1
|
Peng X, Lian Z, O'Brien V, Xiao J, Litchfield BA, Dai Perrard XY, Xu L, Ni J, Mukherjee A, Simmons T, Dong H, Mullick AE, Crooke R, Pownall HJ, Simon SI, Ballantyne CM, Wu H. Foamy monocytes and atherogenesis in mice with combined hyperlipidemia and effects of antisense knockdown of apoCIII. J Lipid Res 2025:100763. [PMID: 39988193 DOI: 10.1016/j.jlr.2025.100763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/06/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025] Open
Abstract
Hypertriglyceridemia (HTG), particularly in combined hyperlipidemia, increases risk for atherosclerotic cardiovascular disease, but the underlying mechanisms remain incompletely understood. We sought to determine contributions of circulating monocytes to atherosclerosis associated with HTG in combined hyperlipidemia, created by transgenic expression of human apoCIII in Ldlr-/- mice (Ldlr-/-ApoCIIItg) fed western high-fat diet (WD). Tissue culture with THP1 and primary human monocytes was used to examine effects of triglyceride (TG)-rich lipoproteins (TGRL) on monocytes. Ldlr-/-ApoCIIItg mice were also treated with apoCIII antisense oligonucleotide (ASO) and examined for foamy monocytes and atherosclerosis. Compared to Ldlr-/- mice, Ldlr-/-ApoCIIItg mice fed WD had early and persistent increases in lipid accumulation within monocytes and enhanced atherosclerosis. Ldlr-/-ApoCIIItg mice vs Ldlr-/- mice had higher levels of CD11c, CD36, and cytokines in foamy monocytes, with increases in foamy monocyte adhesion to VCAM-1 and oxLDL uptake. Monocytes took up TGRL in vivo and in vitro and changed phenotypes. Foamy monocytes infiltrated into atherosclerotic lesions, and specific and sustained depletion of CD11c+ (foamy) monocytes profoundly reduced atherosclerosis in Ldlr-/-ApoCIIItg mice on WD. Treatment with apoCIII ASO lowered plasma TG and cholesterol levels, improved foamy monocyte phenotypes, and reduced atherosclerosis in Ldlr-/-ApoCIIItg mice. In conclusion, HTG in combined hyperlipidemia accelerates atherosclerosis, in part, by increasing foamy monocyte formation and infiltration into atherosclerotic plaques. Treatment with apoCIII ASO is a potential new therapy for improving monocyte phenotypes and reducing atherosclerosis in combined hyperlipidemia.
Collapse
Affiliation(s)
- Xueying Peng
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, P.R. China; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Zeqin Lian
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Veronica O'Brien
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jing Xiao
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Lu Xu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jing Ni
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Aparna Mukherjee
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Timothy Simmons
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Henry Dong
- Department of Pediatrics, Children's Hospital of Pittsburgh UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | - Henry J Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, TX, USA
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
van Tuijl J, van Heck JI, Bahrar H, Broeders W, Wijma J, ten Have YM, Giera M, Zweers-van Essen H, Rodwell L, Joosten LA, Netea MG, Afman LA, Bekkering S, Riksen NP. Single high-fat challenge and trained innate immunity: A randomized controlled cross-over trial. iScience 2024; 27:111103. [PMID: 39493874 PMCID: PMC11530819 DOI: 10.1016/j.isci.2024.111103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2024] [Revised: 05/11/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Brief exposure of monocytes to atherogenic molecules, such as oxidized lipoproteins, triggers a persistent pro-inflammatory phenotype, named trained immunity. In mice, transient high-fat diet leads to trained immunity, which aggravates atherogenesis. We hypothesized that a single high-fat challenge in humans induces trained immunity. In a randomized controlled cross-over study, 14 healthy individuals received a high-fat or reference shake, and blood was drawn before and after 1, 2, 4, 6, 24, and 72 h. Incubation of donor monocytes with the post-high-fat-shake serum induced trained immunity, regulated via Toll-like receptor 4. This was not mediated via triglyceride-rich lipoproteins, C12, 14, and 16, or metabolic endotoxemia. In vivo, however, the high-fat challenge did not affect monocyte phenotype and function. We conclude that a high-fat challenge leads to alterations in the serum composition that have the potential to induce trained immunity in vitro. However, this does not translate into a (persistent) hyperinflammatory monocyte phenotype in vivo.
Collapse
Affiliation(s)
- Julia van Tuijl
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Julia I.P. van Heck
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Wieteke Broeders
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Johan Wijma
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Yvonne M. ten Have
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Heidi Zweers-van Essen
- Department of Gastroenterology and Hepatology-Dietetics, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Laura Rodwell
- Section Biostatics, Department of Health Evidence, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Lydia A. Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University and Research, Wageningen 6700 HB, the Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Niels P. Riksen
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| |
Collapse
|
3
|
Chauhan N, Koli M, Ghosh R, Majumdar AG, Ghosh A, Ghanty TK, Mula S, Patro BS. A BODIPY-Naphtholimine-BF 2 Dyad for Precision Photodynamic Therapy, Targeting, and Dual Imaging of Endoplasmic Reticulum and Lipid Droplets in Cancer. JACS AU 2024; 4:2838-2852. [PMID: 39211629 PMCID: PMC11350743 DOI: 10.1021/jacsau.3c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 09/13/2023] [Revised: 05/11/2024] [Accepted: 05/22/2024] [Indexed: 09/04/2024]
Abstract
Currently, effective therapeutic modalities for pancreatic ductal adenocarcinoma (PDAC) are quite limited, leading to gloomy prognosis and ∼6-months median patient survival. Recent advances showed the promise of photodynamic therapy (PDT) for PDAC patients. Next generation photosensitizers (PS) are based on "organelle-targeted-PDT" and provide new paradigm in the field of precision medicines to address the current challenge for treating PDAC. In this investigation, we have constructed a novel PS, named as N b B, for precise and simultaneous targeting of endoplasmic reticulum (ER) and lipid droplets (LDs) in PDAC, based on the fact that malignant PDAC cells are heavily relying on ER for hormone synthesis. Our live cell imaging and fluorescence recovery after photobleaching (FRAP) experiments revealed that N b B is quickly targeted to ER and subsequently to LDs and shows simultaneous dual fluorescence color due to polar and nonpolar milieu of ER and LDs. Interestingly, the same molecule generates triplet state and singlet oxygen efficiently and causes robust ER stress and cellular lipid peroxidation, leading to apoptosis in two different PDAC cells in the presence of light. Together, we present, for the first time, a potential next generation precision medicine for ER-LD organelle specific imaging and PDT of pancreatic cancer.
Collapse
Affiliation(s)
- Nitish Chauhan
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Mrunesh Koli
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Rajib Ghosh
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Ananda Guha Majumdar
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Ayan Ghosh
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
| | - Tapan K. Ghanty
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Soumyaditya Mula
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Birija Sankar Patro
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| |
Collapse
|
4
|
Bussi C, Lai R, Athanasiadi N, Gutierrez MG. Physiologic medium renders human iPSC-derived macrophages permissive for M. tuberculosis by rewiring organelle function and metabolism. mBio 2024; 15:e0035324. [PMID: 38984828 PMCID: PMC11323749 DOI: 10.1128/mbio.00353-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/05/2024] [Accepted: 06/04/2024] [Indexed: 07/11/2024] Open
Abstract
In vitro studies are crucial for our understanding of the human macrophage immune functions. However, traditional in vitro culture media poorly reflect the metabolic composition of blood, potentially affecting the outcomes of these studies. Here, we analyzed the impact of a physiological medium on human induced pluripotent stem cell (iPSC)-derived macrophages (iPSDM) function. Macrophages cultured in a human plasma-like medium (HPLM) were more permissive to Mycobacterium tuberculosis (Mtb) replication and showed decreased lipid metabolism with increased metabolic polarization. Functionally, we discovered that HPLM-differentiated macrophages showed different metabolic organelle content and activity. Specifically, HPLM-differentiated macrophages displayed reduced lipid droplet and peroxisome content, increased lysosomal proteolytic activity, and increased mitochondrial activity and dynamics. Inhibiting or inducing lipid droplet formation revealed that lipid droplet content is a key factor influencing macrophage permissiveness to Mtb. These findings underscore the importance of using physiologically relevant media in vitro for accurately studying human macrophage function. IMPORTANCE This work compellingly demonstrates that the choice of culture medium significantly influences M. tuberculosis replication outcomes, thus emphasizing the importance of employing physiologically relevant media for accurate in vitro host-pathogen interaction studies. We anticipate that our work will set a precedent for future research with clinical relevance, particularly in evaluating antibiotic efficacy and resistance in cellulo.
Collapse
Affiliation(s)
- Claudio Bussi
- The Francis Crick Institute, London, United Kingdom
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Rachel Lai
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
5
|
Radushev V, Karkossa I, Berg J, von Bergen M, Engelmann B, Rolle-Kampczyk U, Blüher M, Wagner U, Schubert K, Rossol M. Dysregulated cytokine and oxidative response in hyper-glycolytic monocytes in obesity. Front Immunol 2024; 15:1416543. [PMID: 39050851 PMCID: PMC11266186 DOI: 10.3389/fimmu.2024.1416543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/12/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Obesity is associated with a plethora of health complications, including increased susceptibility to infections or decreased vaccine efficacy, partly due to dysregulated immune responses. Monocytes play a crucial role in innate immunity, yet their functional alterations in obesity remain poorly understood. Methods Here, we employed proteomic and metabolomic analyses to investigate monocyte characteristics in individuals with overweight, obesity, impaired glucose tolerance (IGT), and type 2 diabetes (T2D), compared to lean donors. Results and discussion Our results revealed distinct molecular signatures in monocytes from individuals with obesity, with significant alterations in pathways related to metabolism, cellular migration, and phagocytosis. Moreover, LPS-induced activation of monocytes unveiled heightened metabolic reprogramming towards glycolysis in subjects with obesity accompanied by dysregulated cytokine responses and elevated oxidative stress. Additionally, monocytes from donors with obesity exhibited increased lipid droplet accumulation. These findings shed light on the immunometabolic dysregulation underlying obesity-associated immune dysfunction, highlighting potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Veselina Radushev
- Division of Rheumatology, Department of Endocrinology, Nephrology, Rheumatology, Leipzig University, Leipzig, Germany
| | - Isabel Karkossa
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH, Leipzig, Germany
| | - Janina Berg
- Molecular Immunology, Faculty of Health Sciences, BTU Cottbus-Senftenberg, Senftenberg, Germany
| | - Martin von Bergen
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH, Leipzig, Germany
- Institute for Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Beatrice Engelmann
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH, Leipzig, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Ulf Wagner
- Division of Rheumatology, Department of Endocrinology, Nephrology, Rheumatology, Leipzig University, Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH, Leipzig, Germany
| | - Manuela Rossol
- Division of Rheumatology, Department of Endocrinology, Nephrology, Rheumatology, Leipzig University, Leipzig, Germany
- Molecular Immunology, Faculty of Health Sciences, BTU Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Environment and Natural Sciences, BTU Cottbus-Senftenberg, Senftenberg, Germany
| |
Collapse
|
6
|
Cervantes J, Kanter JE. Monocyte and macrophage foam cells in diabetes-accelerated atherosclerosis. Front Cardiovasc Med 2023; 10:1213177. [PMID: 37378396 PMCID: PMC10291141 DOI: 10.3389/fcvm.2023.1213177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/27/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetes results in an increased risk of atherosclerotic cardiovascular disease. This minireview will discuss whether monocyte and macrophage lipid loading contribute to this increased risk, as monocytes and macrophages are critically involved in the progression of atherosclerosis. Both uptake and efflux pathways have been described as being altered by diabetes or conditions associated with diabetes, which may contribute to the increased accumulation of lipids seen in macrophages in diabetes. More recently, monocytes have also been described as lipid-laden in response to elevated lipids, including triglyceride-rich lipoproteins, the class of lipids often elevated in the setting of diabetes.
Collapse
Affiliation(s)
| | - Jenny E. Kanter
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
7
|
Lian Z, Perrard XYD, Antony AK, Peng X, Xu L, Ni J, Zhang B, O’Brien V, Saeed A, Jia X, Hussain A, Yu B, Simon SI, Sacks FM, Hoogeveen RC, Ballantyne CM, Wu H. Dietary Effects on Monocyte Phenotypes in Subjects With Hypertriglyceridemia and Metabolic Syndrome. JACC Basic Transl Sci 2023; 8:460-475. [PMID: 37325398 PMCID: PMC10264566 DOI: 10.1016/j.jacbts.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/13/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 06/17/2023]
Abstract
In patients with hypertriglyceridemia, a short-term low-saturated fat vs high-saturated fat diet induced lower plasma lipids and improved monocyte phenotypes. These findings highlight the role of diet fat content and composition for monocyte phenotypes and possibly cardiovascular disease risk in these patients. (Effects of Dietary Interventions on Monocytes in Metabolic Syndrome; NCT03591588).
Collapse
Affiliation(s)
- Zeqin Lian
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | | | | | - Xueying Peng
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Lu Xu
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jing Ni
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Bingqian Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Veronica O’Brien
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Anum Saeed
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Xiaoming Jia
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, Texas, USA
| | - Aliza Hussain
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, Texas, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, USA
| | - Scott I. Simon
- Department of Biomedical Engineering, University of California, Davis, California, USA
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, and Department of Medicine, Harvard Medical School and Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Ron C. Hoogeveen
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Christie M. Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, Texas, USA
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
8
|
Liu Y, Zhang X, Cheng X, Luo Q, Yu M, Long K, Qu W, Tang Y, Gong M, Liang L, Ke X, Song Y. Characterization of fatty acid metabolism-related lncRNAs in lung adenocarcinoma identifying potential novel prognostic targets. Front Genet 2022; 13:990153. [PMID: 36299578 PMCID: PMC9589892 DOI: 10.3389/fgene.2022.990153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/09/2022] [Accepted: 09/01/2022] [Indexed: 11/15/2022] Open
Abstract
Lung adenocarcinoma (LUAD), a malignant respiratory tumor with an extremely poor prognosis, has troubled the medical community all over the world. According to recent studies, fatty acid metabolism (FAM) and long non-coding RNAs (lncRNAs) regulation have shown exciting results in tumor therapy. In this study, the original LUAD patient data was obtained from the TCGA database, and 12 FAM-related lncRNAs (AL390755.1, AC105020.6, TMPO-AS1, AC016737.2, AC127070.2, LINC01281, AL589986.2, GAS6-DT, AC078993.1, LINC02198, AC007032.1, and AL021026.1) that were highly related to the progression of LUAD were finally identified through bioinformatics analysis, and a risk score model for clinical reference was constructed. The window explores the immunology and molecular mechanism of LUAD, aiming to shed the hoping light on LUAD treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xixian Ke
- *Correspondence: Xixian Ke, ; Yongxiang Song,
| | | |
Collapse
|
9
|
Li Y, Yao R, Ren M, Yuan K, Du Y, He Y, Kang H, Yuan S, Ju W, Qiao J, Xu K, Zeng L. Liposomes trigger bone marrow niche macrophage "foam" cell formation and affect hematopoiesis in mice. J Lipid Res 2022; 63:100273. [PMID: 36084713 PMCID: PMC9587404 DOI: 10.1016/j.jlr.2022.100273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/12/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022] Open
Abstract
Liposomes are the most widely used nanocarrier platform for the delivery of therapeutic and diagnostic agents, and a number of liposomes have been approved for use in clinical practice. After systemic administration, most liposomes are cleared by macrophages in the mononuclear phagocyte system, such as the liver and bone marrow (BM). However, the majority of studies have focused on investigating the therapeutic results of liposomal drugs, and too few studies have evaluated the potential side effects of empty nanocarriers on the functions of macrophages in the mononuclear phagocyte system. Here, we evaluate the potential effects of empty liposomes on the functions of BM niche macrophages. Following liposome administration, we observed lipid droplet (LD) accumulation in cultured primary macrophages and BM niche macrophages. We found that these LD-accumulating macrophages, similar to foam cells, exhibited increased expression of inflammatory cytokines, such as IL-1β and IL-6. We further provided evidence that liposome deposition and degradation induced LD biogenesis on the endoplasmic reticulum membrane and subsequently disturbed endoplasmic reticulum homeostasis and activated the inositol-requiring transmembrane kinase/endoribonuclease 1α/NF-κB signaling pathway, which is responsible for the inflammatory activation of macrophages after liposome engulfment. Finally, we also showed the side effects of dysfunctional BM niche macrophages on hematopoiesis in mice, such as the promotion of myeloid-biased output and impairment of erythropoiesis. This study not only draws attention to the safety of liposomal drugs in clinical practice but also provides new directions for the design of lipid-based drug carriers in preclinical studies.
Collapse
Affiliation(s)
- Yue Li
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ran Yao
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Miao Ren
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ke Yuan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuwei Du
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuan He
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Haiquan Kang
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shengnan Yuan
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
10
|
Poloxamer 407 Induces Hypertriglyceridemia but Decreases Atherosclerosis in Ldlr -/- Mice. Cells 2022; 11:cells11111795. [PMID: 35681489 PMCID: PMC9179832 DOI: 10.3390/cells11111795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/19/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
Background: Hypertriglyceridemia (HTG) increases the risk for atherosclerotic cardiovascular disease, but underlying mechanisms are incompletely understood. Circulating monocytes play an important role in atherogenesis by infiltrating arterial walls, where they differentiate into macrophages. We tested the hypothesis that HTG is mechanistically linked to atherogenesis by altering the monocyte phenotype and infiltration into atherosclerotic lesions in a model of diet-induced atherogenesis in Ldlr−/− mice. Methods: HTG was induced in male Ldlr−/− mice, fed a Western, high-fat high-cholesterol diet, by daily injection of poloxamer 407 (P407), a lipoprotein lipase inhibitor, for seven weeks. Atherosclerosis, monocyte phenotypes, and monocyte migration into atherosclerotic lesions were determined by well-validated methods. Results: Compared with the saline control, P407 injection in Ldlr−/− mice rapidly induced profound and persistent HTG, modestly elevated plasma cholesterol levels, and increased levels of triglyceride and cholesterol carried in very-low-density lipoprotein and low-density lipoprotein. Unexpectedly, mice receiving P407 versus saline control showed less atherosclerosis. Following induction of HTG by P407, CD36+ (also CD11c+), but not CD36− (CD11c−), monocytes showed early increases in lipid accumulation, but the number of CD36+ (not CD36−) monocytes was dramatically decreased afterwards in the circulation until the end of the test. Concurrently, CD36+ (CD11c+) monocyte migration into atherosclerotic lesions was also reduced in mice receiving P407 versus controls. Conclusions: P407 induced severe HTG, but reduced atherosclerosis, in Ldlr−/− mice, possibly because of profound reductions of circulating CD36+ (CD11c+) monocytes, leading to decreased monocyte migration into atherosclerotic lesions.
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Recent studies indicate an association between hypertriglyceridemia (HTG) and atherosclerotic cardiovascular disease (ASCVD). The purpose of this review is to discuss the potential mechanism connecting HTG and ASCVD risk and the potential efficacy of HTG-targeting therapies in ASCVD prevention. RECENT FINDINGS HTG, with elevations in triglyceride-rich lipoproteins (TGRL) and their remnants, are causal ASCVD risk factors. The mechanisms whereby HTG increases ASCVD risk are not well understood but may include multiple factors. Inflammation plays a crucial role in atherosclerosis. TGRL compared to low-density lipoproteins (LDL) correlate better with inflammation. TGRL remnants can penetrate endothelium and interact with macrophages leading to foam cell formation and inflammation in arterial walls, thereby contributing to atherogenesis. In addition, circulating monocytes can take up TGRL and become lipid-laden foamy monocytes, which infiltrate the arterial wall and may also contribute to atherogenesis. Novel therapies targeting HTG or inflammation are in development and have potential of reducing residual ASCVD risk associated with HTG. Clinical and preclinical studies show a causal role of HTG in promoting ASCVD, in which inflammation plays a vital role. Novel therapies targeting HTG or inflammation have potential of reducing residual ASCVD risk.
Collapse
Affiliation(s)
- Xueying Peng
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China.
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
12
|
Lee DG, Kim HJ, Lee Y, Kim JH, Hwang Y, Ha J, Ryoo S. 10-DEBC Hydrochloride as a Promising New Agent against Infection of Mycobacterium abscessus. Int J Mol Sci 2022; 23:591. [PMID: 35054777 PMCID: PMC8775589 DOI: 10.3390/ijms23020591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/09/2021] [Revised: 01/03/2022] [Accepted: 01/03/2022] [Indexed: 11/17/2022] Open
Abstract
Mycobacterium abscessus (M. abscessus) causes chronic pulmonary infections. Its resistance to current antimicrobial drugs makes it the most difficult non-tuberculous mycobacteria (NTM) to treat with a treatment success rate of 45.6%. Therefore, there is a need for new therapeutic agents against M. abscessus. We identified 10-DEBC hydrochloride (10-DEBC), a selective AKT inhibitor that exhibits inhibitory activity against M. abscessus. To evaluate the potential of 10-DEBC as a treatment for lung disease caused by M. abscessus, we measured its effectiveness in vitro. We established the intracellular activity of 10-DEBC against M. abscessus in human macrophages and human embryonic cell-derived macrophages (iMACs). 10-DEBC significantly inhibited the growth of wild-type M. abscessus and clinical isolates and clarithromycin (CLR)-resistant M. abscessus strains. 10-DEBC's drug efficacy did not have cytotoxicity in the infected macrophages. In addition, 10-DEBC operates under anaerobic conditions without replication as well as in the presence of biofilms. The alternative caseum binding assay is a unique tool for evaluating drug efficacy against slow and nonreplicating bacilli in their native caseum media. In the surrogate caseum, the mean undiluted fraction unbound (fu) for 10-DEBC is 5.696. The results of an in vitro study on the activity of M. abscessus suggest that 10-DEBC is a potential new drug for treating M. abscessus infections.
Collapse
Affiliation(s)
- Da-Gyum Lee
- Center for Clinical Research, Masan National Tuberculosis Hospital, Changwon 51755, Korea; (D.-G.L.); (Y.H.)
| | - Hye-Jung Kim
- New Drug Development Center, KBIO OSONG Medical Innovation Foundation, Cheongju 28160, Korea; (H.-J.K.); (J.H.)
| | - Youngsun Lee
- Division of Intractable Diseases Research, Department of Chronic Diseases Convergence Research, Korea National Institute of Health, Cheongju 28160, Korea; (Y.L.); (J.-H.K.)
| | - Jung-Hyun Kim
- Division of Intractable Diseases Research, Department of Chronic Diseases Convergence Research, Korea National Institute of Health, Cheongju 28160, Korea; (Y.L.); (J.-H.K.)
| | - Yoohyun Hwang
- Center for Clinical Research, Masan National Tuberculosis Hospital, Changwon 51755, Korea; (D.-G.L.); (Y.H.)
| | - Jeongyeop Ha
- New Drug Development Center, KBIO OSONG Medical Innovation Foundation, Cheongju 28160, Korea; (H.-J.K.); (J.H.)
| | - Sungweon Ryoo
- Center for Clinical Research, Masan National Tuberculosis Hospital, Changwon 51755, Korea; (D.-G.L.); (Y.H.)
| |
Collapse
|
13
|
Raman Microscopy Techniques to Study Lipid Droplet Composition in Cancer Cells. Methods Mol Biol 2022; 2413:193-209. [PMID: 35044667 PMCID: PMC9939018 DOI: 10.1007/978-1-0716-1896-7_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/19/2022]
Abstract
Raman spectroscopy using feature selection schemes has considerable advantages over gas chromatography for the analysis of fatty acids' composition changes. Here, we introduce an educational methodology to demonstrate the potential of micro-Raman spectroscopy to determine with high accuracy the unsaturation or saturation degrees and composition changes of the fatty acids found in the lipid droplets of the LNCaP prostate cancer cells that were treated with various fatty acids. The methodology uses highly discriminatory wavenumbers among fatty acids present in the sample selected by using the Support Vector Machine algorithm.
Collapse
|
14
|
Lipid Droplets, Phospholipase A 2, Arachidonic Acid, and Atherosclerosis. Biomedicines 2021; 9:biomedicines9121891. [PMID: 34944707 PMCID: PMC8699036 DOI: 10.3390/biomedicines9121891] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/10/2021] [Revised: 12/01/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
Lipid droplets, classically regarded as static storage organelles, are currently considered as dynamic structures involved in key processes of lipid metabolism, cellular homeostasis and signaling. Studies on the inflammatory state of atherosclerotic plaques suggest that circulating monocytes interact with products released by endothelial cells and may acquire a foamy phenotype before crossing the endothelial barrier and differentiating into macrophages. One such compound released in significant amounts into the bloodstream is arachidonic acid, the common precursor of eicosanoids, and a potent inducer of neutral lipid synthesis and lipid droplet formation in circulating monocytes. Members of the family of phospholipase A2, which hydrolyze the fatty acid present at the sn-2 position of phospholipids, have recently emerged as key controllers of lipid droplet homeostasis, regulating their formation and the availability of fatty acids for lipid mediator production. In this paper we discuss recent findings related to lipid droplet dynamics in immune cells and the ways these organelles are involved in regulating arachidonic acid availability and metabolism in the context of atherosclerosis.
Collapse
|
15
|
Chen X, Wu T, Gong Z, Guo J, Liu X, Zhang Y, Li Y, Ferraro P, Li B. Lipid droplets as endogenous intracellular microlenses. LIGHT, SCIENCE & APPLICATIONS 2021; 10:242. [PMID: 34873142 PMCID: PMC8648767 DOI: 10.1038/s41377-021-00687-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/27/2021] [Revised: 10/31/2021] [Accepted: 11/23/2021] [Indexed: 05/24/2023]
Abstract
Using a single biological element as a photonic component with well-defined features has become a new intriguing paradigm in biophotonics. Here we show that endogenous lipid droplets in the mature adipose cells can behave as fully biocompatible microlenses to strengthen the ability of microscopic imaging as well as detecting intra- and extracellular signals. By the assistance of biolenses made of the lipid droplets, enhanced fluorescence imaging of cytoskeleton, lysosomes, and adenoviruses has been achieved. At the same time, we demonstrated that the required excitation power can be reduced by up to 73%. The lipidic microlenses are finely manipulated by optical tweezers in order to address targets and perform their real-time imaging inside the cells. An efficient detecting of fluorescence signal of cancer cells in extracellular fluid was accomplished due to the focusing effect of incident light by the lipid droplets. The lipid droplets acting as endogenous intracellular microlenses open the intriguing route for a multifunctional biocompatible optics tool for biosensing, endoscopic imaging, and single-cell diagnosis.
Collapse
Affiliation(s)
- Xixi Chen
- Institute of Nanophotonics, Jinan University, 511443, Guangzhou, China
| | - Tianli Wu
- Institute of Nanophotonics, Jinan University, 511443, Guangzhou, China
| | - Zhiyong Gong
- Institute of Nanophotonics, Jinan University, 511443, Guangzhou, China
| | - Jinghui Guo
- Department of Physiology, School of Medicine, Jinan University, 510632, Guangzhou, China
| | - Xiaoshuai Liu
- Institute of Nanophotonics, Jinan University, 511443, Guangzhou, China
| | - Yao Zhang
- Institute of Nanophotonics, Jinan University, 511443, Guangzhou, China.
| | - Yuchao Li
- Institute of Nanophotonics, Jinan University, 511443, Guangzhou, China.
| | - Pietro Ferraro
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems «E. Caianiello», Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy.
| | - Baojun Li
- Institute of Nanophotonics, Jinan University, 511443, Guangzhou, China.
| |
Collapse
|
16
|
Potcoava MC, Futia GL, Gibson EA, Schlaepfer IR. Lipid profiling using Raman and a modified support vector machine algorithm. JOURNAL OF RAMAN SPECTROSCOPY : JRS 2021; 52:1910-1922. [PMID: 35814195 PMCID: PMC9269992 DOI: 10.1002/jrs.6238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 11/14/2020] [Accepted: 08/03/2021] [Indexed: 06/15/2023]
Abstract
Lipid droplets are dynamic organelles that play important cellular roles. They are composed of a phospholipid membrane and a core of triglycerides and sterol esters. Fatty acids have important roles in phospholipid membrane formation, signaling, and synthesis of triglycerides as energy storage. Better non-invasive tools for profiling and measuring cellular lipids are needed. Here we demonstrate the potential of Raman spectroscopy to determine with high accuracy the composition changes of the fatty acids and cholesterol found in the lipid droplets of prostate cancer cells treated with various fatty acids. The methodology uses a modified least squares fitting (LSF) routine that uses highly discriminatory wavenumbers between the fatty acids present in the sample using a support vector machine algorithm. Using this new LSF routine, Raman micro-spectroscopy can become a better non-invasive tool for profiling and measuring fatty acids and cholesterol for cancer biology.
Collapse
Affiliation(s)
- Mariana C. Potcoava
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Gregory L. Futia
- Department of Bioengineering, University of Colorado Denver, Aurora, Colorado, USA
| | - Emily A. Gibson
- Department of Bioengineering, University of Colorado Denver, Aurora, Colorado, USA
| | - Isabel R. Schlaepfer
- Division of Medical Oncology, School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Lipid-mediated atherogenesis is hallmarked by a chronic inflammatory state. Low-density lipoprotein cholesterol (LDL-C), triglyceride rich lipoproteins (TRLs), and lipoprotein(a) [Lp(a)] are causally related to atherosclerosis. Within the paradigm of endothelial activation and subendothelial lipid deposition, these lipoproteins induce numerous pro-inflammatory pathways. In this review, we will outline the effects of lipoproteins on systemic inflammatory pathways in atherosclerosis. RECENT FINDINGS Apolipoprotein B-containing lipoproteins exert a variety of pro-inflammatory effects, ranging from the local artery to systemic immune cell activation. LDL-C, TRLs, and Lp(a) induce endothelial dysfunction with concomitant activation of circulating monocytes through enhanced lipid accumulation. The process of trained immunity of the innate immune system, predominantly induced by LDL-C particles, hallmarks the propagation of the low-grade inflammatory response. In concert, bone marrow activation induces myeloid skewing, further contributing to immune cell mobilization and plaque progression. SUMMARY Lipoproteins and inflammation are intertwined in atherogenesis. Elucidating the inflammatory pathways will provide new opportunities for therapeutic agents.
Collapse
Affiliation(s)
- Jordan M. Kraaijenhof
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam
| | - G. Kees Hovingh
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam
| | - Jeffrey Kroon
- Amsterdam UMC, University of Amsterdam, Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Fiorelli S, Anesi A, Porro B, Cosentino N, Werba JP, Di Minno A, Manega CM, Barbieri S, Colombo GI, Marenzi G, Cavalca V, Tremoli E, Eligini S. Lipidomics analysis of monocytes from patients with acute myocardial infarction reveals lactosylceramide as a new player in monocyte migration. FASEB J 2021; 35:e21494. [PMID: 33856696 DOI: 10.1096/fj.202001872rrr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/05/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 11/11/2022]
Abstract
Monocyte recruitment after vascular injury and their migration through the vessel wall represent crucial events in the initiation, progression, and destabilization of atherosclerotic plaque. Circulating monocytes are exposed to stimuli that alter their physiological state, and among them, lipids play a key role. Several studies investigated the mechanisms by which lipids affect monocyte functions promoting coronary atherosclerotic plaque initiation, but information on the relationship between lipid composition and function of monocyte is scant. We aimed at studying the migration of circulating monocytes isolated from patients with acute myocardial infarction (AMI) at hospital presentation and investigating its correlation with cellular lipid profile. The migration of monocytes was tested using both fetal bovine serum (FBS) and autologous serum as chemoattractant stimuli. Monocyte lipid profile was evaluated through an untargeted lipidomics approach, using a liquid chromatography/time-of-flight mass spectrometry platform. We observed that AMI patients' monocytes showed a significant increase in FBS and autologous serum-mediated migration compared to controls. Moreover, a different monocyte lipidomic profile between the two study groups was detected. In particular, AMI patients' monocytes showed an altered composition in ceramides, with an increase in lactosylceramide and in phospholipids (ie, phosphatidylethanolamine and lisophosphatidylethanolamine). Of note, a positive correlation between lactosylceramide levels and monocyte migration was observed. Furthermore, the lactosylceramide synthase inhibition significantly reduced FBS-induced monocyte migration. Our results highlight the influence of lactosylceramide on the monocyte migration capacity, pointing out a new possible mechanism of lipids in the onset of atherothrombosis and, hence, in AMI.
Collapse
Affiliation(s)
| | - Andrea Anesi
- Centro Cardiologico Monzino I.R.C.C.S, Milan, Italy
| | | | | | - José P Werba
- Centro Cardiologico Monzino I.R.C.C.S, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Singh A, Sen P. Lipid droplet: A functionally active organelle in monocyte to macrophage differentiation and its inflammatory properties. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158981. [PMID: 34119681 DOI: 10.1016/j.bbalip.2021.158981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2021] [Revised: 05/06/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022]
Abstract
Lipid droplets (LDs) perform several important functions like inflammatory responses, membrane trafficking, acts as secondary messengers, etc. rather than simply working as an energy reservoir. LDs have been implicated as a controlling factor in the progression of atherosclerosis followed by foam cell formation that derives from macrophages during the differentiation process. However, the role of LDs in monocyte differentiation or its further immunological function is still an area that mandates in-depth investigation. We report that LD dynamics is important for differentiation of monocytes and is absolutely required for sustained and prolonged functional activity of differentiated macrophages. In THP-1 cell line model system, we elucidated that increase in total LD content in monocyte by external lipid supplements, can induce monocyte differentiation independent of classical stimuli, PMA. Differential expression of PLIN2 and ATGL during the event, together with abrogation of de novo lipogenesis further confirmed the fact. Besides, an increase in LD content by free fatty acid supplement was able to exert a synergistic effect with PMA on differentiation and phagocytic activity compared to when they are used alone. Additionally, we have shown Rab5a to play a vital role in LDs biosynthesis/maturation in monocytes and thereby directly affecting differentiation of monocytes into macrophages via AKT pathway. Thus our study reveals the multi-faceted function of LDs during the process of monocyte to macrophage differentiation and thereby helping to maintain the functional activity.
Collapse
Affiliation(s)
- Arpana Singh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Prosenjit Sen
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India.
| |
Collapse
|
20
|
Moore C, Harvey A, Coleman JN, Byrne HJ, McIntyre J. Label-free screening of biochemical changes in macrophage-like cells following MoS 2 exposure using Raman micro-spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 246:118916. [PMID: 33032120 DOI: 10.1016/j.saa.2020.118916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 05/14/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 06/11/2023]
Abstract
The emergence of large scale production techniques for 2D particulate materials has dramatically increased their applications potential. Understanding the interactions of biological cells with such particulate material is therefore of paramount importance, both for toxicological assessment and potential biomedical applications. Conventional in-vitro cytological assays commonly record only a single colorimetric end-point, and do not provide an in-depth analysis of how such materials are uptaken and processed within cells. To demonstrate its potential as an alternative, label free approach, confocal Raman micro-spectroscopy has been used to profile the cellular response of macrophage-like immune cells as a result of exposure to a sub-lethal dose of particulate MoS2, as an example novel 2D material. Particles were seen to be uptaken and trafficked in sub-cellular vesicles, and this sensitive technique allows differences in the biochemical composition of the vesicles to be assessed and monitored as a function of time. Untreated macrophage-like cells contain lipidic vesicles which are found to be relatively rich in the membrane lipid sphingomyelin, key to the process of cell membrane regeneration. After exposure to MoS2, the particulate material is seen to be invaginated in similar vesicles, the most prominent of which now, however, have spectroscopic signatures which are dominated by those of phosphatidyl family lipids, consistent with the phagocytotic pathway. The lipidic content of cells is seen to increase at all time-points (4, 24 and 72 h). although vesicles composed of sphingomyelin become more prominent again following a prolonged incubation of 72 h to a sub-lethal dose of MoS2, as the immune cell has processed the particulate material and initiates recovery to a normal/untreated state. This study reveals Raman micro-spectroscopy is an effective method for monitoring cellular responses and evolution of organelle compositions in response to MoS2 exposure. The additional benefit of using this technique is that cells can be monitored as a function of time, while it can also be used for screening other micro/nano materials for toxicology and/or establishing cell responses.
Collapse
Affiliation(s)
- Caroline Moore
- FOCAS Research Institute, Technological University Dublin, City Centre Campus, Dublin 8, Ireland.
| | - Andrew Harvey
- Centre for Research on Adaptive Nanostructures & Nanodevices (CRANN) and Advanced Materials and BioEngineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| | - Jonathan N Coleman
- Centre for Research on Adaptive Nanostructures & Nanodevices (CRANN) and Advanced Materials and BioEngineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| | - Hugh J Byrne
- FOCAS Research Institute, Technological University Dublin, City Centre Campus, Dublin 8, Ireland
| | | |
Collapse
|
21
|
Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, Yi P, Tang L, Pan Q, Rao S, Liang J, Tang Y, Su M, Luo X, Yang Y, Shi Y, Wang H, Zhou Y, Liao Q. The cancer metabolic reprogramming and immune response. Mol Cancer 2021; 20:28. [PMID: 33546704 PMCID: PMC7863491 DOI: 10.1186/s12943-021-01316-8] [Citation(s) in RCA: 579] [Impact Index Per Article: 144.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/19/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
The overlapping metabolic reprogramming of cancer and immune cells is a putative determinant of the antitumor immune response in cancer. Increased evidence suggests that cancer metabolism not only plays a crucial role in cancer signaling for sustaining tumorigenesis and survival, but also has wider implications in the regulation of antitumor immune response through both the release of metabolites and affecting the expression of immune molecules, such as lactate, PGE2, arginine, etc. Actually, this energetic interplay between tumor and immune cells leads to metabolic competition in the tumor ecosystem, limiting nutrient availability and leading to microenvironmental acidosis, which hinders immune cell function. More interestingly, metabolic reprogramming is also indispensable in the process of maintaining self and body homeostasis by various types of immune cells. At present, more and more studies pointed out that immune cell would undergo metabolic reprogramming during the process of proliferation, differentiation, and execution of effector functions, which is essential to the immune response. Herein, we discuss how metabolic reprogramming of cancer cells and immune cells regulate antitumor immune response and the possible approaches to targeting metabolic pathways in the context of anticancer immunotherapy. We also describe hypothetical combination treatments between immunotherapy and metabolic intervening that could be used to better unleash the potential of anticancer therapies.
Collapse
Affiliation(s)
- Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Pin Yi
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.,University of South China, 421001, Hengyang, Hunan, China
| | - Lu Tang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.,University of South China, 421001, Hengyang, Hunan, China
| | - Qing Pan
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.,University of South China, 421001, Hengyang, Hunan, China
| | - Shan Rao
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Jiaxin Liang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Min Su
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Xia Luo
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yiqing Yang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yingrui Shi
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Hui Wang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.
| |
Collapse
|
22
|
Cyclobutane pyrimidine dimers from UVB exposure induce a hypermetabolic state in keratinocytes via mitochondrial oxidative stress. Redox Biol 2020; 38:101808. [PMID: 33264701 PMCID: PMC7708942 DOI: 10.1016/j.redox.2020.101808] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/28/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Ultraviolet B radiation (UVB) is an environmental complete carcinogen, which induces and promotes keratinocyte carcinomas, the most common human malignancies. UVB induces the formation of cyclobutane pyrimidine dimers (CPDs). Repairing CPDs through nucleotide excision repair is slow and error-prone in placental mammals. In addition to the mutagenic and malignancy-inducing effects, UVB also elicits poorly understood complex metabolic changes in keratinocytes, possibly through CPDs. To determine the effects of CPDs, CPD-photolyase was overexpressed in keratinocytes using an N1-methyl pseudouridine-containing in vitro-transcribed mRNA. CPD-photolyase, which is normally not present in placental mammals, can efficiently and rapidly repair CPDs to block signaling pathways elicited by CPDs. Keratinocytes surviving UVB irradiation turn hypermetabolic. We show that CPD-evoked mitochondrial reactive oxygen species production, followed by the activation of several energy sensor enzymes, including sirtuins, AMPK, mTORC1, mTORC2, p53, and ATM, is responsible for the compensatory metabolic adaptations in keratinocytes surviving UVB irradiation. Compensatory metabolic changes consist of enhanced glycolytic flux, Szent-Györgyi-Krebs cycle, and terminal oxidation. Furthermore, mitochondrial fusion, mitochondrial biogenesis, and lipophagy characterize compensatory hypermetabolism in UVB-exposed keratinocytes. These properties not only support the survival of keratinocytes, but also contribute to UVB-induced differentiation of keratinocytes. Our results indicate that CPD-dependent signaling acutely maintains skin integrity by supporting cellular energy metabolism.
Collapse
|
23
|
Agarwal P, Combes TW, Shojaee-Moradie F, Fielding B, Gordon S, Mizrahi V, Martinez FO. Foam Cells Control Mycobacterium tuberculosis Infection. Front Microbiol 2020; 11:1394. [PMID: 32754123 PMCID: PMC7381311 DOI: 10.3389/fmicb.2020.01394] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/20/2020] [Accepted: 05/29/2020] [Indexed: 12/27/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) infects macrophages and macrophage-derived foam cells, a hallmark of granulomata in tuberculous lesions. We analyzed the effects of lipid accumulation in human primary macrophages and quantified strong triglyceride and phospholipid remodeling which depended on the dietary fatty acid used for the assay. The enrichment of >70% in triglyceride and phospholipids can alter cell membrane properties, signaling and phagocytosis in macrophages. In conventional macrophage cultures, cells are heterogeneous, small or large macrophages. In foam cells, a third population of 30% of cells with increased granularity can be detected. We found that foam cell formation is heterogenous and that lipid accumulation and foam cell formation reduces the phagocytosis of Mtb. Under the conditions tested, cell death was highly prevalent in macrophages, whereas foam cells were largely protected from this effect. Foam cells also supported slower Mtb replication, yet this had no discernible impact on the intracellular efficacy of four different antitubercular drugs. Foam cell formation had a significant impact in the inflammatory potential of the cells. TNF-α, IL-1β, and prototypical chemokines were increased. The ratio of inflammatory IL-1β, TNF-α, and IL-6 vs. anti-inflammatory IL-10 was significantly higher in response to Mtb vs. LPS, and was increased in foam cells compared to macrophages, suggestive of increased pro-inflammatory properties. Cytokine production correlated with NF-κB activation in our models. We conclude that foam cell formation reduces the host cell avidity for, and phagocytosis of, Mtb while protecting the cells from death. This protective effect is associated with enhanced inflammatory potential of foam cells and restricted intracellular growth of Mtb.
Collapse
Affiliation(s)
- Pooja Agarwal
- South African Medical Research Council/National Health Laboratory Service/University of Cape Town, Molecular Mycobacteriology Research Unit, Division of Medical Microbiology, Department of Pathology, Department of Science and Innovation/National Research Foundation, Centre of Excellence for Biomedical TB Research and Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Theo W Combes
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | | | - Barbara Fielding
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Valerie Mizrahi
- South African Medical Research Council/National Health Laboratory Service/University of Cape Town, Molecular Mycobacteriology Research Unit, Division of Medical Microbiology, Department of Pathology, Department of Science and Innovation/National Research Foundation, Centre of Excellence for Biomedical TB Research and Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Fernando O Martinez
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
24
|
Abstract
Caseum, the central necrotic material of tuberculous lesions, is a reservoir of drug-recalcitrant persisting mycobacteria. Caseum is found in closed nodules and in open cavities connecting with an airway. Several commonly accepted characteristics of caseum were established during the preantibiotic era, when autopsies of deceased tuberculosis (TB) patients were common but methodologies were limited. These pioneering studies generated concepts such as acidic pH, low oxygen tension, and paucity of nutrients being the drivers of nonreplication and persistence in caseum. Here we review widely accepted beliefs about the caseum-specific stress factors thought to trigger the shift of Mycobacterium tuberculosis to drug tolerance. Our current state of knowledge reveals that M. tuberculosis is faced with a lipid-rich diet rather than nutrient deprivation in caseum. Variable caseum pH is seen across lesions, possibly transiently acidic in young lesions but overall near neutral in most mature lesions. Oxygen tension is low in the avascular caseum of closed nodules and high at the cavity surface, and a gradient of decreasing oxygen tension likely forms toward the cavity wall. Since caseum is largely made of infected and necrotized macrophages filled with lipid droplets, the microenvironmental conditions encountered by M. tuberculosis in foamy macrophages and in caseum bear many similarities. While there remain a few knowledge gaps, these findings constitute a solid starting point to develop high-throughput drug discovery assays that combine the right balance of oxygen tension, pH, lipid abundance, and lipid species to model the profound drug tolerance of M. tuberculosis in caseum.
Collapse
Affiliation(s)
- Jansy P Sarathy
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine at Seton Hall University, Nutley, New Jersey, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine at Seton Hall University, Nutley, New Jersey, USA
| |
Collapse
|
25
|
Lian Z, Perrard XYD, Peng X, Raya JL, Hernandez AA, Johnson CG, Lagor WR, Pownall HJ, Hoogeveen RC, Simon SI, Sacks FM, Ballantyne CM, Wu H. Replacing Saturated Fat With Unsaturated Fat in Western Diet Reduces Foamy Monocytes and Atherosclerosis in Male Ldlr-/- Mice. Arterioscler Thromb Vasc Biol 2020; 40:72-85. [PMID: 31619061 PMCID: PMC6991890 DOI: 10.1161/atvbaha.119.313078] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/20/2019] [Accepted: 09/29/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE A Mediterranean diet supplemented with olive oil and nuts prevents cardiovascular disease in clinical studies, but the underlying mechanisms are incompletely understood. We investigated whether the preventive effect of the diet could be due to inhibition of atherosclerosis and foamy monocyte formation in Ldlr-/- mice fed with a diet in which milkfat in a Western diet (WD) was replaced with extra-virgin olive oil and nuts (EVOND). Approach and Results: Ldlr-/- mice were fed EVOND or a Western diet for 3 (or 6) months. Compared with the Western diet, EVOND decreased triglyceride and cholesterol levels but increased unsaturated fatty acid concentrations in plasma. EVOND also lowered intracellular lipid accumulation in circulating monocytes, indicating less formation of foamy monocytes, compared with the Western diet. In addition, compared with the Western diet, EVOND reduced monocyte expression of inflammatory cytokines, CD36, and CD11c, with decreased monocyte uptake of oxLDL (oxidized LDL [low-density lipoprotein]) ex vivo and reduced CD11c+ foamy monocyte firm arrest on vascular cell adhesion molecule-1 and E-selectin-coated slides in an ex vivo shear flow assay. Along with these changes, EVOND compared with the Western diet reduced the number of CD11c+ macrophages in atherosclerotic lesions and lowered atherosclerotic lesion area of the whole aorta and aortic sinus. CONCLUSIONS A diet enriched in extra-virgin olive oil and nuts, compared with a Western diet high in saturated fat, lowered plasma cholesterol and triglyceride levels, inhibited foamy monocyte formation, inflammation, and adhesion, and reduced atherosclerosis in Ldlr-/- mice.
Collapse
Affiliation(s)
- Zeqin Lian
- From the Department of Medicine (Z.L., X.D.P., X.P., J.L.R., C.G.J., R.C.H., C.M.B., H.W.), Baylor College of Medicine, Houston, TX
| | - Xiao-Yuan Dai Perrard
- From the Department of Medicine (Z.L., X.D.P., X.P., J.L.R., C.G.J., R.C.H., C.M.B., H.W.), Baylor College of Medicine, Houston, TX
| | - Xueying Peng
- From the Department of Medicine (Z.L., X.D.P., X.P., J.L.R., C.G.J., R.C.H., C.M.B., H.W.), Baylor College of Medicine, Houston, TX
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (X.P)
| | - Joe L Raya
- From the Department of Medicine (Z.L., X.D.P., X.P., J.L.R., C.G.J., R.C.H., C.M.B., H.W.), Baylor College of Medicine, Houston, TX
| | - Alfredo A Hernandez
- Department of Biomedical Engineering, University of California, Davis (A.A.H, S.I.S.)
| | - Collin G Johnson
- From the Department of Medicine (Z.L., X.D.P., X.P., J.L.R., C.G.J., R.C.H., C.M.B., H.W.), Baylor College of Medicine, Houston, TX
| | - William R Lagor
- Department of Molecular Physiology and Biophysics (W.R.L.), Baylor College of Medicine, Houston, TX
| | - Henry J Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX (H.J.P.)
| | - Ron C Hoogeveen
- From the Department of Medicine (Z.L., X.D.P., X.P., J.L.R., C.G.J., R.C.H., C.M.B., H.W.), Baylor College of Medicine, Houston, TX
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis (A.A.H, S.I.S.)
| | - Frank M Sacks
- Department of Nutrition, Harvard School of Public Health, and Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA (F.M.S.)
| | - Christie M Ballantyne
- From the Department of Medicine (Z.L., X.D.P., X.P., J.L.R., C.G.J., R.C.H., C.M.B., H.W.), Baylor College of Medicine, Houston, TX
- Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX
- Center for Cardiometabolic Disease Prevention (C.M.B.), Baylor College of Medicine, Houston, TX
| | - Huaizhu Wu
- From the Department of Medicine (Z.L., X.D.P., X.P., J.L.R., C.G.J., R.C.H., C.M.B., H.W.), Baylor College of Medicine, Houston, TX
- Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX
| |
Collapse
|
26
|
Chang HR, Josefs T, Scerbo D, Gumaste N, Hu Y, Huggins LA, Barett T, Chiang S, Grossman J, Bagdasarov S, Fisher EA, Goldberg IJ. Role of LpL (Lipoprotein Lipase) in Macrophage Polarization In Vitro and In Vivo. Arterioscler Thromb Vasc Biol 2019; 39:1967-1985. [PMID: 31434492 PMCID: PMC6761022 DOI: 10.1161/atvbaha.119.312389] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Fatty acid uptake and oxidation characterize the metabolism of alternatively activated macrophage polarization in vitro, but the in vivo biology is less clear. We assessed the roles of LpL (lipoprotein lipase)-mediated lipid uptake in macrophage polarization in vitro and in several important tissues in vivo. Approach and Results: We created mice with both global and myeloid-cell specific LpL deficiency. LpL deficiency in the presence of VLDL (very low-density lipoproteins) altered gene expression of bone marrow-derived macrophages and led to reduced lipid uptake but an increase in some anti- and some proinflammatory markers. However, LpL deficiency did not alter lipid accumulation or gene expression in circulating monocytes nor did it change the ratio of Ly6Chigh/Ly6Clow. In adipose tissue, less macrophage lipid accumulation was found with global but not myeloid-specific LpL deficiency. Neither deletion affected the expression of inflammatory genes. Global LpL deficiency also reduced the numbers of elicited peritoneal macrophages. Finally, we assessed gene expression in macrophages from atherosclerotic lesions during regression; LpL deficiency did not affect the polarity of plaque macrophages. CONCLUSIONS The phenotypic changes observed in macrophages upon deletion of Lpl in vitro is not mimicked in tissue macrophages.
Collapse
Affiliation(s)
- Hye Rim Chang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Tatjana Josefs
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Diego Scerbo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Namrata Gumaste
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Yunying Hu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Lesley-Ann Huggins
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Tessa Barett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York; Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, New York, New York
| | - Stephanie Chiang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Jennifer Grossman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Svetlana Bagdasarov
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Edward A. Fisher
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| |
Collapse
|
27
|
Tfaili S, Al Assaad A, Fournier N, Allaoui F, Paul JL, Chaminade P, Tfayli A. Investigation of lipid modifications in J774 macrophages by vibrational spectroscopies after eicosapentaenoic acid membrane incorporation in unloaded and cholesterol-loaded cells. Talanta 2019; 199:54-64. [DOI: 10.1016/j.talanta.2019.01.122] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/20/2018] [Revised: 01/20/2019] [Accepted: 01/22/2019] [Indexed: 01/19/2023]
|
28
|
Schnitzler JG, Dallinga-Thie GM, Kroon J. The Role of (Modified) Lipoproteins in Vascular Function: A Duet Between Monocytes and the Endothelium. Curr Med Chem 2019; 26:1594-1609. [PMID: 29546830 DOI: 10.2174/0929867325666180316121015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/01/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Over the last century, many studies have demonstrated that low-density lipoprotein (LDL) is a key risk factor of cardiovascular diseases (CVD) related to atherosclerosis. Thus, for these CVD patients, LDL lowering agents are commonly used in the clinic to reduce the risk for CVD. LDL, upon modification, will develop distinct inflammatory and proatherogenic potential, leading to impaired endothelial integrity, influx of immune cells and subsequent increased foam cell formation. LDL can also directly affect peripheral monocyte composition, rendering them in a more favorable position to migrate and accumulate in the subendothelial space. It has become apparent that other lipoprotein particles, such as triglyceride- rich lipoproteins or remnants (TRL) and lipoprotein(a) [Lp(a)] may also impact on atherogenic pathways. Evidence is accumulating that Lp(a) can promote peripheral monocyte activation, eventually leading to increased transmigration through the endothelium. Similarly, remnant cholesterol has been identified to play a key role in endothelial dysfunction and monocyte behavior. In this review, we will discuss recent developments in understanding the role of different lipoproteins in the context of inflammation at both the level of the monocyte and the endothelium.
Collapse
Affiliation(s)
- Johan G Schnitzler
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Argov-Argaman N. Symposium review: Milk fat globule size: Practical implications and metabolic regulation. J Dairy Sci 2019; 102:2783-2795. [PMID: 30639008 DOI: 10.3168/jds.2018-15240] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/19/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
Abstract
Milk fat globule (MFG) size ranges over 3 orders of magnitude, from less than 200 nm to over 15 µm. The significance of MFG size derives from its tight association with its lipidome and proteome. More specifically, small MFG have relatively higher content of membrane compared with large globules, and this membrane exerts diverse positive health effects, as reported in human and animal studies. In addition, MFG size has industrial significance, as it affects the physicochemical and sensory characteristics of dairy products. Studies on the size regulation of MFG are scarce, mainly because various confounders indirectly affect MFG size. Because MFG size is determined before and during its secretion from mammary epithelial cells, studies on the size regulation of its precursors, the intracellular lipid droplets (LD), have been used as a proxy for understanding the mechanisms controlling MFG size. In this review, we provide evidence for 2 distinct mechanisms regulating LD size in mammary epithelial cells: co-regulation of fat content and triglyceride-synthesis capacity of the cells, and fusion between LD. The latter is controlled by the membrane's polar lipid composition and involves mitochondrial enzymes. Accordingly, this review also discusses MFG size regulation in the in vivo metabolic context, as MFG morphometric features are often modulated under conditions that involve animals' altered energy status.
Collapse
Affiliation(s)
- Nurit Argov-Argaman
- Department of Animal Science, the Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University of Jerusalem, Israel, POB 76100.
| |
Collapse
|
30
|
Norman JE, Aung HH, Wilson DW, Rutledge JC. Inhibition of perilipin 2 expression reduces pro-inflammatory gene expression and increases lipid droplet size. Food Funct 2018; 9:6245-6256. [PMID: 30402637 PMCID: PMC6292725 DOI: 10.1039/c8fo01420e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022]
Abstract
Our lab previously demonstrated that triglyceride-rich lipoprotein (TGRL) lipolysis products induce lipid droplet formation and pro-inflammatory gene expression in monocytes. We hypothesized that the inhibition of perilipin 2 expression in THP-1 monocytes would reduce lipid droplet formation and suppress pro-inflammatory gene expression induced by TGRL lipolysis products. In the current study, we use microarray analysis to identify gene expression altered by TGRL lipolysis products in THP-1 monocytes. We confirmed the expression of selected genes by quantitative reverse transcription PCR and characterized lipid droplet formation in these cells after exposure to TGRL lipolysis products. Using siRNA inhibition of perilipin 2 expression, we examined the role of perilipin 2 in the response of THP-1 monocytes to TGRL lipolysis products. We found that perilipin 2 siRNA increased the intracellular triglyceride content, increased the size of lipid droplets, and reduced pro-atherogenic and pro-inflammatory gene expression. We saw a reduction of serum/glucocorticoid kinase 1, v-maf musculoaponeurotic fibrosarcoma oncogene homolog F (avian), chemokine (C-C motif) ligand 3, and interleukin 8 gene expression induced by TGRL lipolysis products. This study supports previous findings that reduction of perilipin 2 expression is protective against atherogenesis, while finding an unexpected increase in lipid droplet size with reduced perilipin 2 expression.
Collapse
Affiliation(s)
- Jennifer E Norman
- University of California, Davis, School of Medicine, Department of Internal Medicine, Division of Cardiovascular Medicine, GBSF 5404, 451 Health Sciences Dr. Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
31
|
Mitrofanova A, Molina J, Varona Santos J, Guzman J, Morales XA, Ducasa GM, Bryn J, Sloan A, Volosenco I, Kim JJ, Ge M, Mallela SK, Kretzler M, Eddy S, Martini S, Wahl P, Pastori S, Mendez AJ, Burke GW, Merscher S, Fornoni A. Hydroxypropyl-β-cyclodextrin protects from kidney disease in experimental Alport syndrome and focal segmental glomerulosclerosis. Kidney Int 2018; 94:1151-1159. [PMID: 30301568 PMCID: PMC6278936 DOI: 10.1016/j.kint.2018.06.031] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/02/2018] [Revised: 06/14/2018] [Accepted: 06/28/2018] [Indexed: 10/28/2022]
Abstract
Studies suggest that altered renal lipid metabolism plays a role in the pathogenesis of diabetic kidney disease and that genetic or pharmacological induction of cholesterol efflux protects from the development of diabetic kidney disease and focal segmental glomerulosclerosis (FSGS). Here we tested whether altered lipid metabolism contributes to renal failure in the Col4a3 knockout mouse model for Alport Syndrome. There was an eight-fold increase in the cholesterol content in renal cortexes of mice with Alport Syndrome. This was associated with increased glomerular lipid droplets and cholesterol crystals. Treatment of mice with Alport Syndrome with hydroxypropyl-β-cyclodextrin (HPβCD) reduced cholesterol content in the kidneys of mice with Alport Syndrome and protected from the development of albuminuria, renal failure, inflammation and tubulointerstitial fibrosis. Cholesterol efflux and trafficking-related genes were primarily affected in mice with Alport Syndrome and were differentially regulated in the kidney cortex and isolated glomeruli. HPβCD also protected from proteinuria and mesangial expansion in a second model of non-metabolic kidney disease, adriamycin-induced nephropathy. Consistent with our experimental findings, microarray analysis confirmed dysregulation of several lipid-related genes in glomeruli isolated from kidney biopsies of patients with primary FSGS enrolled in the NEPTUNE study. Thus, lipid dysmetabolism occurs in non-metabolic glomerular disorders such as Alport Syndrome and FSGS, and HPβCD improves renal function in experimental Alport Syndrome and FSGS.
Collapse
Affiliation(s)
- Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Judith Molina
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Javier Varona Santos
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Johanna Guzman
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ximena A Morales
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - G Michelle Ducasa
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jonathan Bryn
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alexis Sloan
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ion Volosenco
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shamroop K Mallela
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Matthias Kretzler
- Division of Nephrology, Departments of Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Sean Eddy
- Division of Nephrology, Departments of Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Sebastian Martini
- Division of Nephrology, Departments of Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Patricia Wahl
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Santiago Pastori
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Armando J Mendez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - George W Burke
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA; Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
32
|
Cohen BC, Raz C, Shamay A, Argov-Argaman N. Lipid Droplet Fusion in Mammary Epithelial Cells is Regulated by Phosphatidylethanolamine Metabolism. J Mammary Gland Biol Neoplasia 2017; 22:235-249. [PMID: 29188493 DOI: 10.1007/s10911-017-9386-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/08/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022] Open
Abstract
Mammary epithelial cells (MEC) secrete fat in the form of milk fat globules (MFG) which are found in milk in diverse sizes. MFG originate from intracellular lipid droplets, and the mechanism underlying their size regulation is still elusive. Two main mechanisms have been suggested to control lipid droplet size. The first is a well-documented pathway, which involves regulation of cellular triglyceride content. The second is the fusion pathway, which is less-documented, especially in mammalian cells, and its importance in the regulation of droplet size is still unclear. Using biochemical and molecular inhibitors, we provide evidence that in MEC, lipid droplet size is determined by fusion, independent of cellular triglyceride content. The extent of fusion is determined by the cell membrane's phospholipid composition. In particular, increasing phosphatidylethanolamine (PE) content enhances fusion between lipid droplets and hence increases lipid droplet size. We further identified the underlying biochemical mechanism that controls this content as the mitochondrial enzyme phosphatidylserine decarboxylase; siRNA knockdown of this enzyme reduced the number of large lipid droplets threefold. Further, inhibition of phosphatidylserine transfer to the mitochondria, where its conversion to PE occurs, diminished the large lipid droplet phenotype in these cells. These results reveal, for the first time to our knowledge in mammalian cells and specifically in mammary epithelium, the missing biochemical link between the metabolism of cellular complex lipids and lipid-droplet fusion, which ultimately defines lipid droplet size.
Collapse
Affiliation(s)
- Bat-Chen Cohen
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel.
| | - Chen Raz
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel
| | - Avi Shamay
- Department of Ruminant Science, Agricultural Research Organization, Volcani Center, Bet Dagan, Israel
| | - Nurit Argov-Argaman
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel
| |
Collapse
|
33
|
Dai Perrard XY, Lian Z, Bobotas G, Dicklin MR, Maki KC, Wu H. Effects of n-3 fatty acid treatment on monocyte phenotypes in humans with hypertriglyceridemia. J Clin Lipidol 2017; 11:1361-1371. [PMID: 28942094 PMCID: PMC5698114 DOI: 10.1016/j.jacl.2017.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/31/2017] [Revised: 07/26/2017] [Accepted: 08/22/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Hypertriglyceridemia increases risk for atherosclerotic cardiovascular disease and may contribute to atherosclerosis by changing circulating monocyte phenotypes. High-dose n-3 polyunsaturated fatty acids reduce blood triglyceride levels. Effects of triglyceride-lowering therapy on monocyte phenotypes are not well known. OBJECTIVE We examined effects of n-3 polyunsaturated fatty acid treatments (eicosapentaenoic acid [EPA] plus docosapentaenoic acid [MAT9001] vs EPA ethyl esters [EPA-EE]) on monocyte phenotypes in individuals with hypertriglyceridemia. METHODS Individuals with triglycerides 200 to 400 mg/dL were recruited. Subjects received 2 treatments in randomized order for 14 days each: MAT9001 and EPA-EE, at 4 g/d. At 2 days before the start of, and on the last day of, each treatment, nile red staining for lipids and phenotypes of each monocyte subset were examined by flow cytometry after an overnight fast and postprandially after a high-fat meal. RESULTS Treatment with MAT9001 or EPA-EE reduced fasting triglyceride levels and decreased proportions of intermediate monocytes. Only MAT9001 decreased postprandial blood triglyceride levels, lowered fasting nile red levels, indicating less lipid in classical and intermediate monocytes, and reduced postprandial CD11c levels on nonclassical monocytes. MAT9001 and EPA-EE each reduced fasting and postprandial CD11c and CD36 levels on classical and intermediate monocytes and postprandial CCR5 levels on intermediate and nonclassical monocytes, with no significant differences between the 2 treatments. CONCLUSIONS Treatment with MAT9001 in individuals with hypertriglyceridemia reduced fasting nile red staining for lipids in classical and intermediate monocytes. MAT9001 and EPA-EE each improved fasting and postprandial monocyte phenotypes, which could potentially help to protect against atherosclerosis.
Collapse
Affiliation(s)
| | - Zeqin Lian
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Mary R Dicklin
- Midwest Biomedical Research/Center for Metabolic and Cardiovascular Health, Glen Ellyn, IL, USA
| | - Kevin C Maki
- Midwest Biomedical Research/Center for Metabolic and Cardiovascular Health, Glen Ellyn, IL, USA
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
34
|
Stiebing C, Meyer T, Rimke I, Matthäus C, Schmitt M, Lorkowski S, Popp J. Real-time Raman and SRS imaging of living human macrophages reveals cell-to-cell heterogeneity and dynamics of lipid uptake. JOURNAL OF BIOPHOTONICS 2017; 10:1217-1226. [PMID: 28164480 DOI: 10.1002/jbio.201600279] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 11/01/2016] [Revised: 12/23/2016] [Accepted: 01/18/2017] [Indexed: 06/06/2023]
Abstract
Monitoring living cells in real-time is important in order to unravel complex dynamic processes in life sciences. In particular the dynamics of initiation and progression of degenerative diseases is intensely studied. In atherosclerosis the thickening of arterial walls is related to high lipid levels in the blood stream, which trigger the lipid uptake and formation of droplets as neutral lipid reservoirs in macrophages in the arterial wall. Unregulated lipid uptake finally results in foam cell formation, which is a hallmark of atherosclerosis. In previous studies, the uptake and storage of different fatty acids was monitored by measuring fixed cells. Commonly employed fluorescence staining protocols are often error prone because of cytotoxicity and unspecific fluorescence backgrounds. By following living cells with Raman spectroscopic imaging, lipid uptake of macrophages was studied with real-time data acquisition. Isotopic labeling using deuterated palmitic acid has been combined with spontaneous and stimulated Raman imaging to investigate the dynamic process of fatty acid storage in human macrophages for incubation times from 45 min to 37 h. Striking heterogeneity in the uptake rate and the total concentration of deuterated palmitic acid covering two orders of magnitude is detected in single as well as ensembles of cultured human macrophages. SRS signal of deuterated palmitic acid measured at the CD vibration band after incorporation into living macrophages.
Collapse
Affiliation(s)
- Clara Stiebing
- Leibniz Institute of Photonic Technology (IPHT), Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Tobias Meyer
- Leibniz Institute of Photonic Technology (IPHT), Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Ingo Rimke
- APE Angewandte Physik & Elektronik GmbH, Plauener Straße 163-165, 13053, Berlin
| | - Christian Matthäus
- Leibniz Institute of Photonic Technology (IPHT), Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Michael Schmitt
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Stefan Lorkowski
- Institute of Nutrition and Abbe Center of Photonics, Friedrich Schiller University Jena, Dornburger Straße 25, 07743, Jena, Germany
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Friedrich Schiller University Jena, Dornburger Straße 25, 07743, Jena, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology (IPHT), Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
| |
Collapse
|
35
|
Ameer F, Munir R, Usman H, Rashid R, Shahjahan M, Hasnain S, Zaidi N. Lipid-load in peripheral blood mononuclear cells: Impact of food-consumption, dietary-macronutrients, extracellular lipid availability and demographic factors. Biochimie 2017; 135:104-110. [DOI: 10.1016/j.biochi.2017.01.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/28/2016] [Accepted: 01/29/2017] [Indexed: 11/29/2022]
|
36
|
Rahman MS, Murphy AJ, Woollard KJ. Effects of dyslipidaemia on monocyte production and function in cardiovascular disease. Nat Rev Cardiol 2017; 14:387-400. [PMID: 28300081 DOI: 10.1038/nrcardio.2017.34] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
Monocytes are heterogeneous effector cells involved in the maintenance and restoration of tissue integrity. Monocytes and macrophages are involved in cardiovascular disease progression, and are associated with the development of unstable atherosclerotic plaques. Hyperlipidaemia can accelerate cardiovascular disease progression. However, monocyte responses to hyperlipidaemia are poorly understood. In the past decade, accumulating data describe the relationship between the dynamic blood lipid environment and the heterogeneous circulating monocyte pool, which might have profound consequences for cardiovascular disease. In this Review, we explore the updated view of monocytes in cardiovascular disease and their relationship with macrophages in promoting the homeostatic and inflammatory responses related to atherosclerosis. We describe the different definitions of dyslipidaemia, highlight current theories on the ontogeny of monocyte heterogeneity, discuss how dyslipidaemia might alter monocyte production, and explore the mechanistic interface linking dyslipidaemia with monocyte effector functions, such as migration and the inflammatory response. Finally, we discuss the role of dietary and endogenous lipid species in mediating dyslipidaemic responses, and the role of these lipids in promoting the risk of cardiovascular disease through modulation of monocyte behaviour.
Collapse
Affiliation(s)
- Mohammed Shamim Rahman
- Renal &Vascular Inflammation Section, Division of Immunology and Inflammation, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology Lab, Baker IDI Heart &Diabetes Research Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia.,Department of Immunology, Monash University, 89 Commercial Road, Melbourne, Victoria 3004, Australia
| | - Kevin J Woollard
- Renal &Vascular Inflammation Section, Division of Immunology and Inflammation, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
37
|
Transcriptomic Analysis of THP-1 Macrophages Exposed to Lipoprotein Hydrolysis Products Generated by Lipoprotein Lipase. Lipids 2017; 52:189-205. [DOI: 10.1007/s11745-017-4238-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/07/2016] [Accepted: 02/02/2017] [Indexed: 11/25/2022]
|
38
|
Stiebing C, Schmölz L, Wallert M, Matthäus C, Lorkowski S, Popp J. Raman imaging of macrophages incubated with triglyceride-enriched oxLDL visualizes translocation of lipids between endocytic vesicles and lipid droplets. J Lipid Res 2017; 58:876-883. [PMID: 28143895 DOI: 10.1194/jlr.m071688] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2016] [Revised: 01/15/2017] [Indexed: 01/01/2023] Open
Abstract
Raman spectroscopic imaging was used to investigate the uptake of oxidized LDLs (oxLDLs) by human macrophages. To better understand the endocytic pathway and the intracellular fate of modified lipoproteins is of foremost interest with regard to the development of atherosclerotic plaques. To obtain information on the storage process of lipids caused by oxLDL uptake, Raman spectroscopic imaging was used because of its unique chemical specificity, especially for lipids. For the present study, a protocol was established to incorporate deuterated tripalmitate into oxLDL. Subsequently, human THP-1 macrophages were incubated for different time points and their chemical composition was analyzed using Raman spectroscopic imaging. β-Carotene was found to be a reliable marker molecule for the uptake of lipoproteins into macrophages. In addition, lipoprotein administration led to small endocytic vesicles with different concentrations of deuterated lipids within the cells. For the first time, the translocation of deuterated lipids from endocytic vesicles into lipid droplets over time is reported in mature human THP-1 macrophages.
Collapse
Affiliation(s)
- Clara Stiebing
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Lisa Schmölz
- Institute of Nutrition and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743 Jena, Germany.,Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Maria Wallert
- Institute of Nutrition and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743 Jena, Germany.,Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Christian Matthäus
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Stefan Lorkowski
- Institute of Nutrition and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743 Jena, Germany.,Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany .,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
39
|
Lee LL, Aung HH, Wilson DW, Anderson SE, Rutledge JC, Rutkowsky JM. Triglyceride-rich lipoprotein lipolysis products increase blood-brain barrier transfer coefficient and induce astrocyte lipid droplets and cell stress. Am J Physiol Cell Physiol 2017; 312:C500-C516. [PMID: 28077357 DOI: 10.1152/ajpcell.00120.2016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/03/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/19/2022]
Abstract
Elevation of blood triglycerides, primarily as triglyceride-rich lipoproteins (TGRL), has been linked to cerebrovascular inflammation, vascular dementia, and Alzheimer's disease (AD). Brain microvascular endothelial cells and astrocytes, two cell components of the neurovascular unit, participate in controlling blood-brain barrier (BBB) permeability and regulating neurovascular unit homeostasis. Our studies showed that infusion of high physiological concentrations of TGRL lipolysis products (TGRL + lipoprotein lipase) activate and injure brain endothelial cells and transiently increase the BBB transfer coefficient (Ki = permeability × surface area/volume) in vivo. However, little is known about how blood lipids affect astrocyte lipid accumulation and inflammation. To address this, we first demonstrated TGRL lipolysis products increased lipid droplet formation in cultured normal human astrocytes. We then evaluated the transcriptional pathways activated in astrocytes by TGRL lipolysis products and found upregulated stress and inflammatory-related genes including activating transcription factor 3 (ATF3), macrophage inflammatory protein-3α (MIP-3α), growth differentiation factor-15 (GDF15), and prostaglandin-endoperoxide synthase 2 (COX2). TGRL lipolysis products also activated the JNK/cJUN/ATF3 pathway, induced endoplasmic reticulum stress protein C/EBP homologous protein (CHOP), and the NF-κB pathway, while increasing secretion of MIP-3α, GDF15, and IL-8. Thus our results demonstrate TGRL lipolysis products increase the BBB transfer coefficient (Ki), induce astrocyte lipid droplet formation, activate cell stress pathways, and induce secretion of inflammatory cytokines. Our observations are consistent with evidence for lipid-induced neurovascular injury and inflammation, and we, therefore, speculate that lipid-induced astrocyte injury could play a role in cognitive decline.
Collapse
Affiliation(s)
- Linda L Lee
- Department of Internal Medicine, University of California, Davis, California
| | - Hnin H Aung
- Department of Internal Medicine, University of California, Davis, California
| | - Dennis W Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California; and
| | - Steven E Anderson
- Department of Physiology and Membrane Biology, University of California, Davis, California
| | - John C Rutledge
- Department of Internal Medicine, University of California, Davis, California
| | | |
Collapse
|
40
|
Khan IM, Pokharel Y, Dadu RT, Lewis DE, Hoogeveen RC, Wu H, Ballantyne CM. Postprandial Monocyte Activation in Individuals With Metabolic Syndrome. J Clin Endocrinol Metab 2016; 101:4195-4204. [PMID: 27575945 PMCID: PMC5095236 DOI: 10.1210/jc.2016-2732] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
CONTEXT Postprandial hyperlipidemia has been suggested to contribute to atherogenesis by inducing proinflammatory changes in monocytes. Individuals with metabolic syndrome (MS), shown to have higher blood triglyceride concentration and delayed triglyceride clearance, may thus have increased risk for development of atherosclerosis. OBJECTIVE Our objective was to examine fasting levels and effects of a high-fat meal on phenotypes of monocyte subsets in individuals with obesity and MS and in healthy controls. Design, Setting, Participants, Intervention: Individuals with obesity and MS and gender- and age-matched healthy controls were recruited. Blood was collected from participants after an overnight fast (baseline) and at 3 and 5 hours after ingestion of a high-fat meal. At each time point, monocyte phenotypes were examined by multiparameter flow cytometry. MAIN OUTCOME MEASURES Baseline levels of activation markers and postprandial inflammatory response in each of the three monocyte subsets were measured. RESULTS At baseline, individuals with obesity and MS had higher proportions of circulating lipid-laden foamy monocytes than controls, which were positively correlated with fasting triglyceride levels. Additionally, the MS group had increased counts of nonclassical monocytes, higher CD11c, CX3CR1, and human leukocyte antigen-DR levels on intermediate monocytes, and higher CCR5 and tumor necrosis factor-α levels on classical monocytes in the circulation. Postprandial triglyceride increases in both groups were paralleled by upregulation of lipid-laden foamy monocytes. MS, but not control, subjects had significant postprandial increases of CD11c and percentages of IL-1β+ and tumor necrosis factor-α+ cells in nonclassical monocytes. CONCLUSIONS Compared to controls, individuals with obesity and MS had increased fasting and postprandial monocyte lipid accumulation and activation.
Collapse
Affiliation(s)
- Ilvira M Khan
- Interdepartmental Program in Translational Biology and Molecular Medicine (I.M.K.), Baylor College of Medicine, Houston, Texas; Division of Atherosclerosis and Vascular Medicine (I.M.K., Y.P., R.T.D., R.C.H., H.W., C.M.B.), Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Infectious Diseases (D.E.L.), Department of Internal Medicine, UT Health, Houston, Texas; Section of Leukocyte Biology (H.W., C.M.B.), Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Center for Cardiovascular Disease Prevention (H.W., C.M.B.), Methodist DeBakey Heart and Vascular Center, Houston, Texas
| | - Yashashwi Pokharel
- Interdepartmental Program in Translational Biology and Molecular Medicine (I.M.K.), Baylor College of Medicine, Houston, Texas; Division of Atherosclerosis and Vascular Medicine (I.M.K., Y.P., R.T.D., R.C.H., H.W., C.M.B.), Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Infectious Diseases (D.E.L.), Department of Internal Medicine, UT Health, Houston, Texas; Section of Leukocyte Biology (H.W., C.M.B.), Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Center for Cardiovascular Disease Prevention (H.W., C.M.B.), Methodist DeBakey Heart and Vascular Center, Houston, Texas
| | - Razvan T Dadu
- Interdepartmental Program in Translational Biology and Molecular Medicine (I.M.K.), Baylor College of Medicine, Houston, Texas; Division of Atherosclerosis and Vascular Medicine (I.M.K., Y.P., R.T.D., R.C.H., H.W., C.M.B.), Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Infectious Diseases (D.E.L.), Department of Internal Medicine, UT Health, Houston, Texas; Section of Leukocyte Biology (H.W., C.M.B.), Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Center for Cardiovascular Disease Prevention (H.W., C.M.B.), Methodist DeBakey Heart and Vascular Center, Houston, Texas
| | - Dorothy E Lewis
- Interdepartmental Program in Translational Biology and Molecular Medicine (I.M.K.), Baylor College of Medicine, Houston, Texas; Division of Atherosclerosis and Vascular Medicine (I.M.K., Y.P., R.T.D., R.C.H., H.W., C.M.B.), Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Infectious Diseases (D.E.L.), Department of Internal Medicine, UT Health, Houston, Texas; Section of Leukocyte Biology (H.W., C.M.B.), Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Center for Cardiovascular Disease Prevention (H.W., C.M.B.), Methodist DeBakey Heart and Vascular Center, Houston, Texas
| | - Ron C Hoogeveen
- Interdepartmental Program in Translational Biology and Molecular Medicine (I.M.K.), Baylor College of Medicine, Houston, Texas; Division of Atherosclerosis and Vascular Medicine (I.M.K., Y.P., R.T.D., R.C.H., H.W., C.M.B.), Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Infectious Diseases (D.E.L.), Department of Internal Medicine, UT Health, Houston, Texas; Section of Leukocyte Biology (H.W., C.M.B.), Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Center for Cardiovascular Disease Prevention (H.W., C.M.B.), Methodist DeBakey Heart and Vascular Center, Houston, Texas
| | - Huaizhu Wu
- Interdepartmental Program in Translational Biology and Molecular Medicine (I.M.K.), Baylor College of Medicine, Houston, Texas; Division of Atherosclerosis and Vascular Medicine (I.M.K., Y.P., R.T.D., R.C.H., H.W., C.M.B.), Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Infectious Diseases (D.E.L.), Department of Internal Medicine, UT Health, Houston, Texas; Section of Leukocyte Biology (H.W., C.M.B.), Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Center for Cardiovascular Disease Prevention (H.W., C.M.B.), Methodist DeBakey Heart and Vascular Center, Houston, Texas
| | - Christie M Ballantyne
- Interdepartmental Program in Translational Biology and Molecular Medicine (I.M.K.), Baylor College of Medicine, Houston, Texas; Division of Atherosclerosis and Vascular Medicine (I.M.K., Y.P., R.T.D., R.C.H., H.W., C.M.B.), Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Infectious Diseases (D.E.L.), Department of Internal Medicine, UT Health, Houston, Texas; Section of Leukocyte Biology (H.W., C.M.B.), Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Center for Cardiovascular Disease Prevention (H.W., C.M.B.), Methodist DeBakey Heart and Vascular Center, Houston, Texas
| |
Collapse
|
41
|
Sarathy JP, Zuccotto F, Hsinpin H, Sandberg L, Via LE, Marriner GA, Masquelin T, Wyatt P, Ray P, Dartois V. Prediction of Drug Penetration in Tuberculosis Lesions. ACS Infect Dis 2016; 2:552-63. [PMID: 27626295 DOI: 10.1021/acsinfecdis.6b00051] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
The penetration of antibiotics in necrotic tuberculosis lesions is heterogeneous and drug-specific, but the factors underlying such differential partitioning are unknown. We hypothesized that drug binding to macromolecules in necrotic foci (or caseum) prevents passive drug diffusion through avascular caseum, a critical site of infection. Using a caseum binding assay and MALDI mass spectrometry imaging of tuberculosis drugs, we showed that binding to caseum inversely correlates with passive diffusion into the necrotic core. We developed a high-throughput assay relying on rapid equilibrium dialysis and a caseum surrogate designed to mimic the composition of native caseum. A set of 279 compounds was profiled in this assay to generate a large data set and explore the physicochemical drivers of free diffusion into caseum. Principle component analysis and modeling of the data set delivered an in silico signature predictive of caseum binding, combining 69 molecular descriptors. Among the major positive drivers of binding were high lipophilicity and poor solubility. Determinants of molecular shape such as the number of rings, particularly aromatic rings, number of sp(2) carbon counts, and volume-to-surface ratio negatively correlated with the free fraction, indicating that low-molecular-weight nonflat compounds are more likely to exhibit low caseum binding properties and diffuse effectively through caseum. To provide simple guidance in the property-based design of new compounds, a rule of thumb was derived whereby the sum of the hydrophobicity (clogP) and aromatic ring count is proportional to caseum binding. These tools can be used to ensure desirable lesion partitioning and guide the selection of optimal regimens against tuberculosis.
Collapse
Affiliation(s)
- Jansy P. Sarathy
- Public Health Research
Institute Centre, New Jersey Medical School, Rutgers, 225 Warren Street, Newark, New Jersey 07103, United States
| | - Fabio Zuccotto
- Drug Discovery Unit,
Division of Biological Chemistry and Drug Discovery, Sir James Black
Centre, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Ho Hsinpin
- Public Health Research
Institute Centre, New Jersey Medical School, Rutgers, 225 Warren Street, Newark, New Jersey 07103, United States
| | - Lars Sandberg
- Drug Discovery Unit,
Division of Biological Chemistry and Drug Discovery, Sir James Black
Centre, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Laura E. Via
- Tuberculosis
Research Section, Laboratory of Clinical Infectious Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Gwendolyn A. Marriner
- Tuberculosis
Research Section, Laboratory of Clinical Infectious Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Thierry Masquelin
- Discovery Chemistry Research, Lilly Corporate Center,
Eli Lilly and Company, 893 S. Delaware, MC/87/02/203 G17, Indianapolis, Indiana 46285, United States
| | - Paul Wyatt
- Drug Discovery Unit,
Division of Biological Chemistry and Drug Discovery, Sir James Black
Centre, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Peter Ray
- Drug Discovery Unit,
Division of Biological Chemistry and Drug Discovery, Sir James Black
Centre, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Véronique Dartois
- Public Health Research
Institute Centre, New Jersey Medical School, Rutgers, 225 Warren Street, Newark, New Jersey 07103, United States
| |
Collapse
|
42
|
Dhurga DB, Suresh K, Tan TC. Granular Formation during Apoptosis in Blastocystis sp. Exposed to Metronidazole (MTZ). PLoS One 2016; 11:e0155390. [PMID: 27471855 PMCID: PMC4966910 DOI: 10.1371/journal.pone.0155390] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/19/2016] [Accepted: 04/04/2016] [Indexed: 11/18/2022] Open
Abstract
The role and function of the granular life cycle stage in Blastocystis sp, remains uncertain despite suggestions being made that the granules are metabolic, reproductive and lipid in nature. This present study aims to understand granular formation by triggering apoptosis in Blastocystis sp. by treating them with metronidazole (MTZ). Blastocystis sp.cultures of 4 sub-types namely 1, 2, 3 and 5 when treated with 0.01 and 0.0001 mg/ml of metronidazole (MTZ) respectively showed many of the parasites to be both viable and apoptotic (VA). Treated subtype 3 isolates exhibited the highest number of granular forms i.e. 88% (p<0.001) (0.0001 mg/ml) and 69% (p<0.01) (0.01 mg/ml) respectively at the 72 h in in vitro culture compared to other subtypes. These VA forms showed distinct granules using acridine orange (AO) and 4',6-diamino-2-phenylindole (DAPI) staining with a mean per cell ranging from 5 in ST 5 to as high as 16 in ST 3. These forms showed intact mitochondria in both viable apoptotic (VA) and viable non-apoptotic (VNA) cells with a pattern of accumulation of lipid droplets corresponding to viable cells. Granular VA forms looked ultra-structurally different with prominent presence of mitochondria-like organelle (MLO) and a changed mitochondrial trans-membrane potential with thicker membrane and a highly convoluted inner membrane than the less dense non-viable apoptotic (NVA) cells. This suggests that granular formation during apoptosis is a self-regulatory mechanism to produce higher number of viable cells in response to treatment. This study directs the need to search novel chemotherapeutic approaches by incorporating these findings when developing drugs against the emerging Blastocystis sp. infections.
Collapse
Affiliation(s)
- Devi Balkrishnan Dhurga
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kumar Suresh
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Tian Chye Tan
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
43
|
Ono-Moore KD, Snodgrass RG, Huang S, Singh S, Freytag TL, Burnett DJ, Bonnel EL, Woodhouse LR, Zunino SJ, Peerson JM, Lee JY, Rutledge JC, Hwang DH. Postprandial Inflammatory Responses and Free Fatty Acids in Plasma of Adults Who Consumed a Moderately High-Fat Breakfast with and without Blueberry Powder in a Randomized Placebo-Controlled Trial. J Nutr 2016; 146:1411-9. [PMID: 27306892 PMCID: PMC4926849 DOI: 10.3945/jn.115.223909] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/10/2015] [Accepted: 05/06/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Saturated fatty acids (FAs) released from triglyceride-rich lipoproteins (TGRLs) activate Toll-like receptor 2 (TLR-2) and induce the expression of proinflammatory cytokines in monocytes. Certain plant polyphenols inhibit TLR-mediated signaling pathways. OBJECTIVE We determined whether plasma free FAs (FFAs) after a moderately high-fat (MHF, 40% kcal from fat) breakfast modulate the inflammatory status of postprandial blood, and whether blueberry intake suppresses FFA-induced inflammatory responses in healthy humans. METHODS Twenty-three volunteers with a mean ± SEM age and body mass index (in kg/m(2)) of 30 ± 3 y and 21.9 ± 0.4, respectively, consumed an MHF breakfast with either a placebo powder or 2 or 4 servings of blueberry powder in a randomized crossover design. The placebo powder was provided on the first test day and the blueberry powder doses were randomized with a 2-wk washout period. Plasma concentrations of lipids, glucose, and cytokines were determined. To determine whether FFAs derived from TGRL stimulate monocyte activation, and whether this is inhibited by blueberry intake, whole blood was treated with lipoprotein lipase (LPL). RESULTS The median concentrations of FFAs and cytokines [tumor necrosis factor-α, interleukin (IL)-6 and IL-8] in postprandial plasma (3.5 h) decreased compared with fasting plasma regardless of the blueberry intake (P < 0.001 for FFAs and P < 0.05 for cytokines). However, concentrations of FFAs and cytokines including IL-1β increased in LPL-treated whole blood compared with untreated blood samples from participants who consumed the placebo powder. Blueberry intake suppressed IL-1β and IL-6 production in LPL-treated postprandial blood compared with the placebo control when fasting changes were used as a covariate. CONCLUSIONS The plasma FFA concentration may be an important determinant affecting inflammatory cytokine production in blood. Supplementation with blueberry powder did not affect plasma FFA and cytokine concentrations; however, it attenuated the cytokine production induced by ex vivo treatment of whole blood with LPL. This trial was registered at clinicaltrials.gov as NCT01594008.
Collapse
Affiliation(s)
- Kikumi D Ono-Moore
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA;,Department of Nutrition and
| | - Ryan G Snodgrass
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA;,Department of Nutrition and
| | - Shurong Huang
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | - Shamsher Singh
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | - Tammy L Freytag
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | - Dustin J Burnett
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | | | - Leslie R Woodhouse
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | - Susan J Zunino
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA;,Department of Nutrition and
| | - Janet M Peerson
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA;,Department of Nutrition and
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| | - John C Rutledge
- Department of Internal Medicine, School of Medicine, University of California, Davis, CA; and
| | - Daniel H Hwang
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA; Department of Nutrition and
| |
Collapse
|
44
|
Abstract
Elevated levels of cholesteryl ester (CE)-enriched apoB containing plasma lipoproteins lead to increased foam cell formation, the first step in the development of atherosclerosis. Unregulated uptake of low-density lipoprotein cholesterol by circulating monocytes and other peripheral blood cells takes place through scavenger receptors and over time causes disruption in cellular cholesterol homeostasis. As lipoproteins are taken up, their CE core is hydrolyzed by liposomal lipases to generate free cholesterol (FC). FC can be either re-esterified and stored as CE droplets or shuttled to the plasma membrane for ATP-binding cassette transporter A1-mediated efflux. Because cholesterol is an essential component of all cellular membranes, some FC may be incorporated into microdomains or lipid rafts. These platforms are essential for receptor signaling and transduction, requiring rapid assembly and disassembly. ATP-binding cassette transporter A1 plays a major role in regulating microdomain cholesterol and is most efficient when lipid-poor apolipoprotein AI (apoAI) packages raft cholesterol into soluble particles that are eventually catabolized by the liver. If FC is not effluxed from the cell, it becomes esterified, CE droplets accumulate and microdomain cholesterol content becomes poorly regulated. This dysregulation leads to prolonged activation of immune cell signaling pathways, resulting in receptor oversensitization. The availability of apoAI or other amphipathic α-helix-rich apoproteins relieves the burden of excess microdomain cholesterol in immune cells allowing a reduction in immune cell proliferation and infiltration, thereby stimulating regression of foam cells in the artery. Therefore, cellular balance between FC and CE is essential for proper immune cell function and prevents chronic immune cell overstimulation and proliferation.
Collapse
Affiliation(s)
- Mary G Sorci-Thomas
- From the Division of Endocrinology, Metabolism and Clinical Nutrition, Department of Medicine and Senior Investigator, Blood Research Institute, BloodCenter of Wisconsin (M.G.S.-T.) and Department of Pharmacology and Toxicology (M.J.T.), Medical College of Wisconsin, Milwaukee, WI.
| | - Michael J Thomas
- From the Division of Endocrinology, Metabolism and Clinical Nutrition, Department of Medicine and Senior Investigator, Blood Research Institute, BloodCenter of Wisconsin (M.G.S.-T.) and Department of Pharmacology and Toxicology (M.J.T.), Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
45
|
Aung HH, Altman R, Nyunt T, Kim J, Nuthikattu S, Budamagunta M, Voss JC, Wilson D, Rutledge JC, Villablanca AC. Lipotoxic brain microvascular injury is mediated by activating transcription factor 3-dependent inflammatory and oxidative stress pathways. J Lipid Res 2016; 57:955-68. [PMID: 27087439 DOI: 10.1194/jlr.m061853] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/04/2016] [Indexed: 01/10/2023] Open
Abstract
Dysfunction of the cerebrovasculature plays an important role in vascular cognitive impairment (VCI). Lipotoxic injury of the systemic endothelium in response to hydrolyzed triglyceride-rich lipoproteins (TGRLs; TGRL lipolysis products) or a high-fat Western diet (WD) suggests similar mechanisms may be present in brain microvascular endothelium. We investigated the hypothesis that TGRL lipolysis products cause lipotoxic injury to brain microvascular endothelium by generating increased mitochondrial superoxide radical generation, upregulation of activating transcription factor 3 (ATF3)-dependent inflammatory pathways, and activation of cellular oxidative stress and apoptotic pathways. Human brain microvascular endothelial cells were treated with human TGRL lipolysis products that induced intracellular lipid droplet formation, mitochondrial superoxide generation, ATF3-dependent transcription of proinflammatory, stress response, and oxidative stress genes, as well as activation of proapoptotic cascades. Male apoE knockout mice were fed a high-fat/high-cholesterol WD for 2 months, and brain microvessels were isolated by laser capture microdissection. ATF3 gene transcription was elevated 8-fold in the hippocampus and cerebellar brain region of the WD-fed animals compared with chow-fed control animals. The microvascular injury phenotypes observed in vitro and in vivo were similar. ATF3 plays an important role in mediating brain microvascular responses to acute and chronic lipotoxic injury and may be an important preventative and therapeutic target for endothelial dysfunction in VCI.
Collapse
Affiliation(s)
- Hnin Hnin Aung
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | - Robin Altman
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | - Tun Nyunt
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | - Jeffrey Kim
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | | | - Madhu Budamagunta
- Department of Biochemistry and Molecular Medicine, School of Medicine
| | - John C Voss
- Department of Biochemistry and Molecular Medicine, School of Medicine
| | - Dennis Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
| | - John C Rutledge
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | - Amparo C Villablanca
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| |
Collapse
|
46
|
Abstract
Atherosclerosis is a complex chronic disease. The accumulation of myeloid cells in the arterial intima, including macrophages and dendritic cells (DCs), is a feature of early stages of disease. For decades, it has been known that monocyte recruitment to the intima contributes to the burden of lesion macrophages. Yet, this paradigm may require reevaluation in light of recent advances in understanding of tissue macrophage ontogeny, their capacity for self-renewal, as well as observations that macrophages proliferate throughout atherogenesis and that self-renewal is critical for maintenance of macrophages in advanced lesions. The rate of atherosclerotic lesion formation is profoundly influenced by innate and adaptive immunity, which can be regulated locally within atherosclerotic lesions, as well as in secondary lymphoid organs, the bone marrow and the blood. DCs are important modulators of immunity. Advances in the past decade have cemented our understanding of DC subsets, functions, hematopoietic origin, gene expression patterns, transcription factors critical for differentiation, and provided new tools for study of DC biology. The functions of macrophages and DCs overlap to some extent, thus it is important to reassess the contributions of each of these myeloid cells taking into account strict criteria of cell identification, ontogeny, and determine whether their key roles are within atherosclerotic lesions or secondary lymphoid organs. This review will highlight key aspect of macrophage and DC biology, summarize how these cells participate in different stages of atherogenesis and comment on complexities, controversies, and gaps in knowledge in the field.
Collapse
Affiliation(s)
- Myron I. Cybulsky
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| | - Cheolho Cheong
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| | - Clinton S. Robbins
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| |
Collapse
|
47
|
Very-low and low-density lipoproteins induce neutral lipid accumulation and impair migration in monocyte subsets. Sci Rep 2016; 6:20038. [PMID: 26821597 PMCID: PMC4731823 DOI: 10.1038/srep20038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/19/2015] [Accepted: 12/23/2015] [Indexed: 12/25/2022] Open
Abstract
Blood monocytes are heterogeneous effector cells of the innate immune system. In circulation these cells are constantly in contact with lipid-rich lipoproteins, yet this interaction is poorly characterised. Our aim was to examine the functional effect of hyperlipidaemia on blood monocytes. In the Ldlr−/− mouse monocytes rapidly accumulate cytoplasmic neutral lipid vesicles during hyperlipidaemia. Functional analysis in vivo revealed impaired monocyte chemotaxis towards peritonitis following high fat diet due to retention of monocytes in the greater omentum. In vitro assays using human monocytes confirmed neutral lipid vesicle accumulation after exposure to LDL or VLDL. Neutral lipid accumulation did not inhibit phagocytosis, endothelial adhesion, intravascular crawling and transmigration. However, lipid loading led to a migratory defect towards C5a and disruption of cytoskeletal rearrangement, including an inhibition of RHOA signaling. These data demonstrate distinct effects of hyperlipidaemia on the chemotaxis and cytoskeletal regulation of monocyte subpopulations. These data emphasise the functional consequences of blood monocyte lipid accumulation and reveal important implications for treating inflammation, infection and atherosclerosis in the context of dyslipidaemia.
Collapse
|
48
|
Chen YC, Hsu HC, Lee CM, Sun CK. Third-harmonic generation susceptibility spectroscopy in free fatty acids. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:095013. [PMID: 26405821 DOI: 10.1117/1.jbo.20.9.095013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/04/2015] [Accepted: 08/25/2015] [Indexed: 05/23/2023]
Abstract
Lipid-correlated disease such as atherosclerosis has been an important medical research topic for decades. Many new microscopic imaging techniques such as coherent anti-Stokes Raman scattering and third-harmonic generation (THG) microscopy were verified to have the capability to target lipids in vivo. In the case of THG microscopy, biological cell membranes and lipid bodies in cells and tissues have been shown as good sources of contrast with a laser excitation wavelength around 1200 nm. We report the THG excitation spectroscopy study of two pure free fatty acids including oleic acid and linoleic acid from 1090 to 1330 nm. Different pure fatty acids presented slightly-different THG χ(3) spectra. The measured peak values of THG third-order susceptibility χ(3) in both fatty acids were surprisingly found not to match completely with the resonant absorption wavelengths around 1190 to 1210 nm, suggesting possible wavelengths selection for enhanced THG imaging of lipids while avoiding laser light absorption. Along with the recent advancement in THG imaging, this new window between 1240 to 1290 nm may offer tremendous new opportunities for sensitive label-free lipid imaging in biological tissues.
Collapse
Affiliation(s)
- Yu-Cheng Chen
- National Taiwan University, Molecular Imaging Center, Taipei 10617, Taiwan
| | - Hsun-Chia Hsu
- National Taiwan University, Molecular Imaging Center, Taipei 10617, TaiwanbWashington University in Saint Louis, Department of Biomedical Engineering, Saint Louis, Missouri 63130, United States
| | - Chien-Ming Lee
- National Taiwan University, Department of Electrical Engineering and Graduate Institute of Photonics and Optoelectronics, Taipei 10617, Taiwan
| | - Chi-Kuang Sun
- National Taiwan University, Molecular Imaging Center, Taipei 10617, TaiwancNational Taiwan University, Department of Electrical Engineering and Graduate Institute of Photonics and Optoelectronics, Taipei 10617, TaiwandInstitute of Physics and Research Cen
| |
Collapse
|
49
|
Huser T, Chan J. Raman spectroscopy for physiological investigations of tissues and cells. Adv Drug Deliv Rev 2015; 89:57-70. [PMID: 26144996 DOI: 10.1016/j.addr.2015.06.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/28/2014] [Revised: 06/08/2015] [Accepted: 06/26/2015] [Indexed: 12/29/2022]
Abstract
Raman micro-spectroscopy provides a convenient non-destructive and location-specific means of probing cellular physiology and tissue physiology at sub-micron length scales. By probing the vibrational signature of molecules and molecular groups, the distribution and metabolic products of small molecules that cannot be labeled with fluorescent dyes can be analyzed. This method works well for molecular concentrations in the micro-molar range and has been demonstrated as a valuable tool for monitoring drug-cell interactions. If the small molecule of interest does not contain groups that would allow for a discrimination against cytoplasmic background signals, "labeling" of the molecule by isotope substitution or by incorporating other unique small groups, e.g. alkynes provides a stable signal even for time-lapse imaging such compounds in living cells. In this review we highlight recent progress in assessing the physiology of cells and tissue by Raman spectroscopy and imaging.
Collapse
|
50
|
Xu L, Dai Perrard X, Perrard JL, Yang D, Xiao X, Teng BB, Simon SI, Ballantyne CM, Wu H. Foamy monocytes form early and contribute to nascent atherosclerosis in mice with hypercholesterolemia. Arterioscler Thromb Vasc Biol 2015; 35:1787-97. [PMID: 26112011 DOI: 10.1161/atvbaha.115.305609] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/27/2015] [Accepted: 06/12/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To examine infiltration of blood foamy monocytes, containing intracellular lipid droplets, into early atherosclerotic lesions and its contribution to development of nascent atherosclerosis. APPROACH AND RESULTS In apoE(-/-) mice fed Western high-fat diet (WD), >10% of circulating monocytes became foamy monocytes at 3 days on WD and >20% of monocytes at 1 week. Foamy monocytes also formed early in blood of Ldlr(-/-)Apobec1(-/-) (LDb) mice on WD. Based on CD11c and CD36, mouse monocytes were categorized as CD11c(-)CD36(-), CD11c(-)CD36(+), and CD11c(+)CD36(+). The majority of foamy monocytes were CD11c(+)CD36(+), whereas most nonfoamy monocytes were CD11c(-)CD36(-) or CD11c(-)CD36(+) in apoE(-/-) mice on WD. In wild-type mice, CD11c(+)CD36(+) and CD11c(-)CD36(+), but few CD11c(-)CD36(-), monocytes took up cholesteryl ester-rich very low-density lipoproteins (CE-VLDLs) isolated from apoE(-/-) mice on WD, and CE-VLDL uptake accelerated CD11c(-)CD36(+) to CD11c(+)CD36(+) monocyte differentiation. Ablation of CD36 decreased monocyte uptake of CE-VLDLs. Intravenous injection of DiI-CE-VLDLs in apoE(-/-) mice on WD specifically labeled CD11c(+)CD36(+) foamy monocytes, which infiltrated into nascent atherosclerotic lesions and became CD11c(+) cells that were selectively localized in atherosclerotic lesions. CD11c deficiency reduced foamy monocyte infiltration into atherosclerotic lesions. Specific and consistent depletion of foamy monocytes (for 3 weeks) by daily intravenous injections of low-dose clodrosome reduced development of nascent atherosclerosis. CONCLUSIONS Foamy monocytes, which form early in blood of mice with hypercholesterolemia, infiltrate into early atherosclerotic lesions in a CD11c-dependent manner and play crucial roles in nascent atherosclerosis development.
Collapse
Affiliation(s)
- Lu Xu
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.)
| | - Xiaoyuan Dai Perrard
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.)
| | - Jerry L Perrard
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.)
| | - Donglin Yang
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.)
| | - Xinhua Xiao
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.)
| | - Ba-Bie Teng
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.)
| | - Scott I Simon
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.)
| | - Christie M Ballantyne
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.)
| | - Huaizhu Wu
- From the Department of Medicine (L.X., X.D.P., J.L.P., D.Y., C.M.B., H.W.) and Department of Pediatrics (C.M.B., H.W.), Baylor College of Medicine, Houston, TX; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, TX (C.M.B.); Research Center for Human Genetics, Institute of Molecular Medicine, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston (B.-B.T.); Department of Biomedical Engineering, University of California, Davis (S.I.S.); and Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.X., X.X.).
| |
Collapse
|