1
|
Robichaud S, Rochon V, Emerton C, Laval T, Ouimet M. Trehalose promotes atherosclerosis regression in female mice. Front Cardiovasc Med 2024; 11:1298014. [PMID: 38433753 PMCID: PMC10906268 DOI: 10.3389/fcvm.2024.1298014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/31/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Atherosclerosis is a chronic inflammatory disease caused by the deposition of lipids within the artery wall. During atherogenesis, efficient autophagy is needed to facilitate efferocytosis and cholesterol efflux, limit inflammation and lipid droplet buildup, and eliminate defective mitochondria and protein aggregates. Central to the regulation of autophagy is the transcription factor EB (TFEB), which coordinates the expression of lysosomal biogenesis and autophagy genes. In recent years, trehalose has been shown to promote TFEB activation and protect against atherogenesis. Here, we sought to investigate the role of autophagy activation during atherosclerosis regression. Methods and results Atherosclerosis was established in C57BL/6N mice by injecting AAV-PCSK9 and 16 weeks of Western diet feeding, followed by switching to a chow diet to induce atherosclerosis regression. During the regression period, mice were either injected with trehalose concomitant with trehalose supplementation in their drinking water or injected with saline for 6 weeks. Female mice receiving trehalose had reduced atherosclerosis burden, as evidenced by reduced plaque lipid content, macrophage numbers and IL-1β content in parallel with increased plaque collagen deposition, which was not observed in their male counterparts. In addition, trehalose-treated female mice had lower levels of circulating leukocytes, including inflammatory monocytes and CD4+ T cells. Lastly, we found that autophagy flux in male mice was basally higher than in female mice during atherosclerosis progression. Conclusions Our data demonstrate a sex-specific effect of trehalose in atherosclerosis regression, whereby trehalose reduced lipid content, inflammation, and increased collagen content in female mice but not in male mice. Furthermore, we discovered inherent differences in the autophagy flux capacities between the sexes: female mice exhibited lower plaque autophagy than males, which rendered the female mice more responsive to atherosclerosis regression. Our work highlights the importance of understanding sex differences in atherosclerosis to personalize the development of future therapies to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Sabrina Robichaud
- Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Valérie Rochon
- Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Christina Emerton
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Thomas Laval
- Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Mireille Ouimet
- Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cardiovascular Metabolism and Cell Biology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| |
Collapse
|
2
|
Huang J, Lin Z, Lin J, Xie S, Xia S, Chen G, Zheng Z, Xu Z, Liu F, Wu H, Li S. Causal role of lipid metabolism in pulmonary alveolar proteinosis: an observational and mendelian randomisation study. Thorax 2024; 79:135-143. [PMID: 38124156 DOI: 10.1136/thorax-2023-220789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Pulmonary alveolar proteinosis (PAP) is a rare interstitial lung disease characterised by the accumulation of lipoprotein material in the alveoli. Although dyslipidaemia is a prominet feature, the causal effect of lipid traits on PAP remains unclear. This study aimed to explore the role of lipid traits in PAP and evaluate the potential of lipid-lowering drug targets in PAP. METHODS Clinical outcomes, lipid profiles and lung function tests were analysed in a clinical cohort of diagnosed PAP patients and propensity score-matched healthy controls. Genome-wide association study data on PAP, lipid metabolism, blood cells and variants of genes encoding potential lipid-lowering drug targets were obtained for Mendelian randomisation (MR) and mediation analyses. FINDINGS Observational results showed that higher levels of total cholesterol (TC), triglycerides and low-density lipoprotein (LDL) were associated with increased risks of PAP. Higher levels of TC and LDL were also associated with worse PAP severity. In MR analysis, elevated LDL was associated with an increased risk of PAP (OR: 4.32, 95% CI: 1.63 to 11.61, p=0.018). Elevated monocytes were associated with a lower risk of PAP (OR 0.34, 95% CI: 0.18 to 0.66, p=0.002) and mediated the risk impact of LDL on PAP. Genetic mimicry of PCSK9 inhibition was associated with a reduced risk of PAP (OR 0.03, p=0.007). INTERPRETATION Our results support the crucial role of lipid and metabolism-related traits in PAP risk, emphasising the monocyte-mediated, causal effect of elevated LDL in PAP genetics. PCSK9 mediates the development of PAP by raising LDL. These finding provide evidence for lipid-related mechanisms and promising lipid-lowering drug target for PAP.
Collapse
Affiliation(s)
- Junfeng Huang
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zikai Lin
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinsheng Lin
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shuojia Xie
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shixin Xia
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gengjia Chen
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziwen Zheng
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhe Xu
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fangcheng Liu
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hongkai Wu
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shiyue Li
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Domingues N, Marques ARA, Calado RDA, Ferreira IS, Ramos C, Ramalho J, Soares MIL, Pereira T, Oliveira L, Vicente JR, Wong LH, Simões ICM, Pinho E Melo TMVD, Peden A, Almeida CG, Futter CE, Puertollano R, Vaz WLC, Vieira OV. Oxidized cholesteryl ester induces exocytosis of dysfunctional lysosomes in lipidotic macrophages. Traffic 2023; 24:284-307. [PMID: 37129279 DOI: 10.1111/tra.12888] [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: 12/13/2022] [Revised: 03/29/2023] [Accepted: 04/20/2023] [Indexed: 05/03/2023]
Abstract
A key event in atherogenesis is the formation of lipid-loaded macrophages, lipidotic cells, which exhibit irreversible accumulation of undigested modified low-density lipoproteins (LDL) in lysosomes. This event culminates in the loss of cell homeostasis, inflammation, and cell death. Nevertheless, the exact chemical etiology of atherogenesis and the molecular and cellular mechanisms responsible for the impairment of lysosome function in plaque macrophages are still unknown. Here, we demonstrate that macrophages exposed to cholesteryl hemiazelate (ChA), one of the most prevalent products of LDL-derived cholesteryl ester oxidation, exhibit enlarged peripheral dysfunctional lysosomes full of undigested ChA and neutral lipids. Both lysosome area and accumulation of neutral lipids are partially irreversible. Interestingly, the dysfunctional peripheral lysosomes are more prone to fuse with the plasma membrane, secreting their undigested luminal content into the extracellular milieu with potential consequences for the pathology. We further demonstrate that this phenotype is mechanistically linked to the nuclear translocation of the MiT/TFE family of transcription factors. The induction of lysosome biogenesis by ChA appears to partially protect macrophages from lipid-induced cytotoxicity. In sum, our data show that ChA is involved in the etiology of lysosome dysfunction and promotes the exocytosis of these organelles. This latter event is a new mechanism that may be important in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Neuza Domingues
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - André R A Marques
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Rita Diogo Almeida Calado
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Inês S Ferreira
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Cristiano Ramos
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - José Ramalho
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Maria I L Soares
- CQC and Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - Telmo Pereira
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Luís Oliveira
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - José R Vicente
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Louise H Wong
- Department of Cell Biology, UCL Institute of Ophthalmology, London, UK
| | - Inês C M Simões
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | | | - Andrew Peden
- Department of Biomedical Science & Center for Membrane Interactions and Dynamics, University of Sheffield, UK
| | - Cláudia Guimas Almeida
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Clare E Futter
- Department of Cell Biology, UCL Institute of Ophthalmology, London, UK
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Winchil L C Vaz
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Otília V Vieira
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
4
|
RRP Regulates Autophagy through the AMPK Pathway to Alleviate the Effect of Cell Senescence on Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:9645789. [PMID: 36756298 PMCID: PMC9902129 DOI: 10.1155/2023/9645789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 02/01/2023]
Abstract
Autophagy is closely associated with atherosclerosis and other cardiovascular diseases (CVD). Compound Danshen prescription is widely used as a clinical antiatherosclerotic drug. In our previous studies, we have shown that the combined active component, ginsenoside Rg1-notoginsenoside R1-protocatechualdehyde (RRP), can effectively alleviate endothelial dysfunction and reduce atherosclerotic plaques. However, the association between cellular senescence, caused by reduced autophagy, and atherosclerosis remains unclear. In this study, we investigated whether RRP can enhance autophagy and alleviate cell senescence through the AMPK pathway. Our results showed that RRP reduced the secretion of inflammatory factors in the serum of atherosclerotic mice, enhanced autophagy, and alleviated aortic aging in mice, thus reducing atherosclerotic plaques. In human aortic endothelial cells (HAECs), RRP effectively enhanced autophagy and inhibited senescence by activating the AMPK pathway. When AMPKα was silenced, the effect of RRP was inhibited, thus reversing its antiaging effect. Overall, our results show that RRP regulates autophagy through the AMPK pathway, thereby inhibiting cell senescence and alleviating the progression of atherosclerosis, suggesting that RRP may be a potential candidate drug for the treatment of atherosclerosis.
Collapse
|
5
|
Rezende L, Couto NFD, Fernandes-Braga W, Epshtein Y, Alvarez-Leite JI, Levitan I, Andrade LDO. OxLDL induces membrane structure rearrangement leading to biomechanics alteration and migration deficiency in macrophage. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183951. [PMID: 35504320 DOI: 10.1016/j.bbamem.2022.183951] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022]
Abstract
Cholesterol sequestration from plasma membrane has been shown to induce lipid packing disruption, causing actin cytoskeleton reorganization and polymerization, increasing cell stiffness and inducing lysosomal exocytosis in non-professional phagocytes. Similarly, oxidized form of low-density lipoprotein (oxLDL) has also been shown to disrupt lipid organization and packing in endothelial cells, leading to biomechanics alterations that interfere with membrane injury and repair. For macrophages, much is known about oxLDL effects in cell activation, cytokine production and foam cell formation. However, little is known about its impact in the organization of macrophage membrane structured domains and cellular mechanics, the focus of the present study. Treatment of bone marrow-derived macrophages (BMDM) with oxLDL not only altered membrane structure, and potentially the distribution of raft domains, but also induced actin rearrangement, diffuse integrin distribution and cell shrinkage, similarly to observed upon treatment of these cells with MβCD. Those alterations led to decreased migration efficiency. For both treatments, higher co-localization of actin cytoskeleton and GM1 was observed, indicating a similar mechanism of action involving raft-like domain dynamics. Lastly, like MβCD treatment, oxLDL also induced lysosomal spreading in BMDM. We propose that OxLDL induced re-organization of membrane/cytoskeleton complex in macrophages can be attributed to the insertion of oxysterols into the membrane, which lead to changes in lipid organization and disruption of membrane structure, similar to the effect of cholesterol depletion by MβCD treatment. These results indicate that oxLDL can induce physical alterations in the complex membrane/cytoskeleton of macrophages, leading to significant biomechanical changes that compromise cell behavior.
Collapse
Affiliation(s)
- Luisa Rezende
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natalia Fernanda Do Couto
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Weslley Fernandes-Braga
- Department of Biochemistry and Immunology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Yulia Epshtein
- Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | | | - Irena Levitan
- Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | | |
Collapse
|
6
|
Lorey MB, Öörni K, Kovanen PT. Modified Lipoproteins Induce Arterial Wall Inflammation During Atherogenesis. Front Cardiovasc Med 2022; 9:841545. [PMID: 35310965 PMCID: PMC8927694 DOI: 10.3389/fcvm.2022.841545] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/26/2022] [Indexed: 12/15/2022] Open
Abstract
Circulating apolipoprotein B-containing lipoproteins, notably the low-density lipoproteins, enter the inner layer of the arterial wall, the intima, where a fraction of them is retained and modified by proteases, lipases, and oxidizing agents and enzymes. The modified lipoproteins and various modification products, such as fatty acids, ceramides, lysophospholipids, and oxidized lipids induce inflammatory reactions in the macrophages and the covering endothelial cells, initiating an increased leukocyte diapedesis. Lipolysis of the lipoproteins also induces the formation of cholesterol crystals with strong proinflammatory properties. Modified and aggregated lipoproteins, cholesterol crystals, and lipoproteins isolated from human atherosclerotic lesions, all can activate macrophages and thereby induce the secretion of proinflammatory cytokines, chemokines, and enzymes. The extent of lipoprotein retention, modification, and aggregation have been shown to depend largely on differences in the composition of the circulating lipoprotein particles. These properties can be modified by pharmacological means, and thereby provide opportunities for clinical interventions regarding the prevention and treatment of atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Martina B. Lorey
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- *Correspondence: Katariina Öörni
| | - Petri T. Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| |
Collapse
|
7
|
Alves LS, Marques ARA, Padrão N, Carvalho FA, Ramalho J, Lopes CS, Soares MIL, Futter CE, Pinho E Melo TMVD, Santos NC, Vieira OV. Cholesteryl hemiazelate causes lysosome dysfunction impacting vascular smooth muscle cell homeostasis. J Cell Sci 2022; 135:272202. [PMID: 34528688 DOI: 10.1242/jcs.254631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/07/2021] [Indexed: 01/07/2023] Open
Abstract
In atherosclerotic lesions, vascular smooth muscle cells (VSMCs) represent half of the foam cell population, which is characterized by an aberrant accumulation of undigested lipids within lysosomes. Loss of lysosome function impacts VSMC homeostasis and disease progression. Understanding the molecular mechanisms underlying lysosome dysfunction in these cells is, therefore, crucial. We identify cholesteryl hemiazelate (ChA), a stable oxidation end-product of cholesteryl-polyunsaturated fatty acid esters, as an inducer of lysosome malfunction in VSMCs. ChA-treated VSMCs acquire a foam-cell-like phenotype, characterized by enlarged lysosomes full of ChA and neutral lipids. The lysosomes are perinuclear and exhibit degradative capacity and cargo exit defects. Lysosome luminal pH is also altered. Even though the transcriptional response machinery and autophagy are not activated by ChA, the addition of recombinant lysosomal acid lipase (LAL) is able to rescue lysosome dysfunction. ChA significantly affects VSMC proliferation and migration, impacting atherosclerosis. In summary, this work shows that ChA is sufficient to induce lysosomal dysfunction in VSMCs, that, in ChA-treated VSMCs, neither lysosome biogenesis nor autophagy are triggered, and, finally, that recombinant LAL can be a therapeutic approach for lysosomal dysfunction.
Collapse
Affiliation(s)
- Liliana S Alves
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| | - André R A Marques
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| | - Nuno Padrão
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa 1649-028, Lisboa, Portugal
| | - José Ramalho
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| | - Catarina S Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa 1649-028, Lisboa, Portugal
| | - Maria I L Soares
- CQC and Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Clare E Futter
- Department of Cell Biology, UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | | | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa 1649-028, Lisboa, Portugal
| | - Otília V Vieira
- Chronic Diseases Research Centre (CEDOC), NOVA Medical School, NOVA University Lisbon, 1169-056 Lisboa, Portugal
| |
Collapse
|
8
|
Robichaud S, Rasheed A, Pietrangelo A, Doyoung Kim A, Boucher DM, Emerton C, Vijithakumar V, Gharibeh L, Fairman G, Mak E, Nguyen MA, Geoffrion M, Wirka R, Rayner KJ, Ouimet M. Autophagy Is Differentially Regulated in Leukocyte and Nonleukocyte Foam Cells During Atherosclerosis. Circ Res 2022; 130:831-847. [PMID: 35137605 DOI: 10.1161/circresaha.121.320047] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Atherosclerosis is characterized by an accumulation of foam cells within the arterial wall, resulting from excess cholesterol uptake and buildup of cytosolic lipid droplets (LDs). Autophagy promotes LD clearance by freeing stored cholesterol for efflux, a process that has been shown to be atheroprotective. While the role of autophagy in LD catabolism has been studied in macrophage-derived foam cells, this has remained unexplored in vascular smooth muscle cell (VSMC)-derived foam cells that constitute a large fraction of foam cells within atherosclerotic lesions. OBJECTIVE We performed a comparative analysis of autophagy flux in lipid-rich aortic intimal populations to determine whether VSMC-derived foam cells metabolize LDs similarly to their macrophage counterparts. METHODS AND RESULTS Atherosclerosis was induced in GFP-LC3 transgenic mice by PCSK9 (proprotein convertase subtilisin/kexin type 9)-adeno-associated viral injection and Western diet feeding. Using flow cytometry of aortic digests, we observed a significant increase in dysfunctional autophagy of VSMC-derived foam cells during atherogenesis relative to macrophage-derived foam cells. Using cell culture models of lipid-loaded VSMC and macrophage, we show that autophagy-mediated cholesterol efflux from VSMC foam cells was poor relative to macrophage foam cells, and largely occurs when HDL (high-density lipoprotein) is used as a cholesterol acceptor, as opposed to apoA-1 (apolipoproteinA-1). This was associated with the predominant expression of ABCG1 in VSMC foam cells. Using metformin, an autophagy activator, cholesterol efflux to HDL was significantly increased in VSMC, but not in macrophage, foam cells. CONCLUSIONS These data demonstrate that VSMC and macrophage foam cells perform cholesterol efflux by distinct mechanisms, and that autophagy flux is highly impaired in VSMC foam cells, but can be induced by pharmacological means. Further investigation is warranted into targeting autophagy specifically in VSMC foam cells, the predominant foam cell subtype of advanced atherosclerotic plaques, to promote reverse cholesterol transport and resolution of the atherosclerotic plaque.
Collapse
Affiliation(s)
- Sabrina Robichaud
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Adil Rasheed
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Antonietta Pietrangelo
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Anne Doyoung Kim
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Dominique M Boucher
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Christina Emerton
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
| | - Viyashini Vijithakumar
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Lara Gharibeh
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Garrett Fairman
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Esther Mak
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
| | - Robert Wirka
- University of North Carolina School of Medicine, Chapel Hill (R.W.)
| | - Katey J Rayner
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| | - Mireille Ouimet
- University of Ottawa Heart Institute, ON (S.R., A.R., A.P., A.D.K., D.M.B., C.E., V.V., L.G., G.F., E.M., M.-A.N., M.G., K.J.R., M.O.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON (S.R., A.R., A.P., A.D.K., D.M.B., V.V., L.G., G.F., M.-A.N., K.J.R., M.O.)
| |
Collapse
|
9
|
Marques ARA, Ramos C, Machado-Oliveira G, Vieira OV. Lysosome (Dys)function in Atherosclerosis-A Big Weight on the Shoulders of a Small Organelle. Front Cell Dev Biol 2021; 9:658995. [PMID: 33855029 PMCID: PMC8039146 DOI: 10.3389/fcell.2021.658995] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a progressive insidious chronic disease that underlies most of the cardiovascular pathologies, including myocardial infarction and ischemic stroke. The malfunctioning of the lysosomal compartment has a central role in the etiology and pathogenesis of atherosclerosis. Lysosomes are the degradative organelles of mammalian cells and process endogenous and exogenous substrates in a very efficient manner. Dysfunction of these organelles and consequent inefficient degradation of modified low-density lipoproteins (LDL) and apoptotic cells in atherosclerotic lesions have, therefore, numerous deleterious consequences for cellular homeostasis and disease progression. Lysosome dysfunction has been mostly studied in the context of the inherited lysosomal storage disorders (LSDs). However, over the last years it has become increasingly evident that the consequences of this phenomenon are more far-reaching, also influencing the progression of multiple acquired human pathologies, such as neurodegenerative diseases, cancer, and cardiovascular diseases (CVDs). During the formation of atherosclerotic plaques, the lysosomal compartment of the various cells constituting the arterial wall is under severe stress, due to the tremendous amounts of lipoproteins being processed by these cells. The uncontrolled uptake of modified lipoproteins by arterial phagocytic cells, namely macrophages and vascular smooth muscle cells (VSMCs), is the initial step that triggers the pathogenic cascade culminating in the formation of atheroma. These cells become pathogenic "foam cells," which are characterized by dysfunctional lipid-laden lysosomes. Here, we summarize the current knowledge regarding the origin and impact of the malfunctioning of the lysosomal compartment in plaque cells. We further analyze how the field of LSD research may contribute with some insights to the study of CVDs, particularly how therapeutic approaches that target the lysosomes in LSDs could be applied to hamper atherosclerosis progression and associated mortality.
Collapse
Affiliation(s)
- André R A Marques
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cristiano Ramos
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Gisela Machado-Oliveira
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Otília V Vieira
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
10
|
Effects of lipoproteins on endothelial cells and macrophages function and its possible implications on fetal adverse outcomes associated to maternal hypercholesterolemia during pregnancy. Placenta 2021; 106:79-87. [PMID: 33706211 DOI: 10.1016/j.placenta.2021.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/03/2021] [Accepted: 02/25/2021] [Indexed: 11/23/2022]
Abstract
Hypercholesterolemia is one of the main risk factors associated with atherosclerosis and cardiovascular disease, the leading cause of death worldwide. During pregnancy, maternal hypercholesterolemia develops, and it can occur in a physiological (MPH) or supraphysiological (MSPH) manner, where MSPH is associated with endothelial dysfunction and early atherosclerotic lesions in the fetoplacental vasculature. In the pathogenesis of atherosclerosis, endothelial activation and endothelial dysfunction, characterized by an imbalance in the bioavailability of nitric oxide, contribute to the early stages of this disease. Macrophages conversion to foam cells, cholesterol efflux from these cells and its differentiation into a pro- or anti-inflammatory phenotype are also important processes that contribute to atherosclerosis. In adults it has been reported that native and modified HDL and LDL play an important role in endothelial and macrophage function. In this review it is proposed that fetal lipoproteins could be also relevant factors involved in the detrimental vascular effects described in MSPH. Changes in the composition and function of neonatal lipoproteins compared to adults has been reported and, although in MSPH pregnancies the fetal lipid profile does not differ from MPH, differences in the lipidomic profiles of umbilical venous blood have been reported, which could have implications in the vascular function. In this review we summarize the available information regarding the effects of lipoproteins on endothelial and macrophage function, emphasizing its possible implications on fetal adverse outcomes associated to maternal hypercholesterolemia during pregnancy.
Collapse
|
11
|
Machado-Oliveira G, Ramos C, Marques ARA, Vieira OV. Cell Senescence, Multiple Organelle Dysfunction and Atherosclerosis. Cells 2020; 9:E2146. [PMID: 32977446 PMCID: PMC7598292 DOI: 10.3390/cells9102146] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is an age-related disorder associated with long-term exposure to cardiovascular risk factors. The asymptomatic progression of atherosclerotic plaques leads to major cardiovascular diseases (CVD), including acute myocardial infarctions or cerebral ischemic strokes in some cases. Senescence, a biological process associated with progressive structural and functional deterioration of cells, tissues and organs, is intricately linked to age-related diseases. Cell senescence involves coordinated modifications in cellular compartments and has been demonstrated to contribute to different stages of atheroma development. Senescence-based therapeutic strategies are currently being pursued to treat and prevent CVD in humans in the near-future. In addition, distinct experimental settings allowed researchers to unravel potential approaches to regulate anti-apoptotic pathways, facilitate excessive senescent cell clearance and eventually reverse atherogenesis to improve cardiovascular function. However, a deeper knowledge is required to fully understand cellular senescence, to clarify senescence and atherogenesis intertwining, allowing researchers to establish more effective treatments and to reduce the cardiovascular disorders' burden. Here, we present an objective review of the key senescence-related alterations of the major intracellular organelles and analyze the role of relevant cell types for senescence and atherogenesis. In this context, we provide an updated analysis of therapeutic approaches, including clinically relevant experiments using senolytic drugs to counteract atherosclerosis.
Collapse
Affiliation(s)
- Gisela Machado-Oliveira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| | | | | | - Otília V. Vieira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| |
Collapse
|
12
|
Chen Y, Li Q, Li Q, Xing S, Liu Y, Liu Y, Chen Y, Liu W, Feng F, Sun H. p62/SQSTM1, a Central but Unexploited Target: Advances in Its Physiological/Pathogenic Functions and Small Molecular Modulators. J Med Chem 2020; 63:10135-10157. [DOI: 10.1021/acs.jmedchem.9b02038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ying Chen
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Qi Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Qihang Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yang Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
- Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, Huaian 223005, People’s Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
- Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, Huaian 223005, People’s Republic of China
| |
Collapse
|
13
|
Vrieling F, Wilson L, Rensen PCN, Walzl G, Ottenhoff THM, Joosten SA. Oxidized low-density lipoprotein (oxLDL) supports Mycobacterium tuberculosis survival in macrophages by inducing lysosomal dysfunction. PLoS Pathog 2019; 15:e1007724. [PMID: 30998773 PMCID: PMC6490946 DOI: 10.1371/journal.ppat.1007724] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 04/30/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (DM) is a major risk factor for developing tuberculosis (TB). TB-DM comorbidity is expected to pose a serious future health problem due to the alarming rise in global DM incidence. At present, the causal underlying mechanisms linking DM and TB remain unclear. DM is associated with elevated levels of oxidized low-density lipoprotein (oxLDL), a pathologically modified lipoprotein which plays a key role during atherosclerosis development through the formation of lipid-loaded foamy macrophages, an event which also occurs during progression of the TB granuloma. We therefore hypothesized that oxLDL could be a common factor connecting DM to TB. To study this, we measured oxLDL levels in plasma samples of healthy controls, TB, DM and TB-DM patients, and subsequently investigated the effect of oxLDL treatment on human macrophage infection with Mycobacterium tuberculosis (Mtb). Plasma oxLDL levels were significantly elevated in DM patients and associated with high triglyceride levels in TB-DM. Strikingly, incubation with oxLDL strongly increased macrophage Mtb load compared to native or acetylated LDL (acLDL). Mechanistically, oxLDL -but not acLDL- treatment induced macrophage lysosomal cholesterol accumulation and increased protein levels of lysosomal and autophagy markers, while reducing Mtb colocalization with lysosomes. Importantly, combined treatment of acLDL and intracellular cholesterol transport inhibitor (U18666A) mimicked the oxLDL-induced lysosomal phenotype and impaired macrophage Mtb control, illustrating that the localization of lipid accumulation is critical. Collectively, these results demonstrate that oxLDL could be an important DM-associated TB-risk factor by causing lysosomal dysfunction and impaired control of Mtb infection in human macrophages. Tuberculosis (TB) is an infectious disease of the lungs caused by a bacterium, Mycobacterium tuberculosis (Mtb), and is responsible for over a million deaths per year worldwide. Population studies have demonstrated that type 2 diabetes mellitus (DM) is a risk factor for TB as it triples the risk of developing the disease. DM is a metabolic disorder which is generally associated with obesity, and is characterized by resistance to the pancreatic hormone insulin and high blood glucose and lipid levels. As the global incidence of DM is rising at an alarming rate, especially in regions where TB is common, it is important to understand precisely how DM increases the risk of developing TB. Both TB and DM are associated with the development of foamy macrophages, lipid-loaded white blood cells, which can be the result of a specific lipoprotein particle called oxidized low-density lipoprotein (oxLDL). Here, we demonstrated that DM patients have high blood levels of oxLDL, and generating foamy macrophages with oxLDL supported Mtb survival after infection as a result of faulty intracellular cholesterol accumulation. Our results propose a proof of concept for oxLDL as a risk factor for TB development, encouraging future studies on lipid-lowering therapies for TB-DM.
Collapse
Affiliation(s)
- Frank Vrieling
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Louis Wilson
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Gerhard Walzl
- DST/NRF Center of Excellence for Biomedical Tuberculosis Research, SA MRC Center for TB Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town, South Africa
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
14
|
Trapnell BC, Nakata K, Bonella F, Campo I, Griese M, Hamilton J, Wang T, Morgan C, Cottin V, McCarthy C. Pulmonary alveolar proteinosis. Nat Rev Dis Primers 2019; 5:16. [PMID: 30846703 DOI: 10.1038/s41572-019-0066-3] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pulmonary alveolar proteinosis (PAP) is a syndrome characterized by the accumulation of alveolar surfactant and dysfunction of alveolar macrophages. PAP results in progressive dyspnoea of insidious onset, hypoxaemic respiratory failure, secondary infections and pulmonary fibrosis. PAP can be classified into different types on the basis of the pathogenetic mechanism: primary PAP is characterized by the disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling and can be autoimmune (caused by elevated levels of GM-CSF autoantibodies) or hereditary (due to mutations in CSF2RA or CSF2RB, encoding GM-CSF receptor subunits); secondary PAP results from various underlying conditions; and congenital PAP is caused by mutations in genes involved in surfactant production. In most patients, pathogenesis is driven by reduced GM-CSF-dependent cholesterol clearance in alveolar macrophages, which impairs alveolar surfactant clearance. PAP has a prevalence of at least 7 cases per million individuals in large population studies and affects men, women and children of all ages, ethnicities and geographical locations irrespective of socioeconomic status, although it is more-prevalent in smokers. Autoimmune PAP accounts for >90% of all cases. Management aims at improving symptoms and quality of life; whole-lung lavage effectively removes excessive surfactant. Novel pathogenesis-based therapies are in development, targeting GM-CSF signalling, immune modulation and cholesterol homeostasis.
Collapse
Affiliation(s)
- Bruce C Trapnell
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Koh Nakata
- Bioscience Medical Research Center, Niigata University, Niigata, Japan
| | - Francesco Bonella
- Interstitial and Rare Lung Disease Unit, Pneumology Department, Ruhrlandklinik University Hospital, University of Essen, Essen, Germany
| | - Ilaria Campo
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Matthias Griese
- Pediatric Pneumology, University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - John Hamilton
- University of Melbourne, Parkville, Victoria, Australia
| | - Tisha Wang
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Cliff Morgan
- Department of Critical Care and Anaesthesia, Royal Brompton Hospital, London, UK
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases, University of Lyon, Lyon, France
| | - Cormac McCarthy
- Department of Medicine, St. Vincent's University Hospital and University College Dublin, Dublin, Ireland
| |
Collapse
|
15
|
Exploiting macrophage autophagy-lysosomal biogenesis as a therapy for atherosclerosis. Nat Commun 2017; 8:15750. [PMID: 28589926 PMCID: PMC5467270 DOI: 10.1038/ncomms15750] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/25/2017] [Indexed: 12/15/2022] Open
Abstract
Macrophages specialize in removing lipids and debris present in the atherosclerotic plaque. However, plaque progression renders macrophages unable to degrade exogenous atherogenic material and endogenous cargo including dysfunctional proteins and organelles. Here we show that a decline in the autophagy–lysosome system contributes to this as evidenced by a derangement in key autophagy markers in both mouse and human atherosclerotic plaques. By augmenting macrophage TFEB, the master transcriptional regulator of autophagy–lysosomal biogenesis, we can reverse the autophagy dysfunction of plaques, enhance aggrephagy of p62-enriched protein aggregates and blunt macrophage apoptosis and pro-inflammatory IL-1β levels, leading to reduced atherosclerosis. In order to harness this degradative response therapeutically, we also describe a natural sugar called trehalose as an inducer of macrophage autophagy–lysosomal biogenesis and show trehalose's ability to recapitulate the atheroprotective properties of macrophage TFEB overexpression. Our data support this practical method of enhancing the degradative capacity of macrophages as a therapy for atherosclerotic vascular disease. Dysfunction of autophagy in plaque macrophages aggravates atherosclerosis. Here the authors show that induction of macrophage autophagy–lysosomal biogenesis either genetically by overexpression of the master transcriptional regulator of this process, TFEB, or pharmacologically with trehalose is atheroprotective.
Collapse
|
16
|
Zeya B, Arjuman A, Chandra NC. Lectin-like Oxidized Low-Density Lipoprotein (LDL) Receptor (LOX-1): A Chameleon Receptor for Oxidized LDL. Biochemistry 2016; 55:4437-44. [DOI: 10.1021/acs.biochem.6b00469] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Bushra Zeya
- Department
of Biochemistry, All India Institute of Medical Sciences, Patna 801507, India
| | - Albina Arjuman
- Division of P&I, Indian Council of Medical Research, New Delhi 110 029, India
| | - Nimai Chand Chandra
- Department
of Biochemistry, All India Institute of Medical Sciences, Patna 801507, India
| |
Collapse
|
17
|
Xu X, Yuan X, Li N, Dewey WL, Li PL, Zhang F. Lysosomal cholesterol accumulation in macrophages leading to coronary atherosclerosis in CD38(-/-) mice. J Cell Mol Med 2016; 20:1001-13. [PMID: 26818887 PMCID: PMC4882979 DOI: 10.1111/jcmm.12788] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 12/13/2015] [Indexed: 12/16/2022] Open
Abstract
The disruption in transportation of oxLDL‐derived cholesterol and the subsequent lipid accumulation in macrophages are the hallmark events in atherogenesis. Our recent studies demonstrated that lysosomal Ca2+ messenger of nicotinic acid adenine dinucleotide phosphate (NAADP), an enzymatic product of CD38 ADP‐ribosylcyclase (CD38), promoted lipid endocytic trafficking in human fibroblast cells. The current studies are designed to examine the functional role of CD38/NAADP pathway in the regulation of lysosomal cholesterol efflux in atherosclerosis. Oil red O staining showed that oxLDL concentration‐dependently increased lipid buildup in bone marrow‐derived macrophages from both wild type and CD38−/−, but to a significant higher extent with CD38 gene deletion. Bodipy 493/503 fluorescence staining found that the deposited lipid in macrophages was mainly enclosed in lysosomal organelles and largely enhanced with the blockade of CD38/NAADP pathway. Filipin staining and direct measurement of lysosome fraction further revealed that the free cholesterol constituted a major portion of the total cholesterol segregated in lysosomes. Moreover, in situ assay disclosed that both lysosomal lumen acidity and the acid lipase activity were reduced upon cholesterol buildup in lysosomes. In CD38−/− mice, treatment with Western diet (12 weeks) produced atherosclerotic damage in coronary artery with striking lysosomal cholesterol sequestration in macrophages. These data provide the first experimental evidence that the proper function of CD38/NAADP pathway plays an essential role in promoting free cholesterol efflux from lysosomes and that a defection of this signalling leads to lysosomal cholesterol accumulation in macrophages and results in coronary atherosclerosis in CD38−/− mice.
Collapse
Affiliation(s)
- Xiaoyang Xu
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Xinxu Yuan
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - William L Dewey
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Fan Zhang
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
18
|
Wakita K, Morita SY, Okamoto N, Takata E, Handa T, Nakano M. Chylomicron remnant model emulsions induce intracellular cholesterol accumulation and cell death due to lysosomal destabilization. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:598-604. [DOI: 10.1016/j.bbalip.2015.01.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/15/2015] [Accepted: 01/27/2015] [Indexed: 10/24/2022]
|
19
|
Dubland JA, Francis GA. Lysosomal acid lipase: at the crossroads of normal and atherogenic cholesterol metabolism. Front Cell Dev Biol 2015; 3:3. [PMID: 25699256 PMCID: PMC4313778 DOI: 10.3389/fcell.2015.00003] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/07/2015] [Indexed: 01/01/2023] Open
Abstract
Unregulated cellular uptake of apolipoprotein B-containing lipoproteins in the arterial intima leads to the formation of foam cells in atherosclerosis. Lysosomal acid lipase (LAL) plays a crucial role in both lipoprotein lipid catabolism and excess lipid accumulation as it is the primary enzyme that hydrolyzes cholesteryl esters derived from both low density lipoprotein (LDL) and modified forms of LDL. Evidence suggests that as atherosclerosis progresses, accumulation of excess free cholesterol in lysosomes leads to impairment of LAL activity, resulting in accumulation of cholesteryl esters in the lysosome as well as the cytosol in foam cells. Impaired metabolism and release of cholesterol from lysosomes can lead to downstream defects in ATP-binding cassette transporter A1 regulation, needed to offload excess cholesterol from plaque foam cells. This review focuses on the role LAL plays in normal cholesterol metabolism and how the associated changes in its enzymatic activity may ultimately contribute to atherosclerosis progression.
Collapse
Affiliation(s)
- Joshua A Dubland
- Department of Medicine, Centre for Heart Lung Innovation, Providence Health Care Research Institute at St. Paul's Hospital, University of British Columbia Vancouver, BC, Canada
| | - Gordon A Francis
- Department of Medicine, Centre for Heart Lung Innovation, Providence Health Care Research Institute at St. Paul's Hospital, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
20
|
Trpkovic A, Resanovic I, Stanimirovic J, Radak D, Mousa SA, Cenic-Milosevic D, Jevremovic D, Isenovic ER. Oxidized low-density lipoprotein as a biomarker of cardiovascular diseases. Crit Rev Clin Lab Sci 2014; 52:70-85. [DOI: 10.3109/10408363.2014.992063] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
21
|
Arranz E, Jaime L, García-Risco MR, Fornari T, Reglero G, Santoyo S. Anti-inflammatory activity of rosemary extracts obtained by supercritical carbon dioxide enriched in carnosic acid and carnosol. Int J Food Sci Technol 2014. [DOI: 10.1111/ijfs.12656] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Elena Arranz
- Institute of Food Science Research (CIAL); Universidad Autónoma de Madrid (CEI UAM+CSIC); 28049 Madrid Spain
| | - Laura Jaime
- Institute of Food Science Research (CIAL); Universidad Autónoma de Madrid (CEI UAM+CSIC); 28049 Madrid Spain
| | - Monica R. García-Risco
- Institute of Food Science Research (CIAL); Universidad Autónoma de Madrid (CEI UAM+CSIC); 28049 Madrid Spain
| | - Tiziana Fornari
- Institute of Food Science Research (CIAL); Universidad Autónoma de Madrid (CEI UAM+CSIC); 28049 Madrid Spain
| | - Guillermo Reglero
- Institute of Food Science Research (CIAL); Universidad Autónoma de Madrid (CEI UAM+CSIC); 28049 Madrid Spain
- IMDEA-Food Institute; Universidad Autónoma de Madrid (CEI UAM+CSIC); 28049 Madrid Spain
| | - Susana Santoyo
- Institute of Food Science Research (CIAL); Universidad Autónoma de Madrid (CEI UAM+CSIC); 28049 Madrid Spain
| |
Collapse
|
22
|
Emanuel R, Sergin I, Bhattacharya S, Turner J, Epelman S, Settembre C, Diwan A, Ballabio A, Razani B. Induction of lysosomal biogenesis in atherosclerotic macrophages can rescue lipid-induced lysosomal dysfunction and downstream sequelae. Arterioscler Thromb Vasc Biol 2014; 34:1942-1952. [PMID: 25060788 DOI: 10.1161/atvbaha.114.303342] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Recent reports of a proatherogenic phenotype in mice with macrophage-specific autophagy deficiency have renewed interest in the role of the autophagy-lysosomal system in atherosclerosis. Lysosomes have the unique ability to process both exogenous material, including lipids and autophagy-derived cargo such as dysfunctional proteins/organelles. We aimed to understand the effects of an atherogenic lipid environment on macrophage lysosomes and to evaluate novel ways to modulate this system. APPROACH AND RESULTS Using a variety of complementary techniques, we show that oxidized low-density lipoproteins and cholesterol crystals, commonly encountered lipid species in atherosclerosis, lead to profound lysosomal dysfunction in cultured macrophages. Disruptions in lysosomal pH, proteolytic capacity, membrane integrity, and morphology are readily seen. Using flow cytometry, we find that macrophages isolated from atherosclerotic plaques also display features of lysosome dysfunction. We then investigated whether enhancing lysosomal function can be beneficial. Transcription factor EB (TFEB) is the only known transcription factor that is a master regulator of lysosomal biogenesis although its role in macrophages has not been studied. Lysosomal stress induced by chloroquine or atherogenic lipids leads to TFEB nuclear translocation and activation of lysosomal and autophagy genes. TFEB overexpression in macrophages further augments this prodegradative response and rescues several deleterious effects seen with atherogenic lipid loading as evidenced by blunted lysosomal dysfunction, reduced secretion of the proinflammatory cytokine interleukin-1β, enhanced cholesterol efflux, and decreased polyubiquitinated protein aggregation. CONCLUSIONS Taken together, these data demonstrate that lysosomal function is markedly impaired in atherosclerosis and suggest that induction of a lysosomal biogenesis program in macrophages has antiatherogenic effects.
Collapse
Affiliation(s)
- Roy Emanuel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| | - Ismail Sergin
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| | - Somashubhra Bhattacharya
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| | - Jaleisa Turner
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| | - Slava Epelman
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| | - Carmine Settembre
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| | - Abhinav Diwan
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| | - Andrea Ballabio
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| | - Babak Razani
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (R.E., I.S., S.B., S.E., A.D., B.R.) and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO (J.T., B.R.); John Cochran VA Medical Center, St. Louis, MO (A.D.); Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy (C.S., A.B.); and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (C.S., A.B.)
| |
Collapse
|
23
|
Sergin I, Razani B. Self-eating in the plaque: what macrophage autophagy reveals about atherosclerosis. Trends Endocrinol Metab 2014; 25:225-34. [PMID: 24746519 PMCID: PMC4061377 DOI: 10.1016/j.tem.2014.03.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/22/2014] [Accepted: 03/25/2014] [Indexed: 12/31/2022]
Abstract
Autophagy (or 'self-eating') is the process by which cellular contents are recycled to support downstream metabolism. An explosion in research in the past decade has implicated its role in both health and disease and established the importance of the autophagic response during periods of stress and nutrient deprivation. Atherosclerosis is a state where chronic exposure to cellular stressors promotes disease progression, and alterations in autophagy are predicted to be consequential. Recent reports linking macrophage autophagy to lipid metabolism, blunted inflammatory signaling, and an overall suppression of proatherogenic processes support this notion. We review these data and provide a framework for understanding the role of macrophage autophagy in the pathogenesis of atherosclerosis, one of the most formidable diseases of our time.
Collapse
Affiliation(s)
- Ismail Sergin
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|
24
|
Xie Q, Li F, Zhao SP. Ac‑hE‑18A‑NH2, a novel dual‑domain apolipoprotein mimetic peptide, inhibits apoptosis in macrophages by promoting cholesterol efflux. Mol Med Rep 2014; 9:1851-6. [PMID: 24639032 DOI: 10.3892/mmr.2014.2056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 02/25/2014] [Indexed: 11/06/2022] Open
Abstract
A novel synthetic dual-domain apolipoprotein (apo)-mimetic peptide, Ac-hE-18A-NH2, has been proposed to possess several apo A-I- and apo E-mimetic properties. This study investigated the protective effect of this peptide on oxidized low-density lipoprotein (ox-LDL)-induced apoptosis in RAW264.7 cells. For this purpose, RAW264.7 cells were exposed to 50 µg/ml ox-LDL for 48 h, and then incubated with the peptide Ac-hE-18A-NH2 at various concentrations. Apoptosis was detected using annexin V-fluorescein isothiocyanate staining and flow cytometric analysis. The study revealed that the peptide Ac-hE-18A-NH2 (1, 10 and 50 µg/ml) inhibited ox-LDL-mediated apoptosis, and this was accompanied by an increased rate of intracellular cholesterol efflux, and decreased total cholesterol levels in the cells in a concentration-dependent manner. The peptide also decreased caspase-3 activity and increased B-cell lymphoma 2 protein (Bcl-2) expression in macrophages in a dose-dependent manner. Moreover, blockage of cholesterol efflux by brefeldin A decreased the protective effect of Ac-hE-18A-NH2 against ox-LDL induced apoptosis, while increasing the cholesterol efflux by β-cyclodextrin administration led to a marked decrease in the rate of apoptosis of the cells. These findings demonstrate that the apo-mimetic peptide Ac-hE-18A-NH2 exerts a protective effect against apoptosis by reducing the accumulation of cholesterol.
Collapse
Affiliation(s)
- Qiong Xie
- Department of Cardiology, Hunan Provincial People's Hospital, Changsha, Hunan 410005, P.R. China
| | - Feng Li
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Shui-Ping Zhao
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
25
|
Activation of Nlrp3 inflammasomes enhances macrophage lipid-deposition and migration: implication of a novel role of inflammasome in atherogenesis. PLoS One 2014; 9:e87552. [PMID: 24475307 PMCID: PMC3903678 DOI: 10.1371/journal.pone.0087552] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/21/2013] [Indexed: 12/30/2022] Open
Abstract
Although Nlrp3 inflammasome activation in macrophages has been shown to be critical for the development of atherosclerosis upon atherogenic stimuli, it remains unknown whether activated Nlrp3 inflammasomes by other non-atherogenic stimuli induce alterations in macrophages that may contribute in the concert with other factors to atherogenesis. Thus, the present study tested the hypothesis that activation of Nlrp3 inflammasomes by ATP, which is a classical non-lipid danger stimulus, enhances the migration of macrophage and increases lipids deposition in macrophages accelerating foam cell formation. We first demonstrated that extracellular ATP (2.5 mM) markedly increased the formation and activation of Nlrp3 inflammasomes in bone marrow macrophages (BMMs) from wild type (Asc+/+) mice resulting in activation of caspase-1 and IL-1β production. In these Asc+/+ macrophages, such stimulation of inflammasomes by non-lipid ATP was similar to those induced by atherogenic stimuli such as cholesterol crystals or 7-ketocholesterol. Both non-lipid and lipid forms of stimuli induced formation and activation of Nlrp3 inflammasomes, which were prevented by Asc gene deletion. Interestingly, Asc+/+ BMMs had dramatic lipids accumulation after stimulation with ATP. Further, we demonstrated that large amount of cholesterol was accumulated in lysosomes of Asc+/+ BMMs when inflammasomes were activated by ATP. Such intracellular and lysosomal lipids deposition was not observed in Asc−/− BMMs and also prevented by caspase-1 inhibitor WEHD. In addition, in vitro and in vivo experiments revealed that migration of Asc+/+ BMMs increased due to stimulation of Nlrp3 inflammasomes, which was markedly attenuated in Asc−/− BMMs. Together, these results suggest that activation of Nlrp3 inflammasomes remarkably increases the susceptibility of macrophages to lipid deposition and their migration ability. Such novel action of inflammasomes may facilitate entry or retention of macrophages into the arterial wall, where they form foam cells and ultimately induce atherosclerosis.
Collapse
|
26
|
Bieghs V, Walenbergh SMA, Hendrikx T, van Gorp PJ, Verheyen F, Damink SWO, Masclee AA, Koek GH, Hofker MH, Binder CJ, Shiri-Sverdlov R. Trapping of oxidized LDL in lysosomes of Kupffer cells is a trigger for hepatic inflammation. Liver Int 2013; 33:1056-61. [PMID: 23617943 PMCID: PMC4040540 DOI: 10.1111/liv.12170] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Non-alcoholic steatohepatitis (NASH) is characterized by steatosis and inflammation. The transition from steatosis towards NASH represents a key step in pathogenesis, as it will set the stage for further severe liver damage. Under normal conditions, lipoproteins that are endocytosed by Kupffer cells (KCs) are easily transferred from the lysosomes into the cytoplasm. Oxidized LDL (oxLDL) that is taken up by the macrophages in vitro is trapped within the lysosomes, while acetylated LDL (acLDL) is leading to normal lysosomal hydrolysis, resulting in cytoplasmic storage. We have recently demonstrated that hepatic inflammation is correlated with lysosomal trapping of lipids. So far, a link between lysosomal trapping of oxLDL and inflammation was not established. We hypothesized that lysosomal trapping of oxLDL in KCs will lead to hepatic inflammation. METHODS Ldlr(-/-) mice were injected with LDL, acLDL and oxLDL and sacrificed after 2, 6 and 24 h. RESULTS Electron microscopy of KCs demonstrated that after oxLDL injection, small lipid inclusions were present inside the lysosomes after all time points and were mostly pronounced after 6 and 24 h. In contrast, no lipid inclusions were present inside KCs after LDL or acLDL injection. Hepatic expression of several inflammatory genes and scavenger receptors was higher after oxLDL injections compared with LDL or acLDL. CONCLUSIONS These data suggest that trapping of oxLDL inside lysosomes of KCs in vivo is causally linked to increased hepatic inflammatory gene expression. Our novel observations provide new bases for prevention and treatment of NASH.
Collapse
Affiliation(s)
- Veerle Bieghs
- Department of Molecular Genetics, Maastricht University, Maastricht, the Netherlands
,Department of Internal Medicine, University Hospital (RWTH), Aachen, Germany
| | | | - Tim Hendrikx
- Department of Molecular Genetics, Maastricht University, Maastricht, the Netherlands
| | - Patrick J. van Gorp
- Department of Molecular Genetics, Maastricht University, Maastricht, the Netherlands
| | - Fons Verheyen
- Department of Electron Microscopy Unit, CRISP, Department of Molecular Cell Biology, Maastricht University, Maastricht, the Netherlands
| | - Steven W. Olde Damink
- Department of General Surgery, Maastricht University, Maastricht, the Netherlands
,Department of HPB and Liver Transplantation Surgery, UCL Institute for Liver and Digestive Health, University College London Medical School, Royal Free Campus, London, UK
| | - Ad A. Masclee
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands
| | - Ger H. Koek
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands
| | - Marten H. Hofker
- Department of Pathology & Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
,Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
27
|
Levitan I, Volkov S, Subbaiah PV. Oxidized LDL: diversity, patterns of recognition, and pathophysiology. Antioxid Redox Signal 2010; 13:39-75. [PMID: 19888833 PMCID: PMC2877120 DOI: 10.1089/ars.2009.2733] [Citation(s) in RCA: 311] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 10/09/2009] [Accepted: 11/02/2009] [Indexed: 02/06/2023]
Abstract
Oxidative modification of LDL is known to elicit an array of pro-atherogenic responses, but it is generally underappreciated that oxidized LDL (OxLDL) exists in multiple forms, characterized by different degrees of oxidation and different mixtures of bioactive components. The variable effects of OxLDL reported in the literature can be attributed in large part to the heterogeneous nature of the preparations employed. In this review, we first describe the various subclasses and molecular composition of OxLDL, including the variety of minimally modified LDL preparations. We then describe multiple receptors that recognize various species of OxLDL and discuss the mechanisms responsible for the recognition by specific receptors. Furthermore, we discuss the contentious issues such as the nature of OxLDL in vivo and the physiological oxidizing agents, whether oxidation of LDL is a prerequisite for atherogenesis, whether OxLDL is the major source of lipids in foam cells, whether in some cases it actually induces cholesterol depletion, and finally the Janus-like nature of OxLDL in having both pro- and anti-inflammatory effects. Lastly, we extend our review to discuss the role of LDL oxidation in diseases other than atherosclerosis, including diabetes mellitus, and several autoimmune diseases, such as lupus erythematosus, anti-phospholipid syndrome, and rheumatoid arthritis.
Collapse
Affiliation(s)
- Irena Levitan
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA.
| | | | | |
Collapse
|
28
|
Grandl M, Schmitz G. Fluorescent high-content imaging allows the discrimination and quantitation of E-LDL-induced lipid droplets and Ox-LDL-generated phospholipidosis in human macrophages. Cytometry A 2010; 77:231-42. [PMID: 20014301 DOI: 10.1002/cyto.a.20828] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Macrophage foam cells formed during uptake of atherogenic lipoproteins are a hallmark of atherosclerotic lesion development. In this study, human macrophages were incubated with two prototypic atherogenic LDL modifications enzymatically degraded LDL (E-LDL) and oxidized LDL (Ox-LDL) prepared from the same donor LDL. To detect differences in macrophage lipid storage, fluorescent high-content imaging was used. Lipid droplets were stained using Bodipy 493/503, and the fluorescent phospholipid probe NBD-PE was used to detect endolysosomal phospholipidosis in high-content imaging assays. The phospholipidosis assay was validated using phospholipidosis-inducing cationic amphiphilic drugs. In addition, neutral lipids and phospholipidosis were determined using LipidTOX. Images of 96-well cell culture microtiter plates were captured with multichannel laser-based high-content confocal microscopy, and subsequently cell- and well-based data were analyzed. E-LDL-loaded macrophages show increased intensity of Bodipy 493/503 and LipidTOX-Green neutral lipid droplet staining and a greater mean area and number of lipid droplets per cell compared to Ox-LDL-loaded and M-CSF-differentiated control macrophages. In contrast, Ox-LDL-loaded macrophages show increased intensity of NBD-PE and LipidTOX-Red detectable phospholipidosis in the endolysosomal compartment compared to E-LDL-loaded and M-CSF-differentiated macrophages. Treatment with the peroxisome proliferator-activated receptor-gamma agonist pioglitazone leads to lipid droplet induction depending on the lipid loading state of the macrophages. These results indicate that E-LDL preferentially induces lipid droplets, while Ox-LDL provokes endolysosomal phospholipidosis in human macrophages representing two different lipid storage principles. Therefore, fluorescent high-content imaging is a useful tool to discriminate between and quantify lipid storage compartments in macrophages also in response to drugs affecting cellular lipid metabolism.
Collapse
Affiliation(s)
- Margot Grandl
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | | |
Collapse
|
29
|
Smith KJ, Twal WO, Soodavar F, Virella G, Lopes-Virella MF, Hammad SM. Heat shock protein 70B' (HSP70B') expression and release in response to human oxidized low density lipoprotein immune complexes in macrophages. J Biol Chem 2010; 285:15985-93. [PMID: 20348092 DOI: 10.1074/jbc.m110.113605] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heat shock proteins (HSPs) have been implicated in the activation and survival of macrophages. This study examined the role of HSP70B', a poorly characterized member of the HSP70 family, in response to oxidatively modified LDL (oxLDL) and immune complexes prepared with human oxLDL and purified human antibodies to oxLDL (oxLDL-IC) in monocytic and macrophage cell lines. Immunoblot analysis of cell lysates and conditioned medium from U937 cells treated with oxLDL alone revealed an increase in intracellular HSP70B' protein levels accompanied by a concomitant increase in HSP70B' extracellular levels. Fluorescence immunohistochemistry and confocal microscopy, however, demonstrated that oxLDL-IC stimulated the release of HSP70B', which co-localized with cell-associated oxLDL-IC. In HSP70B'-green fluorescent protein-transfected mouse RAW 264.7 cells, oxLDL-IC-induced HSP70B' co-localized with membrane-associated oxLDL-IC as well as the lipid moiety of internalized oxLDL-IC. Furthermore, the data demonstrated that HSP70B' is involved in cell survival, and this effect could be mediated by sphingosine kinase 1 (SK1) activation. An examination of regularly implicated cytokines revealed a significant relationship between HSP70B' and the release of the anti-inflammatory cytokine interleukin-10 (IL-10). Small interfering RNA knockdown of HSP70B' resulted in a corresponding decrease in SK1 mRNA levels and SK1 phosphorylation as well as increased release of IL-10. In conclusion, these findings suggest that oxLDL-IC induce the synthesis and release of HSP70B', and once stimulated, HSP70B' binds to the cell-associated and internalized lipid moiety of oxLDL-IC. The data also implicate HSP70B' in key cellular functions, such as regulation of SK1 activity and release of IL-10, which influence macrophage activation and survival.
Collapse
Affiliation(s)
- Kent J Smith
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|
30
|
Sun Y, Ishibashi M, Seimon T, Lee M, Sharma SM, Fitzgerald KA, Samokhin AO, Wang Y, Sayers S, Aikawa M, Jerome WG, Ostrowski MC, Bromme D, Libby P, Tabas IA, Welch CL, Tall AR. Free cholesterol accumulation in macrophage membranes activates Toll-like receptors and p38 mitogen-activated protein kinase and induces cathepsin K. Circ Res 2009; 104:455-65. [PMID: 19122179 DOI: 10.1161/circresaha.108.182568] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The molecular events linking lipid accumulation in atherosclerotic plaques to complications such as aneurysm formation and plaque disruption are poorly understood. BALB/c-Apoe(-/-) mice bearing a null mutation in the Npc1 gene display prominent medial erosion and atherothrombosis, whereas their macrophages accumulate free cholesterol in late endosomes and show increased cathepsin K (Ctsk) expression. We now show increased cathepsin K immunostaining and increased cysteinyl proteinase activity using near infrared fluorescence imaging over proximal aortas of Apoe(-/-), Npc1(-/-) mice. In mechanistic studies, cholesterol loading of macrophage plasma membranes (cyclodextrin-cholesterol) or endosomal system (AcLDL+U18666A or Npc1 null mutation) activated Toll-like receptor (TLR) signaling, leading to sustained phosphorylation of p38 mitogen-activated protein kinase and induction of p38 targets, including Ctsk, S100a8, Mmp8, and Mmp14. Studies in macrophages from knockout mice showed major roles for TLR4, following plasma membrane cholesterol loading, and for TLR3, after late endosomal loading. TLR signaling via p38 led to phosphorylation and activation of the transcription factor Microphthalmia transcription factor, acting at E-box elements in the Ctsk promoter. These studies suggest that free cholesterol enrichment of either plasma or endosomal membranes in macrophages leads to activation of signaling via various TLRs, prolonged p38 mitogen-activated protein kinase activation, and induction of Mmps, Ctsk, and S100a8, potentially contributing to plaque complications.
Collapse
Affiliation(s)
- Yu Sun
- Department of Medicine, Columbia University, New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Endolysosomal phospholipidosis and cytosolic lipid droplet storage and release in macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2008; 1791:524-39. [PMID: 19146988 DOI: 10.1016/j.bbalip.2008.12.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 12/08/2008] [Accepted: 12/11/2008] [Indexed: 12/12/2022]
Abstract
This review summarizes the current knowledge of endolysosomal and cytoplasmic lipid storage in macrophages induced by oxidized LDL (Ox-LDL), enzymatically degraded LDL (E-LDL) and other atherogenic lipoprotein modifications, and their relation to the adapter protein 3 (AP-3) dependent ABCA1 and ABCG1 cellular lipid efflux pathways. We compare endolysosomal lipid storage caused either through drug induced phospholipidosis, inheritable endolysosomal and cytosolic lipid storage disorders and Ox-LDL or E-LDL induced phagosomal uptake and cytosolic lipid droplet storage in macrophages. Ox-LDL is resistant to rapid endolysosomal hydrolysis and is trapped within the endolysosomal compartment generating lamellar bodies which resemble the characteristics of phospholipidosis. Various inherited lysosomal storage diseases including sphingolipidosis, glycosphingolipidosis and cholesterylester storage diseases also present a phospholipidosis phenotype. In contrast E-LDL resembling coreless unesterified cholesterol enriched LDL-particles, with a multilamellar, liposome-like structure, lead to rapid phagosomal degradation and cytosolic lipid droplet accumulation. As a consequence the uptake of E-LDL through type I and type II phagocytosis leads to increased lipid droplet formation and moderate upregulation of ABCA1 and ABCG1 while uptake of Ox-LDL leads to a rapid expansion of the lysosomal compartment and a pronounced upregulation of the ABCA1/ABCG1/AP-3 lipid efflux pathway.
Collapse
|
32
|
Jerome WG, Cox BE, Griffin EE, Ullery JC. Lysosomal cholesterol accumulation inhibits subsequent hydrolysis of lipoprotein cholesteryl ester. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2008; 14:138-49. [PMID: 18312718 PMCID: PMC2837357 DOI: 10.1017/s1431927608080069] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Accepted: 08/07/2007] [Indexed: 05/20/2023]
Abstract
Human macrophages incubated for prolonged periods with mildly oxidized LDL (oxLDL) or cholesteryl ester-rich lipid dispersions (DISP) accumulate free and esterified cholesterol within large, swollen lysosomes similar to those in foam cells of atherosclerosis. The cholesteryl ester (CE) accumulation is, in part, the result of inhibition of lysosomal hydrolysis due to increased lysosomal pH mediated by excessive lysosomal free cholesterol (FC). To determine if the inhibition of hydrolysis was long lived and further define the extent of the lysosomal defect, we incubated THP-1 macrophages with oxLDL or DISP to produce lysosome sterol engorgement and then chased with acetylated LDL (acLDL). Unlike oxLDL or DISP, CE from acLDL normally is hydrolyzed rapidly. Three days of incubation with oxLDL or DISP produced an excess of CE in lipid-engorged lysosomes, indicative of inhibition. After prolonged oxLDL or DISP pretreatment, subsequent hydrolysis of acLDL CE was inhibited. Coincident with the inhibition, the lipid-engorged lysosomes failed to maintain an acidic pH during both the initial pretreatment and subsequent acLDL incubation. This indicates that the alterations in lysosomes were general, long lived, and affected subsequent lipoprotein metabolism. This same phenomenon, occurring within atherosclerotic foam cells, could significantly affect lesion progression.
Collapse
Affiliation(s)
- W Gray Jerome
- Department of Pathology, Vanderbilt University School of Medicine, South Nashville, TN 37232-2561, USA.
| | | | | | | |
Collapse
|
33
|
Levitan I, Gooch KJ. Lipid rafts in membrane-cytoskeleton interactions and control of cellular biomechanics: actions of oxLDL. Antioxid Redox Signal 2007; 9:1519-34. [PMID: 17576163 DOI: 10.1089/ars.2007.1686] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Membrane-cytoskeleton coupling is known to play major roles in a plethora of cellular responses, such as cell growth, differentiation, polarization, motility, and others. In this review, the authors discuss the growing amount of evidence indicating that membrane-cytoskeleton interactions are regulated by the lipid composition of the plasma membrane, suggesting that cholesterol-rich membrane domains (lipid rafts), including caveolae, are essential for membrane-cytoskeleton coupling. Several models for raft-cytoskeleton interactions are discussed. Also described is the evidence suggesting that raft-cytoskeleton interactions play key roles in several cytoskeleton-dependent processes, particularly in the regulation of cellular biomechanical properties. To address further the physiological significance of raft-cytoskeleton coupling, the authors focus on the impact of oxidized low density lipoproteins, one of the major cholesterol carriers and proatherogenic factors, on the integrity of lipid rafts/caveolae, and on the organization of the cytoskeleton. Finally, the authors review the recent studies showing that oxLDL and cholesterol depletion have similar impacts on the biomechanical properties of vascular endothelial cells, which in turn affect endothelial angiogenic potential.
Collapse
Affiliation(s)
- Irena Levitan
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA.
| | | |
Collapse
|
34
|
Cox BE, Griffin EE, Ullery JC, Jerome WG. Effects of cellular cholesterol loading on macrophage foam cell lysosome acidification. J Lipid Res 2007; 48:1012-21. [PMID: 17308299 DOI: 10.1194/jlr.m600390-jlr200] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Macrophages incubated with mildly oxidized low density lipoprotein (OxLDL), aggregated low density lipoprotein (AggLDL), or cholesteryl ester-rich lipid dispersions (DISPs) accumulate cholesterol in lysosomes followed by an inhibition of lysosomal cholesteryl ester (CE) hydrolysis. The variety of cholesterol-containing particles producing inhibition of hydrolysis suggests that inhibition may relate to general changes in lysosomes. Lysosome pH is a key mediator of activity and thus is a potential mechanism for lipid-induced inhibition. We investigated the effects of cholesterol accumulation on THP-1 macrophage lysosome pH. Treatment with OxLDL, AggLDL, and DISPs resulted in inhibition of the lysosome's ability to maintain an active pH and concomitant decreases in CE hydrolysis. Consistent with an overall disruption of lysosome function, exposure to OxLDL or AggLDL reduced lysosomal apolipoprotein B degradation. The lysosomal cholesterol sequestration and inactivation are not observed in cholesterol-equivalent cells loaded using acetylated low density lipoprotein (AcLDL). However, AcLDL-derived cholesterol in the presence of progesterone (to block cholesterol egression from lysosomes) inhibited lysosome acidification. Lysosome inhibition was not attributable to a decrease in the overall levels of vacuolar ATPase. However, augmentation of membrane cholesterol in isolated lysosomes inhibited vacuolar ATPase-dependent pumping of H+ ions into lysosomes. These data indicate that lysosomal cholesterol accumulation alters lysosomes in ways that could exacerbate foam cell formation and influence atherosclerotic lesion development.
Collapse
Affiliation(s)
- Brian E Cox
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
35
|
Yancey PG, Jerome WG, Yu H, Griffin EE, Cox BE, Babaev VR, Fazio S, Linton MF. Severely altered cholesterol homeostasis in macrophages lacking apoE and SR-BI. J Lipid Res 2007; 48:1140-9. [PMID: 17299204 DOI: 10.1194/jlr.m600539-jlr200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mice deficient in scavenger receptor class B type I (SR-BI) and apolipoprotein E (apoE) [double knockout (DKO) mice] develop dyslipidemia, accelerated atherosclerosis, and myocardial infarction, and die prematurely. We examined effects of apoE and SR-BI deficiency on macrophage cholesterol homeostasis. DKO macrophages had increased total cholesterol (TC) stores (220-380 microg/mg protein) compared with apoE-/- cells (40 microg/mg), showed significant lysosomal lipid engorgement, and increased their TC by 34% after exposure to HDL. DKO macrophages from apoE-/- mice reconstituted with DKO bone marrow showed less cholesterol accumulation (89 microg/mg), suggesting that the dyslipidemia of DKO mice explains part of the cellular cholesterol defect. However, analyses of DKO and apoE-/- macrophages from transplanted apoE-/- mice revealed a role for macrophage SR-BI, inasmuch as the TC in DKO macrophages increased by 10% in the presence of HDL, whereas apoE-/- macrophage TC decreased by 33%. After incubation with HDL, the free cholesterol (FC) increased by 29% in DKO macrophages, and decreased by 8% in apoE-/- cells, and only DKO cells had FC in large peri-nuclear pools. Similar trends were observed with apoA-I as an acceptor. Thus, the abnormal cholesterol homeostasis of DKO macrophages is due to the plasma lipid environment of DKO mice and to altered trafficking of macrophage cholesterol. Both factors are likely to contribute to the accelerated atherosclerosis in DKO mice.
Collapse
Affiliation(s)
- Patricia G Yancey
- Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Jerome WG. Advanced atherosclerotic foam cell formation has features of an acquired lysosomal storage disorder. Rejuvenation Res 2006; 9:245-55. [PMID: 16706652 DOI: 10.1089/rej.2006.9.245] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis is a disease of large- and medium-sized arteries. Complications from atherosclerosis remain a serious cause of morbidity and mortality in industrialized countries. The disease begins very early in life and effects most people in the West. However, because the progression of the disease is slow, symptoms usually do not occur until after the fifth decade of life. Because atherosclerosis is a ubiquitous occurrence throughout the world, as life expectancy is prolonged most populations will see increasing numbers of deaths from complications of atherosclerosis unless there are dramatic advances in treatment. Because it begins so early in life, current treatment is aimed at slowing or reversing the progression of the disease rather than eliminating the initiating steps. Changes in diet and exercise, cholesterol-lowering drugs, and improvements in surgical treatments have made significant inroads into prolonging life, but much work is still required. To proceed further, a better understanding is needed of the underlying causes of disease progression. In this regard, evidence is mounting that the foam cells of the lesion (a critical cell in atherosclerosis progression) exhibit characteristics of an acquired lysosomal storage disorder. In this review the evidence for this conclusion is reviewed and the ramifications of this conclusion are explored with regard to the understanding of disease progression mechanisms, possible improvements in treatment, and their role in increasing life expectancy.
Collapse
Affiliation(s)
- W Gray Jerome
- Department of Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2561, USA.
| |
Collapse
|
37
|
Zhao HL, Cho KH, Ha YW, Jeong TS, Lee WS, Kim YS. Cholesterol-lowering effect of platycodin D in hypercholesterolemic ICR mice. Eur J Pharmacol 2006; 537:166-73. [PMID: 16626693 DOI: 10.1016/j.ejphar.2006.03.032] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2005] [Revised: 01/03/2006] [Accepted: 03/13/2006] [Indexed: 11/18/2022]
Abstract
This study investigates the in vivo hypocholesterolemic action of platycodin D and its in vitro evidence for the cholesterol-lowering properties. In order to examine the effects of platycodin D on hypercholesterolemia in male ICR mice, platycodin D with doses of 15, 30 or 50 mg/kg was orally administered for 8 weeks. Changes in body weight and daily food intake were measured regularly during the experimental period. Final contents of triglyceride and different types of cholesterol in the serum, livers and feces were determined. The effects of platycodin D on cholesterol metabolism were further investigated with several in vitro assays, including antioxidant effect on low density lipoprotein oxidation, inhibition of human acyl-coenzyme A:cholesterol acyltransferase (hACAT) and serum lipoprotein associated-phospholipase A(2) (Lp-PLA(2)), as well as the regulation of farnesoid X receptor. The formation of insoluble complex between platycodin D and cholesterol was also investigated. Following an eight week experimental period, the body weights of platycodin D-fed mice were less than those of control mice on a high cholesterol diet by 11.2+/-5% (P<0.01) with 15 mg/kg platycodin D, 11.7+/-5% (P<0.01) with 30 mg/kg platycodin D, and 23.4+/-7.9% (P<0.0001) with 50 mg/kg platycodin D, respectively. A decrease in daily food consumption was also noted in most of the treated animals. Triglyceride and cholesterol concentrations were decreased in serums and livers, but increased in feces. Some of the in vitro observations revealed that the hypocholesterolemic effect of platycodin D is partly associated with inhibition to hACAT activity and antagonism to the farnesoid X receptor as well as the formation of insoluble complex with between platycodin D and cholesterol. Both in vivo and in vitro results demonstrate a potential value of platycodin D as a novel cholesterol-lowering and anti-atherogenic candidate.
Collapse
Affiliation(s)
- Hai Lin Zhao
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 110-460, Republic of Korea
| | | | | | | | | | | |
Collapse
|
38
|
Cheng B, Yu Q, Bai Z, Ke L, He P. Mechanisms of increased expression of toll-like receptor-4 in human monocyte/macrophage-derived foam cells. ACTA ACUST UNITED AC 2006; 25:477-9. [PMID: 16463649 DOI: 10.1007/bf02895992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The mechanisms of increased expression of toll-like receptor 4 (TLR-4) during the formation of foam cells were explored. Low density lipoprotein (LDL) was prepared by density gradient ultracentrifugation and oxidized by incubation with CuCl2. The human monocytic leukemia cell line (THP-1) was cultured in RPMI1640. The differentiation of THP-1 cells into macrophages (MPs) was induced by using myristate acetate (PMA) for 48 h. The macrophages were then incubated with oxidized LDL (ox-LDL) to generate foam cells (FCs). The mRNA and protein expression levels of human TLR-4 were detected by immunocytochemistry, Western blotting and reverse transcription polymerase chain reaction (RT-PCR). The results showed that the TLR-4 mRNA and the protein expression levels were significantly increased during the differentiation of monocytes into macrophages (P < 0.05) as well as during the formation of lipid-laden foam cells (P < 0.05). It was concluded that the upregulation of human TLR-4 gene expression during the differentiation of monocytes into macrophages and the differentiation of macrophages into foam cells could increase TLR-4 protein synthesis dramatically, which may enhance the ability of foam cells inflammation reaction in atherosclerosis.
Collapse
Affiliation(s)
- Bei Cheng
- Department of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | | | | | | | | |
Collapse
|
39
|
Jiang P, Yan PK, Chen JX, Zhu BY, Lei XY, Yin WD, Liao DF. High density lipoprotein 3 inhibits oxidized low density lipoprotein-induced apoptosis via promoting cholesterol efflux in RAW264.7 cells. Acta Pharmacol Sin 2006; 27:151-7. [PMID: 16412263 DOI: 10.1111/j.1745-7254.2006.00261.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AIM To investigate the protective effect of high density lipoprotein 3 (HDL3) on oxidized low density lipoprotein (ox-LDL)-induced apoptosis in RAW264.7 cells. METHODS RAW264.7 cells were exposed to 50 mg/L ox-LDL for various durations up to 48 h, and apoptosis was detected using Hoechst 33258 staining and flow cytometric analysis. Total cholesterol levels were detected by high performance liquid chromatography, cholesterol efflux was determined by Tritium labeling, and the cellular lipid droplets were assayed by oil red O staining. RESULTS Treatment with 50 mg/L ox-LDL for 12, 24, and 48 h increased the apoptotic rate of RAW264.7 cells in a time-dependent manner. The peak apoptotic rate (47.7%) was observed after 48 h incubation. HDL3 at various concentrations (50 mg/L, 100 mg/L, and 200 mg/L) inhibited the ox-LDL (50 mg/L for 48 h)-mediated apoptosis that was accompanied by an increased rate of intracellular cholesterol efflux, and decreased total cholesterol levels in cells in a concentration-dependent manner. Blockage of cholesterol efflux by brefeldin decreased the protective effect of HDL3 on ox-LDL-induced apoptosis. Increase of the cholesterol efflux effected by another cholesterol acceptor,beta-cyclodextrin, led to a dramatic decrease in the apoptotic rate of cells. CONCLUSION HDL3 antagonizes ox-LDL-induced apoptosis in RAW264.7 cells, through reducing the accumulation of toxic cholesterol.
Collapse
Affiliation(s)
- Pei Jiang
- Division of Pharmacoproteomics, Institute of Pharmacy and Pharmacology, Nanhua University, Hengyang 421001, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Griffin EE, Ullery JC, Cox BE, Jerome WG. Aggregated LDL and lipid dispersions induce lysosomal cholesteryl ester accumulation in macrophage foam cells. J Lipid Res 2005; 46:2052-60. [PMID: 16024919 DOI: 10.1194/jlr.m500059-jlr200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Macrophage foam cells in atherosclerotic lesions accumulate substantial cholesterol stores within large, swollen lysosomes. Previous studies with mildly oxidized low density lipoprotein (OxLDL)-treated THP-1 macrophages suggest an initial buildup of free cholesterol (FC), followed by an inhibition of lysosomal cholesteryl ester (CE) hydrolysis and a subsequent lysosomal accumulation of unhydrolyzed lipoprotein CE. We examined whether other potential sources of cholesterol found within atherosclerotic lesions could also induce similar lysosomal accumulation. Biochemical analysis combined with microscopic analysis showed that treatment of THP-1 macrophages with aggregated low density lipoprotein (AggLDL) or CE-rich lipid dispersions (DISP) produced a similar lysosomal accumulation of both FC and CE. Co-treatment with an ACAT inhibitor, CP113,818, confirmed that the CE accumulation was primarily the result of the inhibition of lysosomal CE hydrolysis. The rate of unhydrolyzed CE buildup was more rapid with DISP than with AggLDL. However, with both treatments, FC appeared to accumulate in lysosomes before the inhibition in hydrolysis and CE accumulation, a sequence shared with mildly OxLDL. Thus, lysosomal accumulation of FC and CE can be attributable to more general mechanisms than just the inhibition of hydrolysis by oxidized lipids.
Collapse
Affiliation(s)
- Evelyn E Griffin
- Department of Pathology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
41
|
Dove DE, Su YR, Zhang W, Jerome WG, Swift LL, Linton MF, Fazio S. ACAT1 deficiency disrupts cholesterol efflux and alters cellular morphology in macrophages. Arterioscler Thromb Vasc Biol 2004; 25:128-34. [PMID: 15499044 DOI: 10.1161/01.atv.0000148323.94021.e5] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Acyl-coenzyme A: cholesterol acyltransferase (ACAT) converts intracellular free cholesterol (FC) into cholesteryl esters (CE) for storage in lipid droplets. Recent studies in our laboratory have shown that the deletion of the macrophage ACAT1 gene results in apoptosis and increased atherosclerotic lesion area in the aortas of hyperlipidemic mice. The objective of the current study was to elucidate the mechanism of the increased atherosclerosis. METHODS AND RESULTS CE storage and FC efflux were studied in ACAT1(-/-) peritoneal macrophages that were treated with acetylated low-density lipoprotein (acLDL). Our results show that efflux of cellular cholesterol was reduced by 25% in ACAT1-deficient cells compared with wild-type controls. This decrease occurred despite the upregulated expression of ABCA1, an important mediator of cholesterol efflux. In contrast, ACAT1 deficiency increased efflux of the cholesterol derived from acLDL by 32%. ACAT1-deficient macrophages also showed a 26% increase in the accumulation of FC derived from acLDL, which was associated with a 75% increase in the number of intracellular vesicles. CONCLUSIONS Together, these data show that macrophage ACAT1 influences the efflux of both cellular and lipoprotein-derived cholesterol and propose a pathway for the pro-atherogenic transformation of ACAT1(-/-) macrophages.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1
- ATP-Binding Cassette Transporters/biosynthesis
- ATP-Binding Cassette Transporters/metabolism
- Acetyl-CoA C-Acetyltransferase
- Animals
- Biological Transport, Active/physiology
- Cholesterol/metabolism
- Cholesterol/toxicity
- Cholesterol Esters/metabolism
- Cholesterol, LDL/chemistry
- Cholesterol, LDL/metabolism
- Endosomes/chemistry
- Foam Cells/metabolism
- Lysosomes/chemistry
- Macrophages, Peritoneal/chemistry
- Macrophages, Peritoneal/enzymology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/ultrastructure
- Mice
- Microscopy, Electron, Transmission/methods
- Microscopy, Fluorescence/methods
- RNA, Messenger/metabolism
- Sterol O-Acyltransferase/deficiency
- Sterol O-Acyltransferase/physiology
- Tritium/metabolism
Collapse
Affiliation(s)
- Dwayne E Dove
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tenn 37232-6300, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Atherosclerosis remains the leading cause of death in the developed countries. In addition to lipid-lowering drugs - statins, dietary control, and exercise, new approaches are needed for the treatment and prevention of atherosclerosis. This review will focus on the role(s) of lysosomal acid lipase and its use as an enzyme therapy to reduce atherosclerotic lesions in a mouse model and to examine the molecular basis supporting this novel strategy and its mechanism of effect. RECENT FINDINGS Administration of human lysosomal acid lipase via tail vein into mice with atherosclerosis eliminates early aortic and coronary ostial lesions and reduces lesional size in advanced disease. The reduction of advanced lesional area is related to decreases in foamy macrophages, collagen positive areas, and necrotic areas. Compared with sham-treated mice, the human lysosomal acid lipase-treated mice also have reduced levels of plasma cholesteryl esters, and reduced levels of hepatic cholesterol and triglycerides. SUMMARY These studies indicate that administrated lysosomal acid lipase affects the atherogenesis by at least two mechanisms: (1) direct targeting of lesional macrophages with resultant decreases in cholesteryl esters and triglyceride in the lysosomes of macrophages in the lesions; (2) systemic effects that mediate the liver to reduce the hepatic cholesteryl ester and triglyceride release, possibly leading to reduced production of VLDL and LDL.
Collapse
Affiliation(s)
- Hong Du
- The Children's Hospital Research Foundation of Cincinnati Children's Hospital Medical Center, and the Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio 45229-3039, USA
| | | |
Collapse
|
43
|
Whatling C, Björk H, Gredmark S, Hamsten A, Eriksson P. Effect of macrophage differentiation and exposure to mildly oxidized LDL on the proteolytic repertoire of THP-1 monocytes. J Lipid Res 2004; 45:1768-76. [PMID: 15210849 DOI: 10.1194/jlr.m400195-jlr200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lipid-laden monocyte/macrophages in atherosclerotic plaques can produce a range of proteinases capable of degrading components of the plaque extracellular matrix, an event that may weaken plaques, rendering them vulnerable to rupture. The effects of differentiation from monocytes to macrophages and exposure to mildly oxidized LDL (Ox-LDL) on the expression of a range of proteinases and their inhibitors were assessed in the human THP-1 cell line. Of 56 proteinases/inhibitors investigated, 17 were upregulated during macrophage differentiation, including several matrix metalloproteinases (MMPs) and cathepsins along with their native inhibitors. Similarly, expression of matrix-degrading proteinases was also increased during differentiation of human primary macrophages. In conjunction, the proteolytic capacity of the cells increased, as assessed by substrate zymography. Subsequent exposure of differentiated THP-1 cells to mildly Ox-LDL increased the expression of a control gene (adipocyte lipid binding protein) and increased the activity of nuclear factor-kappaB and activator protein-1 in serum-free conditions but did not significantly affect the expression of any of the proteinases or inhibitors investigated. These results indicate that in this model macrophage differentiation, rather than exposure to Ox-LDL, has a more important effect on the expression of genes involved in extracellular matrix remodeling.
Collapse
Affiliation(s)
- Carl Whatling
- Atherosclerosis Research Unit, King Gustaf V Research Institute, Karolinska Hospital, S-171 76 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
44
|
Yang L, Yang JB, Chen J, Yu GY, Zhou P, Lei L, Wang ZZ, Cy Chang C, Yang XY, Chang TY, Li BL. Enhancement of human ACAT1 gene expression to promote the macrophage-derived foam cell formation by dexamethasone. Cell Res 2004; 14:315-23. [PMID: 15353128 DOI: 10.1038/sj.cr.7290231] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In macrophages, the accumulation of cholesteryl esters synthesized by the activated acyl-coenzyme A:cholesterol acyltransferase-1 (ACAT1) results in the foam cell formation, a hallmark of early atherosclerotic lesions. In this study, with the treatment of a glucocorticoid hormone dexamethasone (Dex), lipid staining results clearly showed the large accumulation of lipid droplets containing cholesteryl esters in THP-1-derived macrophages exposed to lower concentration of the oxidized low-density lipoprotein (ox-LDL). More notably, when treated together with specific anti-ACAT inhibitors, the abundant cholesteryl ester accumulation was markedly diminished in THP-1-derived macrophages, confirming that ACAT is the key enzyme responsible for intracellular cholesteryl ester synthesis. RT-PCR and Western blot results indicated that Dex caused up-regulation of human ACAT1 expression at both the mRNA and protein levels in THP-1 and THP-1-derived macrophages. The luciferase activity assay demonstrated that Dex could enhance the activity of human ACAT1 gene P1 promoter, a major factor leading to the ACAT1 activation, in a cell-specific manner. Further experimental evidences showed that a glucocorticoid response element (GRE) located within human ACAT1 gene P1 promoter to response to the elevation of human ACAT1 gene expression by Dex could be functionally bound with glucocorticoid receptor (GR) proteins. These data supported the hypothesis that the clinical treatment with Dex, which increased the incidence of atherosclerosis, may in part due to enhancing the ACAT1 expression to promote the accumulation of cholesteryl esters during the macrophage-derived foam cell formation, an early stage of atherosclerosis.
Collapse
Affiliation(s)
- Li Yang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, 320 Yueyang Rd, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kawai Y, Saito A, Shibata N, Kobayashi M, Yamada S, Osawa T, Uchida K. Covalent binding of oxidized cholesteryl esters to protein: implications for oxidative modification of low density lipoprotein and atherosclerosis. J Biol Chem 2003; 278:21040-9. [PMID: 12663661 DOI: 10.1074/jbc.m212426200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
It has been proposed that plasma low density lipoproteins (LDL) undergo oxidative modification before they can produce foam cells in atherosclerosis. The oxidation of LDL generates a variety of reactive aldehydic products, which covalently bind to the LDL apolipoprotein B-100 (apoB). In the present study, to investigate the mechanisms contributing to the modification of LDL, we analyzed oxidized cholesteryl esters generated during the autoxidation of LDL and characterized their covalent binding to the lysine residues of LDL apoB. In addition, we raised a monoclonal antibody specific to a lysine-bound oxidized cholesteryl ester and determined its production in human atherosclerotic lesions. The peroxidation of LDL with Cu2+ produced 9-oxononanoylcholesterol (9-ONC) and 5-oxovaleroylcholesterol as the major oxidized cholesteryl esters. We observed that the levels of 9-ONC and 5-oxovaleroylcholesterol peaked at 12 h and significantly decreased thereafter. The reduction of the core aldehyde levels was accompanied by (i) the formation of free 7-ketocholesterol and 7-ketocholesteryl ester core aldehydes and (ii) an increase in the amounts of apoB-bound cholesterol and 7-ketocholesterol, suggesting that the cholesteryl ester core aldehydes were further converted to their 7-ketocholesterol- and apoB-bound derivatives. To detect the protein-bound 9-ONC, we raised the monoclonal antibody 2A81, directed against 9-ONC-modified protein, and found that it extensively recognized protein-bound cholesteryl ester core aldehydes. Agarose gel electrophoresis followed by immunoblot analysis of the oxidized LDL clearly demonstrated the formation of antigenic structures. Furthermore, immunohistochemical analysis of the atherosclerotic lesions from the human aorta showed that immunoreactive materials with mAb 2A81 were indeed present in the lesions, in which the intense immunoreactivity was mainly located in the macrophage-derived foam cells and the thickening neointima of the arterial walls. The results of this study suggest that the binding of cholesteryl ester core aldehydes to LDL might represent the process common to the oxidative modification of lipoproteins.
Collapse
Affiliation(s)
- Yoshichika Kawai
- Laboratory of Food and Biodynamics, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Argmann CA, Sawyez CG, McNeil CJ, Hegele RA, Huff MW. Activation of peroxisome proliferator-activated receptor gamma and retinoid X receptor results in net depletion of cellular cholesteryl esters in macrophages exposed to oxidized lipoproteins. Arterioscler Thromb Vasc Biol 2003; 23:475-82. [PMID: 12615696 DOI: 10.1161/01.atv.0000058860.62870.6e] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Peroxisome proliferator-activated receptor gamma (PPARgamma), a ligand-activated transcription factor, has pleiotropic effects, including regulation of macrophage differentiation and lipid homeostasis. The PPARgamma ligands, thiazolidinediones (TZDs), attenuate atherosclerosis in mice by uncertain mechanisms. The objective of this study was to determine whether activation of PPARgamma or its obligate heterodimer, retinoid X receptor (RXR), modulates macrophage foam cell formation induced by oxidized (ox) lipoproteins. METHODS AND RESULTS Incubation of THP-1 macrophages with oxHTG-VLDL, oxREM, or oxLDL increased cellular cholesteryl ester over 6-fold. Preincubation with the TZD, ciglitazone, the RXR-specific ligand, 9-cis retinoic acid (9cRA) or the combination reduced CE mass accumulation by up to 65%. Ciglitazone and 9cRA increased CD36 mRNA (up to 4-fold); however, uptake of [125I]oxLDL was only modestly enhanced (up to 1.8-fold) becaues of a concomitant PPARgamma:RXR-induced decrease in SRAI/II activity (up to 40%). This suggested that PPARgamma:RXR activation inhibited cholesteryl ester accumulation by enhancing cholesterol efflux. Ciglitazone and 9cRA were found to increase the expression of ATP-binding cassette proteins A1 and G1, resulting in enhanced cholesterol efflux to lipoprotein-deficient serum, apoAI and HDL3. CONCLUSIONS PPARgamma and/or RXR activation inhibit foam cell formation through enhanced cholesterol efflux despite increased oxLDL uptake. These observations explain the reduced atherosclerosis in TZD-treated mice and may extend the therapeutic implications of these ligands.
Collapse
Affiliation(s)
- C A Argmann
- Department of Medicine and Robarts Research Institute, University of Western Ontario, London, Ontario, Canada N6A 5K8
| | | | | | | | | |
Collapse
|
47
|
Lysosomal cholesterol derived from mildly oxidized low density lipoprotein is resistant to efflux. J Lipid Res 2001. [DOI: 10.1016/s0022-2275(20)31654-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
48
|
Vandaveer SS, Erf GF, Durdik JM. Avian T helper one/two immune response balance can be shifted toward inflammation by antigen delivery to scavenger receptors. Poult Sci 2001; 80:172-81. [PMID: 11233005 DOI: 10.1093/ps/80.2.172] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Whether immune responses are dominated by inflammation or antibody production is often key to surviving infections. Therefore, differential control of these immune pathways determined by CD4 T cells is of fundamental interest for vaccine design. Little is known about how inflammatory [T helper cell (Th) type 1 (Th1)] versus antibody-inducing (Th2) choices are controlled in domestic fowl. To address this, MHC-matched chickens were immunized to test whether antibody-dominated Th2 or inflammatory Th1 responses could be preferentially activated, and our findings subsequently extended to outbred broiler breeders. Strategies used were known to shift the response in mice from Th2 to Th1 by delivering the injected antigen preferentially to macrophages. The model antigen, BSA, was maleylated to allow binding to scavenger receptors (SR) present on mammalian macrophages. Maleyl-BSA bound well in receptor-specific fashion to a chicken macrophage cell line. Compared with native BSA, immunization with SR-binding, maleyl-BSA modulated the immune response toward the Th1 pathway, as evident by increases in the magnitude of in vivo inflammatory reactions and declines in antibody-making responses. Initiation of a maleyl-BSA Th1 pathway is further supported by the enhanced ability of splenocytes to express mRNA for interferon-gamma in response to antigens. Together, these data establish the presence and functional relevance of SR in domestic fowl as well as provide a system for investigating the mechanisms controlling Th1/Th2 pathways in chickens. Moreover, the ability to direct immune responses toward either pathway by antigen maleylation will contribute significantly to the development of better vaccines for poultry diseases.
Collapse
Affiliation(s)
- S S Vandaveer
- Department of Biological Sciences, University of Arkansas, Fayetteville 72701, USA
| | | | | |
Collapse
|
49
|
Intracellular trafficking of pigeon β-very low density lipoprotein and low density lipoprotein at low and high concentrations in pigeon macrophages. J Lipid Res 2000. [DOI: 10.1016/s0022-2275(20)31976-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
50
|
Abstract
Oxidation products of lipids and proteins are found in atherosclerotic plaque and in macrophage foam cells. Macrophages avidly endocytose in-vitro oxidized LDL and accumulate sterols. What is the evidence that such a process is involved in in-vivo foam cell formation? The present review surveys current knowledge on the metabolism of oxidized LDL by macrophages, and the types, amounts and location of oxidation products that accumulate in these cells. Comparable studies of lesion lipoproteins and foam cells indicate that limited extracellular lipoprotein oxidation, perhaps followed by more extensive intracellular oxidation subsequent to uptake by macrophages, is a more likely scenario in vivo.
Collapse
Affiliation(s)
- W Jessup
- Cell Biology Group, Heart Research Institute, Sydney, New South Wales, Australia.
| | | |
Collapse
|