1
|
Wang H, Zhang L, Liu H, Yang Y, Lu W, Cao X, Yang X, Qin Q, Song R, Feng D, Wang S, Bai T, He J. PDZK1 confers sensitivity to sunitinib in clear cell renal cell carcinoma by suppressing the PDGFR-β pathway. Br J Cancer 2024; 131:347-360. [PMID: 38822145 PMCID: PMC11263541 DOI: 10.1038/s41416-024-02725-4] [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: 09/11/2023] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Sunitinib has emerged as the primary treatment for advanced or metastatic clear cell renal cell carcinoma (ccRCC) due to its significant improvement in patients' average survival time. However, drug resistance and adverse effects of sunitinib pose challenges to its clinical benefits. METHODS The differentially expressed genes (DEGs) associated with sunitinib sensitivity and resistance in ccRCC were investigated. Cell counting kit-8, plate colony formation, flow cytometry and subcutaneous xenograft tumor model assays were employed to explore the effects of PDZK1 on ccRCC. Further research on the molecular mechanism was conducted through western blot, co-immunoprecipitation, immunofluorescence co-localization and immunohistochemical staining. RESULTS We elucidated that PDZK1 is significantly downregulated in sunitinib-resistant ccRCC specimens, and PDZK1 negatively regulates the phosphorylation of PDGFR-β and the activation of its downstream pathways through interaction with PDGFR-β. The dysregulated low levels of PDZK1 contribute to inadequate inhibition of cell proliferation, tumor growth, and insensitivity to sunitinib treatment. Notably, our preclinical investigations showed that miR-15b antagomirs enhance sunitinib cytotoxic effects against ccRCC cells by upregulating PDZK1 levels, suggesting their potential in overcoming sunitinib resistance. CONCLUSIONS Our findings establish the miR-15b/PDZK1/PDGFR-β axis as a promising therapeutic target and a novel predictor for ccRCC patients' response to sunitinib treatment.
Collapse
MESH Headings
- Sunitinib/pharmacology
- Sunitinib/therapeutic use
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/metabolism
- Humans
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/pathology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Animals
- Drug Resistance, Neoplasm/genetics
- Mice
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Xenograft Model Antitumor Assays
- MicroRNAs/genetics
- Signal Transduction/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Male
- Mice, Nude
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
Collapse
Affiliation(s)
- Haibo Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Lijie Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hua Liu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Yumeng Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Wenxiu Lu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Xuedi Cao
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Qiong Qin
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Ran Song
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Duiping Feng
- Department of Interventional Radiology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Songlin Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
- Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy and Tooth Regeneration, School of Stomatology, Capital Medical University, Beijing, People's Republic of China
| | - Tao Bai
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Junqi He
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
2
|
Wang Y, Li J, Hao P, Li J, Han R, Lin J, Li X. Integrated Whole-Exome and Transcriptome Sequencing Indicated Dysregulation of Cholesterol Metabolism in Eyelid Sebaceous Gland Carcinoma. Transl Vis Sci Technol 2023; 12:4. [PMID: 36735267 PMCID: PMC9907373 DOI: 10.1167/tvst.12.2.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Purpose To identify the molecular background of eyelid sebaceous gland carcinomas (SCs), we conducted the integrated whole-exome sequencing and transcriptome sequencing for eyelid SCs in this study. Methods The genetic alterations were studied by whole-exome sequencing, and the messenger RNA expression was studied using Oxford Nanopore Technologies (ONT) in five paired fresh eyelid SC tissues and adjacent normal tissues. Integrated analysis of exome and transcriptomic information was conducted for filtering candidate driver genes. Protein-protein interaction (PPI) network of filtered candidate genes was analyzed by STRING. The protein expression was verified by immunohistochemistry in 29 eyelid SCs and 17 compared normal sebaceous gland tissues. Results The average numbers of pathogenic somatic single-nucleotide variants (SNVs) and indels in eyelid SCs were 75 and 28, respectively. Tumor protein p53 (TP53), zinc finger protein 750 (ZNF750), filaggrin 2 (FLG2), valosin-containing protein (VCP), and zinc finger protein 717 (ZNF717) were recurrent mutated genes. A mean of 844 differentially expressed genes (DEGs) were upregulated, and 1401 DEGs were downregulated in SC samples. The intersection of DEG-based pathways and mutation-based pathways was mainly involved in microbial infection and inflammation, immunodeficiency, cancer, lipid metabolism, and the other pathways. The intersection of DEGs and mutated genes consisted of 55 genes, of which 15 genes formed a PPI network with 4 clusters. The PPI cluster composed of scavenger receptor class B member 1 (SCARB1), peroxisome proliferator-activated receptor γ (PPARG), peroxisome proliferator-activated receptor γ coactivator 1α (PPARGC1A) was involved in cholesterol metabolism. The expression of SCARB1 protein was found to be increased, whereas that of PPARG protein was decreased in eyelid SCs compared to that in the normal sebaceous glands. Conclusions Increased SCARB1 and decreased PPARG indicated that dysregulation of cholesterol metabolism might be involved in carcinogenesis of eyelid SCs. Translational Relevance The malfunction in cholesterol metabolism might advance our knowledge of the carcinogenesis of eyelid SCs.
Collapse
Affiliation(s)
- Yuchuan Wang
- Tianjin Eye Hospital, Tianjin Key lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China,Nankai University Affiliated Eye Hospital, Tianjin, China,Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jun Li
- Tianjin Eye Hospital, Tianjin Key lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China,Nankai University Affiliated Eye Hospital, Tianjin, China,Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Peng Hao
- Tianjin Eye Hospital, Tianjin Key lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China,Nankai University Affiliated Eye Hospital, Tianjin, China,Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jing Li
- Tianjin Eye Hospital, Tianjin Key lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China,Nankai University Affiliated Eye Hospital, Tianjin, China,Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Ruifang Han
- Tianjin Eye Hospital, Tianjin Key lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China,Nankai University Affiliated Eye Hospital, Tianjin, China,Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jinyong Lin
- Tianjin Eye Hospital, Tianjin Key lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China,Nankai University Affiliated Eye Hospital, Tianjin, China,Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Xuan Li
- Tianjin Eye Hospital, Tianjin Key lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China,Nankai University Affiliated Eye Hospital, Tianjin, China,Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
3
|
Li J, Hui D, Yang L, Hou J, Xie H. Effect of high density lipoprotein cholesterol (HDL-C) on renal outcome in patients with nephrotic syndrome complicated with steroid-induced diabetes mellitus(SIDM). BMC Nephrol 2023; 24:2. [PMID: 36597028 PMCID: PMC9809113 DOI: 10.1186/s12882-022-03042-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE We aimed to investigate the renal prognosis of patients with idiopathic nephrotic syndrome (INS) complicated with steroid-induced diabetes mellitus (SIDM), the association of high-density lipoprotein cholesterol (HDL-C) before glucocorticoid treatment with renal prognosis, and the risk for persistent diabetes among patients with INS who had withdrawn from steroid therapy. MATERIALS AND METHODS We retrospectively analyzed 239 patients with INS complicated with SIDM at the National Clinical Research Center of Kidney Diseases, Jinling Hospital, from January 2008 to December 2019. The primary endpoint was the composite renal outcome defined as the development of end-stage renal disease (ESRD) or a 50% decrease in estimated glomerular filtration rate (eGFR) for more than 24 months after glucocorticoid withdrawal. The secondary endpoint was persistent diabetes, defined as fulfilling the criteria for diagnosing diabetes or using antidiabetic medications for at least 24 months after glucocorticoid withdrawal. RESULTS After glucocorticoid withdrawal for over 24 months, 35 (14.6%) patients reached the composite renal endpoint: end-stage renal disease (n = 14) or a 50% decrease in eGFR (n = 21). Before glucocorticoid therapy, a level of HDL-C greater than 1.45 mmol/L worsened renal survival in patients with INS complicated with SIDM. The log10 the level of HDL-C before glucocorticoid treatment was an independent risk factor for the renal outcome. A prediction model was generated: Hazard ratio (renal outcome) = 0.94 * hypertension before glucocorticoid therapy + 2.29 * log10 level of HDL-C before glucocorticoid treatment + 0.90 * the grade of interstitial tubule injury (AUROC, 0.75; 95% CI, 0.63 to 0.87; P < 0.01). Meanwhile, a level of fasting plasma glucose (FPG) before glucocorticoid treatment greater than 5.2 mmol/L enhanced the likelihood of persistent diabetes for at least 24 months after glucocorticoid withdrawal. CONCLUSIONS Increased level of HDL-C before glucocorticoid therapy was independently associated with a higher risk for renal outcome and thus may be useful in the renal prognosis of patients with INS complicated with SIDM.
Collapse
Affiliation(s)
- Jiarong Li
- grid.41156.370000 0001 2314 964XNational Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing Univerisity School of Medicine, Nanjing, 210016 China
| | - Di Hui
- grid.41156.370000 0001 2314 964XNational Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing Univerisity School of Medicine, Nanjing, 210016 China
| | - Liu Yang
- grid.41156.370000 0001 2314 964XNational Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing Univerisity School of Medicine, Nanjing, 210016 China
| | - Jinhua Hou
- grid.41156.370000 0001 2314 964XNational Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing Univerisity School of Medicine, Nanjing, 210016 China
| | - Honglang Xie
- grid.41156.370000 0001 2314 964XNational Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing Univerisity School of Medicine, Nanjing, 210016 China
| |
Collapse
|
4
|
Kluck GE, Qian AS, Sakarya EH, Quach H, Deng YD, Trigatti BL. Apolipoprotein A1 Protects Against Necrotic Core Development in Atherosclerotic Plaques: PDZK1-Dependent High-Density Lipoprotein Suppression of Necroptosis in Macrophages. Arterioscler Thromb Vasc Biol 2023; 43:45-63. [PMID: 36353992 PMCID: PMC9762725 DOI: 10.1161/atvbaha.122.318062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Atherosclerosis is a chronic disease affecting artery wall and a major contributor to cardiovascular diseases. Large necrotic cores increase risk of plaque rupture leading to thrombus formation. Necrotic cores are rich in debris from dead macrophages. Programmed necrosis (necroptosis) contributes to necrotic core formation. HDL (high-density lipoprotein) exerts direct atheroprotective effects on different cells within atherosclerotic plaques. Some of these depend on the SR-B1 (scavenger receptor class B type I) and the adapter protein PDZK1 (postsynaptic density protein/Drosophila disc-large protein/Zonula occludens protein containing 1). However, a role for HDL in protecting against necroptosis and necrotic core formation in atherosclerosis is not completely understood. METHODS Low-density lipoprotein receptor-deficient mice engineered to express different amounts of ApoA1 (apolipoprotein A1), or to lack PDZK1 were fed a high fat diet for 10 weeks. Atherosclerotic plaque areas, necrotic cores, and key necroptosis mediators, RIPK3 (receptor interacting protein kinase 3), and MLKL (mixed lineage kinase domain-like protein) were characterized. Cultured macrophages were treated with HDL to determine its effects, as well as the roles of SR-B1, PDZK1, and the PI3K (phosphoinositide 3-kinase) signaling pathway on necroptotic cell death. RESULTS Genetic overexpression reduced, and ApoA1 knockout increased necrotic core formation and RIPK3 and MLKL within atherosclerotic plaques. Macrophages were protected against necroptosis by HDL and this protection required SR-B1, PDZK1, and PI3K/Akt pathway. PDZK1 knockout increased atherosclerosis in LDLRKO mice, increasing necrotic cores and phospho-MLKL; both of which were reversed by restoring PDZK1 in BM-derived cells. CONCLUSIONS Our findings demonstrate that HDL in vitro and ApoA1, in vivo, protect against necroptosis in macrophages and necrotic core formation in atherosclerosis, suggesting a pathway that could be a target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- George E.G. Kluck
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Alexander S. Qian
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Emmanuel H. Sakarya
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Henry Quach
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Yak D. Deng
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| |
Collapse
|
5
|
Pan H, Huan C, Zhang W, Hou Y, Zhou Z, Yao J, Gao S. PDZK1 upregulates nitric oxide production through the PI3K/ERK2 pathway to inhibit porcine circovirus type 2 replication. Vet Microbiol 2022; 272:109514. [PMID: 35917623 DOI: 10.1016/j.vetmic.2022.109514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/01/2022] [Accepted: 07/14/2022] [Indexed: 10/17/2022]
Abstract
Porcine circovirus type 2 (PCV2) is the causative agent of porcine circovirus-associated disease. Changes in host cell gene expression are induced by PCV2 infection. Here, we showed that porcine PDZ Domain-Containing 1 (PDZK1) expression was enhanced during PCV2 infection and that overexpression of PDZK1 inhibited the expression of PCV2 Cap protein. PCV2 genomic DNA copy number and viral titers were decreased in PDZK1-overexpressing PK-15B6 cells. PDZK1 knockdown enhanced the replication of PCV2. Overexpression of PDZK1 activated the phosphoinositide 3-kinase (PI3K)/ERK2 signaling pathway to enhance nitric oxide (NO) levels, while PDZK1 knockdown had the opposite effects. A PI3K inhibitor (LY294002) and a NO synthase inhibitor (L-NAME hydrochloride) decreased the activity of PDZK1 in restricting PCV2 replication. ERK2 knockdown enhanced the proliferation of PCV2 by decreasing levels of NO. Levels of interleukin (IL)- 4 mRNA were reduced in PDZK1 knockdown and ERK2 knockdown PK-15B6 cells. Increased IL-4 mRNA levels were unable to decrease NO production in PDZK1-overexpressing cells. Thus, we conclude that PDZK1 affected PCV2 replication by regulating NO production via PI3K/ERK2 signaling. PDZK1 affected IL-4 expression through the PI3K/ERK2 pathway, but PDZK1 modulation of PCV2 replication occurred independently of IL-4. Our results contribute to understanding the biological functions of PDZK1 and provide a theoretical basis for the pathogenic mechanisms of PCV2.
Collapse
Affiliation(s)
- Haochun Pan
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Changchao Huan
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Wei Zhang
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Yutong Hou
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Ziyan Zhou
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Jingting Yao
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Song Gao
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, China.
| |
Collapse
|
6
|
Diab A, Valenzuela Ripoll C, Guo Z, Javaheri A. HDL Composition, Heart Failure, and Its Comorbidities. Front Cardiovasc Med 2022; 9:846990. [PMID: 35350538 PMCID: PMC8958020 DOI: 10.3389/fcvm.2022.846990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Although research on high-density lipoprotein (HDL) has historically focused on atherosclerotic coronary disease, there exists untapped potential of HDL biology for the treatment of heart failure. Anti-oxidant, anti-inflammatory, and endothelial protective properties of HDL could impact heart failure pathogenesis. HDL-associated proteins such as apolipoprotein A-I and M may have significant therapeutic effects on the myocardium, in part by modulating signal transduction pathways and sphingosine-1-phosphate biology. Furthermore, because heart failure is a complex syndrome characterized by multiple comorbidities, there are complex interactions between heart failure, its comorbidities, and lipoprotein homeostatic mechanisms. In this review, we will discuss the effects of heart failure and associated comorbidities on HDL, explore potential cardioprotective properties of HDL, and review novel HDL therapeutic targets in heart failure.
Collapse
|
7
|
HDL and Endothelial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:27-47. [DOI: 10.1007/978-981-19-1592-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
8
|
Powers HR, Sahoo D. SR-B1's Next Top Model: Structural Perspectives on the Functions of the HDL Receptor. Curr Atheroscler Rep 2022; 24:277-288. [PMID: 35107765 PMCID: PMC8809234 DOI: 10.1007/s11883-022-01001-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 02/04/2023]
Abstract
PURPOSE OF REVIEW The binding of high-density lipoprotein (HDL) to its primary receptor, scavenger receptor class B type 1 (SR-B1), is critical for lowering plasma cholesterol levels and reducing cardiovascular disease risk. This review provides novel insights into how the structural elements of SR-B1 drive efficient function with an emphasis on bidirectional cholesterol transport. RECENT FINDINGS We have generated a new homology model of full-length human SR-B1 based on the recent resolution of the partial structures of other class B scavenger receptors. Interrogating this model against previously published observations allows us to generate structurally informed hypotheses about SR-B1's ability to mediate HDL-cholesterol (HDL-C) transport. Furthermore, we provide a structural perspective as to why human variants of SR-B1 may result in impaired HDL-C clearance. A comprehensive understanding of SR-B1's structure-function relationships is critical to the development of therapeutic agents targeting SR-B1 and modulating cardiovascular disease risk.
Collapse
Affiliation(s)
- Hayley R. Powers
- grid.30760.320000 0001 2111 8460Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI USA
| | - Daisy Sahoo
- grid.30760.320000 0001 2111 8460Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI USA ,grid.30760.320000 0001 2111 8460Department of Medicine, Division of Endocrinology & Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI USA ,grid.30760.320000 0001 2111 8460Cardiovascular Center, H4930 Health Research Center, Medical College of Wisconsin, 8701 W. Watertown Plank Road, Milwaukee, WI 53226 USA
| |
Collapse
|
9
|
Tao J, Li C, Zheng Y, Wang F, Zhang M, Wu X, Chen Y, Zeng Q, Chen F, Fei W. Biological protein mediated ferroptotic tumor nanotherapeutics. J Mater Chem B 2021; 9:9262-9284. [PMID: 34730601 DOI: 10.1039/d1tb01289d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ferroptosis, a cell death pathway involving iron-related generation of lipid hydroperoxides for achieving incredible tumor suppression, has reignited the hope of chemotherapy in tumor treatment in the past decade. With extensive research studies, various bioactive proteins and cellular pathways have been demonstrated to regulate the occurrence and development of ferroptosis. The gradually established ferroptotic regulatory network is conducive to find effective proteins from a holistic perspective and guides better designs for future ferroptotic tumor therapies. The first section of this review summarizes the recent advances in ferroptotic regulatory mechanisms of proteins and attempts to clarify their latent function in the ferroptotic regulatory network. Second, the existing protein-mediated ferroptotic tumor nanotherapeutic strategies were reviewed, including the protein-mediated iron supplement, cell membrane transporter inhibition, glutathione peroxidase 4 interference, glutathione depletion, bioenzyme-mediated reactive oxygen species generation, heat shock protein inhibition, and tumor-overexpressed protein-triggered drug release for ferroptotic therapy. Finally, the future expectations and challenges of ferroptotic tumor nanotherapeutics for clinical cancer therapy are highlighted.
Collapse
Affiliation(s)
- Jiaoyang Tao
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Chaoqun Li
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Yongquan Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Fengmei Wang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Xiaodong Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Qingquan Zeng
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Fengying Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
10
|
Mineo C. Lipoprotein receptor signalling in atherosclerosis. Cardiovasc Res 2021; 116:1254-1274. [PMID: 31834409 DOI: 10.1093/cvr/cvz338] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/01/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The founding member of the lipoprotein receptor family, low-density lipoprotein receptor (LDLR) plays a major role in the atherogenesis through the receptor-mediated endocytosis of LDL particles and regulation of cholesterol homeostasis. Since the discovery of the LDLR, many other structurally and functionally related receptors have been identified, which include low-density lipoprotein receptor-related protein (LRP)1, LRP5, LRP6, very low-density lipoprotein receptor, and apolipoprotein E receptor 2. The scavenger receptor family members, on the other hand, constitute a family of pattern recognition proteins that are structurally diverse and recognize a wide array of ligands, including oxidized LDL. Among these are cluster of differentiation 36, scavenger receptor class B type I and lectin-like oxidized low-density lipoprotein receptor-1. In addition to the initially assigned role as a mediator of the uptake of macromolecules into the cell, a large number of studies in cultured cells and in in vivo animal models have revealed that these lipoprotein receptors participate in signal transduction to modulate cellular functions. This review highlights the signalling pathways by which these receptors influence the process of atherosclerosis development, focusing on their roles in the vascular cells, such as macrophages, endothelial cells, smooth muscle cells, and platelets. Human genetics of the receptors is also discussed to further provide the relevance to cardiovascular disease risks in humans. Further knowledge of the vascular biology of the lipoprotein receptors and their ligands will potentially enhance our ability to harness the mechanism to develop novel prophylactic and therapeutic strategies against cardiovascular diseases.
Collapse
Affiliation(s)
- Chieko Mineo
- Department of Pediatrics and Cell Biology, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| |
Collapse
|
11
|
Abumrad NA, Cabodevilla AG, Samovski D, Pietka T, Basu D, Goldberg IJ. Endothelial Cell Receptors in Tissue Lipid Uptake and Metabolism. Circ Res 2021; 128:433-450. [PMID: 33539224 DOI: 10.1161/circresaha.120.318003] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipid uptake and metabolism are central to the function of organs such as heart, skeletal muscle, and adipose tissue. Although most heart energy derives from fatty acids (FAs), excess lipid accumulation can cause cardiomyopathy. Similarly, high delivery of cholesterol can initiate coronary artery atherosclerosis. Hearts and arteries-unlike liver and adrenals-have nonfenestrated capillaries and lipid accumulation in both health and disease requires lipid movement from the circulation across the endothelial barrier. This review summarizes recent in vitro and in vivo findings on the importance of endothelial cell receptors and uptake pathways in regulating FAs and cholesterol uptake in normal physiology and cardiovascular disease. We highlight clinical and experimental data on the roles of ECs in lipid supply to tissues, heart, and arterial wall in particular, and how this affects organ metabolism and function. Models of FA uptake into ECs suggest that receptor-mediated uptake predominates at low FA concentrations, such as during fasting, whereas FA uptake during lipolysis of chylomicrons may involve paracellular movement. Similarly, in the setting of an intact arterial endothelial layer, recent and historic data support a role for receptor-mediated processes in the movement of lipoproteins into the subarterial space. We conclude with thoughts on the need to better understand endothelial lipid transfer for fuller comprehension of the pathophysiology of hyperlipidemia, and lipotoxic diseases such as some forms of cardiomyopathy and atherosclerosis.
Collapse
Affiliation(s)
- Nada A Abumrad
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Dmitri Samovski
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Terri Pietka
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| |
Collapse
|
12
|
Rink JS, Lin AY, McMahon KM, Calvert AE, Yang S, Taxter T, Moreira J, Chadburn A, Behdad A, Karmali R, Thaxton CS, Gordon LI. Targeted reduction of cholesterol uptake in cholesterol-addicted lymphoma cells blocks turnover of oxidized lipids to cause ferroptosis. J Biol Chem 2021; 296:100100. [PMID: 33208460 PMCID: PMC7949030 DOI: 10.1074/jbc.ra120.014888] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
Normal human cells can either synthesize cholesterol or take it up from lipoproteins to meet their metabolic requirements. In some malignant cells, de novo cholesterol synthesis genes are transcriptionally silent or mutated, meaning that cholesterol uptake from lipoproteins is required for survival. Recent data suggest that lymphoma cells dependent upon lipoprotein-mediated cholesterol uptake are also subject to ferroptosis, an oxygen- and iron-dependent cell death mechanism triggered by accumulation of oxidized lipids in cell membranes unless the lipid hydroperoxidase, glutathione peroxidase 4 (GPX4), reduces these toxic lipid species. To study mechanisms linking cholesterol uptake with ferroptosis and determine the potential role of the high-density lipoprotein (HDL) receptor as a target for cholesterol depleting therapy, we treated lymphoma cell lines known to be sensitive to the reduction of cholesterol uptake with HDL-like nanoparticles (HDL NPs). HDL NPs are a cholesterol-poor ligand that binds to the receptor for cholesterol-rich HDLs, scavenger receptor type B1 (SCARB1). Our data reveal that HDL NP treatment activates a compensatory metabolic response in treated cells toward increased de novo cholesterol synthesis, which is accompanied by nearly complete reduction in expression of GPX4. As a result, oxidized membrane lipids accumulate, leading to cell death through a mechanism consistent with ferroptosis. We obtained similar results in vivo after systemic administration of HDL NPs in mouse lymphoma xenografts and in primary samples obtained from patients with lymphoma. In summary, targeting SCARB1 with HDL NPs in cholesterol uptake-addicted lymphoma cells abolishes GPX4, resulting in cancer cell death by a mechanism consistent with ferroptosis.
Collapse
Affiliation(s)
- Jonathan S Rink
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Adam Yuh Lin
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Kaylin M McMahon
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA; Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | - Andrea E Calvert
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA; Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | - Shuo Yang
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Tim Taxter
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jonathan Moreira
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Amy Chadburn
- Department of Pathology, Weill Cornell Medical Center, New York, New York, USA
| | - Amir Behdad
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Reem Karmali
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - C Shad Thaxton
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA; Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA; International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, USA.
| | - Leo I Gordon
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
13
|
High-Density Lipoprotein-Targeted Therapies for Heart Failure. Biomedicines 2020; 8:biomedicines8120620. [PMID: 33339429 PMCID: PMC7767106 DOI: 10.3390/biomedicines8120620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 02/08/2023] Open
Abstract
The main and common constituents of high-density lipoproteins (HDLs) are apolipoprotein A-I, cholesterol, and phospholipids. Biochemical heterogeneity of HDL particles is based on the variable presence of one or more representatives of at least 180 proteins, 200 lipid species, and 20 micro RNAs. HDLs are circulating multimolecular platforms that perform divergent functions whereby the potential of HDL-targeted interventions for treatment of heart failure can be postulated based on its pleiotropic effects. Several murine studies have shown that HDLs exert effects on the myocardium, which are completely independent of any impact on coronary arteries. Overall, HDL-targeted therapies exert a direct positive lusitropic effect on the myocardium, inhibit the development of cardiac hypertrophy, suppress interstitial and perivascular myocardial fibrosis, increase capillary density in the myocardium, and prevent the occurrence of heart failure. In four distinct murine models, HDL-targeted interventions were shown to be a successful treatment for both pre-existing heart failure with reduced ejection fraction (HFrEF) and pre-existing heart failure with preserved ejection fraction (HFrEF). Until now, the effect of HDL-targeted interventions has not been evaluated in randomized clinical trials in heart failure patients. As HFpEF represents an important unmet therapeutic need, this is likely the preferred therapeutic domain for clinical translation.
Collapse
|
14
|
Ma B, Jia J, Wang X, Zhang R, Niu S, Ni L, Di X, Liu C. Differential roles of Scavenger receptor class B type I: A protective molecule and a facilitator of atherosclerosis (Review). Mol Med Rep 2020; 22:2599-2604. [PMID: 32945418 PMCID: PMC7453654 DOI: 10.3892/mmr.2020.11383] [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: 04/15/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
The scavenger receptor class B type I (SR-BI) is a multi-ligand membrane protein receptor that binds to high-density lipoprotein (HDL) under physiological conditions, promoting the selective uptake of cholesterol esters from HDL into cells. SR-BI also promotes the reverse transport of excess cholesterol from peripheral tissues to the liver, contributing to the synthesis of bile acids for excretion and the removal of excess cholesterol from the body, thereby lowering the cholesterol load and exerting anti-atherosclerotic effects. Studies in mice and humans have demonstrated that a functional defect of SR-BI can cause atherosclerotic lesions and cardiovascular diseases, such as myocardial infarction and stroke. Additionally, SR-BI in vascular endothelial cells promoted the deposition of low-density lipoprotein under the endothelium. Although SR-BI is widely expressed in various tissues and cell types throughout the body, its expression level and function vary accordingly. The present review focuses on the biological functions and mechanisms of SR-BI in regulating atherosclerosis.
Collapse
Affiliation(s)
- Baitao Ma
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Jing Jia
- Department of Obstetrics and Gynaecology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xuebin Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Rui Zhang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Shuai Niu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Leng Ni
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xiao Di
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Changwei Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
15
|
Khadge S, Sharp JG, Thiele GM, McGuire TR, Talmadge JE. Fatty Acid Mediators in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1259:125-153. [PMID: 32578175 DOI: 10.1007/978-3-030-43093-1_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patients with cancer frequently overexpress inflammatory cytokines with an associated neutrophilia both of which may be downregulated by diets with high omega-3 polyunsaturated fatty acids (ω-3 PUFA). The anti-inflammatory activity of dietary ω-3 PUFA has been suggested to have anticancer properties and to improve survival of cancer patients. Currently, the majority of dietary research efforts do not differentiate between obesity and dietary fatty acid consumption as mediators of inflammatory cell expansion and tumor microenvironmental infiltration, initiation, and progression. In this chapter, we discuss the relationships between dietary lipids, inflammation, neoplasia and strategies to regulate these relationships. We posit that dietary composition, notably the ratio of ω-3 vs. ω-6 PUFA, regulates tumor initiation and progression and the frequency and sites of metastasis that, together, impact overall survival (OS). We focus on three broad topics: first, the role of dietary lipids in chronic inflammation and tumor initiation, progression, and regression; second, lipid mediators linking inflammation and cancer; and third, dietary lipid regulation of murine and human tumor initiation, progression, and metastasis.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Vanderbilt University, Nashville, TN, USA
| | - John Graham Sharp
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - James E Talmadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
16
|
Takaeko Y, Matsui S, Kajikawa M, Maruhashi T, Yamaji T, Harada T, Han Y, Hashimoto H, Kihara Y, Hida E, Chayama K, Goto C, Aibara Y, Yusoff FM, Kishimoto S, Nakashima A, Higashi Y. Relationship between high-density lipoprotein cholesterol levels and endothelial function in women: a cross-sectional study. BMJ Open 2020; 10:e038121. [PMID: 32641366 PMCID: PMC7342861 DOI: 10.1136/bmjopen-2020-038121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES The purpose of this study was to evaluate the relationship between high-density lipoprotein cholesterol (HDL-C) levels and endothelial function in women. DESIGN Cross-sectional study. SETTING 22 university hospitals and affiliated clinics in Japan. PARTICIPANTS 1719 Japanese women aged 17-90 years who were not receiving lipid-lowering therapy. MEASURES We evaluated flow-mediated vasodilation (FMD) and serum levels of HDL-C. All participants were divided into four groups by HDL-C level: low HDL-C (<40 mg/dL), moderate HDL-C (40-59 mg/dL), high HDL-C (60-79 md/dL) and extremely high HDL-C (≥80 mg/dL). RESULTS Univariate regression analysis revealed a significant relationship between FMD and HDL-C (r=0.12, p<0.001). FMD values were significantly smaller in the low HDL-C group (5.2%±3.8%) and moderate HDL-C group (5.2%±3.8%) than in the extremely high HDL-C group (6.7%±3.4%) (p=0.024 and p=0.003, respectively), while there was no significant difference in FMD between the high HDL-C group and the extremely high HDL-C group. Multiple logistic regression analysis did not show a significant association between HDL-C levels and FMD. CONCLUSIONS Endothelial function increased in relation to HDL-C levels. However, there was no association of HDL-C levels with endothelial function after adjustment of traditional cardiovascular risk factors in women. TRIAL REGISTRATION NUMBER UMIN000012950; Results.
Collapse
Affiliation(s)
- Yuji Takaeko
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shogo Matsui
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takayuki Yamaji
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yiming Han
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Haruki Hashimoto
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Eisuke Hida
- Department of Biostatistics and Data Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Chikara Goto
- Department of Physical Therapy, Hiroshima International University, HigashiHiroshima, Hiroshima, Japan
| | - Yoshiki Aibara
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| |
Collapse
|
17
|
Biological convergence of three human and animal model quantitative trait loci for blood pressure. J Hypertens 2020; 38:322-331. [DOI: 10.1097/hjh.0000000000002267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
18
|
Sharma B, Agnihotri N. Role of cholesterol homeostasis and its efflux pathways in cancer progression. J Steroid Biochem Mol Biol 2019; 191:105377. [PMID: 31063804 DOI: 10.1016/j.jsbmb.2019.105377] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/09/2019] [Accepted: 05/04/2019] [Indexed: 12/27/2022]
Abstract
Tumor cells show high avidity for cholesterol in order to support their inherent nature to divide and proliferate. This results in the rewiring of cholesterol homeostatic pathways by influencing not only de novo synthesis but also uptake or efflux pathways of cholesterol. Recent findings have pointed towards the importance of cholesterol efflux in tumor pathogenesis. Cholesterol efflux is the first and foremost step in reverse cholesterol transport and any perturbation in this pathway may lead to the accumulation of intracellular cholesterol, thereby altering the cellular equilibrium. This review addresses the different mechanisms of cholesterol efflux from the cell and highlights their role and regulation in context to tumor development. There are four different routes by which cholesterol can be effluxed from the cell namely, 1) passive diffusion of cholesterol to mature HDL particles, 2) SR-B1 mediated facilitated diffusion, 3) Active efflux to apo A1 via ABCA1 and 4) ABCG1 mediated efflux to mature HDL. These molecular players facilitating cholesterol efflux are engaged in a complex interplay with different signaling pathways. Thus, an understanding of the efflux pathways, their regulation and cross-talk with signaling molecules may provide novel prognostic markers and therapeutic targets to combat the onset of carcinogenesis.
Collapse
Affiliation(s)
- Bhoomika Sharma
- Department of Biochemistry, BMS-Block II, Panjab University, Sector-25, Chandigarh, 160014, India.
| | - Navneet Agnihotri
- Department of Biochemistry, BMS-Block II, Panjab University, Sector-25, Chandigarh, 160014, India.
| |
Collapse
|
19
|
Muresan XM, Sticozzi C, Belmonte G, Cervellati F, Ferrara F, Lila MA, Valacchi G. SR-B1 involvement in keratinocytes in vitro wound closure. Arch Biochem Biophys 2018; 658:1-6. [PMID: 30240595 DOI: 10.1016/j.abb.2018.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 12/17/2022]
Abstract
Skin represents the most extended organ of human body, having as main function the protection of our body from outdoor stressors. Its protective ability is compromised when the skin is disrupted as a consequence of mechanical insults. For this purpose, cutaneous tissue is equipped with an efficient and fine mechanism involved in repairing the wounded area. Among the numerous players that take part in the wound healing process, SR-B1 has been recently shown to have a role in keratinocyte re-epithelialization. SR-B1 is a mediator of cholesterol uptake from HDLs, whereas it is implicated in other cellular processes such as vitamins absorption, vesicle trafficking or pathogen identification. The aim of this study was to investigate the mechanisms involved in SR-B1 role in skin wound closure. Our in vitro data demonstrated that SR-B1 influenced keratinocyte proliferation and migration through a downregulation of nuclear cyclin D1 levels and active MMP9 expression respectively possibly in an NF-kB-dependent mechanism. In addition, SR-B1 was also able to modulate keratinocyte morphology into a pro-migratory cytoskeleton rearrangement. The present in vitro study suggests a new role of SRB1 as a possible new key player in cutaneous wound healing mechanism.
Collapse
Affiliation(s)
- Ximena M Muresan
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Claudia Sticozzi
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Belmonte
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesca Ferrara
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mary Ann Lila
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA
| | - Giuseppe Valacchi
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA.
| |
Collapse
|
20
|
Yu P, Qian AS, Chathely KM, Trigatti BL. PDZK1 in leukocytes protects against cellular apoptosis and necrotic core development in atherosclerotic plaques in high fat diet fed ldl receptor deficient mice. Atherosclerosis 2018; 276:171-181. [DOI: 10.1016/j.atherosclerosis.2018.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 04/20/2018] [Accepted: 05/02/2018] [Indexed: 02/09/2023]
|
21
|
Shen WJ, Asthana S, Kraemer FB, Azhar S. Scavenger receptor B type 1: expression, molecular regulation, and cholesterol transport function. J Lipid Res 2018; 59:1114-1131. [PMID: 29720388 DOI: 10.1194/jlr.r083121] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/26/2018] [Indexed: 12/16/2022] Open
Abstract
Cholesterol is required for maintenance of plasma membrane fluidity and integrity and for many cellular functions. Cellular cholesterol can be obtained from lipoproteins in a selective pathway of HDL-cholesteryl ester (CE) uptake without parallel apolipoprotein uptake. Scavenger receptor B type 1 (SR-B1) is a cell surface HDL receptor that mediates HDL-CE uptake. It is most abundantly expressed in liver, where it provides cholesterol for bile acid synthesis, and in steroidogenic tissues, where it delivers cholesterol needed for storage or steroidogenesis in rodents. SR-B1 transcription is regulated by trophic hormones in the adrenal gland, ovary, and testis; in the liver and elsewhere, SR-B1 is subject to posttranscriptional and posttranslational regulation. SR-B1 operates in several metabolic processes and contributes to pathogenesis of atherosclerosis, inflammation, hepatitis C virus infection, and other conditions. Here, we summarize characteristics of the selective uptake pathway and involvement of microvillar channels as facilitators of selective HDL-CE uptake. We also present the potential mechanisms of SR-B1-mediated selective cholesterol transport; the transcriptional, posttranscriptional, and posttranslational regulation of SR-B1; and the impact of gene variants on expression and function of human SR-B1. A better understanding of this unique pathway and SR-B1's role may yield improved therapies for a wide variety of conditions.
Collapse
Affiliation(s)
- Wen-Jun Shen
- Geriatric Research, Education, and Clinical Research Center (GRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 and Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305
| | - Shailendra Asthana
- Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad 121001, Haryana, India
| | - Fredric B Kraemer
- Geriatric Research, Education, and Clinical Research Center (GRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 and Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305
| | - Salman Azhar
- Geriatric Research, Education, and Clinical Research Center (GRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 and Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
22
|
He D, Zhao M, Wu C, Zhang W, Niu C, Yu B, Jin J, Ji L, Willard B, Mathew AV, Chen YE, Pennathur S, Yin H, He Y, Pan B, Zheng L. Apolipoprotein A-1 mimetic peptide 4F promotes endothelial repairing and compromises reendothelialization impaired by oxidized HDL through SR-B1. Redox Biol 2017; 15:228-242. [PMID: 29277016 PMCID: PMC5975068 DOI: 10.1016/j.redox.2017.11.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/24/2017] [Accepted: 11/29/2017] [Indexed: 01/24/2023] Open
Abstract
Disruption of endothelial monolayer integrity is the primary instigating factor for many cardiovascular diseases. High density lipoprotein (HDL) oxidized by heme enzyme myeloperoxidase (MPO) is dysfunctional in promoting endothelial repair. Apolipoprotein A-1 mimetic 4F with its pleiotropic benefits has been proven effective in many in vivo models. In this study we investigated whether 4F promotes endothelial repair and restores the impaired function of oxidized HDL (Cl/NO2-HDL) in promoting re-endothelialization. We demonstrate that 4F and Cl/NO2-HDL act on scavenger receptor type I (SR-B1) using human aorta endothelial cells (HAEC) and SR-B1 (-/-) mouse aortic endothelial cells. Wound healing, transwell migration, lamellipodia formation and single cell migration assay experiments show that 4F treatment is associated with a recovery of endothelial cell migration and associated with significantly increased endothelial nitric oxide synthase (eNOS) activity, Akt phosphorylation and SR-B1 expression. 4F increases NO generation and diminishes oxidative stress. In vivo, 4F can stimulate cell proliferation and re-endothelialization in the carotid artery after treatment with Cl/NO2-HDL in a carotid artery electric injury model but fails to do so in SR-B1(-/-) mice. These findings demonstrate that 4F promotes endothelial cell migration and has a potential therapeutic benefit against early endothelial injury in cardiovascular diseases. 4F restores the decreased ability of Cl/NO2-HDL in promoting endothelial repair. 4F increases NO generation and diminishes oxidative stress. 4F increases eNOS activity, Akt phosphorylation and SR-B1 expression. 4F can stimulate re-endothelialization in a carotid artery electric injury model.
Collapse
Affiliation(s)
- Dan He
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China
| | - Congying Wu
- The Institute of Systems Biomedicine, Department of Medical Genetics, Peking University Health Science Center, Beijing 100191, China
| | - Wenjing Zhang
- The Military General Hospital of Beijing, Beijing 100700, China
| | - Chenguang Niu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China
| | - Baoqi Yu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China
| | - Jingru Jin
- The Military General Hospital of Beijing, Beijing 100700, China
| | - Liang Ji
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China
| | - Belinda Willard
- Proteomics Laboratory, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Anna V Mathew
- Department of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Department of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS), Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Yuan He
- National Research Institute for Health and Family Planning, Beijing 100081, China
| | - Bing Pan
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China.
| |
Collapse
|
23
|
Fung KYY, Fairn GD, Lee WL. Transcellular vesicular transport in epithelial and endothelial cells: Challenges and opportunities. Traffic 2017; 19:5-18. [PMID: 28985008 DOI: 10.1111/tra.12533] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/29/2017] [Accepted: 09/29/2017] [Indexed: 12/31/2022]
Abstract
Vesicle-mediated transcellular transport or simply "transcytosis" is a cellular process used to shuttle macromolecules such as lipoproteins, antibodies, and albumin from one surface of a polarized cell to the other. This mechanism is in contrast to the transit of small molecules such as anions, cations and amino acids that occur via uptake, diffusion through the cytosol and release and is also distinct from paracellular leak between cells. Importantly, transcytosis has evolved as a process to selectively move macromolecules between 2 neighboring yet unique microenvironments within a multicellular organism. Examples include the movement of lipoproteins out of the circulatory system and into tissues and the delivery of immunoglobulins to mucosal surfaces. Regardless of whether the transport is conducted by endothelial or epithelial cells, the process often involves receptor-mediated uptake of a ligand into an endocytic vesicle, regulated transit of the carrier through the cytoplasm and release of the cargo via an exocytic event. While transcytosis has been examined in detail in epithelial cells, for both historical and technical reasons, the process is less understood in endothelial cells. Here, we spotlight aspects of epithelial transcytosis including recent findings and review the comparative dearth of knowledge regarding the process in endothelial cells highlighting the opportunity for further study.
Collapse
Affiliation(s)
- Karen Y Y Fung
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Gregory D Fairn
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery & Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Institute for Biomedical Engineering and Science Technology (iBEST), Ryerson University and St Michael's Hospital, Toronto, Ontario, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Institute for Biomedical Engineering and Science Technology (iBEST), Ryerson University and St Michael's Hospital, Toronto, Ontario, Canada.,Departments of Medicine, Laboratory Medicine and Pathobiology,& Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Shen WJ, Azhar S, Kraemer FB. SR-B1: A Unique Multifunctional Receptor for Cholesterol Influx and Efflux. Annu Rev Physiol 2017; 80:95-116. [PMID: 29125794 DOI: 10.1146/annurev-physiol-021317-121550] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The scavenger receptor, class B type 1 (SR-B1), is a multiligand membrane receptor protein that functions as a physiologically relevant high-density lipoprotein (HDL) receptor whose primary role is to mediate selective uptake or influx of HDL-derived cholesteryl esters into cells and tissues. SR-B1 also facilitates the efflux of cholesterol from peripheral tissues, including macrophages, back to liver. As a regulator of plasma membrane cholesterol content, SR-B1 promotes the uptake of lipid soluble vitamins as well as viral entry into host cells. These collective functions of SR-B1 ultimately affect programmed cell death, female fertility, platelet function, vasculature inflammation, and diet-induced atherosclerosis and myocardial infarction. SR-B1 has also been identified as a potential marker for cancer diagnosis and prognosis. Finally, the SR-B1-linked selective HDL-cholesteryl ester uptake pathway is now being evaluated as a gateway for the delivery of therapeutic and diagnostic agents. In this review, we focus on the regulation and functional significance of SR-B1 in mediating cholesterol movement into and out of cells.
Collapse
Affiliation(s)
- Wen-Jun Shen
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| | - Salman Azhar
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| | - Fredric B Kraemer
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| |
Collapse
|
25
|
Fung KY, Wang C, Nyegaard S, Heit B, Fairn GD, Lee WL. SR-BI Mediated Transcytosis of HDL in Brain Microvascular Endothelial Cells Is Independent of Caveolin, Clathrin, and PDZK1. Front Physiol 2017; 8:841. [PMID: 29163190 PMCID: PMC5670330 DOI: 10.3389/fphys.2017.00841] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/09/2017] [Indexed: 01/19/2023] Open
Abstract
The vascular endothelium supplying the brain exhibits very low paracellular and transcellular permeability and is a major constituent of the blood-brain barrier. High-density lipoprotein (HDL) crosses the blood-brain barrier by transcytosis, but technical limitations have made it difficult to elucidate its regulation. Using a combination of spinning-disc confocal and total internal reflection fluorescence microscopy, we examined the uptake and transcytosis of HDL by human primary brain microvascular endothelial cell monolayers. Using these approaches, we report that HDL internalization requires dynamin but not clathrin heavy chain and that its internalization and transcytosis are saturable. Internalized HDL partially co-localized with the scavenger receptor BI (SR-BI) and knockdown of SR-BI significantly attenuated HDL internalization. However, we observed that the adaptor protein PDZK1—which is critical to HDL-SR-BI signaling in other tissues—is not required for HDL uptake in these cells. Additionally, while these cells express caveolin, the abundance of caveolae in this tissue is negligible and we find that SR-BI and caveolin do not co-fractionate. Furthermore, direct silencing of caveolin-1 had no impact on the uptake of HDL. Finally, inhibition of endothelial nitric oxide synthase increased HDL internalization while increasing nitric oxide levels had no impact. Together, these data indicate that SR-BI-mediated transcytosis in brain microvascular endothelial cells is distinct from uptake and signaling pathways described for this receptor in other cell types.
Collapse
Affiliation(s)
- Karen Y Fung
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Changsen Wang
- Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Steffen Nyegaard
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Bryan Heit
- Department of Microbiology and Immunology, Centre for Human Immunology, University of Western Ontario, London, ON, Canada
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Surgery, University of Toronto, ON, Canada
| | - Warren L Lee
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Medicine and Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| |
Collapse
|
26
|
Wu BJ, Li Y, Ong KL, Sun Y, Shrestha S, Hou L, Johns D, Barter PJ, Rye KA. Reduction of In-Stent Restenosis by Cholesteryl Ester Transfer Protein Inhibition. Arterioscler Thromb Vasc Biol 2017; 37:2333-2341. [PMID: 29025709 DOI: 10.1161/atvbaha.117.310051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/02/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Angioplasty and stent implantation, the most common treatment for atherosclerotic lesions, have a significant failure rate because of restenosis. This study asks whether increasing plasma high-density lipoprotein (HDL) levels by inhibiting cholesteryl ester transfer protein activity with the anacetrapib analog, des-fluoro-anacetrapib, prevents stent-induced neointimal hyperplasia. APPROACH AND RESULTS New Zealand White rabbits received normal chow or chow supplemented with 0.14% (wt/wt) des-fluoro-anacetrapib for 6 weeks. Iliac artery endothelial denudation and bare metal steel stent deployment were performed after 2 weeks of des-fluoro-anacetrapib treatment. The animals were euthanized 4 weeks poststent deployment. Relative to control, dietary supplementation with des-fluoro-anacetrapib reduced plasma cholesteryl ester transfer protein activity and increased plasma apolipoprotein A-I and HDL cholesterol levels by 53±6.3% and 120±19%, respectively. Non-HDL cholesterol levels were unaffected. Des-fluoro-anacetrapib treatment reduced the intimal area of the stented arteries by 43±5.6% (P<0.001), the media area was unchanged, and the arterial lumen area increased by 12±2.4% (P<0.05). Des-fluoro-anacetrapib treatment inhibited vascular smooth muscle cell proliferation by 41±4.5% (P<0.001). Incubation of isolated HDLs from des-fluoro-anacetrapib-treated animals with human aortic smooth muscle cells at apolipoprotein A-I concentrations comparable to their plasma levels inhibited cell proliferation and migration. These effects were dependent on scavenger receptor-B1, the adaptor protein PDZ domain-containing protein 1, and phosphatidylinositol-3-kinase/Akt activation. HDLs from des-fluoro-anacetrapib-treated animals also inhibited proinflammatory cytokine-induced human aortic smooth muscle cell proliferation and stent-induced vascular inflammation. CONCLUSIONS Inhibiting cholesteryl ester transfer protein activity in New Zealand White rabbits with iliac artery balloon injury and stent deployment increases HDL levels, inhibits vascular smooth muscle cell proliferation, and reduces neointimal hyperplasia in an scavenger receptor-B1, PDZ domain-containing protein 1- and phosphatidylinositol-3-kinase/Akt-dependent manner.
Collapse
Affiliation(s)
- Ben J Wu
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.).
| | - Yue Li
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.)
| | - Kwok L Ong
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.)
| | - Yidan Sun
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.)
| | - Sudichhya Shrestha
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.)
| | - Liming Hou
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.)
| | - Douglas Johns
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.)
| | - Philip J Barter
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.)
| | - Kerry-Anne Rye
- From the School of Medical Sciences, The University of New South Wales Sydney, Australia (B.J.W., K.L.O., Y.S., S.S., L.H., P.J.B., K.-A.R.); Institute of Pathophysiology and Immunology, Medical University of Graz, Austria (Y.S.); and Merck & Co., Inc, Kenilworth, NJ (D.J.).
| |
Collapse
|
27
|
Linton MF, Tao H, Linton EF, Yancey PG. SR-BI: A Multifunctional Receptor in Cholesterol Homeostasis and Atherosclerosis. Trends Endocrinol Metab 2017; 28:461-472. [PMID: 28259375 PMCID: PMC5438771 DOI: 10.1016/j.tem.2017.02.001] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/31/2017] [Accepted: 02/01/2017] [Indexed: 02/07/2023]
Abstract
The HDL receptor scavenger receptor class B type I (SR-BI) plays crucial roles in cholesterol homeostasis, lipoprotein metabolism, and atherosclerosis. Hepatic SR-BI mediates reverse cholesterol transport (RCT) by the uptake of HDL cholesterol for routing to the bile. Through the selective uptake of HDL lipids, hepatic SR-BI modulates HDL composition and preserves HDL's atheroprotective functions of mediating cholesterol efflux and minimizing inflammation and oxidation. Macrophage and endothelial cell SR-BI inhibits the development of atherosclerosis by mediating cholesterol trafficking to minimize atherosclerotic lesion foam cell formation. SR-BI signaling also helps limit inflammation and cell death and mediates efferocytosis of apoptotic cells in atherosclerotic lesions thereby preventing vulnerable plaque formation. SR-BI is emerging as a multifunctional therapeutic target to reduce atherosclerosis development.
Collapse
Affiliation(s)
- MacRae F Linton
- Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6300, USA; Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6300, USA.
| | - Huan Tao
- Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6300, USA
| | - Edward F Linton
- Perelman School of Medicine, University of Pennsylvania, Jordan Medical Education Center, 6th Floor, 3400 Civic Center Blvd, Philadelphia, PA 19104-6055, USA
| | - Patricia G Yancey
- Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6300, USA.
| |
Collapse
|
28
|
Velagapudi S, Yalcinkaya M, Piemontese A, Meier R, Nørrelykke SF, Perisa D, Rzepiela A, Stebler M, Stoma S, Zanoni P, Rohrer L, von Eckardstein A. VEGF-A Regulates Cellular Localization of SR-BI as Well as Transendothelial Transport of HDL but Not LDL. Arterioscler Thromb Vasc Biol 2017; 37:794-803. [DOI: 10.1161/atvbaha.117.309284] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/20/2017] [Indexed: 11/16/2022]
Abstract
Objective—
Low- and high-density lipoproteins (LDL and HDL) must pass the endothelial layer to exert pro- and antiatherogenic activities, respectively, within the vascular wall. However, the rate-limiting factors that mediate transendothelial transport of lipoproteins are yet little known. Therefore, we performed a high-throughput screen with kinase drug inhibitors to identify modulators of transendothelial LDL and HDL transport.
Approach and Results—
Microscopy-based high-content screening was performed by incubating human aortic endothelial cells with 141 kinase-inhibiting drugs and fluorescent-labeled LDL or HDL. Inhibitors of vascular endothelial growth factor (VEGF) receptors (VEGFR) significantly decreased the uptake of HDL but not LDL. Silencing of VEGF receptor 2 significantly decreased cellular binding, association, and transendothelial transport of
125
I-HDL but not
125
I-LDL. RNA interference with VEGF receptor 1 or VEGF receptor 3 had no effect. Binding, uptake, and transport of HDL but not LDL were strongly reduced in the absence of VEGF-A from the cell culture medium and were restored by the addition of VEGF-A. The restoring effect of VEGF-A on endothelial binding, uptake, and transport of HDL was abrogated by pharmacological inhibition of phosphatidyl-inositol 3 kinase/protein kinase B or p38 mitogen-activated protein kinase, as well as silencing of scavenger receptor BI. Moreover, the presence of VEGF-A was found to be a prerequisite for the localization of scavenger receptor BI in the plasma membrane of endothelial cells.
Conclusions—
The identification of VEGF as a regulatory factor of transendothelial transport of HDL but not LDL supports the concept that the endothelium is a specific and, hence, druggable barrier for the entry of lipoproteins into the vascular wall.
Collapse
Affiliation(s)
- Srividya Velagapudi
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Mustafa Yalcinkaya
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Antonio Piemontese
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Roger Meier
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Simon Flyvbjerg Nørrelykke
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Damir Perisa
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Andrzej Rzepiela
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Michael Stebler
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Szymon Stoma
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Paolo Zanoni
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Lucia Rohrer
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| | - Arnold von Eckardstein
- From the Institute of Clinical Chemistry, University and University Hospital of Zurich, Schlieren, Switzerland (S.V., M.Y., A.P., D.P., P.Z., L.R., A.v.E.); Competence Center for Integrated Human Physiology, University of Zurich, Switzerland (S.V., M.Y., D.P., P.Z., L.R., A.v.E.); Department of Pharmacy, University of Parma, Italy (A.P.); and Scientific Center for Optical and Electron Microscopy, ETH Zurich, Switzerland (R.M., S.F.N., A.R., M.S., S.S.)
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW To outline the roles of SR-B1 and PDZK1 in hepatic selective HDL cholesterol uptake and reverse cholesterol transport and the consequences for atherosclerosis development. RECENT FINDINGS Much of our understanding of the physiological roles of SR-B1 and PDZK1 in HDL metabolism and atherosclerosis comes from studies of genetically manipulated mice. These show SR-B1 and PDZK1 play key roles in HDL metabolism and protection against atherosclerosis. The recent identification of rare loss of function mutations in the human SCARB1 gene verifies that it plays similar roles in HDL metabolism in humans. Other rare mutations in both the human SCARB1 and PDZK1 genes remain to be characterized but may have potentially devastating consequences to SR-B1 function. SUMMARY Identification of carriers of rare mutations in human SCARB1 and PDZK1 that impair the function of their gene products and characterization of the effects of these mutations on HDL cholesterol levels and atherosclerosis will add to our understanding of the importance of HDL function and cholesterol flux, as opposed to HDL-cholesterol levels, per se, for protection against cardiovascular disease.
Collapse
Affiliation(s)
- Bernardo L Trigatti
- aDepartment of Biochemistry and Biomedical Sciences, McMaster University bThrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
30
|
Hoekstra M. SR-BI as target in atherosclerosis and cardiovascular disease - A comprehensive appraisal of the cellular functions of SR-BI in physiology and disease. Atherosclerosis 2017; 258:153-161. [DOI: 10.1016/j.atherosclerosis.2017.01.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 12/12/2022]
|
31
|
Gutierrez-Pajares JL, Ben Hassen C, Chevalier S, Frank PG. SR-BI: Linking Cholesterol and Lipoprotein Metabolism with Breast and Prostate Cancer. Front Pharmacol 2016; 7:338. [PMID: 27774064 PMCID: PMC5054001 DOI: 10.3389/fphar.2016.00338] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022] Open
Abstract
Studies have demonstrated the significant role of cholesterol and lipoprotein metabolism in the progression of cancer. The SCARB1 gene encodes the scavenger receptor class B type I (SR-BI), which is an 82-kDa glycoprotein with two transmembrane domains separated by a large extracellular loop. SR-BI plays an important role in the regulation of cholesterol exchange between cells and high-density lipoproteins. Accordingly, hepatic SR-BI has been shown to play an essential role in the regulation of the reverse cholesterol transport pathway, which promotes the removal and excretion of excess body cholesterol. In the context of atherosclerosis, SR-BI has been implicated in the regulation of intracellular signaling, lipid accumulation, foam cell formation, and cellular apoptosis. Furthermore, since lipid metabolism is a relevant target for cancer treatment, recent studies have focused on examining the role of SR-BI in this pathology. While signaling pathways have initially been explored in non-tumoral cells, studies with cancer cells have now demonstrated SR-BI's function in tumor progression. In this review, we will discuss the role of SR-BI during tumor development and malignant progression. In addition, we will provide insights into the transcriptional and post-transcriptional regulation of the SCARB1 gene. Overall, studying the role of SR-BI in tumor development and progression should allow us to gain useful information for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Jorge L Gutierrez-Pajares
- Université François Rabelais de Tours, Faculté de Médecine-INSERM UMR1069 "Nutrition, Croissance et Cancer" Tours, France
| | - Céline Ben Hassen
- Université François Rabelais de Tours, Faculté de Médecine-INSERM UMR1069 "Nutrition, Croissance et Cancer" Tours, France
| | - Stéphan Chevalier
- Université François Rabelais de Tours, Faculté de Médecine-INSERM UMR1069 "Nutrition, Croissance et Cancer" Tours, France
| | - Philippe G Frank
- Université François Rabelais de Tours, Faculté de Médecine-INSERM UMR1069 "Nutrition, Croissance et Cancer" Tours, France
| |
Collapse
|
32
|
Pal R, Ke Q, Pihan GA, Yesilaltay A, Penman ML, Wang L, Chitraju C, Kang PM, Krieger M, Kocher O. Carboxy-terminal deletion of the HDL receptor reduces receptor levels in liver and steroidogenic tissues, induces hypercholesterolemia, and causes fatal heart disease. Am J Physiol Heart Circ Physiol 2016; 311:H1392-H1408. [PMID: 27694217 DOI: 10.1152/ajpheart.00463.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/13/2016] [Indexed: 01/15/2023]
Abstract
The HDL receptor SR-BI mediates the transfer of cholesteryl esters from HDL to cells and controls HDL abundance and structure. Depending on the genetic background, loss of SR-BI causes hypercholesterolemia, anemia, reticulocytosis, splenomegaly, thrombocytopenia, female infertility, and fatal coronary heart disease (CHD). The carboxy terminus of SR-BI (505QEAKL509) must bind to the cytoplasmic adaptor PDZK1 for normal hepatic-but not steroidogenic cell-expression of SR-BI protein. To determine whether SR-BI's carboxy terminus is also required for normal protein levels in steroidogenic cells, we introduced into SR-BI's gene a 507Ala/STOP mutation that produces a truncated receptor (SR-BIΔCT). As expected, the dramatic reduction of hepatic receptor protein in SR-BIΔCT mice was similar to that in PDZK1 knockout (KO) mice. Unlike SR-BI KO females, SR-BIΔCT females were fertile. The severity of SR-BIΔCT mice's hypercholesterolemia was intermediate between those of SR-BI KO and PDZK1 KO mice. Substantially reduced levels of the receptor in adrenal cortical cells, ovarian cells, and testicular Leydig cells in SR-BIΔCT mice suggested that steroidogenic cells have an adaptor(s) functionally analogous to hepatic PDZK1. When SR-BIΔCT mice were crossed with apolipoprotein E KO mice (SR-BIΔCT/apoE KO), pathologies including hypercholesterolemia, macrocytic anemia, hepatic and splenic extramedullary hematopoiesis, massive splenomegaly, reticulocytosis, thrombocytopenia, and rapid-onset and fatal occlusive coronary arterial atherosclerosis and CHD (median age of death: 9 wk) were observed. These results provide new insights into the control of SR-BI in steroidogenic cells and establish SR-BIΔCT/apoE KO mice as a new animal model for the study of CHD.
Collapse
Affiliation(s)
- Rinku Pal
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Qingen Ke
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - German A Pihan
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ayce Yesilaltay
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Marsha L Penman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Li Wang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Chandramohan Chitraju
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Peter M Kang
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Olivier Kocher
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
33
|
ApoA-I induces S1P release from endothelial cells through ABCA1 and SR-BI in a positive feedback manner. J Physiol Biochem 2016; 72:657-667. [PMID: 27377933 DOI: 10.1007/s13105-016-0504-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 06/23/2016] [Indexed: 10/21/2022]
Abstract
Sphingosine-1-phosphate (S1P), which has emerged as a pivotal signaling mediator that participates in the regulation of multiple cellular processes, is derived from various cells, including vascular endothelial cells. S1P accumulates in lipoproteins, especially HDL, and the majority of free plasma S1P is bound to HDL. We hypothesized that HDL-associated S1P is released through mechanisms associated with the HDL maturation process. ApoA-I, a major HDL apolipoprotein, is a critical factor for nascent HDL formation and lipid trafficking via ABCA1. Moreover, apoA-I is capable of promoting bidirectional lipid movement through SR-BI. In the present study, we confirmed that apoA-I can facilitate the production and release of S1P by HUVECs. Furthermore, we demonstrated that ERK1/2 and SphK activation induced by apoA-I is involved in the release of S1P from HUVECs. Inhibitor and siRNA experiments showed that ABCA1 and SR-BI are required for S1P release and ERK1/2 phosphorylation induced by apoA-I. However, the effects triggered by apoA-I were not suppressed by inhibiting ABCA1/JAK2 or the SR-BI/Src pathway. S1P released due to apoA-I activation can stimulate the (ERK1/2)/SphK1 pathway through S1PR (S1P receptor) 1/3. These results indicated that apoA-I not only promotes S1P release through ABCA1 and SR-BI but also indirectly activates the (ERK1/2)/SphK1 pathway by releasing S1P to trigger their receptors. In conclusion, we suggest that release of S1P induced by apoA-I from endothelial cells through ABCA1 and SR-BI is a self-positive-feedback process: apoA-I-(ABCA1 and SR-BI)-(S1P release)-S1PR-ERK1/2-SphK1-(S1P production)-(more S1P release induced by apoA-I).
Collapse
|
34
|
Annema W, von Eckardstein A. Dysfunctional high-density lipoproteins in coronary heart disease: implications for diagnostics and therapy. Transl Res 2016; 173:30-57. [PMID: 26972566 DOI: 10.1016/j.trsl.2016.02.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 12/18/2022]
Abstract
Low plasma levels of high-density lipoprotein (HDL) cholesterol are associated with increased risks of coronary heart disease. HDL mediates cholesterol efflux from macrophages for reverse transport to the liver and elicits many anti-inflammatory and anti-oxidative activities which are potentially anti-atherogenic. Nevertheless, HDL has not been successfully targeted by drugs for prevention or treatment of cardiovascular diseases. One potential reason is the targeting of HDL cholesterol which does not capture the structural and functional complexity of HDL particles. Hundreds of lipid species and dozens of proteins as well as several microRNAs have been identified in HDL. This physiological heterogeneity is further increased in pathologic conditions due to additional quantitative and qualitative molecular changes of HDL components which have been associated with both loss of physiological function and gain of pathologic dysfunction. This structural and functional complexity of HDL has prevented clear assignments of molecules to the functions of normal HDL and dysfunctions of pathologic HDL. Systematic analyses of structure-function relationships of HDL-associated molecules and their modifications are needed to test the different components and functions of HDL for their relative contribution in the pathogenesis of atherosclerosis. The derived biomarkers and targets may eventually help to exploit HDL for treatment and diagnostics of cardiovascular diseases.
Collapse
Affiliation(s)
- Wijtske Annema
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
35
|
Vaziri ND. Disorders of lipid metabolism in nephrotic syndrome: mechanisms and consequences. Kidney Int 2016; 90:41-52. [PMID: 27165836 DOI: 10.1016/j.kint.2016.02.026] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/02/2016] [Accepted: 02/11/2016] [Indexed: 12/17/2022]
Abstract
Nephrotic syndrome results in hyperlipidemia and profound alterations in lipid and lipoprotein metabolism. Serum cholesterol, triglycerides, apolipoprotein B (apoB)-containing lipoproteins (very low-density lipoprotein [VLDL], immediate-density lipoprotein [IDL], and low-density lipoprotein [LDL]), lipoprotein(a) (Lp[a]), and the total cholesterol/high-density lipoprotein (HDL) cholesterol ratio are increased in nephrotic syndrome. This is accompanied by significant changes in the composition of various lipoproteins including their cholesterol-to-triglyceride, free cholesterol-to-cholesterol ester, and phospholipid-to-protein ratios. These abnormalities are mediated by changes in the expression and activities of the key proteins involved in the biosynthesis, transport, remodeling, and catabolism of lipids and lipoproteins including apoproteins A, B, C, and E; 3-hydroxy-3-methylglutaryl-coenzyme A reductase; fatty acid synthase; LDL receptor; lecithin cholesteryl ester acyltransferase; acyl coenzyme A cholesterol acyltransferase; HDL docking receptor (scavenger receptor class B, type 1 [SR-B1]); HDL endocytic receptor; lipoprotein lipase; and hepatic lipase, among others. The disorders of lipid and lipoprotein metabolism in nephrotic syndrome contribute to the development and progression of cardiovascular and kidney disease. In addition, by limiting delivery of lipid fuel to the muscles for generation of energy and to the adipose tissues for storage of energy, changes in lipid metabolism contribute to the reduction of body mass and impaired exercise capacity. This article provides an overview of the mechanisms, consequences, and treatment of lipid disorders in nephrotic syndrome.
Collapse
Affiliation(s)
- Nosratola D Vaziri
- Division of Nephrology and Hypertension, Departments of Medicine, Physiology, and Biophysics, University of California, Irvine, Irvine, California.
| |
Collapse
|
36
|
Li Y, Zhao M, He D, Zhao X, Zhang W, Wei L, Huang E, Ji L, Zhang M, Willard B, Fu Z, Wang L, Pan B, Zheng L, Ji L. HDL in diabetic nephropathy has less effect in endothelial repairing than diabetes without complications. Lipids Health Dis 2016; 15:76. [PMID: 27074994 PMCID: PMC4831084 DOI: 10.1186/s12944-016-0246-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/08/2016] [Indexed: 12/13/2022] Open
Abstract
Background Diabetic nephropathy has a high cardiovascular risk with a low-level HDL(high density lipoprotein) in epidemiologic studies. Glycated HDL in diabetes can diminish the capacity to stimulate endothelial cell migration, but the mechanism has not been adequately explored in diabetic nephropathy. We performed this study to find out whether HDL in diabetic nephropathy is more dysfunctional than HDL in diabetes without complications. Methods Endothelial cells were treated with N-HDL (normal), D-HDL (T2DM[type 2 diabetes mellitus] without complications), DN-HDL (T2DM nephropathy), N-apoA-I (normal apoA-I), and G-apoA-I (glycated apoA-I in vitro). Cell migration capacity was measured with wound-healing and transwell migration assay in vitro and electric carotid injury model in vivo. Protein glycation levels were measured with nanoLC-MS/MS. PI3K expression and Akt phosphorylation were analyzed by western blot. Results In wound-healing assay, DN-HDL showed a 17.12 % decrease compared with D-HDL (p < 0.05). DN-HDL showed a 29.85 % decrease in comparison with D-HDL (p < 0.001) in transwell assay. In the electric carotid injury model, D-HDL and DN-HDL impaired the re-endothelialization capacity; DN-HDL was less effective than D-HDL. Meanwhile, DN-HDL was found to have a significantly higher protein glycation level than D-HDL (p < 0.001). PI3K expression and Akt phosphorylation were reduced significantly in DN-HDL in comparison with D-HDL and N-HDL. Conclusions We found that HDL from diabetic nephropathy has a higher level of glycation and induced less cell migration in vitro and in vivo compared with that from diabetes without nephropathy. This finding suggests that diabetic nephropathy has higher levels of glycated HDL and partially explains why patients with DN have a higher risk of cardiovascular disease. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0246-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yufeng Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No.11 Xizhimen Nan Dajie, Xicheng District, Beijing, 100044, China.,Department of Endocrinology and Metabolism, Capital Medical University Pinggu Teaching Hospital, Beijing, 101200, China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Dan He
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Xuyang Zhao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Wenjing Zhang
- Department of Obstetrics, The Military General Hospital of Beijing, Beijing, 100700, China
| | - Lixin Wei
- Department of Nephrology, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, China
| | - Edgar Huang
- School of Informatics and Computing, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Liang Ji
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Meng Zhang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Belinda Willard
- Cleveland Clinic Lerner Research Institute Mass Spectrometry Laboratory for Protein Sequencing, Cleveland, Ohio, USA
| | - Zuodi Fu
- Department of Endocrinology and Metabolism, Capital Medical University Pinggu Teaching Hospital, Beijing, 101200, China
| | - Lijuan Wang
- Department of Endocrinology and Metabolism, Capital Medical University Pinggu Teaching Hospital, Beijing, 101200, China
| | - Bing Pan
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China.
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No.11 Xizhimen Nan Dajie, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
37
|
Baranova IN, Souza ACP, Bocharov AV, Vishnyakova TG, Hu X, Vaisman BL, Amar MJ, Chen Z, Kost Y, Remaley AT, Patterson AP, Yuen PST, Star RA, Eggerman TL. Human SR-BI and SR-BII Potentiate Lipopolysaccharide-Induced Inflammation and Acute Liver and Kidney Injury in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:3135-47. [PMID: 26936883 PMCID: PMC4856165 DOI: 10.4049/jimmunol.1501709] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/24/2016] [Indexed: 12/18/2022]
Abstract
The class B scavenger receptors BI (SR-BI) and BII (SR-BII) are high-density lipoprotein receptors that recognize various pathogens, including bacteria and their products. It has been reported that SR-BI/II null mice are more sensitive than normal mice to endotoxin-induced inflammation and sepsis. Because the SR-BI/II knockout model demonstrates multiple immune and metabolic disorders, we investigated the role of each receptor in the LPS-induced inflammatory response and tissue damage using transgenic mice with pLiv-11-directed expression of human SR-BI (hSR-BI) or human SR-BII (hSR-BII). At 6 h after i.p. LPS injection, transgenic hSR-BI and hSR-BII mice demonstrated markedly higher serum levels of proinflammatory cytokines and 2- to 3-fold increased expression levels of inflammatory mediators in the liver and kidney, compared with wild-type (WT) mice. LPS-stimulated inducible NO synthase expression was 3- to 6-fold higher in the liver and kidney of both transgenic strains, although serum NO levels were similar in all mice. Despite the lower high-density lipoprotein plasma levels, both transgenic strains responded to LPS by a 5-fold increase of plasma corticosterone levels, which were only moderately lower than in WT animals. LPS treatment resulted in MAPK activation in tissues of all mice; however, the strongest response was detected for hepatic extracellular signal-regulated protein kinase 1 and 2 and kidney JNK of both transgenic mice. Histological examination of hepatic and renal tissue from LPS-challenged mice revealed more injury in hSR-BII, but not hSR-BI, transgenic mice versus WT controls. Our findings demonstrate that hSR-BII, and to a lesser extent hSR-BI, significantly increase LPS-induced inflammation and contribute to LPS-induced tissue injury in the liver and kidney, two major organs susceptible to LPS toxicity.
Collapse
Affiliation(s)
- Irina N Baranova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Ana C P Souza
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Alexander V Bocharov
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892;
| | - Tatyana G Vishnyakova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Xuzhen Hu
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Boris L Vaisman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Marcelo J Amar
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Zhigang Chen
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Yana Kost
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Alan T Remaley
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Amy P Patterson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Peter S T Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Robert A Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Thomas L Eggerman
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892; Division of Diabetes, Endocrinology, and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
38
|
Lv P, Tong X, Peng Q, Liu Y, Jin H, Liu R, Sun W, Pan B, Zheng L, Huang Y. Treatment with the herbal medicine, naoxintong improves the protective effect of high-density lipoproteins on endothelial function in patients with type 2 diabetes. Mol Med Rep 2016; 13:2007-16. [PMID: 26781332 PMCID: PMC4768949 DOI: 10.3892/mmr.2016.4792] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 12/23/2015] [Indexed: 12/28/2022] Open
Abstract
The protective effect of high-density lipoprotein (HDL) on endothelial function is impaired in patients with type 2 diabetes mellitus (T2DM), which may result in atherosclerotic complications. Naoxintong (NXT) is a compound preparation that includes Radix Astragali, Angelicae sinensis, Radix Paeoniae Rubra and Ligusticum wallichii. It is widely administered in China to prevent atherosclerotic complications. In the present study, NXT was administered to 69 patients with T2DM. HDLs were isolated from patient blood samples prior to and following the intervention. In vitro endothelial functions of HDL, including proliferation, migration, angiogenesis, and anti-apoptosis were investigated by bromodeoxyuridine, wound healing, Transwell and Matrigel tube formation assays on human umbilical vein endothelial cells (HUVECs). The results from the present study demonstrated that HUVECs treated with HDL isolated from diabetic patients following NXT therapy exhibited increased proliferative effects (10–27%; P<0.05), and improved migration ability (15–35%; P<0.05), anti-apoptotic function (23–34%; P<0.05) and angiogenesis (30–54%; P<0.001). Furthermore, the phosphorylation levels of Akt (26–36%; P<0.01) and extracellular signal-regulated kinase (16–80%; P<0.01) were increased following NXT therapy. The present in vitro study demonstrates that the protective effect of HDL on endothelial function is markedly impaired in diabetic patients who tend to develop atherosclerosis, and the impaired function may be partly abrogated by NXT.
Collapse
Affiliation(s)
- Pu Lv
- Department of Neurology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Xunliang Tong
- Department of Neurology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Qing Peng
- Department of Neurology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Yuanyuan Liu
- Department of Neurology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Ran Liu
- Department of Neurology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Wei Sun
- Department of Neurology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Bing Pan
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
39
|
Abstract
The adrenal gland is one of the prominent sites for steroid hormone synthesis. Lipoprotein-derived cholesterol esters (CEs) delivered via SR-B1 constitute the dominant source of cholesterol for steroidogenesis, particularly in rodents. Adrenocorticotropic hormone (ACTH) stimulates steroidogenesis through downstream actions on multiple components involved in steroidogenesis. Both acute and chronic ACTH treatments can modulate SR-B1 function, including its transcription, posttranscriptional stability, phosphorylation and dimerization status, as well as the interaction with other protein partners, all of which result in changes in the ability of SR-B1 to mediate HDL-CE uptake and the supply of cholesterol for conversion to steroids. Here, we provide a review of the recent findings on the regulation of adrenal SR-B1 function by ACTH.
Collapse
Affiliation(s)
- Wen-Jun Shen
- The Division of Endocrinology, Stanford University, Stanford, CA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Salman Azhar
- The Division of Endocrinology, Stanford University, Stanford, CA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Fredric B. Kraemer
- The Division of Endocrinology, Stanford University, Stanford, CA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- *Correspondence: Fredric B. Kraemer,
| |
Collapse
|
40
|
Cameron SJ, Morrell CN, Bao C, Swaim AF, Rodriguez A, Lowenstein CJ. A Novel Anti-Inflammatory Effect for High Density Lipoprotein. PLoS One 2015; 10:e0144372. [PMID: 26680360 PMCID: PMC4683005 DOI: 10.1371/journal.pone.0144372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 11/17/2015] [Indexed: 11/21/2022] Open
Abstract
High density lipoprotein has anti-inflammatory effects in addition to mediating reverse cholesterol transport. While many of the chronic anti-inflammatory effects of high density lipoprotein (HDL) are attributed to changes in cell adhesion molecules, little is known about acute signal transduction events elicited by HDL in endothelial cells. We now show that high density lipoprotein decreases endothelial cell exocytosis, the first step in leukocyte trafficking. ApoA-I, a major apolipoprotein of HDL, mediates inhibition of endothelial cell exocytosis by interacting with endothelial scavenger receptor-BI which triggers an intracellular protective signaling cascade involving protein kinase C (PKC). Other apolipoproteins within the HDL particle have only modest effects upon endothelial exocytosis. Using a human primary culture of endothelial cells and murine apo-AI knockout mice, we show that apo-AI prevents endothelial cell exocytosis which limits leukocyte recruitment. These data suggest that high density lipoprotein may inhibit diseases associated with vascular inflammation in part by blocking endothelial exocytosis.
Collapse
Affiliation(s)
- Scott J. Cameron
- Departments of Medicine, Division of Cardiology, University of Rochester School of Medicine, Box 679, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
- * E-mail:
| | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
- Department of Comparative Medicine, The Johns Hopkins University School of Medicine 733 N. Broadway, MRB 827, Baltimore, MD, 21205, United States of America
| | - Clare Bao
- Department of Medicine, The Johns Hopkins University School of Medicine, 950 Ross Building, 720 Rutland Ave, Baltimore, MD, 21205, United States of America
| | - AnneMarie F. Swaim
- Department of Comparative Medicine, The Johns Hopkins University School of Medicine 733 N. Broadway, MRB 827, Baltimore, MD, 21205, United States of America
| | - Annabelle Rodriguez
- Department of Cell Biology, University of Connecticut School of Medicine, E5050, 263 Farmington Avenue, Farmington, CT, 06030, United States of America
| | - Charles J. Lowenstein
- Departments of Medicine, Division of Cardiology, University of Rochester School of Medicine, Box 679, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, 601 Elmwood Avenue, Rochester, NY, 14652, United States of America
| |
Collapse
|
41
|
Angius F, Spolitu S, Uda S, Deligia S, Frau A, Banni S, Collu M, Accossu S, Madeddu C, Serpe R, Batetta B. High-density lipoprotein contribute to G0-G1/S transition in Swiss NIH/3T3 fibroblasts. Sci Rep 2015; 5:17812. [PMID: 26640042 PMCID: PMC4671069 DOI: 10.1038/srep17812] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 10/19/2015] [Indexed: 01/08/2023] Open
Abstract
High density lipoproteins (HDLs) play a crucial role in removing excess cholesterol from peripheral tissues. Although their concentration is lower during conditions of high cell growth rate (cancer and infections), their involvement during cell proliferation is not known. To this aim, we investigated the replicative cycles in synchronised Swiss 3T3 fibroblasts in different experimental conditions: i) contact-inhibited fibroblasts re-entering cell cycle after dilution; ii) scratch-wound assay; iii) serum-deprived cells induced to re-enter G1 by FCS, HDL or PDGF. Analyses were performed during each cell cycle up to quiescence. Cholesterol synthesis increased remarkably during the replicative cycles, decreasing only after cells reached confluence. In contrast, cholesteryl ester (CE) synthesis and content were high at 24 h after dilution and then decreased steeply in the successive cycles. Flow cytometry analysis of DiO-HDL, as well as radiolabeled HDL pulse, demonstrated a significant uptake of CE-HDL in 24 h. DiI-HDL uptake, lipid droplets (LDs) and SR-BI immunostaining and expression followed the same trend. Addition of HDL or PDGF partially restore the proliferation rate and significantly increase SR-BI and pAKT expression in serum-deprived cells. In conclusion, cell transition from G0 to G1/S requires CE-HDL uptake, leading to CE-HDL/SR-BI pathway activation and CEs increase into LDs.
Collapse
Affiliation(s)
- Fabrizio Angius
- Unit of Experimental Medicine, University of Cagliari, Cagliari, Italy
| | - Stefano Spolitu
- Unit of Experimental Medicine, University of Cagliari, Cagliari, Italy
| | - Sabrina Uda
- Unit of Experimental Medicine, University of Cagliari, Cagliari, Italy
| | - Stefania Deligia
- Unit of Experimental Medicine, University of Cagliari, Cagliari, Italy
| | - Alessandra Frau
- Unit of Experimental Medicine, University of Cagliari, Cagliari, Italy
| | - Sebastiano Banni
- Divisions of Physiology, University of Cagliari, Cagliari, Italy
| | - Maria Collu
- Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Simonetta Accossu
- Unit of Experimental Medicine, University of Cagliari, Cagliari, Italy
| | - Clelia Madeddu
- Department of Biomedical Sciences, Department of Medical Sciences "Mario Aresu", University of Cagliari, Cagliari, Italy
| | - Roberto Serpe
- Department of Biomedical Sciences, Department of Medical Sciences "Mario Aresu", University of Cagliari, Cagliari, Italy
| | - Barbara Batetta
- Unit of Experimental Medicine, University of Cagliari, Cagliari, Italy
| |
Collapse
|
42
|
Dysfunctional High-Density Lipoprotein: An Innovative Target for Proteomics and Lipidomics. CHOLESTEROL 2015; 2015:296417. [PMID: 26634153 PMCID: PMC4655037 DOI: 10.1155/2015/296417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/12/2015] [Accepted: 10/12/2015] [Indexed: 02/02/2023]
Abstract
High-Density Lipoprotein-Cholesterol (HDL-C) is regarded as an important protective factor against cardiovascular disease, with abundant evidence of an inverse relationship between its serum levels and risk of cardiovascular disease, as well as various antiatherogenic, antioxidant, and anti-inflammatory properties. Nevertheless, observations of hereditary syndromes featuring scant HDL-C concentration in absence of premature atherosclerotic disease suggest HDL-C levels may not be the best predictor of cardiovascular disease. Indeed, the beneficial effects of HDL may not depend solely on their concentration, but also on their quality. Distinct subfractions of this lipoprotein appear to be constituted by specific protein-lipid conglomerates necessary for different physiologic and pathophysiologic functions. However, in a chronic inflammatory microenvironment, diverse components of the HDL proteome and lipid core suffer alterations, which propel a shift towards a dysfunctional state, where HDL-C becomes proatherogenic, prooxidant, and proinflammatory. This heterogeneity highlights the need for further specialized molecular studies in this aspect, in order to achieve a better understanding of this dysfunctional state; with an emphasis on the potential role for proteomics and lipidomics as valuable methods in the search of novel therapeutic approaches for cardiovascular disease.
Collapse
|
43
|
Prostacyclin receptors: Transcriptional regulation and novel signalling mechanisms. Prostaglandins Other Lipid Mediat 2015; 121:70-82. [DOI: 10.1016/j.prostaglandins.2015.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/25/2015] [Accepted: 04/18/2015] [Indexed: 12/24/2022]
|
44
|
Trigatti BL, Fuller M. HDL signaling and protection against coronary artery atherosclerosis in mice. J Biomed Res 2015; 30:94-100. [PMID: 26642235 PMCID: PMC4820886 DOI: 10.7555/jbr.30.20150079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/29/2015] [Indexed: 01/18/2023] Open
Abstract
Atherosclerosis is a leading underlying factor in cardiovascular disease and stroke, important causes of morbidity and mortality across the globe. Abundant epidemiological studies demonstrate that high levels of high density lipoprotein (HDL) are associated with reduced risk of atherosclerosis and preclinical, animal model studies demonstrate that this association is causative. Understanding the molecular mechanisms underlying the protective effects of HDL will allow more strategic approaches to development of HDL based therapeutics. Recent evidence suggests that an important aspect of the ability of HDL to protect against atherosclerosis is its ability to trigger signaling responses in a variety of target cells including endothelial cells and macrophages in the vessel wall. These signaling responses require the HDL receptor, scavenger receptor class B type 1 (SR-B1), an adaptor protein (PDZK1) that binds to the cytosolic C terminus of SR-B1, Akt1 activation and (at least in endothelial cells) activation of endothelial NO synthase (eNOS). Mouse models of atherosclerosis, exemplified by apolipoprotein E or low density lipoprotein receptor gene inactivated mice (apoE or LDLR KO) develop atherosclerosis in their aortas but appear generally resistant to coronary artery atherosclerosis. On the other hand, inactivation of each of the components of HDL signaling (above) in either apoE or LDLR KO mice renders them susceptible to extensive coronary artery atherosclerosis suggesting that HDL signaling may play an important role in protection against coronary artery disease.
Collapse
Affiliation(s)
- Bernardo L Trigatti
- Department of Biochemistry and Biomedical Sciences, McMaster University and Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences., Hamilton, Ontario L8L 2X2, Canada;
| | - Mark Fuller
- Department of Biochemistry and Biomedical Sciences, McMaster University and Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences., Hamilton, Ontario L8L 2X2, Canada
| |
Collapse
|
45
|
Tao H, Yancey PG, Babaev VR, Blakemore JL, Zhang Y, Ding L, Fazio S, Linton MF. Macrophage SR-BI mediates efferocytosis via Src/PI3K/Rac1 signaling and reduces atherosclerotic lesion necrosis. J Lipid Res 2015; 56:1449-60. [PMID: 26059978 PMCID: PMC4513986 DOI: 10.1194/jlr.m056689] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Indexed: 12/13/2022] Open
Abstract
Macrophage apoptosis and efferocytosis are key determinants of atherosclerotic plaque inflammation and necrosis. Bone marrow transplantation studies in ApoE- and LDLR-deficient mice revealed that hematopoietic scavenger receptor class B type I (SR-BI) deficiency results in severely defective efferocytosis in mouse atherosclerotic lesions, resulting in a 17-fold higher ratio of free to macrophage-associated dead cells in lesions containing SR-BI−/− cells, 5-fold more necrosis, 65.2% less lesional collagen content, nearly 7-fold higher dead cell accumulation, and 2-fold larger lesion area. Hematopoietic SR-BI deletion elicited a maladaptive inflammatory response [higher interleukin (IL)-1β, IL-6, and TNF-α lower IL-10 and transforming growth factor β]. Efferocytosis of apoptotic thymocytes was reduced by 64% in SR-BI−/− versus WT macrophages, both in vitro and in vivo. In response to apoptotic cells, macrophage SR-BI bound with phosphatidylserine and induced Src phosphorylation and cell membrane recruitment, which led to downstream activation of phosphoinositide 3-kinase (PI3K) and Ras-related C3 botulinum toxin substrate 1 (Rac1) for engulfment and clearance of apoptotic cells, as inhibition of Src decreased PI3K, Rac1-GTP, and efferocytosis in WT cells. Pharmacological inhibition of Rac1 reduced macrophage efferocytosis in a SR-BI-dependent fashion, and activation of Rac1 corrected the defective efferocytosis in SR-BI−/− macrophages. Thus, deficiency of macrophage SR-BI promotes defective efferocytosis signaling via the Src/PI3K/Rac1 pathway, resulting in increased plaque size, necrosis, and inflammation.
Collapse
Affiliation(s)
- Huan Tao
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Patricia G Yancey
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Vladimir R Babaev
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John L Blakemore
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Youmin Zhang
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Lei Ding
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Sergio Fazio
- Department of Medicine, Physiology, and Pharmacology, Center of Preventive Cardiology, Oregon Health and Science University, Portland, OR 97239
| | - MacRae F Linton
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232 Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
46
|
Lee WR, Sacharidou A, Behling-Kelly E, Oltmann SC, Zhu W, Ahmed M, Gerard RD, Hui DY, Abe JI, Shaul PW, Mineo C. PDZK1 prevents neointima formation via suppression of breakpoint cluster region kinase in vascular smooth muscle. PLoS One 2015; 10:e0124494. [PMID: 25886360 PMCID: PMC4401672 DOI: 10.1371/journal.pone.0124494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 03/02/2015] [Indexed: 01/21/2023] Open
Abstract
Scavenger receptor class B, type I (SR-BI) and its adaptor protein PDZK1 mediate responses to HDL cholesterol in endothelium. Whether the receptor-adaptor protein tandem serves functions in other vascular cell types is unknown. The current work determined the roles of SR-BI and PDZK1 in vascular smooth muscle (VSM). To evaluate possible VSM functions of SR-BI and PDZK1 in vivo, neointima formation was assessed 21 days post-ligation in the carotid arteries of wild-type, SR-BI-/- or PDZK1-/- mice. Whereas neointima development was negligible in wild-type and SR-BI-/-, there was marked neointima formation in PDZK1-/- mice. PDZK1 expression was demonstrated in primary mouse VSM cells, and compared to wild-type cells, PDZK1-/- VSM displayed exaggerated proliferation and migration in response to platelet derived growth factor (PDGF). Tandem affinity purification-mass spectrometry revealed that PDZK1 interacts with breakpoint cluster region kinase (Bcr), which contains a C-terminal PDZ binding sequence and is known to enhance responses to PDGF in VSM. PDZK1 interaction with Bcr in VSM was demonstrated by pull-down and by coimmunoprecipitation, and the augmented proliferative response to PDGF in PDZK1-/- VSM was abrogated by Bcr depletion. Furthermore, compared with wild-type Bcr overexpression, the introduction of a Bcr mutant incapable of PDZK1 binding into VSM cells yielded an exaggerated proliferative response to PDGF. Thus, PDZK1 has novel SR-BI-independent function in VSM that affords protection from neointima formation, and this involves PDZK1 suppression of VSM cell proliferation via an inhibitory interaction with Bcr.
Collapse
Affiliation(s)
- Wan Ru Lee
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Erica Behling-Kelly
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Sarah C. Oltmann
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Weifei Zhu
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Mohamed Ahmed
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Robert D. Gerard
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - David Y. Hui
- Department of Pathology, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Jun-ichi Abe
- Department of Medicine and the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Philip W. Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail: (PS); (CM)
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail: (PS); (CM)
| |
Collapse
|
47
|
Tan JTM, Ng MKC, Bursill CA. The role of high-density lipoproteins in the regulation of angiogenesis. Cardiovasc Res 2015; 106:184-93. [PMID: 25759067 DOI: 10.1093/cvr/cvv104] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 02/22/2015] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is important for postnatal physiological processes including tissue neovascularization in response to an ischaemic injury. Conversely, uncontrolled inflammatory-driven angiogenesis can accelerate atherosclerotic plaque and tumour growth. Angiogenesis-associated diseases are highly prevalent globally, with cardiovascular-related disorders and cancer being the leading causes of mortality worldwide. A vast amount of research has been conducted on the vasculoprotective effects of high-density lipoproteins (HDLs) and while current HDL-raising therapies to date have not yielded the desired benefits clinically, its role in angiogenesis is yet to be fully elucidated. Epidemiological studies report positive correlations between elevated HDL levels and improved prognosis in both ischaemia- and inflammatory-driven pathologies, in which angiogenesis plays a key role. This review focuses on current evidence from epidemiological and prospective studies, coupled with animal models and mechanistic studies that highlight the ability of HDL to conditionally regulate angiogenesis.
Collapse
Affiliation(s)
- Joanne T M Tan
- The Heart Research Institute, 7 Eliza Street, Newtown, Sydney, New South Wales 2042, Australia Sydney Medical School, University of Sydney, Sydney, Australia
| | - Martin K C Ng
- The Heart Research Institute, 7 Eliza Street, Newtown, Sydney, New South Wales 2042, Australia Sydney Medical School, University of Sydney, Sydney, Australia Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Christina A Bursill
- The Heart Research Institute, 7 Eliza Street, Newtown, Sydney, New South Wales 2042, Australia Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
48
|
Wu BJ, Shrestha S, Ong KL, Johns D, Hou L, Barter PJ, Rye KA. Cholesteryl ester transfer protein inhibition enhances endothelial repair and improves endothelial function in the rabbit. Arterioscler Thromb Vasc Biol 2015; 35:628-36. [PMID: 25633313 DOI: 10.1161/atvbaha.114.304747] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE High-density lipoproteins (HDLs) can potentially protect against atherosclerosis by multiple mechanisms, including enhancement of endothelial repair and improvement of endothelial function. This study asks if increasing HDL levels by inhibiting cholesteryl ester transfer protein activity with the anacetrapib analog, des-fluoro-anacetrapib, enhances endothelial repair and improves endothelial function in New Zealand White rabbits with balloon injury of the abdominal aorta. APPROACH AND RESULTS New Zealand White rabbits received chow or chow supplemented with 0.07% or 0.14% (wt/wt) des-fluoro-anacetrapib for 8 weeks. Endothelial denudation of the abdominal aorta was carried out after 2 weeks. The animals were euthanized 6 weeks postinjury. Treatment with 0.07% and 0.14% des-fluoro-anacetrapib reduced cholesteryl ester transfer protein activity by 81±4.9% and 92±12%, increased plasma apolipoprotein A-I levels by 1.4±0.1-fold and 1.5±0.1-fold, increased plasma HDL-cholesterol levels by 1.8±0.2-fold and 1.9±0.1-fold, reduced intimal hyperplasia by 37±11% and 51±10%, and inhibited vascular cell proliferation by 25±6.1% and 35±6.7%, respectively. Re-endothelialization of the injured aorta increased from 43±6.7% (control) to 69±6.6% and 76±7.7% in the 0.07% and 0.14% des-fluoro-anacetrapib-treated animals, respectively. Aortic ring relaxation and guanosine 3',5'-cyclic monophosphate production in response to acetylcholine were also improved. Incubation of HDLs from the des-fluoro-anacetrapib-treated animals with human coronary artery endothelial cells increased cell proliferation and migration relative to control. These effects were abolished by knockdown of scavenger receptor-B1 and PDZ domain-containing protein 1 and by pharmacological inhibition of phosphatidylinositol-4,5-bisphosphate 3-kinase/Akt. CONCLUSIONS Increasing HDL levels by inhibiting cholesteryl ester transfer protein reduces intimal thickening and regenerates functional endothelium in damaged New Zealand White rabbit aortas in an scavenger receptor-B1-dependent and phosphatidylinositol-4,5-bisphosphate 3-kinase/Akt-dependent manner.
Collapse
Affiliation(s)
- Ben J Wu
- From the Centre for Vascular Research, The University of New South Wales, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.); Merck & Co, Inc, Kenilworth, NJ (D.J.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (B.J.W., K.L.O., P.J.B., K.-A.R.); and Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.).
| | - Sudichhya Shrestha
- From the Centre for Vascular Research, The University of New South Wales, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.); Merck & Co, Inc, Kenilworth, NJ (D.J.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (B.J.W., K.L.O., P.J.B., K.-A.R.); and Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.)
| | - Kwok L Ong
- From the Centre for Vascular Research, The University of New South Wales, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.); Merck & Co, Inc, Kenilworth, NJ (D.J.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (B.J.W., K.L.O., P.J.B., K.-A.R.); and Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.)
| | - Douglas Johns
- From the Centre for Vascular Research, The University of New South Wales, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.); Merck & Co, Inc, Kenilworth, NJ (D.J.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (B.J.W., K.L.O., P.J.B., K.-A.R.); and Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.)
| | - Liming Hou
- From the Centre for Vascular Research, The University of New South Wales, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.); Merck & Co, Inc, Kenilworth, NJ (D.J.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (B.J.W., K.L.O., P.J.B., K.-A.R.); and Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.)
| | - Philip J Barter
- From the Centre for Vascular Research, The University of New South Wales, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.); Merck & Co, Inc, Kenilworth, NJ (D.J.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (B.J.W., K.L.O., P.J.B., K.-A.R.); and Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.)
| | - Kerry-Anne Rye
- From the Centre for Vascular Research, The University of New South Wales, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.); Merck & Co, Inc, Kenilworth, NJ (D.J.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (B.J.W., K.L.O., P.J.B., K.-A.R.); and Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (B.J.W., S.S., K.L.O., L.H., P.J.B., K.-A.R.).
| |
Collapse
|
49
|
Abstract
High-density lipoproteins (HDLs) exert many beneficial effects which may help to protect against the development or progression of atherosclerosis or even facilitate lesion regression. These activities include promoting cellular cholesterol efflux, protecting low-density lipoproteins (LDLs) from modification, preserving endothelial function, as well as anti-inflammatory and antithrombotic effects. However, questions remain about the relative importance of these activities for atheroprotection. Furthermore, the many molecules (both lipids and proteins) associated with HDLs exert both distinct and overlapping activities, which may be compromised by inflammatory conditions, resulting in either loss of function or even gain of dysfunction. This complexity of HDL functionality has so far precluded elucidation of distinct structure-function relationships for HDL or its components. A better understanding of HDL metabolism and structure-function relationships is therefore crucial to exploit HDLs and its associated components and cellular pathways as potential targets for anti-atherosclerotic therapies and diagnostic markers.
Collapse
Affiliation(s)
- Wijtske Annema
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland,
| | | | | |
Collapse
|
50
|
Abstract
Numerous epidemiologic studies revealed that high-density lipoprotein (HDL) is an important risk factor for coronary heart disease. There are several well-documented HDL functions such as reversed cholesterol transport, inhibition of inflammation, or inhibition of platelet activation that may account for the atheroprotective effects of this lipoprotein. Mechanistically, these functions are carried out by a direct interaction of HDL particle or its components with receptors localized on the cell surface followed by generation of intracellular signals. Several HDL-associated receptor ligands such as apolipoprotein A-I (apoA-I) or sphingosine-1-phosphate (S1P) have been identified in addition to HDL holoparticles, which interact with surface receptors such as ATP-binding cassette transporter A1 (ABCA1); S1P receptor types 1, 2, and 3 (S1P1, S1P2, and S1P3); or scavenger receptor type I (SR-BI) and activate intracellular signaling cascades encompassing kinases, phospholipases, trimeric and small G-proteins, and cytoskeletal proteins such as actin or junctional protein such as connexin43. In addition, depletion of plasma cell cholesterol mediated by ABCA1, ATP-binding cassette transporter G1 (ABCG1), or SR-BI was demonstrated to indirectly inhibit signaling over proinflammatory or proliferation-stimulating receptors such as Toll-like or growth factor receptors. The present review summarizes the current knowledge regarding the HDL-induced signal transduction and its relevance to athero- and cardioprotective effects as well as other physiological effects exerted by HDL.
Collapse
|