1
|
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
|
2
|
Elimam H, Gauvin J, Huynh DN, Ménard L, Al-Hawat ML, Harb D, Lubell WD, Carpentier AC, Ong H, Marleau S. Targeting CD36 With EP 80317 Reduces Remote Inflammatory Response to Hind Limb Ischemia-Reperfusion in Mice. J Biochem Mol Toxicol 2024; 38:e70057. [PMID: 39552437 PMCID: PMC11582942 DOI: 10.1002/jbt.70057] [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: 09/19/2024] [Revised: 10/26/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
Reperfusion of ischemic skeletal muscle triggers oxidative stress and an immediate inflammatory reaction, leading to damage of distant organs such as the lungs. The inflammatory process implicates numerous mediators, including cytokines, chemokines, and arachidonic acid metabolites. In the orchestration of the inflammatory cascade, a critical role is played by the cluster of differentiation-36 receptor (CD36), a scavenger receptor class B protein (SR-B2) which is expressed on macrophages and functions as a Toll-like receptor coreceptor. A mouse model of hind limb ischemia-reperfusion has been used to investigate the interplay between CD36 signaling and remote inflammation: leukocyte recruitment, regulation of the nucleotide-binding domain leucin-rich repeat and pyrin-containing receptor 3 (NLRP3) inflammasome, and release of nuclear factor-kappa B (NF-ĸB) and arachidonic acid metabolites. Levels of reactive oxygen species, inflammatory mediators, and gene expression were measured in blood and lung tissue samples collected from anesthetized mice on which unilateral hind limb ischemia was induced by rubber band constriction for 30 min followed by reperfusion for 3 h. The CD36 modulator EP 80317, a member of the growth hormone releasing peptide 6 family, was employed as a pharmacological agent to mitigate distant lung injury following skeletal limb ischemia-reperfusion. Targeting CD36 on monocytes/macrophages, EP 80317 abated pro-inflammatory signaling and transcriptional activity encompassing lipid and cytokine mediators. Targeting CD36 was shown to offer promise for curtailing tissue injury following hind limb ischemia-reperfusion.
Collapse
Affiliation(s)
- Hanan Elimam
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Jade Gauvin
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - David N Huynh
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Liliane Ménard
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | | | - Diala Harb
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - André C Carpentier
- Department of Medicine, Division of Endocrinology, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
3
|
Yue N, Jin Q, Li C, Zhang L, Cao J, Wu C. CD36: a promising therapeutic target in hematologic tumors. Leuk Lymphoma 2024; 65:1749-1765. [PMID: 38982639 DOI: 10.1080/10428194.2024.2376178] [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: 05/20/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024]
Abstract
Cluster of differentiation 36 (CD36) is a multiligand receptor with important roles in lipid metabolism, angiogenesis and innate immunity, and its diverse effects may depend on the binding of specific ligands in different contexts. CD36 is expressed not only on immune cells in the tumor microenvironment (TME) but also on some hematopoietic cells. CD36 is associated with the growth, metastasis and drug resistance in some hematologic tumors, such as leukemia, lymphoma and myelodysplastic syndrome. Currently, some targeted therapeutic agents against CD36 have been developed, such as anti-CD36 antibodies, CD36 antagonists (small molecules) and CD36 expression inhibitors. This paper not only innovatively addresses the role of CD36 in some hematopoietic cells, such as erythrocytes, hematopoietic stem cells and platelets, but also pays special attention to the role of CD36 in the development of hematologic tumors, and suggests that CD36 may be a potential cancer therapeutic target in hematologic tumors.
Collapse
Affiliation(s)
- Ningning Yue
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qiqi Jin
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Cuicui Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Litian Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jiajia Cao
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Chongyang Wu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
4
|
Feng M, Zhou Q, Xie H, Liu C, Zheng M, Zhang S, Zhou S, Zhao J. Role of CD36 in central nervous system diseases. Neural Regen Res 2024; 19:512-518. [PMID: 37721278 PMCID: PMC10581564 DOI: 10.4103/1673-5374.380821] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/12/2023] [Accepted: 05/04/2023] [Indexed: 09/19/2023] Open
Abstract
CD36 is a highly glycosylated integral membrane protein that belongs to the scavenger receptor class B family and regulates the pathological progress of metabolic diseases. CD36 was recently found to be widely expressed in various cell types in the nervous system, including endothelial cells, pericytes, astrocytes, and microglia. CD36 mediates a number of regulatory processes, such as endothelial dysfunction, oxidative stress, mitochondrial dysfunction, and inflammatory responses, which are involved in many central nervous system diseases, such as stroke, Alzheimer's disease, Parkinson's disease, and spinal cord injury. CD36 antagonists can suppress CD36 expression or prevent CD36 binding to its ligand, thereby achieving inhibition of CD36-mediated pathways or functions. Here, we reviewed the mechanisms of action of CD36 antagonists, such as Salvianolic acid B, tanshinone IIA, curcumin, sulfosuccinimidyl oleate, antioxidants, and small-molecule compounds. Moreover, we predicted the structures of binding sites between CD36 and antagonists. These sites can provide targets for more efficient and safer CD36 antagonists for the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Min Feng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Qiang Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Huimin Xie
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Mengru Zheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Shuyu Zhang
- Medical College of Nantong University, Nantong, Jiangsu Province, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Jian Zhao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
5
|
Chen S, Gao JJ, Liu YJ, Mo ZW, Wu FY, Hu ZJ, Peng YM, Zhang XQ, Ma ZS, Liu ZL, Yan JY, Ou ZJ, Li Y, Ou JS. The oxidized phospholipid PGPC impairs endothelial function by promoting endothelial cell ferroptosis via FABP3. J Lipid Res 2024; 65:100499. [PMID: 38218337 PMCID: PMC10864338 DOI: 10.1016/j.jlr.2024.100499] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 01/15/2024] Open
Abstract
Ferroptosis is a novel cell death mechanism that is mediated by iron-dependent lipid peroxidation. It may be involved in atherosclerosis development. Products of phospholipid oxidation play a key role in atherosclerosis. 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphocholine (PGPC) is a phospholipid oxidation product present in atherosclerotic lesions. It remains unclear whether PGPC causes atherosclerosis by inducing endothelial cell ferroptosis. In this study, human umbilical vein endothelial cells (HUVECs) were treated with PGPC. Intracellular levels of ferrous iron, lipid peroxidation, superoxide anions (O2•-), and glutathione were detected, and expression of fatty acid binding protein-3 (FABP3), glutathione peroxidase 4 (GPX4), and CD36 were measured. Additionally, the mitochondrial membrane potential (MMP) was determined. Aortas from C57BL6 mice were isolated for vasodilation testing. Results showed that PGPC increased ferrous iron levels, the production of lipid peroxidation and O2•-, and FABP3 expression. However, PGPC inhibited the expression of GPX4 and glutathione production and destroyed normal MMP. These effects were also blocked by ferrostatin-1, an inhibitor of ferroptosis. FABP3 silencing significantly reversed the effect of PGPC. Furthermore, PGPC stimulated CD36 expression. Conversely, CD36 silencing reversed the effects of PGPC, including PGPC-induced FABP3 expression. Importantly, E06, a direct inhibitor of the oxidized 1-palmitoyl-2-arachidonoyl-phosphatidylcholine IgM natural antibody, inhibited the effects of PGPC. Finally, PGPC impaired endothelium-dependent vasodilation, ferrostatin-1 or FABP3 inhibitors inhibited this impairment. Our data demonstrate that PGPC impairs endothelial function by inducing endothelial cell ferroptosis through the CD36 receptor to increase FABP3 expression. Our findings provide new insights into the mechanisms of atherosclerosis and a therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Si Chen
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Jian-Jun Gao
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Yu-Jia Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Zhi-Wei Mo
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Fang-Yuan Wu
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zuo-Jun Hu
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Xiao-Qin Zhang
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhen-Sheng Ma
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Ze-Long Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Jian-Yun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Yan Li
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China.
| | - Jing-Song Ou
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
6
|
Xia L, Zhou Z, Chen X, Luo W, Ding L, Xie H, Zhuang W, Ni K, Li G. Ligand-dependent CD36 functions in cancer progression, metastasis, immune response, and drug resistance. Biomed Pharmacother 2023; 168:115834. [PMID: 37931517 DOI: 10.1016/j.biopha.2023.115834] [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: 07/12/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023] Open
Abstract
CD36, a multifunctional glycoprotein, has been shown to play critical roles in tumor initiation, progression, metastasis, immune response, and drug resistance. CD36 serves as a receptor for a wide range of ligands, including lipid-related ligands (e.g., long-chain fatty acid (LCFA), oxidized low-density lipoprotein (oxLDL), and oxidized phospholipids), as well as protein-related ligands (e.g., thrombospondins, amyloid proteins, collagens I and IV). CD36 is overexpressed in various cancers and may act as an independent prognostic marker. While it was initially identified as a mediator of anti-angiogenesis through its interaction with thrombospondin-1 (TSP1), recent research has highlighted its role in promoting tumor growth, metastasis, drug resistance, and immune suppression. The varied impact of CD36 on cancer is likely ligand-dependent. Therefore, we focus specifically on the ligand-dependent role of CD36 in cancer to provide a critical review of recent advances, perspectives, and challenges.
Collapse
Affiliation(s)
- Liqun Xia
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhenwei Zhou
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianjiong Chen
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenqin Luo
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifeng Ding
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyun Xie
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhuang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Kangxin Ni
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
7
|
Ullah S, Ahmad T, Ikram M, Rasheed HM, Khan MI, Khan T, Alsahli TG, Alzarea SI, Althobaiti M, Shah AJ. 7-Hydroxy Frullanolide Ameliorates Isoproterenol-Induced Myocardial Injury through Modification of iNOS and Nrf2 Genes. Biomedicines 2023; 11:2470. [PMID: 37760913 PMCID: PMC10526241 DOI: 10.3390/biomedicines11092470] [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: 07/23/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Myocardial infarction (MI) is the principal cause of premature death. Protecting myocardium from ischemia is the main focus of intense research. 7-hydroxy frullanolide (7-HF) is a potent anti-inflammatory agent, showing its efficacy in different acute and chronic inflammatory disorders such as atherosclerosis, suggesting it can be a potential cardioprotective agent. For the induction of MI, Sprague-Dawley rats (n = 5) were administered isoproterenol (ISO) 85 mg/kg s.c at 24 h intervals for two days. The potential cardioprotective effect of 7-HF and its mechanisms were explored by in vivo and in vitro methods. 7-HF significantly prevented the extent of myocardial injury by decreasing the infarct size, preserving the histology of myocardial tissue, and reducing the release of cardiac biomarkers. Further, 7-HF increased the mRNA expression of cardioprotective gene Nrf2 and reduced the mRNA expression of iNOS. 7-HF also improved cardiac function by decreasing the cardiac workload through its negative chronotropic and negative ionotropic effect, as well as by reducing peripheral vascular resistance due to the inhibition of voltage-dependent calcium channels and the release of calcium from intracellular calcium stores. In conclusion, 7-HF showed cardioprotective effects in the MI model, which might be due to modulating the expression of iNOS and Nrf2 genes as well as improving cardiac functions.
Collapse
Affiliation(s)
- Saif Ullah
- Cardiovascular Research Group, Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, University Road, Abbottabad 22060, Pakistan; (S.U.); (M.I.); (T.K.)
| | - Taseer Ahmad
- Department of Pharmacology, College of Pharmacy, University of Sargodha, University Road, Sargodha 40100, Pakistan;
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Muhammad Ikram
- Cardiovascular Research Group, Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, University Road, Abbottabad 22060, Pakistan; (S.U.); (M.I.); (T.K.)
| | | | | | - Taous Khan
- Cardiovascular Research Group, Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, University Road, Abbottabad 22060, Pakistan; (S.U.); (M.I.); (T.K.)
| | - Tariq G. Alsahli
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia; (T.G.A.); (S.I.A.)
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia; (T.G.A.); (S.I.A.)
| | - Musaad Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
| | - Abdul Jabbar Shah
- Cardiovascular Research Group, Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, University Road, Abbottabad 22060, Pakistan; (S.U.); (M.I.); (T.K.)
| |
Collapse
|
8
|
Feng WW, Zuppe HT, Kurokawa M. The Role of CD36 in Cancer Progression and Its Value as a Therapeutic Target. Cells 2023; 12:1605. [PMID: 37371076 PMCID: PMC10296821 DOI: 10.3390/cells12121605] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Cluster of differentiation 36 (CD36) is a cell surface scavenger receptor that plays critical roles in many different types of cancer, notably breast, brain, and ovarian cancers. While it is arguably most well-known for its fatty acid uptake functions, it is also involved in regulating cellular adhesion, immune response, and apoptosis depending on the cellular and environmental contexts. Here, we discuss the multifaceted role of CD36 in cancer biology, such as its role in mediating metastasis, drug resistance, and immune evasion to showcase its potential as a therapeutic target. We will also review existing approaches to targeting CD36 in pre-clinical studies, as well as discuss the only CD36-targeting drug to advance to late-stage clinical trials, VT1021. Given the roles of CD36 in the etiology of metabolic disorders, such as atherosclerosis, diabetes, and non-alcoholic fatty liver disease, the clinical implications of CD36-targeted therapy are wide-reaching, even beyond cancer.
Collapse
Affiliation(s)
- William W. Feng
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Hannah T. Zuppe
- School of Biomedical Sciences, Kent State University, Kent, OH 44240, USA
| | - Manabu Kurokawa
- School of Biomedical Sciences, Kent State University, Kent, OH 44240, USA
- Department of Biological Sciences, Kent State University, Kent, OH 44240, USA
| |
Collapse
|
9
|
Gauvin J, Frégeau G, Elimam H, Ménard L, Huynh D, Lê C, Ahsanullah A, Lubell WD, Ong H, Marleau S. A cyclic azapeptide ligand of the scavenger receptor CD36/SR-B2 reduces the atherosclerotic lesion progression and enhances plaque stability in apolipoprotein E-deficient mice. Front Pharmacol 2023; 14:1204905. [PMID: 37332345 PMCID: PMC10270736 DOI: 10.3389/fphar.2023.1204905] [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: 04/12/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial walls that develops at predisposed sites. As a major risk factor for adverse cardiovascular pathology, atherosclerosis can progress to myocardial infarction and stroke, due to the rupture of unstable atherosclerotic lesions. Macrophage uptake of modified lipoproteins and metabolic dysfunction contributes significantly to the initiation and development of atherosclerotic lesions. The cluster of differentiation 36 receptor [CD36 (SR-B2)] plays a key role in atherosclerotic lesion progression and acts as an efferocytic molecule in the resolution of advanced plaque. In previous studies, linear azapeptide CD36 ligands were shown to exhibit anti-atherosclerotic properties. In the present study, a novel potent and selective macrocyclic azapeptide CD36 ligand, MPE-298, has proven effective in protecting against atherosclerosis progression. Features of greater plaque stability were observed after 8 weeks of daily injections with the cyclic azapeptide in apolipoprotein E-deficient mice fed a high-fat high-cholesterol diet.
Collapse
Affiliation(s)
- Jade Gauvin
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | | | - Hanan Elimam
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Sinai University-Kantara Branch, Ismailia, Egypt
| | - Liliane Ménard
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - David Huynh
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Catherine Lê
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Ahsanullah Ahsanullah
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - William D. Lubell
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
10
|
Callegari IOM, Rocha GZ, Oliveira AG. Physical exercise, health, and disease treatment: The role of macrophages. Front Physiol 2023; 14:1061353. [PMID: 37179836 PMCID: PMC10166825 DOI: 10.3389/fphys.2023.1061353] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
Subclinical inflammation is linked to comorbidities and risk factors, consolidating the diagnosis of chronic non-communicable diseases, such as insulin resistance, atherosclerosis, hepatic steatosis, and some types of cancer. In this context, the role of macrophages is highlighted as a marker of inflammation as well as for the high power of plasticity of these cells. Macrophages can be activated in a wide range between classical or proinflammatory, named M1, and alternative or anti-inflammatory, also known as M2 polarization. All nuances between M1 and M2 macrophages orchestrate the immune response by secreting different sets of chemokines, while M1 cells promote Th1 response, the M2 macrophages recruit Th2 and Tregs lymphocytes. In turn, physical exercise has been a faithful tool in combating the proinflammatory phenotype of macrophages. This review proposes to investigate the cellular and molecular mechanisms in which physical exercise can help control inflammation and infiltration of macrophages within the non-communicable diseases scope. During obesity progress, proinflammatory macrophages predominate in adipose tissue inflammation, which reduces insulin sensitivity until the development of type 2 diabetes, progression of atherosclerosis, and diagnosis of non-alcoholic fatty liver disease. In this case, physical activity restores the balance between the proinflammatory/anti-inflammatory macrophage ratio, reducing the level of meta-inflammation. In the case of cancer, the tumor microenvironment is compatible with a high level of hypoxia, which contributes to the advancement of the disease. However, exercise increases the level of oxygen supply, favoring macrophage polarization in favor of disease regression.
Collapse
Affiliation(s)
- Irineu O. M. Callegari
- Department of Physical Education, Bioscience Institute, São Paulo State University (UNESP), São Paulo, Brazil
| | - Guilherme Z. Rocha
- Department of Internal Medicine, State University of Campinas, Campinas, Brazil
| | - Alexandre G. Oliveira
- Department of Physical Education, Bioscience Institute, São Paulo State University (UNESP), São Paulo, Brazil
| |
Collapse
|
11
|
Boccanegra B, Cappellari O, Mantuano P, Trisciuzzi D, Mele A, Tulimiero L, De Bellis M, Cirmi S, Sanarica F, Cerchiara AG, Conte E, Meanti R, Rizzi L, Bresciani E, Denoyelle S, Fehrentz JA, Cruciani G, Nicolotti O, Liantonio A, Torsello A, De Luca A. Growth hormone secretagogues modulate inflammation and fibrosis in mdx mouse model of Duchenne muscular dystrophy. Front Immunol 2023; 14:1119888. [PMID: 37122711 PMCID: PMC10130389 DOI: 10.3389/fimmu.2023.1119888] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Growth hormone secretagogues (GHSs) exert multiple actions, being able to activate GHS-receptor 1a, control inflammation and metabolism, to enhance GH/insulin-like growth factor-1 (IGF-1)-mediated myogenesis, and to inhibit angiotensin-converting enzyme. These mechanisms are of interest for potentially targeting multiple steps of pathogenic cascade in Duchenne muscular dystrophy (DMD). Methods Here, we aimed to provide preclinical evidence for potential benefits of GHSs in DMD, via a multidisciplinary in vivo and ex vivo comparison in mdx mice, of two ad hoc synthesized compounds (EP80317 and JMV2894), with a wide but different profile. 4-week-old mdx mice were treated for 8 weeks with EP80317 or JMV2894 (320 µg/kg/d, s.c.). Results In vivo, both GHSs increased mice forelimb force (recovery score, RS towards WT: 20% for EP80317 and 32% for JMV2894 at week 8). In parallel, GHSs also reduced diaphragm (DIA) and gastrocnemius (GC) ultrasound echodensity, a fibrosis-related parameter (RS: ranging between 26% and 75%). Ex vivo, both drugs ameliorated DIA isometric force and calcium-related indices (e.g., RS: 40% for tetanic force). Histological analysis highlighted a relevant reduction of fibrosis in GC and DIA muscles of treated mice, paralleled by a decrease in gene expression of TGF-β1 and Col1a1. Also, decreased levels of pro-inflammatory genes (IL-6, CD68), accompanied by an increment in Sirt-1, PGC-1α and MEF2c expression, were observed in response to treatments, suggesting an overall improvement of myofiber metabolism. No detectable transcript levels of GHS receptor-1a, nor an increase of circulating IGF-1 were found, suggesting the presence of a novel receptor-independent mechanism in skeletal muscle. Preliminary docking studies revealed a potential binding capability of JMV2894 on metalloproteases involved in extracellular matrix remodeling and cytokine production, such as ADAMTS-5 and MMP-9, overactivated in DMD. Discussion Our results support the interest of GHSs as modulators of pathology progression in mdx mice, disclosing a direct anti-fibrotic action that may prove beneficial to contrast pathological remodeling.
Collapse
Affiliation(s)
- Brigida Boccanegra
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Ornella Cappellari
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Paola Mantuano
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Daniela Trisciuzzi
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonietta Mele
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Lisamaura Tulimiero
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Michela De Bellis
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Santa Cirmi
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Francesca Sanarica
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Elena Conte
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Ramona Meanti
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Severine Denoyelle
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Orazio Nicolotti
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonella Liantonio
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Annamaria De Luca
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
12
|
The Role of Macrophages in the Pathogenesis of Atherosclerosis. Cells 2023; 12:cells12040522. [PMID: 36831189 PMCID: PMC9954519 DOI: 10.3390/cells12040522] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
A wide variety of cell populations, including both immune and endothelial cells, participate in the pathogenesis of atherosclerosis. Among these groups, macrophages deserve special attention because different populations of them can have completely different effects on atherogenesis and inflammation in atherosclerosis. In the current review, the significance of different phenotypes of macrophages in the progression or regression of atherosclerosis will be considered, including their ability to become the foam cells and the consequences of this event, as well as their ability to create a pro-inflammatory or anti-inflammatory medium at the site of atherosclerotic lesions as a result of cytokine production. In addition, several therapeutic strategies directed to the modulation of macrophage activity, which can serve as useful ideas for future drug developments, will be considered.
Collapse
|
13
|
Zhang J, Ding W, Liu J, Wan J, Wang M. Scavenger Receptors in Myocardial Infarction and Ischemia/Reperfusion Injury: The Potential for Disease Evaluation and Therapy. J Am Heart Assoc 2023; 12:e027862. [PMID: 36645089 PMCID: PMC9939064 DOI: 10.1161/jaha.122.027862] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Scavenger receptors (SRs) are a structurally heterogeneous superfamily of evolutionarily conserved receptors that are divided into classes A to J. SRs can recognize multiple ligands, such as modified lipoproteins, damage-associated molecular patterns, and pathogen-associated molecular patterns, and regulate lipid metabolism, immunity, and homeostasis. According to the literature, SRs may play a critical role in myocardial infarction and ischemia/reperfusion injury, and the soluble types of SRs may be a series of promising biomarkers for the diagnosis and prognosis of patients with acute coronary syndrome or acute myocardial infarction. In this review, we briefly summarize the structure and function of SRs and discuss the association between each SR and ischemic cardiac injury in patients and animal models in detail. A better understanding of the effect of SRs on ischemic cardiac injury will inspire novel ideas for therapeutic drug discovery and disease evaluation in patients with myocardial infarction.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina,Cardiovascular Research InstituteWuhan UniversityWuhanChina,Hubei Key Laboratory of CardiologyWuhanChina
| | - Wen Ding
- Department of RadiologyThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Jianfang Liu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina,Cardiovascular Research InstituteWuhan UniversityWuhanChina,Hubei Key Laboratory of CardiologyWuhanChina
| | - Jun Wan
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina,Cardiovascular Research InstituteWuhan UniversityWuhanChina,Hubei Key Laboratory of CardiologyWuhanChina
| | - Menglong Wang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina,Cardiovascular Research InstituteWuhan UniversityWuhanChina,Hubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
14
|
Yang R, Liu Q, Zhang M. The Past and Present Lives of the Intraocular Transmembrane Protein CD36. Cells 2022; 12:cells12010171. [PMID: 36611964 PMCID: PMC9818597 DOI: 10.3390/cells12010171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Cluster of differentiation 36 (CD36) belongs to the B2 receptors of the scavenger receptor class B family, which is comprised of single-chain secondary transmembrane glycoproteins. It is present in a variety of cell types, including monocytes, macrophages, microvascular endothelial cells, adipocytes, hepatocytes, platelets, skeletal muscle cells, kidney cells, cardiomyocytes, taste bud cells, and a variety of other cell types. CD36 can be localized on the cell surface, mitochondria, endoplasmic reticulum, and endosomes, playing a role in lipid accumulation, oxidative stress injury, apoptosis, and inflammatory signaling. Recent studies have found that CD36 is expressed in a variety of ocular cells, including retinal pigment epithelium (RPE), retinal microvascular endothelial cells, retinal ganglion cells (RGC), Müller cells, and photoreceptor cells, playing an important role in eye diseases, such as age-related macular degeneration (AMD), diabetic retinopathy (DR), and glaucoma. Therefore, a comprehensive understanding of CD36 function and downstream signaling pathways is of great significance for the prevention and treatment of eye diseases. This article reviews the molecular characteristics, distribution, and function of scavenger receptor CD36 and its role in ophthalmology in order to deepen the understanding of CD36 in eye diseases and provide new ideas for treatment strategies.
Collapse
Affiliation(s)
- Rucui Yang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
- Department of Ophthalmology, Shantou University Medical College, Shantou University, Shantou 515041, China
| | - Qingping Liu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
| |
Collapse
|
15
|
Zia A, Wu Y, Nguyen T, Wang X, Peter K, Ta HT. The choice of targets and ligands for site-specific delivery of nanomedicine to atherosclerosis. Cardiovasc Res 2021; 116:2055-2068. [PMID: 32077918 DOI: 10.1093/cvr/cvaa047] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/23/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022] Open
Abstract
As nanotechnologies advance into clinical medicine, novel methods for applying nanomedicine to cardiovascular diseases are emerging. Extensive research has been undertaken to unlock the complex pathogenesis of atherosclerosis. However, this complexity presents challenges to develop effective imaging and therapeutic modalities for early diagnosis and acute intervention. The choice of ligand-receptor system vastly influences the effectiveness of nanomedicine. This review collates current ligand-receptor systems used in targeting functionalized nanoparticles for diagnosis and treatment of atherosclerosis. Our focus is on the binding affinity and selectivity of ligand-receptor systems, as well as the relative abundance of targets throughout the development and progression of atherosclerosis. Antibody-based targeting systems are currently the most commonly researched due to their high binding affinities when compared with other ligands, such as antibody fragments, peptides, and other small molecules. However, antibodies tend to be immunogenic due to their size. Engineering antibody fragments can address this issue but will compromise their binding affinity. Peptides are promising ligands due to their synthetic flexibility and low production costs. Alongside the aforementioned binding affinity of ligands, the choice of target and its abundance throughout distinct stages of atherosclerosis and thrombosis is relevant to the intended purpose of the nanomedicine. Further studies to investigate the components of atherosclerotic plaques are required as their cellular and molecular profile shifts over time.
Collapse
Affiliation(s)
- Adil Zia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yuao Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Tuan Nguyen
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xiaowei Wang
- Baker Heart and Diabetes Institute, Melbourne, VIC 3000, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, VIC 3000, Australia
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
16
|
Recognition of Oxidized Lipids by Macrophages and Its Role in Atherosclerosis Development. Biomedicines 2021; 9:biomedicines9080915. [PMID: 34440119 PMCID: PMC8389651 DOI: 10.3390/biomedicines9080915] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a multifactorial chronic disease that has a prominent inflammatory component. Currently, atherosclerosis is regarded as an active autoimmune process that involves both innate and adaptive immune pathways. One of the drivers of this process is the presence of modified low-density lipoprotein (LDL). For instance, lipoprotein oxidation leads to the formation of oxidation-specific epitopes (OSE) that can be recognized by the immune cells. Macrophage response to OSEs is recognized as a key trigger for initiation and a stimulator of progression of the inflammatory process in the arteries. At the same time, the role of oxidized LDL components is not limited to pro-inflammatory stimulation, but includes immunoregulatory effects that can have protective functions. It is, therefore, important to better understand the complexity of oxidized LDL effects in atherosclerosis in order to develop new therapeutic approaches to correct the inflammatory and metabolic imbalance associated with this disorder. In this review, we discuss the process of oxidized LDL formation, mechanisms of OSE recognition by macrophages and the role of these processes in atherosclerosis.
Collapse
|
17
|
Huang X, Khoong Y, Han C, Su D, Ma H, Gu S, Li Q, Zan T. Targeting Dermal Fibroblast Subtypes in Antifibrotic Therapy: Surface Marker as a Cellular Identity or a Functional Entity? Front Physiol 2021; 12:694605. [PMID: 34335301 PMCID: PMC8319956 DOI: 10.3389/fphys.2021.694605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/16/2021] [Indexed: 02/01/2023] Open
Abstract
Fibroblasts are the chief effector cells in fibrotic diseases and have been discovered to be highly heterogeneous. Recently, fibroblast heterogeneity in human skin has been studied extensively and several surface markers for dermal fibroblast subtypes have been identified, holding promise for future antifibrotic therapies. However, it has yet to be confirmed whether surface markers should be looked upon as merely lineage landmarks or as functional entities of fibroblast subtypes, which may further complicate the interpretation of cellular function of these fibroblast subtypes. This review aims to provide an update on current evidence on fibroblast surface markers in fibrotic disorders of skin as well as of other organ systems. Specifically, studies where surface markers were treated as lineage markers and manipulated as functional membrane proteins are both evaluated in parallel, hoping to reveal the underlying mechanism behind the pathogenesis of tissue fibrosis contributed by various fibroblast subtypes from multiple angles, shedding lights on future translational researches.
Collapse
Affiliation(s)
- Xin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yimin Khoong
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chengyao Han
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dai Su
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Ma
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuchen Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Tam J, Thankam F, Agrawal DK, Radwan MM. Critical Role of LOX-1-PCSK9 Axis in the Pathogenesis of Atheroma Formation and Its Instability. Heart Lung Circ 2021; 30:1456-1466. [PMID: 34092505 DOI: 10.1016/j.hlc.2021.05.085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/15/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is a major contributor to annual deaths globally. Atherosclerosis is a prominent risk factor for CVD. Although significant developments have been recently made in the prevention and treatment, the molecular pathology of atherosclerosis remains unknown. Interestingly, the recent discovery of proprotein convertase subtilisin/kexin type 9 (PCSK9) introduced a new avenue to explore the molecular pathogenesis and novel management strategies for atherosclerosis. Initial research focussed on the PCSK9-mediated degradation of low density lipoprotein receptor (LDLR) and subsequent activation of pro-inflammatory pathways by oxidised low density lipoprotein (ox-LDL). Recently, PCSK9 and lectin-like oxidised low-density lipoprotein receptor-1 (LOX-1) were shown to positively amplify each other pro-inflammatory activity and gene expression in endothelial cells, macrophages and vascular smooth muscle cells. In this literature review, we provide insight into the reciprocal relationship between PCSK9 and LOX-1 in the pathogenesis of atheroma formation and plaque instability in atherosclerosis. Further understanding of the LOX-1-PCSK9 axis possesses tremendous translational potential to design novel management approaches for atherosclerosis.
Collapse
Affiliation(s)
- Jonathan Tam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Finosh Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Mohamed M Radwan
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
19
|
Liu YG, Yan JL, Ji YQ, Nie WJ, Jiang Y. Black mulberry ethanol extract attenuates atherosclerosis-related inflammatory factors and downregulates PPARγ and CD36 genes in experimental atherosclerotic rats. Food Funct 2021; 11:2997-3005. [PMID: 32236255 DOI: 10.1039/c9fo02736j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atherosclerosis (AS) is the pathological basis of various vascular diseases and currently is seriously affecting human health. Numerous studies have paid more attention to natural medicines with anti-AS properties. As a traditional Uygur folk medicine, black mulberry fruits are conventionally used in the prevention and treatment of cardiovascular diseases in southern Xinjiang of China, and their underlying mechanisms remain unknown. Our previous study revealed that the ethanol extract of black mulberry (EEBM) inhibited AS development by improving lipid metabolism abnormalities, enhancing anti-oxidative activities, and reducing atherosclerotic lesions of atherosclerotic rats. Based on this, our objective was to further investigate the effects of EEBM on the expression of AS-related inflammatory factors and the key genes PPARγ and CD36 of the ox-LDL-PPARγ-CD36 feed-forward cycle in experimental atherosclerotic rats. Black mulberry fruits were extracted with acid ethanol and chromatographed on an AB-8 macroporous resin to obtain EEBM. All experimental rats were randomly divided into five groups: normal, model, model plus simvastatin (5 mg/kg d·body weight), and model plus low-dose and high-dose EEBM groups (105 and 210 mg/kg d·body weight, respectively). Serum levels of the inflammatory factors were determined by enzyme-linked immunosorbent assay (ELISA). The mRNA and protein expression of PPARγ and CD36 in atherosclerotic rats' liver tissue and thoracic aorta were determined by Q-PCR and western blot analysis, respectively. EEBM at high dose effectively attenuated the abnormally expressed AS-related inflammatory factors of TNF-α, IL-6, MMP-9, and CRP in atherosclerotic rats by 41.5%, 66.1%, 77.5%, and 79.5%, respectively. After treatment with high dose EEBM, the elevated-expressions of PPARγ and CD36 at the mRNA and protein levels in atherosclerotic rats were found to be obviously downregulated at both levels. These results demonstrate that EEBM might lessen the AS-related inflammatory reaction, and then inhibit the formation of ox-LDL, consequently downregulating the expression of PPARγ and CD36 at the mRNA and protein levels, thus reducing macrophage-foam-cell formation and prohibiting the development of atherosclerotic plaque through the ox-LDL-PPARγ-CD36 feed-forward cycle, which can effectively prevent the occurrence and development of AS in atherosclerotic rats.
Collapse
Affiliation(s)
- Yun-Guo Liu
- College of Life Sciences, Linyi University, Linyi 276005, China
| | - Jia-Li Yan
- Department of Laboratory Medicine, Chengdu Medical College, Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu, 610500, China.
| | - Yan-Qing Ji
- College of Life Sciences, Linyi University, Linyi 276005, China
| | - Wen-Jing Nie
- Changji Vocational and Technical College, Changji, 831100, Xinjiang, China
| | - Yan Jiang
- Department of Laboratory Medicine, Chengdu Medical College, Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu, 610500, China.
| |
Collapse
|
20
|
Juan CC, Li LH, Hou SK, Liu PS, Kao WF, Chiu YH, How CK. Expression of ABC transporter and scavenger receptor mRNAs in PBMCs in 100-km ultramarathon runners. Eur J Clin Invest 2021; 51:e13365. [PMID: 32725886 DOI: 10.1111/eci.13365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Cholesterol metabolism is tightly regulated at the cellular level. This study was to measure the expression levels of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1), scavenger receptor class B type I (SR-BI) and class A (SRA), and CD36 mRNAs in peripheral blood mononuclear cells (PBMCs) in response to 100-km ultramarathon event and determine any correlation between these ABC transporters/scavenger receptor expression levels and plasma cholesterol homeostasis. MATERIALS AND METHODS Twenty-six participants were enrolled. Blood was drawn from each individual 1 week prior, immediately after, and 24 hours after the race. The expression levels of ABCA1, ABCG1, SR-BI, SRA and CD36 in PBMCs were measured by using real-time quantitative reverse transcription polymerase chain reaction. RESULTS Plasma triglyceride levels were significantly increased immediately after the race and dropped at 24-hour post-race compared with pre-race values. The 100-km ultramarathon boosted high-density lipoprotein cholesterol (HDL-C) levels and decreased low-density lipoprotein cholesterol (LDL-C) levels 24-hour post-race. The expression levels of ABCA1, ABCG1 and SR-BI were markedly decreased, whereas that of CD36 was slightly but significantly upregulated in runners' PBMCs immediately after the race. Ultramarathon resulted in immediate large-scale stimulation of inflammatory cytokines with increased plasma interleukin-6 and tumour necrosis factor-alpha levels. Moreover, by using in vitro models with human monocytic cell lines, incubation of runners' plasma immediately after the race significantly downregulated ABCA1 and ABCG1, and upregulated CD36 expression in these cells. CONCLUSIONS ABCA1, ABCG1 and CD36 gene expressions in PBMCS might be associated with endurance exercise-induced plasma cholesterol homeostasis and systemic inflammatory response.
Collapse
Affiliation(s)
- Chi-Chang Juan
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Li-Hua Li
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Ph.D. Program of Medical Biotechnology, Taipei Medical University, Taipei, Taiwan
| | - Sen-Kuang Hou
- Department of Emergency Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Emergency and Critical Care Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ping-Shiou Liu
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Fong Kao
- Department of Emergency Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Emergency and Critical Care Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yu-Hui Chiu
- Department of Emergency Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Emergency Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chorng-Kuang How
- Emergency Department, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Emergency Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Kinmen Hospital, Ministry of Health and Welfare, Kinmen, Taiwan
| |
Collapse
|
21
|
Reich N, Hölscher C. Acylated Ghrelin as a Multi-Targeted Therapy for Alzheimer's and Parkinson's Disease. Front Neurosci 2020; 14:614828. [PMID: 33381011 PMCID: PMC7767977 DOI: 10.3389/fnins.2020.614828] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Much thought has been given to the impact of Amyloid Beta, Tau and Alpha-Synuclein in the development of Alzheimer's disease (AD) and Parkinson's disease (PD), yet the clinical failures of the recent decades indicate that there are further pathological mechanisms at work. Indeed, besides amyloids, AD and PD are characterized by the culminative interplay of oxidative stress, mitochondrial dysfunction and hyperfission, defective autophagy and mitophagy, systemic inflammation, BBB and vascular damage, demyelination, cerebral insulin resistance, the loss of dopamine production in PD, impaired neurogenesis and, of course, widespread axonal, synaptic and neuronal degeneration that leads to cognitive and motor impediments. Interestingly, the acylated form of the hormone ghrelin has shown the potential to ameliorate the latter pathologic changes, although some studies indicate a few complications that need to be considered in the long-term administration of the hormone. As such, this review will illustrate the wide-ranging neuroprotective properties of acylated ghrelin and critically evaluate the hormone's therapeutic benefits for the treatment of AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical & Life Sciences Division, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, A Second Hospital, Shanxi Medical University, Taiyuan, China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
22
|
Cuthbert GA, Shaik F, Harrison MA, Ponnambalam S, Homer-Vanniasinkam S. Scavenger Receptors as Biomarkers and Therapeutic Targets in Cardiovascular Disease. Cells 2020; 9:cells9112453. [PMID: 33182772 PMCID: PMC7696859 DOI: 10.3390/cells9112453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
The process of atherosclerosis leads to the formation of plaques in the arterial wall, resulting in a decreased blood supply to tissues and organs and its sequelae: morbidity and mortality. A class of membrane-bound proteins termed scavenger receptors (SRs) are closely linked to the initiation and progression of atherosclerosis. Increasing interest in understanding SR structure and function has led to the idea that these proteins could provide new routes for cardiovascular disease diagnosis, management, and treatment. In this review, we consider the main classes of SRs that are implicated in arterial disease. We consider how our understanding of SR-mediated recognition of diverse ligands, including modified lipid particles, lipids, and carbohydrates, has enabled us to better target SR-linked functionality in disease. We also link clinical studies on vascular disease to our current understanding of SR biology and highlight potential areas that are relevant to cardiovascular disease management and therapy.
Collapse
Affiliation(s)
- Gary A. Cuthbert
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK;
- Correspondence: ; Tel.:+44 113 3433007
| | - Faheem Shaik
- School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; (F.S.); (S.P.)
| | | | - Sreenivasan Ponnambalam
- School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; (F.S.); (S.P.)
| | | |
Collapse
|
23
|
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: 123] [Impact Index Per Article: 24.6] [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
|
24
|
Frégeau G, Sarduy R, Elimam H, Esposito CL, Mellal K, Ménard L, Leitão da Graça SD, Proulx C, Zhang J, Febbraio M, Soto Y, Lubell WD, Ong H, Marleau S. Atheroprotective and atheroregressive potential of azapeptide derivatives of GHRP-6 as selective CD36 ligands in apolipoprotein E-deficient mice. Atherosclerosis 2020; 307:52-62. [PMID: 32721647 DOI: 10.1016/j.atherosclerosis.2020.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 06/08/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Scavenger receptor class B member 3, also known as cluster of differentiation-36 (CD36) receptor, is involved in the uptake and accumulation of modified lipoprotein in macrophages, driving atherosclerosis progression. Azapeptide analogs of growth hormone-releasing peptide-6 (GHRP-6) have been developed as selective CD36 ligands and evaluated for their anti-atherosclerotic properties in apoe-/- mice. METHODS From 4 to 19 weeks of age, male apoe-/- mice were fed a high fat high cholesterol (HFHC) diet, then switched to normal chow and treated daily with 300 nmol/kg of MPE-001 ([aza-Tyr4]-GHRP-6) or MPE-003 ([aza-(N,N-diallylaminobut-2-ynyl)Gly4]-GHRP-6) for 9 weeks. In another protocol, mice were fed a HFHC diet throughout the study. RESULTS Azapeptides decreased lesion progression in the aortic arch and reduced aortic sinus lesion areas below pre-existing lesions levels in apoe-/- mice which were switched to chow diet. In mice fed a HFHC throughout the study, azapeptides reduced lesion progression in the aortic vessel and sinus. The anti-atherosclerotic effect of azapeptides was associated with a reduced ratio of iNOS+/CD206+ macrophages within lesions, and lowered plasma inflammatory cytokine levels. Monocytes from azapeptide-treated mice showed altered mitochondrial oxygen consumption rates, consistent with an M2-like phenotype. These effects were dependent on CD36, and not observed in apoe-/-cd36-/- mice. CONCLUSIONS Azapeptides MPE-001 and MPE-003 diminished aortic lesion progression and reduced, below pre-existing levels, lesions in the aortic sinus of atherosclerotic mice. A relative increase of M2-like macrophages was observed in lesions, associated with reduced systemic inflammation. Development of CD36-selective azapeptide ligands merits consideration for treating atherosclerotic disease.
Collapse
Affiliation(s)
- Geneviève Frégeau
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Roger Sarduy
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Hanan Elimam
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada; Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Cloé L Esposito
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Katia Mellal
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Liliane Ménard
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | | | - Caroline Proulx
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - Jinqiang Zhang
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - Maria Febbraio
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yosdel Soto
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
25
|
Kim E, Yang J, Woo Park K, Cho S. Preventative, but not post-stroke, inhibition of CD36 attenuates brain swelling in hyperlipidemic stroke. J Cereb Blood Flow Metab 2020; 40:885-894. [PMID: 31092085 PMCID: PMC7168788 DOI: 10.1177/0271678x19850004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The lack of inclusion of comorbidities in animal models of stroke may underlie the limited development of therapy in stroke. Previous studies in mice deficient of CD36, an immune receptor, indicated its contribution to stroke-induced inflammation and injury in hyperlipidemic conditions. The current study, therefore, tested whether pharmacological inhibition of CD36 provides neuroprotection in hyperlipidemic stroke. The hyperlipidemic mice subjected to stroke showed an exacerbation of infarct size and profound brain swelling. However, post-stroke treatment with CD36 inhibitors did not reduce, and in some cases worsened, acute stroke outcome, suggesting potential benefits of elevated CD36 in the post-stroke brain in a hyperlipidemic condition. On the other hand, chronic treatment of a CD36 inhibitor prior to stroke significantly reduced stroke-induced brain swelling. There was a trend toward infarct reduction, although it did not reach statistical significance. The observed benefit of preventative CD36 inhibition is in line with previously reported smaller infarct volume and swelling in CD36 KO mice. Thus, the current findings suggest that insights gained from the genetic models should be carefully considered before the implementation of pharmacological interventions, as a potential therapeutic strategy may depend on preventative treatment or a post-stroke acute treatment paradigm.
Collapse
Affiliation(s)
- Eunhee Kim
- Burke Neurological Institute, White Plains, NY, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.,Vivian L Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jiwon Yang
- Burke Neurological Institute, White Plains, NY, USA
| | | | - Sunghee Cho
- Burke Neurological Institute, White Plains, NY, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
26
|
Abstract
Cardiovascular disease (CVD) is the number one cause of death in the United States and worldwide. The most common cause of cardiovascular disease is atherosclerosis, or formation of fatty plaques in the arteries. Low-density lipoprotein (LDL), termed "bad cholesterol", is a large molecule comprised of many proteins as well as lipids including cholesterol, phospholipids, and triglycerides. Circulating levels of LDL are directly associated with atherosclerosis disease severity. Once thought to simply be caused by passive retention of LDL in the vasculature, atherosclerosis studies over the past 40-50 years have uncovered a much more complex mechanism. It has now become well established that within the vasculature, LDL can undergo many different types of oxidative modifications such as esterification and lipid peroxidation. The resulting oxidized LDL (oxLDL) has been found to have antigenic potential and contribute heavily to atherosclerosis associated inflammation, activating both innate and adaptive immunity. This review discusses the many proposed mechanisms by which oxidized LDL modulates inflammatory responses and how this might modulate atherosclerosis.
Collapse
Affiliation(s)
- Jillian P Rhoads
- Department of Medicine, Division of Rheumatology, Vanderbilt Medical Center, Nashville, TN 37232
| | - Amy S Major
- Department of Medicine, Division of Rheumatology, Vanderbilt Medical Center, Nashville, TN 37232; Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN 37212
| |
Collapse
|
27
|
Zoccal KF, Gardinassi LG, Bordon KCF, Arantes EC, Marleau S, Ong H, Faccioli LH. EP80317 Restrains Inflammation and Mortality Caused by Scorpion Envenomation in Mice. Front Pharmacol 2019; 10:171. [PMID: 30886580 PMCID: PMC6409428 DOI: 10.3389/fphar.2019.00171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/11/2019] [Indexed: 11/13/2022] Open
Abstract
Over 1 million cases of scorpion stings are estimated every year, whereas current treatment is limited to antivenom serum combined with supportive therapy. Tityus serrulatus scorpion venom (TsV) is composed of diverse molecules, including toxins that induce a catecholamine storm and mediate classical symptoms of scorpion envenomation. However, the same toxins promote an intense inflammatory response coordinated by innate immune cells, such as macrophages, contributing significantly to the lung edema and mortality caused by TsV injection. Macrophages sense TsV via innate immune receptors, including TLR2, TLR4, and CD14 that promote inflammation and mortality via PGE2/cAMP/PKA/NF-κB/IL-1β axis. The scavenger receptor CD36 also recognizes TsV, but in contrast to the other receptors, it drives the production of leukotriene B4 (LTB4). This lipid mediator operates via BLT1 receptor to reduce cAMP production and consequently IL-1β release, which results in resistance to fatal outcomes of experimental scorpion envenomation. EP80317 is an hexapeptide that serves as a ligand for CD36 and features protective effects under conditions such as atherosclerosis and vascular inflammation. In this study, we evaluated the effects of EP80317 treatment during experimental scorpion envenomation. EP80317 treatment suppressed mouse peritoneal macrophage production of IL-1β, IL-6, tumor necrosis factor (TNF-α), CCL3, and PGE2 in vitro. EP80317 treatment also boosted the production of LTB4 and IL-10 in response to TsV. Importantly, EP80317 restrained lung inflammation and mortality caused by TsV in vivo. Taken together, these data indicate a strong therapeutic potential of EP80317 as a supportive treatment to control inflammation induced by scorpion envenomation.
Collapse
Affiliation(s)
- Karina F Zoccal
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.,Centro Universitário Barão de Mauá, Ribeirão Preto, Brazil
| | - Luiz G Gardinassi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Karla C F Bordon
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Eliane C Arantes
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Sylvie Marleau
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Huy Ong
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
28
|
Håversen L, Sundelin JP, Mardinoglu A, Rutberg M, Ståhlman M, Wilhelmsson U, Hultén LM, Pekny M, Fogelstrand P, Bentzon JF, Levin M, Borén J. Vimentin deficiency in macrophages induces increased oxidative stress and vascular inflammation but attenuates atherosclerosis in mice. Sci Rep 2018; 8:16973. [PMID: 30451917 PMCID: PMC6242955 DOI: 10.1038/s41598-018-34659-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022] Open
Abstract
The aim was to clarify the role of vimentin, an intermediate filament protein abundantly expressed in activated macrophages and foam cells, in macrophages during atherogenesis. Global gene expression, lipid uptake, ROS, and inflammation were analyzed in bone-marrow derived macrophages from vimentin-deficient (Vim-/-) and wild-type (Vim+/+) mice. Atherosclerosis was induced in Ldlr-/- mice transplanted with Vim-/- and Vim+/+ bone marrow, and in Vim-/- and Vim+/+ mice injected with a PCSK9 gain-of-function virus. The mice were fed an atherogenic diet for 12-15 weeks. We observed impaired uptake of native LDL but increased uptake of oxLDL in Vim-/- macrophages. FACS analysis revealed increased surface expression of the scavenger receptor CD36 on Vim-/- macrophages. Vim-/- macrophages also displayed increased markers of oxidative stress, activity of the transcription factor NF-κB, secretion of proinflammatory cytokines and GLUT1-mediated glucose uptake. Vim-/- mice displayed decreased atherogenesis despite increased vascular inflammation and increased CD36 expression on macrophages in two mouse models of atherosclerosis. We demonstrate that vimentin has a strong suppressive effect on oxidative stress and that Vim-/- mice display increased vascular inflammation with increased CD36 expression on macrophages despite decreased subendothelial lipid accumulation. Thus, vimentin has a key role in regulating inflammation in macrophages during atherogenesis.
Collapse
Affiliation(s)
- Liliana Håversen
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jeanna Perman Sundelin
- Strategic planning and operations, Cardiovascular and metabolic diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, SE1 9RT, United Kingdom
| | - Mikael Rutberg
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ulrika Wilhelmsson
- Department of Clinical Neuroscience/Center for Brain Repair, University of Gothenburg, Gothenburg, Sweden
| | - Lillemor Mattsson Hultén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Milos Pekny
- Department of Clinical Neuroscience/Center for Brain Repair, University of Gothenburg, Gothenburg, Sweden
| | - Per Fogelstrand
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jacob Fog Bentzon
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark, and Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Malin Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
29
|
Maguire EM, Pearce SWA, Xiao Q. Foam cell formation: A new target for fighting atherosclerosis and cardiovascular disease. Vascul Pharmacol 2018; 112:54-71. [PMID: 30115528 DOI: 10.1016/j.vph.2018.08.002] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/17/2018] [Accepted: 08/03/2018] [Indexed: 12/23/2022]
Abstract
During atherosclerosis, the gradual accumulation of lipids into the subendothelial space of damaged arteries results in several lipid modification processes followed by macrophage uptake in the arterial wall. The way in which these modified lipoproteins are dealt with determines the likelihood of cholesterol accumulation within the monocyte-derived macrophage and thus its transformation into the foam cell that makes up the characteristic fatty streak observed in the early stages of atherosclerosis. The unique expression of chemokine receptors and cellular adhesion molecules expressed on the cell surface of monocytes points to a particular extravasation route that they can take to gain entry into atherosclerotic site, in order to undergo differentiation into the phagocytic macrophage. Indeed several GWAS and animal studies have identified key genes and proteins required for monocyte recruitment as well cholesterol handling involving lipid uptake, cholesterol esterification and cholesterol efflux. A re-examination of the previously accepted paradigm of macrophage foam cell origin has been called into question by recent studies demonstrating shared expression of scavenger receptors, cholesterol transporters and pro-inflammatory cytokine release by alternative cell types present in the neointima, namely; endothelial cells, vascular smooth muscle cells and stem/progenitor cells. Thus, therapeutic targets aimed at a more heterogeneous foam cell population with shared functions, such as enhanced protease activity, and signalling pathways, mediated by non-coding RNA molecules, may provide greater therapeutic outcome in patients. Finally, studies targeting each aspect of foam cell formation and death using both genetic knock down and pharmacological inhibition have provided researchers with a clearer understanding of the cellular processes at play, as well as helped researchers to identify key molecular targets, which may hold significant therapeutic potential in the future.
Collapse
Affiliation(s)
- Eithne M Maguire
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Stuart W A Pearce
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
30
|
Maréchal L, Laviolette M, Rodrigue-Way A, Sow B, Brochu M, Caron V, Tremblay A. The CD36-PPARγ Pathway in Metabolic Disorders. Int J Mol Sci 2018; 19:1529. [PMID: 29883404 PMCID: PMC5983591 DOI: 10.3390/ijms19051529] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/08/2018] [Accepted: 05/16/2018] [Indexed: 12/21/2022] Open
Abstract
Uncovering the biological role of nuclear receptor peroxisome proliferator-activated receptors (PPARs) has greatly advanced our knowledge of the transcriptional control of glucose and energy metabolism. As such, pharmacological activation of PPARγ has emerged as an efficient approach for treating metabolic disorders with the current use of thiazolidinediones to improve insulin resistance in diabetic patients. The recent identification of growth hormone releasing peptides (GHRP) as potent inducers of PPARγ through activation of the scavenger receptor CD36 has defined a novel alternative to regulate essential aspects of lipid and energy metabolism. Recent advances on the emerging role of CD36 and GHRP hexarelin in regulating PPARγ downstream actions with benefits on atherosclerosis, hepatic cholesterol biosynthesis and fat mitochondrial biogenesis are summarized here. The response of PPARγ coactivator PGC-1 is also discussed in these effects. The identification of the GHRP-CD36-PPARγ pathway in controlling various tissue metabolic functions provides an interesting option for metabolic disorders.
Collapse
Affiliation(s)
- Loïze Maréchal
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Physiology, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Maximilien Laviolette
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Amélie Rodrigue-Way
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Baly Sow
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Michèle Brochu
- Department of Physiology, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Véronique Caron
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
| | - André Tremblay
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
- Centre de Recherche en Reproduction et Fertilité, University of Montreal, Saint Hyacinthe, QC J2S 7C6, Canada.
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1C5, Canada.
| |
Collapse
|
31
|
Huynh DN, Bessi VL, Ménard L, Piquereau J, Proulx C, Febbraio M, Lubell WD, Carpentier AC, Burelle Y, Ong H, Marleau S. Adiponectin has a pivotal role in the cardioprotective effect of CP-3(iv), a selective CD36 azapeptide ligand, after transient coronary artery occlusion in mice. FASEB J 2018; 32:807-818. [PMID: 29018142 DOI: 10.1096/fj.201700505r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CD36 is a multiligand receptor involved in lipid metabolism. We investigated the mechanisms underlying the cardioprotective effect of CP-3(iv), an azapeptide belonging to a new class of selective CD36 ligands. The role of CP-3(iv) in mediating cardioprotection was investigated because CD36 signaling leads to activation of peroxisome proliferator-activated receptor-γ, a transcriptional regulator of adiponectin. CP-3(iv) pretreatment reduced infarct size by 54% and preserved hemodynamics in C57BL/6 mice subjected to 30 min coronary ligation and reperfusion but had no effect in CD36-deficient mice. The effects of CP-3(iv) were associated with an increase in circulating adiponectin levels, epididymal fat adiponectin gene expression, and adiponectin transcriptional regulators ( Pparg, Cebpb, Sirt1) after 6 h of reperfusion. Reduced myocardial oxidative stress and apoptosis were observed along with an increase in expression of myocardial adiponectin target proteins, including cyclooxygenase-2, phospho-AMPK, and phospho-Akt. Moreover, CP-3(iv) increased myocardial performance in isolated hearts, whereas blockade of adiponectin with an anti-adiponectin antibody abrogated it. CP-3(iv) exerts cardioprotection against myocardial ischemia and reperfusion (MI/R) injury and dysfunction, at least in part, by increasing circulating and myocardial adiponectin levels. Hence, both paracrine and endocrine effects of adiponectin may contribute to reduced reactive oxygen species generation and apoptosis after MI/R, in a CD36-dependent manner.-Huynh, D. N., Bessi, V. L., Ménard, L., Piquereau, J., Proulx, C., Febbraio, M., Lubell, W. D., Carpentier, A. C., Burelle, Y., Ong, H., Marleau, S. Adiponectin has a pivotal role in the cardioprotective effect of CP-3(iv), a selective CD36 azapeptide ligand, after transient coronary artery occlusion in mice.
Collapse
Affiliation(s)
- David N Huynh
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Valérie L Bessi
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Liliane Ménard
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Jérôme Piquereau
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Caroline Proulx
- Department of Chemistry, Université de Montréal, Montreal, Quebec, Canada
| | - Maria Febbraio
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montreal, Quebec, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Yan Burelle
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
32
|
Ooi BK, Goh BH, Yap WH. Oxidative Stress in Cardiovascular Diseases: Involvement of Nrf2 Antioxidant Redox Signaling in Macrophage Foam Cells Formation. Int J Mol Sci 2017; 18:ijms18112336. [PMID: 29113088 PMCID: PMC5713305 DOI: 10.3390/ijms18112336] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/11/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is an important risk factor contributing to the pathogenesis of cardiovascular diseases. Oxidative stress that results from excessive reactive oxygen species (ROS) production accounts for impaired endothelial function, a process which promotes atherosclerotic lesion or fatty streaks formation (foam cells). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor involved in cellular redox homeostasis. Upon exposure to oxidative stress, Nrf2 is dissociated from its inhibitor Keap-1 and translocated into the nucleus, where it results in the transcriptional activation of cell defense genes. Nrf2 has been demonstrated to be involved in the protection against foam cells formation by regulating the expression of antioxidant proteins (HO-1, Prxs, and GPx1), ATP-binding cassette (ABC) efflux transporters (ABCA1 and ABCG1) and scavenger receptors (scavenger receptor class B (CD36), scavenger receptor class A (SR-A) and lectin-type oxidized LDL receptor (LOX-1)). However, Nrf2 has also been reported to exhibit pro-atherogenic effects. A better understanding on the mechanism of Nrf2 in oxidative stress-induced cardiac injury, as well as the regulation of cholesterol uptake and efflux, are required before it can serve as a novel therapeutic target for cardiovascular diseases prevention and treatment.
Collapse
Affiliation(s)
- Bee Kee Ooi
- School of Biosciences, Taylor's University, Subang Jaya, Selangor Darul Ehsan 47500, Malaysia.
| | - Bey Hing Goh
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Wei Hsum Yap
- School of Biosciences, Taylor's University, Subang Jaya, Selangor Darul Ehsan 47500, Malaysia.
| |
Collapse
|
33
|
Oxidative Stress in Cardiovascular Diseases: Involvement of Nrf2 Antioxidant Redox Signaling in Macrophage Foam Cells Formation. Int J Mol Sci 2017. [PMID: 29113088 DOI: 10.3390/ijms18112336.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Oxidative stress is an important risk factor contributing to the pathogenesis of cardiovascular diseases. Oxidative stress that results from excessive reactive oxygen species (ROS) production accounts for impaired endothelial function, a process which promotes atherosclerotic lesion or fatty streaks formation (foam cells). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor involved in cellular redox homeostasis. Upon exposure to oxidative stress, Nrf2 is dissociated from its inhibitor Keap-1 and translocated into the nucleus, where it results in the transcriptional activation of cell defense genes. Nrf2 has been demonstrated to be involved in the protection against foam cells formation by regulating the expression of antioxidant proteins (HO-1, Prxs, and GPx1), ATP-binding cassette (ABC) efflux transporters (ABCA1 and ABCG1) and scavenger receptors (scavenger receptor class B (CD36), scavenger receptor class A (SR-A) and lectin-type oxidized LDL receptor (LOX-1)). However, Nrf2 has also been reported to exhibit pro-atherogenic effects. A better understanding on the mechanism of Nrf2 in oxidative stress-induced cardiac injury, as well as the regulation of cholesterol uptake and efflux, are required before it can serve as a novel therapeutic target for cardiovascular diseases prevention and treatment.
Collapse
|
34
|
Mosa R, Huang L, Wu Y, Fung C, Mallawakankanamalage O, LeRoith D, Chen C. Hexarelin, a Growth Hormone Secretagogue, Improves Lipid Metabolic Aberrations in Nonobese Insulin-Resistant Male MKR Mice. Endocrinology 2017; 158:3174-3187. [PMID: 28977588 PMCID: PMC5659698 DOI: 10.1210/en.2017-00168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/10/2017] [Indexed: 12/16/2022]
Abstract
Despite the occurrence of dyslipidemia and its contribution to the development of insulin resistance in obese subjects, a growing number of studies have described abnormal lipid profiles among leaner persons. For example, individuals with an abnormal paucity or distribution of fat (lipodystrophy) develop severe insulin resistance, dyslipidemia, and hepatic steatosis. Deranged adipocyte metabolism and differentiation contribute to ectopic fat deposition and consequent development of insulin resistance. Growth hormone (GH) therapy has been shown to correct body composition abnormalities in some lipodystrophy patients. However, little is known about the effects of GH-releasing peptides in this regard. Hexarelin, a GH secretagogue, has recently been shown to have beneficial effects on fat metabolism via the CD36 receptor. In this study, the effects of twice daily intraperitoneal injections of hexarelin (200 μg/kg body weight) were examined in nonobese insulin-resistant MKR mice and corresponding wild-type FVB mice for 12 days. Hexarelin treatment significantly improved glucose and insulin intolerance and decreased plasma and liver triglycerides in MKR mice. These beneficial metabolic effects could be due to the improved lipid metabolism and enhanced adipocyte differentiation of white adipose tissue with hexarelin treatment. Interestingly, although food intake of hexarelin-treated MKR mice was significantly increased, this did not change total body weight. Moreover, hexarelin treatment corrected the abnormal body composition of MKR mice, as demonstrated by a decrease in fat mass and an increase in lean mass. Our results suggest a possible application of hexarelin in treatment of lipid disorders associated with the metabolic syndrome.
Collapse
Affiliation(s)
- Rasha Mosa
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Lili Huang
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Yeda Wu
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Chungyan Fung
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Oshini Mallawakankanamalage
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Derek LeRoith
- Clinical Endocrinology Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Chen Chen
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
35
|
CD36 in chronic kidney disease: novel insights and therapeutic opportunities. Nat Rev Nephrol 2017; 13:769-781. [DOI: 10.1038/nrneph.2017.126] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Chistiakov DA, Melnichenko AA, Orekhov AN, Bobryshev YV. How do macrophages sense modified low-density lipoproteins? Int J Cardiol 2017; 230:232-240. [DOI: 10.1016/j.ijcard.2016.12.164] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/19/2016] [Accepted: 12/25/2016] [Indexed: 01/18/2023]
|
37
|
Watson DC, Bayik D, Srivatsan A, Bergamaschi C, Valentin A, Niu G, Bear J, Monninger M, Sun M, Morales-Kastresana A, Jones JC, Felber BK, Chen X, Gursel I, Pavlakis GN. Efficient production and enhanced tumor delivery of engineered extracellular vesicles. Biomaterials 2016; 105:195-205. [PMID: 27522254 DOI: 10.1016/j.biomaterials.2016.07.003] [Citation(s) in RCA: 284] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EV), including exosomes and microvesicles, are nano-sized intercellular communication vehicles that participate in a multitude of physiological processes. Due to their biological properties, they are also promising candidates for the systemic delivery of therapeutic compounds, such as cytokines, chemotherapeutic drugs, siRNAs and viral vectors. However, low EV production yield and rapid clearance of administered EV by liver macrophages limit their potential use as therapeutic vehicles. We have used a hollow-fiber bioreactor for the efficient production of bioactive EV bearing the heterodimeric cytokine complex Interleukin-15:Interleukin-15 receptor alpha. Bioreactor culture yielded ∼40-fold more EV per mL conditioned medium, as compared to conventional cell culture. Biophysical analysis and comparative proteomics suggested a more diverse population of EV in the bioreactor preparations, while serum protein contaminants were detectable only in conventional culture EV preparations. We also identified the Scavenger Receptor Class A family (SR-A) as a novel monocyte/macrophage uptake receptor for EV. In vivo blockade of SR-A with dextran sulfate dramatically decreased EV liver clearance in mice, while enhancing tumor accumulation. These findings facilitate development of EV therapeutic methods.
Collapse
Affiliation(s)
- Dionysios C Watson
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States; Department of Medicine, University of Patras, Greece
| | - Defne Bayik
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States; Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800 Turkey
| | - Avinash Srivatsan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, United States
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, United States
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
| | - Mitchell Monninger
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, United States
| | - Mei Sun
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, United States
| | - Aizea Morales-Kastresana
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States
| | - Jennifer C Jones
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ihsan Gursel
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800 Turkey
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States.
| |
Collapse
|
38
|
Zhang J, Zu Y, Dhanasekara CS, Li J, Wu D, Fan Z, Wang S. Detection and treatment of atherosclerosis using nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27241794 DOI: 10.1002/wnan.1412] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/25/2016] [Accepted: 04/12/2016] [Indexed: 01/10/2023]
Abstract
Atherosclerosis is the key pathogenesis of cardiovascular disease, which is a silent killer and a leading cause of death in the United States. Atherosclerosis starts with the adhesion of inflammatory monocytes on the activated endothelial cells in response to inflammatory stimuli. These monocytes can further migrate into the intimal layer of the blood vessel where they differentiate into macrophages, which take up oxidized low-density lipoproteins and release inflammatory factors to amplify the local inflammatory response. After accumulation of cholesterol, the lipid-laden macrophages are transformed into foam cells, the hallmark of the early stage of atherosclerosis. Foam cells can die from apoptosis or necrosis, and the intracellular lipid is deposed in the artery wall forming lesions. The angiogenesis for nurturing cells is enhanced during lesion development. Proteases released from macrophages, foam cells, and other cells degrade the fibrous cap of the lesion, resulting in rupture of the lesion and subsequent thrombus formation. Thrombi can block blood circulation, which represents a major cause of acute heart events and stroke. There are generally no symptoms in the early stages of atherosclerosis. Current detection techniques cannot easily, safely, and effectively detect the lesions in the early stages, nor can they characterize the lesion features such as the vulnerability. While the available therapeutic modalities cannot target specific molecules, cells, and processes in the lesions, nanoparticles appear to have a promising potential in improving atherosclerosis detection and treatment via targeting the intimal macrophages, foam cells, endothelial cells, angiogenesis, proteolysis, apoptosis, and thrombosis. Indeed, many nanoparticles have been developed in improving blood lipid profile and decreasing inflammatory response for enhancing therapeutic efficacy of drugs and decreasing their side effects. WIREs Nanomed Nanobiotechnol 2017, 9:e1412. doi: 10.1002/wnan.1412 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | | | - Jun Li
- Laboratory Animal Center, Peking University, Beijing, PR China
| | - Dayong Wu
- Nutritional Immunology Laboratory, Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Zhaoyang Fan
- Department of Electrical and Computer Engineering and Nano Tech Center, Texas Tech University, Lubbock, TX, USA
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
39
|
Xue XH, Shi FF, Chen T, Wei W, Zhou XM, Chen LD. Inhibition of ERK1/2 improves lipid balance in rat macrophages via ABCA1/G1 and CD36. Mol Med Rep 2015; 13:1533-40. [PMID: 26707062 DOI: 10.3892/mmr.2015.4697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 12/07/2015] [Indexed: 11/06/2022] Open
Abstract
ATP-binding cassette transporters A1 (ABCA1) and G1 (ABCG1), and macrophage scavenger receptor, cluster of differentiation (CD)36, function as key mediators of cholesterol efflux and influx from macrophages. In addition, they are associated with foam cell formation and the development of atherosclerosis (AS). The aim of the present study was to investigate the effects of extracellular signal-regulated kinases 1/2 (ERK1/2) inhibition on lipid balance in oxidized-low-density lipoprotein (Ox-LDL)-stimulated rat macrophages, and to examine the role of ERK1/2 inhibitors in AS. Rat peritoneal macrophages were treated with Ox-LDL alone or in combination with an ERK1/2 inhibitor, U0126, and untreated cells served as controls. Ox-LDL-induced lipid accumulation was detected by DiI fluorescence and oil red O staining. In addition, the mRNA and protein expression levels of ABCA1, ABCG1 and CD36 were determined using polymerase chain reaction and western blotting, respectively. Treatment with Ox-LDL significantly increased lipid accumulation and upregulated the mRNA and protein expression levels of ABCA1, ABCG1 and CD36 in macrophages. The addition of U0126 resulted in a marked reduction of lipid deposition, upregulation of ABCA1/G1 expression and suppression of CD36 expression in Ox-LDL-stimulated macrophages. The results of the present study indicated a novel association between ERK1/2 signaling and lipid metabolism, thus suggesting that inhibition of ERK1/2 may be considered a promising therapeutic strategy against AS.
Collapse
Affiliation(s)
- Xie-Hua Xue
- Department of Neurology, Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Feng-Fei Shi
- Institute of Rehabilitation Medicine, Rehabilitation Technology Collaborative Innovation Center, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Tong Chen
- Institute of Rehabilitation Medicine, Rehabilitation Technology Collaborative Innovation Center, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Wei Wei
- Department of Neurology, Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Xiao-Mao Zhou
- Institute of Rehabilitation Medicine, Rehabilitation Technology Collaborative Innovation Center, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Li-Dian Chen
- Institute of Rehabilitation Medicine, Rehabilitation Technology Collaborative Innovation Center, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| |
Collapse
|
40
|
Mosa RMH, Zhang Z, Shao R, Deng C, Chen J, Chen C. Implications of ghrelin and hexarelin in diabetes and diabetes-associated heart diseases. Endocrine 2015; 49:307-23. [PMID: 25645463 DOI: 10.1007/s12020-015-0531-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/12/2015] [Indexed: 02/07/2023]
Abstract
Ghrelin and its synthetic analog hexarelin are specific ligands of growth hormone secretagogue (GHS) receptor. GHS have strong growth hormone-releasing effect and other neuroendocrine activities such as stimulatory effects on prolactin and adrenocorticotropic hormone secretion. Recently, several studies have reported other beneficial functions of GHS that are independent of GH. Ghrelin and hexarelin, for examples, have been shown to exert GH-independent cardiovascular activity. Hexarelin has been reported to regulate peroxisome proliferator-activated receptor gamma (PPAR-γ) in macrophages and adipocytes. PPAR-γ is an important regulator of adipogenesis, lipid metabolism, and insulin sensitization. Ghrelin also shows protective effects on beta cells against lipotoxicity through activation of phosphatidylinositol-3 kinase/protein kinase B, c-Jun N-terminal kinase (JNK) inhibition, and nuclear exclusion of forkhead box protein O1. Acylated ghrelin (AG) and unacylated ghrelin (UAG) administration reduces glucose levels and increases insulin-producing beta cell number, and insulin secretion in pancreatectomized rats and in newborn rats treated with streptozotocin, suggesting a possible role of GHS in pancreatic regeneration. Therefore, the discovery of GHS has opened many new perspectives in endocrine, metabolic, and cardiovascular research areas, suggesting the possible therapeutic application in diabetes and diabetic complications especially diabetic cardiomyopathy. Here, we review the physiological roles of ghrelin and hexarelin in the protection and regeneration of beta cells and their roles in the regulation of insulin release, glucose, and fat metabolism and present their potential therapeutic effects in the treatment of diabetes and diabetic-associated heart diseases.
Collapse
|
41
|
Han X, Boisvert WA. Interleukin-10 protects against atherosclerosis by modulating multiple atherogenic macrophage function. Thromb Haemost 2014; 113:505-12. [PMID: 25373619 DOI: 10.1160/th14-06-0509] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/22/2014] [Indexed: 01/15/2023]
Abstract
Atherosclerosis is primarily a disorder of lipid metabolism, but there is also a prominent chronic inflammatory component that drives the atherosclerotic lesion progression in the artery wall. During hyperlipidaemic conditions, there is a rapid influx of circulating monocytes into the atherosclerosis-prone areas of the arterial intima. These infiltrated monocytes differentiate into macrophages and take up the atherogenic lipoproteins in the intima of the vessel wall that have been modified within the lesion environment. Interleukin (IL)-10 is a prototypic anti-inflammatory cytokine made primarily by the macrophages and Th2 subtype T lymphocytes. In terms of atherosclerosis its major roles include inhibition of macrophage activation as well as inhibition of matrix metalloproteinase, pro-inflammatory cytokines and cyclooxygenase-2 expression in lipid-loaded and activated macrophage foam cells. Recent discoveries suggest another important role of IL-10 in atherosclerosis: its ability to alter lipid metabolism in macrophages. The current review will highlight the present knowledge on multiple ways in which IL-10 mediates atherosclerosis. As macrophages play a critical role in all stages of atherosclerosis, the review will concentrate on how IL-10 regulates the activities of macrophages that are especially important in the development of atherosclerosis.
Collapse
Affiliation(s)
| | - William A Boisvert
- William A. Boisvert, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, USA, Tel.: +1 808 692 1567, Fax: +1 808 692 1973, E-mail:
| |
Collapse
|
42
|
Traoré M, Doan ND, Lubell WD. Diversity-Oriented Synthesis of Azapeptides with Basic Amino Acid Residues: Aza-Lysine, Aza-Ornithine, and Aza-Arginine. Org Lett 2014; 16:3588-91. [DOI: 10.1021/ol501586y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mariam Traoré
- Département de Chimie, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7
| | - Ngoc-Duc Doan
- Département de Chimie, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7
| | - William D. Lubell
- Département de Chimie, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7
| |
Collapse
|
43
|
Xu Y, Liu Q, Xu Y, Liu C, Wang X, He X, Zhu N, Liu J, Wu Y, Li Y, Li N, Feng T, Lai F, Zhang M, Hong B, Jiang JD, Si S. Rutaecarpine suppresses atherosclerosis in ApoE-/- mice through upregulating ABCA1 and SR-BI within RCT. J Lipid Res 2014; 55:1634-47. [PMID: 24908654 DOI: 10.1194/jlr.m044198] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Indexed: 11/20/2022] Open
Abstract
ABCA1 and scavenger receptor class B type I (SR-BI)/CD36 and lysosomal integral membrane protein II analogous 1 (CLA-1) are the key transporter and receptor in reverse cholesterol transport (RCT). Increasing the expression level of ABCA1 and SR-BI/CLA-1 is antiatherogenic. The aim of the study was to find novel antiatherosclerotic agents upregulating expression of ABCA1 and SR-BI/CLA-1 from natural compounds. Using the ABCA1p-LUC and CLA-1p-LUC HepG2 cell lines, we found that rutaecarpine (RUT) triggered promoters of ABCA1 and CLA-1 genes. RUT increased ABCA1 and SR-BI/CLA-1 expression in vitro related to liver X receptor alpha and liver X receptor beta. RUT induced cholesterol efflux in RAW264.7 cells. ApoE-deficient (ApoE(-/-)) mice treated with RUT for 8 weeks showed ∼68.43, 70.23, and 85.56% less en face lesions for RUT (L), RUT (M), and RUT (H) groups, respectively, compared with the model group. Mouse macrophage-specific antibody and filipin staining indicated that RUT attenuated macrophages and cholesterol accumulations in atherosclerotic lesions, respectively. Additionally, ABCA1 and SR-BI expression was highly induced by RUT in livers of ApoE(-/-) mice. Meanwhile, RUT treatment significantly increased the fecal (3)H-cholesterol excretion, which demonstrated that RUT could promote RCT in vivo. RUT was identified to be a candidate that protected ApoE(-/-) mice from developing atherosclerosis through preferentially promoting activities of ABCA1 and SR-BI within RCT.
Collapse
Affiliation(s)
- Yanni Xu
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qi Liu
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yang Xu
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chang Liu
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiao Wang
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiaobo He
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ningyu Zhu
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jikai Liu
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yexiang Wu
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongzhen Li
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ni Li
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Tingting Feng
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Fangfang Lai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Murui Zhang
- Sir Runrun Shaw Hospital of Zhejiang University, Hangzhou, Zhejiang Province 310016, China
| | - Bin Hong
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jian-Dong Jiang
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shuyi Si
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
44
|
Park YM. CD36, a scavenger receptor implicated in atherosclerosis. Exp Mol Med 2014; 46:e99. [PMID: 24903227 PMCID: PMC4081553 DOI: 10.1038/emm.2014.38] [Citation(s) in RCA: 377] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 03/17/2014] [Accepted: 04/08/2014] [Indexed: 12/17/2022] Open
Abstract
CD36 is a membrane glycoprotein that is present on various types of cells, including monocytes, macrophages, microvascular endothelial cells, adipocytes and platelets. Macrophage CD36 participates in atherosclerotic arterial lesion formation through its interaction with oxidized low-density lipoprotein (oxLDL), which triggers signaling cascades for inflammatory responses. CD36 functions in oxLDL uptake and foam cell formation, which is the initial critical stage of atherosclerosis. In addition, oxLDL via CD36 inhibits macrophage migration, which may be a macrophage-trapping mechanism in atherosclerotic lesions. The role of CD36 was examined in in vitro studies and in vivo experiments, which investigated various functions of CD36 in atherosclerosis and revealed that CD36 deficiency reduces atherosclerotic lesion formation. Platelet CD36 also promotes atherosclerotic inflammatory processes and is involved in thrombus formation after atherosclerotic plaque rupture. Because CD36 is an essential component of atherosclerosis, defining the function of CD36 and its corresponding signaling pathway may lead to a new treatment strategy for atherosclerosis.
Collapse
Affiliation(s)
- Young Mi Park
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
- Ewha Global Top 5 Research Program, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
45
|
Rodrigue‐Way A, Caron V, Bilodeau S, Keil S, Hassan M, Lévy E, Mitchell GA, Tremblay A. Scavenger receptor CD36 mediates inhibition of cholesterol synthesis viaactivation of the PPARγ/PGC‐1α pathway and Insig1/2 expression in hepatocytes. FASEB J 2014; 28:1910-1923. [DOI: 10.1096/fj.13-240168] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Amélie Rodrigue‐Way
- Research CenterSainte‐Justine HospitalMontréalQuébecCanada
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontréalQuébecCanada
| | | | - Stéphanie Bilodeau
- Research CenterSainte‐Justine HospitalMontréalQuébecCanada
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontréalQuébecCanada
| | - Sarah Keil
- Research CenterSainte‐Justine HospitalMontréalQuébecCanada
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontréalQuébecCanada
| | - Meryl Hassan
- Research CenterSainte‐Justine HospitalMontréalQuébecCanada
| | - Emile Lévy
- Research CenterSainte‐Justine HospitalMontréalQuébecCanada
| | - Grant A. Mitchell
- Research CenterSainte‐Justine HospitalMontréalQuébecCanada
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontréalQuébecCanada
- Department of PediatricsUniversity of MontrealMontréalQuébecCanada
| | - André Tremblay
- Research CenterSainte‐Justine HospitalMontréalQuébecCanada
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontréalQuébecCanada
- Department of Obstetrics and Gynecology, Faculty of MedicineUniversity of MontrealMontréalQuébecCanada
| |
Collapse
|
46
|
Valanti E, Tsompanidis A, Sanoudou D. Pharmacogenomics in the development and characterization of atheroprotective drugs. Methods Mol Biol 2014; 1175:259-300. [PMID: 25150873 DOI: 10.1007/978-1-4939-0956-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Atherosclerosis is the main cause of cardiovascular disease (CVD) and can lead to stroke, myocardial infarction, and death. The clinically available atheroprotective drugs aim mainly at reducing the levels of circulating low-density lipoprotein (LDL), increasing high-density lipoprotein (HDL), and attenuating inflammation. However, the cardiovascular risk remains high, along with morbidity, mortality, and incidence of adverse drug events. Pharmacogenomics is increasingly contributing towards the characterization of existing atheroprotective drugs, the evaluation of novel ones, and the identification of promising, unexplored therapeutic targets, at the global molecular pathway level. This chapter presents highlights of pharmacogenomics investigations and discoveries that have contributed towards the elucidation of pharmacological atheroprotection, while opening the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Efi Valanti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens, 115 27, Greece
| | | | | |
Collapse
|
47
|
Bujold K, Mellal K, Zoccal KF, Rhainds D, Brissette L, Febbraio M, Marleau S, Ong H. EP 80317, a CD36 selective ligand, promotes reverse cholesterol transport in apolipoprotein E-deficient mice. Atherosclerosis 2013; 229:408-14. [DOI: 10.1016/j.atherosclerosis.2013.05.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 05/08/2013] [Accepted: 05/31/2013] [Indexed: 01/05/2023]
|
48
|
Takahashi M, Yagyu H, Tazoe F, Nagashima S, Ohshiro T, Okada K, Osuga JI, Goldberg IJ, Ishibashi S. Macrophage lipoprotein lipase modulates the development of atherosclerosis but not adiposity. J Lipid Res 2013; 54:1124-34. [PMID: 23378601 DOI: 10.1194/jlr.m035568] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The role of macrophage lipoprotein lipase (LpL) in the development of atherosclerosis and adiposity was examined in macrophage LpL knockout (MLpLKO) mice. MLpLKO mice were generated using cre-loxP gene targeting. Loss of LpL in macrophages did not alter plasma LpL activity or lipoprotein levels. Incubation of apolipoprotein E (ApoE)-deficient β-VLDL with peritoneal macrophages from ApoE knockout mice lacking macrophage LpL (MLpLKO/ApoEKO) led to less cholesteryl ester formation than that found with ApoEKO macrophages. MLpLKO/ApoEKO macrophages had reduced intracellular triglyceride levels, with decreased CD36 and carnitine palmitoyltransferase-1 mRNA levels compared with ApoEKO macrophages, when incubated with VLDL. Although both MLpLKO/ApoEKO and ApoEKO mice developed comparable hypercholesterolemia in response to feeding with a Western-type diet for 12 weeks, atherosclerosis was less in MLpLKO/ApoEKO mice. Epididymal fat mass and gene expression levels associated with inflammation did not differ between the two groups. In conclusion, macrophage LpL plays an important role in the development of atherosclerosis but not adiposity.
Collapse
Affiliation(s)
- Manabu Takahashi
- Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Glatz JFC, Angin Y, Steinbusch LKM, Schwenk RW, Luiken JJFP. CD36 as a target to prevent cardiac lipotoxicity and insulin resistance. Prostaglandins Leukot Essent Fatty Acids 2013; 88:71-7. [PMID: 22580174 DOI: 10.1016/j.plefa.2012.04.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 04/19/2012] [Accepted: 04/20/2012] [Indexed: 11/17/2022]
Abstract
The fatty acid transporter and scavenger receptor CD36 is increasingly being implicated in the pathogenesis of insulin resistance and its progression towards type 2 diabetes and associated cardiovascular complications. The redistribution of CD36 from intracellular stores to the plasma membrane is one of the earliest changes occurring in the heart during diet induced obesity and insulin resistance. This elicits an increased rate of fatty acid uptake and enhanced incorporation into triacylglycerol stores and lipid intermediates to subsequently interfere with insulin-induced GLUT4 recruitment (i.e., insulin resistance). In the present paper we discuss the potential of CD36 to serve as a target to rectify abnormal myocardial fatty acid uptake rates in cardiac lipotoxic diseases. Two approaches are described: (i) immunochemical inhibition of CD36 present at the sarcolemma and (ii) interference with the subcellular recycling of CD36. Using in vitro model systems of high-fat diet induced insulin resistance, the results indicate the feasibility of using CD36 as a target for adaptation of cardiac metabolic substrate utilization. In conclusion, CD36 deserves further attention as a promising therapeutic target to redirect fatty acid fluxes in the body.
Collapse
Affiliation(s)
- Jan F C Glatz
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht CARIM, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
50
|
Li W, Wang D, Chi Y, Wang R, Zhang F, Ma G, Chen Z, Li J, Liu Z, Matsuura E, Liu Q. 7-Ketocholesteryl-9-carboxynonanoate enhances the expression of ATP-binding cassette transporter A1 via CD36. Atherosclerosis 2012. [PMID: 23200840 DOI: 10.1016/j.atherosclerosis.2012.10.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND CD36 signal transductions have been reported by a variety of lipid moiety of oxidized low-density lipoprotein (oxLDL), however, CD36 signal induced by 7-ketocholesteryl-9-carboxynonanoate (oxLig-1), a lipid moiety of oxLDL has not been elucidated. METHODS AND RESULTS OxLig-1 leads to activation and recruitment of Src kinase Fyn, Lyn and caveolin-1 to CD36 in J774A.1 cells, but not in CD36 knockdown cells. The mitogen-activated protein (MAP) kinases c-Jun N-terminal kinase 2 (JNK2) and extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) are specifically phosphorylated in J774A.1 cells after treatment with oxLig-1 and inhibited by pretreatment of Src inhibitor, AG1879. The expression of ABCA1 is up-regulated by treatment with oxLig-1in J774A.1 cells, and the increased expression of ABCA1 is dramatically down-regulated by pretreatment with pharmacological inhibitors of ERK (PD98059) and JNK (SP600125). CONCLUSIONS The specific CD36 signal induced by oxLig-1 initiated the activation of Fyn, Lyn, caveolin-1, JNK2 and ERK1/2, and resulted in the up-regulation of ABCA1.
Collapse
Affiliation(s)
- Wenzhe Li
- College of Life Science and Technology, Key Laboratory of Carbohydrate and Lipid Metabolism Research, Dalian University, 10-Xuefu Avenue, Dalian Economical and Technological Development Zone, Liaoning 116622, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|