1
|
Wang Z, Pan BS, Manne RK, Chen J, Lv D, Wang M, Tran P, Weldemichael T, Yan W, Zhou H, Martinez GM, Shao J, Hsu CC, Hromas R, Zhou D, Qin Z, Lin HK, Li HY. CD36-mediated endocytosis of proteolysis-targeting chimeras. Cell 2025:S0092-8674(25)00386-1. [PMID: 40250420 DOI: 10.1016/j.cell.2025.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 01/25/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Passive diffusion does not explain why many drugs are too large and/or too polar for rule-breaking membrane penetration, such as proteolysis-targeting chimeras (PROTACs, generally of a molecular weight > 800 Da). Here, using biotinylated chemical-probe-based target fishing and genetic knockdown/knockin approaches, we discovered that the membrane cluster of differentiation 36 (CD36) binds to and facilitates the uptake and efficacy of diverse PROTACs (e.g., SIM1-Me, MZ1, and clinical ARV-110) and large and/or polar small-molecule drugs (e.g., rapalink-1, rapamycin, navitoclax, birinapant, tubacin, and doxorubicin) via the CD36-mediated early endosome antigen 1 (EEA1)/Ras-related protein 5A (Rab5) endosomal cascade in vitro and/or in vivo. We then devised a novel chemical endocytic medicinal chemistry strategy to improve binding of PROTACs to CD36 using structural modifications via the prodrug approach, markedly enhancing PROTAC anti-tumor efficacy through spontaneously augmenting permeability and solubility.
Collapse
Affiliation(s)
- Zhengyu Wang
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Bo-Syong Pan
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
| | - Rajesh Kumar Manne
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
| | - Jungang Chen
- Department of Pathology and Winthrop P. Rockefeller Cancer Institute, School of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Dongwen Lv
- Department of Biochemistry and Structural Biology, Center for Innovative Drug Discovery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Minmin Wang
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Phuc Tran
- Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tsigereda Weldemichael
- Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wei Yan
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Hongfei Zhou
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Gloria M Martinez
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jingwei Shao
- Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Che-Chia Hsu
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
| | - Robert Hromas
- Department of Medicine and the Mays Cancer, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology, Center for Innovative Drug Discovery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Zhiqiang Qin
- Department of Pathology and Winthrop P. Rockefeller Cancer Institute, School of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Hui-Kuan Lin
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA.
| | - Hong-Yu Li
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
2
|
Yu H, Guo J, Li B, Ma J, Abebe BK, Mei C, Raza SHA, Cheng G, Zan L. Erucic acid promotes intramuscular fat deposition through the PPARγ-FABP4/CD36 pathway. Int J Biol Macromol 2025; 298:140121. [PMID: 39837435 DOI: 10.1016/j.ijbiomac.2025.140121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
The regulation of intramuscular fat (IMF) accumulation plays a crucial role in determining meat quality in the beef industry. In humans, fat deposition in skeletal muscle is closely associated with insulin resistance and obesity. However, its underlying mechanisms are not fully elucidated. We previously identified erucic acid (EA) as a key metabolite that may affect IMF deposition of beef using omics strategies. By utilizing bovine intramuscular preadipocytes in vitro, the study demonstrates a dose-dependent increase in lipid storage induced by EA, along with mRNA expression levels of transporters FABP4 and CD36. At a mechanistic level, EA triggers ERK1/2 phosphorylation and enhances the expression of PPARγ, FABP4, and CD36, thereby facilitating the formation of lipid droplets within preadipocytes. In vivo experiments conducted in mice support these findings, indicating that EA stimulates fat accumulation in skeletal muscles and enhances the levels of FABP4 and CD36 proteins. These outcomes not only enhance our comprehension of the molecular mechanisms governing IMF deposition but also provide insights into potential strategies for enhancing meat quality and addressing metabolic disorders linked to fat accumulation in skeletal muscles. The findings of the study contribute to existing scholarly knowledge and lay the groundwork for future research endeavors aimed at improving meat quality and metabolic well-being.
Collapse
Affiliation(s)
- Hengwei Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Juntao Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Bingzhi Li
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shaanxi Provinc, Yangling vocational & technical college, Yangling 712100, China
| | - Jing Ma
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Belete Kuraz Abebe
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chugang Mei
- College of Grassland Agriculture, Northwest A&F University, Yangling 712100, China; National Beef Cattle Improvement Center, Yangling 712100, China
| | - Sayed Haidar Abbas Raza
- Guangdong Provincial Key Laboratory of Food Quality and Safety / Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou 510642, China.
| | - Gong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; National Beef Cattle Improvement Center, Yangling 712100, China.
| |
Collapse
|
3
|
Mata-Cruz C, Guerrero-Rodríguez SL, Gómez-Castellano K, Carballo-Uicab G, Almagro JC, Pérez-Tapia SM, Velasco-Velázquez MA. Discovery and in vitro characterization of a human anti-CD36 scFv. Front Immunol 2025; 16:1531171. [PMID: 39967671 PMCID: PMC11832482 DOI: 10.3389/fimmu.2025.1531171] [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] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction CD36 is a membrane receptor that participates in the cellular uptake of fatty acids and lipid metabolism. CD36 overexpression favors progression of different pathologies, such as atherosclerosis and cancer. Thus, CD36 targeting has medicinal relevance. Herein, we aimed to identify human anti-CD36 single-chain variable fragment (scFv) with therapeutic potential. Methods The semisynthetic ALTHEA Gold Plus Libraries™ were panned using recombinant human CD36. Clone selection was performed by ELISA. Analysis of scFv binding and blocking function was evaluated by flow cytometry in macrophage-like THP-1 cells and hepatocellular carcinoma HepG2 cells. The phenotypic changes induced by CD36 ligands were assessed in vitro by: i) oil red staining, ii) tumorsphere assays, and iii) RT-qPCR. Results We identified an anti-CD36 scFv, called D11, that competes with a commercial anti-CD36 antibody with proven efficacy in disease models. D11 binds to CD36 expressed in the membrane of the cellular models employed and reduces the uptake of CD36 ligands. In macrophage-like THP-1 cells, D11 impaired the acquisition of foam cell phenotype induced by oxLDL, decreasing lipid droplet content and the expression of lipid metabolism genes. Treatment of HepG2 cells with D11 reduced lipid accumulation and the enhanced clonogenicity stimulated by palmitate. Conclusion We discovered a new fully human scFv that is an effective blocker of CD36. Since D11 reduces the acquisition of pathogenic features induced by CD36 ligands, it could support the generation of therapeutic proteins targeting CD36.
Collapse
Affiliation(s)
- Cecilia Mata-Cruz
- School of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Graduate Program in Biochemical Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Keyla Gómez-Castellano
- Research and Development in Biotherapeutics Unit (UDIBI), National School of Biological Sciences, National Polytechnic Institute, Mexico City, Mexico
| | - Gregorio Carballo-Uicab
- Research and Development in Biotherapeutics Unit (UDIBI), National School of Biological Sciences, National Polytechnic Institute, Mexico City, Mexico
| | - Juan Carlos Almagro
- Research and Development in Biotherapeutics Unit (UDIBI), National School of Biological Sciences, National Polytechnic Institute, Mexico City, Mexico
- GlobalBio, Inc., Cambridge, MA, United States
| | - S. Mayra Pérez-Tapia
- Research and Development in Biotherapeutics Unit (UDIBI), National School of Biological Sciences, National Polytechnic Institute, Mexico City, Mexico
- National Laboratory for Specialized Services of Investigation, Development and Innovation (I+D+i) for Pharma Chemicals and Biotechnological Products, LANSEIDI-FarBiotec-CONAHCYT, Mexico City, Mexico
- Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City, Mexico
| | | |
Collapse
|
4
|
Li J, Chen J, Yang G, Zhang S, Li P, Ye L. CD36 as a Therapeutic Target in Tumor Microenvironment and Lipid Metabolism. Anticancer Agents Med Chem 2025; 25:447-459. [PMID: 39754780 DOI: 10.2174/0118715206353634241111113338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 01/06/2025]
Abstract
Dysregulated lipid metabolism within the tumor microenvironment (TME) is a critical hallmark of cancer progression, with lipids serving as a major energy source for tumor cells. Beyond their role in cell membrane synthesis, lipids also provide essential substrates for biomolecule production and activate signaling pathways that regulate various cellular processes. Aberrant lipid metabolism impacts not only function but also alters the behavior of immune and stromal cells within the TME. CD36, a key lipid transporter, plays a crucial role in regulating fatty acid sensing and lipid metabolism, and its dysregulated expression has been associated with poor prognosis in several cancers. Studies have demonstrated that elevated CD 36 expression in the TME is closely linked to abnormal lipid metabolism, promoting tumor growth, migration, and metastasis. In recent years, significant progress has been made in developing CD36-targeted therapies, including small-molecule inhibitors, antibodies, and nanoparticle-based drugs, with many entering experimental or preclinical stages. This review comprehensively summarizes the latest advances in understanding the role of CD36 in the TME, focusing on its metabolic regulatory mechanisms in tumor cells, immune cells, and stromal cells. Additionally, it highlights the contribution of CD36 to immune evasion, drug resistance, and cancer stem cell maintenance while discussing several therapeutic strategies targeting CD36, including novel therapies currently in clinical trials. By exploring the therapeutic potential of CD36, this review provides critical insights for the future development of CD36-targeted cancer therapies.
Collapse
Affiliation(s)
- Jiaxuan Li
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Jiaqi Chen
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Guang Yang
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Shulin Zhang
- School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Peiyao Li
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Lan Ye
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| |
Collapse
|
5
|
Najjar SM, Shively JE. Regulation of lipid storage and inflammation in the liver by CEACAM1. Eur J Clin Invest 2024; 54 Suppl 2:e14338. [PMID: 39674882 DOI: 10.1111/eci.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/11/2024] [Indexed: 12/17/2024]
Abstract
This review focuses on a special aspect of hepatic lipid storage and inflammation that occurs during nutritional excess in obesity. Mounting evidence supports that prolonged excess fatty acid (FA) uptake in the liver is strongly associated with hepatic lipid storage and inflammation and that the two processes are closely linked by a homeostatic mechanism. There is also strong evidence that bacterial lipids may enter the gut by a common mechanism with lipid absorption and that there is a set point to determine when their uptake triggers an inflammatory response in the liver. In fact, the progression from high uptake of FAs in the liver resulting in Metabolic dysfunction-associated steatotic liver disease (MASLD) to the development of the more serious Metabolic dysfunction-associated steatohepatitis (MASH) depends on the degree of inflammation and its progression from an acute to a chronic state. Thus, MASLD/MASH implicates both excess fatty acids and progressive inflammation in the aetiology of liver disease. We start the discussion by introduction of CD36, a major player in FA and lipopolysaccharide (LPS) uptake in the duodenum, liver and adipose tissue. We will then introduce CEACAM1, a major player in the regulation of hepatic de novo lipogenesis and the inflammatory response in the liver, and its dual association with CD36 in enterocytes and hepatocytes. We conclude that CEACAM1 and CD36 together regulate lipid droplet formation and inflammation in the liver.
Collapse
Affiliation(s)
- Sonia M Najjar
- Department of Biomedical Sciences and the Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - John E Shively
- Department of Immunology and Theranostics, Arthur D. Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
6
|
Ajoolabady A, Pratico D, Lin L, Mantzoros CS, Bahijri S, Tuomilehto J, Ren J. Inflammation in atherosclerosis: pathophysiology and mechanisms. Cell Death Dis 2024; 15:817. [PMID: 39528464 PMCID: PMC11555284 DOI: 10.1038/s41419-024-07166-8] [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] [Scholar Register] [Received: 02/21/2024] [Revised: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Atherosclerosis imposes a heavy burden on cardiovascular health due to its indispensable role in the pathogenesis of cardiovascular disease (CVD) such as coronary artery disease and heart failure. Ample clinical and experimental evidence has corroborated the vital role of inflammation in the pathophysiology of atherosclerosis. Hence, the demand for preclinical research into atherosclerotic inflammation is on the horizon. Indeed, the acquisition of an in-depth knowledge of the molecular and cellular mechanisms of inflammation in atherosclerosis should allow us to identify novel therapeutic targets with translational merits. In this review, we aimed to critically discuss and speculate on the recently identified molecular and cellular mechanisms of inflammation in atherosclerosis. Moreover, we delineated various signaling cascades and proinflammatory responses in macrophages and other leukocytes that promote plaque inflammation and atherosclerosis. In the end, we highlighted potential therapeutic targets, the pros and cons of current interventions, as well as anti-inflammatory and atheroprotective mechanisms.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ling Lin
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | | | - Suhad Bahijri
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Jaakko Tuomilehto
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Public Health, University of Helsinki, Helsinki, Finland.
- Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
7
|
Chung A, Chang HK, Pan H, Bashore AC, Shuck K, Matias CV, Gomez J, Yan H, Li M, Bauer RC. ADAMTS7 Promotes Smooth Muscle Foam Cell Expansion in Atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582156. [PMID: 38463994 PMCID: PMC10925101 DOI: 10.1101/2024.02.26.582156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Human genetic studies have repeatedly associated ADAMTS7 with atherosclerotic cardiovascular disease. Subsequent investigations in mice demonstrated that ADAMTS7 is proatherogenic and induced in response to vascular injury and that the proatherogenicity of ADAMTS7, a secreted protein, is due to its catalytic activity. However, the cell-specific mechanisms governing ADAMTS7 proatherogenicity remain unclear. To determine which vascular cell types express ADAMTS7, we interrogated single-cell RNA sequencing of human carotid atherosclerosis and found ADAMTS7 expression in smooth muscle cells (SMCs), endothelial cells (ECs), and fibroblasts. We subsequently created SMC- and EC-specific Adamts7 conditional knockout and transgenic mice. Conditional knockout of Adamts7 in either cell type is insufficient to reduce atherosclerosis, whereas transgenic induction in either cell type increases atherosclerosis. In SMC transgenic mice, this increase coincides with an expansion of lipid-laden SMC foam cells and decreased fibrous cap formation. RNA-sequencing in SMCs revealed an upregulation of lipid uptake genes typically assigned to macrophages. Subsequent experiments demonstrated that ADAMTS7 increases SMC oxLDL uptake through increased CD36 levels. Furthermore, Cd36 expression is increased due to increased levels of PU.1, a transcription factor typically associated with myeloid fate determination. In summary, Adamts7 expression in either SMCs or ECs promotes SMC foam cell formation and atherosclerosis. In SMCs, ADAMTS7 promotes oxLDL uptake via increased PU.1 and Cd36 expression, thereby increasing SMC foam cell formation and atherosclerosis.
Collapse
|
8
|
Xia Q, Liu F, Zhou Y, Yang G, Li F, Liang T, Liu J, Li W, Huang Y, Zhu C. CD47-SIRPα signaling-inspired engineered monocytes for preventing the progression of atherosclerotic plaques. Mater Today Bio 2024; 28:101178. [PMID: 39211288 PMCID: PMC11357865 DOI: 10.1016/j.mtbio.2024.101178] [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] [Scholar Register] [Received: 05/16/2024] [Revised: 07/14/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
The accumulation of foam cells in the subendothelial space of the vascular wall to form plaques is the real cause of atherosclerotic lesions. Conventional interventions, such as statins and anti-cytokine or anti-inflammatory therapies, suffer problems in terms of their short therapeutic outcomes and potential disruption of the immune system. The development of more efficient therapeutics to restrict the initial progression of plaques appears to be crucial for treating and preventing atherosclerosis. Decreasing foam cell formation by reversing the excessive phagocytosis of modified low-density lipoprotein (LDL) in macrophages is highly desirable. Here, we developed a strategy based on engineered monocytes to dynamically regulate lipid uptake by macrophages inspired by a CD47-SIRPα signaling-induced defect in the phagocytosis of lesional macrophages at the advanced stage of AS. Briefly, a complex called CD47p-GQDs-miR223, which is designed to interact with SIRPα, was synthesized to remodel monocytes by decreasing the uptake of oxidized LDL through the activation of CD47-SIRPα signaling. After injection, these monocytes compete for recruitment to atherosclerotic plaques, release gene drugs and mediate anti-inflammatory phenotypic remodeling of the aboriginal macrophages, effectively inhibiting the development of foam cells. Our strategy provides a new therapeutic for preventing the progression of atherosclerosis.
Collapse
Affiliation(s)
- Qing Xia
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Guanyuan Yang
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, National and Regional Engineering Laboratory of Tissue Engineering, Third Military Medical University, Chongqing, 400038, China
| | - Fangzhou Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Tingting Liang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Wanling Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yaqing Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Chuhong Zhu
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, National and Regional Engineering Laboratory of Tissue Engineering, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
9
|
Jawabri AA, John A, Ghattas MA, Mahgoub RE, Hamad MIK, Barakat MT, Shobi B, Daggag H, Ali BR. Cellular and functional evaluation of LDLR missense variants reported in hypercholesterolemic patients demonstrates their hypomorphic impacts on trafficking and LDL internalization. Front Cell Dev Biol 2024; 12:1412236. [PMID: 39114568 PMCID: PMC11303217 DOI: 10.3389/fcell.2024.1412236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/10/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is an autosomal dominant disorder characterized by increased LDL-cholesterol levels. About 85% of FH cases are caused by LDLR mutations encoding the low-density lipoprotein receptor (LDLR). LDLR is synthesized in the endoplasmic reticulum (ER) where it undergoes post-translational modifications and then transported through Golgi apparatus to the plasma membrane. Over 2900 LDLR variants have been reported in FH patients with limited information on the pathogenicity and functionality of many of them. This study aims to elucidate the cellular trafficking and functional implications of LDLR missense variants identified in suspected FH patients using biochemical and functional methods. METHODS We used HeLa, HEK293T, and LDLR-deficient-CHO-ldlA7 cells to evaluate the subcellular localization and LDL internalization of ten LDLR missense variants (p.C167F, p.D178N, p.C243Y, p.E277K, p.G314R, p.H327Y, p.D477N, p.D622G, p.R744Q, and p.R814Q) reported in multiethnic suspected FH patients. We also analyzed the functional impact of three variants (p.D445E, p.D482H, and p.C677F), two of which previously shown to be retained in the ER. RESULTS We show that p.D622G, p.D482H, and p.C667F are largely retained in the ER whereas p.R744Q is partially retained. The other variants were predominantly localized to the plasma membrane. LDL internalization assays in CHO-ldlA7 cells indicate that p.D482H, p.C243Y, p.D622G, and p.C667F have quantitatively lost their ability to internalize Dil-LDL with the others (p.C167F, p.D178N, p.G314R, p.H327Y, p.D445E, p.D477N, p.R744Q and p.R814Q) showing significant losses except for p.E277K which retained full activity. However, the LDL internalization assay is only to able evaluate the impact of the variants on LDL internalization and not the exact functional defects such as failure to bind LDL. The data represented illustrate the hypomorphism nature of variants causing FH which may explain some of the variable expressivity of FH. CONCLUSION Our combinatorial approach of in silico, cellular, and functional analysis is a powerful strategy to determine pathogenicity and FH disease mechanisms which may provide opportunitites for novel therapeutic strategies.
Collapse
Affiliation(s)
- Aseel A. Jawabri
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Anne John
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | | | - Radwa E. Mahgoub
- College of Pharmacy, Al-Ain University, Abu Dhabi, United Arab Emirates
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Maha T. Barakat
- Research Institute, Imperial College London Diabetes Centre (ICLDC), Abu Dhabi, United Arab Emirates
| | - Bindu Shobi
- Research Institute, Imperial College London Diabetes Centre (ICLDC), Abu Dhabi, United Arab Emirates
| | - Hinda Daggag
- Research Institute, Imperial College London Diabetes Centre (ICLDC), Abu Dhabi, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
10
|
Zheng Y, Feng J, Ling M, Yu Y, Tao Y, Wang X. A comprehensive review on targeting cluster of differentiation: An attractive strategy for inhibiting viruses through host proteins. Int J Biol Macromol 2024; 269:132200. [PMID: 38723834 DOI: 10.1016/j.ijbiomac.2024.132200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/20/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Viral infections continue to pose a significant global public health threat. Targeting host proteins, such as cluster of differentiation (CD) macromolecules, may offer a promising alternative approach to developing antiviral treatments. CDs are cell-surface biological macromolecules mainly expressed on leukocytes that viruses can use to enter cells, thereby evading immune detection and promoting their replication. The manipulation of CDs by viruses may represent an effective and clever means of survival through the prolonged co-evolution of hosts and viruses. Targeting of CDs is anticipated to hinder the invasion of related viruses, modulate the body's immune system, and diminish the incidence of subsequent inflammation. They have become crucial for biomedical diagnosis, and some have been used as valuable tools for resisting viral infections. However, a summary of the structures and functions of CDs involved in viral infection is currently lacking. The development of drugs targeting these biological macromolecules is restricted both in terms of their availability and the number of compounds currently identified. This review provides a comprehensive analysis of the critical role of CD proteins in virus invasion and a list of relevant targeted antiviral agents, which will serve as a valuable reference for future research in this field.
Collapse
Affiliation(s)
- Youle Zheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Min Ling
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yixin Yu
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yanfei Tao
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
11
|
Bao Q, Zhang B, Zhou L, Yang Q, Mu X, Liu X, Zhang S, Yuan M, Zhang Y, Che J, Wei W, Liu T, Li G, He J. CNP Ameliorates Macrophage Inflammatory Response and Atherosclerosis. Circ Res 2024; 134:e72-e91. [PMID: 38456298 DOI: 10.1161/circresaha.123.324086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND CNP (C-type natriuretic peptide), an endogenous short peptide in the natriuretic peptide family, has emerged as an important regulator to govern vascular homeostasis. However, its role in the development of atherosclerosis remains unclear. This study aimed to investigate the impact of CNP on the progression of atherosclerotic plaques and elucidate its underlying mechanisms. METHODS Plasma CNP levels were measured in patients with acute coronary syndrome. The potential atheroprotective role of CNP was evaluated in apolipoprotein E-deficient (ApoE-/-) mice through CNP supplementation via osmotic pumps, genetic overexpression, or LCZ696 administration. Various functional experiments involving CNP treatment were performed on primary macrophages derived from wild-type and CD36 (cluster of differentiation 36) knockout mice. Proteomics and multiple biochemical analyses were conducted to unravel the underlying mechanism. RESULTS We observed a negative correlation between plasma CNP concentration and the burden of coronary atherosclerosis in patients. In early atherosclerotic plaques, CNP predominantly accumulated in macrophages but significantly decreased in advanced plaques. Supplementing CNP via osmotic pumps or genetic overexpression ameliorated atherosclerotic plaque formation and enhanced plaque stability in ApoE-/- mice. CNP promoted an anti-inflammatory macrophage phenotype and efferocytosis and reduced foam cell formation and necroptosis. Mechanistically, we found that CNP could accelerate HIF-1α (hypoxia-inducible factor 1-alpha) degradation in macrophages by enhancing the interaction between PHD (prolyl hydroxylase domain-containing protein) 2 and HIF-1α. Furthermore, we observed that CD36 bound to CNP and mediated its endocytosis in macrophages. Moreover, we demonstrated that the administration of LCZ696, an orally bioavailable drug recently approved for treating chronic heart failure with reduced ejection fraction, could amplify the bioactivity of CNP and ameliorate atherosclerotic plaque formation. CONCLUSIONS Our study reveals that CNP enhanced plaque stability and alleviated macrophage inflammatory responses by promoting HIF-1α degradation, suggesting a novel atheroprotective role of CNP. Enhancing CNP bioactivity may offer a novel pharmacological strategy for treating related diseases.
Collapse
Affiliation(s)
- Qiankun Bao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Bangying Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Lu Zhou
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Qian Yang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Xiaofeng Mu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Xing Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Shiying Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Meng Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Yue Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Jingjin Che
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Wen Wei
- Center for Mechanisms of Evolution, Biodesign Institute, Arizona State University, Tempe (W.W.)
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, China (J.H.)
| |
Collapse
|
12
|
Park JH, Kwon S, Park YM. Extracellular Vimentin Alters Energy Metabolism And Induces Adipocyte Hypertrophy. Diabetes Metab J 2024; 48:215-230. [PMID: 37750184 PMCID: PMC10995492 DOI: 10.4093/dmj.2022.0332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 06/19/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGRUOUND Previous studies have reported that oxidative stress contributes to obesity characterized by adipocyte hypertrophy. However, mechanism has not been studied extensively. In the current study, we evaluated role of extracellular vimentin secreted by oxidized low-density lipoprotein (oxLDL) in energy metabolism in adipocytes. METHODS We treated 3T3-L1-derived adipocytes with oxLDL and measured vimentin which was secreted in the media. We evaluated changes in uptake of glucose and free fatty acid, expression of molecules functioning in energy metabolism, synthesis of adenosine triphosphate (ATP) and lactate, markers for endoplasmic reticulum (ER) stress and autophagy in adipocytes treated with recombinant vimentin. RESULTS Adipocytes secreted vimentin in response to oxLDL. Microscopic evaluation revealed that vimentin treatment induced increase in adipocyte size and increase in sizes of intracellular lipid droplets with increased intracellular triglyceride. Adipocytes treated with vimentin showed increased uptake of glucose and free fatty acid with increased expression of plasma membrane glucose transporter type 1 (GLUT1), GLUT4, and CD36. Vimentin treatment increased transcription of GLUT1 and hypoxia-inducible factor 1α (Hif-1α) but decreased GLUT4 transcription. Adipose triglyceride lipase (ATGL), peroxisome proliferator-activated receptor γ (PPARγ), sterol regulatory element-binding protein 1 (SREBP1), diacylglycerol O-acyltransferase 1 (DGAT1) and 2 were decreased by vimentin treatment. Markers for ER stress were increased and autophagy was impaired in vimentin-treated adipocytes. No change was observed in synthesis of ATP and lactate in the adipocytes treated with vimentin. CONCLUSION We concluded that extracellular vimentin regulates expression of molecules in energy metabolism and promotes adipocyte hypertrophy. Our results show that vimentin functions in the interplay between oxidative stress and metabolism, suggesting a mechanism by which adipocyte hypertrophy is induced in oxidative stress.
Collapse
Affiliation(s)
- Ji-Hae Park
- Department of Medicine, Graduate School, Ewha Womans University, Seoul, Korea
| | - Soyeon Kwon
- Department of Medicine, Graduate School, Ewha Womans University, Seoul, Korea
| | - Young Mi Park
- Department of Medicine, Graduate School, Ewha Womans University, Seoul, Korea
- Department of Molecular Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Ashaq MS, Zhang S, Xu M, Li Y, Zhao B. The regulatory role of CD36 in hematopoiesis beyond fatty acid uptake. Life Sci 2024; 339:122442. [PMID: 38244916 DOI: 10.1016/j.lfs.2024.122442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/07/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
CD36 is a transmembrane glycoprotein, located on surface of numerous cell types. This review is aimed to explore regulatory role of CD36 in hematopoiesis beyond fatty acid uptake. CD36 acts as a pattern recognition receptor, regulates cellular fatty acid homeostasis, and negatively monitors angiogenesis. CD36 also mediates free fatty acid transportation to hematopoietic stem cells in response to infections. During normal physiology and pathophysiology, CD36 significantly participates in the activation and metabolic needs of platelets, macrophages, monocytes, T cells, B cells, and dendritic cells. CD36 has shown a unique relationship with Plasmodium falciparum-infected erythrocytes (PfIEs) as a beneficiary for both parasite and host. CD36 actively participates in pathogenesis of various hematological cancers as a significant prognostic biomarker including AML, HL, and NHL. CD36-targeting antibodies, CD36 antagonists (small molecules), and CD36 expression inhibitors/modulators are used to target CD36, depicting its therapeutic potential. Many preclinical studies or clinical trials were performed to assess CD36 as a therapeutic target; some are still under investigation. This review reflects the role of CD36 in hematopoiesis which requires more consideration in future research.
Collapse
Affiliation(s)
- Muhammad Sameer Ashaq
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shujing Zhang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Miaomiao Xu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuan Li
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Baobing Zhao
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
14
|
Asoudeh M, Nguyen N, Raith M, Denman DS, Anozie UC, Mokhtarnejad M, Khomami B, Skotty KM, Isaac S, Gebhart T, Vaigneur L, Gelgie A, Dego OK, Freeman T, Beever J, Dalhaimer P. PEGylated nanoparticles interact with macrophages independently of immune response factors and trigger a non-phagocytic, low-inflammatory response. J Control Release 2024; 366:282-296. [PMID: 38123071 PMCID: PMC10922886 DOI: 10.1016/j.jconrel.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Poly-ethylene-glycol (PEG)-based nanoparticles (NPs) - including cylindrical micelles (CNPs), spherical micelles (SNPs), and PEGylated liposomes (PLs) - are hypothesized to be cleared in vivo by opsonization followed by liver macrophage phagocytosis. This hypothesis has been used to explain the rapid and significant localization of NPs to the liver after administration into the mammalian vasculature. Here, we show that the opsonization-phagocytosis nexus is not the major factor driving PEG-NP - macrophage interactions. First, mouse and human blood proteins had insignificant affinity for PEG-NPs. Second, PEG-NPs bound macrophages in the absence of serum proteins. Third, lipoproteins blocked PEG-NP binding to macrophages. Because of these findings, we tested the postulate that PEG-NPs bind (apo)lipoprotein receptors. Indeed, PEG-NPs triggered an in vitro macrophage transcription program that was similar to that triggered by lipoproteins and different from that triggered by lipopolysaccharide (LPS) and group A Streptococcus. Unlike LPS and pathogens, PLs did not increase transcripts involved in phagocytosis or inflammation. High-density lipoprotein (HDL) and SNPs triggered remarkably similar mouse bone-marrow-derived macrophage transcription programs. Unlike opsonized pathogens, CNPs, SNPs, and PLs lowered macrophage autophagosome levels and either reduced or did not increase the secretion of key macrophage pro-inflammatory cytokines and chemokines. Thus, the sequential opsonization and phagocytosis process is likely a minor aspect of PEG-NP - macrophage interactions. Instead, PEG-NP interactions with (apo)lipoprotein and scavenger receptors appear to be a strong driving force for PEG-NP - macrophage binding, entry, and downstream effects. We hypothesize that the high presence of these receptors on liver macrophages and on liver sinusoidal endothelial cells is the reason PEG-NPs localize rapidly and strongly to the liver.
Collapse
Affiliation(s)
- Monireh Asoudeh
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Nicole Nguyen
- School of Medical Laboratory Science, University of Tennessee Medical Center, Knoxville, TN 37996, USA
| | - Mitch Raith
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Desiree S Denman
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Uche C Anozie
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Mahshid Mokhtarnejad
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Bamin Khomami
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Kaitlyn M Skotty
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Sami Isaac
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | - Aga Gelgie
- Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | | | - Trevor Freeman
- Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Jon Beever
- Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Paul Dalhaimer
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA.
| |
Collapse
|
15
|
Tsuzuki S, Sugawara T, Manabe Y, Yamasaki M, Sasaki T. Binding Interaction Between Lauric Acid and Cluster of Differentiation 36 Underpinned by a Fluorescence- Intensifying Assay. J Oleo Sci 2024; 73:113-119. [PMID: 38171727 DOI: 10.5650/jos.ess23156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
Abstract
Cluster of differentiation 36 (CD36) is a scavenger receptor expressed in various vertebrate cells that contains diverse ligands, including long-chain fatty acids. This receptor has recently been suggested as a captor of specific volatile odorants (e.g., aliphatic acetates) in the mammalian nasal epithelium. This study used a fluorescence-intensifying assay to produce the first evidence that lauric acid, an odorous fatty acid, directly binds to CD36. This expansion of the repertoire of volatile ligands supports potential applications for nasal CD36. Our present findings could promote future research aimed at understanding the mechanisms of fatty acid interactions with CD36.
Collapse
Affiliation(s)
- Satoshi Tsuzuki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| | - Tatsuya Sugawara
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| | - Yuki Manabe
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| | - Masayuki Yamasaki
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Ryukoku University
| | - Tsutomu Sasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| |
Collapse
|
16
|
Wang H, Wang L, Liu Y, Men W, Hao W, Fang C, Li C, Zhang L. Plasma levels of CD36 and glutathione as biomarkers for ruptured intracranial aneurysm. Open Life Sci 2023; 18:20220757. [PMID: 38196515 PMCID: PMC10775171 DOI: 10.1515/biol-2022-0757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/18/2023] [Accepted: 09/24/2023] [Indexed: 01/11/2024] Open
Abstract
Evidence has proved that intracranial aneurysm (IA) formation and rupture might be closely related to inflammatory response and oxidative stress. Our objective was to evaluate the potential of CD36 and glutathione (GSH) as biomarkers for IA. In this study, the enzyme-linked immunosorbent assay was used to measure the plasma levels of CD36 and GSH in 30 IA patients and 30 healthy controls. Then, correlation analysis, receiver operating characteristic (ROC) curve, and logistic regression analysis were performed. The results showed that the plasma level of CD36 in IA patients was significantly higher than that in the control group (P < 0.0001), and plasma GSH was significantly lower compared with that in the control group (P < 0.0001). ROC analysis showed that CD36 and GSH had high sensitivity (90.0 and 96.6%) and specificity (96.6 and 86.6%) for IA diagnosis. The combined sensitivity and specificity achieved were 100 and 100%, respectively. The plasma levels of CD36 and GSH did not show a significant correlation with age, the Glasgow Coma Scale, Hunter-Hess score, aneurysm size, aneurysm height, aneurysm neck, and aspect ratio. The AUC of the logistic regression model based on CD36 and GSH was 0.505. Our results suggested that the combination of plasma CD36 and GSH could serve as potential biomarkers for IA rupture.
Collapse
Affiliation(s)
- Hanbin Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Luxuan Wang
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Yunmei Liu
- Department of Reproductive Medicine, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Weidong Men
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Wanjiao Hao
- Department of Reproductive Medicine, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Chuan Fang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Chunhui Li
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Lijian Zhang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| |
Collapse
|
17
|
Pope LE, Dixon SJ. Regulation of ferroptosis by lipid metabolism. Trends Cell Biol 2023; 33:1077-1087. [PMID: 37407304 PMCID: PMC10733748 DOI: 10.1016/j.tcb.2023.05.003] [Citation(s) in RCA: 195] [Impact Index Per Article: 97.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 07/07/2023]
Abstract
Ferroptosis is an iron-dependent lethal mechanism that can be activated in disease and is a proposed target for cancer therapy. Ferroptosis is defined by the overwhelming accumulation of membrane lipid peroxides. Ferroptotic lipid peroxidation is initiated on internal membranes and then appears at the plasma membrane, triggering lethal ion imbalances and membrane permeabilization. Sensitivity to ferroptosis is governed by the levels of peroxidizable polyunsaturated lipids and associated lipid metabolic enzymes. A different network of enzymes and endogenous metabolites restrains lipid peroxidation by interfering with the initiation or propagation of this process. This emerging understanding is informing new approaches to treat disease by modulating lipid metabolism to enhance or inhibit ferroptosis.
Collapse
Affiliation(s)
- Lauren E Pope
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
18
|
Boohar RT, Vandepas LE, Traylor-Knowles N, Browne WE. Phylogenetic and Protein Structure Analyses Provide Insight into the Evolution and Diversification of the CD36 Domain "Apex" among Scavenger Receptor Class B Proteins across Eukarya. Genome Biol Evol 2023; 15:evad218. [PMID: 38035778 PMCID: PMC10715195 DOI: 10.1093/gbe/evad218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 11/07/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023] Open
Abstract
The cluster of differentiation 36 (CD36) domain defines the characteristic ectodomain associated with class B scavenger receptor (SR-B) proteins. In bilaterians, SR-Bs play critical roles in diverse biological processes including innate immunity functions such as pathogen recognition and apoptotic cell clearance, as well as metabolic sensing associated with fatty acid uptake and cholesterol transport. Although previous studies suggest this protein family is ancient, SR-B diversity across Eukarya has not been robustly characterized. We analyzed SR-B homologs identified from the genomes and transcriptomes of 165 diverse eukaryotic species. The presence of highly conserved amino acid motifs across major eukaryotic supergroups supports the presence of a SR-B homolog in the last eukaryotic common ancestor. Our comparative analyses of SR-B protein structure identify the retention of a canonical asymmetric beta barrel tertiary structure within the CD36 ectodomain across Eukarya. We also identify multiple instances of independent lineage-specific sequence expansions in the apex region of the CD36 ectodomain-a region functionally associated with ligand-sensing. We hypothesize that a combination of both sequence expansion and structural variation in the CD36 apex region may reflect the evolution of SR-B ligand-sensing specificity between diverse eukaryotic clades.
Collapse
Affiliation(s)
- Reed T Boohar
- Department of Biology, University of Miami, Coral Gables, Florida, USA
| | - Lauren E Vandepas
- Department of Biology, University of Miami, Coral Gables, Florida, USA
| | - Nikki Traylor-Knowles
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, Florida, USA
| | - William E Browne
- Department of Biology, University of Miami, Coral Gables, Florida, USA
| |
Collapse
|
19
|
Villalobos-Labra R, Liu R, Spaans F, Sáez T, Semeria Maitret T, Quon A, Sawamura T, Cooke CLM, Davidge ST. Placenta-Derived Extracellular Vesicles From Preeclamptic Pregnancies Impair Vascular Endothelial Function via Lectin-Like Oxidized LDL Receptor-1. Hypertension 2023; 80:2226-2238. [PMID: 37615097 DOI: 10.1161/hypertensionaha.123.21205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Preeclampsia is a complex syndrome that includes maternal vascular dysfunction. Syncytiotrophoblast-derived extracellular vesicles from preeclampsia placentas (preeclampsia-STBEVs) were shown to induce endothelial dysfunction, but an endothelial transmembrane mediator is still unexplored. The LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1) is a transmembrane scavenger receptor that can cause endothelial dysfunction, and its expression is increased in the endothelium of preeclampsia women. In this study, we hypothesized that LOX-1 mediates the effects of preeclampsia-STBEVs on endothelial function. METHODS Preeclampsia-STBEVs were collected by perfusion of placentas from women with preeclampsia and in vitro and ex vivo endothelial cell function were assessed. RESULTS In human umbilical vein endothelial cells, inhibition of LOX-1 with LOX-1 blocking antibody (TS20) reduced the uptake of preeclampsia-STBEVs (61.3±8.8%). TS20 prevented the activation of ERK (extracellular signal-regulated kinase, a kinase downstream of LOX-1) and reduced the activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells; 21.1±8.0%) and nitrative stress (23.2±10.3%) that was induced by preeclampsia-STBEVs. Vascular function was assessed by wire myography in isolated mesenteric arteries from pregnant rats that were incubated overnight with preeclampsia-STBEVs±TS20. TS20 prevented endothelium-dependent vasodilation impairment induced by preeclampsia-STBEVs. Nitric oxide contribution to the relaxation was reduced by preeclampsia-STBEVs, which was prevented by TS20. Superoxide dismutase or apocynin, an inhibitor of NOX (nicotinamide adenine dinucleotide phosphate oxidase), restored the impaired endothelium-dependent vasodilation in arteries exposed to preeclampsia-STBEVs. CONCLUSIONS Taken together, our findings demonstrate that LOX-1 mediates the endothelial dysfunction induced by preeclampsia-STBEVs. Our study further expands on the mechanisms that may lead to adverse outcomes in preeclampsia and proposes LOX-1 as a potential target for future interventions.
Collapse
Affiliation(s)
- Roberto Villalobos-Labra
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Escuela de Medicina sede San Felipe (R.V.-L.), Universidad de Valparaíso, Chile
| | - Ricky Liu
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tamara Sáez
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Departamento de Medicina Interna (T. Sáez), Universidad de Valparaíso, Chile
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina (T. Sáez), Universidad de Valparaíso, Chile
| | - Tamara Semeria Maitret
- Department of Laboratory Medicine and Pathology (T.S.M.), University of Alberta, Edmonton, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tatsuya Sawamura
- Departments of Molecular Pathophysiology and Life Innovation, Shinshu University, Matsumoto, Japan (T. Sawamura)
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| |
Collapse
|
20
|
Powers HR, Jenjak SE, Volkman BF, Sahoo D. Development and validation of a purification system for functional full-length human SR-B1 and CD36. J Biol Chem 2023; 299:105187. [PMID: 37625590 PMCID: PMC10509710 DOI: 10.1016/j.jbc.2023.105187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/31/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Scavenger receptor class B type 1 (SR-B1) and CD36 are both members of the class B scavenger receptor family that play important roles in lipoprotein metabolism and atherosclerotic disease. SR-B1 is the primary receptor for high-density lipoproteins, while CD36 is the receptor responsible for the internalization of oxidized low-density lipoproteins. Despite their importance, class B scavenger receptor structure has only been studied by functional domain or peptide fragments-there are currently no reports of utilizing purified full-length protein. Here we report the successful expression and purification of full-length human SR-B1 and CD36 using an Spodoptera frugiperda insect cell system. We demonstrate that both SR-B1 and CD36 retained their normal functions in Spodoptera frugiperda cells, including lipoprotein binding, lipid transport, and the formation of higher order oligomers in the plasma membrane. Purification schemes for both scavenger receptors were optimized and their purity was confirmed by SDS-PAGE. Both purified scavenger receptors were assessed for stability by thermal shift assay and shown to maintain stable melting temperatures up to 6 weeks post-purification. Microscale thermophoresis was used to demonstrate that purified SR-B1 and CD36 were able to bind their native lipoprotein ligands. Further, there was no difference in affinity of SR-B1 for high-density lipoprotein or CD36 for oxidized low-density lipoprotein, when comparing glycosylated and deglycosylated receptors. These studies mark a significant step forward in creating physiologically relevant tools to study scavenger receptor function and lay the groundwork for future functional studies and determination of receptor structure.
Collapse
Affiliation(s)
- Hayley R Powers
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Shawn E Jenjak
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Daisy Sahoo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Division of Endocrinology & Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
21
|
He Y, Liu T. Oxidized low-density lipoprotein regulates macrophage polarization in atherosclerosis. Int Immunopharmacol 2023; 120:110338. [PMID: 37210916 DOI: 10.1016/j.intimp.2023.110338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Abstract
Atherosclerosis is the pathological basis of acute cardiovascular and cerebrovascular diseases. Oxidized LDL has been recognized as a major atherogenic factor in the vessel wall for decades. A growing body of evidence suggests that oxidized LDL modulates macrophage phenotypes in atherosclerosis. This article reviews the research progress on the regulation of macrophage polarization by oxidized LDL. Mechanistically, oxidized LDL induces macrophage polarization via cell signaling, metabolic reprogramming, epigenetic regulation, and intercellular regulation. This review is expected to provide new targets for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yonghang He
- The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808, China
| | - Tingting Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, No. 42 Jiaoping Road, Tangxia Town, Dongguan City, Guangdong Province 523710, China; The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
22
|
Nóbrega-Pereira S, Santos F, Oliveira Santos M, Serafim TL, Lopes AP, Coutinho D, Carvalho FS, Domingues RM, Domingues P, Bernardes de Jesus B, Morais VA, Dias S. Mitochondrial Metabolism Drives Low-density Lipoprotein-induced Breast Cancer Cell Migration. CANCER RESEARCH COMMUNICATIONS 2023; 3:709-724. [PMID: 37377750 PMCID: PMC10132314 DOI: 10.1158/2767-9764.crc-22-0394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 06/29/2023]
Abstract
Most cancer-related deaths are due to metastases. Systemic factors, such as lipid-enriched environments [as low-density lipoprotein (LDL)-cholesterol], favor breast cancer, including triple-negative breast cancer (TNBC) metastasis formation. Mitochondria metabolism impacts TNBC invasive behavior but its involvement in a lipid-enriched setting is undisclosed. Here we show that LDL increases lipid droplets, induces CD36 and augments TNBC cells migration and invasion in vivo and in vitro. LDL induces higher mitochondrial mass and network spread in migrating cells, in an actin remodeling-dependent manner, and transcriptomic and energetic analyses revealed that LDL renders TNBC cells dependent on fatty acids (FA) usage for mitochondrial respiration. Indeed, engagement on FA transport into the mitochondria is required for LDL-induced migration and mitochondrial remodeling. Mechanistically, LDL treatment leads to mitochondrial long-chain fatty acid accumulation and increased reactive oxygen species (ROS) production. Importantly, CD36 or ROS blockade abolished LDL-induced cell migration and mitochondria metabolic adaptations. Our data suggest that LDL induces TNBC cells migration by reprogramming mitochondrial metabolism, revealing a new vulnerability in metastatic breast cancer. Significance LDL induces breast cancer cell migration that relies on CD36 for mitochondrial metabolism and network remodeling, providing an antimetastatic metabolic strategy.
Collapse
Affiliation(s)
- Sandrina Nóbrega-Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Instituto de Biomedicina (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Francisco Santos
- Instituto de Biomedicina (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Miguel Oliveira Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Teresa L. Serafim
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Ana Patrícia Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Diogo Coutinho
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Filipa S. Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Rosário M. Domingues
- Mass Spectrometry Center, QOPNA, University of Aveiro, Aveiro, Portugal
- Department of Chemistry and CESAM&ECOMARE, University of Aveiro, Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Center, QOPNA, University of Aveiro, Aveiro, Portugal
| | - Bruno Bernardes de Jesus
- Instituto de Biomedicina (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Vanessa A. Morais
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Sérgio Dias
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
23
|
Cong Y, Hong Y, Wang D, Cheng P, Wang Z, Xing C, Sun W, Xu G. 2,3,7,8-Tetrachlorodibenzo-p-dioxin induces liver lipid metabolism disorder via the ROS/AMPK/CD36 signaling pathway. Toxicol Sci 2023; 191:276-284. [PMID: 36534932 DOI: 10.1093/toxsci/kfac133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is widely considered as the most toxic and common carcinogen in the world. Exposure to TCDD causes liver lipid metabolism disorder and steatosis. However, the molecular mechanism of TCDD-induced liver lipid accumulation is not completely clear. Here, we found that a 5 μg/kg TCDD exposure for 3 weeks induced hepatocyte lipid deposition, increased CD36 expression, and promoted AMP-activated protein kinase (AMPK) ɑ phosphorylation in the liver of C57BL/6J mice. Furthermore, sulfo-N-succinimidyl oleate, a CD36 inhibiter, blunted TCDD-induced lipid deposition in Huh7 cells, confirming the critical role of CD36 in TCDD-induced hepatic steatosis. In terms of molecular mechanisms, we found that TCDD exposure increased reactive oxygen species (ROS) levels in Huh7 cells, which activated AMPK. Moreover, the activated AMPK upregulated CD36 expression. Therefore, we can see that the increase in CD36 expression induced by TCDD was regulated by ROS/AMPK/CD36 signaling pathway. Our results help to clarify the molecular mechanism of TCDD-induced hepatic steatosis.
Collapse
Affiliation(s)
- Yewen Cong
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yujing Hong
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226001, P.R. China.,Department of Clinical Nutrition, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu 226006, P.R. China
| | - Dandan Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226001, P.R. China.,Haian Center for Disease Control and Prevention, Haian, Jiangsu 226600, P.R. China
| | - Pei Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226001, P.R. China.,Department of Clinical Nutrition, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221000, P. R. China
| | - Zhisheng Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Changming Xing
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wenxing Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Guangfei Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
24
|
Tsuzuki S. A point of view on human fat olfaction - do fatty derivatives serve as cues for awareness of dietary fats? Biomed Res 2023; 44:127-146. [PMID: 37544735 DOI: 10.2220/biomedres.44.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Fat (triglycerides) consumption is critical for the survival of animals, including humans. Being able to smell fat can be advantageous in judging food value. However, fat has poor volatility; thus, olfaction of fat seems impossible. What about fatty acids that comprise fat? Humans smell and discriminate medium-chain fatty acids. However, no conclusive evidence has been provided for the olfactory sense of long-chain fatty acids, including essential acids such as linoleic acid (LA). Instead, humans likely perceive the presence of essential fatty acids through the olfaction of volatile compounds generated by their oxidative breakdown (e.g., hexanal and γ-decalactone). For some people, such scents are pleasing, especially when they come from fruit. Nonetheless, it remains unclear whether the olfaction of these volatiles leads to the recognition of fat per se. Nowadays, people often smell LA-borne aldehydes such as E,E-2,4-decadienal that occur appreciably, for example, from edible oils during deep frying, and are pronely captivated by their characteristic "fatty" note, which can be considered a "pseudo-perception" of fat. However, our preference for such LA-borne aldehyde odors may be a potential cause behind the modern overdose of n-6 fatty acids. This review aims to provide a view of whether and, if any, how we olfactorily perceive dietary fats and raises future purposes related to human fat olfaction, such as investigating sub-olfactory systems for detecting long-chain fatty acids.
Collapse
Affiliation(s)
- Satoshi Tsuzuki
- Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| |
Collapse
|
25
|
The Potential Importance of CXCL1 in the Physiological State and in Noncancer Diseases of the Cardiovascular System, Respiratory System and Skin. Int J Mol Sci 2022; 24:ijms24010205. [PMID: 36613652 PMCID: PMC9820720 DOI: 10.3390/ijms24010205] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
In this paper, we present a literature review of the role of CXC motif chemokine ligand 1 (CXCL1) in physiology, and in selected major non-cancer diseases of the cardiovascular system, respiratory system and skin. CXCL1, a cytokine belonging to the CXC sub-family of chemokines with CXC motif chemokine receptor 2 (CXCR2) as its main receptor, causes the migration and infiltration of neutrophils to the sites of high expression. This implicates CXCL1 in many adverse conditions associated with inflammation and the accumulation of neutrophils. The aim of this study was to describe the significance of CXCL1 in selected diseases of the cardiovascular system (atherosclerosis, atrial fibrillation, chronic ischemic heart disease, hypertension, sepsis including sepsis-associated encephalopathy and sepsis-associated acute kidney injury), the respiratory system (asthma, chronic obstructive pulmonary disease (COPD), chronic rhinosinusitis, coronavirus disease 2019 (COVID-19), influenza, lung transplantation and ischemic-reperfusion injury and tuberculosis) and the skin (wound healing, psoriasis, sunburn and xeroderma pigmentosum). Additionally, the significance of CXCL1 is described in vascular physiology, such as the effects of CXCL1 on angiogenesis and arteriogenesis.
Collapse
|
26
|
Martin-Perez M, Urdiroz-Urricelqui U, Bigas C, Benitah SA. The role of lipids in cancer progression and metastasis. Cell Metab 2022; 34:1675-1699. [PMID: 36261043 DOI: 10.1016/j.cmet.2022.09.023] [Citation(s) in RCA: 213] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Lipids have essential biological functions in the body (e.g., providing energy storage, acting as a signaling molecule, and being a structural component of membranes); however, an excess of lipids can promote tumorigenesis, colonization, and metastatic capacity of tumor cells. To metastasize, a tumor cell goes through different stages that require lipid-related metabolic and structural adaptations. These adaptations include altering the lipid membrane composition for invading other niches and overcoming cell death mechanisms and promoting lipid catabolism and anabolism for energy and oxidative stress protective purposes. Cancer cells also harness lipid metabolism to modulate the activity of stromal and immune cells to their advantage and to resist therapy and promote relapse. All this is especially worrying given the high fat intake in Western diets. Thus, metabolic interventions aiming to reduce lipid availability to cancer cells or to exacerbate their metabolic vulnerabilities provide promising therapeutic opportunities to prevent cancer progression and treat metastasis.
Collapse
Affiliation(s)
- Miguel Martin-Perez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08028 Barcelona, Spain.
| | - Uxue Urdiroz-Urricelqui
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Claudia Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
27
|
Goldberg AR, Ferguson M, Pal S, Cohen R, Orlicky DJ, McCullough RL, Rutkowski JM, Burchill MA, Tamburini BAJ. Oxidized low density lipoprotein in the liver causes decreased permeability of liver lymphatic- but not liver sinusoidal-endothelial cells via VEGFR-3 regulation of VE-Cadherin. Front Physiol 2022; 13:1021038. [PMID: 36338478 PMCID: PMC9626955 DOI: 10.3389/fphys.2022.1021038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/05/2022] [Indexed: 01/27/2023] Open
Abstract
The lymphatic vasculature of the liver is vital for liver function as it maintains fluid and protein homeostasis and is important for immune cell transport to the lymph node. Chronic liver disease is associated with increased expression of inflammatory mediators including oxidized low-density lipoprotein (oxLDL). Intrahepatic levels of oxLDL are elevated in nonalcoholic fatty liver disease (NAFLD), chronic hepatitis C infection (HCV), alcohol-associated liver disease (ALD), and cholestatic liver diseases. To determine if liver lymphatic function is impaired in chronic liver diseases, in which increased oxLDL has been documented, we measured liver lymphatic function in murine models of NAFLD, ALD and primary sclerosing cholangitis (PSC). We found that Mdr2-/- (PSC), Lieber-DeCarli ethanol fed (ALD) and high fat and high cholesterol diet fed (NAFLD) mice all had a significant impairment in the ability to traffic FITC labeled dextran from the liver parenchyma to the liver draining lymph nodes. Utilizing an in vitro permeability assay, we found that oxLDL decreased the permeability of lymphatic endothelial cells (LEC)s, but not liver sinusoidal endothelial cells (LSEC)s. Here we demonstrate that LECs and LSECs differentially regulate SRC-family kinases, MAPK kinase and VE-Cadherin in response to oxLDL. Furthermore, Vascular Endothelial Growth Factor (VEGF)C or D (VEGFR-3 ligands) appear to regulate VE-Cadherin expression as well as decrease cellular permeability of LECs in vitro and in vivo after oxLDL treatment. These findings suggest that oxLDL acts to impede protein transport through the lymphatics through tightening of the cell-cell junctions. Importantly, engagement of VEGFR-3 by its ligands prevents VE-Cadherin upregulation and improves lymphatic permeability. These studies provide a potential therapeutic target to restore liver lymphatic function and improve liver function.
Collapse
Affiliation(s)
- Alyssa R. Goldberg
- Department of Pediatrics, Section of Pediatric Gastroenterology, Hepatology & Nutrition. Children’s Hospital Colorado, Digestive Health Institute- Pediatric Liver Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Megan Ferguson
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Sarit Pal
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Rachel Cohen
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - David J. Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rebecca L. McCullough
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado School of Medicine, Aurora, CO, United States
| | - Joseph M. Rutkowski
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX, United States
| | - Matthew A. Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Beth A. Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
28
|
Interplay between fat cells and immune cells in bone: Impact on malignant progression and therapeutic response. Pharmacol Ther 2022; 238:108274. [DOI: 10.1016/j.pharmthera.2022.108274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022]
|
29
|
Farías MA, Diethelm-Varela B, Navarro AJ, Kalergis AM, González PA. Interplay between Lipid Metabolism, Lipid Droplets, and DNA Virus Infections. Cells 2022; 11:2224. [PMID: 35883666 PMCID: PMC9324743 DOI: 10.3390/cells11142224] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/05/2022] [Accepted: 07/09/2022] [Indexed: 12/10/2022] Open
Abstract
Lipid droplets (LDs) are cellular organelles rich in neutral lipids such as triglycerides and cholesterol esters that are coated by a phospholipid monolayer and associated proteins. LDs are known to play important roles in the storage and availability of lipids in the cell and to serve as a source of energy reserve for the cell. However, these structures have also been related to oxidative stress, reticular stress responses, and reduced antigen presentation to T cells. Importantly, LDs are also known to modulate viral infection by participating in virus replication and assembly. Here, we review and discuss the interplay between neutral lipid metabolism and LDs in the replication cycle of different DNA viruses, identifying potentially new molecular targets for the treatment of viral infections.
Collapse
Affiliation(s)
- Mónica A. Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile; (M.A.F.); (B.D.-V.); (A.J.N.); (A.M.K.)
| | - Benjamín Diethelm-Varela
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile; (M.A.F.); (B.D.-V.); (A.J.N.); (A.M.K.)
| | - Areli J. Navarro
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile; (M.A.F.); (B.D.-V.); (A.J.N.); (A.M.K.)
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile; (M.A.F.); (B.D.-V.); (A.J.N.); (A.M.K.)
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile; (M.A.F.); (B.D.-V.); (A.J.N.); (A.M.K.)
| |
Collapse
|
30
|
Qin S, Li B, Ming H, Nice EC, Zou B, Huang C. Harnessing redox signaling to overcome therapeutic-resistant cancer dormancy. Biochim Biophys Acta Rev Cancer 2022; 1877:188749. [PMID: 35716972 DOI: 10.1016/j.bbcan.2022.188749] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023]
Abstract
Dormancy occurs when cells preserve viability but stop proliferating, which is considered an important cause of tumor relapse, which may occur many years after clinical remission. Since the life cycle of dormant cancer cells is affected by both intracellular and extracellular factors, gene mutation or epigenetic regulation of tumor cells may not fully explain the mechanisms involved. Recent studies have indicated that redox signaling regulates the formation, maintenance, and reactivation of dormant cancer cells by modulating intracellular signaling pathways and the extracellular environment, which provides a molecular explanation for the life cycle of dormant tumor cells. Indeed, redox signaling regulates the onset of dormancy by balancing the intrinsic pathways, the extrinsic environment, and the response to therapy. In addition, redox signaling sustains dormancy by managing stress homeostasis, maintaining stemness and immunogenic equilibrium. However, studies on dormancy reactivation are still limited, partly explained by redox-mediated activation of lipid metabolism and the transition from the tumor microenvironment to inflammation. Encouragingly, several drug combination strategies based on redox biology are currently under clinical evaluation. Continuing to gain an in-depth understanding of redox regulation and develop specific methods targeting redox modification holds the promise to accelerate the development of strategies to treat dormant tumors and benefit cancer patients.
Collapse
Affiliation(s)
- Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Hui Ming
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Bingwen Zou
- Department of Thoracic Oncology and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China.
| |
Collapse
|
31
|
Chen Y, Zhang J, Cui W, Silverstein RL. CD36, a signaling receptor and fatty acid transporter that regulates immune cell metabolism and fate. J Exp Med 2022; 219:e20211314. [PMID: 35438721 PMCID: PMC9022290 DOI: 10.1084/jem.20211314] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/13/2022] Open
Abstract
CD36 is a type 2 cell surface scavenger receptor widely expressed in many immune and non-immune cells. It functions as both a signaling receptor responding to DAMPs and PAMPs, as well as a long chain free fatty acid transporter. Recent studies have indicated that CD36 can integrate cell signaling and metabolic pathways through its dual functions and thereby influence immune cell differentiation and activation, and ultimately help determine cell fate. Its expression along with its dual functions in both innate and adaptive immune cells contribute to pathogenesis of common diseases, including atherosclerosis and tumor progression, which makes CD36 and its downstream effectors potential therapeutic targets. This review comprehensively examines the dual functions of CD36 in a variety of immune cells, especially macrophages and T cells. We also briefly discuss CD36 function in non-immune cells, such as adipocytes and platelets, which impact the immune system via intercellular communication. Finally, outstanding questions in this field are provided for potential directions of future studies.
Collapse
Affiliation(s)
- Yiliang Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Versiti, Blood Research Institute, Milwaukee, WI
| | - Jue Zhang
- Versiti, Blood Research Institute, Milwaukee, WI
| | - Weiguo Cui
- Versiti, Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Roy L. Silverstein
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Versiti, Blood Research Institute, Milwaukee, WI
| |
Collapse
|
32
|
Lu L, Cai Y, Luo X, Wang Z, Fung SH, Jia H, Yu CL, Chan WY, Miu KK, Xiao W. A Core Omnigenic Non-coding Trait Governing Dex-Induced Osteoporotic Effects Identified Without DEXA. Front Pharmacol 2021; 12:750959. [PMID: 34899306 PMCID: PMC8651565 DOI: 10.3389/fphar.2021.750959] [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] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022] Open
Abstract
Iatrogenic glucocorticoid (GC)-induced osteoporosis (GIO) is an idiosyncratic form of secondary osteoporosis. Genetic predisposition among individuals may give rise to variant degree of phenotypic changes but there has yet been a documented unified pathway to explain the idiosyncrasy. In this study, we argue that the susceptibility to epigenetic changes governing molecular cross talks along the BMP and PI3K/Akt pathway may underline how genetic background dictate GC-induced bone loss. Concordantly, osteoblasts from BALB/c or C57BL/6 neonatal mice were treated with dexamethasone for transcriptome profiling. Furthermore, we also confirmed that GC-pre-conditioned mesenchymal stem cells (MSCs) would give rise to defective osteogenesis by instigating epigenetic changes which affected the accessibility of enhancer marks. In line with these epigenetic changes, we propose that GC modulates a key regulatory network involving the scavenger receptor Cd36 in osteoblasts pre-conditioning pharmacological idiosyncrasy in GIO.
Collapse
Affiliation(s)
- Li Lu
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanzhen Cai
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoling Luo
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhangting Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR China
| | - Sin Hang Fung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR China
| | - Huanhuan Jia
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmacy, Guangdong Pharmaceutical University, Guangzhou, China.,Guangdong Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Chi Lam Yu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR China
| | - Wai Yee Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR China
| | - Kai Kei Miu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR China
| | - Wende Xiao
- Department of Orthopedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
33
|
Lands B. Lipid nutrition: "In silico" studies and undeveloped experiments. Prog Lipid Res 2021; 85:101142. [PMID: 34818526 DOI: 10.1016/j.plipres.2021.101142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022]
Abstract
This review examines lipids and lipid-binding sites on proteins in relation to cardiovascular disease. Lipid nutrition involves food energy from ingested fatty acids plus fatty acids formed from excess ingested carbohydrate and protein. Non-esterified fatty acids (NEFA) and lipoproteins have many detailed attributes not evident in their names. Recognizing attributes of lipid-protein interactions decreases unexpected outcomes. Details of double bond position and configuration interacting with protein binding sites have unexpected consequences in acyltransferase and cell replication events. Highly unsaturated fatty acids (HUFA) have n-3 and n-6 motifs with documented differences in intensity of destabilizing positive feedback loops amplifying pathophysiology. However, actions of NEFA have been neglected relative to cholesterol, which is co-produced from excess food. Native low-density lipoproteins (LDL) bind to a high-affinity cell surface receptor which poorly recognizes biologically modified LDLs. NEFA increase negative charge of LDL and decrease its processing by "normal" receptors while increasing processing by "scavenger" receptors. A positive feedback loop in the recruitment of monocytes and macrophages amplifies chronic inflammatory pathophysiology. Computer tools combine multiple components in lipid nutrition and predict balance of energy and n-3:n-6 HUFA. The tools help design and execute precise clinical nutrition monitoring that either supports or disproves expectations.
Collapse
Affiliation(s)
- Bill Lands
- Fellow ASN, AAAS, SFRBM, ISSFAL, College Park, MD, USA.
| |
Collapse
|
34
|
Felici N, Liu D, Maret J, Restrepo M, Borovskiy Y, Hajj J, Chung W, Laudanski K. Long-Term Abnormalities of Lipid Profile After a Single Episode of Sepsis. Front Cardiovasc Med 2021; 8:674248. [PMID: 34869619 PMCID: PMC8634493 DOI: 10.3389/fcvm.2021.674248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 09/29/2021] [Indexed: 01/15/2023] Open
Abstract
Background: Acute disturbances of the lipid profile are commonplace during acute sepsis episode. However, their long-term persistence has not to be investigated despite pivotal role of dyslipidemia in several comorbidities excessively noted in sepsis survivors (stroke, cardiomyopathy). Methods: A total of 9,861 individuals hospitalized for a singular episode of sepsis between 2009 and 2019 were identified from electronic medical records. Lab measurements of total cholesterol (Tchol), high-density lipoprotein (HDL-c), low-density lipoprotein (LDL-c), very low-density lipoprotein (VLDL), triglycerides (TG), lipoprotein(a) [Lp (a)], apolipoprotein B (ApoB), and C-reactive protein (CRP). The data were examined as baseline values before sepsis, during hospitalization, and <3 months, 3-6 months, 6-12 months, 1-2 years, and more than 2 years from initial sepsis. Results: Significant reductions in HDL-c (HDLbaseline = 44.06 vs. HDLsepsis = 28.2; U = -37.79, p < 0.0001, Cohen's d = 0.22) and LDL-c serum levels were observed during and up to three months post sepsis, with females much less affected. In contrast, male subjects had derangement in HDL present for up to two years after a singular septic episode. Total cholesterol levels were slightly yet significantly elevated for up to two years after sepsis. TG were elevated up to one year [TGbaseline = 128.26 vs. TGsepsis = 170.27, t(8255) = -21.33, p < 0.0001, Cohen's d = 0.49] and normalized. Lp(a) was elevated up to two years after initial episode [Lp(a)baseline = 24.6 ± 16.06; Lp(a)sepsis-2year = 8.25 ± 5.17; Lp(a)morethan2years = 61.4 ± 40.1; ANOVA F (2, 24) = 7.39; p = 0.0032]. Response to statin therapy was blunted in sepsis survivors for several years after sepsis resolution. Significant drop-out in prescription of statins and niacin after sepsis was observed. Serum high sensitivity C-reactive protein was elevated for up to five years after sepsis resolution (H [6;1685] = 502.2; p < 0.0001). Discussion: Lipid abnormalities persisted long after the initial septic insult suggesting potential role in accelerating atherosclerosis and other abnormalities. In addition, sepsis seems to blunt statin effectiveness. Additionally, a significant and unexplained drop in statin use was seen in post-septic period. Conclusions: Our study suggests that persistent derangements of lipid profile components for up to two years after sepsis may be associated with altered risk of atherosclerosis-related events among sepsis survivors.
Collapse
Affiliation(s)
| | - Da Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Josh Maret
- College Arts and Sciences, Drexel University, Philadelphia, PA, United States
| | - Mariana Restrepo
- College Arts and Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Yuliya Borovskiy
- Corporate Informational Service, Penn Medicine, Philadelphia, PA, United States
- Data Analytics Core, Penn Medicine, Philadelphia, PA, United States
| | - Jihane Hajj
- Department of Nursing, Widener University, Chester, PA, United States
| | - Wesley Chung
- Society for HealthCare Innovation, San Francisco, CA, United States
| | - Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
- Leonard Davis Institute of Health Economics, Philadelphia, PA, United States
| |
Collapse
|
35
|
Redox Control of the Dormant Cancer Cell Life Cycle. Cells 2021; 10:cells10102707. [PMID: 34685686 PMCID: PMC8535080 DOI: 10.3390/cells10102707] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/11/2021] [Accepted: 09/28/2021] [Indexed: 02/05/2023] Open
Abstract
Following efficient tumor therapy, some cancer cells may survive through a dormancy process, contributing to tumor recurrence and worse outcomes. Dormancy is considered a process where most cancer cells in a tumor cell population are quiescent with no, or only slow, proliferation. Recent advances indicate that redox mechanisms control the dormant cancer cell life cycle, including dormancy entrance, long-term dormancy, and metastatic relapse. This regulatory network is orchestrated mainly through redox modification on key regulators or global change of reactive oxygen species (ROS) levels in dormant cancer cells. Encouragingly, several strategies targeting redox signaling, including sleeping, awaking, or killing dormant cancer cells are currently under early clinical evaluation. However, the molecular mechanisms underlying redox control of the dormant cancer cell cycle are poorly understood and need further exploration. In this review, we discuss the underlying molecular basis of redox signaling in the cell life cycle of dormant cancer and the potential redox-based targeting strategies for eliminating dormant cancer cells.
Collapse
|
36
|
Tsuzuki S, Kimoto Y, Yamasaki M, Sugawara T, Manabe Y, Inoue K, Sasaki T. Assessment of direct binding interaction between CD36 and its potential lipid ligands using a peptide mimic of the receptor labeled with a fluorophore. Biomed Res 2021; 42:181-191. [PMID: 34544994 DOI: 10.2220/biomedres.42.181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cluster of differentiation 36 (CD36) is a cell-surface receptor that recognizes diverse substances. We have presented indirect evidence that a short segment of the receptor comprising amino acids 149-168 contains a site for binding of its lipid ligands (e.g., distinct fatty acids and aldehydes). However, experimental support for their direct interactions is yet to be achieved. For this, we devised a fluorescence intensity assay, where a synthetic peptide consisting of CD36 amino acids 149-168 labeled with fluorescein isothiocyanate (FITC-CD36149-168) and its variant peptides were used as positive and negative probes, respectively. First, we obtained results indicating that 1-palmitoyl-2-(5-keto-6-octenedioyl)phosphatidylcholine (an established CD36 ligand) but not 1-palmitoyl-2-arachidonyl-phosphatidylcholine (a non-ligand of the receptor) bound in a saturable and specific manner to FITC-CD36149-168. Strikingly, the assay allowed us to provide the first evidence supporting direct and specific binding between the CD36 segment and fatty aldehydes (e.g., Z-11-hexadecenal). However, this method failed to illustrate specific interactions of the segment with fatty acids, such as oleic acid. Nonetheless, our findings offer further insight into the biologically relevant ligands and the role of CD36. In addition, we suggest that this fluorescence-based technique provides a convenient means to evaluate protein (peptide)-lipid interactions.
Collapse
Affiliation(s)
- Satoshi Tsuzuki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| | - Yusaku Kimoto
- Department of Food Science and Biotechnology, Faculty of Agriculture, Kyoto University
| | - Masayuki Yamasaki
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Ryukoku University
| | - Tatsuya Sugawara
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| | - Yuki Manabe
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| | - Tsutomu Sasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| |
Collapse
|
37
|
Shen X, Liang X, Ji X, You J, Zhuang X, Song Y, Yin H, Zhao M, Zhao L. CD36 and DGAT2 facilitate the lipid-lowering effect of chitooligosaccharides via fatty acid intake and triglyceride synthesis signaling. Food Funct 2021; 12:8681-8693. [PMID: 34351342 DOI: 10.1039/d1fo01472b] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This study examined the impact of chitobiose (GlcN)2 and chitotriose (GlcN)3 on lipid accumulation modification and their inhibitory functionalities. (GlcN)2 and (GlcN)3 significantly inhibited the total cholesterol (TC), triglyceride (TG), and low-density lipid cholesterol (LDL-c) levels in the liver of the ob/ob-/- mice fed a non-high-fat diet. This phenomenon was associated with a reduction in the mRNA and protein expression of TG synthesis and fatty acid uptake-related signaling, significantly affecting the cluster of differentiation 36 (CD36) and diacylglycerol acyltransferase 2 (DGAT2). Furthermore, the CD36 and DGAT2 genes were overexpressed by constructing a plasmid and transfecting it into HepG2 cells, after which the phenotypic traits of lipid accumulation were assessed in vitro. Consequently, it was evident that (GlcN)2 and (GlcN)3 reduced the overexpression of these proteins and relieved cellular lipid accumulation. In conclusion, these results indicated that (GlcN)2 and (GlcN)3 acted positively against NAFLD while regulating steatosis in the non-high-fat diet NAFLD model. The potential NAFLD treatment strategies, such as targeting CD36 and DGAT2 signaling, could provide scientific insight into further applying food-derived ingredients to reduce the risk of high-fat metabolism.
Collapse
Affiliation(s)
- Xin Shen
- State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China.
| | - Xinyi Liang
- State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China.
| | - Xiaoguo Ji
- State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China.
| | - Jiangshan You
- State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China.
| | - Xinye Zhuang
- State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China.
| | - Yudong Song
- State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China.
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Mengyao Zhao
- State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China.
| | - Liming Zhao
- State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China. and Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China
| |
Collapse
|
38
|
Noels H, Lehrke M, Vanholder R, Jankowski J. Lipoproteins and fatty acids in chronic kidney disease: molecular and metabolic alterations. Nat Rev Nephrol 2021; 17:528-542. [PMID: 33972752 DOI: 10.1038/s41581-021-00423-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) induces modifications in lipid and lipoprotein metabolism and homeostasis. These modifications can promote, modulate and/or accelerate CKD and secondary cardiovascular disease (CVD). Lipid and lipoprotein abnormalities - involving triglyceride-rich lipoproteins, LDL and/or HDL - not only involve changes in concentration but also changes in molecular structure, including protein composition, incorporation of small molecules and post-translational modifications. These alterations modify the function of lipoproteins and can trigger pro-inflammatory and pro-atherogenic processes, as well as oxidative stress. Serum fatty acid levels are also often altered in patients with CKD and lead to changes in fatty acid metabolism - a key process in intracellular energy production - that induce mitochondrial dysfunction and cellular damage. These fatty acid changes might not only have a negative impact on the heart, but also contribute to the progression of kidney damage. The presence of these lipoprotein alterations within a biological environment characterized by increased inflammation and oxidative stress, as well as the competing risk of non-atherosclerotic cardiovascular death as kidney function declines, has important therapeutic implications. Additional research is needed to clarify the pathophysiological link between lipid and lipoprotein modifications, and kidney dysfunction, as well as the genesis and/or progression of CVD in patients with kidney disease.
Collapse
Affiliation(s)
- Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Michael Lehrke
- Department of Internal Medicine I, RWTH Aachen University, University Hospital, Aachen, Germany
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Pediatrics, University Hospital, Ghent, Belgium
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany.
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
39
|
Xu S, Chaudhary O, Rodríguez-Morales P, Sun X, Chen D, Zappasodi R, Xu Z, Pinto AFM, Williams A, Schulze I, Farsakoglu Y, Varanasi SK, Low JS, Tang W, Wang H, McDonald B, Tripple V, Downes M, Evans RM, Abumrad NA, Merghoub T, Wolchok JD, Shokhirev MN, Ho PC, Witztum JL, Emu B, Cui G, Kaech SM. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8 + T cells in tumors. Immunity 2021; 54:1561-1577.e7. [PMID: 34102100 PMCID: PMC9273026 DOI: 10.1016/j.immuni.2021.05.003] [Citation(s) in RCA: 420] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/03/2021] [Accepted: 05/04/2021] [Indexed: 01/21/2023]
Abstract
A common metabolic alteration in the tumor microenvironment (TME) is lipid accumulation, a feature associated with immune dysfunction. Here, we examined how CD8+ tumor infiltrating lymphocytes (TILs) respond to lipids within the TME. We found elevated concentrations of several classes of lipids in the TME and accumulation of these in CD8+ TILs. Lipid accumulation was associated with increased expression of CD36, a scavenger receptor for oxidized lipids, on CD8+ TILs, which also correlated with progressive T cell dysfunction. Cd36-/- T cells retained effector functions in the TME, as compared to WT counterparts. Mechanistically, CD36 promoted uptake of oxidized low-density lipoproteins (OxLDL) into T cells, and this induced lipid peroxidation and downstream activation of p38 kinase. Inhibition of p38 restored effector T cell functions in vitro, and resolution of lipid peroxidation by overexpression of glutathione peroxidase 4 restored functionalities in CD8+ TILs in vivo. Thus, an oxidized lipid-CD36 axis promotes intratumoral CD8+ T cell dysfunction and serves as a therapeutic avenue for immunotherapies.
Collapse
Affiliation(s)
- Shihao Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Omkar Chaudhary
- Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Patricia Rodríguez-Morales
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Xiaoli Sun
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dan Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ziyan Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Antonio F M Pinto
- Mass Spectrometry Core for Proteomics and Metabolomics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - April Williams
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Isabell Schulze
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yagmur Farsakoglu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Siva Karthik Varanasi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jun Siong Low
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA; Fondazione per l'istituto di ricerca in biomedicina, Bellinzona, Switzerland
| | - Wenxi Tang
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Haiping Wang
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Bryan McDonald
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Victoria Tripple
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nada A Abumrad
- Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maxim N Shokhirev
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Joseph L Witztum
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brinda Emu
- Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Guoliang Cui
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
40
|
Proprotein Convertase Subtilisin Kexin Type 9 Inhibitors Reduce Platelet Activation Modulating ox-LDL Pathways. Int J Mol Sci 2021; 22:ijms22137193. [PMID: 34281247 PMCID: PMC8267646 DOI: 10.3390/ijms22137193] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Proprotein convertase subtilisin kexin type 9 inhibitors (PCSK9i) lower LDL-cholesterol and slow atherosclerosis preventing cardiovascular events. While it is known that circulating PCSK9 enhances platelet activation (PA) and that PCSK9i reduce it, the underlying mechanism is not still clarified. Methods: In a multicenter before–after study in 80 heterozygous familial hypercholesterolemia (HeFH) patients on treatment with maximum tolerated statin dose ± ezetimibe, PA, soluble-NOX2-derived peptide (sNOX2-dp), and oxidized-LDL (ox-LDL) were measured before and after six months of PCSK9i treatment. In vitro study investigates the effects of plasma from HeFH patients before and after PCK9i on PA in washed platelets (wPLTs) from healthy subjects. Results: Compared to baseline, PCSK9i reduced the serum levels of LDL-c, ox-LDL, Thromboxane (Tx) B2, sNOX2-dp, and PCSK9 (p < 0.001). The decrease of TxB2 correlates with that of ox-LDL, while ox-LDL reduction correlated with PCSK9 and sNOX2-dp delta. In vitro study demonstrated that wPLTs resuspended in plasma from HeFH after PCSK9i treatment induced lower PA and sNOX2-dp release than those obtained using plasma before PCSK9i treatment. This reduction was vanished by adding ox-LDL. ox-LDL-induced PA was blunted by CD36, LOX1, and NOX2 inhibition. Conclusions: PCSK9i treatment reduces PA modulating NOX2 activity and in turn ox-LDL formation in HeFH patients.
Collapse
|
41
|
Hsu YW, Hsu FF, Chiang MT, Tsai DL, Li FA, Angata T, Crocker PR, Chau LY. Siglec-E retards atherosclerosis by inhibiting CD36-mediated foam cell formation. J Biomed Sci 2021; 28:5. [PMID: 33397354 PMCID: PMC7784283 DOI: 10.1186/s12929-020-00698-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/21/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The accumulation of lipid-laden macrophages, foam cells, within sub-endothelial intima is a key feature of early atherosclerosis. Siglec-E, a mouse orthologue of human Siglec-9, is a sialic acid binding lectin predominantly expressed on the surface of myeloid cells to transduce inhibitory signal via recruitment of SH2-domain containing protein tyrosine phosphatase SHP-1/2 upon binding to its sialoglycan ligands. Whether Siglec-E expression on macrophages impacts foam cell formation and atherosclerosis remains to be established. METHODS ApoE-deficient (apoE-/-) and apoE/Siglec-E-double deficient (apoE-/-/Siglec-E-/-) mice were placed on high fat diet for 3 months and their lipid profiles and severities of atherosclerosis were assessed. Modified low-density lipoprotein (LDL) uptake and foam cell formation in wild type (WT) and Siglec-E-/-- peritoneal macrophages were examined in vitro. Potential Siglec-E-interacting proteins were identified by proximity labeling in conjunction with proteomic analysis and confirmed by coimmunoprecipitation experiment. Impacts of Siglec-E expression and cell surface sialic acid status on oxidized LDL uptake and signaling involved were examined by biochemical assays. RESULTS Here we show that genetic deletion of Siglec-E accelerated atherosclerosis without affecting lipid profile in apoE-/- mice. Siglec-E deficiency promotes foam cell formation by enhancing acetylated and oxidized LDL uptake without affecting cholesterol efflux in macrophages in vitro. By performing proximity labeling and proteomic analysis, we identified scavenger receptor CD36 as a cell surface protein interacting with Siglec-E. Further experiments performed in HEK293T cells transiently overexpressing Siglec-E and CD36 and peritoneal macrophages demonstrated that depletion of cell surface sialic acids by treatment with sialyltransferase inhibitor or sialidase did not affect interaction between Siglec-E and CD36 but retarded Siglec-E-mediated inhibition on oxidized LDL uptake. Subsequent experiments revealed that oxidized LDL induced transient Siglec-E tyrosine phosphorylation and recruitment of SHP-1 phosphatase in macrophages. VAV, a downstream effector implicated in CD36-mediated oxidized LDL uptake, was shown to interact with SHP-1 following oxidized LDL treatment. Moreover, oxidized LDL-induced VAV phosphorylation was substantially lower in WT macrophages comparing to Siglec-E-/- counterparts. CONCLUSIONS These data support the protective role of Siglec-E in atherosclerosis. Mechanistically, Siglec-E interacts with CD36 to suppress downstream VAV signaling involved in modified LDL uptake.
Collapse
Affiliation(s)
- Yaw-Wen Hsu
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Fu-Fei Hsu
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Ming-Tsai Chiang
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Dong-Lin Tsai
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Fu-An Li
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Takashi Angata
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Paul R Crocker
- Division of Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Lee-Young Chau
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan.
| |
Collapse
|
42
|
Kim JJ, David JM, Wilbon SS, Santos JV, Patel DM, Ahmad A, Mitrofanova A, Liu X, Mallela SK, Ducasa GM, Ge M, Sloan AJ, Al-Ali H, Boulina M, Mendez AJ, Contreras GN, Prunotto M, Sohail A, Fridman R, Miner JH, Merscher S, Fornoni A. Discoidin domain receptor 1 activation links extracellular matrix to podocyte lipotoxicity in Alport syndrome. EBioMedicine 2020; 63:103162. [PMID: 33340991 PMCID: PMC7750578 DOI: 10.1016/j.ebiom.2020.103162] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Background Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase that is activated by collagens that is involved in the pathogenesis of fibrotic disorders. Interestingly, de novo production of the collagen type I (Col I) has been observed in Col4a3 knockout mice, a mouse model of Alport Syndrome (AS mice). Deletion of the DDR1 in AS mice was shown to improve survival and renal function. However, the mechanisms driving DDR1-dependent fibrosis remain largely unknown. Methods Podocyte pDDR1 levels, Collagen and cluster of differentiation 36 (CD36) expression was analyzed by Real-time PCR and Western blot. Lipid droplet accumulation and content was determined using Bodipy staining and enzymatic analysis. CD36 and DDR1 interaction was determined by co-immunoprecipitation. Creatinine, BUN, albuminuria, lipid content, and histological and morphological assessment of kidneys harvested from AS mice treated with Ezetimibe and/or Ramipril or vehicle was performed. Findings We demonstrate that Col I-mediated DDR1 activation induces CD36-mediated podocyte lipotoxic injury. We show that Ezetimibe interferes with the CD36/DDR1 interaction in vitro and prevents lipotoxicity in AS mice thus preserving renal function similarly to ramipril. Interpretation Our study suggests that Col I/DDR1-mediated lipotoxicity contributes to renal failure in AS and that targeting this pathway may represent a new therapeutic strategy for patients with AS and with chronic kidney diseases (CKD) associated with Col4 mutations. Funding This study is supported by the NIH grants R01DK117599, R01DK104753, R01CA227493, U54DK083912, UM1DK100846, U01DK116101, UL1TR000460 (Miami Clinical Translational Science Institute, National Center for Advancing Translational Sciences and the National Institute on Minority Health and Health Disparities), F32DK115109, Hoffmann-La Roche and Alport Syndrome Foundation.
Collapse
Affiliation(s)
- Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States.
| | - Judith M David
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Sydney S Wilbon
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Javier V Santos
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Devang M Patel
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Anis Ahmad
- Department of Radiation Oncology, University of Miami, FL 33136, United States
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Xiaochen Liu
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Shamroop K Mallela
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Gloria M Ducasa
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Alexis J Sloan
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Hassan Al-Ali
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Marcia Boulina
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Armando J Mendez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Gabriel N Contreras
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Marco Prunotto
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Anjum Sohail
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Rafael Fridman
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States.
| |
Collapse
|
43
|
Shu H, Peng Y, Hang W, Nie J, Zhou N, Wang DW. The role of CD36 in cardiovascular disease. Cardiovasc Res 2020; 118:115-129. [PMID: 33210138 PMCID: PMC8752351 DOI: 10.1093/cvr/cvaa319] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
CD36, also known as the scavenger receptor B2, is a multifunctional receptor widely expressed in various organs. CD36 plays a crucial role in the uptake of long-chain fatty acids, the main metabolic substrate in myocardial tissue. The maturation and transportation of CD36 is regulated by post-translational modifications, including phosphorylation, ubiquitination, glycosylation, and palmitoylation. CD36 is decreased in pathological cardiac hypertrophy caused by ischaemia-reperfusion and pressure overload, and increased in diabetic cardiomyopathy and atherosclerosis. Deficiency of CD36 alleviates diabetic cardiomyopathy and atherosclerosis, while overexpression of CD36 eliminates ischaemia-reperfusion damage, together suggesting that CD36 is closely associated with the progression of cardiovascular diseases and may be a new therapeutic target. This review summarizes the regulation and post-translational modifications of CD36 and evaluates its role in cardiovascular diseases and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yizhong Peng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Weijian Hang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
44
|
Nie J, Yang J, Wei Y, Wei X. The role of oxidized phospholipids in the development of disease. Mol Aspects Med 2020; 76:100909. [PMID: 33023753 DOI: 10.1016/j.mam.2020.100909] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/29/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023]
Abstract
Oxidized phospholipids (OxPLs), complex mixtures of phospholipid oxidation products generated during normal or pathological processes, are increasingly recognized to show bioactive effects on many cellular signalling pathways. There is a growing body of evidence showing that OxPLs play an important role in many diseases, so it is essential to define the specific role of OxPLs in different diseases for the design of disease therapies. In vastly diverse pathological processes, OxPLs act as pro-inflammatory agents and contribute to the progression of many diseases; in addition, they play a role in anti-inflammatory processes, promoting the dissipation of inflammation and inhibiting the progression of some diseases. In addition to participating in the regulation of inflammatory responses, OxPLs affect the occurrence and development of diseases through other pathways, such as apoptosis promotion. In this review, the different and even opposite effects of different OxPL molecular species are discussed. Furthermore, the specific effects of OxPLs in various diseases, as well as the receptor and cellular mechanisms involved, are summarized.
Collapse
Affiliation(s)
- Ji Nie
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Respiration, First People's Hospital of Yunnan Province, Yunnan, 650032, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Jing Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
45
|
Bandaru S, Ala C, Ekstrand M, Akula MK, Pedrelli M, Liu X, Bergström G, Håversen L, Borén J, Bergo MO, Akyürek LM. Lack of RAC1 in macrophages protects against atherosclerosis. PLoS One 2020; 15:e0239284. [PMID: 32941503 PMCID: PMC7498073 DOI: 10.1371/journal.pone.0239284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022] Open
Abstract
The Rho GTPase RAC1 is an important regulator of cytoskeletal dynamics, but the role of macrophage-specific RAC1 has not been explored during atherogenesis. We analyzed RAC1 expression in human carotid atherosclerotic plaques using immunofluorescence and found higher macrophage RAC1 expression in advanced plaques compared with intermediate human atherosclerotic plaques. We then produced mice with Rac1-deficient macrophages by breeding conditional floxed Rac1 mice (Rac1fl/fl) with mice expressing Cre from the macrophage-specific lysosome M promoter (LC). Atherosclerosis was studied in vivo by infecting Rac1fl/fl and Rac1fl/fl/LC mice with AdPCSK9 (adenoviral vector overexpressing proprotein convertase subtilisin/kexin type 9). Rac1fl/fl/LC macrophages secreted lower levels of IL-6 and TNF-α and exhibited reduced foam cell formation and lipid uptake. The deficiency of Rac1 in macrophages reduced the size of aortic atherosclerotic plaques in AdPCSK9-infected Rac1fl/fl/LC mice. Compare with controls, intima/media ratios, the size of necrotic cores, and numbers of CD68-positive macrophages in atherosclerotic plaques were reduced in Rac1-deficient mice. Moreover, we found that RAC1 interacts with actin-binding filamin A. Macrophages expressed increased RAC1 levels in advanced human atherosclerosis. Genetic inactivation of RAC1 impaired macrophage function and reduced atherosclerosis in mice, suggesting that drugs targeting RAC1 may be useful in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Sashidar Bandaru
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Chandu Ala
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Matias Ekstrand
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Murali K. Akula
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Matteo Pedrelli
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Xi Liu
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Västra Götalandregionen, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Liliana Håversen
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Martin O. Bergo
- Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Levent M. Akyürek
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Pathology, Västra Götalandregionen, Sahlgrenska University Hospital, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Atherosclerosis is a chronic disease characterized by lipid retention and inflammation in the artery wall. The retention and oxidation of low-density lipoprotein (LDL) in sub-endothelial space play a critical role in atherosclerotic plaque formation and destabilization. Oxidized LDL (ox-LDL) and other modified LDL particles are avidly taken up by endothelial cells, smooth muscle cells, and macrophages mainly through several scavenger receptors, including CD36 which is a class B scavenger receptor and membrane glycoprotein. RECENT FINDINGS Animal studies performed on CD36-deficient mice suggest that deficiency of CD36 prevents the development of atherosclerosis, though with some debate. CD36 serves as a signaling hub protein at the crossroad of inflammation, lipid metabolism, and fatty acid metabolism. In addition, the level of soluble CD36 (unattached to cells) in the circulating blood was elevated in patients with atherosclerosis and other metabolic disorders. We performed a state-of-the-art review on the structure, ligands, functions, and regulation of CD36 in the context of atherosclerosis by focusing on the pathological role of CD36 in the dysfunction of endothelial cells, smooth muscle cells, monocytes/macrophages, and platelets. Finally, we highlight therapeutic possibilities to target CD36 expression/activity in atherosclerosis.
Collapse
|
47
|
Grajchen E, Wouters E, van de Haterd B, Haidar M, Hardonnière K, Dierckx T, Van Broeckhoven J, Erens C, Hendrix S, Kerdine-Römer S, Hendriks JJA, Bogie JFJ. CD36-mediated uptake of myelin debris by macrophages and microglia reduces neuroinflammation. J Neuroinflammation 2020; 17:224. [PMID: 32718316 PMCID: PMC7384221 DOI: 10.1186/s12974-020-01899-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/15/2020] [Indexed: 11/24/2022] Open
Abstract
Background The presence of foamy macrophages and microglia containing intracellular myelin remnants is a pathological hallmark of neurodegenerative disorders such as multiple sclerosis (MS). Despite the importance of myelin internalization in affecting both central nervous system repair and neuroinflammation, the receptors involved in myelin clearance and their impact on the phagocyte phenotype and lesion progression remain to be clarified. Methods Flow cytometry, quantitative PCR, and immunohistochemistry were used to define the mRNA and protein abundance of CD36 in myelin-containing phagocytes. The impact of CD36 and nuclear factor erythroid 2–related factor 2 (NRF2) on the phagocytic and inflammatory features of macrophages and microglia was assessed using a pharmacological CD36 inhibitor (sulfo-N-succinimidyl oleate) and Nrf2−/− bone marrow-derived macrophages. Finally, the experimental autoimmune encephalomyelitis (EAE) model was used to establish the impact of CD36 inhibition on neuroinflammation and myelin phagocytosis in vivo. Results Here, we show that the fatty acid translocase CD36 is required for the uptake of myelin debris by macrophages and microglia, and that myelin internalization increased CD36 expression through NRF2. Pharmacological inhibition of CD36 promoted the inflammatory properties of myelin-containing macrophages and microglia in vitro, which was paralleled by a reduced activity of the anti-inflammatory lipid-sensing liver X receptors and peroxisome proliferator-activated receptors. By using the EAE model, we provide evidence that CD36 is essential for myelin debris clearance in vivo. Importantly, CD36 inhibition markedly increased the neuroinflammatory burden and disease severity in the EAE model. Conclusion Altogether, we show for the first time that CD36 is crucial for clearing myelin debris and suppressing neuroinflammation in demyelinating disorders such as MS.
Collapse
Affiliation(s)
- Elien Grajchen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Elien Wouters
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Britt van de Haterd
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Mansour Haidar
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Kévin Hardonnière
- Inflammation, Microbiome and Immunosurveillance, INSERM UMR99, Université Paris-Saclay, Châtenay-Malabry, France
| | - Tess Dierckx
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Celine Erens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sven Hendrix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Saadia Kerdine-Römer
- Inflammation, Microbiome and Immunosurveillance, INSERM UMR99, Université Paris-Saclay, Châtenay-Malabry, France
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
48
|
Vazquez MM, Gutierrez MV, Salvatore SR, Puiatti M, Dato VA, Chiabrando GA, Freeman BA, Schopfer FJ, Bonacci G. Nitro-oleic acid, a ligand of CD36, reduces cholesterol accumulation by modulating oxidized-LDL uptake and cholesterol efflux in RAW264.7 macrophages. Redox Biol 2020; 36:101591. [PMID: 32531545 PMCID: PMC7287307 DOI: 10.1016/j.redox.2020.101591] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 01/09/2023] Open
Abstract
Macrophages play a pivotal role in the early stages of atherosclerosis development; they excessively accumulate cholesterol in the cytosol in response to modified Low Density Lipoprotein (mLDL). The mLDL are incorporated through scavenger receptors. CD36 is a high-affinity cell surface scavenger receptor that facilitates the binding and uptake of long-chain fatty acids and mLDL into the cell. Numerous structurally diverse ligands can initiate signaling responses through CD36 to regulate cell metabolism, migration, and angiogenesis. Nitro-fatty acids are endogenous electrophilic lipid mediators that react with and modulate the function of multiple enzymes and transcriptional regulatory proteins. These actions induce the expression of several anti-inflammatory and cytoprotective genes and limit pathologic responses in experimental models of atherosclerosis, cardiac ischemia/reperfusion, and inflammatory diseases. Pharmacological and genetic approaches were used to explore the actions of nitro-oleic acid (NO2-OA) on macrophage lipid metabolism. Pure synthetic NO2-OA dose-dependently increased CD36 expression in RAW264.7 macrophages and this up-regulation was abrogated in BMDM from Nrf2-KO mice. Ligand binding analysis revealed that NO2-OA specifically interacts with CD36, thus limiting the binding and uptake of mLDL. Docking analysis shows that NO2-OA establishes a low binding energy interaction with the alpha helix containing Lys164 in CD36. NO2-OA also restored autophagy flux in mLDL-loaded macrophages, thus reversing cholesterol deposition within the cell. In aggregate, these results indicate that NO2-OA reduces cholesterol uptake by binding to CD36 and increases cholesterol efflux by restoring autophagy.
Collapse
Affiliation(s)
- Matias M Vazquez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina
| | - Maria V Gutierrez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina
| | - Sonia R Salvatore
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, United States
| | - Marcelo Puiatti
- Departamento de Química Orgánica, INFIQC, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Virginia Actis Dato
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina
| | - Gustavo A Chiabrando
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, United States
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, United States
| | - Gustavo Bonacci
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.
| |
Collapse
|
49
|
Yuvashree M, Ganesh RN, Viswanathan P. Potential application of nanoemulsified garlic oil blend in mitigating the progression of type 2 diabetes-mediated nephropathy in Wistar rats. 3 Biotech 2020; 10:272. [PMID: 32523866 PMCID: PMC7260324 DOI: 10.1007/s13205-020-02262-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/14/2020] [Indexed: 02/05/2023] Open
Abstract
The renoprotective potential of nanoemulsified garlic oil blend (GNE) in alleviating the progressive stages of hyperlipidemia-mediated diabetic nephropathy was examined. The study was carried out in high fat-fed, streptozotocin-induced type 2 diabetic Wistar rats for five months. The diabetic rats showed a significant increase of area under the curve in OGTT (p < 0.01) and IPITT (p < 0.01), increased urinary albumin (p < 0.01), urinary microprotein (p < 0.001), total cholesterol (p < 0.01), triglycerides (p < 0.001) and LDL cholesterol (p < 0.001), with decreased serum albumin (p < 0.01), serum protein (p < 0.001) and HDL-cholesterol levels (p < 0.05) than the control rats. The histopathological analysis evidenced mesangial expansion and hypercellularity at the end of the first and third month, and glomerulosclerosis and tubular atrophy at the end of the fifth month in diabetic rats. Moreover, on disease progression, increase in urinary podocalyxin, NGAL and CD36 was observed, and the renal mRNA and protein expression of podocalyxin decreased significantly with a concomitant increase in NGAL and CD36 expression from first till fifth month end. The treatment with GNE (20 mg/kg) significantly ameliorated the serum albumin (p < 0.001) and urine albumin (p < 0.01) from the end of the third month with significant attenuation in the lipid profile than GO (20 mg/kg) or Ator (8 mg/kg). Moreover, GNE reverted the histopathological alterations and attenuated the aberrant mRNA, protein expression and urinary excretion level of renal CD36, podocalyxin and NGAL in diabetic rats from an early stage of disease till the end of the study period. This study demonstrated the enhanced efficacy of GO in nanoemulsified form in mitigating the progression of nephropathy in type 2 diabetic rats.
Collapse
Affiliation(s)
- Muralidaran Yuvashree
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632 014 India
| | - Rajesh Nachiappa Ganesh
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Dhanvantrinagar, Puducherry India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632 014 India
| |
Collapse
|
50
|
Yuvashree M, Ganesh RN, Viswanathan P. Potential application of nanoemulsified garlic oil blend in mitigating the progression of type 2 diabetes-mediated nephropathy in Wistar rats. 3 Biotech 2020. [DOI: https://doi.org/10.1007/s13205-020-02262-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|