1
|
Zhang J, Chen C, Geng Q, Li H, Wu M, Chan B, Wang S, Sheng W. ZNF263 cooperates with ZNF31 to promote the drug resistance and EMT of pancreatic cancer through transactivating RNF126. J Cell Physiol 2024; 239:e31259. [PMID: 38515383 DOI: 10.1002/jcp.31259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
The poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is attribute to the aggressive local invasion, distant metastasis and drug resistance of PDAC patients, which was strongly accelerated by epithelial-mesenchymal transition (EMT). In current study, we systematically investigate the role of ZNF263/RNF126 axis in the initiation of EMT in PDAC in vitro and vivo. ZNF263 is firstly identified as a novel transactivation factor of RNF126. Both ZNF263 and RNF126 were overexpressed in PDAC tissues, which were associated with multiple advanced clinical stages and poor prognosis of PDAC patients. ZNF263 overexpression promoted cell proliferation, drug resistance and EMT in vitro via activating RNF126 following by the upregulation of Cyclin D1, N-cad, and MMP9, and the downregulation of E-cad, p21, and p27. ZNF263 silencing contributed to the opposite phenotype. Mechanistically, ZNF263 transactivated RNF126 via binding to its promoter. Further investigations revealed that ZNF263 interacted with ZNF31 to coregulate the transcription of RNF126, which in turn promoted ubiquitination-mediated degradation of PTEN. The downregulation of PTEN activated AKT/Cyclin D1 and AKT/GSK-3β/β-catenin signaling, thereby promoting the malignant phenotype of PDAC. Finally, the coordination of ZNF263 and RNF126 promotes subcutaneous tumor size and distant liver metastasis in vivo. ZNF263, as an oncogene, promotes proliferation, drug resistance and EMT of PDAC through transactivating RNF126.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chuanping Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qilong Geng
- Department of Clinical Medicine, The First Clinical College, Anhui Medical University, Hefei, Anhui, China
| | - Haoyu Li
- Department of Clinical Medicine, The First Clinical College, Anhui Medical University, Hefei, Anhui, China
| | - Mengcheng Wu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Boyuan Chan
- Department of Clinical Medicine, The First Clinical College, Anhui Medical University, Hefei, Anhui, China
| | - Shiyang Wang
- Department of Geriatric Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Weiwei Sheng
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
2
|
Li Z, Zhang W, Wang QR, Yang YJ, Liu XH, Cheng G, Chang FJ. Effect of Thrombolysis on Circulating Microparticles in Patients with ST-Segment Elevation Myocardial Infarction. Cardiovasc Ther 2023; 2023:5559368. [PMID: 38024103 PMCID: PMC10676276 DOI: 10.1155/2023/5559368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/11/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Objective We demonstrated that circulating microparticles (MPs) are increased in patients with coronary heart disease (both chronic coronary syndrome (CCS) and acute coronary syndrome). Whether thrombolysis affects MPs in patients with ST-segment elevation myocardial infarction (STEMI) with or without percutaneous coronary intervention (PCI) is unknown. Methods This study was divided into three groups: STEMI patients with thrombolysis (n = 18) were group T, patients with chronic coronary syndrome (n = 20) were group CCS, and healthy volunteers (n = 20) were the control group. Fasting venous blood was extracted from patients in the CCS and control groups, and venous blood was extracted from patients in the T group before (pre-T) and 2 hours after (post-T) thrombolysis. MPs from each group were obtained by centrifugation. After determining the concentration, the effects of MPs on endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) in rat myocardial tissue in vitro were detected by immunohistochemistry and western blotting. Changes in nitric oxide (NO) and oxygen free radicals (O2•-) were also detected. The effect of MPs on vasodilation in isolated rat thoracic aortae was detected. Results Compared with that in the control group (2.60 ± 0.38 mg/ml), the concentration of MPs was increased in patients with CCS (3.49 ± 0.72 mg/ml) and in STEMI patients before thrombolysis (4.17 ± 0.58 mg/ml). However, thrombolysis did not further increase MP levels (post-T, 4.23 ± 1.01 mg/ml) compared with those in STEMI patients before thrombolysis. Compared with those in the control group, MPs in both CCS and STEMI patients before thrombolysis inhibited the expression of eNOS (both immunohistochemistry and western blot analysis of phosphorylation at Ser1177), NO production in the isolated myocardium and vasodilation in vitro and stimulated the expression of iNOS (immunohistochemistry and western blot analysis of phosphorylation at Thr495), and the generation of O2•- in the isolated myocardium. The effects of MPs were further enhanced by MPs from STEMI patients 2 hours after thrombolysis. Conclusion Changes in MP function after thrombolysis may be one of the mechanisms leading to ischemia-reperfusion after thrombolysis.
Collapse
Affiliation(s)
- Zhe Li
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Wei Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Qun-Rang Wang
- Department of Cardiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xian'yang, China
| | - Yu-juan Yang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xin-Hong Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Gong Cheng
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Feng-Jun Chang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
3
|
Schoch L, Alcover S, Padró T, Ben-Aicha S, Mendieta G, Badimon L, Vilahur G. Update of HDL in atherosclerotic cardiovascular disease. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2023; 35:297-314. [PMID: 37940388 DOI: 10.1016/j.arteri.2023.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Epidemiologic evidence supported an inverse association between HDL (high-density lipoprotein) cholesterol (HDL-C) levels and atherosclerotic cardiovascular disease (ASCVD), identifying HDL-C as a major cardiovascular risk factor and postulating diverse HDL vascular- and cardioprotective functions beyond their ability to drive reverse cholesterol transport. However, the failure of several clinical trials aimed at increasing HDL-C in patients with overt cardiovascular disease brought into question whether increasing the cholesterol cargo of HDL was an effective strategy to enhance their protective properties. In parallel, substantial evidence supports that HDLs are complex and heterogeneous particles whose composition is essential for maintaining their protective functions, subsequently strengthening the "HDL quality over quantity" hypothesis. The following state-of-the-art review covers the latest understanding as per the roles of HDL in ASCVD, delves into recent advances in understanding the complexity of HDL particle composition, including proteins, lipids and other HDL-transported components and discusses on the clinical outcomes after the administration of HDL-C raising drugs with particular attention to CETP (cholesteryl ester transfer protein) inhibitors.
Collapse
Affiliation(s)
- Leonie Schoch
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Sebastián Alcover
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
| | | | - Guiomar Mendieta
- Cardiology Unit, Cardiovascular Clinical Institute, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; Cardiovascular Research Chair, UAB, 08025 Barcelona, Spain; CiberCV, Institute of Health Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; CiberCV, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Katsimardou A, Patoulias D, Zografou I, Siskos F, Stavropoulos K, Imprialos K, Tegou Z, Boulmpou A, Georgopoulou V, Hatzipapa N, Papadopoulos C, Doumas M. The Impact of Metabolic Syndrome Components on Erectile Function in Patients with Type 2 Diabetes. Metabolites 2023; 13:metabo13050617. [PMID: 37233658 DOI: 10.3390/metabo13050617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/31/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Erectile dysfunction is commonly encountered in diabetic patients and in patients with metabolic syndrome; however, only a few studies have assessed patients with metabolic syndrome and type 2 diabetes mellitus (T2DM) regarding their sexual function. The purpose of this study is to examine the effect of metabolic syndrome and its components on the erectile function of T2DM patients. A cross-sectional study including T2DM patients was conducted from November 2018 until November 2020. Participants were evaluated for the presence of metabolic syndrome and their sexual function was assessed using the International Index of Erectile Function (IIEF) questionnaire. A total of 45 consecutive male patients participated in this study. Metabolic syndrome was diagnosed in 84.4% and erectile dysfunction (ED) in 86.7% of them. Metabolic syndrome was not associated with ED or ED severity. Among metabolic syndrome components, only high-density lipoprotein cholesterol (HDL) was associated with ED [x2 (1, n = 45) = 3.894, p = 0.048; OR = 5.5 (95% CI: 0.890-33.99)] and with the IIEF erectile function scores (median 23 vs. 18, U = 75, p = 0.012). Multiple regression analyses showed that HDL was non-significantly associated with the IIEF erectile function scores. In conclusion, among T2DM patients HDL is associated with ED.
Collapse
Affiliation(s)
- Alexandra Katsimardou
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Dimitrios Patoulias
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Ioanna Zografou
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Fotios Siskos
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Konstantinos Stavropoulos
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Konstantinos Imprialos
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Zoi Tegou
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Aristi Boulmpou
- 3rd Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Vivian Georgopoulou
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Nikoleta Hatzipapa
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Christodoulos Papadopoulos
- 3rd Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Michael Doumas
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
- Veterans Affairs Medical Center, George Washington University, Washington, DC 20422, USA
| |
Collapse
|
5
|
Kim M, An G, Lim W, Song G. Alachlor breaks down intracellular calcium homeostasis and leads to cell cycle arrest through JNK/MAPK and PI3K/AKT signaling mechanisms in bovine mammary gland epithelial cells. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2022; 184:105063. [PMID: 35715071 DOI: 10.1016/j.pestbp.2022.105063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/31/2022] [Accepted: 02/22/2022] [Indexed: 06/15/2023]
Abstract
Alachlor is a widely used herbicide for the cultivation of various grains employed as food for cattle. The mechanisms leading to the toxic effects of alachlor on epithelial cells of the bovine mammary gland are not well known. Thus, this study was conducted to clarify the toxicological effects of alachlor on the immortalized epithelial cell line of the bovine mammary gland (MAC-T) cells. After treatment, many factors related to cell viability, proliferation, and cellular homeostasis were evaluated. Alachlor arrested cell cycle progression by blocking the expression of cyclin and cyclin-dependent kinases, and induced the breakdown of Ca2+ homeostasis. The cytosolic and mitochondrial levels of Ca2+ were also abnormally increased after the treatment of cells with alachlor, ultimately leading to the depolarization of mitochondrial membrane potential in MAC-T cells. The signaling cascade was found to be dysregulated by the abnormal phosphorylation of signaling molecules involved in PI3K/AKT (AKT, p70S6K, and S6) and MAPK/JNK (JNK and c-Jun) pathways. In these mechanisms, exposure to alachlor led to a reduction in the viability and proliferation of MAC-T cells. Altogether, the toxic effects of alachlor can lead to abnormal conditions in epithelial cells of the bovine mammary gland, which might hinder these cells from performing their main role, such as producing milk.
Collapse
Affiliation(s)
- Miji Kim
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
6
|
Hooshdaran B, Pressly BB, Alferiev IS, Smith JD, Zoltick PW, Tschabrunn CM, Wilensky RL, Gorman RC, Levy RJ, Fishbein I. Stent-based delivery of AAV2 vectors encoding oxidation-resistant apoA1. Sci Rep 2022; 12:5464. [PMID: 35361857 PMCID: PMC8971450 DOI: 10.1038/s41598-022-09524-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/16/2022] [Indexed: 12/14/2022] Open
Abstract
In-stent restenosis (ISR) complicates revascularization in the coronary and peripheral arteries. Apolipoprotein A1 (apoA1), the principal protein component of HDL possesses inherent anti-atherosclerotic and anti-restenotic properties. These beneficial traits are lost when wild type apoA1(WT) is subjected to oxidative modifications. We investigated whether local delivery of adeno-associated viral (AAV) vectors expressing oxidation-resistant apoA1(4WF) preserves apoA1 functionality. The efflux of 3H-cholesterol from macrophages to the media conditioned by endogenously produced apoA1(4WF) was 2.1-fold higher than for apoA1(WT) conditioned media in the presence of hypochlorous acid emulating conditions of oxidative stress. The proliferation of apoA1(WT)- and apoA1(4FW)-transduced rat aortic smooth muscle cells (SMC) was inhibited by 66% ± 10% and 65% ± 11%, respectively, in comparison with non-transduced SMC (p < 0.001). Conversely, the proliferation of apoA1(4FW)-transduced, but not apoA1(WT)-transduced rat blood outgrowth endothelial cells (BOEC) was increased 41% ± 5% (p < 0.001). Both apoA1 transduction conditions similarly inhibited basal and TNFα-induced reactive oxygen species in rat aortic endothelial cells (RAEC) and resulted in the reduced rat monocyte attachment to the TNFα-activated endothelium. AAV2-eGFP vectors immobilized reversibly on stainless steel mesh surfaces through the protein G/anti-AAV2 antibody coupling, efficiently transduced cells in culture modeling stent-based delivery. In vivo studies in normal pigs, deploying AAV2 gene delivery stents (GDS) preloaded with AAV2-eGFP in the coronary arteries demonstrated transduction of the stented arteries. However, implantation of GDS formulated with AAV2-apoA1(4WF) failed to prevent in-stent restenosis in the atherosclerotic vasculature of hypercholesterolemic diabetic pigs. It is concluded that stent delivery of AAV2-4WF while feasible, is not effective for mitigation of restenosis in the presence of severe atherosclerotic disease.
Collapse
Affiliation(s)
- Bahman Hooshdaran
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
| | - Benjamin B Pressly
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
| | - Ivan S Alferiev
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Jonathan D Smith
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, USA
| | - Philip W Zoltick
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
| | - Cory M Tschabrunn
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Robert L Wilensky
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Robert C Gorman
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Robert J Levy
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Ilia Fishbein
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA.
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA.
| |
Collapse
|
7
|
Homo Sapiens (Hsa)-microRNA (miR)-6727-5p Contributes to the Impact of High-Density Lipoproteins on Fibroblast Wound Healing In Vitro. MEMBRANES 2022; 12:membranes12020154. [PMID: 35207076 PMCID: PMC8876102 DOI: 10.3390/membranes12020154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/26/2022]
Abstract
Chronic, non-healing wounds are a significant cause of global morbidity and mortality, and strategies to improve delayed wound closure represent an unmet clinical need. High-density lipoproteins (HDL) can enhance wound healing, but exploitation of this finding is challenging due to the complexity and instability of these heterogeneous lipoproteins. The responsiveness of primary human neonatal keratinocytes, and neonatal and human dermal fibroblasts (HDF) to HDL was confirmed by cholesterol efflux, but promotion of ‘scrape’ wound healing occurred only in primary human neonatal (HDFn) and adult fibroblasts (HDFa). Treatment of human fibroblasts with HDL induced multiple changes in the expression of small non-coding microRNA sequences, determined by microchip array, including hsa-miR-6727-5p. Intriguingly, levels of hsa-miR-6727-5p increased in HDFn, but decreased in HDFa, after exposure to HDL. Delivery of a hsa-miR-6727-5p mimic elicited repression of different target genes in HDFn (ZNF584) and HDFa (EDEM3, KRAS), and promoted wound closure in HDFn. By contrast, a hsa-miR-6727-5p inhibitor promoted wound closure in HDFa. We conclude that HDL treatment exerts distinct effects on the expression of hsa-miR-6727-5p in neonatal and adult fibroblasts, and that this is a sequence which plays differential roles in wound healing in these cell types, but cannot replicate the myriad effects of HDL.
Collapse
|
8
|
Grao-Cruces E, Lopez-Enriquez S, Martin ME, Montserrat-de la Paz S. High-density lipoproteins and immune response: A review. Int J Biol Macromol 2022; 195:117-123. [PMID: 34896462 DOI: 10.1016/j.ijbiomac.2021.12.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 01/04/2023]
Abstract
High-density lipoproteins (HDLs) are heterogeneous lipoproteins that modify their composition and functionality depending on physiological or pathological conditions. The main roles of HDL are cholesterol efflux, and anti-inflammatory and antioxidant functions. These functions can be compromised under pathological conditions. HDLs play a role in the immune system as anti-inflammatory molecules but when inflammation occurs, HDLs change their composition and carry pro-inflammatory cargo. Hence, many molecular intermediates that influence inflammatory microenvironments and cell signaling pathways can modulate HDLs structural modification and function. This review provides a comprehensive assessment of the importance of HDL composition and anti-inflammatory function in the onset and progression of atherosclerotic cardiovascular diseases. On the other hand, immune cell activation during progression of atheroma plaque formation can be influenced by HDLs through HDL-derived cholesterol depletion from lipid rafts and through HDL interaction with HDL receptors expressed on T and B lymphocytes. Cholesterol efflux is mediated by HDL receptors located in lipid rafts in peripheral cells, which undergo membrane structural modifications, and interferes with subsequent molecules interactions or intracellular signaling cascades. Regarding antigen-presentation cells such as macrophages or dendritic cells, HDL function may then modulate lymphocytes activation in immune response. Our review also contributes to the understanding of the effects exerted by HDLs in signal transduction associated to our immune cell population during chronic diseases progression.
Collapse
Affiliation(s)
- Elena Grao-Cruces
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Soledad Lopez-Enriquez
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Maria E Martin
- Department of Cell Biology, Faculty of Biology, University of Seville, Av. Reina Mercedes s/n, 41012 Seville, Spain
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain.
| |
Collapse
|
9
|
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]
|
10
|
Robert J, Osto E, von Eckardstein A. The Endothelium Is Both a Target and a Barrier of HDL's Protective Functions. Cells 2021; 10:1041. [PMID: 33924941 PMCID: PMC8146309 DOI: 10.3390/cells10051041] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 12/11/2022] Open
Abstract
The vascular endothelium serves as a barrier between the intravascular and extravascular compartments. High-density lipoproteins (HDL) have two kinds of interactions with this barrier. First, bloodborne HDL must pass the endothelium to access extravascular tissues, for example the arterial wall or the brain, to mediate cholesterol efflux from macrophages and other cells or exert other functions. To complete reverse cholesterol transport, HDL must even pass the endothelium a second time to re-enter circulation via the lymphatics. Transendothelial HDL transport is a regulated process involving scavenger receptor SR-BI, endothelial lipase, and ATP binding cassette transporters A1 and G1. Second, HDL helps to maintain the integrity of the endothelial barrier by (i) promoting junction closure as well as (ii) repair by stimulating the proliferation and migration of endothelial cells and their progenitor cells, and by preventing (iii) loss of glycocalix, (iv) apoptosis, as well as (v) transmigration of inflammatory cells. Additional vasoprotective functions of HDL include (vi) the induction of nitric oxide (NO) production and (vii) the inhibition of reactive oxygen species (ROS) production. These vasoprotective functions are exerted by the interactions of HDL particles with SR-BI as well as specific agonists carried by HDL, notably sphingosine-1-phophate (S1P), with their specific cellular counterparts, e.g., S1P receptors. Various diseases modify the protein and lipid composition and thereby the endothelial functionality of HDL. Thorough understanding of the structure-function relationships underlying the multiple interactions of HDL with endothelial cells is expected to elucidate new targets and strategies for the treatment or prevention of various diseases.
Collapse
Affiliation(s)
| | | | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, 8091 Zurich, Switzerland; (J.R.); (E.O.)
| |
Collapse
|
11
|
Cole J, Blackhurst DM, Solomon GAE, Ratanjee BD, Benjamin R, Marais AD. Atherosclerotic cardiovascular disease in hyperalphalipoproteinemia due to LIPG variants. J Clin Lipidol 2021; 15:142-150.e2. [PMID: 33414088 DOI: 10.1016/j.jacl.2020.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND High density lipoprotein cholesterol (HDL-C) concentration correlates inversely with atherosclerotic cardiovascular disease (ASCVD) risk and is included in risk calculations. Endothelial lipase (EL) is a phospholipase that remodels HDL. Deficiency of EL due to mutations in its gene, LIPG, is associated with hyperalphalipoproteinemia. The effects of EL on HDL function and ASCVD risk remain poorly understood. OBJECTIVES To determine whether hyperalphalipoproteinemia due to EL deficiency is protective against ASCVD. METHODS We identified LIPG variants amongst patients with severe hyperalphalipoproteinemia (HDL-C >2.5 mmol/L) attending a referral lipid clinic in the Western Cape Province of South Africa. We analysed the clinical and biochemical phenotypes amongst primary hyperalphalipoproteinemia cases (males HDL-C >1.6 mmol/L; females HDL-C >1.8 mmol/L) due to LIPG variants, and the distribution of variants in normal and hyperalphalipoproteinemia ranges of HDL-C. RESULTS 1007 patients with HDL-C concentration ranging from 1.2 to 4.5 mmol/L were included. Seventeen females had primary hyperalphalipoproteinemia. Vascular disease was prominent, but not associated with HDL-C concentration, LDL-C concentration or carotid artery intima media thickness. Two novel and three known LIPG variants were identified in severe hyperalphalipoproteinemia. Four additional variants were identified in the extended cohort. Two common variants appeared normally distributed across the HDL-C concentration range, while six less-common variants were found only at higher HDL-C concentrations. One rare variant had a moderate effect. CONCLUSION Hyperalphalipoproteinemia due to LIPG variants is commoner in females and may not protect against ASCVD. Use of current risk calculations may be inappropriate in patients with hyperalphalipoproteinemia due to EL deficiency. Our study cautions targeting EL to reduce risk.
Collapse
Affiliation(s)
- Justine Cole
- Division of Chemical Pathology, University of Cape Town Faculty of Health Sciences, Anzio Road, Observatory, 7925, Cape Town, South Africa; Chemical Pathology, National Health Laboratory Service, C17 Groote Schuur Hospital, Main Road, Observatory, 7925, Cape Town, South Africa.
| | - Diane Mary Blackhurst
- Division of Chemical Pathology, University of Cape Town Faculty of Health Sciences, Anzio Road, Observatory, 7925, Cape Town, South Africa
| | - Gabriele Anna Eva Solomon
- Division of Chemical Pathology, University of Cape Town Faculty of Health Sciences, Anzio Road, Observatory, 7925, Cape Town, South Africa
| | - Bharati Dhanluxmi Ratanjee
- Division of Chemical Pathology, University of Cape Town Faculty of Health Sciences, Anzio Road, Observatory, 7925, Cape Town, South Africa
| | - Ryan Benjamin
- Division of Chemical Pathology, University of Cape Town Faculty of Health Sciences, Anzio Road, Observatory, 7925, Cape Town, South Africa; Chemical Pathology, National Health Laboratory Service, C17 Groote Schuur Hospital, Main Road, Observatory, 7925, Cape Town, South Africa
| | - Adrian David Marais
- Division of Chemical Pathology, University of Cape Town Faculty of Health Sciences, Anzio Road, Observatory, 7925, Cape Town, South Africa.
| |
Collapse
|
12
|
Abstract
The prevalence of heart failure (HF), including reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF), has increased significantly worldwide. However, the prognosis and treatment of HF are still not good. Recent studies have demonstrated that high-density lipoprotein (HDL) plays an important role in cardiac repair during HF. The exact role and mechanism of HDL in the regulation of HF remain unexplained. Here, we discuss recent findings regarding HDL in the progression of HF, such as the regulation of excitation-contraction coupling, energy homeostasis, inflammation, neurohormone activation, and microvascular dysfunction. The effects of HDL on the regulation of cardiac-related cells, such as endothelial cells (ECs), cardiomyocytes (CMs), and on cardiac resident immune cell dysfunction in HF are also explained. An in-depth understanding of HDL function in the heart may provide new strategies for the prevention and treatment of HF.
Collapse
|
13
|
The Role and Function of HDL in Patients with Chronic Kidney Disease and the Risk of Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21020601. [PMID: 31963445 PMCID: PMC7014265 DOI: 10.3390/ijms21020601] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health problem with steadily increasing occurrence. Significantly elevated cardiovascular morbidity and mortality have been observed in CKD. Cardiovascular diseases are the most important and frequent cause of death of CKD patients globally. The presence of CKD is related to disturbances in lipoprotein metabolism whose consequences are dyslipidemia and the accumulation of atherogenic particles. CKD not only fuels the reduction of high-density lipoprotein (HDL) cholesterol concentration, but also it modifies the composition of this lipoprotein. The key role of HDL is the participation in reverse cholesterol transport from peripheral tissues to the liver. Moreover, HDL prevents the oxidation of low-density lipoprotein (LDL) cholesterol by reactive oxygen species (ROS) and protects against the adverse effects of oxidized LDL (ox-LDL) on the endothelium. Numerous studies have demonstrated the ability of HDL to promote the production of nitric oxide (NO) by endothelial cells (ECs) and to exert antiapoptotic and anti-inflammatory effects. Increasing evidence suggests that in patients with chronic inflammatory disorders, HDLs may lose important antiatherosclerotic properties and become dysfunctional. So far, no therapeutic strategy to raise HDL, or alter the ratio of HDL subfractions, has been successful in slowing the progression of CKD or reducing cardiovascular disease in patients either with or without CKD.
Collapse
|
14
|
Yang N, Tian H, Zhan E, Zhai L, Jiao P, Yao S, Lu G, Mu Q, Wang J, Zhao A, Zhou Y, Qin S. Reverse-D-4F improves endothelial progenitor cell function and attenuates LPS-induced acute lung injury. Respir Res 2019; 20:131. [PMID: 31242908 PMCID: PMC6595601 DOI: 10.1186/s12931-019-1099-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/13/2019] [Indexed: 12/22/2022] Open
Abstract
Background Patients with acute lung injury (ALI) have increased levels of pro-inflammatory mediators, which impair endothelial progenitor cell (EPC) function. Increasing the number of EPC and alleviating EPC dysfunction induced by pro-inflammatory mediators play important roles in suppressing ALI development. Because the high density lipoprotein reverse-D-4F (Rev-D4F) improves EPC function, we hypothesized that it might repair lipopolysaccharide (LPS)-induced lung damage by improving EPC numbers and function in an LPS-induced ALI mouse model. Methods LPS was used to induce ALI in mice, and then the mice received intraperitoneal injections of Rev-D4F. Immunohistochemical staining, flow cytometry, MTT, transwell, and western blotting were used to assess the effect of Rev-D4F on repairment of lung impairment, and improvement of EPC numbers and function, as well as the signaling pathways involved. Results Rev-D4F inhibits LPS-induced pulmonary edema and decreases plasma levels of the pro-inflammatory mediators TNF-α and ET-1 in ALI mice. Rev-D4F inhibited infiltration of red and white blood cells into the interstitial space, reduced lung injury-induced inflammation, and restored injured pulmonary capillary endothelial cells. In addition, Rev-D4F increased numbers of circulating EPC, stimulated EPC differentiation, and improved EPC function impaired by LPS. Rev-D4F also acted via a PI3-kinase-dependent mechanism to restore levels of phospho-AKT, eNOS, and phospho-eNOS suppressed by LPS. Conclusions These findings indicate that Rev-D4F has an important vasculoprotective role in ALI by improving the EPC numbers and functions, and Rev-D4F reverses LPS-induced EPC dysfuncion partially through PI3K/AKT/eNOS signaling pathway. Electronic supplementary material The online version of this article (10.1186/s12931-019-1099-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nana Yang
- Experimental Center for Medical Research, Weifang Medical University, Weifang City, People's Republic of China.
| | - Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China
| | - Enxin Zhan
- Institute of Preschool Education, Jinan Preschool Education College, Jinan City, People's Republic of China
| | - Lei Zhai
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China
| | - Guohua Lu
- Experimental Center for Medical Research, Weifang Medical University, Weifang City, People's Republic of China
| | - Qingjie Mu
- Experimental Center for Medical Research, Weifang Medical University, Weifang City, People's Republic of China
| | - Juan Wang
- Department of Pharmaceutical Sciences, Binzhou Medical College, Yantai City, People's Republic of China
| | - Aihua Zhao
- Department of Emergency Medicine, the second Affiliated Hospital of Shandong First Medical University, Tai-an City, People's Republic of China
| | - Yadong Zhou
- Department of Emergency Medicine, the second Affiliated Hospital of Shandong First Medical University, Tai-an City, People's Republic of China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China. .,Heart Center of Shandong First Medical University, Tai-an City, People's Republic of China.
| |
Collapse
|
15
|
Eshaghi FS, Ghazizadeh H, Kazami-Nooreini S, Timar A, Esmaeily H, Mehramiz M, Avan A, Ghayour-Mobarhan M. Association of a genetic variant in AKT1 gene with features of the metabolic syndrome. Genes Dis 2019; 6:290-295. [PMID: 32042868 PMCID: PMC6997569 DOI: 10.1016/j.gendis.2019.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/28/2019] [Accepted: 03/15/2019] [Indexed: 11/30/2022] Open
Abstract
Metabolic syndrome (MetS) is a clustering of metabolic abnormalities that is associated with increased risk of developing cardiovascular disease and type 2 diabetes. There is growing body of data showing the associations of genetic variants of the genes involved in the PI3K/AKT/mTOR pathway with diabetes and obesity. We aimed to investigate the association between MetS and its components with the genetic polymorphism in AKT1, rs1130233 (T > C). Total of 618 participants, recruited from Mashhad stroke and heart atherosclerosis disorder cohort (MASHAD study). Patients with MetS were defined by using international diabetes federation (IDF) criteria (n = 326) and those without MetS (n = 261) were recruited. Anthropometric and biochemical parameters were measured in all subjects. Genetic analysis for the rs1130233 polymorphism was performed, using the ABI-StepOne instruments with SDS version-2.0 software. Individuals with MetS had a significantly higher levels of BMI, waist-circumference, total cholesterol, triglyceride, high sensitivity-c reactive protein (hs-CRP) and blood-pressure, and lower concentrations of high density lipoprotein (HDL-C), compared to non-MetS individuals (P < 0.05). The association between the rs1130233 and MetS was not significant. Subjects with a CC or CT genotypes had a significantly higher serum hs-CRP-level (OR: 1.5; 95% CI (1.05–2.1), P = 0.02). Additionally, subjects who carried the TC genotype had a higher BMI compared to the CC genotype (p value = 0.045). Our findings demonstrated that AKT1, rs1130233 (T > C) polymorphism was associated with major components of MetS such as hs-CRP, and BMI, indicating further investigation in a multi-center setting to explore its value as an emerging biomarker of risk stratification marker.
Collapse
Affiliation(s)
- Fateme Sadat Eshaghi
- Department of Biochemistry, Faculty of Basic Sciences, Hakim Sabzevary University, Sabzevar, Iran
| | - Hamideh Ghazizadeh
- Metabolic Syndrome and Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sakine Kazami-Nooreini
- Department of Biochemistry, Faculty of Basic Sciences, Hakim Sabzevary University, Sabzevar, Iran
| | - Ameneh Timar
- Department of Biochemistry, Faculty of Basic Sciences, Hakim Sabzevary University, Sabzevar, Iran
| | - Habibollah Esmaeily
- Social Department of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrane Mehramiz
- Metabolic Syndrome and Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome and Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome and Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Shi Y, Lv X, Liu Y, Li B, Liu M, Yan M, Liu Y, Li Q, Zhang X, He S, Zhu M, He J, Zhu Y, Zhu Y, Ai D. Elevating ATP‐binding cassette transporter G1 improves re‐endothelialization function of endothelial progenitor cells
via
Lyn/Akt/eNOS in diabetic mice. FASEB J 2018; 32:6525-6536. [DOI: 10.1096/fj.201800248rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Ying Shi
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Xue Lv
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Yanan Liu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Bochuan Li
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Mingming Liu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Meng Yan
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Yajin Liu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Qi Li
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Xuejiao Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Shuang He
- Tianjin Institute of Cardiovascular DiseaseTianjin Chest HospitalTianjinChina
| | - Mason Zhu
- Department of Molecular BiologyUniversity of CaliforniaSan Diego La JollaCaliforniaUSA
| | - Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Yan Zhu
- Tianjin Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| | - Ding Ai
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and PathophysiologyTianjin Medical UniversityTianjinChina
| |
Collapse
|
17
|
Orsatti CL, Sobreira ML, Sandrim VC, Nahas-Neto J, Orsatti FL, Nahas EAP. Autophagy-related 16-like 1gene polymorphism, risk factors for cardiovascular disease and associated carotid intima-media thickness in postmenopausal women. Clin Biochem 2018; 61:12-17. [PMID: 30236831 DOI: 10.1016/j.clinbiochem.2018.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Early identification of asymptomatic postmenopausal women (PW), who are more predisposed to developing cardiovascular disease (CVD), is an important preventive strategy. Autophagy-related 16-like 1 (ATG16L1) is an autophagy gene known to control host immune responses and is associated with a variety of diseases, including CVD. OBJECTIVE The aim of the study was to associate the ATG16L1 polymorphism variant with subclinical carotid atherosclerosis in asymptomatic PW. STUDY DESIGN This cross-sectional study included 210 Brazilian postmenopausal women (age ≥ 45 years with amenorrhea ≥12 months). Clinical, anthropometric and biochemical assessments were performed to evaluate the cardiovascular risk factors. DNA was extracted from buccal cells and the ATG16L1 (T300A) polymorphism was determined by the polymerase chain reaction (PCR). The carotid intima-media thickness and/or the presence of plaques were evaluated by carotid duplex ultrasound. For statistical analysis, the t-test, logistic regression and analysis of covariance (ANCOVA) were used. RESULTS The presence of the polymorphic allele forATG16L1 (T300A) was found in 77.47% (A/G = 49.87%, G/G = 27.60%). The ATG16L1 (T300A) polymorphism is significantly associated with increased carotid intima-media thickness (IMT) after adjustments of the confounding variables (P < .037). No significant associations were observed between the polymorphism with other risk factors for CVD in PW. CONCLUSION In postmenopausal women, the ATG16L1 (T300A) polymorphism is significantly associated with increased carotid IMT (marker of atherosclerotic disease) after adjustments of the confounding variables (P < .037). Thus, identifying the ATG16L1 polymorphism is an important strategy for screening asymptomatic PW who are more predisposed to developing CVD.
Collapse
Affiliation(s)
- Claudio Lera Orsatti
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil.
| | - Marcone Lima Sobreira
- Department of Surgery, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Valéria Cristina Sandrim
- Department of Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Jorge Nahas-Neto
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Fabio Lera Orsatti
- Department of Sport Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Eliana Aguiar Petri Nahas
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| |
Collapse
|
18
|
Syu JN, Yang MD, Tsai SY, Chiang EPI, Chiu SC, Chao CY, Rodriguez RL, Tang FY. S-allylcysteine Improves Blood Flow Recovery and Prevents Ischemic Injury by Augmenting Neovasculogenesis. Cell Transplant 2018; 26:1636-1647. [PMID: 29251114 PMCID: PMC5753981 DOI: 10.1177/0963689717724792] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Studies suggest that a low level of circulating human endothelial progenitor cells (EPCs) is a risk factor for ischemic injury and coronary artery disease (CAD). Consumption of S-allylcysteine (SAC) is known to prevent CAD. However, the protective effects of SAC on the ischemic injury are not yet clear. In this study, we examined whether SAC could improve blood flow recovery in ischemic tissues through EPC-mediated neovasculogenesis. The results demonstrate that SAC significantly enhances the neovasculogenesis of EPCs in vitro. The molecular mechanisms for SAC enhancement of neovasculogenesis include the activation of Akt/endothelial nitric oxide synthase signaling cascades. SAC increased the expression of c-kit, β-catenin, cyclin D1, and Cyclin-dependent kinase 4 (CDK4) proteins in EPCs. Daily intake of SAC at dosages of 0.2 and 2 mg/kg body weight significantly enhanced c-kit protein levels in vivo. We conclude that dietary consumption of SAC improves blood flow recovery and prevents ischemic injury by inducing neovasculogenesis in experimental models.
Collapse
Affiliation(s)
- Jia-Ning Syu
- Biomedical Science Laboratory, Department of Nutrition, China Medical University, Taichung, Taiwan, Republic of China
| | - Mei-Due Yang
- Department of Clinical Nutrition, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China
- Department of Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China
- Department of Medicine, China Medical University, Taichung, Taiwan, Republic of China
| | - Shu-Yao Tsai
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan, Republic of China
| | - En-Pei Isabel Chiang
- Department of Food Science and Biotechnology, NCHU-UCD Plant and Food Biotechnology Center and Agricultural Biotechnology Center (ABC), National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Shao-Chih Chiu
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan, Republic of China
- Center for Cell Therapy, China Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Che-Yi Chao
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan, Republic of China
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China
| | - Raymond L. Rodriguez
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Feng-Yao Tang
- Biomedical Science Laboratory, Department of Nutrition, China Medical University, Taichung, Taiwan, Republic of China
- Feng-Yao Tang, Biomedical Science Laboratory, Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan, Republic of China.
| |
Collapse
|
19
|
Lucchesi D, Popa SG, Sancho V, Giusti L, Garofolo M, Daniele G, Pucci L, Miccoli R, Penno G, Del Prato S. Influence of high density lipoprotein cholesterol levels on circulating monocytic angiogenic cells functions in individuals with type 2 diabetes mellitus. Cardiovasc Diabetol 2018; 17:78. [PMID: 29866130 PMCID: PMC5987640 DOI: 10.1186/s12933-018-0720-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/21/2018] [Indexed: 02/07/2023] Open
Abstract
Background High-density lipoproteins (HDLs) can exert anti-atherogenic effects. On top of removing excess cholesterol through reverse cholesterol transport, HDLs play beneficial actions on endothelial function and integrity. In particular, HDLs are strong determinant of endothelial progenitor cells (EPCs) number and function. To gain further insights into such an effect we characterized in vitro functionality of circulating “early” EPCs obtained from 60 type 2 diabetes individuals with low HDL-cholesterol (HDL-C) and 59 with high HDL-C levels. Methods After an overnight fast, venous blood was drawn in EDTA tubes and processed within 2-h from sampling. Peripheral blood mononuclear cells were isolated and plated on fibronectin coated culture dishes; after 3 days culture, adherent cells positive for Dil-ac-LDL/Lectin dual fluorescent staining were identified as monocytic angiogenic cells (MACs). After 5–7 days culture in EBM-2 medium, adherent cells were evaluated for viability/proliferation (MTT assay), senescence (beta-galactosidase activity detection), migration (modified Boyden chamber using VEGF as chemoattractant), adhesion capacity (on fibronectin-coated culture dishes) and ROS production (ROS-sensitive fluorescent probe CM-H2DCFDA). Results MACs obtained from diabetic individuals with high HDL-C had 23% higher viability compared to low HDL-C (111.6 ± 32.7% vs. 90.5 ± 28.6% optical density; p = 0.002). H2O2 exposure impaired MACs viability to a similar extent in both groups (109.2 ± 31.7% vs. 74.5 ± 40.8% in high HDL-C, p < 0.0001; 88.3 ± 25.5% vs. 72.3 ± 22.5% in low-HDL, p = 0.004). MACs senescence was comparable in the two groups (102.7 ± 29.8% vs. 99.2 ± 27.8%; p = 0.703) and was only slightly modified by exposure to H2O2. There was no difference in the MACs migration capacity between the two groups (91.3 ± 34.2% vs. 108.7 ± 39.5%; p = 0.111), as well as in MACs adhesion capacity (105.2 ± 32.7% vs. 94.1 ± 26.1%; p = 0.223). Finally, ROS production was slightly thought not significantly higher in MACs from type 2 diabetes individuals with low- than high-HDL. After stratification of HDL-C levels into quartiles, viability (p < 0.0001) and adhesion (p = 0.044) were higher in Q4 than in Q1–Q3. In logistic regression analysis, HDL-C was correlated to MACs viability and adhesion independently of HbA1c or BMI, respectively. Conclusions Our data suggest that in type 2 diabetes subjects, HDL-cholesterol is an independent determinant of circulating MACs functional capacities—mainly viability, to a lesser extent adhesion—likely contributing also through this mechanism to cardiovascular protection even in type 2 diabetes.
Collapse
Affiliation(s)
- Daniela Lucchesi
- Section of Diabetes and Metabolic Disease, Department of Clinical and Experimental Medicine, University of Pisa and Azienda Ospedaliero-Universitaria Pisana, Via Paradisa, 2, 56124, Pisa, Italy
| | - Simona Georgiana Popa
- Diabetes, Nutrition and Metabolic Diseases, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Veronica Sancho
- Section of Diabetes and Metabolic Disease, Department of Clinical and Experimental Medicine, University of Pisa and Azienda Ospedaliero-Universitaria Pisana, Via Paradisa, 2, 56124, Pisa, Italy
| | - Laura Giusti
- Section of Diabetes and Metabolic Disease, Department of Clinical and Experimental Medicine, University of Pisa and Azienda Ospedaliero-Universitaria Pisana, Via Paradisa, 2, 56124, Pisa, Italy
| | - Monia Garofolo
- Section of Diabetes and Metabolic Disease, Department of Clinical and Experimental Medicine, University of Pisa and Azienda Ospedaliero-Universitaria Pisana, Via Paradisa, 2, 56124, Pisa, Italy
| | - Giuseppe Daniele
- Section of Diabetes and Metabolic Disease, Department of Clinical and Experimental Medicine, University of Pisa and Azienda Ospedaliero-Universitaria Pisana, Via Paradisa, 2, 56124, Pisa, Italy
| | - Laura Pucci
- Institute of Agricultural Biology and Biotechnology, National Research Council (CNR), Pisa, Italy
| | - Roberto Miccoli
- Section of Diabetes and Metabolic Disease, Department of Clinical and Experimental Medicine, University of Pisa and Azienda Ospedaliero-Universitaria Pisana, Via Paradisa, 2, 56124, Pisa, Italy
| | - Giuseppe Penno
- Section of Diabetes and Metabolic Disease, Department of Clinical and Experimental Medicine, University of Pisa and Azienda Ospedaliero-Universitaria Pisana, Via Paradisa, 2, 56124, Pisa, Italy
| | - Stefano Del Prato
- Section of Diabetes and Metabolic Disease, Department of Clinical and Experimental Medicine, University of Pisa and Azienda Ospedaliero-Universitaria Pisana, Via Paradisa, 2, 56124, Pisa, Italy.
| |
Collapse
|
20
|
Cannizzo CM, Adonopulos AA, Solly EL, Ridiandries A, Vanags LZ, Mulangala J, Yuen SCG, Tsatralis T, Henriquez R, Robertson S, Nicholls SJ, Di Bartolo BA, Ng MKC, Lam YT, Bursill CA, Tan JTM. VEGFR2 is activated by high-density lipoproteins and plays a key role in the proangiogenic action of HDL in ischemia. FASEB J 2018; 32:2911-2922. [PMID: 29401597 DOI: 10.1096/fj.201700617r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
High-density lipoproteins augment hypoxia-induced angiogenesis by inducing the key angiogenic vascular endothelial growth factor A (VEGFA) and total protein levels of its receptor 2 (VEGFR2). The activation/phosphorylation of VEGFR2 is critical for mediating downstream, angiogenic signaling events. This study aimed to determine whether reconstituted high-density lipoprotein (rHDL) activates VEGFR2 phosphorylation and the downstream signaling events and the importance of VEGFR2 in the proangiogenic effects of rHDL in hypoxia. In vitro, rHDL increased VEGFR2 activation and enhanced phosphorylation of downstream, angiogenic signaling proteins ERK1/2 and p38 MAPK in hypoxia. Incubation with a VEGFR2-neutralizing antibody attenuated rHDL-induced phosphorylation of VEGFR2, ERK1/2, p38 MAPK, and tubule formation. In a murine model of ischemia-driven neovascularization, rHDL infusions enhanced blood perfusion and augmented capillary and arteriolar density. Infusion of a VEGFR2-neutralizing antibody ablated those proangiogenic effects of rHDL. Circulating Sca1+/CXCR4+ angiogenic progenitor cell levels, important for neovascularization in response to ischemia, were higher in rHDL-infused mice 3 d after ischemic induction, but that did not occur in mice that also received the VEGFR2-neutralizing antibody. In summary, VEGFR2 has a key role in the proangiogenic effects of rHDL in hypoxia/ischemia. These findings have therapeutic implications for angiogenic diseases associated with an impaired response to tissue ischemia.-Cannizzo, C. M., Adonopulos, A. A., Solly, E. L., Ridiandries, A., Vanags, L. Z., Mulangala, J., Yuen, S. C. G., Tsatralis, T., Henriquez, R., Robertson, S., Nicholls, S. J., Di Bartolo, B. A., Ng, M. K. C., Lam, Y. T., Bursill, C. A., Tan, J. T. M. VEGFR2 is activated by high-density lipoproteins and plays a key role in the proangiogenic action of HDL in ischemia.
Collapse
Affiliation(s)
- Carla M Cannizzo
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Aaron A Adonopulos
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Emma L Solly
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Anisyah Ridiandries
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Laura Z Vanags
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Jocelyne Mulangala
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| | - Sui Ching G Yuen
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Tania Tsatralis
- The Heart Research Institute, Newtown, New South Wales, Australia
| | - Rodney Henriquez
- The Heart Research Institute, Newtown, New South Wales, Australia
| | - Stacy Robertson
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Stephen J Nicholls
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| | - Belinda A Di Bartolo
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| | - Martin K C Ng
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Yuen Ting Lam
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Christina A Bursill
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia.,Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| | - Joanne T M Tan
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia.,Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| |
Collapse
|
21
|
Regulator of G protein signaling 4 inhibits human melanoma cells proliferation and invasion through the PI3K/AKT signaling pathway. Oncotarget 2017; 8:78530-78544. [PMID: 29108247 PMCID: PMC5667980 DOI: 10.18632/oncotarget.20825] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/26/2017] [Indexed: 11/25/2022] Open
Abstract
Melanoma is a tumor produced by skin melanocytes, which has a high metastatic rate and poor prognosis. So far, plenty of work has been done on melanoma, but mechanisms underlying melanoma development have not been fully elucidated. Here we identified regulator of G protein signaling 4(RGS4) as novel therapeutic target for malignant melanoma and its regulating effect on melanoma. We found that endogenous RGS4 expression was much lower in melanoma tissues and cells. In A375 cell line with low endogenous RGS4 expression, the function of RGS4 was detected by up-regulation its expression with pcDNA3.1-RGS4 and knockdown its expression with siRNA. Our results showed that RGS4 could significantly reduce the proliferation, migration and invasion of melanoma cells. RGS4 is an important regulator for the apoptosis of melanocyte, and the apoptosis rate is significantly decreased in low RGS4 enviroment. RGS4 induced non-activation of PI3K/AKT pathway, resulting in decreased expression of E2F1 and Cyclin D1, thus constraining cell proliferation and invasion. These results were further confirmed in M14 cell lines. Collectively, our findings show that RGS4 plays an important role in multiple cellular functions of melanoma development and is valuable to be a therapeutic target.
Collapse
|
22
|
Effect of Atorvastatin Therapy on the Level of CD34 +CD133 +CD309 + Endothelial Progenitor Cells in Patients with Coronary Heart Disease. Bull Exp Biol Med 2017; 163:133-136. [PMID: 28577105 DOI: 10.1007/s10517-017-3753-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Indexed: 02/07/2023]
Abstract
In 58 patients with coronary heart disease, the count of CD34+CD133+CD309+ endothelial progenitor cells in the blood was determined and the dynamics of the content of endothelial progenitor cells, angiogenic growth factors, and lipid parameters over 3 months of atorvastatin therapy was analyzed. Atorvastatin was administered in daily doses of 10 mg (26 patients) and 40 mg (32 patients). Control group comprised 15 healthy volunteers. In patients with coronary heart disease, the count of endothelial progenitor cells was lower by 4 times, the level of VEGF was higher by 52%, and the level of endostatin was lower by 13% than in healthy volunteers. Atorvastatin therapy significantly reduced the levels of VEGF (by 11%), C-reactive protein (by 26%), total cholesterol (by 30%), LDL cholesterol (by 35%), and triglycerides (by 18%); the levels of endostatin, MCP-1, and HDL cholesterol remained unchanged. The count of endothelial progenitor cells increased significantly by 72% irrespectively on the statin dose, but the changes were more pronounced in patients with lower initial endothelial progenitor cell counts and in patients with more drastic decrease in LDL cholesterol.
Collapse
|
23
|
Helkin A, Stein JJ, Lin S, Siddiqui S, Maier KG, Gahtan V. Dyslipidemia Part 1--Review of Lipid Metabolism and Vascular Cell Physiology. Vasc Endovascular Surg 2016; 50:107-18. [PMID: 26983667 DOI: 10.1177/1538574416628654] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Dyslipidemia, more specifically, high-serum low-density lipoproteins and low-serum high-density lipoproteins, are known risk factors for cardiovascular disease. The current clinical treatment of dyslipidemia represents the outcome of a large body of fundamental basic science research on lipids, lipid metabolism, and the effects of different lipids on cellular components of the artery, inflammatory cells, and platelets. In general, lower density lipids activate intracellular pathways to increase local and systemic inflammation, monocyte adhesion, endothelial cell dysfunction and apoptosis, and smooth muscle cell proliferation, resulting in foam cell formation and genesis of atherosclerotic plaque. In contrast, higher density lipids prevent or attenuate atherosclerosis. This article is part 1 of a 2-part review, with part 1 focusing on lipid metabolism and the downstream effects of lipids on the development of atherosclerosis, and part 2 on the clinical treatment of dyslipidemia and the role of these drugs for patients with arterial disease exclusive of the coronary arteries.
Collapse
Affiliation(s)
- Alex Helkin
- Department of Veterans Affairs Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jeffery J Stein
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Stacey Lin
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Sufyan Siddiqui
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Kristopher G Maier
- Department of Veterans Affairs Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Vivian Gahtan
- Department of Veterans Affairs Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
24
|
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
|
25
|
Schraufstatter IU, Khaldoyanidi SK, DiScipio RG. Complement activation in the context of stem cells and tissue repair. World J Stem Cells 2015; 7:1090-1108. [PMID: 26435769 PMCID: PMC4591784 DOI: 10.4252/wjsc.v7.i8.1090] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 07/27/2015] [Indexed: 02/06/2023] Open
Abstract
The complement pathway is best known for its role in immune surveillance and inflammation. However, its ability of opsonizing and removing not only pathogens, but also necrotic and apoptotic cells, is a phylogenetically ancient means of initiating tissue repair. The means and mechanisms of complement-mediated tissue repair are discussed in this review. There is increasing evidence that complement activation contributes to tissue repair at several levels. These range from the chemo-attraction of stem and progenitor cells to areas of complement activation, to increased survival of various cell types in the presence of split products of complement, and to the production of trophic factors by cells activated by the anaphylatoxins C3a and C5a. This repair aspect of complement biology has not found sufficient appreciation until recently. The following will examine this aspect of complement biology with an emphasis on the anaphylatoxins C3a and C5a.
Collapse
|
26
|
Deroyer C, Magne J, Moonen M, Le Goff C, Dupont L, Hulin A, Radermecker M, Colige A, Cavalier E, Kolh P, Pierard L, Lancellotti P, Merville MP, Fillet M. New biomarkers for primary mitral regurgitation. Clin Proteomics 2015; 12:25. [PMID: 26405438 PMCID: PMC4581160 DOI: 10.1186/s12014-015-9097-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/14/2015] [Indexed: 12/24/2022] Open
Abstract
Background Mitral regurgitation is a frequent valvular heart disease affecting around 2.5 % of the population with prevalence directly related to aging. Degeneration of mitral valve is broadly considered as a passive ongoing pathophysiological process and little is known about its physiological deregulation. The purpose of this study was to highlight new biomarkers of mitral regurgitation in order to decipher the underlying pathological mechanism as well as to allow the diagnosis and the monitoring of the disease. Results Modulation of various blood proteins expression was examined in patients suffering from different grades of mitral regurgitation (mild, moderate and severe) compared to healthy controls. To this end, several routine clinical assays and the multi analyte profile technology targeting 184 proteins were used. High-density lipoprotein, apolipoprotein-A1, haptoglobin and haptoglobin-α2 chain levels significantly decreased proportionally to the degree of mitral regurgitation when compared to controls. High-density lipoprotein and apolipoprotein-A1 levels were associated with effective regurgitant orifice area and regurgitant volume. Apolipoprotein-A1 was an independent predictor of severe mitral regurgitation. Moreover, with ordinal logistic regression, apolipoprotein-A1 remained the only independent factor associated with mitral regurgitation. In addition, myxomatous mitral valves were studied by immunocytochemistry. We observed an increase of LC3, the marker of autophagy, in myxomatous mitral valves compared with healthy mitral valves. Conclusion These potential biomarkers of mitral regurgitation highlighted different cellular processes that could be modified in myxomatous degenerescence: reverse cholesterol transport, antioxidant properties and autophagy. Electronic supplementary material The online version of this article (doi:10.1186/s12014-015-9097-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Céline Deroyer
- GIGA Proteomic Unit, Clinical Chemistry Laboratory, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Julien Magne
- GIGA Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Marie Moonen
- GIGA Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Caroline Le Goff
- Department of Clinical Chemistry, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Laura Dupont
- GIGA-Cancer, Laboratory of Connective Tissues Biology, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Alexia Hulin
- GIGA-Cancer, Laboratory of Connective Tissues Biology, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Marc Radermecker
- Department of Cardiovascular and Thoracic Surgery and Human Anatomy, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Alain Colige
- GIGA-Cancer, Laboratory of Connective Tissues Biology, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Philippe Kolh
- Department of Biomedical and Preclinical Sciences, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Luc Pierard
- GIGA Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Patrizio Lancellotti
- GIGA Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Marie-Paule Merville
- Department of Clinical Chemistry, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Marianne Fillet
- GIGA Proteomic Unit, Clinical Chemistry Laboratory, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium ; Laboratory for the Analysis of Medicines, CIRM, University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| |
Collapse
|
27
|
Shen H, Zhou E, Wei X, Fu Z, Niu C, Li Y, Pan B, Mathew AV, Wang X, Pennathur S, Zheng L, Wang Y. High density lipoprotein promotes proliferation of adipose-derived stem cells via S1P1 receptor and Akt, ERK1/2 signal pathways. Stem Cell Res Ther 2015; 6:95. [PMID: 25976318 PMCID: PMC4453044 DOI: 10.1186/s13287-015-0090-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 03/26/2015] [Accepted: 05/07/2015] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Adipose-derived stem cells (ADSC) are non-hematopoietic mesenchymal stem cells that have shown great promise in their ability to differentiate into multiple cell lineages. Their ubiquitous nature and the ease of harvesting have attracted the attention of many researchers, and they pose as an ideal candidate for applications in regenerative medicine. Several reports have demonstrated that transplanting ADSC can promote repair of injured tissue and angiogenesis in animal models. Survival of these cells after transplant remains a key limiting factor for the success of ADSC transplantation. Circulating factors like High Density Lipoprotein (HDL) has been known to promote survival of other stems cells like bone marrow derived stem cells and endothelial progenitor cells, both by proliferation and by inhibiting cell apoptosis. The effect of HDL on transplanted adipose-derived stem cells in vivo is largely unknown. METHODS This study focused on exploring the effects of plasma HDL on ADSC and delineating the mechanisms involved in their proliferation after entering the bloodstream. Using the MTT and BrdU assays, we tested the effects of HDL on ADSC proliferation. We probed the downstream intracellular Akt and ERK1/2 signaling pathways and expression of cyclin proteins in ADSC using western blot. RESULTS Our study found that HDL promotes proliferation of ADSC, by binding to sphingosine-1- phosphate receptor-1(S1P1) on the cell membrane. This interaction led to activation of intracellular Akt and ERK1/2 signaling pathways, resulting in increased expression of cyclin D1 and cyclin E, and simultaneous reduction in expression of cyclin-dependent kinase inhibitors p21 and p27, therefore promoting cell cycle progression and cell proliferation. CONCLUSIONS These studies raise the possibility that HDL may be a physiologic regulator of stem cells and increasing HDL concentrations may be valuable strategy to promote ADSC transplantation.
Collapse
Affiliation(s)
- Haitao Shen
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, 515041, China.
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Enchen Zhou
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Education Ministry, Peking University Health Science Center, Beijing, 100191, China.
| | - Xiujing Wei
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, 515041, China.
- Hutchison Medi Pharma (Suzhou), Jiangsu, 215125, China.
| | - Zhiwei Fu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Education Ministry, Peking University Health Science Center, Beijing, 100191, 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 Education Ministry, Peking University Health Science Center, Beijing, 100191, China.
| | - Yang Li
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Education Ministry, Peking University Health Science Center, Beijing, 100191, 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 Education Ministry, Peking University Health Science Center, Beijing, 100191, China.
| | - Anna V Mathew
- Department of Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Xu Wang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Education Ministry, Peking University Health Science Center, 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 Education Ministry, Peking University Health Science Center, Beijing, 100191, China.
| | - Yongyu Wang
- Institute of Hypoxia Medicine, Wenzhou Medical University, Zhejiang, 325035, China.
| |
Collapse
|
28
|
González-Pecchi V, Valdés S, Pons V, Honorato P, Martinez LO, Lamperti L, Aguayo C, Radojkovic C. Apolipoprotein A-I enhances proliferation of human endothelial progenitor cells and promotes angiogenesis through the cell surface ATP synthase. Microvasc Res 2015; 98:9-15. [DOI: 10.1016/j.mvr.2014.11.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/28/2014] [Accepted: 11/02/2014] [Indexed: 12/14/2022]
|
29
|
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
|
30
|
Tran-Dinh A, Diallo D, Delbosc S, Varela-Perez LM, Dang QB, Lapergue B, Burillo E, Michel JB, Levoye A, Martin-Ventura JL, Meilhac O. HDL and endothelial protection. Br J Pharmacol 2014; 169:493-511. [PMID: 23488589 DOI: 10.1111/bph.12174] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/07/2013] [Accepted: 02/24/2013] [Indexed: 12/23/2022] Open
Abstract
High-density lipoproteins (HDLs) represent a family of particles characterized by the presence of apolipoprotein A-I (apoA-I) and by their ability to transport cholesterol from peripheral tissues back to the liver. In addition to this function, HDLs display pleiotropic effects including antioxidant, anti-apoptotic, anti-inflammatory, anti-thrombotic or anti-proteolytic properties that account for their protective action on endothelial cells. Vasodilatation via production of nitric oxide is also a hallmark of HDL action on endothelial cells. Endothelial cells express receptors for apoA-I and HDLs that mediate intracellular signalling and potentially participate in the internalization of these particles. In this review, we will detail the different effects of HDLs on the endothelium in normal and pathological conditions with a particular focus on the potential use of HDL therapy to restore endothelial function and integrity.
Collapse
|
31
|
Chang FJ, Yuan HY, Hu XX, Ou ZJ, Fu L, Lin ZB, Wang ZP, Wang SM, Zhou L, Xu YQ, Wang CP, Xu Z, Zhang X, Zhang CX, Ou JS. High density lipoprotein from patients with valvular heart disease uncouples endothelial nitric oxide synthase. J Mol Cell Cardiol 2014; 74:209-19. [DOI: 10.1016/j.yjmcc.2014.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 05/04/2014] [Accepted: 05/21/2014] [Indexed: 11/29/2022]
|
32
|
Yin K, Agrawal DK. High-density lipoprotein: a novel target for antirestenosis therapy. Clin Transl Sci 2014; 7:500-11. [PMID: 25043950 DOI: 10.1111/cts.12186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Restenosis is an integral pathological process central to the recurrent vessel narrowing after interventional procedures. Although the mechanisms for restenosis are diverse in different pathological conditions, endothelial dysfunction, inflammation, vascular smooth muscle cell (SMC) proliferation, and myofibroblasts transition have been thought to play crucial role in the development of restenosis. Indeed, there is an inverse relationship between high-density lipoprotein (HDL) levels and risk for coronary heart disease (CHD). However, relatively studies on the direct assessment of HDL effect on restenosis are limited. In addition to involvement in the cholesterol reverse transport, many vascular protective effects of HDL, including protection of endothelium, antiinflammation, antithrombus actions, inhibition of SMC proliferation, and regulation by adventitial effects may contribute to the inhibition of restenosis, though the exact relationships between HDL and restenosis remain to be elucidated. This review summarizes the vascular protective effects of HDL, emphasizing the potential role of HDL in intimal hyperplasia and vascular remodeling, which may provide novel prophylactic and therapeutic strategies for antirestenosis.
Collapse
Affiliation(s)
- Kai Yin
- Center for Clinical & Translational Science, Creighton University School of Medicine, Omaha, Nebraska, USA
| | | |
Collapse
|
33
|
|
34
|
Rossi F, Bertone C, Michelon E, Bianco MJ, Santiemma V. High-density lipoprotein cholesterol affects early endothelial progenitor cell number and endothelial function in obese women. Obesity (Silver Spring) 2013; 21:2356-61. [PMID: 23512890 DOI: 10.1002/oby.20367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 12/21/2012] [Indexed: 11/10/2022]
Abstract
OBJECTIVE In order to improve our understanding of high-density lipoprotein cholesterol (HDL-C) cardiovascular (CV) impact in obesity, the association of HDL-C plasma level with circulating early endothelial progenitor cell (early-EPC) number and endothelium-dependent vasodilatation (EDV) in obese women with normal or high low-density lipoprotein cholesterol (LDL-C) plasma levels was evaluated. DESIGN AND METHODS One hundred thirteen obese female subjects and a control group of 78 healthy female subjects were recruited. Circulating early-EPC were assessed by single- and two-color flow cytometric analyses with a fluorescence activated cell sorting (FACScan) flow cytometer. EDV was evaluated as response to ischemia by strain gauge plethysmography. RESULTS Both early-EPC number and EDV were significantly decreased in obese women compared with the control group. Obese women with low HDL-C showed a further decrease of early-EPC and EDV in the presence of both high or normal LDL-C plasmatic levels. In the normal HDL-C level subgroup, hypercholesterolemic and nonhypercholesterolemic subjects showed no difference in early-EPC number, whereas slight EDV impairment was present in hypercholesterolemic subjects. CONCLUSION In obese women, low HDL-C is associated to decreased early-EPC number and impaired EDV, suggesting the need to assess whether evaluation of early-EPC and EDV may increase HDL-C prognostic value in the stratification of CV risk.
Collapse
Affiliation(s)
- F Rossi
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | | | | | | | | |
Collapse
|
35
|
Prosser HCG, Tan JTM, Dunn LL, Patel S, Vanags LZ, Bao S, Ng MKC, Bursill CA. Multifunctional regulation of angiogenesis by high-density lipoproteins. Cardiovasc Res 2013; 101:145-54. [PMID: 24130189 DOI: 10.1093/cvr/cvt234] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIMS High-density lipoproteins (HDL) exert striking anti-inflammatory effects and emerging evidence suggests that they may augment ischaemia-mediated neovascularization. We sought to determine whether HDL conditionally regulates angiogenesis, depending on the pathophysiological context by (i) inhibiting inflammation-induced angiogenesis, but also; (ii) enhancing ischaemia-mediated angiogenesis. METHODS AND RESULTS Intravenously delivered apolipoprotein (apo) A-I attenuated neovascularization in the murine femoral collar model of inflammation-induced angiogenesis, compared with phosphate-buffered saline infused C57BL6/J mice (58%), P < 0.05. Conversely, apoA-I delivery augmented neovessel formation (75%) and enhanced blood perfusion (45%) in the murine hindlimb ischaemia model, P < 0.05. Reconstituted HDL (rHDL) was tested on key angiogenic cell functions in vitro. rHDL inhibited human coronary artery endothelial cell migration (37.9 and 76.9%), proliferation (15.7 and 40.4%), and tubulogenesis on matrigel (52 and 98.7%) when exposed to two inflammatory stimuli: tumour necrosis factor-α (TNF-α) and macrophage-conditioned media (MCM). In contrast, rHDL significantly augmented hypoxia-stimulated migration (36.9%), proliferation (135%), and tubulogenesis (22.9%), P < 0.05. Western blot and RT-PCR analyses revealed that these divergent actions of rHDL were associated with conditional regulation of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and VEGF receptor 2, which were attenuated in response to TNF-α (40.4, 41.0, and 33.2%) and MCM (72.5, 30.7, and 69.5%), but augmented by rHDL in hypoxia (39.8, 152.6, and 15.7%%), all P < 0.05. CONCLUSION HDL differentially regulates angiogenesis dependent upon the pathophysiological setting, characterized by suppression of inflammation-associated angiogenesis, and conversely, by the enhancement of hypoxia-mediated angiogenesis. This has significant implications for therapeutic modulation of neovascularization.
Collapse
Affiliation(s)
- Hamish C G Prosser
- The Heart Research Institute, 7 Eliza Street, Newtown, Sydney, NSW 2042, Australia
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Tan JTM, Prosser HCG, Vanags LZ, Monger SA, Ng MKC, Bursill CA. High-density lipoproteins augment hypoxia-induced angiogenesis via regulation of post-translational modulation of hypoxia-inducible factor 1α. FASEB J 2013; 28:206-17. [PMID: 24022405 DOI: 10.1096/fj.13-233874] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Increasing evidence suggests that high-density lipoproteins (HDLs) promote hypoxia-induced angiogenesis. The hypoxia-inducible factor 1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway is important in hypoxia and is modulated post-translationally by prolyl hydroxylases (PHD1-PHD3) and E3 ubiquitin ligases (Siah1 and Siah2). We aimed to elucidate the mechanisms by which HDLs augment hypoxia-induced angiogenesis. Preincubation (16 h) of human coronary artery endothelial cells with reconstituted high-density lipoprotein (rHDL) containing apolipoprotein A-I (apoA-I) and phosphatidylcholine (20 μM, final apoA-I concentration), before hypoxia, increased Siah1 (58%) and Siah2 (88%) mRNA levels and suppressed PHD2 (32%) and PHD3 (45%) protein levels compared with hypoxia-induced control levels. After Siah1/2 small interfering RNA knockdown, rHDL was unable to suppress PHD2/3 and failed to induce HIF-1α, VEGF, and tubulogenesis in hypoxia. Inhibition of the upstream phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway also abrogated the effects of rHDL. Furthermore, knockdown of the scavenger receptor SR-BI attenuated rHDL-induced elevations in Siah1/2 and tubulogenesis in hypoxia, indicating that SR-BI plays a key role. Finally, the importance of VEGF in mediating the ability of rHDL to drive hypoxia-induced angiogenesis was confirmed using a VEGF-neutralizing antibody. In summary, rHDL augments the HIF-1α/VEGF pathway via SR-BI and modulation of the post-translational regulators of HIF-1α (PI3K/Siahs/PHDs). HDL-induced augmentation of angiogenesis in hypoxia may have implications for therapeutic modulation of ischemic injury.
Collapse
Affiliation(s)
- Joanne T M Tan
- 1Heart Research Institute, 7 Eliza Street, Newtown, NSW 2042, Australia.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Multiple human population studies have established the concentration of high density lipoprotein (HDL) cholesterol as an independent, inverse predictor of the risk of having a cardiovascular event. Furthermore, HDLs have several well-documented functions with the potential to protect against cardiovascular disease. These include an ability to promote the efflux of cholesterol from macrophages in the artery wall, inhibit the oxidative modification of low density lipoproteins (LDLs), inhibit vascular inflammation, inhibit thrombosis, promote endothelial repair, promote angiogenesis, enhance endothelial function, improve diabetic control, and inhibit hematopoietic stem cell proliferation. There are undoubtedly other beneficial functions of HDLs yet to be identified. The HDL fraction in human plasma is heterogeneous, consisting of several subpopulations of particles of varying size, density, and composition. The functions of the different HDL subpopulations remain largely unknown. Given that therapies that increase the concentration of HDL cholesterol have varying effects on the levels of specific HDL subpopulations, it is of great importance to understand how distribution of different HDL subpopulations contribute to the potentially cardioprotective functions of this lipoprotein fraction. This review summarizes current understanding of the relationship of HDL subpopulations to their cardioprotective properties and highlights the gaps in current knowledge regarding this important aspect of HDL biology.
Collapse
Affiliation(s)
- Kerry-Anne Rye
- Lipid Research Group, Centre for Vascular Research, University of New South Wales, Sydney, New South Wales, Australia 2052
| | | |
Collapse
|
38
|
Chiang EPI, Chiu SC, Pai MH, Wang YC, Wang FY, Kuo YH, Tang FY. Organosulfur garlic compounds induce neovasculogenesis in human endothelial progenitor cells through a modulation of MicroRNA 221 and the PI3-K/Akt signaling pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:4839-4849. [PMID: 23663050 DOI: 10.1021/jf304951p] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Human endothelial progenitor cells (EPCs) play crucial roles in the prevention of ischemic injury via neovasculogenesis. Frequent garlic consumption is reportedly associated with a low incidence of cardiovascular diseases (CVD). However, the molecular mechanisms by which garlic extracts, including diallyl disulfide (DADS) and diallyl trisulfide (DATS), exert an effect on neovasculogenesis have not been elucidated yet. The current study investigated the effects of these organosulfur compounds on neovasculogenesis by using vascular tube formation assay, Western blotting assay, real-time polymerase chain reaction (RT-PCR), and immunohistochemical (IHC) staining assays in both in vitro and in vivo models. The current study demonstrates that DADS and DATS dose-dependently enhance the neovasculogenesis of human EPCs in vitro. The mechanism of actions included the up-regulation of the c-kit protein, as well as the phosphorylation (i.e., activation) of the Akt and ERK 1/2 signaling molecules in human EPCs. Furthermore, DATS suppressed the expression of microRNA (miR) 221 in vitro. In a mouse xenograft model of neovasculogenesis, DATS consumption induced the formation of new blood vessels at a dosage of 10 mg/kg of body weight/day. It is suggested that garlic consumption enhances neovasculogenesis in human EPCs and thereby probably exerts a preventive effect against ischemic injuries.
Collapse
Affiliation(s)
- En-Pei Isabel Chiang
- Department of Food Science and Biotechnology and ‡Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
High density lipoprotein (HDL) cholesterol has direct effects on numerous cell types that influence cardiovascular and metabolic health. These include endothelial cells, vascular smooth-muscle cells, leukocytes, platelets, adipocytes, skeletal muscle myocytes, and pancreatic β cells. The effects of HDL or apoA-I, its major apolipoprotein, occur through the modulation of intracellular calcium, oxygen-derived free-radical production, numerous kinases, and enzymes, including endothelial nitric-oxide synthase (eNOS). ApoA-I and HDL also influence gene expression, particularly genes encoding mediators of inflammation in vascular cells. In many paradigms, the change in intracellular signaling occurs as a result of cholesterol efflux, with the cholesterol acceptor methyl-β-cyclodextrin often invoking responses identical to HDL or apoA-I. The ABC transporters ABCA1 and ABCG1 and scavenger receptor class B, type I (SR-BI) frequently participate in the cellular responses. Structure-function relationships are emerging for signal initiation by ABCA1 and SR-BI, with plasma membrane cholesterol binding by the C-terminal transmembrane domain of SR-BI uniquely enabling it to serve as a sensor of changes in membrane cholesterol. Further investigation of the processes underlying HDL and apoA-I modulation of intracellular signaling will potentially reveal new prophylactic and therapeutic strategies to optimize both cardiovascular and metabolic health.
Collapse
Affiliation(s)
- Chieko Mineo
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
40
|
Lin CP, Lin FY, Huang PH, Chen YL, Chen WC, Chen HY, Huang YC, Liao WL, Huang HC, Liu PL, Chen YH. Endothelial progenitor cell dysfunction in cardiovascular diseases: role of reactive oxygen species and inflammation. BIOMED RESEARCH INTERNATIONAL 2012; 2013:845037. [PMID: 23484163 PMCID: PMC3591199 DOI: 10.1155/2013/845037] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 11/13/2012] [Indexed: 12/31/2022]
Abstract
Endothelial progenitor cells (EPCs) move towards injured endothelium or inflamed tissues and incorporate into foci of neovascularisation, thereby improving blood flow and tissue repair. Patients with cardiovascular diseases have been shown to exhibit reduced EPC number and function. It has become increasingly apparent that these changes may be effected in response to enhanced oxidative stress, possibly as a result of systemic and localised inflammatory responses. The interplay between inflammation and oxidative stress affects the initiation, progression, and complications of cardiovascular diseases. Recent studies suggest that inflammation and oxidative stress modulate EPC bioactivity. Clinical medications with anti-inflammatory and antioxidant properties, such as statins, thiazolidinediones, angiotensin II receptor 1 blockers, and angiotensin-converting enzyme inhibitors, are currently administered to patients with cardiovascular diseases. These medications appear to exert beneficial effects on EPC biology. This review focuses on EPC biology and explores the links between oxidative stress, inflammation, and development of cardiovascular diseases.
Collapse
Affiliation(s)
- Chih-Pei Lin
- Department of Biotechnology and Laboratory Science in Medicine and Institute of Biotechnology in Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Pathology and Laboratory Medicine, Department of Internal Medicine and Divisions of Biochemistry and Cardiology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei 112, Taiwan
| | - Feng-Yen Lin
- Department of Internal Medicine, School of Medicine, Taipei Medical University and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Po-Hsun Huang
- Department of Pathology and Laboratory Medicine, Department of Internal Medicine and Divisions of Biochemistry and Cardiology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei 112, Taiwan
- Faculty of Medicine and Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Wen-Chi Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Huey-Yi Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Yu-Chuen Huang
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Huey-Chun Huang
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
41
|
Abstract
In addition to its role in reverse cholesterol transport, high-density lipoprotein (HDL) cholesterol has direct action on numerous cell types that influence cardiovascular and metabolic health. Cellular responses to HDL entail its capacity to invoke cholesterol efflux that causes signal initiation via scavenger receptor class B, type I, and plasma membrane receptor activation by HDL cargo molecules. In endothelial cells and their progenitors, HDL attenuates apoptosis and stimulates proliferation and migration. HDL also has diverse anti-inflammatory actions in both endothelial cells and leukocytes. In vascular smooth muscles, HDL tempers proinflammatory, promigratory, and degradative processes, and through actions on endothelium and platelets HDL is antithrombotic. There are additional actions of HDL of potential cardiovascular consequence that are indirect, including the capacities to promote pancreatic β-cell insulin secretion, to protect pancreatic β cells from apoptosis, and to enhance glucose uptake by skeletal muscle myocytes. Furthermore, HDL decreases white adipose tissue mass, increases energy expenditure, and promotes the production of adipose-derived cytokine adiponectin that has its own vascular-protective properties. Many of these numerous actions of HDL have been observed not only in cell culture and animal models but also in human studies, and assessments of these functions are now being applied to patient populations to better-elucidate which actions of HDL may contribute to its cardioprotective potential and how they can be quantified and targeted. Further work on the many mechanisms of HDL action promises to reveal new prophylactic and therapeutic strategies to optimize both cardiovascular and metabolic health.
Collapse
Affiliation(s)
- Chieko Mineo
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
42
|
Abstract
In addition to its role in reverse cholesterol transport, high-density lipoprotein (HDL) cholesterol has direct action on numerous cell types that influence cardiovascular and metabolic health. Cellular responses to HDL entail its capacity to invoke cholesterol efflux that causes signal initiation via scavenger receptor class B, type I, and plasma membrane receptor activation by HDL cargo molecules. In endothelial cells and their progenitors, HDL attenuates apoptosis and stimulates proliferation and migration. HDL also has diverse anti-inflammatory actions in both endothelial cells and leukocytes. In vascular smooth muscles, HDL tempers proinflammatory, promigratory, and degradative processes, and through actions on endothelium and platelets HDL is antithrombotic. There are additional actions of HDL of potential cardiovascular consequence that are indirect, including the capacities to promote pancreatic β-cell insulin secretion, to protect pancreatic β cells from apoptosis, and to enhance glucose uptake by skeletal muscle myocytes. Furthermore, HDL decreases white adipose tissue mass, increases energy expenditure, and promotes the production of adipose-derived cytokine adiponectin that has its own vascular-protective properties. Many of these numerous actions of HDL have been observed not only in cell culture and animal models but also in human studies, and assessments of these functions are now being applied to patient populations to better-elucidate which actions of HDL may contribute to its cardioprotective potential and how they can be quantified and targeted. Further work on the many mechanisms of HDL action promises to reveal new prophylactic and therapeutic strategies to optimize both cardiovascular and metabolic health.
Collapse
Affiliation(s)
- Chieko Mineo
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
43
|
Wang S, Peng D. Regulation of adipocyte autophagy--the potential anti-obesity mechanism of high density lipoprotein and ApolipoproteinA-I. Lipids Health Dis 2012; 11:131. [PMID: 23039759 PMCID: PMC3478219 DOI: 10.1186/1476-511x-11-131] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 09/30/2012] [Indexed: 01/01/2023] Open
Abstract
Obesity is reaching epidemic worldwide and is risk factor for cardiovascular disease and type 2 diabetes. Although plasma high density lipoprotein (HDL) and apolipoprotein A-I (apoA-I) are inversely correlated to obesity, whether HDLs have anti-obesity effect remains unclear until a recent study reporting the direct anti-obesity effect of apoA-I and its mimetic peptide. However, the mechanism is not fully understood. Increasing adipose energy expenditure through attainment of brown adipocyte phenotype in white adipose tissue is considered a potential strategy to combat obesity. Specific inhibition of autophagy in adipose tissue is associated with reduced adiposity which is attributed to the attainment of brown adipocyte phenotype in white adipose tissue and the increased energy expenditure. HDL and apoA-I could activate PI3K-Akt-mTORC1 signaling which negatively regulates autophagy. The links between HDL/apoA-I and autophagy brings a new understanding on the anti-obesity effect of HDL and apoA-I.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Cardiology, The Second Xiangya Hospital of Central South University, 139 Middle Ren-Min Rd, Changsha, Hunan 410011, China.
| | | |
Collapse
|
44
|
High density lipoprotein cholesterol promotes the proliferation of bone-derived mesenchymal stem cells via binding scavenger receptor-B type I and activation of PI3K/Akt, MAPK/ERK1/2 pathways. Mol Cell Biochem 2012; 371:55-64. [PMID: 22886428 DOI: 10.1007/s11010-012-1422-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 08/01/2012] [Indexed: 12/16/2022]
Abstract
High-density lipoprotein (HDL) possesses protective properties in cardiovascular diseases. However, the effect of HDL on the mesenchymal stem cells (MSCs), which could be mobilized to the damaged myocardial tissue, has not been well elucidated yet. In the current study, we investigated the effect of HDL on the proliferation of MSCs so as to reveal its molecular mechanisms. MSCs derived from rats were treated with HDL in different concentrations and for different periods. The proliferation of MSCs was measured with MTT and BrdU cell proliferation assay. The phosphorylation of Akt, ERK1/2 and the expression of p21 were evaluated by Western blotting. After the activity of respective pathways was down-regulated by the specific inhibitor and the gene of scavenger receptor-B type I (SR-BI) was knocked down by RNA interference, BrdU assay was performed to examine this effect of HDL on MSCs. We found that the proliferation of MSCs induced by HDL, in a time- and concentration-dependent manner, was the phosphorylation of Akt- and ERK1/2-dependent, which was significantly attenuated by the specific inhibitor to respective pathways. Moreover, MAPK/ERK1/2 pathway exerted a more dominating effect on this process. SR-BI contributed to HDL-induced proliferation of MSCs, which was effectively abolished by the silencing of SR-BI. The results suggested that HDL was capable of improving MSCs proliferation, in which MAPK/ERK1/2 and PI3K/Akt pathways involved and SR-BI played a critical role as well.
Collapse
|
45
|
|
46
|
Farmer JA, Liao J. Evolving concepts of the role of high-density lipoprotein in protection from atherosclerosis. Curr Atheroscler Rep 2011; 13:107-14. [PMID: 21380938 DOI: 10.1007/s11883-011-0166-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
High-density lipoprotein (HDL) is classified as a negative risk factor due to the inverse relationship between elevated levels of HDL cholesterol and atherosclerosis. The mechanism by which HDL can mediate protection from atherosclerosis is complex and multifactorial. The primary role of reverse cholesterol transport in the reduction of risk for coronary artery disease is supported by a considerable amount of experimental data. HDL is able to interact with and remove cholesterol from the lipid-laden foam cells in the peripheral vasculature with subsequent transportation to the liver for excretion. However, HDL has multiple other physiologic effects that may play a significant role in protection from atherosclerosis. HDL has been demonstrated to exhibit multiple beneficial effects on the coagulation system. Platelet function is improved by both direct and indirect mechanisms. HDL has a complex interaction with the protein C and protein S system. Thrombolytic balance is also improved by HDL. HDL has been demonstrated to have a significant natural antioxidant effect that inhibits the oxidative step required for low-density lipoprotein uptake by the macrophage. Additionally, HDL has also been demonstrated to exert multiple beneficial effects on endothelial function, including decreased apoptosis and endothelial repair.
Collapse
Affiliation(s)
- John A Farmer
- Baylor College of Medicine, Baylor Clinic, Houston, TX, 77030, USA.
| | | |
Collapse
|
47
|
Current world literature. Curr Opin Lipidol 2011; 22:231-6. [PMID: 21562387 DOI: 10.1097/mol.0b013e328347aeca] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Abstract
PURPOSE OF REVIEW Our purpose is to review recent findings highlighting the metabolic and functional diversity of HDL subspecies. RECENT FINDINGS HDL heterogeneity - both structural and functional - is the main focus of this review. Recent work indicates that the metabolism and functionality of HDL particles differ greatly among HDL subspecies. With the introduction of new and improved methodology (e.g., proteomics), new aspects of the structural complexity and functionality of HDL have been revealed. It has been confirmed that HDL functions - including, but not limited to decreasing inflammation, apoptosis, macrophage adhesion to the endothelium and insulin resistance - are due to HDL's ability to remove cholesterol from cells (reverse cholesterol transport). A new level of HDL complexity has recently been revealed by investigating the lipid composition of HDL with gas chromatography, gas chromatography-mass spectrometry and liquid chromatography-mass spectrometry. There are about 100 different HDL-associated proteins; however, there are many more lipid species potentially associated with HDL particles. SUMMARY The most important recent findings disclose that HDL is more complex than previously thought. HDL subclasses differ in physical-chemical properties, protein and lipid composition, metabolism, physiological functions and pathophysiological significance. The staggering complexity of HDL demands significantly more investigation before we can truly begin to understand HDL metabolism and function in humans.
Collapse
Affiliation(s)
- Bela F Asztalos
- Lipid Metabolism Laboratory, Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA.
| | | | | |
Collapse
|