(s)
- Rachel J. Roth Flach
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, Massachusetts, United States of America
- * E-mail:
| | - Chunyan Su
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, Massachusetts, United States of America
| | - Eliza Bollinger
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, Massachusetts, United States of America
| | - Christian Cortes
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, Massachusetts, United States of America
| | - Andrew W. Robertson
- Drug Safety Research and Development Global Pathology, Pfizer Inc., Groton, Connecticut, United States of America
| | - Alan C. Opsahl
- Drug Safety Research and Development Global Pathology, Pfizer Inc., Groton, Connecticut, United States of America
| | - Timothy M. Coskran
- Drug Safety Research and Development Global Pathology, Pfizer Inc., Groton, Connecticut, United States of America
| | - Kevin P. Maresca
- Early Clinical Development, Pfizer Inc., Cambridge, Massachusetts, United States of America
| | - Edmund J. Keliher
- Early Clinical Development, Pfizer Inc., Cambridge, Massachusetts, United States of America
| | - Phillip D. Yates
- Early Clinical Development, Pfizer Inc., Cambridge, Massachusetts, United States of America
| | - Albert M. Kim
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, Massachusetts, United States of America
| | - Amit S. Kalgutkar
- Medicine Design, Pfizer Inc., Cambridge, Massachusetts, United States of America
| | - Leonard Buckbinder
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, Massachusetts, United States of America
| |
Collapse
|
105
|
Anti-atherosclerotic action of GW9508 - Free fatty acid receptors activator - In apoE-knockout mice. Pharmacol Rep 2019; 71:551-555. [PMID: 31129318 DOI: 10.1016/j.pharep.2019.02.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND In the past two decades, enhanced understanding of the biology of G-protein-coupled receptors (GPRs) has led to the identification of several such receptors as novel targets for free fatty acids (FFAs). Two GPRs, FFAR1 and FFAR4, have received special attention in the context of chronic inflammatory diseases, thanks to their anti-inflammatory activities. METHODS The present study investigates the influence of prolonged treatment with GW9508 - agonist of FFAR1 and FFAR4 - on the development of atherosclerosis plaque in apoE-knockout mice, using morphometric and molecular methods. RESULTS GW9508 administration has led to the reduction of atheroscletoric plaque size in an apoE-knockout mice model. Moreover, a FFAR1/FFAR4 agonist reduced the content of macrophages by almost 20%, attributed by immunohistochemical phenotyping to the pro-inflammatory M1-like activation state macrophages. CONCLUSIONS Prolonged administration of GW9508 resulted in significant amelioration of atherogenesis, providing evidence that the strategy based on macrophage phenotype switching toward an M2-like activation state via stimulation of FFAR1/FFAR4 receptors holds promise for a new approach to the prevention or treatment of atherosclerosis.
Collapse
|
106
|
Liu Y, Wang X, Pang J, Zhang H, Luo J, Qian X, Chen Q, Ling W. Attenuation of Atherosclerosis by Protocatechuic Acid via Inhibition of M1 and Promotion of M2 Macrophage Polarization. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:807-818. [PMID: 30592218 DOI: 10.1021/acs.jafc.8b05719] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Macrophage polarization has a vital impact on the progression of atherosclerosis (AS). Protocatechuic acid (PCA), a flavonol, displays notable atheroprotective effects, but its mechanisms have not been clearly defined. We investigated whether PCA attenuated AS by regulating macrophage polarization. PCA consumption inhibited HCD-induced plaque formation (17.84 and 8.21% in the HCD and HCD with PCA groups, respectively, p < 0.05) and inflammatory responses in apolipoprotein E deficient (ApoE-/-) mice. Moreover, PCA suppressed classically activated macrophage (M1) polarization, which decreased the secretion of nitric oxide synthase (54.63 and 32.86% in the HCD and HCD with PCA groups, respectively, p < 0.05) and proinflammatory factors. PCA promoted alternatively activated macrophage (M2) activation, which increased the expression of arginine I (6.97 and 26.19% in the HCD and HCD with PCA groups, respectively, p < 0.001) and anti-inflammatory factors. PCA also regulated M1-M2 polarization in J774 cells and mouse-bone-marrow-derived macrophages. Finally, PCA reduced PI3K-Akt-mediated nuclear-factor-κB activation, thereby suppressing M1 polarization, and provoked signal-transducers-and-activators-of-transcription-6 phosphorylation and peroxisome-proliferator-activated-receptor-γ activation, leading to enhanced M2 activation. Our data revealed that PCA alleviated AS by regulating M1-M2 conversion.
Collapse
Affiliation(s)
- Yao Liu
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Xu Wang
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Juan Pang
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Hanyue Zhang
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Jing Luo
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Xiaoyun Qian
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Qian Chen
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
- Guangdong Engineering Technology Center of Nutrition Transformation , Guangzhou 510080 , PR China
| |
Collapse
|
107
|
Orecchioni M, Ghosheh Y, Pramod AB, Ley K. Macrophage Polarization: Different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. Alternatively Activated Macrophages. Front Immunol 2019; 10:1084. [PMID: 31178859 PMCID: PMC6543837 DOI: 10.3389/fimmu.2019.01084] [Citation(s) in RCA: 1333] [Impact Index Per Article: 222.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/29/2019] [Indexed: 12/30/2022] Open
Abstract
Macrophages are found in tissues, body cavities, and mucosal surfaces. Most tissue macrophages are seeded in the early embryo before definitive hematopoiesis is established. Others are derived from blood monocytes. The macrophage lineage diversification and plasticity are key aspects of their functionality. Macrophages can also be generated from monocytes in vitro and undergo classical (LPS+IFN-γ) or alternative (IL-4) activation. In vivo, macrophages with different polarization and different activation markers coexist in tissues. Certain mouse strains preferentially promote T-helper-1 (Th1) responses and others Th2 responses. Their macrophages preferentially induce iNOS or arginase and have been called M1 and M2, respectively. In many publications, M1 and classically activated and M2 and alternatively activated are used interchangeably. We tested whether this is justified by comparing the gene lists positively [M1(=LPS+)] or negatively [M2(=LPS-)] correlated with the ratio of IL-12 and arginase 1 in transcriptomes of LPS-treated peritoneal macrophages with in vitro classically (LPS, IFN-γ) vs. alternatively activated (IL-4) bone marrow derived macrophages, both from published datasets. Although there is some overlap between in vivo M1(=LPS+) and in vitro classically activated (LPS+IFN-γ) and in vivo M2(=LPS-) and in vitro alternatively activated macrophages, many more genes are regulated in opposite or unrelated ways. Thus, M1(=LPS+) macrophages are not equivalent to classically activated, and M2(=LPS-) macrophages are not equivalent to alternatively activated macrophages. This fundamental discrepancy explains why most surface markers identified on in vitro generated macrophages do not translate to the in vivo situation. Valid in vivo M1/M2 surface markers remain to be discovered.
Collapse
Affiliation(s)
- Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Yanal Ghosheh
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Akula Bala Pramod
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- *Correspondence: Klaus Ley
| |
Collapse
|
108
|
Labonte AC, Kegerreis B, Geraci NS, Bachali P, Madamanchi S, Robl R, Catalina MD, Lipsky PE, Grammer AC. Identification of alterations in macrophage activation associated with disease activity in systemic lupus erythematosus. PLoS One 2018; 13:e0208132. [PMID: 30562343 PMCID: PMC6298676 DOI: 10.1371/journal.pone.0208132] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by abnormalities in B cell and T cell function, but the role of disturbances in the activation status of macrophages (Mϕ) has not been well described in human patients. To address this, gene expression profiles from isolated lymphoid and myeloid populations were analyzed to identify differentially expressed (DE) genes between healthy controls and patients with either inactive or active SLE. While hundreds of DE genes were identified in B and T cells of active SLE patients, there were no DE genes found in B or T cells from patients with inactive SLE compared to healthy controls. In contrast, large numbers of DE genes were found in myeloid cells (MC) from both active and inactive SLE patients. Among the DE genes were several known to play roles in Mϕ activation and polarization, including the M1 genes STAT1 and SOCS3 and the M2 genes STAT3, STAT6, and CD163. M1-associated genes were far more frequent in data sets from active versus inactive SLE patients. To characterize the relationship between Mϕ activation and disease activity in greater detail, weighted gene co-expression network analysis (WGCNA) was used to identify modules of genes associated with clinical activity in SLE patients. Among these were disease activity-correlated modules containing activation signatures of predominantly M1-associated genes. No disease activity-correlated modules were enriched in M2-associated genes. Pathway and upstream regulator analysis of DE genes from both active and inactive SLE MC were cross-referenced with high-scoring hits from the drug discovery Library of Integrated Network-based Cellular Signatures (LINCS) to identify new strategies to treat both stages of SLE. A machine learning approach employing MC gene modules and a generalized linear model was able to predict the disease activity status in unrelated gene expression data sets. In summary, altered MC gene expression is characteristic of both active and inactive SLE. However, disease activity is associated with an alteration in the activation of MC, with a bias toward the M1 proinflammatory phenotype. These data suggest that while hyperactivity of B cells and T cells is associated with active SLE, MC potentially direct flare-ups and remission by altering their activation status toward the M1 state.
Collapse
Affiliation(s)
- Adam C. Labonte
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Brian Kegerreis
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Nicholas S. Geraci
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Prathyusha Bachali
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
| | - Sushma Madamanchi
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Robert Robl
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Michelle D. Catalina
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Peter E. Lipsky
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Amrie C. Grammer
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
109
|
Cervadoro A, Palomba R, Vergaro G, Cecchi R, Menichetti L, Decuzzi P, Emdin M, Luin S. Targeting Inflammation With Nanosized Drug Delivery Platforms in Cardiovascular Diseases: Immune Cell Modulation in Atherosclerosis. Front Bioeng Biotechnol 2018; 6:177. [PMID: 30542650 PMCID: PMC6277804 DOI: 10.3389/fbioe.2018.00177] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis (AS) is a disorder of large and medium-sized arteries; it consists in the formation of lipid-rich plaques in the intima and inner media, whose pathophysiology is mostly driven by inflammation. Currently available interventions and therapies for treating atherosclerosis are not always completely effective; side effects associated with treatments, mainly caused by immunodepression for anti-inflammatory molecules, limit the systemic administration of these and other drugs. Given the high degree of freedom in the design of nanoconstructs, in the last decades researchers have put high effort in the development of nanoparticles (NPs) formulations specifically designed for either drug delivery, visualization of atherosclerotic plaques, or possibly the combination of both these and other functionalities. Here we will present the state of the art of these subjects, the knowledge of which is necessary to rationally address the use of NPs for prevention, diagnosis, and/or treatment of AS. We will analyse the work that has been done on: (a) understanding the role of the immune system and inflammation in cardiovascular diseases, (b) the pathological and biochemical principles in atherosclerotic plaque formation, (c) the latest advances in the use of NPs for the recognition and treatment of cardiovascular diseases, (d) the cellular and animal models useful to study the interactions of NPs with the immune system cells.
Collapse
Affiliation(s)
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Giuseppe Vergaro
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Roberta Cecchi
- NEST Laboratory, Scuola Normale Superiore, Pisa, Italy.,Center for Nanotechnology Innovation (CNI@NEST), Istituto Italiano di Tecnologia, Pisa, Italy
| | | | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Michele Emdin
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Stefano Luin
- NEST Laboratory, Scuola Normale Superiore, Pisa, Italy.,NEST Laboratory, Istituto Nanoscienze, CNR, Pisa, Italy
| |
Collapse
|
110
|
Mueller PA, Zhu L, Tavori H, Huynh K, Giunzioni I, Stafford JM, Linton MF, Fazio S. Deletion of Macrophage Low-Density Lipoprotein Receptor-Related Protein 1 (LRP1) Accelerates Atherosclerosis Regression and Increases C-C Chemokine Receptor Type 7 (CCR7) Expression in Plaque Macrophages. Circulation 2018; 138:1850-1863. [PMID: 29794082 PMCID: PMC6343494 DOI: 10.1161/circulationaha.117.031702] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 05/14/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND We previously showed that mice lacking MΦLRP1-/- (low-density lipoprotein receptor-related protein 1 in macrophages) undergo accelerated atherosclerotic plaque formation due to changes in macrophages including increased apoptosis, decreased efferocytosis, and exaggerated transition to the inflammatory M1 phenotype. Here we sought to explore the role of macrophage low-density lipoprotein receptor-related protein 1 during regression of atherosclerosis since regressing plaques are characterized by transitioning of macrophages to M2 status as inflammation resolves. METHODS Apolipoprotein E-/- mice on a high-fat diet for 12 weeks were reconstituted with bone marrow from apolipoprotein E-producing wild-type or MΦLRP1-/- mice, and then placed on a chow diet for 10 weeks (n=9 to 11 mice/group). A cohort of apolipoprotein E-/- mice reconstituted with apolipoprotein E-/- bone marrow served as baseline controls (n=9). RESULTS Plaques of both wild-type and MΦLRP1-/- bone marrow recipients regressed compared with controls (11% and 22%, respectively; P<0.05), and plaques of MΦLRP1-/- recipients were 13% smaller than those of wild-type recipients ( P<0.05). Recipients of MΦLRP1-/- marrow had 36% fewer M1 macrophages ( P<0.01) and 2.5-fold more CCR7 (C-C chemokine receptor type 7)-positive macrophages in the plaque relative to wild-type mice ( P<0.01). Additionally, in vivo studies of cellular egress showed a 4.6-fold increase in 5-ethynyl-2´-deoxyuridine-labeled CCR7+ macrophages in mediastinal lymph nodes. Finally, in vivo studies of reverse cholesterol transport showed a 1.4-fold higher reverse cholesterol transport in MΦLRP1-/- recipient mice ( P<0.01). CONCLUSIONS Absence of macrophage low-density lipoprotein receptor-related protein 1 unexpectedly accelerates atherosclerosis regression, enhances reverse cholesterol transport, and increases expression of the motility receptor CCR7, which drives macrophage egress from lesions.
Collapse
Affiliation(s)
- Paul A. Mueller
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Lin Zhu
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN
- Division of Diabetes, Metabolism and Endocrinology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Tennessee Valley Healthcare System, Nashville, TN
| | - Hagai Tavori
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Katherine Huynh
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Ilaria Giunzioni
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - John M. Stafford
- Division of Diabetes, Metabolism and Endocrinology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Tennessee Valley Healthcare System, Nashville, TN
| | - MacRae F. Linton
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| |
Collapse
|
111
|
Halu A, Wang JG, Iwata H, Mojcher A, Abib AL, Singh SA, Aikawa M, Sharma A. Context-enriched interactome powered by proteomics helps the identification of novel regulators of macrophage activation. eLife 2018; 7:37059. [PMID: 30303482 PMCID: PMC6179386 DOI: 10.7554/elife.37059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
The role of pro-inflammatory macrophage activation in cardiovascular disease (CVD) is a complex one amenable to network approaches. While an indispensible tool for elucidating the molecular underpinnings of complex diseases including CVD, the interactome is limited in its utility as it is not specific to any cell type, experimental condition or disease state. We introduced context-specificity to the interactome by combining it with co-abundance networks derived from unbiased proteomics measurements from activated macrophage-like cells. Each macrophage phenotype contributed to certain regions of the interactome. Using a network proximity-based prioritization method on the combined network, we predicted potential regulators of macrophage activation. Prediction performance significantly increased with the addition of co-abundance edges, and the prioritized candidates captured inflammation, immunity and CVD signatures. Integrating the novel network topology with transcriptomics and proteomics revealed top candidate drivers of inflammation. In vitro loss-of-function experiments demonstrated the regulatory role of these proteins in pro-inflammatory signaling. When human cells or tissues are injured, the body triggers a response known as inflammation to repair the damage and protect itself from further harm. However, if the same issue keeps recurring, the tissues become inflamed for longer periods of time, which may ultimately lead to health problems. This is what could be happening in cardiovascular diseases, where long-term inflammation could damage the heart and blood vessels. Many different proteins interact with each other to control inflammation; gaining an insight into the nature of these interactions could help to pinpoint the role of each molecular actor. Researchers have used a combination of unbiased, large-scale experimental and computational approaches to develop the interactome, a map of the known interactions between all proteins in humans. However, interactions between proteins can change between cell types, or during disease. Here, Halu et al. aimed to refine the human interactome and identify new proteins involved in inflammation, especially in the context of cardiovascular disease. Cells called macrophages produce signals that trigger inflammation whey they detect damage in other cells or tissues. The experiments used a technique called proteomics to measure the amounts of all the proteins in human macrophages. Combining these data with the human interactome made it possible to predict new links between proteins known to have a role in inflammation and other proteins in the interactome. Further analysis using other sets of data from macrophages helped identify two new candidate proteins – GBP1 and WARS – that may promote inflammation. Halu et al. then used a genetic approach to deactivate the genes and decrease the levels of these two proteins in macrophages, which caused the signals that encourage inflammation to drop. These findings suggest that GBP1 and WARS regulate the activity of macrophages to promote inflammation. The two proteins could therefore be used as drug targets to treat cardiovascular diseases and other disorders linked to inflammation, but further studies will be needed to precisely dissect how GBP1 and WARS work in humans.
Collapse
Affiliation(s)
- Arda Halu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States.,Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Jian-Guo Wang
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Hiroshi Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Alexander Mojcher
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Ana Luisa Abib
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Amitabh Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
112
|
Intermittent Hypoxia Enhances THP-1 Monocyte Adhesion and Chemotaxis and Promotes M1 Macrophage Polarization via RAGE. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1650456. [PMID: 30402462 PMCID: PMC6196992 DOI: 10.1155/2018/1650456] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/20/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023]
Abstract
Intermittent hypoxia (IH) that resulted from obstructive sleep apnea (OSA) has been found to be a risk factor of coronary artery disease. IH and the receptor for advanced glycation end products (RAGE) expression are known to activate monocyte/macrophage and associated with atherosclerosis development, while their effects on monocyte adhesion, chemotaxis to the endothelium, and macrophage polarization remain unknown. In the present study, RAGE in THP-1 monocytes was inhibited by shRNA lentiviral particles, followed by exposure to IH. Cell adhesion assay, transwell migration assay, and macrophage polarization assays were performed to study the effects of IH and RAGE. The mRNA and protein expression levels were investigated by RT/real-time PCR and western blot analysis, respectively. We found that IH increased RAGE expression and activated NF-кB signalling in THP-1 monocytes. The results also revealed that IH enhanced the MCP-1-mediated THP-1 monocyte adhesion and chemotaxis and promoted macrophage polarization toward a proinflammatory phenotype, which was mediated by RAGE activity. Additionally, inhibition of chemokine receptor type 2 (CCR2) suppressed the IH-induced monocyte adhesion and chemotaxis. These results demonstrated a potential role of monocyte adhesion, chemotaxis, and macrophage polarization in the development cardiovascular diseases induced by IH and identified that RAGE could be a promising therapeutic target to prevent atherosclerosis in patients with OSA.
Collapse
|
113
|
Modulation of the monocyte/macrophage system in heart failure by targeting heme oxygenase-1. Vascul Pharmacol 2018; 112:79-90. [PMID: 30213580 DOI: 10.1016/j.vph.2018.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/26/2018] [Accepted: 08/29/2018] [Indexed: 01/14/2023]
Abstract
Upon myocardial infarction (MI) immune system becomes activated by extensive necrosis of cardiomyocytes releasing intracellular molecules called damage-associated molecular patterns. Overactive and prolonged immune responses are likely to be responsible for heart failure development and progression in patients surviving the ischemic episode. Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. This stress-inducible enzyme is induced by various oxidative and inflammatory signals. Consequently, biological actions of HO-1 are not limited to degradation of a toxic heme released from hemoproteins, but also provide an adaptive cellular response against chronic inflammation and oxidative injury. Indeed, the immunomodulatory and anti-inflammatory properties of HO-1 were demonstrated in several experimental studies, as well as in human cases of genetic HO-1 deficiency. HO-1 was shown to suppress the production, myocardial infiltration and inflammatory properties of monocytes and macrophages what resulted in limitation of post-MI cardiac damage. This review specifically addresses the role of HO-1, heme and its degradation products in macrophage biology and post-ischemic cardiac repair. A more complete understanding of these mechanisms is essential to develop new therapeutic approaches.
Collapse
|
114
|
Tajbakhsh A, Rezaee M, Kovanen PT, Sahebkar A. Efferocytosis in atherosclerotic lesions: Malfunctioning regulatory pathways and control mechanisms. Pharmacol Ther 2018; 188:12-25. [DOI: 10.1016/j.pharmthera.2018.02.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
115
|
Zhao Y, Zheng J, Yu Y, Wang L. Panax notoginseng Saponins Regulate Macrophage Polarization under Hyperglycemic Condition via NF- κB Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9239354. [PMID: 30151392 PMCID: PMC6091338 DOI: 10.1155/2018/9239354] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/10/2018] [Accepted: 07/11/2018] [Indexed: 01/30/2023]
Abstract
Panax notoginseng saponins (PNS), the principal constituents derived from Panax notoginseng, have been extensively used for treating cardiocerebral vascular diseases in China and other Asian countries. The main effects of PNS were anti-inflammatory properties, inhibition of platelet aggregation, improvement of blood flow and insulin resistance, and so on. This study was carried out to explore the effects of PNS on macrophage polarization under hyperglycemic conditions. Human acute monocyte leukemia cell line THP-1 cells were induced into macrophages with Phorbol ester (PMA). Macrophages were then divided into five groups as follows: control (5.5mMol/l glucose), hyperglycemia group (15mMol/l glucose), hyperglycemia plus low-dose PNS (20ug/ml), hyperglycemia plus moderate-dose PNS (40ug/ml), and hyperglycemia plus high-dose PNS (60ug/ml). After 48-hour cell culture, the percentages of M1- and M2-polarized macrophages were measured by flow cytometry analysis. Reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) was used to evaluate the Ym1 and arginase 1 expression in macrophages. Protein expression of arginase 1, NF-κB p50, p65, and inhibitor of κB (IκB) alpha phosphorylation in macrophages was identified with Western blotting. PNS, especially the high-dose PNS, remarkably increased M2 phenotype ratio in macrophages cultured with hyperglycemia, and the mRNA expression of Ym1 and arginase 1 in macrophages was also upregulated. Meanwhile, PNS remarkably increased the protein expression of arginase 1 and decreased IκB-alpha phosphorylation and subunits of NF-κB p50 and p65 from macrophages in culture medium with hyperglycemia. Taken together, our work demonstrated that PNS promote macrophages to transform M2 phenotype under hyperglycemic conditions through downregulating NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, China
| | - Jianlei Zheng
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, China
| | - Yongmei Yu
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, China
| | - Lihong Wang
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
116
|
Improvement of Adipose Macrophage Polarization in High Fat Diet-Induced Obese GHSR Knockout Mice. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4924325. [PMID: 30112394 PMCID: PMC6077514 DOI: 10.1155/2018/4924325] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/06/2018] [Accepted: 05/28/2018] [Indexed: 12/16/2022]
Abstract
Purpose Adipose tissue inflammation is the key linking obesity to insulin resistance. Over 50% of the interstitial cells in adipose tissue are macrophages, which produce inflammatory cytokines and therefore play an important role in the progression of insulin resistance. Within this classification view, macrophage biology is driven by two polarization phenotypes, M1 (proinflammatory) and M2 (anti-inflammatory). The unique functional receptor of ghrelin, growth hormone secretagogue receptor (GHSR), is a classic seven-transmembrane G protein-coupled receptor that is linked to multiple intracellular signaling pathways. Knockout of GHSR improves the obesity and glucose metabolic disorders, suggesting a crucial role of ghrelin activity in insulin resistance. Here, we discussed whether macrophage polarization phenotypes in adipose tissue were changed in GHSR knockout (GHSR-/-) mice. Methods GHSR-/- mice were fed with normal chow diet (NCD) or high fat diet (HFD). Markers of different macrophage polarization phenotypes were detected by real-time RT-PCR. Results The size of adipocytes decreased and interstitial cells, especially infiltrated macrophages, reduced in epididymal adipose tissue of GHSR-/- mice fed with HFD. Compared with wild type mice, the mRNA levels of inflammatory adipokines such as resistin, IL-6, and PAI-1 were significantly lower in epididymal adipose tissue of GHSR-/- mice, whereas anti-inflammatory adipokine, adiponectin, was significantly higher. M1 markers, MCP-1, TNF-α, and iNOS, were significantly lower in epididymal adipose tissue of GHSR-/- mice, whereas M2 markers, Arg-1, Mgl-1, were Mrc1, were significantly higher. Conclusion The GHSR-/- mice fed with HFD showed suppressed adipose inflammation, reduced macrophage infiltration, and enhanced M2 polarization of macrophages in adipose tissue, which improved insulin sensitivity.
Collapse
|
117
|
Zhu X, Tu Y, Chen H, Jackson AO, Patel V, Yin K. Micro-environment and intracellular metabolism modulation of adipose tissue macrophage polarization in relation to chronic inflammatory diseases. Diabetes Metab Res Rev 2018; 34:e2993. [PMID: 29475214 DOI: 10.1002/dmrr.2993] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 12/13/2022]
Abstract
The accumulation and pro-inflammatory polarization of immune cells, mainly macrophages, in adipose tissue (AT) are considered crucial factors for obesity-induced chronic inflammatory diseases. In this review, we highlighted the role of adipose tissue macrophage (ATM) polarization on AT function in the obese state and the effect of the micro-environment and intracellular metabolism on the dynamic switch of ATMs into their pro-inflammatory or anti-inflammatory phenotypes, which may have distinct influences on obesity-related chronic inflammatory diseases. Obesity-associated metabolic dysfunctions, including those of glucose, fatty acid, cholesterol, and other nutrient substrates such as vitamin D and iron in AT, promote the pro-inflammatory polarization of ATMs and AT inflammation via regulating the interaction between ATMs and adipocytes and intracellular metabolic pathways, including glycolysis, fatty acid oxidation, and reverse cholesterol transportation. Focusing on the regulation of ATM metabolism will provide a novel target for the treatment of obesity-related chronic inflammatory diseases, including insulin resistance, cardiovascular diseases, and cancers.
Collapse
Affiliation(s)
- Xiao Zhu
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang, China
| | - Yixuan Tu
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
| | - Hainan Chen
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang, China
| | - Ampadu O Jackson
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
| | - Vaibhav Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang, China
| |
Collapse
|
118
|
Jadapalli JK, Halade GV. Unified nexus of macrophages and maresins in cardiac reparative mechanisms. FASEB J 2018; 32:5227-5237. [PMID: 29750575 DOI: 10.1096/fj.201800254r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Macrophages are immune-sensing "big eater" phagocytic cells responsible for an innate, adaptive, and regenerative response. After myocardial infarction, macrophages predominantly clear the deceased cardiomyocyte apoptotic or necrotic neutrophils to develop a regenerative and reparative program with the activation of the lipoxygenase-mediated maresin (MaR) metabolome at the site of ischemic injury. The specialized proresolving molecule and macrophage mediator in resolving inflammation, MaR-1, produced by human macrophages, has potent defining effects that limit polymorphonuclear neutrophil infiltration, enhance uptake of apoptotic PMNs, regulate inflammation resolution and tissue regeneration, and reduce pain. In addition to proresolving and anti-inflammatory actions, MaR-1 displays potent tissue regenerative effects in stroke and is an antinociceptive. Macrophages actively participate in the biosynthesis of bioactive MaR-2, which exhibits anti-inflammatory, proresolving, and atherosclerotic effects. A new class of macrophage-derived molecules, MaR conjugates in tissue regeneration, is identified that regulates phagocytosis and the repair and regeneration of damaged tissue. The presented review provides a current summary of the effect of MaR in resolution pathophysiology, with relevance to a cardiac repair program.-Jadapalli, J. K., Halade, G. V. Unified nexus of macrophages and maresins in cardiac reparative mechanisms.
Collapse
Affiliation(s)
- Jeevan Kumar Jadapalli
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama, Birmingham, Alabama, USA
| | - Ganesh V Halade
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama, Birmingham, Alabama, USA
| |
Collapse
|
119
|
Adamson SE, Polanowska-Grabowska R, Marqueen K, Griffiths R, Angdisen J, Breevoort SR, Schulman IG, Leitinger N. Deficiency of Dab2 (Disabled Homolog 2) in Myeloid Cells Exacerbates Inflammation in Liver and Atherosclerotic Plaques in LDLR (Low-Density Lipoprotein Receptor)-Null Mice-Brief Report. Arterioscler Thromb Vasc Biol 2018; 38:1020-1029. [PMID: 29599136 PMCID: PMC5920703 DOI: 10.1161/atvbaha.117.310467] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 03/06/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Inflammatory macrophages promote the development of atherosclerosis. We have identified the adaptor protein Dab2 (disabled homolog 2) as a regulator of phenotypic polarization in macrophages. The absence of Dab2 in myeloid cells promotes an inflammatory phenotype, but the impact of myeloid Dab2 deficiency on atherosclerosis has not been shown. APPROACH AND RESULTS To determine the role of myeloid Dab2 in atherosclerosis, Ldlr-/- mice were reconstituted with either Dab2-positive or Dab2-deficient bone marrow and fed a western diet. Consistent with our previous finding that Dab2 inhibits NFκB (nuclear factor κ-light-chain-enhancer of activated B cells) signaling in macrophages, Ldlr-/- mice reconstituted with Dab2-deficient bone marrow had increased systemic inflammation as evidenced by increased serum IL-6 (interleukin-6) levels and increased inflammatory cytokine expression levels in liver. Serum lipid levels were significantly lower in Ldlr-/- mice reconstituted with Dab2-deficient bone marrow, and further examination of livers from these mice revealed drastically increased inflammatory tissue damage and massive infiltration of immune cells. Surprisingly, the atherosclerotic lesion burden in Ldlr-/- mice reconstituted with Dab2-deficient bone marrow was decreased compared with Ldlr-/- mice reconstituted with wild-type bone marrow. Further analysis of aortic root sections revealed increased macrophage content and evidence of increased apoptosis in lesions from Ldlr-/- mice reconstituted with Dab2-deficient bone marrow but no difference in collagen or α-smooth muscle actin content. CONCLUSIONS Dab2 deficiency in myeloid cells promotes inflammation in livers and atherosclerotic plaques in a mouse model of atherosclerosis. Nevertheless, decreased serum lipids as a result of massive inflammatory liver damage may preclude an appreciable increase in atherosclerotic lesion burden in mice reconstituted with Dab2-deficient bone marrow.
Collapse
Affiliation(s)
- Samantha E Adamson
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (S.E.A., R.P.-G., R.G., N.L.)
| | - Renata Polanowska-Grabowska
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (S.E.A., R.P.-G., R.G., N.L.)
| | - Kathryn Marqueen
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
| | - Rachael Griffiths
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (S.E.A., R.P.-G., R.G., N.L.)
| | - Jerry Angdisen
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
| | - Sarah R Breevoort
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
| | - Ira G Schulman
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
| | - Norbert Leitinger
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (S.E.A., R.P.-G., R.G., N.L.)
| |
Collapse
|
120
|
Sieve I, Ricke-Hoch M, Kasten M, Battmer K, Stapel B, Falk CS, Leisegang MS, Haverich A, Scherr M, Hilfiker-Kleiner D. A positive feedback loop between IL-1β, LPS and NEU1 may promote atherosclerosis by enhancing a pro-inflammatory state in monocytes and macrophages. Vascul Pharmacol 2018; 103-105:16-28. [DOI: 10.1016/j.vph.2018.01.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/13/2017] [Accepted: 01/19/2018] [Indexed: 01/22/2023]
|
121
|
Jingjing Z, Nan Z, Wei W, Qinghe G, Weijuan W, Peng W, Xiangpeng W. MicroRNA-24 Modulates Staphylococcus aureus-Induced Macrophage Polarization by Suppressing CHI3L1. Inflammation 2018; 40:995-1005. [PMID: 28303416 DOI: 10.1007/s10753-017-0543-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Macrophages play a crucial role in host innate anti-Staphylococcus aureus defense, which is tightly regulated by multiple factors, including microRNAs. A recent study showed that miR-24 plays an important role in macrophage polarization. Here, we investigated the biological function of miR-24 in S. aureus-stimulated macrophages. The results revealed that miR-24 expression was significantly decreased in both human and mouse macrophage cell lines with S. aureus stimulation in a time-dependent manner. Moreover, miR-24 overexpression significantly decreased the production of M1 phenotype markers, such as IL-6, iNOS, TNF-α, CD86, and CD80, whereas it increased the production of M2 markers, such as Arg1, CCL17, CCL22, CD163, and CD206, in S. aureus-stimulated macrophages. Conversely, knockdown of miR-24 promoted M1 macrophage polarization but diminished M2 macrophage polarization in S. aureus-stimulated macrophages. Furthermore, CHI3L1 was predicted as a target gene of miR-24 using bioinformatics software and identified by luciferase reporter assay. Additionally, miR-24 overexpression inhibited CHI3L1 expression and downregulated the downstream MAPK pathway in S. aureus-stimulated macrophages. Finally, CHI3L1 overexpression rescued macrophage polarization and MAPK pathway inhibition induced by miR-24 mimic transfection in S. aureus-stimulated macrophages. In conclusion, the data suggest that miR-24 serves as a molecular regulator in S. aureus-induced macrophage polarization through targeting of CHI3L1 and regulation of the MAPK pathway, which may provide a promising therapeutic target for S. aureus-related infections and inflammatory diseases.
Collapse
Affiliation(s)
- Zhang Jingjing
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, No. 601 Jinsui Road, Hongqi District, Xinxiang, 453003, Henan Province, China
| | - Zhang Nan
- Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Wu Wei
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Guo Qinghe
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, No. 601 Jinsui Road, Hongqi District, Xinxiang, 453003, Henan Province, China
| | - Wang Weijuan
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Wang Peng
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Wang Xiangpeng
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, No. 601 Jinsui Road, Hongqi District, Xinxiang, 453003, Henan Province, China.
| |
Collapse
|
122
|
Abstract
Apolipoprotein E (apoE) is a 34-kDa glycoprotein that is secreted from many cells throughout the body. ApoE is best known for its role in lipoprotein metabolism. Recent studies underline the association of circulating lipoprotein-associated apoE levels and the development for cardiovascular disease (CVD). Besides its well-established role in pathology of CVD, it is also implicated in neurodegenerative diseases and recent new data on adipose-produced apoE point to a novel metabolic role for apoE in obesity. The regulation of apoE production and secretion is remarkably cell and tissue specific. Here, we summarize recent insights into the differential regulation apoE production and secretion by hepatocytes, monocytes/macrophages, adipocytes, and the central nervous system and relevant variations in apoE biochemistry and function.
Collapse
Affiliation(s)
- Maaike Kockx
- Concord Repatriation General Hospital, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Mathew Traini
- Concord Repatriation General Hospital, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Leonard Kritharides
- Concord Repatriation General Hospital, ANZAC Research Institute, Sydney, Australia.
- Sydney Medical School, University of Sydney, Sydney, Australia.
- Department of Cardiology, Concord Repatriation General Hospital, Concord, NSW, 2139, Australia.
| |
Collapse
|
123
|
Liu M, Yang W, Liu S, Hock D, Zhang B, Huo RY, Tong X, Yan H. LXRα is expressed at higher levels in healthy people compared to atherosclerosis patients and its over-expression polarizes macrophages towards an anti-inflammatory MΦ2 phenotype. Clin Exp Hypertens 2018; 40:213-217. [PMID: 29420090 DOI: 10.1080/10641963.2017.1288740] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mengyuan Liu
- Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin, 300060 China
- Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin, 300060 China
| | - Weijian Yang
- Graduate School of Tianjin Medical University, Tianjin, China
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Shuling Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, 300060 China
| | - Duncan Hock
- Medicine School, University of South Florida, Tampa, 33620-9951 United States
| | - Bohao Zhang
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Ruth Ya Huo
- School of Medicine, University of New South Wales, Sydney, 2052 Australia
| | - Xiaoguang Tong
- Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin, 300060 China
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Hua Yan
- Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin, 300060 China
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
124
|
Evdokimenko AN, Anufriev PL, Kulichenkova KN, Gulevskaya TS, Tanashyan MM. [Morphometric characteristics of neovascularization of carotid atherosclerotic plaques]. Arkh Patol 2018; 80:24-29. [PMID: 29697668 DOI: 10.17116/patol201880224-29] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
OBJECTIVE to identify the relationship between the magnitude of neovascularization of an atherosclerotic plaque and its other processes, such as atheromatosis, macrophage responses, hemorrhages, and calcification. MATERIAL AND METHODS 48 carotid atherosclerotic plaques all their way underwent morphopathological examination, by assessing the magnitude of neovascularization (total vessel density per plague cm2; density of vessels of different diameters; localization in section), atheromatosis (a volume fraction), foam cells (a score of 0 to 5), hemorrhages (a score of 0 to 4), and calcification (a score of 0 to 4). The Mann-Whitney U test and the Spearman correlation coefficient with p<0.05 were used in the statistical analysis. RESULTS There was a substantial variability in vessel diameters and densities in the plaque with a drastic predominance of vessels smaller than 20 µm in diameter (69.8±14.3%). The vessels with a diameter as large as 20 µm were more frequently located in the area of accumulation of foam cells or organizing hemorrhages (p<0.03), unlike those with a diameter of more than 40 µm, which were more common in the areas of connective tissue, especially adjacent to large calcifications, and in the foci of organized hemorrhages (p<0.008). Intensified calcification was accompanied by the higher density of vessels having a diameter of more than 40 µm (p<0.038) and particularly of those with a diameter of more than 60 µm (p=0.006). The degree of hemorrhages and the number of foam cells were unrelated to the density of vessels, but these increased with their localization in the surface areas of a plaque (p=0.000342). With the increasing severity of atheromatosis, there was a decrease in the density of vessels larger than 20 µm in diameter (p=0.00042). CONCLUSION The number, diameter, and location of vessels in the plaque serve as a dynamic reflection of its destructive and reparative processes; and a larger number of vessels having a diameter of more than 40 µm can be an indicator for the activation of reparative processes.
Collapse
|
125
|
Casaburi I, Chimento A, De Luca A, Nocito M, Sculco S, Avena P, Trotta F, Rago V, Sirianni R, Pezzi V. Cholesterol as an Endogenous ERRα Agonist: A New Perspective to Cancer Treatment. Front Endocrinol (Lausanne) 2018; 9:525. [PMID: 30254608 PMCID: PMC6141749 DOI: 10.3389/fendo.2018.00525] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/21/2018] [Indexed: 01/01/2023] Open
Abstract
The estrogen-related receptors (ERRs) are important members of nuclear receptors which contain three isoforms (α, β, and γ). ERRα is the best-characterized isoform expressed mainly in high-energy demanding tissues where it preferentially works in association with the peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α) and PGC-1β. ERRα together with its cofactors modulates cellular metabolism, supports the growth of rapidly dividing cells, directs metabolic programs required for cell differentiation and maintains cellular energy homeostasis in differentiated cells. In cancer cells, the functional association between ERRα and PGC-1s is further influenced by oncogenic signals and induces metabolic programs favoring cell growth and proliferation as well as tumor progression. Recently, cholesterol has been identified as a natural ERRα ligand using a combined biochemical strategy. This new finding highlighted some important physiological aspects related to the use of cholesterol-lowering drugs such as statins and bisphosphonates. Even more meaningful is the link between increased cholesterol levels and certain cancer phenotypes characterized by an overexpressed ERRα such as mammary, prostatic, and colorectal cancers, where the metabolic adaptation affects many cancer processes. Moreover, high-energy demanding cancer-related processes are strictly related to the cross-talk between tumor cells and some key players of tumor microenvironment, such as tumor-associated macrophage that fuels cancer progression. Some evidence suggests that high cholesterol content and ERRα activity favor the inflammatory environment by the production of different cytokines. In this review, starting from the most recent observations on the physiological role of the new signaling activated by the natural ligand of ERRα, we propose a new hypothesis on the suitability to control cholesterol levels as a chance in modulating ERRα activity in those tumors in which its expression and activity are increased.
Collapse
|
126
|
Wolf D, Stachon P, Bode C, Zirlik A. Inflammatory mechanisms in atherosclerosis. Hamostaseologie 2017; 34:63-71. [DOI: 10.5482/hamo-13-09-0050] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/05/2013] [Indexed: 01/13/2023] Open
Abstract
SummaryThroughout the last two decades inflammation has been recognized as the central mechanism underlying atherogenesis. A multitude of basic science work demonstrates the pivotal role of inflammatory processes during every step of atherosclerotic plaque formation: From initiation via propagation to complication.This review describes some of the key mechanisms involved with a particular focus on the diverse group of inflammatory cells and their subsets that distinctly contribute to atherogenic and anti-atherogenic phenomena. Furthermore, we summarize the controlling action of a tight network of co-stimulatory molecules and cytokines orchestrating the inflammatory and anti-inflammatory effector functions. Finally, the current status of clinical trials evaluating anti-inflammatory/ immune-modulatory treatment strategies is summarized and an outlook for future therapeutic implications is provided.
Collapse
|
127
|
Li J, Lei HT, Cao L, Mi YN, Li S, Cao YX. Crocin alleviates coronary atherosclerosis via inhibiting lipid synthesis and inducing M2 macrophage polarization. Int Immunopharmacol 2017; 55:120-127. [PMID: 29248792 DOI: 10.1016/j.intimp.2017.11.037] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/06/2017] [Accepted: 11/27/2017] [Indexed: 12/15/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease arising from an imbalance in lipid levels and the accumulation of cholesterol-laden macrophages in the artery wall. Crocin is an active ingredient of Crocus sativus L. This study established a rat coronary atherosclerosis model induced by vitamin D3 (VD3), to explore the effect of Crocin on lipid metabolism, macrophage polarization and the activity of inflammatory proteins. The results revealed that Crocin decreased blood lipid levels by decreasing the levels of endothelin (ET), total cholesterol (TC), triglyceridelow (TG) and low-density lipoprotein cholesterol (LDL-c), elevating the level of high-density lipoprotein cholesterin (HDL-c). Crocin also inhibited lipogenesis by suppressing the expression of lipogenesis-related proteins and elevating lipid catabolism-related proteins. Moreover, Crocin effectively alleviated inflammation by suppressing the expression of pro-inflammatory cytokines and increasing levels of anti-inflammatory cytokines. We further found that Crocin promoted macrophage polarization to the M2 phenotype by reducing M1 markers (CD40+ and CD11c+) and elevating M2 markers (CD68+ and CD206+). Finally, Crocin strongly inhibited the expression of NF-κB p65 and its translocation into the nucleus. Crocin partially counteracted nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 expression and the nuclei accumulation caused by NF-κB p65 overexpression. Taken together, our research indicated that Crocin inhibited lipogenesis and alleviated the inflammation in a VD3-induced rat coronary atherosclerosis model by promoting M2 macrophage polarization and maybe by inhibiting NF-κB p65 nuclear translocation. This study implicates Crocin as a potential therapeutic strategy for coronary atherosclerosis.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China; Department of Traditional Chinese Medicine, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Hong-Tao Lei
- Department of Neonatal, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Lei Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Yan-Ni Mi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Sen Li
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Yong-Xiao Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
128
|
Bras JP, Silva AM, Calin GA, Barbosa MA, Santos SG, Almeida MI. miR-195 inhibits macrophages pro-inflammatory profile and impacts the crosstalk with smooth muscle cells. PLoS One 2017; 12:e0188530. [PMID: 29166412 PMCID: PMC5699821 DOI: 10.1371/journal.pone.0188530] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/08/2017] [Indexed: 12/16/2022] Open
Abstract
Macrophages are a main component of atherosclerotic plaques. Recent studies suggest that pro-inflammatory M1 macrophages are pro-atherogenic while M2 macrophages promote plaque stability. Moreover, toll-like receptor signalling pathways are implicated in atherosclerotic plaque formation, evolution and regression. We propose microRNAs as key regulators of these processes. In this context, our goal is to promote inflammation resolution using miR-195 to reduce M1-like macrophage polarization and to evaluate the molecular mechanisms underlying such effect, as well as to explore the functional consequences for smooth muscle cell recruitment. Human primary macrophages were differentiated from peripheral blood monocytes and stimulated with LPS or IL-10 to promote M1 or M2c polarization, respectively. miR-195 levels were upregulated in M2c macrophages compared with M1 macrophages. In THP-1 macrophages stimulated with LPS and IFN-γ, results show that TLR2 levels were reduced by miR-195 overexpression compared with scrambled control. In addition, phosphorylated forms of p54 JNK, p46 JNK and p38 MAPK were decreased by miR-195 in macrophages following M1 stimulation. Moreover, miR-195 significantly decreased levels of IL-1β, IL-6 and TNF-α pro-inflammatory cytokines in the supernatants of M1-stimulated macrophage cultures. At the functional level, results from smooth muscle cell recruitment and migration models showed that miR-195 impairs the capacity of M1 macrophages to promote smooth muscle cells migration. In conclusion, miR-195 is involved in macrophage polarization and inhibits TLR2 inflammatory pathway mediators. Moreover, miR-195 impairs the effect of macrophages on smooth muscle cells recruitment capacity and migration profile. Thus, miR-195 might be used as a new potential tool to promote inflammation resolution in cardiovascular research.
Collapse
Affiliation(s)
- Joao Paulo Bras
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia da Universidade do Porto, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Andreia Machado Silva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - George A. Calin
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Mario Adolfo Barbosa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Susana Gomes Santos
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Maria Ines Almeida
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| |
Collapse
|
129
|
Otsuka F, Zhao X, Trout HH, Qiao Y, Wasserman BA, Nakano M, Macphee CH, Brandt M, Krug-Gourley S, Guo L, Ladich ER, Cheng Q, Davis HR, Finn AV, Virmani R, Kolodgie FD. Community-based statins and advanced carotid plaque: Role of CD163 positive macrophages in lipoprotein-associated phospholipase A 2 activity in atherosclerotic plaque. Atherosclerosis 2017; 267:78-89. [PMID: 29101839 DOI: 10.1016/j.atherosclerosis.2017.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 10/09/2017] [Accepted: 10/12/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS Lipoprotein-associated phospholipase A2 (Lp-PLA2), an enzymatic inflammatory biomarker primarily bound to low-density lipoprotein cholesterol, is associated with an approximate twofold increased risk of cardiovascular disease and stroke. Despite indications that circulating Lp-PLA2 is sensitive to statins, it remains largely unknown whether statin usage exerts local effects on Lp-PLA2 expression at the site of atheromatous plaque. METHODS Carotid plaques (n = 38) were prospectively collected from symptomatic (n = 18) and asymptomatic (n = 20) patients with (n = 20) or without (n = 18) documented statin history. In all cases, endarterectomy was performed where the primary stenosis was removed in an undisturbed manner. Serial cryosections of the presenting lesion were assessed histologically for macrophages, Lp-PLA2, and cell death (apoptotic index). RESULTS Symptomatic lesions exhibited less calcification, with greater inflammation characterized by increased expression of CD68+ and CD163+ macrophage subsets, and Lp-PLA2. Symptomatic plaques also exhibited greater necrotic core area and increased apoptosis, as compared with asymptomatic lesions. In contrast, statin treatment did not appear to influence any of these parameters, except for the extent of apoptosis, which was less in statin treated as compared with statin naïve lesions. Overall, Lp-PLA2 expression correlated positively with necrotic core area, CD68+ and CD163+ macrophage area, and cell death. Finally, in vitro assays and dual immunofluorescence staining confirmed CD163-expressing monocytes/macrophages are also a major source of Lp-PLA2. CONCLUSIONS Statin treatment has no effect on local atherosclerotic lesion Lp-PLA2 activity, therefore, the addition of anti-inflammatory treatments to further decrease macrophage Lp-PLA2 expression in atherosclerotic lesions may reduce lesional inflammation and cell death, and prevent necrotic core expansion and lesion progression.
Collapse
Affiliation(s)
| | | | - Hugh H Trout
- Department of Surgery, Suburban Hospital, Bethesda, MD, USA
| | - Ye Qiao
- Department of Radiology and Radiological Sciences, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Bruce A Wasserman
- Department of Radiology and Radiological Sciences, The Johns Hopkins Hospital, Baltimore, MD, USA
| | | | | | | | | | - Liang Guo
- CVPath Institute, Inc., Gaithersburg, MD, USA
| | | | - Qi Cheng
- CVPath Institute, Inc., Gaithersburg, MD, USA
| | | | | | | | | |
Collapse
|
130
|
Lin L, Hu K. Tissue-type plasminogen activator modulates macrophage M2 to M1 phenotypic change through annexin A2-mediated NF-κB pathway. Oncotarget 2017; 8:88094-88103. [PMID: 29152144 PMCID: PMC5675696 DOI: 10.18632/oncotarget.21510] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/31/2017] [Indexed: 11/25/2022] Open
Abstract
Macrophage accumulation is one of the hallmarks of progressive kidney disease. In response to injury, macrophages undergo a phenotypic polarization to become two functionally distinct subsets: M1 and M2 macrophages. Macrophage polarization is a dynamic process, and recent work indicates that macrophages, in response to kidney injury, can shift their polarity. However, the underlying mechanisms remain largely unknown. Tissue-type plasminogen activator (tPA), a protease up-regulated in the chronically injured kidneys, has been shown to preferably promote M1 macrophage accumulation and renal inflammation. We hypothesized that tPA may be an endogenous factor that modulates macrophage M2 to M1 phenotypic change contributing to the accumulation of M1 macrophages in the injured kidneys. It was found that obstruction-induced renal M1 chemokine expression was alleviated in tPA knockout mice, and these knockout mice displayed increased M2 markers. In vitro, resting J774 macrophages were treated with IL-4 to induce M2 phenotype as indicated by de novo expression of arginase 1, Ym1, and IL-10, as well as suppression of iNOS, TNF-α, and IL-1β. Intriguingly, these IL-4-induced M2 macrophages, after tPA treatment, not only lost their M2 markers such as arginase 1, Ym1, and IL-10, but also displayed increased M1 chemokines including iNOS, TNF-α, and IL-1β. Possible endotoxin contamination was also excluded as heat-inactivated tPA lost its effect. Additionally, tPA-mediated macrophage M2 to M1 phenotypic change required its receptor annexin A2, and SN50, a specific NF-κB inhibitor, abolished tPA's effect. Thus, it's clear that tPA promotes macrophage M2 to M1 phenotypic change through annexin A2-mediated NF-κB pathway.
Collapse
Affiliation(s)
- Ling Lin
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Kebin Hu
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, Pennsylvania, USA.,Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
131
|
Serbulea V, DeWeese D, Leitinger N. The effect of oxidized phospholipids on phenotypic polarization and function of macrophages. Free Radic Biol Med 2017; 111:156-168. [PMID: 28232205 PMCID: PMC5511074 DOI: 10.1016/j.freeradbiomed.2017.02.035] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/09/2017] [Accepted: 02/15/2017] [Indexed: 12/26/2022]
Abstract
Oxidized phospholipids are products of lipid oxidation that are found on oxidized low-density lipoproteins and apoptotic cell membranes. These biologically active lipids were shown to affect a variety of cell types and attributed pro-as well as anti-inflammatory effects. In particular, macrophages exposed to oxidized phospholipids drastically change their gene expression pattern and function. These 'Mox,'macrophages were identified in atherosclerotic lesions, however, it remains unclear how lipid oxidation products are sensed by macrophages and how they influence their biological function. Here, we review recent developments in the field that provide insight into the structure, recognition, and downstream signaling of oxidized phospholipids in macrophages.
Collapse
Affiliation(s)
- Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center and Department of Pharmacology, University of Virginia, USA
| | - Dory DeWeese
- Robert M. Berne Cardiovascular Research Center and Department of Pharmacology, University of Virginia, USA
| | - Norbert Leitinger
- Robert M. Berne Cardiovascular Research Center and Department of Pharmacology, University of Virginia, USA
| |
Collapse
|
132
|
The Effects of Selective Hematopoietic Expression of Human IL-37 on Systemic Inflammation and Atherosclerosis in LDLr-Deficient Mice. Int J Mol Sci 2017; 18:ijms18081672. [PMID: 28792474 PMCID: PMC5578062 DOI: 10.3390/ijms18081672] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/21/2017] [Accepted: 07/28/2017] [Indexed: 12/28/2022] Open
Abstract
The human cytokine interleukin (IL)-37 has potent anti-inflammatory capacities, and hematopoietic cell-specific transgenic overexpression of IL-37 in mice protects against septic shock and colitis. In the present study we investigated the effect of hematopoietic expression of IL-37 on atherosclerosis development under low-grade inflammatory conditions. Low-density lipoprotein receptor (LDLr)-deficient mice were lethally irradiated and transplanted with bone marrow from IL-37-transgenic or control wild-type mice and fed a Western-type diet (WTD; 1% cholesterol) for eight weeks. Metabolic and inflammatory parameters were monitored and atherosclerosis was assessed in the aortic valve area. Hematopoietic IL-37 expression did not influence body weight, food intake and plasma cholesterol levels during the study. Plasma soluble E-selectin levels were increased with WTD-feeding as compared to chow-feeding, but were not influenced by IL-37 expression. IL-37 expression reduced the inflammatory state as indicated by reduced white blood cell counts and by reduced basal and lipopolysaccharide-induced cytokine response by peritoneal macrophages ex vivo. IL-37 expression did not influence the atherosclerotic lesion area. Lesion composition was marginally affected. Smooth muscle cell content was decreased, but macrophage and collagen content were not different. We conclude that under low-grade inflammatory conditions, hematopoietic IL-37 expression reduces the inflammatory state, but does not influence atherosclerosis development in hyperlipidemic LDLr-deficient mice.
Collapse
|
133
|
Lin J, Liu Q, Zhang H, Huang X, Zhang R, Chen S, Wang X, Yu B, Hou J. C1q/Tumor necrosis factor-related protein-3 protects macrophages against LPS-induced lipid accumulation, inflammation and phenotype transition via PPARγ and TLR4-mediated pathways. Oncotarget 2017; 8:82541-82557. [PMID: 29137283 PMCID: PMC5669909 DOI: 10.18632/oncotarget.19657] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/22/2017] [Indexed: 12/11/2022] Open
Abstract
Macrophage inflammation and foam cell formation are critical events during the initiation and development of atherosclerosis (AS). C1q/tumor necrosis factor-related protein-3 (CTRP3) is a novel adipokine with anti-inflammatory and cardioprotection properties; however, little is known regarding the influence of CTRP3 on AS. As macrophages play a key role in AS, this study investigated the effects of CTRP3 on macrophage lipid metabolism, inflammatory reactions, and phenotype transition, as well as underlying mechanisms, to reveal the relationship between CTRP3 and AS. CTRP3 reduced the number of lipid droplets, lowered cholesteryl ester (CE), total cholesterol (TC), and free cholesterol (FC) levels, reduced the CE/TC ratio, and dose-dependently inhibited TNFα, IL-6, MCP-1, MMP-9 and IL-1β release in lipopolysaccharide (LPS)-stimulated THP-1 macrophages and mouse peritoneal macrophages. Pretreatment with CTRP3 effectively increased macrophage transformation to M2 macrophages rather than M1 macrophages. Western blotting showed that the specific NF-κB pathway inhibitor ammonium pyrrolidine dithiocarbamate (PDTC) or siRNA targeting PPARγ/LXRα markedly strengthened or abolished the above-mentioned effects of CTRP3, respectively. These results show that CTRP3 inhibits TLR4-NF-κB pro-inflammatory pathways but activates the PPARγ-LXRα-ABCA1/ABCG1 cholesterol efflux pathway. Taken together, CTRP3 participates in anti-lipid accumulation, anti-inflammation and macrophage phenotype conversion via the TLR4-NF-κB and PPARγ-LXRα-ABCA1/ABCG1 pathways and, thus, may have anti-atherosclerotic properties.
Collapse
Affiliation(s)
- Jiale Lin
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Qi Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Hui Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Xingtao Huang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Ruoxi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Shuyuan Chen
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Xuedong Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Jingbo Hou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
134
|
Yang Y, Luo NS, Ying R, Xie Y, Chen JY, Wang XQ, Gu ZJ, Mai JT, Liu WH, Wu MX, Chen ZT, Fang YB, Zhang HF, Zuo ZY, Wang JF, Chen YX. Macrophage-derived foam cells impair endothelial barrier function by inducing endothelial-mesenchymal transition via CCL-4. Int J Mol Med 2017; 40:558-568. [PMID: 28656247 PMCID: PMC5504989 DOI: 10.3892/ijmm.2017.3034] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 06/06/2017] [Indexed: 12/20/2022] Open
Abstract
Recently, endothelial-mesenchymal transition (EndMT) has been demonstrated to play an important role in the development of atherosclerosis, the molecular mechanisms of which remain unclear. In the present study, scanning electron microscopy directly revealed a widened endothelial space and immunohistofluorescence demonstrated that EndMT was increased in human aorta atherosclerotic plaques. M1 macrophage-derived foam cell (M1-FC) supernatants, but not M2 macrophage-derived foam cell (M2-FC) supernatants, induced EndMT. A protein array and enzyme-linked immunosorbent assay identified that the levels of several cytokines, including C-C motif chemokine ligand 4 (CCL-4) were increased in M1-FC supernatants, in which EndMT was promoted, accompanied by increased endothelial permeability and monocyte adhesion. Furthermore, anti-CCL-4 antibody abolished the effects of M1-FC supernatants on EndMT. At the same time, CCL-4 activated its receptor, C-C motif chemokine receptor-5 (CCR-5), and upregulated transforming growth factor-β (TGF-β) expression. Further experiments revealed that EndMT induced by CCL-4 was reversed by treatment with CCR-5 antagonist and the RNA-mediated knockdown of TGF-β. On the whole, the data of the present study suggest that M1-FCs induce EndMT by upregulating CCL-4, and increase endothelial permeability and monocyte adhesion. These data may help to elucidate the important role of EndMT in the development of atherosclerosis.
Collapse
Affiliation(s)
- Ying Yang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Nian-Sang Luo
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Ru Ying
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jia-Yuan Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiao-Qiao Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhen-Jie Gu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jing-Ting Mai
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Wen-Hao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Mao-Xiong Wu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhi-Teng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yong-Biao Fang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhi-Yi Zuo
- Laboratory of RNA and Major Disease of Heart and Brain, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yang-Xin Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
135
|
Rostam HM, Reynolds PM, Alexander MR, Gadegaard N, Ghaemmaghami AM. Image based Machine Learning for identification of macrophage subsets. Sci Rep 2017; 7:3521. [PMID: 28615717 PMCID: PMC5471192 DOI: 10.1038/s41598-017-03780-z] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/03/2017] [Indexed: 11/29/2022] Open
Abstract
Macrophages play a crucial rule in orchestrating immune responses against pathogens and foreign materials. Macrophages have remarkable plasticity in response to environmental cues and are able to acquire a spectrum of activation status, best exemplified by pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes at the two ends of the spectrum. Characterisation of M1 and M2 subsets is usually carried out by quantification of multiple cell surface markers, transcription factors and cytokine profiles. These approaches are time-consuming, require large numbers of cells and are resource intensive. In this study, we used machine learning algorithms to develop a simple and fast imaging-based approach that enables automated identification of different macrophage functional phenotypes using their cell size and morphology. Fluorescent microscopy was used to assess cell morphology of different cell types which were stained for nucleus and actin distribution using DAPI and phalloidin respectively. By only analysing their morphology we were able to identify M1 and M2 phenotypes effectively and could distinguish them from naïve macrophages and monocytes with an average accuracy of 90%. Thus we suggest high-content and automated image analysis can be used for fast phenotyping of functionally diverse cell populations with reasonable accuracy and without the need for using multiple markers.
Collapse
Affiliation(s)
- Hassan M Rostam
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.,Department of Biology, University of Garmian, Kalar, Kurdistan, Iraq
| | - Paul M Reynolds
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK
| | - Morgan R Alexander
- Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Nikolaj Gadegaard
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK.
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
136
|
Schulman IG. Liver X receptors link lipid metabolism and inflammation. FEBS Lett 2017; 591:2978-2991. [PMID: 28555747 DOI: 10.1002/1873-3468.12702] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/23/2017] [Indexed: 12/14/2022]
Abstract
The response of immune cells to pathogens is often associated with changes in the flux through basic metabolic pathways. Indeed, in many cases changes in metabolism appear to be necessary for a robust immune response. The Liver X receptors (LXRs) are members of the nuclear hormone receptor superfamily that regulate gene networks controlling cholesterol and lipid metabolism. In immune cells, particularly in macrophages, LXRs also inhibit proinflammatory gene expression. This Review will highlight recent studies that connect LXR-dependent control of lipid metabolism to regulation of the immune response.
Collapse
Affiliation(s)
- Ira G Schulman
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
137
|
Cimmino G, Loffredo FS, Morello A, D'Elia S, De Palma R, Cirillo P, Golino P. Immune-Inflammatory Activation in Acute Coronary Syndromes: A Look into the Heart of Unstable Coronary Plaque. Curr Cardiol Rev 2017; 13:110-117. [PMID: 27758696 PMCID: PMC5452145 DOI: 10.2174/1573403x12666161014093812] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/24/2016] [Accepted: 10/06/2016] [Indexed: 12/21/2022] Open
Abstract
In the last twenty years, our comprehension of the molecular mechanisms involved in the formation, progression and complication of atherosclerotic plaque has advanced significantly and the main role of inflammation and immunity in this phenomenon is now largely accepted. Accumulating evidence highlight the crucial role of different inflammatory and immune cells, such as monocytes and T-lymphocytes, in the pathophysiology of atherosclerotic lesion, particularly in contributing to its com-plications, such as rupture or ulceration. According to the new terminology, “vulnerable plaque” identi-fies an inflamed atherosclerotic lesion that is particularly prone to rupture. Once disrupted, prothrom-botic material is exposed to the flowing blood, thus activating coagulation cascade and platelet aggrega-tion, ultimately leading to acute thrombus formation within the coronary vessel. To date this is the key event underlying the clinical manifestations of acute coronary syndromes (ACS). The degree of vessel occlusion (complete vs. incomplete) and the time of blood flow cessation will define the severity of clinical picture. This phenomenon seems to be the final effect of a complex inter-action between different local and systemic factors, involving the degree of inflammation, type of cells infiltration and the rheological characteristics of blood flow at the site of plaque rupture, thrombogenic substrates within the atherosclerotic lesion and different soluble mediators, already present or acutely released in the circulating blood. This article will review currently available data on the pathophysiology of ACS, emphasizing the immunological and inflammatory aspects of vulnerable plaque. We may pos-tulate that intraplaque antigens and local microenvironment will define the immune-inflammatory re-sponse and cells phenotype, thus determining the severity of clinical manifestations.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| | - Francesco S Loffredo
- Molecular Cardiology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Alberto Morello
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| | - Saverio D'Elia
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| | - Raffaele De Palma
- Department of Clinical and Experimental Medicine, Section of Immunology, Second University of Naples, Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Golino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| |
Collapse
|
138
|
He H, Ghosh S, Yang H. Nanomedicines for dysfunctional macrophage-associated diseases. J Control Release 2017; 247:106-126. [PMID: 28057522 PMCID: PMC5360184 DOI: 10.1016/j.jconrel.2016.12.032] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/28/2016] [Indexed: 12/13/2022]
Abstract
Macrophages play vital functions in host inflammatory reaction, tissue repair, homeostasis and immunity. Dysfunctional macrophages have significant pathophysiological impacts on diseases such as cancer, inflammatory diseases (rheumatoid arthritis and inflammatory bowel disease), metabolic diseases (atherosclerosis, diabetes and obesity) and major infections like human immunodeficiency virus infection. In view of this common etiology in these diseases, targeting the recruitment, activation and regulation of dysfunctional macrophages represents a promising therapeutic strategy. With the advancement of nanotechnology, development of nanomedicines to efficiently target dysfunctional macrophages can strengthen the effectiveness of therapeutics and improve clinical outcomes. This review discusses the specific roles of dysfunctional macrophages in various diseases and summarizes the latest advances in nanomedicine-based therapeutics and theranostics for treating diseases associated with dysfunctional macrophages.
Collapse
Affiliation(s)
- Hongliang He
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA 23219, United States
| | - Shobha Ghosh
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States.
| | - Hu Yang
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA 23219, United States; Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, United States; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States.
| |
Collapse
|
139
|
Li G, Qiao W, Zhang W, Li F, Shi J, Dong N. The shift of macrophages toward M1 phenotype promotes aortic valvular calcification. J Thorac Cardiovasc Surg 2017; 153:1318-1327.e1. [PMID: 28283241 DOI: 10.1016/j.jtcvs.2017.01.052] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 12/31/2016] [Accepted: 01/25/2017] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The purpose of the present study was to comprehensively compare the phenotype profile of infiltrated macrophages in human noncalcified and calcific aortic valves, and to determine whether the shift of macrophage polarization modulates valvular calcification in vitro. METHODS Cell surface markers of macrophages and inflammatory cytokines expression in 90 cases of human noncalcified and calcific aortic valve leaflets were analyzed. The normal aortic valve interstitial cells were isolated and cultured in vitro. After incubation with nonconditioned medium and conditioned medium from unstimulated or lipopolysaccharide-stimulated U937 monocytes, valve interstitial cells were evaluated by osteogenic differentiation markers. RESULTS Infiltration of macrophages was enhanced in the calcific aortic valves, and M1 phenotype was the predominant macrophage subsets. In addition, both proinflammatory and anti-inflammatory cytokines were significantly upregulated in the calcific aortic valves. Furthermore, lipopolysaccharide-stimulated monocytes presented with increased expression of inducible nitric oxide synthase and high proportional CD11c-positive (M1) macrophages. Conditioned medium from unstimulated monocytes promoted the osteogenic differentiation of valve interstitial cells in vitro, as evidenced by increased markers such as bone morphogenetic protein 2, osteopontin, and alkaline phosphatase. Conditioned medium from M1 macrophages further enhanced valve interstitial cells calcification. Enzyme-linked immunosorbent assay showed that M1 phenotype macrophages secreted tumor necrosis factors α and interleukin 6, and neutralizing antibodies to these 2 proinflammatory cytokines attenuated induction of osteogenic differentiation and calcification by the conditioned media. CONCLUSIONS Both total numbers and polarization of macrophage influence the process of calcification in human aortic valve. The shift toward M1 phenotype might promote valve interstitial cell calcification.
Collapse
Affiliation(s)
- Geng Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wenjing Zhang
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
140
|
Xu R, Li C, Wu Y, Shen L, Ma J, Qian J, Ge J. Role of KCa3.1 Channels in Macrophage Polarization and Its Relevance in Atherosclerotic Plaque Instability. Arterioscler Thromb Vasc Biol 2017; 37:226-236. [PMID: 28062499 DOI: 10.1161/atvbaha.116.308461] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 12/05/2016] [Indexed: 12/22/2022]
Abstract
Objective—
Emerging evidence indicates that proinflammatory macrophage polarization imbalance plays a key role in atherosclerotic plaque progression and instability. The calcium-activated potassium channel KCa3.1 is critically involved in macrophage activation and function. However, the role of KCa3.1 in macrophage polarization is unknown. This study investigates the potential role of KCa3.1 in transcriptional regulation in macrophage polarization and its relationship to plaque instability.
Approach and Results—
Human monocytes were differentiated into macrophages using macrophage colony-stimulating factor. Macrophages were then polarized into proinflammatory M1 cells by interferon-γ and lipopolysaccharide and into alternative M2 macrophages by interleukin-4. A model for plaque instability was induced by combined partial ligation of the left renal artery and left common carotid artery in apolipoprotein E knockout mice. Significant upregulation of KCa3.1 expression was observed during the differentiation of human monocytes into macrophages. Blocking KCa3.1 significantly reduced the expression of proinflammatory genes during macrophages polarization. Further mechanistic studies indicated that blocking KCa3.1 inhibited macrophage differentiation toward the M1 phenotype by downregulating signal transducer and activator of transcription-1 phosphorylation. In animal models, KCa3.1 blockade therapy strikingly reduced the incidence of plaque rupture and luminal thrombus in carotid arteries, decreased the expression of markers associated with M1 macrophage polarization, and enhanced the expression of M2 markers within atherosclerotic lesions.
Conclusions—
These results suggest that blocking KCa3.1 suppresses plaque instability in advanced stages of atherosclerosis by inhibiting macrophage polarization toward an M1 phenotype.
Collapse
Affiliation(s)
- Rende Xu
- From the Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenguang Li
- From the Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yizhe Wu
- From the Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Shen
- From the Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianying Ma
- From the Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Juying Qian
- From the Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- From the Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
141
|
Brophy ML, Dong Y, Wu H, Rahman HNA, Song K, Chen H. Eating the Dead to Keep Atherosclerosis at Bay. Front Cardiovasc Med 2017; 4:2. [PMID: 28194400 PMCID: PMC5277199 DOI: 10.3389/fcvm.2017.00002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/12/2017] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is the primary cause of coronary heart disease (CHD), ischemic stroke, and peripheral arterial disease. Despite effective lipid-lowering therapies and prevention programs, atherosclerosis is still the leading cause of mortality in the United States. Moreover, the prevalence of CHD in developing countries worldwide is rapidly increasing at a rate expected to overtake those of cancer and diabetes. Prominent risk factors include the hardening of arteries and high levels of cholesterol, which lead to the initiation and progression of atherosclerosis. However, cell death and efferocytosis are critical components of both atherosclerotic plaque progression and regression, yet, few currently available therapies focus on these processes. Thus, understanding the causes of cell death within the atherosclerotic plaque, the consequences of cell death, and the mechanisms of apoptotic cell clearance may enable the development of new therapies to treat cardiovascular disease. Here, we review how endoplasmic reticulum stress and cholesterol metabolism lead to cell death and inflammation, how dying cells affect plaque progression, and how autophagy and the clearance of dead cells ameliorates the inflammatory environment of the plaque. In addition, we review current research aimed at alleviating these processes and specifically targeting therapeutics to the site of the plaque.
Collapse
Affiliation(s)
- Megan L Brophy
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Yunzhou Dong
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Hao Wu
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - H N Ashiqur Rahman
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Kai Song
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Hong Chen
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| |
Collapse
|
142
|
Liberale L, Dallegri F, Montecucco F, Carbone F. Pathophysiological relevance of macrophage subsets in atherogenesis. Thromb Haemost 2017; 117:7-18. [PMID: 27683760 DOI: 10.1160/th16-08-0593] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
Macrophages are highly heterogeneous and plastic cells. They were shown to play a critical role in all stages of atherogenesis, from the initiation to the necrotic core formation and plaque rupture. Lesional macrophages primarily derive from blood monocyte, but local macrophage proliferation as well as differentiation from smooth muscle cells have also been described. Within atherosclerotic plaques, macrophages rapidly respond to changes in the microenvironment, shifting between pro- (M1) or anti-inflammatory (M2) functional phenotypes. Furthermore, different stimuli have been associated with differentiation of newly discovered M2 subtypes: IL-4/IL-13 (M2a), immune-complex (M2b), IL-10/glucocorticoids (M2c), and adenosine receptor agonist (M2d). More recently, additional intraplaque macrophage phenotypes were also recognized in response to CXCL4 (M4), oxidized phospholipids (Mox), haemoglobin/haptoglobin complexes (HA-mac/M(Hb)), and heme (Mhem). Such macrophage polarization was described as a progression among multiple phenotypes, which reflect the activity of different transcriptional factors and the cross-talk between intracellular signalling. Finally, the distribution of macrophage subsets within different plaque areas was markedly associated with cardiovascular (CV) vulnerability. The aim of this review is to update the current knowledge on the role of macrophage subsets in atherogenesis. In addition, the molecular mechanisms underlying macrophage phenotypic shift will be summarised and discussed. Finally, the role of intraplaque macrophages as predictors of CV events and the therapeutic potential of these cells will be discussed.
Collapse
Affiliation(s)
| | | | - Fabrizio Montecucco
- Fabrizio Montecucco, MD, PhD, Department of Internal Medicine, University of Genoa School of Medicine, 6 viale Benedetto XV, 16132 Genoa, Italy, Tel.: +39 010 353 8694, Fax: +39 010 353 8686, E-mail:
| | | |
Collapse
|
143
|
Guo Y, Luo F, Yi Y, Xu D. Fibroblast growth factor 21 potentially inhibits microRNA-33 expression to affect macrophage actions. Lipids Health Dis 2016; 15:208. [PMID: 27905947 PMCID: PMC5134291 DOI: 10.1186/s12944-016-0381-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 11/24/2016] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease with complex pathological processes. MicroRNA-33 (miR-33), a novel non-coding RNA that coexpresses with sterol regulatory element-binding proteins (SREBPs), affects macrophage actions to prevent atherosclerosis. Fibroblast growth factor 21 (FGF21) is an important regulator of lipid metabolism, especially for macrophage-related cholesterol export, but the mechanism is not fully studied. Interestingly, FGF21 has been evidenced to prevent atherosclerosis via inhibiting SREBP-2 expression. Therefore, we speculate that FGF21 may be a potential regulator for miR-33 with an aim of insight into novel anti-atherosclerotic mechanisms and research fields.
Collapse
Affiliation(s)
- Yuan Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Fei Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Yuhong Yi
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| |
Collapse
|
144
|
Li HY, Su YY, Zhang YF, Liu ZQ, Hua BJ. Involvement of peroxisome proliferator activated receptor-γ in the anti-inflammatory effects of atorvastatin in oxygen-glucose deprivation/reperfusion-stimulated RAW264.7 murine macrophages. Mol Med Rep 2016; 14:4055-4062. [PMID: 27633957 PMCID: PMC5101877 DOI: 10.3892/mmr.2016.5742] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 06/03/2016] [Indexed: 02/05/2023] Open
Abstract
Ischemia-reperfusion (I/R) injury is important in the pathogenesis and/or progression of various diseases, including stroke, cardiovascular disease and acute renal injury. Increasing evidence indicates that atorvastatin exerts protective effects in I/R injury-associated diseases; however, the underlying mechanisms remain to be fully elucidated. In the present study, oxygen-glucose deprivation (OGD)/reperfusion-stimulated. RAW264.7 murine macrophages served as a model of I/R injury. The knockdown of peroxisome proliferator activated receptor-γ (PPARγ) expression in these cells increased OGD/reperfusion-induced expression of inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), and enhanced OGD/reperfusion-induced downregulation of the expression of cluster of differentiation (CD) 206, at the mRNA and protein levels. Conversely, overexpression of PPARγ significantly attenuated OGD/reperfusion-induced alterations in the expression of iNOS, TNF-α, IFN-γ and CD206 at the mRNA and protein levels. Notably, atorvastatin inhibited OGD/reperfusion-induced iNOS expression and reversed OGD/reperfusion-induced downregulation of the expression of CD206 and PPARγ at the mRNA and protein levels. The results of the present study indicate that atorvastatin exhibits significant anti-inflammatory effects in OGD/reperfusion-stimulated RAW264.7 cells, possibly via PPARγ regulation. The findings of the present study may reveal a novel mechanism underlying the protective effects of atorvastatin in I/R injury-associated diseases.
Collapse
Affiliation(s)
- Hong-Yan Li
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, Guangdong 510800, P.R. China
| | - Yan-Yan Su
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, Guangdong 510800, P.R. China
| | - Yun-Fang Zhang
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, Guangdong 510800, P.R. China
| | - Zhi-Qiang Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Bao-Jun Hua
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, Guangdong 510800, P.R. China
| |
Collapse
|
145
|
Myeloperoxidase-Oxidized LDLs Enhance an Anti-Inflammatory M2 and Antioxidant Phenotype in Murine Macrophages. Mediators Inflamm 2016; 2016:8249476. [PMID: 27656049 PMCID: PMC5021486 DOI: 10.1155/2016/8249476] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/28/2016] [Accepted: 08/02/2016] [Indexed: 12/02/2022] Open
Abstract
Macrophages and oxidized LDLs play a key role in atherogenesis but their heterogeneity has been neglected up to now. Macrophages are prone to polarization and subsets of polarized macrophages have been described in atheromas. LDLs can be oxidized not only chemically by copper (Ox-LDLs) but also enzymatically by myeloperoxidase (MpOx-LDLs) resulting in oxidized LDLs poor in lipid peroxides. The effects of physiologically relevant myeloperoxidase-oxidized LDLs on macrophage polarization or on polarized macrophages remain largely unknown. In this study, the effects of LDLs on macrophage polarization were investigated by monitoring the expression of M1 and M2 genes following stimulation with native LDLs, Ox-LDLs, or MpOx-LDLs in RAW 264.7 cells. Except for MRC1, which is induced only by Ox-LDLs, MpOx-LDLs induced an overexpression of most of the selected marker genes at the mRNA level. MpOx-LDLs also modulate marker gene expression in polarized macrophages favoring notably anti-inflammatory Arg1 expression in M2 cells and also in the other phenotypes. Noteworthy, MpOx-LDLs were the most efficient to accumulate lipids intracellularly in (un)polarized macrophages whatever the phenotype. These data were largely confirmed in murine bone marrow-derived macrophages. Our data suggest that MpOx-LDLs were the most efficient to accumulate within cells and to enhance an anti-inflammatory and antioxidant phenotype in M2 cells and also in the other macrophage phenotypes.
Collapse
|
146
|
Meshkani R, Vakili S. Tissue resident macrophages: Key players in the pathogenesis of type 2 diabetes and its complications. Clin Chim Acta 2016; 462:77-89. [PMID: 27570063 DOI: 10.1016/j.cca.2016.08.015] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/21/2016] [Accepted: 08/23/2016] [Indexed: 02/07/2023]
Abstract
There is increasing evidence showing that chronic inflammation is an important pathogenic mediator of the development of type 2 diabetes (T2D). It is now generally accepted that tissue-resident macrophages play a major role in regulation of tissue inflammation. T2D-associated inflammation is characterized by an increased abundance of macrophages in different tissues along with production of inflammatory cytokines. The complexity of macrophage phenotypes has been reported from different human tissues. Macrophages exhibit a phenotypic range that is intermediate between two extremes, M1 (pro-inflammatory) and M2 (anti-inflammatory). Cytokines and chemokines produced by macrophages generate local and systemic inflammation and this condition leads to pancreatic β-cell dysfunction and insulin resistance in liver, adipose and skeletal muscle tissues. Data from human and animal studies also suggest that macrophages contribute to T2D complications such as nephropathy, neuropathy, retinopathy and cardiovascular diseases through cell-cell interactions and the release of pro-inflammatory cytokines, chemokines, and proteases to induce inflammatory cell recruitment, cell apoptosis, angiogenesis, and matrix protein remodeling. In this review we focus on the functions of macrophages and the importance of these cells in the pathogenesis of T2D. In addition, the contribution of macrophages to diabetes complications such as nephropathy, neuropathy, retinopathy and cardiovascular diseases is discussed.
Collapse
Affiliation(s)
- Reza Meshkani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Sanaz Vakili
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| |
Collapse
|
147
|
Aryal B, Rotllan N, Araldi E, Ramírez CM, He S, Chousterman BG, Fenn AM, Wanschel A, Madrigal-Matute J, Warrier N, Martín-Ventura JL, Swirski FK, Suárez Y, Fernández-Hernando C. ANGPTL4 deficiency in haematopoietic cells promotes monocyte expansion and atherosclerosis progression. Nat Commun 2016; 7:12313. [PMID: 27460411 PMCID: PMC4974469 DOI: 10.1038/ncomms12313] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 06/21/2016] [Indexed: 12/27/2022] Open
Abstract
Lipid accumulation in macrophages has profound effects on macrophage gene expression and contributes to the development of atherosclerosis. Here, we report that angiopoietin-like protein 4 (ANGPTL4) is the most highly upregulated gene in foamy macrophages and it's absence in haematopoietic cells results in larger atherosclerotic plaques, characterized by bigger necrotic core areas and increased macrophage apoptosis. Furthermore, hyperlipidemic mice deficient in haematopoietic ANGPTL4 have higher blood leukocyte counts, which is associated with an increase in the common myeloid progenitor (CMP) population. ANGPTL4-deficient CMPs have higher lipid raft content, are more proliferative and less apoptotic compared with the wild-type (WT) CMPs. Finally, we observe that ANGPTL4 deficiency in macrophages promotes foam cell formation by enhancing CD36 expression and reducing ABCA1 localization in the cell surface. Altogether, these findings demonstrate that haematopoietic ANGPTL4 deficiency increases atherogenesis through regulating myeloid progenitor cell expansion and differentiation, foam cell formation and vascular inflammation. Angiopoietin-like 4 protein (ANGPTL4) is a regulator of lipoprotein metabolism whose role in atherosclerosis has been controversial. Here the authors show that ANGPTL4 deficiency in haematopoietic cells increases atherogenesis by promoting myeloid progenitor cell expansion and differentiation, foam cell formation and vascular inflammation.
Collapse
Affiliation(s)
- Binod Aryal
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520, USA.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA.,Departments of Medicine and Cell Biology, Leon H. Charney Division of Cardiology and Cell Biology, New York University School of Medicine, New York, New York 10016, USA
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520, USA.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Elisa Araldi
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520, USA.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Cristina M Ramírez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520, USA.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Shun He
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Benjamin G Chousterman
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Ashley M Fenn
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Amarylis Wanschel
- Departments of Medicine and Cell Biology, Leon H. Charney Division of Cardiology and Cell Biology, New York University School of Medicine, New York, New York 10016, USA
| | - Julio Madrigal-Matute
- Departments of Medicine and Cell Biology, Leon H. Charney Division of Cardiology and Cell Biology, New York University School of Medicine, New York, New York 10016, USA
| | - Nikhil Warrier
- Departments of Medicine and Cell Biology, Leon H. Charney Division of Cardiology and Cell Biology, New York University School of Medicine, New York, New York 10016, USA
| | - Jose L Martín-Ventura
- Vascular Research Lab, IIS-Fundación Jimenez-Díaz, Universidad Autónoma de Madrid, Madrid 28040, Spain
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520, USA.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520, USA.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
148
|
9-cis β-Carotene Increased Cholesterol Efflux to HDL in Macrophages. Nutrients 2016; 8:nu8070435. [PMID: 27447665 PMCID: PMC4963911 DOI: 10.3390/nu8070435] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/29/2016] [Accepted: 07/14/2016] [Indexed: 12/15/2022] Open
Abstract
Cholesterol efflux from macrophages is a key process in reverse cholesterol transport and, therefore, might inhibit atherogenesis. 9-cis-β-carotene (9-cis-βc) is a precursor for 9-cis-retinoic-acid (9-cis-RA), which regulates macrophage cholesterol efflux. Our objective was to assess whether 9-cis-βc increases macrophage cholesterol efflux and induces the expression of cholesterol transporters. Enrichment of a mouse diet with βc from the alga Dunaliella led to βc accumulation in peritoneal macrophages. 9-cis-βc increased the mRNA levels of CYP26B1, an enzyme that regulates RA cellular levels, indicating the formation of RA from βc in RAW264.7 macrophages. Furthermore, 9-cis-βc, as well as all-trans-βc, significantly increased cholesterol efflux to high-density lipoprotein (HDL) by 50% in RAW264.7 macrophages. Likewise, food fortification with 9-cis-βc augmented cholesterol efflux from macrophages ex vivo. 9-cis-βc increased both the mRNA and protein levels of ABCA1 and apolipoprotein E (APOE) and the mRNA level of ABCG1. Our study shows, for the first time, that 9-cis-βc from the diet accumulates in peritoneal macrophages and increases cholesterol efflux to HDL. These effects might be ascribed to transcriptional induction of ABCA1, ABCG1, and APOE. These results highlight the beneficial effect of βc in inhibition of atherosclerosis by improving cholesterol efflux from macrophages.
Collapse
|
149
|
Poulsen CB, Al-Mashhadi AL, von Wachenfeldt K, Bentzon JF, Nielsen LB, Al-Mashhadi RH, Thygesen J, Tolbod L, Larsen JR, Frøkiær J, Tawakol A, Vucic E, Fredrickson J, Baruch A, Frendéus B, Robertson AKL, Moestrup SK, Drouet L, Falk E. Treatment with a human recombinant monoclonal IgG antibody against oxidized LDL in atherosclerosis-prone pigs reduces cathepsin S in coronary lesions. Int J Cardiol 2016; 215:506-15. [PMID: 27135822 DOI: 10.1016/j.ijcard.2016.03.222] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 03/26/2016] [Indexed: 12/15/2022]
|
150
|
Cytokines: roles in atherosclerosis disease progression and potential therapeutic targets. Future Med Chem 2016; 8:1317-30. [PMID: 27357616 DOI: 10.4155/fmc-2016-0072] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis, the primary cause of cardiovascular disease (CVD), is a chronic inflammatory disorder in the walls of medium and large arteries. CVD is currently responsible for about one in three global deaths and this is expected to rise in the future due to an increase in the prevalence of obesity and diabetes. Current therapies for atherosclerosis mainly modulate lipid homeostasis and while successful at reducing the risk of a CVD-related death, they are associated with considerable residual risk and various side effects. There is, therefore, a need for alternative therapies aimed at regulating inflammation in order to reduce atherogenesis. This review will highlight the key role cytokines play during disease progression as well as potential therapeutic strategies to target them.
Collapse
|