1
|
Faneuff EE, Kim MJ, Blackman A, Karunakaran KA, Bader JE, Zhen X, Gallagher KS, Durst TJ, Connelly JA, Rathmell JC, Salina A, Martinez-Barricarte R, Serezani CH. PTEN inhibits scavenger receptor-mediated phagocytosis of methicillin-resistant Staphylococcus aureus. Immunohorizons 2025; 9:vlaf011. [PMID: 40288813 PMCID: PMC12034382 DOI: 10.1093/immhor/vlaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/18/2025] [Indexed: 04/29/2025] Open
Abstract
Phagocytosis requires the coordination of various classes of receptors and the activation of multiple signaling programs, culminating in actin cytoskeletal rearrangement and ingestion. Given the pleiotropic nature of the events necessary for proper microbial ingestion, identifying molecules that control distinct steps of phagocytosis could reveal potential strategies to enhance microbial clearance. PTEN is a lipid/protein phosphatase traditionally recognized as a tumor suppressor. While PTEN inhibits various arms of the innate immune response, its role during Staphylococcus aureus infection remains unclear. We hypothesize that PTEN inhibits the functions of scavenger receptors (SRs) and the actin cytoskeleton during methicillin-resistant S. aureus (MRSA) infection in macrophages. RNAseq analysis of PTEN KO immortalized bone marrow-derived macrophages (iBMDMs) unveiled increased expression of genes involved in actin polymerization, pathogen recognition, and SRs, which leads to enhanced MRSA phagocytosis in both iBMDMs and primary peritoneal macrophages lacking PTEN. PTEN is physically associated with 2 SRs, MARCO and CD36, and blocking these receptors prevents the increased phagocytosis seen in PTEN KO macrophages. PTEN binds to the actin depolymerizing factor cofilin-1 during infection, inhibiting F-actin (the essential form of actin for phagocytosis) while increasing G-actin pools. Cytometry by time of flight (CyTOF) analysis of human myeloid cell populations from a PTEN-haploinsufficient patient suggests that PTEN is necessary for generating specific monocyte and dendritic subclasses. This study identifies the role of PTEN in macrophage phagocytosis of a gram-positive pathogen and in the development of monocyte subsets. This highlights the spectrum of PTEN importance in host defense mechanisms in both murine and human phagocytes.
Collapse
Affiliation(s)
- Eden E Faneuff
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Min Joo Kim
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Amondrea Blackman
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kirti A Karunakaran
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jackie E Bader
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Xin Zhen
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kaitlyn S Gallagher
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tanner J Durst
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James A Connelly
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ana Salina
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ruben Martinez-Barricarte
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Carlos Henrique Serezani
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
2
|
Aerbajinai W, Zhu J, Chin K, Rodgers GP. Glia maturation factor-γ regulates amyloid-β42 phagocytosis through scavenger receptor class A type I in murine macrophages. J Leukoc Biol 2024; 117:qiae197. [PMID: 39243388 PMCID: PMC11685041 DOI: 10.1093/jleuko/qiae197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/05/2024] [Indexed: 09/09/2024] Open
Abstract
Dysfunctional phagocytic clearance of β-amyloid (Aβ) in microglia and peripheral macrophages/monocytes has been implicated in Alzheimer's disease, but the mechanisms underlying this dysfunction are not yet well understood. In this study, we examined the role of glia maturation factor-γ (GMFG), an actin-disassembly protein, i.e. highly expressed in immune cells, in macrophage Aβ phagocytosis and in regulating type I class A scavenger receptor, a cell-surface receptor that has previously been implicated in Aβ clearance. GMFG knockdown-increased phagocytosis of Aβ42 in bone marrow-derived macrophages and RAW264.7 murine macrophages, while GMFG overexpression reduced Aβ42 uptake in these cells. Blocking with anti-type I class A scavenger receptor antibodies inhibited Aβ42 uptake in GMFG-knockdown cells, establishing a role for type I class A scavenger receptor in Aβ42 phagocytosis. GMFG knockdown-increased type I class A scavenger receptor protein expression under both basal conditions and in response to Aβ42 treatment via both the transcriptional and posttranscriptional levels in RAW264.7 macrophages. GMFG knockdown modulated Aβ42-induced K48-linked and K63-polyubiquitination of type I class A scavenger receptor, the phosphorylation of type I class A scavenger receptor and c-Jun N-Terminal kinase (JNK), suggesting that GMFG plays a role for intracellular signaling in the type I class A scavenger receptor--mediated uptake of Aβ. Further, GMFG-knockdown cells displayed increased levels of the transcriptional factor MafB, and silencing of MafB in these cells reduced their type I class A scavenger receptor expression. Finally, GMFG was found to interact with the nuclear pore complex component RanBP2, and silencing of RanBP2 in GMFG-knockdown cells reduced their type I class A scavenger receptor expression. Collectively, these data support the role of GMFG as a novel regulator of type I class A scavenger receptor in macrophage Aβ phagocytosis and may provide insight into therapeutic approaches to potentially slow or prevent the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Wulin Aerbajinai
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, BG 10, RM 9N113, 10 Center Dr., Bethesda, MD 20892, United States
| | - Jianqiong Zhu
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, BG 10, RM 9N113, 10 Center Dr., Bethesda, MD 20892, United States
| | - Kyung Chin
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, BG 10, RM 9N113, 10 Center Dr., Bethesda, MD 20892, United States
| | - Griffin P Rodgers
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, BG 10, RM 9N113, 10 Center Dr., Bethesda, MD 20892, United States
| |
Collapse
|
3
|
Heydeck D, Kakularam KR, Labuz D, Machelska H, Rohwer N, Weylandt K, Kuhn H. Transgenic mice overexpressing human ALOX15 under the control of the aP2 promoter are partly protected in the complete Freund's adjuvant-induced paw inflammation model. Inflamm Res 2023; 72:1649-1664. [PMID: 37498393 PMCID: PMC10499711 DOI: 10.1007/s00011-023-01770-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND, OBJECTIVES AND DESIGN Arachidonic acid 15-lipoxygenase (ALOX15) has been implicated in the pathogenesis of inflammatory diseases but since pro- and anti-inflammatory roles have been suggested, the precise function of this enzyme is still a matter of discussion. To contribute to this discussion, we created transgenic mice, which express human ALOX15 under the control of the activating protein 2 promoter (aP2-ALOX15 mice) and compared the sensitivity of these gain-of-function animals in two independent mouse inflammation models with Alox15-deficient mice (loss-of-function animals) and wildtype control animals. MATERIALS AND METHODS Transgenic aP2-ALOX15 mice were tested in comparison with Alox15 knockout mice (Alox15-/-) and corresponding wildtype control animals (C57BL/6J) in the complete Freund's adjuvant induced hind-paw edema model and in the dextran sulfate sodium induced colitis (DSS-colitis) model. In the paw edema model, the degree of paw swelling and the sensitivity of the inflamed hind-paw for mechanic (von Frey test) and thermal (Hargreaves test) stimulation were quantified as clinical readout parameters. In the dextran sodium sulfate induced colitis model the loss of body weight, the colon lengths and the disease activity index were determined. RESULTS In the hind-paw edema model, systemic inactivation of the endogenous Alox15 gene intensified the inflammatory symptoms, whereas overexpression of human ALOX15 reduced the degree of hind-paw inflammation. These data suggest anti-inflammatory roles for endogenous and transgenic ALOX15 in this particular inflammation model. As mechanistic reason for the protective effect downregulation of the pro-inflammatory ALOX5 pathways was suggested. However, in the dextran sodium sulfate colitis model, in which systemic inactivation of the Alox15 gene protected female mice from DSS-induced colitis, transgenic overexpression of human ALOX15 did hardly impact the intensity of the inflammatory symptoms. CONCLUSION The biological role of ALOX15 in the pathogenesis of inflammation is variable and depends on the kind of the animal inflammation model.
Collapse
Affiliation(s)
- Dagmar Heydeck
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Kumar R. Kakularam
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Dominika Labuz
- Department of Experimental Anesthesiology, Charité ˗ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Halina Machelska
- Department of Experimental Anesthesiology, Charité ˗ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Nadine Rohwer
- Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, Medical Department B, Brandenburg Medical School, University Hospital Ruppin-Brandenburg, Fehrbelliner Straße 38, 16816 Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, Brandenburg Medical School and University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Karsten Weylandt
- Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, Medical Department B, Brandenburg Medical School, University Hospital Ruppin-Brandenburg, Fehrbelliner Straße 38, 16816 Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, Brandenburg Medical School and University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
| | - Hartmut Kuhn
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
4
|
Functional Characterization of Transgenic Mice Overexpressing Human 15-Lipoxygenase-1 (ALOX15) under the Control of the aP2 Promoter. Int J Mol Sci 2023; 24:ijms24054815. [PMID: 36902243 PMCID: PMC10003068 DOI: 10.3390/ijms24054815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Arachidonic acid lipoxygenases (ALOX) have been implicated in the pathogenesis of inflammatory, hyperproliferative, neurodegenerative, and metabolic diseases, but the physiological function of ALOX15 still remains a matter of discussion. To contribute to this discussion, we created transgenic mice (aP2-ALOX15 mice) expressing human ALOX15 under the control of the aP2 (adipocyte fatty acid binding protein 2) promoter, which directs expression of the transgene to mesenchymal cells. Fluorescence in situ hybridization and whole-genome sequencing indicated transgene insertion into the E1-2 region of chromosome 2. The transgene was highly expressed in adipocytes, bone marrow cells, and peritoneal macrophages, and ex vivo activity assays proved the catalytic activity of the transgenic enzyme. LC-MS/MS-based plasma oxylipidome analyses of the aP2-ALOX15 mice suggested in vivo activity of the transgenic enzyme. The aP2-ALOX15 mice were viable, could reproduce normally, and did not show major phenotypic alterations when compared with wildtype control animals. However, they exhibited gender-specific differences with wildtype controls when their body-weight kinetics were evaluated during adolescence and early adulthood. The aP2-ALOX15 mice characterized here can now be used for gain-of-function studies evaluating the biological role of ALOX15 in adipose tissue and hematopoietic cells.
Collapse
|
5
|
Chen Y, Yang X, Kitajima S, Quan L, Wang Y, Zhu M, Liu E, Lai L, Yan H, Fan J. Macrophage elastase derived from adventitial macrophages modulates aortic remodeling. Front Cell Dev Biol 2023; 10:1097137. [PMID: 36704203 PMCID: PMC9871815 DOI: 10.3389/fcell.2022.1097137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/26/2022] [Indexed: 01/12/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is pathologically characterized by intimal atherosclerosis, disruption and attenuation of the elastic media, and adventitial inflammatory infiltrates. Although all these pathological events are possibly involved in the pathogenesis of AAA, the functional roles contributed by adventitial inflammatory macrophages have not been fully documented. Recent studies have revealed that increased expression of matrix metalloproteinase-12 (MMP-12) derived from macrophages may be particularly important in the pathogenesis of both atherosclerosis and AAA. In the current study, we developed a carrageenan-induced abdominal aortic adventitial inflammatory model in hypercholesterolemic rabbits and evaluated the effect of adventitial macrophage accumulation on the aortic remodeling with special reference to the influence of increased expression of MMP-12. To accomplish this, we compared the carrageenan-induced aortic lesions of transgenic (Tg) rabbits that expressed high levels of MMP-12 in the macrophage lineage to those of non-Tg rabbits. We found that the aortic medial and adventitial lesions of Tg rabbits were greater in degree than those of non-Tg rabbits, with the increased infiltration of macrophages and prominent destruction of elastic lamellae accompanied by the frequent appearance of dilated lesions, while the intimal lesions were slightly increased. Enhanced aortic lesions in Tg rabbits were focally associated with increased dilation of the aortic lumens. RT-PCR and Western blotting revealed high levels of MMP-12 in the lesions of Tg rabbits that were accompanied by elevated levels of MMP-2 and -3, which was caused by increased number of macrophages. Our results suggest that adventitial inflammation constitutes a major stimulus to aortic remodeling and increased expression of MMP-12 secreted from adventitial macrophages plays an important role in the pathogenesis of vascular diseases such as AAA.
Collapse
Affiliation(s)
- Yajie Chen
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Xiawen Yang
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Shuji Kitajima
- Analytical Research Center for Experimental Sciences, Saga University, Saga, Japan
| | - Longquan Quan
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yao Wang
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Maobi Zhu
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease and Laboratory Animal Center, Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Liangxue Lai
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Haizhao Yan
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China,*Correspondence: Haizhao Yan, ; Jianglin Fan,
| | - Jianglin Fan
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan,*Correspondence: Haizhao Yan, ; Jianglin Fan,
| |
Collapse
|
6
|
Gudgeon J, Marín-Rubio JL, Trost M. The role of macrophage scavenger receptor 1 (MSR1) in inflammatory disorders and cancer. Front Immunol 2022; 13:1012002. [PMID: 36325338 PMCID: PMC9618966 DOI: 10.3389/fimmu.2022.1012002] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/28/2022] [Indexed: 08/27/2023] Open
Abstract
Macrophage scavenger receptor 1 (MSR1), also named CD204, holds key inflammatory roles in multiple pathophysiologic processes. Present primarily on the surface of various types of macrophage, this receptor variably affects processes such as atherosclerosis, innate and adaptive immunity, lung and liver disease, and more recently, cancer. As highlighted throughout this review, the role of MSR1 is often dichotomous, being either host protective or detrimental to the pathogenesis of disease. We will discuss the role of MSR1 in health and disease with a focus on the molecular mechanisms influencing MSR1 expression, how altered expression affects disease process and macrophage function, the limited cell signalling pathways discovered thus far, the emerging role of MSR1 in tumour associated macrophages as well as the therapeutic potential of targeting MSR1.
Collapse
Affiliation(s)
| | - José Luis Marín-Rubio
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Matthias Trost
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
7
|
Ponnusamy N, Arumugam M. Interaction of Host Pattern Recognition Receptors (PRRs) with Mycobacterium Tuberculosis and Ayurvedic Management of Tuberculosis: A Systemic Approach. Infect Disord Drug Targets 2022; 22:e130921196420. [PMID: 34517809 DOI: 10.2174/1871526521666210913110834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/15/2021] [Accepted: 06/11/2021] [Indexed: 06/13/2023]
Abstract
Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (Mtb), infects the lungs' alveolar surfaces through aerosol droplets. At this stage, the disease progression may have many consequences, determined primarily by the reactions of the human immune system. However, one approach will be to more actively integrate the immune system, especially the pattern recognition receptor (PRR) systems of the host, which notices pathogen-associated molecular patterns (PAMPs) of Mtb. Several types of PRRs are involved in the detection of Mtb, including Toll-like receptors (TLRs), C-type lectin receptors (CLRs), Dendritic cell (DC) -specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), Mannose receptor (MR), and NOD-like receptors (NLRs) related to inflammasome activation. In this study, we focus on reviewing the Mtb pathophysiology and interaction of host PPRs with Mtb as well as adverse drug effects of anti-tuberculosis drugs (ATDs) and systematic TB treatment via Ayurvedic medicine.
Collapse
Affiliation(s)
- Nirmaladevi Ponnusamy
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Mohanapriya Arumugam
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
8
|
Leerach N, Munesue S, Harashima A, Kimura K, Oshima Y, Kawano S, Tanaka M, Niimura A, Sakulsak N, Yamamoto H, Hori O, Yamamoto Y. RAGE signaling antagonist suppresses mouse macrophage foam cell formation. Biochem Biophys Res Commun 2021; 555:74-80. [PMID: 33813279 DOI: 10.1016/j.bbrc.2021.03.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 01/23/2023]
Abstract
The engagement of the receptor for advanced glycation end-products (receptor for AGEs, RAGE) with diverse ligands could elicit chronic vascular inflammation, such as atherosclerosis. Binding of cytoplasmic tail RAGE (ctRAGE) to diaphanous-related formin 1 (Diaph1) is known to yield RAGE intracellular signal transduction and subsequent cellular responses. However, the effectiveness of an inhibitor of the ctRAGE/Diaph1 interaction in attenuating the development of atherosclerosis is unclear. In this study, using macrophages from Ager+/+ and Ager-/- mice, we validated the effects of an inhibitor on AGEs-RAGE-induced foam cell formation. The inhibitor significantly suppressed AGEs-RAGE-evoked Rac1 activity, cell invasion, and uptake of oxidized low-density lipoprotein, as well as AGEs-induced NF-κB activation and upregulation of proinflammatory gene expression. Moreover, expression of Il-10, an anti-inflammatory gene, was restored by this antagonist. These findings suggest that the RAGE-Diaph1 inhibitor could be a potential therapeutic drug against RAGE-related diseases, such as chronic inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Nontaphat Leerach
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Seiichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Ai Harashima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Kumi Kimura
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Yu Oshima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Shuhei Kawano
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Mariko Tanaka
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Akane Niimura
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Natthiya Sakulsak
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Hiroshi Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan; Komatsu University, Komatsu, 923-0921, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan.
| |
Collapse
|
9
|
Peng H, Guo Q, Su T, Xiao Y, Li CJ, Huang Y, Luo XH. Identification of SCARA3 with potential roles in metabolic disorders. Aging (Albany NY) 2020; 13:2149-2167. [PMID: 33318306 PMCID: PMC7880357 DOI: 10.18632/aging.202228] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/22/2020] [Indexed: 04/11/2023]
Abstract
Obesity is characterized by the expansion of adipose tissue which is partially modulated by adipogenesis. In the present study, we identified five differentially expressed genes by incorporating two adipogenesis-related datasets from the GEO database and their correlation with adipogenic markers. However, the role of scavenger receptor class A member 3 (SCARA3) in obesity-related disorders has been rarely reported. We found that Scara3 expression in old adipose tissue-derived mesenchymal stem cells (Ad-MSCs) was lower than it in young Ad-MSCs. Obese mice caused by deletion of the leptin receptor gene (db/db) or by a high-fat diet both showed reduced Scara3 expression in inguinal white adipose tissue. Moreover, hypermethylation of SCARA3 was observed in patients with type 2 diabetes and atherosclerosis. Data from the CTD database indicated that SCARA3 is a potential target for metabolic diseases. Mechanistically, JUN was predicted as a transcriptional factor of SCARA3 in different databases which is consistent with our further bioinformatics analysis. Collectively, our study suggested that SCARA3 is potentially associated with age-related metabolic dysfunction, which provided new insights into the pathogenesis and treatment of obesity as well as other obesity-associated metabolic complications.
Collapse
Affiliation(s)
- Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
10
|
Morita Y, Senokuchi T, Yamada S, Wada T, Furusho T, Matsumura T, Ishii N, Nishida S, Nishida S, Motoshima H, Komohara Y, Yamagata K, Araki E. Impact of tissue macrophage proliferation on peripheral and systemic insulin resistance in obese mice with diabetes. BMJ Open Diabetes Res Care 2020; 8:8/1/e001578. [PMID: 33087339 PMCID: PMC7580054 DOI: 10.1136/bmjdrc-2020-001578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/06/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Obesity-related insulin resistance is a widely accepted pathophysiological feature in type 2 diabetes. Systemic metabolism and immunity are closely related, and obesity represents impaired immune function that predisposes individuals to systemic chronic inflammation. Increased macrophage infiltration and activation in peripheral insulin target tissues in obese subjects are strongly related to insulin resistance. Using a macrophage-specific proliferation inhibition mouse model (mac-p27Tg), we previously reported that suppressed plaque inflammation reduced atherosclerosis and improved plaque stabilization. However, the direct evidence that proliferating macrophages are responsible for inducing insulin resistance was not provided. RESEARCH DESIGN AND METHODS The mac-p27Tg mice were fed a high-fat diet, and glucose metabolism, histological changes, macrophage polarization, and tissue functions were investigated to reveal the significance of tissue macrophage proliferation in insulin resistance and obesity. RESULTS The mac-p27Tg mice showed improved glucose tolerance and insulin sensitivity, along with a decrease in the number and ratio of inflammatory macrophages. Obesity-induced inflammation and oxidative stress was attenuated in white adipose tissue, liver, and gastrocnemius. Histological changes related to insulin resistance, such as liver steatosis/fibrosis, adipocyte enlargement, and skeletal muscle fiber transformation to fast type, were ameliorated in mac-p27Tg mice. Serum tumor necrosis factor alpha and free fatty acid were decreased, which might partially impact improved insulin sensitivity and histological changes. CONCLUSIONS Macrophage proliferation in adipose tissue, liver, and skeletal muscle was involved in promoting the development of systemic insulin resistance. Controlling the number of tissue macrophages by inhibiting macrophage proliferation could be a therapeutic target for insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Yutaro Morita
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Takafumi Senokuchi
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Sarie Yamada
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Toshiaki Wada
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Tatsuya Furusho
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Takeshi Matsumura
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Norio Ishii
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Saiko Nishida
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Syuhei Nishida
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Hiroyuki Motoshima
- Department of Metabolic Medicine and Endocrinology, Kikuchi Medical Association Hospital, Kikuchi, Kumamoto, Japan
| | - Yoshihiro Komohara
- Cell Pathology Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Kazuya Yamagata
- Medical Biochemistry, Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| | - Eiichi Araki
- Department of Metabolic Medicine Faculty of Life Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto, Japan
| |
Collapse
|
11
|
Post GR, Yuan Y, Holthoff ER, Quick CM, Post SR. Identification of a novel monocytic phenotype in Classic Hodgkin Lymphoma tumor microenvironment. PLoS One 2019; 14:e0224621. [PMID: 31714922 PMCID: PMC6850552 DOI: 10.1371/journal.pone.0224621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/17/2019] [Indexed: 01/07/2023] Open
Abstract
Classic Hodgkin lymphoma (CHL) characteristically shows few malignant cells in a microenvironment comprised of mixed inflammatory cells. Although CHL is associated with a high cure rate, recent studies have associated poor prognosis with absolute monocyte count in peripheral blood and increased monocyte/macrophages in involved lymph nodes. Thus, the role of monocytic infiltration and macrophage differentiation in the tumor microenvironment of CHL may be more relevant than absolute macrophage numbers to defining prognosis in CHL patients and potentially have therapeutic implications. Most studies identify tumor-associated macrophages (TAMs) using markers (e.g., CD68) expressed by macrophages and other mononuclear phagocytes, such as monocytes. In contrast, Class A Scavenger Receptor (SR-A/CD204) is expressed by tissue macrophages but not monocytic precursors. In this study, we examined SR-A expression in CHL (n = 43), and compared its expression with that of other macrophage markers. We confirmed a high prevalence of mononuclear cells that stained with CD68, CD163, and CD14 in CHL lymph nodes. However, SR-A protein expression determined by immunohistochemistry was limited to macrophages localized in sclerotic bands characteristic of nodular sclerosis CHL. In contrast, SR-A protein was readily detectable in lymph nodes with metastatic tumor, extra-nodal CHL, T cell/histiocyte-rich large B cell lymphoma, and resident macrophages in non-malignant tissues, including spleen, lymph node, liver and lung. The results of SR-A protein expression paralleled the expression of SR-A mRNA determined by quantitative RT-PCR. These data provide evidence that tumor-infiltrating monocyte/macrophages in CHL have a unique phenotype that likely depends on the microenvironment of nodal CHL.
Collapse
Affiliation(s)
- Ginell R. Post
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Youzhong Yuan
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Emily R. Holthoff
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Charles M. Quick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Steven R. Post
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
12
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
13
|
Cheng N, Liang Y, Du X, Ye RD. Serum amyloid A promotes LPS clearance and suppresses LPS-induced inflammation and tissue injury. EMBO Rep 2018; 19:embr.201745517. [PMID: 30126923 DOI: 10.15252/embr.201745517] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 07/24/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022] Open
Abstract
Lipopolysaccharide (LPS) is a major microbial mediator for tissue injury and sepsis resulting from Gram-negative bacterial infection. LPS is an external factor that induces robust expression of serum amyloid A (SAA), a major constituent of the acute-phase proteins, but the relationship between SAA expression and LPS-induced tissue injury remains unclear. Here, we report that mice with inducible transgenic expression of human SAA1 are partially protected against inflammatory response and lung injury caused by LPS and cecal ligation and puncture (CLP). In comparison, transgenic SAA1 does not attenuate TNFα-induced lung inflammation and injury. The SAA1 expression level correlates inversely with the endotoxin concentrations in serum and lung tissues since SAA1 binds directly to LPS to form a complex that promotes LPS uptake by macrophages. Disruption of the SAA1-LPS interaction with a SAA1-derived peptide partially reduces the protective effect and exacerbates inflammation. These findings demonstrate that acute-phase SAA provides innate feedback protection against LPS-induced inflammation and tissue injury.
Collapse
Affiliation(s)
- Ni Cheng
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Yurong Liang
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Xiaoping Du
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Richard D Ye
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA .,State Key Laboratory for Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region, China
| |
Collapse
|
14
|
Cheng N, Liang Y, Du X, Ye RD. Serum amyloid A promotes
LPS
clearance and suppresses
LPS
‐induced inflammation and tissue injury. EMBO Rep 2018. [DOI: 10.15252/embr.201745517 (e45517):14 pp] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Ni Cheng
- Department of Pharmacology and Center for Lung and Vascular Biology College of Medicine University of Illinois Chicago IL USA
| | - Yurong Liang
- Department of Pharmacology and Center for Lung and Vascular Biology College of Medicine University of Illinois Chicago IL USA
| | - Xiaoping Du
- Department of Pharmacology and Center for Lung and Vascular Biology College of Medicine University of Illinois Chicago IL USA
| | - Richard D Ye
- Department of Pharmacology and Center for Lung and Vascular Biology College of Medicine University of Illinois Chicago IL USA
- State Key Laboratory for Quality Research in Chinese Medicine Institute of Chinese Medical Sciences University of Macau Macau Special Administrative Region China
| |
Collapse
|
15
|
Cui Z, Liao J, Cheong N, Longoria C, Cao G, DeLisser HM, Savani RC. The Receptor for Hyaluronan-Mediated Motility (CD168) promotes inflammation and fibrosis after acute lung injury. Matrix Biol 2018; 78-79:255-271. [PMID: 30098420 PMCID: PMC6368477 DOI: 10.1016/j.matbio.2018.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 07/09/2018] [Accepted: 08/04/2018] [Indexed: 12/15/2022]
Abstract
Acute lung injury results in early inflammation and respiratory distress, and later fibrosis. The glycosaminoglycan hyaluronan (HA) and the Receptor for Hyaluronan-Mediated Motility (RHAMM, CD168) have been implicated in the response to acute lung injury. We hypothesized that, compared to wild type (WT) mice, RHAMM knockout (KO) mice would be protected from, whereas mice with macrophage-specific transgenic overexpression of RHAMM (TG) would have worse inflammation, respiratory distress and fibrosis after intratracheal (IT) bleomycin. Compared to WT mice, 10 days after IT bleomycin, RHAMM KO mice had less weight loss, less increase in respiratory rate, and fewer CD45+ cells in the lung. At day 28, compared to injured WT animals, injured RHAMM KO mice had lower M1 macrophage content, as well as decreased fibrosis as determined by trichrome staining, Ashcroft scores and lung HPO content. Four lines of transgenic mice with selective overexpression of RHAMM in macrophages were generated using the Scavenger Receptor A promoter driving a myc-tagged full length RHAMM cDNA. Baseline expression of RHAMM and CD44 was the same in WT and TG mice. By flow cytometry, TG bone marrow-derived macrophages (BMDM) had increased cell surface RHAMM and myc, but equal CD44 expression. TG BMDM also had 2-fold increases in both chemotaxis to HA and proliferation in fetal bovine serum. In TG mice, increased inflammation after thioglycollate-induced peritonitis was restricted to macrophages and not neutrophils. For lung injury studies, non-transgenic mice given bleomycin had respiratory distress with increased respiratory rates from day 7 to 21. However, TG mice had higher respiratory rates from 4 days after bleomycin and continued to increase respiratory rates up to day 21. At 21 days after IT bleomycin, TG mice had increased lung macrophage accumulation. Lavage HA concentrations were 6-fold higher in injured WT mice, but 30-fold higher in injured TG mice. At 21 days after IT bleomycin, WT mice had developed fibrosis, but TG mice showed exaggerated fibrosis with increased Ashcroft scores and HPO content. We conclude that RHAMM is a critical component of the inflammatory response, respiratory distress and fibrosis after acute lung injury. We speculate that RHAMM is a potential therapeutic target to limit the consequences of acute lung injury.
Collapse
Affiliation(s)
- Zheng Cui
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Jie Liao
- Center for Pulmonary & Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Naeun Cheong
- Center for Pulmonary & Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher Longoria
- Center for Pulmonary & Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gaoyuan Cao
- Perelmen Center for Advanced Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Horace M DeLisser
- Perelmen Center for Advanced Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Rashmin C Savani
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, USA; Center for Pulmonary & Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
16
|
Zanoni P, Velagapudi S, Yalcinkaya M, Rohrer L, von Eckardstein A. Endocytosis of lipoproteins. Atherosclerosis 2018; 275:273-295. [PMID: 29980055 DOI: 10.1016/j.atherosclerosis.2018.06.881] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
During their metabolism, all lipoproteins undergo endocytosis, either to be degraded intracellularly, for example in hepatocytes or macrophages, or to be re-secreted, for example in the course of transcytosis by endothelial cells. Moreover, there are several examples of internalized lipoproteins sequestered intracellularly, possibly to exert intracellular functions, for example the cytolysis of trypanosoma. Endocytosis and the subsequent intracellular itinerary of lipoproteins hence are key areas for understanding the regulation of plasma lipid levels as well as the biological functions of lipoproteins. Indeed, the identification of the low-density lipoprotein (LDL)-receptor and the unraveling of its transcriptional regulation led to the elucidation of familial hypercholesterolemia as well as to the development of statins, the most successful therapeutics for lowering of cholesterol levels and risk of atherosclerotic cardiovascular diseases. Novel limiting factors of intracellular trafficking of LDL and the LDL receptor continue to be discovered and to provide drug targets such as PCSK9. Surprisingly, the receptors mediating endocytosis of high-density lipoproteins or lipoprotein(a) are still a matter of controversy or even new discovery. Finally, the receptors and mechanisms, which mediate the uptake of lipoproteins into non-degrading intracellular itineraries for re-secretion (transcytosis, retroendocytosis), storage, or execution of intracellular functions, are largely unknown.
Collapse
Affiliation(s)
- Paolo Zanoni
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Srividya Velagapudi
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Mustafa Yalcinkaya
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Lucia Rohrer
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
17
|
Yamada S, Senokuchi T, Matsumura T, Morita Y, Ishii N, Fukuda K, Murakami-Nishida S, Nishida S, Kawasaki S, Motoshima H, Furukawa N, Komohara Y, Fujiwara Y, Koga T, Yamagata K, Takeya M, Araki E. Inhibition of Local Macrophage Growth Ameliorates Focal Inflammation and Suppresses Atherosclerosis. Arterioscler Thromb Vasc Biol 2018; 38:994-1006. [PMID: 29496659 DOI: 10.1161/atvbaha.117.310320] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 02/18/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Macrophages play a central role in various stages of atherosclerotic plaque formation and progression. The local macrophages reportedly proliferate during atherosclerosis, but the pathophysiological significance of macrophage proliferation in this context remains unclear. Here, we investigated the involvement of local macrophage proliferation during atherosclerosis formation and progression using transgenic mice, in which macrophage proliferation was specifically suppressed. APPROACH AND RESULTS Inhibition of macrophage proliferation was achieved by inducing the expression of cyclin-dependent kinase inhibitor 1B, also known as p27kip, under the regulation of a scavenger receptor promoter/enhancer. The macrophage-specific human p27kip Tg mice were subsequently crossed with apolipoprotein E-deficient mice for the atherosclerotic plaque study. Results showed that a reduced number of local macrophages resulted in marked suppression of atherosclerotic plaque formation and inflammatory response in the plaque. Moreover, fewer local macrophages in macrophage-specific human p27kip Tg mice helped stabilize the plaque, as evidenced by a reduced necrotic core area, increased collagenous extracellular matrix, and thickened fibrous cap. CONCLUSIONS These results provide direct evidence of the involvement of local macrophage proliferation in formation and progression of atherosclerotic plaques and plaque stability. Thus, control of macrophage proliferation might represent a therapeutic target for treating atherosclerotic diseases.
Collapse
Affiliation(s)
- Sarie Yamada
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Takafumi Senokuchi
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Takeshi Matsumura
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Yutaro Morita
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Norio Ishii
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Kazuki Fukuda
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Saiko Murakami-Nishida
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Shuhei Nishida
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Shuji Kawasaki
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Hiroyuki Motoshima
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | - Noboru Furukawa
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| | | | | | - Tomoaki Koga
- Department of Medical Cell Biology (T.K.), Faculty of Life Sciences, Kumamoto University, Japan
| | | | | | - Eiichi Araki
- From the Department of Metabolic Medicine (S.Y., T.S., T.M., Y.M., N.I., K.F., S.M.-N., S.N., S.K., H.M., N.F., E.A.)
| |
Collapse
|
18
|
Gupta RK, Gupta K, Sharma A, Das M, Ansari IA, Dwivedi PD. Maillard reaction in food allergy: Pros and cons. Crit Rev Food Sci Nutr 2017; 58:208-226. [PMID: 26980434 DOI: 10.1080/10408398.2016.1152949] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Food allergens have a notable potential to induce various health concerns in susceptible individuals. The majority of allergenic foods are usually subjected to thermal processing prior to their consumption. However, during thermal processing and long storage of foods, Maillard reaction (MR) often takes place. The MR is a non-enzymatic glycation reaction between the carbonyl group of reducing sugars and compounds having free amino groups. MR may sometimes be beneficial by damaging epitope of allergens and reducing allergenic potential, while exacerbation in allergic reactions may also occur due to changes in the motifs of epitopes or neoallergen generation. Apart from these modulations, non-enzymatic glycation can also modify the food protein(s) with various type of advance glycation end products (AGEs) such as Nϵ-(carboxymethyl-)lysine (CML), pentosidine, pyrraline, and methylglyoxal-H1 derived from MR. These Maillard products may act as immunogen by inducing the activation and proliferation of various immune cells. Literature is available to understand pathogenesis of glycation in the context of various diseases but there is hardly any review that can provide a thorough insight on the impact of glycation in food allergy. Therefore, present review explores the pathogenesis with special reference to food allergy caused by non-enzymatic glycation as well as AGEs.
Collapse
Affiliation(s)
- Rinkesh Kumar Gupta
- a Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research , Lucknow -, India.,b Department of Biosciences , Integral University , Lucknow , India
| | - Kriti Gupta
- a Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research , Lucknow -, India
| | - Akanksha Sharma
- a Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research , Lucknow -, India.,c Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Capmus , Lucknow , India
| | - Mukul Das
- a Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research , Lucknow -, India
| | | | | |
Collapse
|
19
|
Macrophage-specific overexpression of interleukin-5 attenuates atherosclerosis in LDL receptor-deficient mice. Gene Ther 2015; 22:645-52. [PMID: 25871825 DOI: 10.1038/gt.2015.33] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 03/24/2015] [Accepted: 04/07/2015] [Indexed: 12/19/2022]
Abstract
Interleukin-5 (IL-5) increases the secretion of natural T15/EO6 IgM antibodies that inhibit the uptake of oxidized low-density lipoprotein (LDL) by macrophages. This study aimed to determine whether macrophage-specific expression of IL-5 in LDL receptor-deficient mice (Ldlr(-/-)) could improve cholesterol metabolism and reduce atherosclerosis. To induce macrophage-specific IL-5 expression, the pLVCD68-IL5 lentivirus was delivered into Ldlr(-/-) mice via bone marrow transplantation. The recipient mice were fed a Western-type diet for 12 weeks to induce lesion formation. We found that IL-5 was efficiently and specifically overexpressed in macrophages in recipients of pLVCD68-IL5-transduced bone marrow cells (BMC). Plasma titers of T15/EO6 IgM antibodies were significantly elevated by 58% compared with control mice transplanted with pLVCD68 lacking the IL-5 coding sequence. Plaque areas of aortas in IL-5-overexpressing mice were reduced by 43% and associated with a 2.4-fold decrease in lesion size at the aortic roots when compared with mice receiving pLVCD68-transduced BMCs. The study showed that macrophage-specific overexpression of IL-5 inhibited the progression of atherosclerotic lesions. These findings suggest that modulation of IL-5 cytokine expression represents a potential strategy for intervention of familial hypercholesterolemia and other cardiovascular diseases.
Collapse
|
20
|
Kang WS, Kwon JS, Kim HB, Jeong HY, Kang HJ, Jeong MH, Cho JG, Park JC, Kim YS, Ahn Y. A macrophage-specific synthetic promoter for therapeutic application of adiponectin. Gene Ther 2014; 21:353-62. [PMID: 24500526 PMCID: PMC3975813 DOI: 10.1038/gt.2014.3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/19/2013] [Accepted: 12/16/2013] [Indexed: 11/09/2022]
Abstract
Foam cell formation from macrophage is a major cause of atherosclerosis. An efficient macrophage-specific promoter is required for the targeting to macrophages. In this study, we develop a macrophage-specific synthetic promoter for the therapeutic application of adiponectin (APN), an antiatherogenic gene. Synthetic promoter-146 (SP146), registered on the NCBI website (http://www.ncbi.nlm.nih.gov/nuccore/DQ107383), was tested for promoter activities in two non-macrophage cell lines (293 T, HeLa) and a macrophage cell line (RAW264.7, bone marrow-derived macrophages). To enforce macrophage specificity, partial elements of p47(phox) including the PU.1 site with various lengths (-C1, -C2 and -C3) were inserted next to the synthetic promoters. SP146-C1 showed the highest specificity and efficacy in RAW264.7 cells and was selected for development of an APN-carrying macrophage-specific promoter. Green fluorescent protein (GFP)- or APN-expressing lentivirus under SP146-C1 (Lenti-SP-GFP or Lenti-SP-APN, respectively) showed the highest expression efficacy in RAW264.7 cells compared with the non-macrophage cell lines. APN overexpression in RAW264.7 cells successfully inhibited intracellular lipid accumulation, and atherosclerotic lesions and lipid accumulation were significantly reduced by Lenti-SP-APN in ApoE-/- atherosclerosis mice. In conclusion, the synthetic promoter SP146-C1, combined with a p47(phox) promoter element, was successfully developed to target macrophage, and macrophage-specific introduction of APN under SP146-C1 was shown to ameliorate the atherosclerotic pathology.
Collapse
Affiliation(s)
- W S Kang
- 1] Research Laboratory of Cardiovascular Regeneration, Chonnam National University Hospital, Gwangju, Korea [2] Center of Molecular Medicine, Graduate School, Chonnam National University, Gwangju, Korea
| | - J S Kwon
- 1] Research Laboratory of Cardiovascular Regeneration, Chonnam National University Hospital, Gwangju, Korea [2] Heart Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - H B Kim
- Research Laboratory of Cardiovascular Regeneration, Chonnam National University Hospital, Gwangju, Korea
| | - H-Y Jeong
- Research Laboratory of Cardiovascular Regeneration, Chonnam National University Hospital, Gwangju, Korea
| | - H J Kang
- Research Laboratory of Cardiovascular Regeneration, Chonnam National University Hospital, Gwangju, Korea
| | - M H Jeong
- 1] Heart Research Center, Chonnam National University Hospital, Gwangju, Korea [2] Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - J G Cho
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - J C Park
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Y S Kim
- 1] Research Laboratory of Cardiovascular Regeneration, Chonnam National University Hospital, Gwangju, Korea [2] Heart Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Y Ahn
- 1] Research Laboratory of Cardiovascular Regeneration, Chonnam National University Hospital, Gwangju, Korea [2] Heart Research Center, Chonnam National University Hospital, Gwangju, Korea [3] Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| |
Collapse
|
21
|
Liu Z, Wang J, Huang E, Gao S, Li H, Lu J, Tian K, Little PJ, Shen X, Xu S, Liu P. Tanshinone IIA suppresses cholesterol accumulation in human macrophages: role of heme oxygenase-1. J Lipid Res 2013; 55:201-13. [PMID: 24302760 DOI: 10.1194/jlr.m040394] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Accumulation of foam cells in the neointima represents a key event in atherosclerosis. We previously demonstrated that Tanshinone IIA (Tan), a lipophilic bioactive compound extracted from Salvia miltiorrhiza Bunge, inhibits experimental atherogenesis, yet the detailed mechanisms are not fully understood. In this study, we sought to explore the potential effects of Tan on lipid accumulation in macrophage foam cells and the underlying molecular mechanisms. Our data indicate that Tan treatment reduced the content of macrophages, cholesterol accumulation, and the development of atherosclerotic plaque in apolipoprotein E-deficient mice. In human macrophages, Tan ameliorated oxidized low density lipoporotein (oxLDL)-elicited foam cell formation by inhibiting oxLDL uptake and promoting cholesterol efflux. Mechanistically, Tan markedly reduced the expression of scavenger receptor class A and increased the expression of ATP-binding cassette transporter A1 (ABCA1) and ABCG1 in lipid-laden macrophages via activation of the extracellular signal-regulated kinase (ERK)/nuclear factor-erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. Tan treatment induced the phosphorylation and nuclear translocation of Nrf2 and subsequently increased the expression of HO-1, and these effects were abolished by the specific ERK inhibitors, PD98059 and U0126. Moreover, HO-1 small interfering RNA or zinc protoporphyrin (a HO-1 inhibitor) abrogated Tan-mediated suppression of lipid accumulation in macrophages. Our current findings demonstrate that a novel HO-1-dependent mechanism is involved in the regulation of cholesterol balance by Tan.
Collapse
Affiliation(s)
- Zhiping Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shi Y, Gochuico BR, Yu G, Tang X, Osorio JC, Fernandez IE, Risquez CF, Patel AS, Shi Y, Wathelet MG, Goodwin AJ, Haspel JA, Ryter SW, Billings EM, Kaminski N, Morse D, Rosas IO. Syndecan-2 exerts antifibrotic effects by promoting caveolin-1-mediated transforming growth factor-β receptor I internalization and inhibiting transforming growth factor-β1 signaling. Am J Respir Crit Care Med 2013; 188:831-41. [PMID: 23924348 DOI: 10.1164/rccm.201303-0434oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
RATIONALE Alveolar transforming growth factor (TGF)-β1 signaling and expression of TGF-β1 target genes are increased in patients with idiopathic pulmonary fibrosis (IPF) and in animal models of pulmonary fibrosis. Internalization and degradation of TGF-β receptor TβRI inhibits TGF-β signaling and could attenuate development of experimental lung fibrosis. OBJECTIVES To demonstrate that after experimental lung injury, human syndecan-2 confers antifibrotic effects by inhibiting TGF-β1 signaling in alveolar epithelial cells. METHODS Microarray assays were performed to identify genes differentially expressed in alveolar macrophages of patients with IPF versus control subjects. Transgenic mice that constitutively overexpress human syndecan-2 in macrophages were developed to test the antifibrotic properties of syndecan-2. In vitro assays were performed to determine syndecan-2-dependent changes in epithelial cell TGF-β1 signaling, TGF-β1, and TβRI internalization and apoptosis. Wild-type mice were treated with recombinant human syndecan-2 during the fibrotic phase of bleomycin-induced lung injury. MEASUREMENTS AND MAIN RESULTS We observed significant increases in alveolar macrophage syndecan-2 levels in patients with IPF. Macrophage-specific overexpression of human syndecan-2 in transgenic mice conferred antifibrotic effects after lung injury by inhibiting TGF-β1 signaling and downstream expression of TGF-β1 target genes, reducing extracellular matrix production and alveolar epithelial cell apoptosis. In vitro, syndecan-2 promoted caveolin-1-dependent internalization of TGF-β1 and TβRI in alveolar epithelial cells, which inhibited TGF-β1 signaling and epithelial cell apoptosis. Therapeutic administration of human syndecan-2 abrogated lung fibrosis in mice. CONCLUSIONS Alveolar macrophage syndecan-2 exerts antifibrotic effects by promoting caveolin-1-dependent TGF-β1 and TβRI internalization and inhibiting TGF-β1 signaling in alveolar epithelial cells. Hence, molecules that facilitate TβRI degradation via endocytosis represent potential therapies for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yuanyuan Shi
- 1 Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sato-Nishiwaki M, Aida Y, Abe S, Shibata Y, Kimura T, Yamauchi K, Kishi H, Igarashi A, Inoue S, Sato M, Nakajima O, Kubota I. Reduced number and morphofunctional change of alveolar macrophages in MafB gene-targeted mice. PLoS One 2013; 8:e73963. [PMID: 24040127 PMCID: PMC3765310 DOI: 10.1371/journal.pone.0073963] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 07/25/2013] [Indexed: 01/19/2023] Open
Abstract
Alveolar macrophages (AMs) play an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD). We previously demonstrated that the transcription factor, MafB, increased in the AMs of mice exposed to cigarette smoke, and in those of human patients with COPD. The aim of this study was to evaluate the role of MafB in AMs using newly established transgenic (TG) mice that specifically express dominant negative (DN) MafB in macrophages under the control of macrophage scavenger receptor (MSR) enhancer-promoter. We performed cell differential analyses in bronchoalveolar lavage cells, morphological analyses with electron microscopy, and flow cytometry-based analyses of surface markers and a phagocytic capacity assay in macrophages. AM number in the TG mice was significantly decreased compared with wild-type (WT) mice. Morphologically, the high electron density area in the nucleus increased, the shape of pseudopods on the AMs was altered, and actin filament was less localized in the pseudopods of AMs of TG mice, compared with WT mice. The expression of surface markers, F4/80 and CD11b, on peritoneal macrophages in TG mice was reduced compared with WT mice, while those on AMs remained unchanged. Phagocytic capacity was decreased in AMs from TG mice, compared with WT mice. In conclusion, MafB regulates the phenotype of macrophages with respect to the number of alveolar macrophages, the nuclear compartment, cellular shape, surface marker expression, and phagocytic function. MSR-DN MafB TG mice may present a useful model to clarify the precise role of MafB in macrophages.
Collapse
MESH Headings
- Animals
- Antigens, Surface/metabolism
- Apoptosis
- Bronchoalveolar Lavage Fluid/cytology
- Gene Expression Regulation
- Genes, Dominant
- Humans
- Immunophenotyping
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/ultrastructure
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- MafB Transcription Factor/genetics
- MafB Transcription Factor/metabolism
- Mice
- Mice, Transgenic
- Phagocytosis/immunology
- Promoter Regions, Genetic
- Receptors, Fc/metabolism
- Receptors, Scavenger/genetics
- Spleen/immunology
- Spleen/metabolism
Collapse
Affiliation(s)
- Michiko Sato-Nishiwaki
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Yasuko Aida
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Shuichi Abe
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Yoko Shibata
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
- * E-mail:
| | - Tomomi Kimura
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Keiko Yamauchi
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Hiroyuki Kishi
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Akira Igarashi
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Sumito Inoue
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Masamichi Sato
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Osamu Nakajima
- Research Laboratory for Molecular Genetics, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| | - Isao Kubota
- Department of Cardiology, Pulmonology and Nephrology, School of Medicine, Yamagata University, Yamagata City, Yamagata, Japan
| |
Collapse
|
24
|
Neyen C, Plüddemann A, Mukhopadhyay S, Maniati E, Bossard M, Gordon S, Hagemann T. Macrophage scavenger receptor a promotes tumor progression in murine models of ovarian and pancreatic cancer. THE JOURNAL OF IMMUNOLOGY 2013; 190:3798-805. [PMID: 23447685 DOI: 10.4049/jimmunol.1203194] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alternatively activated macrophages express the pattern recognition receptor scavenger receptor A (SR-A). We demonstrated previously that coculture of macrophages with tumor cells upregulates macrophage SR-A expression. We show in this study that macrophage SR-A deficiency inhibits tumor cell migration in a coculture assay. We further demonstrate that coculture of tumor-associated macrophages and tumor cells induces secretion of factors that are recognized by SR-A on tumor-associated macrophages. We tentatively identified several potential ligands for the SR-A receptor in tumor cell-macrophage cocultures by mass spectrometry. Competing with the coculture-induced ligand in our invasion assay recapitulates SR-A deficiency and leads to similar inhibition of tumor cell invasion. In line with our in vitro findings, tumor progression and metastasis are inhibited in SR-A(-/-) mice in two in vivo models of ovarian and pancreatic cancer. Finally, treatment of tumor-bearing mice with 4F, a small peptide SR-A ligand able to compete with physiological SR-A ligands in vitro, recapitulates the inhibition of tumor progression and metastasis observed in SR-A(-/-) mice. Our observations suggest that SR-A may be a potential drug target in the prevention of metastatic cancer progression.
Collapse
Affiliation(s)
- Claudine Neyen
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
25
|
Huang S, Miao R, Zhou Z, Wang T, Liu J, Liu G, Chen YE, Xin HB, Zhang J, Fu M. MCPIP1 negatively regulates toll-like receptor 4 signaling and protects mice from LPS-induced septic shock. Cell Signal 2013; 25:1228-34. [PMID: 23422584 DOI: 10.1016/j.cellsig.2013.02.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 02/03/2013] [Accepted: 02/08/2013] [Indexed: 12/20/2022]
Abstract
Septic shock is one of leading causes of morbidity and mortality in hospital patients. However, genetic factors predisposing to septic shock are not fully understood. Our previous work showed that MCP-induced protein 1 (MCPIP1) was induced by lipopolysaccharides (LPSs), which then negatively regulates LPS-induced inflammatory signaling in vitro. Here we report that although MCPIP1 was induced by various toll-like receptor (TLR) ligands in macrophages, MCPIP1-deficient mice are extremely susceptible to TLR4 ligand (LPS)-induced septic shock and death, but not to the TLR2, 3, 5 and 9 ligands-induced septic shock. Consistently, LPS induced tumor necrosis factor α (TNFα) production in MCPIP1-deficient mice was 20-fold greater than that in their wild-type littermates. Further analysis revealed that MCPIP1-deficient mice developed severe acute lung injury after LPS injection and JNK signaling was highly activated in MCPIP1-deficient lungs after LPS stimulation. Finally, macrophage-specific MCPIP1 transgenic mice were partially protected from LPS-induced septic shock, suggesting that inflammatory cytokines from sources other than macrophages may significantly contribute to the pathogenesis of LPS-induced septic shock. Taken together, these results suggest that MCPIP1 selectively suppresses TLR4 signaling pathway and protects mice from LPS-induced septic shock.
Collapse
Affiliation(s)
- Shengping Huang
- Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Levin MC, Lidberg U, Jirholt P, Adiels M, Wramstedt A, Gustafsson K, Greaves DR, Li S, Fazio S, Linton MF, Olofsson SO, Borén J, Gjertsson I. Evaluation of macrophage-specific promoters using lentiviral delivery in mice. Gene Ther 2012; 19:1041-7. [PMID: 22130447 PMCID: PMC3697098 DOI: 10.1038/gt.2011.195] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 10/07/2011] [Accepted: 10/19/2011] [Indexed: 12/22/2022]
Abstract
In gene therapy, tissue-specific promoters are useful tools to direct transgene expression and improve efficiency and safety. Macrophage-specific promoters (MSPs) have previously been published using different delivery systems. In this study, we evaluated five different MSPs fused with green fluorescent protein (GFP) to delineate the one with highest specificity using lentiviral delivery. We compared three variants of the CD68 promoter (full length, the 343-bp proximal part and the 150-bp proximal part) and two variants (in forward and reverse orientation) of a previously characterized synthetic promoter derived from elements of transcription factor genes. We transduced a number of cell lines and primary cells in vitro. In addition, hematopoietic stem cells were transduced with MSPs and transferred into lethally irradiated recipient mice. Fluorescence activated cell sorting analysis was performed to determine the GFP expression in different cell populations both in vitro and in vivo. We showed that MSPs can efficiently be used for lentiviral gene delivery and that the 150-bp proximal part of the CD68 promoter provides primarily macrophage-specific expression of GFP. We propose that this is the best currently available MSP to use for directing transgene expression to macrophage populations in vivo using lentiviral vectors.
Collapse
Affiliation(s)
- M C Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, Göteborg, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Reifenberg K, Cheng F, Orning C, Crain J, Küpper I, Wiese E, Protschka M, Blessing M, Lackner KJ, Torzewski M. Overexpression of TGF-ß1 in macrophages reduces and stabilizes atherosclerotic plaques in ApoE-deficient mice. PLoS One 2012; 7:e40990. [PMID: 22829904 PMCID: PMC3400574 DOI: 10.1371/journal.pone.0040990] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 06/19/2012] [Indexed: 12/28/2022] Open
Abstract
Although macrophages represent the hallmark of both human and murine atherosclerotic lesions and have been shown to express TGF-ß1 (transforming growth factor β1) and its receptors, it has so far not been experimentally addressed whether the pleiotropic cytokine TGF-ß1 may influence atherogenesis by a macrophage specific mechanism. We developed transgenic mice with macrophage specific TGF-ß1 overexpression, crossed the transgenics to the atherosclerotic ApoE (apolipoprotein E) knock-out strain and quantitatively analyzed both atherosclerotic lesion development and composition of the resulting double mutants. Compared with control ApoE−/− mice, animals with macrophage specific TGF-ß1 overexpression developed significantly less atherosclerosis after 24 weeks on the WTD (Western type diet) as indicated by aortic plaque area en face (p<0.05). Reduced atherosclerotic lesion development was associated with significantly less macrophages (p<0.05 after both 8 and 24 weeks on the WTD), significantly more smooth muscle cells (SMCs; p<0.01 after 24 weeks on the WTD), significantly more collagen (p<0.01 and p<0.05 after 16 and 24 weeks on the WTD, respectively) without significant differences of inner aortic arch intima thickness or the number of total macrophages in the mice pointing to a plaque stabilizing effect of macrophage-specific TGF-ß1 overexpression. Our data shows that macrophage specific TGF-ß1 overexpression reduces and stabilizes atherosclerotic plaques in ApoE-deficient mice.
Collapse
Affiliation(s)
- Kurt Reifenberg
- Central Laboratory Animal Facility, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Fei Cheng
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Carolin Orning
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Jeanine Crain
- Central Laboratory Animal Facility, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Ines Küpper
- Central Laboratory Animal Facility, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Elena Wiese
- Central Laboratory Animal Facility, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Martina Protschka
- Center for Biotechnology and Biomedicine, Veterinary Faculty, University of Leipzig, Leipzig, Germany
| | - Manfred Blessing
- Center for Biotechnology and Biomedicine, Veterinary Faculty, University of Leipzig, Leipzig, Germany
| | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Michael Torzewski
- Department of Laboratory Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
28
|
Zhu Y, van Essen D, Saccani S. Cell-Type-Specific Control of Enhancer Activity by H3K9 Trimethylation. Mol Cell 2012; 46:408-23. [DOI: 10.1016/j.molcel.2012.05.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 04/15/2012] [Accepted: 05/08/2012] [Indexed: 11/16/2022]
|
29
|
Kzhyshkowska J, Neyen C, Gordon S. Role of macrophage scavenger receptors in atherosclerosis. Immunobiology 2012; 217:492-502. [PMID: 22437077 DOI: 10.1016/j.imbio.2012.02.015] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Accepted: 02/19/2012] [Indexed: 12/22/2022]
Abstract
Accumulating evidence indicates that atherosclerosis is a chronic inflammatory disease. The key innate immune cells that are involved in the pathogenesis of atherosclerosis are circulating monocytes and plaque macrophages. Complex interplay between immune and metabolic processes results in pathological activity of these cells. The best understood pathological process mediated by macrophages is their inability to process modified lipoproteins properly resulting in the formation of foamy cells, which are a dangerous component of atherosclerotic plaques. Key molecules involved in the recognition and processing of modified lipoproteins are scavenger receptors (SR). This is a large family of surface expressed structurally heterogeneous receptors with a broad spectrum of endogenous and exogenous ligands. The common functional feature of SR is internalisation of extracellular components and targeting them for lysosomal degradation. However, these relatively simple functions can have complex consequences, since they are linked to diverse specific signalling pathways and to other membrane transport pathways. Moreover, scavenger receptors can co-operate with other types of receptors increasing the variability of the macrophage response to multiple extracellular ligands. At least some SRs respond to modified lipoproteins by amplification of inflammation and accumulation of macrophages in the plaque, while some SRs may support tolerogenic reactions. Outcome of different SR activities will be the decision of monocytes and macrophage to guard homeostatic balance, support atherosclerosis progression and plaque instability by inflammatory reactions, or support rapid fibrotic processes in the plaque that stabilise it. Despite the accumulating knowledge about the molecular mechanisms of scavenger receptor action, their role in the progression of atherosclerosis remains controversial. The activities of scavenger receptors that can contribute to each of these processes are a subject of current review.
Collapse
Affiliation(s)
- Julia Kzhyshkowska
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Germany.
| | | | | |
Collapse
|
30
|
Gu X, Zhang W, Liu J, Shaw JP, Shen Y, Xu Y, Lu H, Wu Z. Preparation and characterization of a lovastatin-loaded protein-free nanostructured lipid carrier resembling high-density lipoprotein and evaluation of its targeting to foam cells. AAPS PharmSciTech 2011; 12:1200-8. [PMID: 21927961 PMCID: PMC3225525 DOI: 10.1208/s12249-011-9668-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 07/28/2011] [Indexed: 11/30/2022] Open
Abstract
This study was designed to investigate whether a non-protein nanostructured lipid carrier (NLC) resembling high-density lipoprotein (HDL) could deliver a hydrophobic anti-atherogenic drug, lovastatin, to foam cells. Lovastatin-loaded NLC (LT-NLC) was prepared by a nanoprecipitation/solvent diffusion method. The LT-NLC-apoprotein (LT-NLC-apo) was prepared by incubating LT-NLC with native HDL. The physicochemical parameters of LT-NLC were characterized in terms of particle size, zeta potential, morphology, entrapment efficiency, and crystallization behavior. Targeting behavior and mechanism were demonstrated by the incubation of LT-NLC-apo with a RAW 264.7 macrophage-derived foam cell model in the presence or absence of very-low-density lipoprotein (VLDL) and lipase. The results showed that LT-NLC was solid spherical or oval in shape with an average diameter of 13.8 ± 2.2 nm, zeta potential of −29.3 ± 0.2 mV and entrapment efficiency of 96.2 ± 1.3%. Phagocytosis studies showed that uptake of LT-NLC-apo by macrophages was significantly lower than LT-NLC (p < 0.01), suggesting that LT-NLC-apo could possibly escape recognition from macrophages in vivo. The uptake was increased twofold when LT-NLC-apo was incubated with transfected foam cells containing VLDL and lipase. These results indicated that non-protein NLC resembling HDL could be a useful tool to deliver lipophilic anti-atherogenic drugs to foam cells, and that uptake could be enhanced by the VLDL receptor pathway.
Collapse
|
31
|
Novak ML, Bryer SC, Cheng M, Nguyen MH, Conley KL, Cunningham AK, Xue B, Sisson TH, You JS, Hornberger TA, Koh TJ. Macrophage-specific expression of urokinase-type plasminogen activator promotes skeletal muscle regeneration. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1448-57. [PMID: 21709151 PMCID: PMC3140545 DOI: 10.4049/jimmunol.1004091] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Macrophages (Mp) and the plasminogen system play important roles in tissue repair following injury. We hypothesized that Mp-specific expression of urokinase-type plasminogen activator (uPA) is sufficient for Mp to migrate into damaged muscle and for efficient muscle regeneration. We generated transgenic mice expressing uPA only in Mp, and we assessed the ability of these mice to repair muscle injury. Mp-only uPA expression was sufficient to induce wild-type levels of Mp accumulation, angiogenesis, and new muscle fiber formation. In mice with wild-type uPA expression, Mp-specific overexpression further increased Mp accumulation and enhanced muscle fiber regeneration. Furthermore, Mp expression of uPA regulated the level of active hepatocyte growth factor, which is required for muscle fiber regeneration, in damaged muscle. In vitro studies demonstrated that uPA promotes Mp migration through proteolytic and nonproteolytic mechanisms, including proteolytic activation of hepatocyte growth factor. In summary, Mp-derived uPA promotes muscle regeneration by inducing Mp migration, angiogenesis, and myogenesis.
Collapse
Affiliation(s)
- Margaret L. Novak
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | - Scott C. Bryer
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | - Ming Cheng
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | - Mai-Huong Nguyen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | - Kevin L. Conley
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | | | - Bing Xue
- Department of Medicine, University of Michigan, Ann Arbor
| | | | - Jae-Sung You
- Department of Comparative Biosciences, University of Wisconsin, Madison
| | | | - Timothy J. Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| |
Collapse
|
32
|
Hume DA. Applications of myeloid-specific promoters in transgenic mice support in vivo imaging and functional genomics but do not support the concept of distinct macrophage and dendritic cell lineages or roles in immunity. J Leukoc Biol 2011; 89:525-38. [PMID: 21169519 DOI: 10.1189/jlb.0810472] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Myeloid lineage cells contribute to innate and acquired immunity, homeostasis, wound repair, and inflammation. There is considerable interest in manipulation of their function in transgenic mice using myeloid-specific promoters. This review considers the applications and specificity of some of the most widely studied transgenes, driven by promoter elements of the lysM, csf1r, CD11c, CD68, macrophage SRA, and CD11b genes, as well as several others. Transgenes have been used in mice to generate myeloid lineage-specific cell ablation, expression of genes of interest, including fluorescent reporters, or deletion via recombination. In general, the specificity of such transgenes has been overinterpreted, and none of them provide well-documented, reliable, differential expression in any specific myeloid cell subset, macrophages, granulocytes, or myeloid DCs. Nevertheless, they have proved valuable in cell isolation, functional genomics, and live imaging of myeloid cell behavior in many different pathologies.
Collapse
Affiliation(s)
- David A Hume
- The Roslin Institute, University of Edinburgh, Roslin, Midlothian EH25 9PS, UK.
| |
Collapse
|
33
|
Fishbein I, Chorny M, Levy RJ. Site-specific gene therapy for cardiovascular disease. CURRENT OPINION IN DRUG DISCOVERY & DEVELOPMENT 2010; 13:203-13. [PMID: 20205054 PMCID: PMC2845151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Gene therapy holds considerable promise for the treatment of cardiovascular disease and may provide novel therapeutic solutions for both genetic disorders and acquired pathophysiologies such as arteriosclerosis, heart failure and arrhythmias. Recombinant DNA technology and the sequencing of the human genome have made a plethora of candidate therapeutic genes available for cardiovascular diseases. However, progress in the field of gene therapy for cardiovascular disease has been modest; one of the key reasons for this limited progress is the lack of gene delivery systems for localizing gene therapy to specific sites to optimize transgene expression and efficacy. This review summarizes progress made toward the site-specific delivery of cardiovascular gene therapy and highlights selected promising novel approaches.
Collapse
Affiliation(s)
- Ilia Fishbein
- The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
34
|
Mehra D, Sternberg DI, Jia Y, Canfield S, Lemaitre V, Nkyimbeng T, Wilder J, Sonett J, D'Armiento J. Altered lymphocyte trafficking and diminished airway reactivity in transgenic mice expressing human MMP-9 in a mouse model of asthma. Am J Physiol Lung Cell Mol Physiol 2009; 298:L189-96. [PMID: 19940022 DOI: 10.1152/ajplung.00042.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) is hypothesized to facilitate leukocyte extravasation and extracellular remodeling in asthmatic airways. Careful descriptive studies have shown that MMP-9 levels are higher in the sputum of asthmatics; however, the consequence of increased MMP-9 activity has not been determined in this disease. We induced asthma in transgenic mice that express human MMP-9 in the murine lung tissue macrophage to determine the direct effect of human MMP-9 expression on airway inflammation. Transgenic (TG) and wild-type (WT) mice were immunized and challenged with ovalbumin. Forty-eight hours after the ovalbumin challenge, airway hyperresponsiveness (AHR) was measured, and inflammatory cell infiltration was evaluated in bronchoalveolar lavage fluid (BALF) and lung tissue. Baseline levels of inflammation were similar in the TG and WT groups of mice, and pulmonary eosinophilia was established in both groups by sensitization and challenge with ovalbumin. There was a significant reduction in AHR in sensitized and challenged trangenics compared with WT controls. Although total BALF cell counts were similar in both groups, the lymphocyte number in the lavage of the TG group was significantly diminished compared with the WT group (0.25 +/- 0.08 vs. 0.89 +/- 0.53; P = 0.0032). In addition, the draining lymphocytes were found to be larger in the TG animals compared with the WT mice. Equal numbers of macrophages, eosinophils, and neutrophils were seen in both groups. IL-13 levels were found to be lower in the sensitized TG compared with the WT mice. These results demonstrate an inverse relationship between human MMP-9 and AHR and suggest that MMP-9 expression alters leukocyte extravasation by reducing lymphocyte accumulation in the walls of asthmatic airways.
Collapse
Affiliation(s)
- Divya Mehra
- Dept. of Medicine, Division of Pulmonary and Molecular Medicine, Columbia Presbyterian Medical Center, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Krishnan R, Kremen M, Hu JH, Emery I, Farris SD, Slezicki KI, Chu T, Du L, Dichek HL, Dichek DA. Level of macrophage uPA expression is an important determinant of atherosclerotic lesion growth in Apoe-/- mice. Arterioscler Thromb Vasc Biol 2009; 29:1737-44. [PMID: 19729604 PMCID: PMC2766014 DOI: 10.1161/atvbaha.109.195529] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Enhanced plasminogen activation, mediated by overexpression of urokinase-type plasminogen activator (uPA), accelerates atherosclerosis in apolipoprotein E-null mice. However, the mechanisms through which uPA acts remain unclear. In addition, although elevated uPA expression can accelerate murine atherosclerosis, there is not yet any evidence that decreased uPA expression would retard atherosclerosis. METHODS AND RESULTS We used a bone marrow transplant (BMT) approach and apolipoprotein E-deficient (Apoe(-/-)) mice to investigate cellular mechanisms of uPA-accelerated atherosclerosis, aortic dilation, and sudden death. We also used BMT to determine whether postnatal loss of uPA expression in macrophages retards atherosclerosis. BMT from uPA-overexpressing mice yielded recipients with macrophage-specific uPA overexpression; whereas BMT from uPA knockout mice yielded recipients with macrophage-specific loss of uPA expression. Recipients of uPA-overexpressing BM acquired all the vascular phenotypes (accelerated atherosclerosis, aortic medial destruction and dilation, severe coronary stenoses) as well as the sudden death phenotype of uPA-overexpressing mice. Moreover, fat-fed 37-week-old recipients of uPA-null BM had significantly less atherosclerosis than recipients of uPA wild-type marrow (40% less aortic surface lesion area; P=0.03). CONCLUSIONS The level of uPA expression by macrophages-over a broad range-is an important determinant of atherosclerotic lesion growth in Apoe(-/-) mice.
Collapse
Affiliation(s)
| | - Michal Kremen
- Department of Medicine, University of Washington, Seattle, WA
| | - Jie Hong Hu
- Department of Medicine, University of Washington, Seattle, WA
| | - Isaac Emery
- Department of Medicine, University of Washington, Seattle, WA
| | | | | | - Talyn Chu
- Department of Medicine, University of Washington, Seattle, WA
| | - Liang Du
- Department of Medicine, University of Washington, Seattle, WA
| | - Helén L. Dichek
- Department of Pediatrics, University of Washington, Seattle, WA
| | - David A. Dichek
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
36
|
Inducible transgenes under the control of the hCD68 promoter identifies mouse macrophages with a distribution that differs from the F4/80 - and CSF-1R-expressing populations. Exp Hematol 2009; 37:1387-92. [PMID: 19772887 DOI: 10.1016/j.exphem.2009.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 08/26/2009] [Accepted: 09/16/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Macrophages are critical components of diverse microenvironments (ME) in adulthood, as well as during embryogenesis. Their role in development precludes the use of gene-targeting and knockout approaches for studying their function. Hence, we proposed to create a macrophage-specific inducible transgenic mouse where genes can be turned on or off at will. MATERIALS AND METHODS A transgenic mouse in which the reverse tetracycline activator (rtTA-M2) is expressed under the hCD68 promoter for macrophage-specific gene induction was developed and crossed with a second transgenic reporter mouse strain in which the gene for green fluorescent protein (GFP) is under the control of tetracycline responsive element promoter. After doxycycline induction of the double transgenic animals (designated CD68-rtTA-tet-GFP), inducible expression of GFP was characterized by multicolor flow cytometric analysis of blood, marrow, and spleen cells and by demonstration of GFP expression in fresh-frozen sections in diverse tissues. RESULTS In bone marrow, inducible GFP expression was not confined to, or inclusive of, all cells expressing the classical macrophage markers, such as F4/80. However, GFP-expressing cells in thioglycollate-elicited peritoneal macrophages were also positive for F4/80 and monocyte-macrophage-specific 2 antigen. Interestingly, flow analysis also indicated little overlap between the F4/80 and CSF-1R-positive populations. Fresh-frozen samples of tissues known to contain macrophages revealed GFP-expressing cells with variable morphologies. CONCLUSION Our results show that the hCD68 promoter directs gene expression in a macrophage population distinct from that defined by classical monocyte-macrophage markers or promoters. Whether this population is functionally distinct remains to be established.
Collapse
|
37
|
Lemaître V, Kim HE, Forney-Prescott M, Okada Y, D’Armiento J. Transgenic expression of matrix metalloproteinase-9 modulates collagen deposition in a mouse model of atherosclerosis. Atherosclerosis 2009; 205:107-12. [DOI: 10.1016/j.atherosclerosis.2008.11.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Revised: 11/18/2008] [Accepted: 11/21/2008] [Indexed: 10/21/2022]
|
38
|
Suganami T, Yuan X, Shimoda Y, Uchio-Yamada K, Nakagawa N, Shirakawa I, Usami T, Tsukahara T, Nakayama K, Miyamoto Y, Yasuda K, Matsuda J, Kamei Y, Kitajima S, Ogawa Y. Activating transcription factor 3 constitutes a negative feedback mechanism that attenuates saturated Fatty acid/toll-like receptor 4 signaling and macrophage activation in obese adipose tissue. Circ Res 2009; 105:25-32. [PMID: 19478204 DOI: 10.1161/circresaha.109.196261] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Obese adipose tissue is markedly infiltrated by macrophages, suggesting that they may participate in the inflammatory pathways that are activated in obese adipose tissue. Evidence has suggested that saturated fatty acids released via adipocyte lipolysis serve as a naturally occurring ligand that stimulates Toll-like receptor (TLR)4 signaling, thereby inducing the inflammatory responses in macrophages in obese adipose tissue. Through a combination of cDNA microarray analyses of saturated fatty acid-stimulated macrophages in vitro and obese adipose tissue in vivo, here we identified activating transcription factor (ATF)3, a member of the ATF/cAMP response element-binding protein family of basic leucine zipper-type transcription factors, as a target gene of saturated fatty acids/TLR4 signaling in macrophages in obese adipose tissue. Importantly, ATF3, when induced by saturated fatty acids, can transcriptionally repress tumor necrosis factor-alpha production in macrophages in vitro. Chromatin immunoprecipitation assay revealed that ATF3 is recruited to the region containing the activator protein-1 site of the endogenous tumor necrosis factor-alpha promoter. Furthermore, transgenic overexpression of ATF3 specifically in macrophages results in the marked attenuation of proinflammatory M1 macrophage activation in the adipose tissue from genetically obese KKA(y) mice fed high-fat diet. This study provides evidence that ATF3, which is induced in obese adipose tissue, acts as a transcriptional repressor of saturated fatty acids/TLR4 signaling, thereby revealing the negative feedback mechanism that attenuates obesity-induced macrophage activation. Our data also suggest that activation of ATF3 in macrophages offers a novel therapeutic strategy to prevent or treat obesity-induced adipose tissue inflammation.
Collapse
Affiliation(s)
- Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Merched AJ, Ko K, Gotlinger KH, Serhan CN, Chan L. Atherosclerosis: evidence for impairment of resolution of vascular inflammation governed by specific lipid mediators. FASEB J 2008; 22:3595-606. [PMID: 18559988 DOI: 10.1096/fj.08-112201] [Citation(s) in RCA: 332] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is now recognized as an inflammatory disease involving the vascular wall. Recent results indicate that acute inflammation does not simply passively resolve as previously assumed but is actively terminated by a homeostatic process that is governed by specific lipid-derived mediators initiated by lipoxygenases. Experiments with animals and humans support a proinflammatory role for the 5-lipoxygenase system. In contrast, results from animal experiments show a range of responses with the 12/15-lipoxygenase pathways in atherosclerosis. To date, the only two clinical epidemiology human studies both support an antiatherogenic role for 12/15-lipoxygenase downstream actions. We tested the hypothesis that atherosclerosis results from a failure in the resolution of local inflammation by analyzing apolipoprotein E-deficient mice with 1) global leukocyte 12/15-lipoxygenase deficiency, 2) normal enzyme expression, or 3) macrophage-specific 12/15-lipoxygenase overexpression. Results from these indicate that 12/15-lipoxygenase expression protects mice against atherosclerosis via its role in the local biosynthesis of lipid mediators, including lipoxin A(4), resolvin D1, and protectin D1. These mediators exert potent agonist actions on macrophages and vascular endothelial cells that can control the magnitude of the local inflammatory response. Taken together, these findings suggest that a failure of local endogenous resolution mechanisms may underlie the unremitting inflammation that fuels atherosclerosis.
Collapse
Affiliation(s)
- Aksam J Merched
- Department of Molecular and Cellular Biology, Baylor College of Medicine and St. Luke's Episcopal Hospital, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
40
|
Foronjy R, Nkyimbeng T, Wallace A, Thankachen J, Okada Y, Lemaitre V, D'Armiento J. Transgenic expression of matrix metalloproteinase-9 causes adult-onset emphysema in mice associated with the loss of alveolar elastin. Am J Physiol Lung Cell Mol Physiol 2008; 294:L1149-57. [PMID: 18408070 DOI: 10.1152/ajplung.00481.2007] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Matrix metalloproteinase (MMP)-9 has been consistently identified in the lungs of patients with chronic obstructive pulmonary disease (COPD). However, its role in the development of the disease remains undefined. Mice that specifically express human MMP-9 in their macrophages were generated, and morphometric, biochemical, and histological analyses were conducted on the transgenic and littermate control mice over 1 yr to determine the effect of macrophage MMP-9 expression on emphysema formation and lung matrix content. Lung morphometry was normal in transgenic mice at 2 mo of age (mean linear intercept = 50+/-3 littermate mice vs. 51+/-2 transgenic mice). However, after 12 mo of age, the MMP-9 transgenic mice developed significant air space enlargement (mean linear intercept = 53+/-3 littermate mice vs. 61+/-2 MMP-9 transgenic mice; P<0.04). Lung hydroxyproline content was not significantly different between wild-type and transgenic mice, but MMP-9 did significantly decrease alveolar wall elastin at 1 yr of age (4.9+/-0.3% area of alveolar wall in the littermate mice vs. 3.3+/-0.3% area of alveolar wall in the MMP-9 mice; P<0.004). Thus these results establish a central role for MMP-9 in the pathogenesis of this disease by demonstrating that expression of this protease in macrophages can alter the extracellular matrix and induce progressive air space enlargement in mice.
Collapse
|
41
|
Yamada S, Wang KY, Tanimoto A, Fan J, Shimajiri S, Kitajima S, Morimoto M, Tsutsui M, Watanabe T, Yasumoto K, Sasaguri Y. Matrix metalloproteinase 12 accelerates the initiation of atherosclerosis and stimulates the progression of fatty streaks to fibrous plaques in transgenic rabbits. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1419-29. [PMID: 18403602 DOI: 10.2353/ajpath.2008.070604] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Whether fatty streaks are directly followed by fibrous plaque formation in atherosclerosis remains controversial. Disruption of the basement membrane and elastic layers is thought to be essential for this process. Matrix metalloproteinase 12 (MMP-12) can degrade a broad spectrum of substrates, but the role of MMP-12 in the early stage of atherosclerosis is unclear. To investigate MMP-12 function in the initiation and progression of atherosclerosis, we investigated macrophage migration and elastolysis in relation to fatty streaks in human MMP-12 transgenic (hMMP-12 Tg) rabbits. Fatty streaks in hMMP-12 Tg rabbits fed a 1% cholesterol diet for 6 weeks (cholesterol-induced model of atherosclerosis) were more pronounced and were associated with more significant degradation of the internal elastic layer compared with wild-type (WT) animals. Numbers of infiltrating macrophages and smooth muscle cells in the lesions were increased in hMMP-12 Tg compared with WT animals. In both cuff- and ligation-induced models of atherosclerosis, smooth muscle cell-predominant atherosclerotic lesions were elevated with significant elastolysis of the internal elastic lamina in Tg compared with WT animals; "microelastolytic sites" were recognized before formation of the neointima in the cuff model only. These results indicate that MMP-12 may be critical to the initiation and progression of atherosclerosis via degradation of the elastic layers and/or basement membrane. Therefore, a specific MMP-12 inhibitor might prove useful for the treatment of progressive atherosclerosis.
Collapse
Affiliation(s)
- Sohsuke Yamada
- Department of Pathology and Cell Biology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pan H, Mostoslavsky G, Eruslanov E, Kotton DN, Kramnik I. Dual-promoter lentiviral system allows inducible expression of noxious proteins in macrophages. J Immunol Methods 2008; 329:31-44. [PMID: 17967462 PMCID: PMC2244810 DOI: 10.1016/j.jim.2007.09.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 09/14/2007] [Accepted: 09/19/2007] [Indexed: 11/23/2022]
Abstract
In-depth studies of innate immunity require efficient genetic manipulation of macrophages, which is especially difficult in primary macrophages. We have developed a lentiviral system for inducible gene expression both in macrophage cell lines and in primary macrophages. A transgenic mouse strain C3H.TgN(SRA-rtTA) that expresses reverse tetracycline transactivator (rtTA) under the control of macrophage-specific promoter, a modified human Scavenger Receptor A (SRA) promoter was generated. For gene delivery, we constructed a dual-promoter lentiviral vector, in which expression of a "gene-of-interest" is driven by a doxycycline-inducible promoter and the expression of a selectable surface marker is driven by an independent constitutive promoter UBC. This vector is used for transduction of bone marrow-derived macrophage precursors. The transduced cells can be enriched to 95-99% purity using marker-specific monoclonal antibodies, expanded and differentiated into mature macrophages or myeloid dendritic cells. We also successfully used this approach for inducible protein expression in hard to transfect macrophage cell lines. Because many proteins, which are expressed by activated or infected macrophages, possess cytotoxic, anti-proliferative or pro-apoptotic activities, generation of stable macrophage cell lines that constitutively express those proteins is impossible. Our method will be especially useful to study immunity-related macrophage proteins in their physiological context during macrophage activation or infection.
Collapse
Affiliation(s)
- Hui Pan
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 667 Huntington Avenue, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
43
|
Choi MS, Lee WH, Kwon EY, Kang MA, Lee MK, Park YB, Jeon SM. Effects of soy pinitol on the pro-inflammatory cytokines and scavenger receptors in oxidized low-density lipoprotein-treated THP-1 macrophages. J Med Food 2007; 10:594-601. [PMID: 18158828 DOI: 10.1089/jmf.2006.220] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Pinitol, a methylated form of D-chiro-inositol, acts as a insulin mediator. We investigated the effects of soy pinitol on the factors involved in foam cell formation using differentiated THP-1 macrophages. Pinitol slightly inhibited the lipid-laden foam cell formation by oxidized low-density lipoprotein (oxLDL) in a dose-dependent manner. Tumor necrosis factor-alpha and monocyte chemoattractant protein-1 releases were significantly reduced by pinitol treatment (0.05-0.5 mM), whereas interleukin-1beta and interleukin-8 secretions were significantly reduced in low-dose pinitol (0.05 or 0.1 mM) and 0.5 mM pinitol-treated cells, respectively, compared to no pinitol-treated cells. Gene expressions of CD36 and CD68 were significantly down-regulated by 0.05-0.5 mM pinitol compared to the oxLDL-treated control cells. Matrix metalloproteinase-9 gene expression was significantly decreased in 0.05-0.5 mM pinitol-treated cells compared to the no pinitol-treated macrophages. We conclude that pinitol has some inhibitory effects on foam cell formation by reducing lipid accumulation, secretion, and expression of some cytokines and macrophage scavenger receptor expression via its insulin-like action.
Collapse
Affiliation(s)
- Myung-Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
44
|
Zhao B, Song J, Chow WN, St. Clair RW, Rudel LL, Ghosh S. Macrophage-specific transgenic expression of cholesteryl ester hydrolase significantly reduces atherosclerosis and lesion necrosis in Ldlr mice. J Clin Invest 2007; 117:2983-92. [PMID: 17885686 PMCID: PMC1978419 DOI: 10.1172/jci30485] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Accepted: 06/26/2007] [Indexed: 01/20/2023] Open
Abstract
Accumulation of cholesteryl esters (CEs) in macrophage foam cells, central to atherosclerotic plaque formation, occurs as a result of imbalance between the cholesterol influx and efflux pathways. While the uptake, or influx, of modified lipoproteins is largely unregulated, extracellular acceptor-mediated free cholesterol (FC) efflux is rate limited by the intracellular hydrolysis of CE. We previously identified and cloned a neutral CE hydrolase (CEH) from human macrophages and demonstrated its role in cellular CE mobilization. In the present study, we examined the hypothesis that macrophage-specific overexpression of CEH in atherosclerosis-susceptible Ldlr(-/-) mice will result in reduction of diet-induced atherosclerosis. Transgenic mice overexpressing this CEH specifically in the macrophages (driven by scavenger receptor promoter/enhancer) were developed and crossed into the Ldlr(-/-) background (Ldlr(-/-)CEHTg mice). Macrophage-specific overexpression of CEH led to a significant reduction in the lesion area and cholesterol content of high-fat, high-cholesterol diet-induced atherosclerotic lesions. The lesions from Ldlr(-/-)CEHTg mice did not have increased FC, were less necrotic, and contained significantly higher numbers of viable macrophage foam cells. Higher CEH-mediated FC efflux resulted in enhanced flux of FC from macrophages to gall bladder bile and feces in vivo. These studies demonstrate that by enhancing cholesterol efflux and reverse cholesterol transport, macrophage-specific overexpression of CEH is antiatherogenic.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Internal Medicine and
Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA.
Department of Pathology, Lipid Sciences Section, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jingmei Song
- Department of Internal Medicine and
Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA.
Department of Pathology, Lipid Sciences Section, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Woon N. Chow
- Department of Internal Medicine and
Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA.
Department of Pathology, Lipid Sciences Section, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Richard W. St. Clair
- Department of Internal Medicine and
Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA.
Department of Pathology, Lipid Sciences Section, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Lawrence L. Rudel
- Department of Internal Medicine and
Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA.
Department of Pathology, Lipid Sciences Section, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Shobha Ghosh
- Department of Internal Medicine and
Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA.
Department of Pathology, Lipid Sciences Section, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
45
|
Macrophage-specific inhibition of NF-κB activation reduces foam-cell formation. Atherosclerosis 2007; 192:283-90. [DOI: 10.1016/j.atherosclerosis.2006.07.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 07/19/2006] [Accepted: 07/19/2006] [Indexed: 10/24/2022]
|
46
|
Kisgati M, Asmis R. Generation of retroviruses for the overexpression of cytosolic and mitochondrial glutathione reductase in macrophages in vivo. Cytotechnology 2007; 54:5-14. [PMID: 19003013 DOI: 10.1007/s10616-007-9046-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Accepted: 01/17/2007] [Indexed: 02/06/2023] Open
Abstract
Retroviral gene transfer and bone marrow transplantation has been used by many investigators to study the role of macrophage proteins in different mouse models of human disease. While this approach is faster and less expensive than generating transgenic mice with macrophage-specific promoters and applicable to a wider array of mouse models, it has been hampered by two major drawbacks: labor-intensive cloning procedures involved in generating retroviral vectors for each gene of interest and low viral titers. Here we describe the construction of a MSCV-based retroviral vector that can serve as an acceptor vector for commercially available Cre-lox-compatible donor vectors. Using this new retroviral vector in combination with a FACS approach to enhance viral titers, we generated high-titer retroviruses carrying either EGFP-tagged cytosolic or EGFP-tagged mitochondria-targeted glutathione reductase. We show that the introduction of these constructs via retroviral gene transfer and bone marrow transplantation into atherosclerosis-prone LDL receptor-null mice results in the long-term increase in macrophage glutathione reductase activity.
Collapse
Affiliation(s)
- Marta Kisgati
- Department of Laboratory Medicine, Kenezy Gyula Hospital, Debrecen, Hungary
| | | |
Collapse
|
47
|
He W, Qiang M, Ma W, Valente AJ, Quinones MP, Wang W, Reddick RL, Xiao Q, Ahuja SS, Clark RA, Freeman GL, Li S. Development of a synthetic promoter for macrophage gene therapy. Hum Gene Ther 2006; 17:949-59. [PMID: 16972763 DOI: 10.1089/hum.2006.17.949] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Macrophages have the potential to deliver therapeutic genes to many target tissues. Macrophage-specific synthetic promoters (SPs) generated by random ligation of myeloid/macrophage cis elements had activity up to 100-fold that of a native macrophage promoter in macrophage cell lines, but were minimally active in nonmyeloid cells. Mouse bone marrow cells (BMCs) transduced ex vivo with lentivectors expressing green fluorescent protein (GFP) driven either by an SP (SP-GFP) or a cytomegalovirus (CMV) promoter (CMV-GFP) were used for syngeneic transplantation of lethally irradiated mice. Blood leukocytes showed stable GFP expression for up to 15 months after transplantation. SP-GFP expression was selective for CD11b+ macrophages, whereas CMV-GFP expression was observed in erythrocytes, as well as in both CD11b+ and CD11b- leukocytes. Furthermore, SP-GFP expression was much stronger than CMV-GFP expression in CD11b+ macrophages. apoE-/- BMCs transduced with the lentiviral vector encoding human apoE were used to transplant apoE-/- mice. Macrophage expression of apoE from 10 to 26 weeks of age significantly reduced atherosclerotic lesions in recipient apoE-/- mice. Thus, the novel SPs, especially when combined with lentivectors, are useful for macrophage-specific delivery of therapeutic genes.
Collapse
Affiliation(s)
- Weijing He
- Department of Medicine, University of Texas Health Science Center at San Antonio, and South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
He W, Qiang M, Ma W, Valente AJ, Quinones MP, Wang W, Reddick RL, Xiao Q, Ahuja SS, Clark RA, Freeman GL, Li S. Development of a Synthetic Promoter for Macrophage Gene Therapy. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
49
|
Liang J, Liu E, Yu Y, Kitajima S, Koike T, Jin Y, Morimoto M, Hatakeyama K, Asada Y, Watanabe T, Sasaguri Y, Watanabe S, Fan J. Macrophage Metalloelastase Accelerates the Progression of Atherosclerosis in Transgenic Rabbits. Circulation 2006; 113:1993-2001. [PMID: 16636188 DOI: 10.1161/circulationaha.105.596031] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Macrophage metalloelastase (matrix metalloproteinase [MMP]-12) is upregulated in atherosclerotic lesions and aneurysm; thus, increased MMP-12 activity may play an important role in the pathogenesis of atherosclerosis. However, the pathological roles of MMP-12 in the initiation and progression of atherosclerosis have not been defined. METHODS AND RESULTS We compared the susceptibility of MMP-12 transgenic (Tg) rabbits to cholesterol-rich diet-induced atherosclerosis with that of non-Tg littermate rabbits. The rabbits were maintained at either relatively lower levels of hypercholesterolemia for shorter periods or higher levels of hypercholesterolemia for longer periods through a diet containing different amounts of cholesterol. We found no significant difference in the aortic atherosclerotic lesion size or quality between Tg and non-Tg rabbits at lower hypercholesterolemia. At higher hypercholesterolemia for longer periods, however, Tg rabbits developed more extensive atherosclerosis in the aortas and coronary arteries than did non-Tg rabbits. Histological examinations revealed that atherosclerotic lesions of Tg rabbits contained prominent macrophage infiltration associated with marked disruption of the elastic lamina in the tunica media with occasional formation of aneurysm-like lesions. Furthermore, increased expression of MMP-12 derived from macrophages was associated with elevated expression of MMP-3, suggesting that MMP-12 may play a pivotal role in the cascade activation of other MMPs, thereby exacerbating extracellular matrix degradation during the progression of atherosclerosis. CONCLUSIONS Overexpression of MMP-12 causes accelerated atherosclerosis in Tg rabbits. These results suggest that macrophage-derived MMP-12 participates in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Jingyan Liang
- Department of Pathology, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bradshaw G, Gutierrez A, Miyake JH, Davis KR, Li AC, Glass CK, Curtiss LK, Davis RA. Facilitated replacement of Kupffer cells expressing a paraoxonase-1 transgene is essential for ameliorating atherosclerosis in mice. Proc Natl Acad Sci U S A 2005; 102:11029-34. [PMID: 16043712 PMCID: PMC1182426 DOI: 10.1073/pnas.0502677102] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Resident macrophages (i.e., Kupffer cells) are derived from hematopoietic stem cells (HSCs) and are primarily responsible for the removal from plasma of oxidized forms of low-density lipoprotein (LDL). The therapeutic potential of Kupffer cell expression of a transgene encoding paraoxonase-1 (PON1), whose plasma activity correlates with the protection from atherosclerosis, was examined in mice rendered atherosclerosis-susceptible through genetic deletion of the LDL receptor. Mice having their bone marrow engrafted with HSCs expressing the PON1 transgene (PON1-Tg) driven by a macrophage-specific promoter were injected i.v. with saline (vehicle only) or with gadolinium chloride (GdCl(3)), an agent that rapidly causes Kupffer cell apoptosis. One month later, GdCl(3)-facilitated Kupffer cell apoptosis increased the hepatic expression of transgenic PON1 mRNA by 9-fold. After 12 weeks of being fed a cholesterol-enriched atherogenic diet, mice injected with GdCl(3) exhibited 50% reductions in both aortic sinus atherosclerotic lesions (P < 0.0097) and surface lesions of the abdominal aorta (P < 0.006). In contrast, mice receiving HSCs expressing the PON1-Tg but not treated with GdCl(3) showed no protection from atherosclerosis. In addition, mice engrafted with HSCs not expressing the PON1-Tg but injected with GdCl(3) also showed no protection from atherosclerosis. These findings, showing that GdCl(3)-enhanced hepatic expression of the PON1-Tg is essential for reducing atherosclerosis, indicate that Kupffer cells play an important role in atherogenesis. GdCl(3)-facilated replacement of Kupffer cells may enhance the efficacy of other HSC-based gene therapies.
Collapse
Affiliation(s)
- Gary Bradshaw
- Department of Biology and Heart Institute, San Diego State University, San Diego, CA 92182, USA
| | | | | | | | | | | | | | | |
Collapse
|