1
|
He RR, Ma CR, He X, Dong YX, Li H, Chu ZX, Yang XH, Wang JQ, Wang T, Wang FQ, Du FF, Rao Y, Yu WX, Gao XM, Fan GW, Cheng C, Li C. Circulating metabolites of Borneolum syntheticum (Bingpian) ameliorate atherosclerosis in ApoE -/- mice via inhibiting macrophage foam-cell formation. Acta Pharmacol Sin 2025; 46:759-776. [PMID: 39472494 PMCID: PMC11845446 DOI: 10.1038/s41401-024-01406-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/01/2024] [Indexed: 02/23/2025]
Abstract
Translational pharmacological research on traditional medicines lays the foundation for precisely understanding how the medicines function in the body to deliver therapeutic benefits. Borneolum syntheticum (Bingpian) is commonly used in Chinese herbal medicines for coronary heart disease, but its specific cardiovascular impact remains poorly understood. Isoborneol, a constituent of Bingpian, has been found to reduce lipid accumulation in macrophages in vitro, but its oral bioavailability is limited. This investigation aimed to evaluate anti-atherosclerotic effects of Bingpian, based on understanding its first-pass metabolism. Human subjects orally received an herbal medicine containing Bingpian and their plasma samples were analyzed to identify the major circulating compounds of Bingpian, with the metabolism that was also characterized in vitro and in mice. The identified compounds were evaluated for their ability to inhibit macrophage foam-cell formation induced by oxidized low-density lipoprotein. Furthermore, the anti-atherosclerotic effect of repeatedly dosed Bingpian was assessed in ApoE-/- mice fed a high-fat diet. In human subjects, the major circulating compounds of Bingpian were metabolites, rather than their precursor constituents borneol and isoborneol. These constituents were efficiently absorbed in the intestinal tract but underwent significant first-pass metabolism, involving UGT2B7-mediated glucuronidation into borneol-2-O-glucuronide and isoborneol-2-O-glucuronide, respectively, and CYP2A6/2B6/3A-mediated oxidation both into camphor. Despite their poor membrane permeability, hepatic efflux of borneol-2-O-glucuronide and isoborneol-2-O-glucuronide into the systemic circulation was enhanced by MRP3/4. The circulating metabolites, particularly their combinations, markedly inhibited macrophage foam-cell formation induced by oxidized low-density lipoprotein in vitro. Sub-chronic administration of Bingpian (30 mg·kg-1·d-1, i.g.) for 12 weeks significantly decreased atherosclerotic lesion size and enhanced plaque stability in ApoE-/- mice. Systemic exposure to Bingpian metabolites in mice closely resembles that in humans, suggesting that the pharmacodynamic effects of Bingpian in mice are likely applicable to humans. Overall, the cardiovascular benefits of Bingpian involve reducing atherosclerosis by inhibiting foam-cell formation through its metabolites. This investigation supports that oral Bingpian could be a druggable agent for reducing atherosclerosis.
Collapse
Affiliation(s)
- Rong-Rong He
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chuan-Rui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300073, China
| | - Xin He
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yan-Xi Dong
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hui Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zi-Xuan Chu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xi-He Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jia-Qi Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ting Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Shanghai, 201203, China
| | - Feng-Qing Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Fei-Fei Du
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ying Rao
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
| | - Wen-Xuan Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiu-Mei Gao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Guan-Wei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300073, China.
| | - Chen Cheng
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Chuan Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China.
| |
Collapse
|
2
|
Mester P, Birner C, Schmid S, Müller M, Pavel V, Buechler C. Elevated plasma soluble lectin-like oxidised low-density lipoprotein receptor 1 as an independent prognostic biomarker in sepsis. Lipids Health Dis 2025; 24:47. [PMID: 39948564 PMCID: PMC11823166 DOI: 10.1186/s12944-025-02462-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/02/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Soluble lectin-like oxidised low-density lipoprotein receptor 1 (sLOX-1) is overproduced during inflammation, with its expression and release triggered by C-reactive protein (CRP). As CRP levels are typically elevated in sepsis, this study aimed to investigate whether sLOX-1 levels increase in parallel. METHODS Plasma sLOX-1 levels of 52 patients with systemic inflammatory response syndrome (SIRS), 45 patients with sepsis, 88 patients with septic shock and 37 controls were measured by ELISA. Associations with CRP, underlying diseases, severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and bacterial infections were analysed. RESULTS Plasma sLOX-1 levels were similarly elevated in patients with SIRS, sepsis, or septic shock compared to controls. Plasma sLOX-1 levels did not differ between male and female controls or patients. Plasma sLOX-1 levels were comparable in patients infected with SARS-CoV-2, Gram-negative bacteria, or Gram-positive bacteria. No association was observed between sLOX-1 levels and underlying liver cirrhosis or pancreatitis. Notably, plasma sLOX-1 levels correlated positively with leukocyte and basophil counts but showed no correlation with CRP or procalcitonin. Of clinical relevance, positive correlations were also found with aspartate aminotransferase (AST) and bilirubin levels. Among the 41 patients who did not survive, sLOX-1, AST, and bilirubin levels were significantly higher compared to those of survivors. CONCLUSIONS Plasma levels of sLOX-1 are elevated in patients with SIRS or sepsis and are significantly higher in non-survivors. Of note, they do not correlate with classical inflammatory markers, suggesting that sLOX-1 may function as an independent prognostic biomarker for predicting poor outcomes in patients with SIRS or sepsis.
Collapse
Affiliation(s)
- Patricia Mester
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Charlotte Birner
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Stephan Schmid
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Martina Müller
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Vlad Pavel
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
3
|
Malamud M, Brown GD. The Dectin-1 and Dectin-2 clusters: C-type lectin receptors with fundamental roles in immunity. EMBO Rep 2024; 25:5239-5264. [PMID: 39482490 PMCID: PMC11624271 DOI: 10.1038/s44319-024-00296-2] [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: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024] Open
Abstract
The ability of myeloid cells to recognize and differentiate endogenous or exogenous ligands rely on the presence of different transmembrane protein receptors. C-type lectin receptors (CLRs), defined by the presence of a conserved structural motif called C-type lectin-like domain (CTLD), are a crucial family of receptors involved in this process, being able to recognize a diverse range of ligands from glycans to proteins or lipids and capable of initiating an immune response. The Dectin-1 and Dectin-2 clusters involve two groups of CLRs, with genes genomically linked within the natural killer cluster of genes in both humans and mice, and all characterized by the presence of a single extracellular CTLD. Fundamental immune cell functions such as antimicrobial effector mechanisms as well as internalization and presentation of antigens are induced and/or regulated through activatory, or inhibitory signalling pathways triggered by these receptors after ligand binding. In this review, we will discuss the most recent concepts regarding expression, ligands, signaling pathways and functions of each member of the Dectin clusters of CLRs, highlighting the importance and diversity of their functions.
Collapse
Affiliation(s)
- Mariano Malamud
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Gordon D Brown
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| |
Collapse
|
4
|
Giacaglia MB, Pires V, Santana MFM, Passarelli M. Unraveling the Pleiotropic Role of High-Density Lipoproteins (HDLs) in Autoimmune Rheumatic Diseases. Int J Rheumatol 2024; 2024:1896817. [PMID: 39574464 PMCID: PMC11581784 DOI: 10.1155/2024/1896817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Autoimmune rheumatic diseases (ARDs) exhibit an elevated incidence of cardiovascular disease (CVD). The elevation of inflammatory and immune stress accompanying ARDs contributes to atherosclerosis development and alterations in lipid metabolism and lipoprotein profile add to cardiovascular (CV) risk. The plasma concentration of high-density lipoprotein cholesterol (HDLc) is inversely related to CVD and serves as a discriminator of CV risk. However, this association is not unequivocal, and changes in HDL functionality appear to emerge as a better indicator of CV risk, albeit difficult to measure and monitor clinically. The modulation of HDLc itself can bring benefits in controlling autoimmunity and reducing ARD activity. Understanding HDL function and each peculiarity involved in ARDs enables to seek means to prevent ischemic outcomes associated with CVD, in the face of the residual CV risk persisting even with controlled disease activity and classic risk factors. By comprehending HDL's structural and functional nuances, it will be possible to develop more effective strategies to manage the evolution and outcomes of ARDs. It is also necessary to standardize diagnostic methods and establish different markers for each specific disease allowing the design of intervention strategies to restore HDL functionality, reduce residual CV, and prevent, alleviate, or even suppress ARD activity.
Collapse
Affiliation(s)
- Marcia B. Giacaglia
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE) 01525-000, São Paulo, Brazil
| | - Vitória Pires
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| | - Monique F. M. Santana
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| | - Marisa Passarelli
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE) 01525-000, São Paulo, Brazil
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| |
Collapse
|
5
|
Akkuş M, Solak H. Elevated levels of oxLDL and LOX-1: Implications for schizophrenia pathophysiology. J Psychiatr Res 2024; 177:140-146. [PMID: 39013288 DOI: 10.1016/j.jpsychires.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/21/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
Inflammation and oxidative stress are both considered to be factors in the etiopathogenesis of schizophrenia. LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1) and ox-LDL (oxidized low-density lipoprotein) have been reported to be active in neuroinflammation pathways in which they are involved in oxidative stress and inflammation. However, its relationship with schizophrenia is unclear. This study aimed to assess the potential connection between serum ox-LDL and LOX-1 levels in schizophrenia patients, their unaffected first-degree relatives, and healthy controls. The study comprised 63 schizophrenia patients, 57 first-degree relatives, and 63 healthy controls who were age, gender, and BMI-matched. Serum ox-LDL and LOX-1 levels were measured. PANSS was used to assess the severity of the disease. Levels of both ox-LDL and LOX-1 were markedly elevated in individuals diagnosed with schizophrenia when compared to both their relatives and a control group. While ox-LDL levels were significantly higher in relatives of patients compared to controls, there was no significant difference between relatives of patients and control groups for LOX-1 levels. Significant correlations were observed between PANNS general and total and ox-LDL levels and PANNS negative and LOX-1 levels. The relationship between ox-LDL and LOX-1 and schizophrenia is quite limited in the literature and is a new field of study. Future studies are needed to evaluate their role in etiopathogenesis.
Collapse
Affiliation(s)
- Merve Akkuş
- Department of Psychiatry, Kütahya Health Sciences University, Faculty of Medicine, Vefa Alayunt Street, 43100, Kutahya Province, Kütahya, Turkey.
| | - Hatice Solak
- Department of Physiology, Faculty of Medicine, Kütahya Health Science University, Evliya Çelebi Campus, Tavşanlı Road 10th Km, 43100, Kutahya Province, Kütahya, Turkey.
| |
Collapse
|
6
|
Munno M, Mallia A, Greco A, Modafferi G, Banfi C, Eligini S. Radical Oxygen Species, Oxidized Low-Density Lipoproteins, and Lectin-like Oxidized Low-Density Lipoprotein Receptor 1: A Vicious Circle in Atherosclerotic Process. Antioxidants (Basel) 2024; 13:583. [PMID: 38790688 PMCID: PMC11118168 DOI: 10.3390/antiox13050583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Atherosclerosis is a complex condition that involves the accumulation of lipids and subsequent plaque formation in the arterial intima. There are various stimuli, cellular receptors, and pathways involved in this process, but oxidative modifications of low-density lipoprotein (ox-LDL) are particularly important in the onset and progression of atherosclerosis. Ox-LDLs promote foam-cell formation, activate proinflammatory pathways, and induce smooth-muscle-cell migration, apoptosis, and cell death. One of the major receptors for ox-LDL is LOX-1, which is upregulated in several cardiovascular diseases, including atherosclerosis. LOX-1 activation in endothelial cells promotes endothelial dysfunction and induces pro-atherogenic signaling, leading to plaque formation. The binding of ox-LDLs to LOX-1 increases the generation of reactive oxygen species (ROS), which can induce LOX-1 expression and oxidize LDLs, contributing to ox-LDL generation and further upregulating LOX-1 expression. This creates a vicious circle that is amplified in pathological conditions characterized by high plasma levels of LDLs. Although LOX-1 has harmful effects, the clinical significance of inhibiting this protein remains unclear. Further studies both in vitro and in vivo are needed to determine whether LOX-1 inhibition could be a potential therapeutic target to counteract the atherosclerotic process.
Collapse
Affiliation(s)
- Marco Munno
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Alice Mallia
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
- Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Università di Pavia, 27100 Pavia, Italy
| | - Arianna Greco
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Gloria Modafferi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Sonia Eligini
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| |
Collapse
|
7
|
Pyrpyris N, Dimitriadis K, Beneki E, Iliakis P, Soulaidopoulos S, Tsioufis P, Adamopoulou E, Kasiakogias A, Sakalidis A, Koutsopoulos G, Aggeli K, Tsioufis K. LOX-1 Receptor: A Diagnostic Tool and Therapeutic Target in Atherogenesis. Curr Probl Cardiol 2024; 49:102117. [PMID: 37802161 DOI: 10.1016/j.cpcardiol.2023.102117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/08/2023]
Abstract
Low-density lipoprotein (LDL) and oxidized LDL (oxLDL) are major contributors to atherogenesis, as endogenous antigens, via several receptors such as LOX 1. A PubMed search was conducted in order to identify relevant articles regarding LOX-1's role in the atherosclerosis, diagnosis, prognostic use and molecules that could be used for therapy. The references of the manuscripts obtained were also reviewed, in order to find additional relevant bibliography. LOX-1 is a lectin-like pattern recognition receptor, mostly expressed in endothelial cells (ECs) which can bind a variety of molecules, including oxLDL and C-reactive protein (CRP). LOX-1 plays a key role in oxLDL's role as a causative agent of atherosclerosis through several pathologic mechanisms, such as oxLDL deposition in the subintima, foam cell formation and endothelial dysfunction. Additionally, LOX-1 acts a scavenger receptor for oxLDL in macrophages and can be responsible for oxLDL uptake, when stimulated. Serum LOX-1 (sLOX-1) has emerged as a new, potential biomarker for diagnosis of acute coronary syndromes, and it seems promising for use along with other common biomarkers in everyday clinical practice. In a therapeutic perspective, natural as well as synthetic molecules exert anti-LOX-1 properties and attain the receptor's pathophysiological effects, thus extensive research is ongoing to further evaluate molecules with therapeutic potential. However, most of these molecules need further trials in order to properly assess their safety and efficacy for clinical use. The aim of this review is to investigate LOX-1 role in atherogenesis and explore its potential as diagnostic tool and therapeutic target.
Collapse
Affiliation(s)
- Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece.
| | - Eirini Beneki
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Panagiotis Iliakis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Stergios Soulaidopoulos
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Panagiotis Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Elena Adamopoulou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Alexandros Kasiakogias
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Athanasios Sakalidis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - George Koutsopoulos
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Konstantina Aggeli
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| |
Collapse
|
8
|
Villalobos-Labra R, Liu R, Spaans F, Sáez T, Semeria Maitret T, Quon A, Sawamura T, Cooke CLM, Davidge ST. Placenta-Derived Extracellular Vesicles From Preeclamptic Pregnancies Impair Vascular Endothelial Function via Lectin-Like Oxidized LDL Receptor-1. Hypertension 2023; 80:2226-2238. [PMID: 37615097 DOI: 10.1161/hypertensionaha.123.21205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Preeclampsia is a complex syndrome that includes maternal vascular dysfunction. Syncytiotrophoblast-derived extracellular vesicles from preeclampsia placentas (preeclampsia-STBEVs) were shown to induce endothelial dysfunction, but an endothelial transmembrane mediator is still unexplored. The LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1) is a transmembrane scavenger receptor that can cause endothelial dysfunction, and its expression is increased in the endothelium of preeclampsia women. In this study, we hypothesized that LOX-1 mediates the effects of preeclampsia-STBEVs on endothelial function. METHODS Preeclampsia-STBEVs were collected by perfusion of placentas from women with preeclampsia and in vitro and ex vivo endothelial cell function were assessed. RESULTS In human umbilical vein endothelial cells, inhibition of LOX-1 with LOX-1 blocking antibody (TS20) reduced the uptake of preeclampsia-STBEVs (61.3±8.8%). TS20 prevented the activation of ERK (extracellular signal-regulated kinase, a kinase downstream of LOX-1) and reduced the activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells; 21.1±8.0%) and nitrative stress (23.2±10.3%) that was induced by preeclampsia-STBEVs. Vascular function was assessed by wire myography in isolated mesenteric arteries from pregnant rats that were incubated overnight with preeclampsia-STBEVs±TS20. TS20 prevented endothelium-dependent vasodilation impairment induced by preeclampsia-STBEVs. Nitric oxide contribution to the relaxation was reduced by preeclampsia-STBEVs, which was prevented by TS20. Superoxide dismutase or apocynin, an inhibitor of NOX (nicotinamide adenine dinucleotide phosphate oxidase), restored the impaired endothelium-dependent vasodilation in arteries exposed to preeclampsia-STBEVs. CONCLUSIONS Taken together, our findings demonstrate that LOX-1 mediates the endothelial dysfunction induced by preeclampsia-STBEVs. Our study further expands on the mechanisms that may lead to adverse outcomes in preeclampsia and proposes LOX-1 as a potential target for future interventions.
Collapse
Affiliation(s)
- Roberto Villalobos-Labra
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Escuela de Medicina sede San Felipe (R.V.-L.), Universidad de Valparaíso, Chile
| | - Ricky Liu
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tamara Sáez
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Departamento de Medicina Interna (T. Sáez), Universidad de Valparaíso, Chile
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina (T. Sáez), Universidad de Valparaíso, Chile
| | - Tamara Semeria Maitret
- Department of Laboratory Medicine and Pathology (T.S.M.), University of Alberta, Edmonton, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tatsuya Sawamura
- Departments of Molecular Pathophysiology and Life Innovation, Shinshu University, Matsumoto, Japan (T. Sawamura)
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| |
Collapse
|
9
|
Puddu A, Montecucco F, Maggi D. Caveolin-1 and Atherosclerosis: Regulation of LDLs Fate in Endothelial Cells. Int J Mol Sci 2023; 24:8869. [PMID: 37240214 PMCID: PMC10219015 DOI: 10.3390/ijms24108869] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Caveolae are 50-100 nm cell surface plasma membrane invaginations observed in terminally differentiated cells. They are characterized by the presence of the protein marker caveolin-1. Caveolae and caveolin-1 are involved in regulating several signal transduction pathways and processes. It is well recognized that they have a central role as regulators of atherosclerosis. Caveolin-1 and caveolae are present in most of the cells involved in the development of atherosclerosis, including endothelial cells, macrophages, and smooth muscle cells, with evidence of either pro- or anti-atherogenic functions depending on the cell type examined. Here, we focused on the role of caveolin-1 in the regulation of the LDLs' fate in endothelial cells.
Collapse
Affiliation(s)
- Alessandra Puddu
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy; (F.M.); (D.M.)
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy; (F.M.); (D.M.)
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Davide Maggi
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy; (F.M.); (D.M.)
| |
Collapse
|
10
|
Dillon GA, Stanhewicz AE, Serviente C, Flores VA, Stachenfeld N, Alexander LM. Seven days of statin treatment improves nitric-oxide mediated endothelial-dependent cutaneous microvascular function in women with endometriosis. Microvasc Res 2022; 144:104421. [PMID: 35970408 PMCID: PMC9527706 DOI: 10.1016/j.mvr.2022.104421] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 10/31/2022]
Abstract
INTRODUCTION Endometriosis is associated with systemic inflammation and increased risk of cardiovascular disease (CVD). Endothelial dysfunction is one of the first manifestations of CVD but is unexplored in women with endometriosis. HMG-CoA-reductase inhibitors (statins) exert potent anti-inflammatory effects, and have been proposed as an adjunctive therapy in women with endometriosis. We hypothesized that microvascular endothelial function would be impaired in otherwise healthy women with endometriosis mediated by reduced nitric oxide (NO)-dependent dilation and that short term statin administration would improve endothelial function. METHODS In 8 healthy control (HC: 33 ± 9 yr) and 8 women with endometriosis (EN: 34 ± 9 yr), laser-Doppler flux (LDF) was measured continuously during graded intradermal microdialysis perfusion of the endothelium-dependent agonist acetylcholine (Ach: 10-10-10-1 M) alone and in combination with the NO synthase inhibitor (L-NAME: 0.015 M). 6 EN repeated the microdialysis experiment following 7 days of oral atorvastatin treatment (10 mg). Cutaneous vascular conductance was calculated (CVC = LDF*mmHg-1) and normalized to site-specific maximum (28 mM sodium nitroprusside, 43 °C). The NO-dependent dilation was calculated as the difference between the areas under the dose response curves. RESULTS Ach-induced vasodilation was blunted in women with endometriosis (main effect p < 0.01), indicating impaired endothelial function. NO-dependent vasodilation was also reduced in women with endometriosis (HC: 217 ± 120.3 AUC vs. EN: 88 ± 97 AUC, p = 0.03). Oral atorvastatin improved Ach-induced (main effect p < 0.01) and NO-dependent (295 ± 153 AUC; p = 0.05) vasodilation in women with endometriosis. CONCLUSION Microcirculatory endothelium-dependent vasodilation is impaired in women with endometriosis, mediated in part by reductions in NO. Short-term oral atorvastatin improved endothelium-dependent vasodilation, suggesting that statin therapy may be a viable intervention strategy to mitigate accelerated CVD risk in women with endometriosis.
Collapse
Affiliation(s)
- Gabrielle A Dillon
- Noll Laboratory, Department of Kinesiology, The Pennsylvania State University, University Park, PA, United States of America; Center for Healthy Aging, The Pennsylvania State University, University Park, PA, United States of America
| | - Anna E Stanhewicz
- Noll Laboratory, Department of Kinesiology, The Pennsylvania State University, University Park, PA, United States of America; Department of Health and Human Physiology, The University of Iowa, Iowa City, IA, United States of America
| | - Corinna Serviente
- Noll Laboratory, Department of Kinesiology, The Pennsylvania State University, University Park, PA, United States of America; Center for Healthy Aging, The Pennsylvania State University, University Park, PA, United States of America; Department of Kinesiology, University of Massachusetts Amherst, MA, United States of America; Institute for Applied Life Sciences, University of Massachusetts Amherst, MA, United States of America
| | - Valerie A Flores
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States of America
| | - Nina Stachenfeld
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States of America; John B. Pierce Laboratory, Yale University, New Haven, CT, United States of America
| | - Lacy M Alexander
- Noll Laboratory, Department of Kinesiology, The Pennsylvania State University, University Park, PA, United States of America; Center for Healthy Aging, The Pennsylvania State University, University Park, PA, United States of America.
| |
Collapse
|
11
|
Tian Y, Chang S, Xu J, Gong P, Yu B, Qi J. Investigation of the effective components inhibited macrophage foam cell formation in Ophiopogonis Radix. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114678. [PMID: 34563614 DOI: 10.1016/j.jep.2021.114678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/12/2021] [Accepted: 09/22/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ophiopogonis Radix, the commonly used traditional Chinese medicine in clinic for treating cardiovascular diseases, is returned to the stomach, lung and heart meridian. It is reported to nourish yin, moisten lung and is used to treat heart yin deficiency syndromes and asthenia of heart and lung, which indicated that Ophiopogonis Radix may have a protective effect on heart disorders. Atherosclerosisis is an important process in the development of cardiovascular diseases and abnormal lipid deposition induced macrophage foam cells is its crucial foundation. Our previous study showed the extract of Ophiopogonis Radix (EOR) ameliorates atherosclerosis in vitro. However, it may protect against cardiovascular diseases through inhibiting macrophage foam cell formation and its potential effective components and mechanisms are still unclear. AIM OF THE STUDY Our study aimed to investigate the effect of Ophiopogonis Radix on macrophage foam cell formation and its potential active constituents and mechanisms. MATERIALS AND METHODS Ox-LDL induced macrophage cells were employed to evaluate the effect of Ophiopogonis Radix on macrophage foam cell formation. Then the potential active constituents inhibited formation of macrophage foam cells were screened by biospecific cell extraction and its underlying mechanisms were also explored by Western blot. RESULTS The extract of Ophiopogonis Radix was found to significantly inhibit macrophage foam cell formation, evidenced by the decrease of TG and TC and Oil Red O staining analysis in macrophage cells, which indicated that EOR reduced the formation of macrophage foam cells. At the same time, EOR was showed to increase antioxidant capacity in macrophage cells. After treatment with EOR, two potential active components interacted with macrophage foam cells specifically were identified to inhibit macrophage foam cell formation including methylophiopogonanone A and methylophiopogonanone B. Methylophiopogonanone A was then proved to decrease the expression of CD36, Lox-1 and SREBP2, increase the expression of ABCA1 obviously, while the expression of ABCG1 and SREBP1 had no changes. CONCLUSIONS In our study, Ophiopogonis Radix was found to protect against atherosclerosis through suppressing ox-LDL induced macrophage foam cell formation and two potential compounds were identified by biospecific cell extraction including methylophiopogonanone A and methylophiopogonanone B. Moreover, methylophiopogonanone A was proved to inhibit foam cells through reducing uptake, synthesis and increasing efflux, which may provide guidance and reference for application of Ophiopogonis Radix and investigation of the effective components of TCMs.
Collapse
Affiliation(s)
- YuShan Tian
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Shanquan Chang
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Juntao Xu
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Puyang Gong
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - BoYang Yu
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| | - Jin Qi
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| |
Collapse
|
12
|
Theodoros K, Sharma M, Anton P, Hugo C, Ellen O, Hultgren NW, Ritou E, Williams DS, Orian S S, Srinivasa T R. The ApoA-I mimetic peptide 4F attenuates in vitro replication of SARS-CoV-2, associated apoptosis, oxidative stress and inflammation in epithelial cells. Virulence 2021; 12:2214-2227. [PMID: 34494942 PMCID: PMC8437485 DOI: 10.1080/21505594.2021.1964329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/12/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
An oral antiviral against SARS-CoV-2 that also attenuates inflammatory instigators of severe COVID-19 is not available to date. Herein, we show that the apoA-I mimetic peptide 4 F inhibits Spike mediated viral entry and has antiviral activity against SARS-CoV-2 in human lung epithelial Calu3 and Vero-E6 cells. In SARS-CoV-2 infected Calu3 cells, 4 F upregulated inducers of the interferon pathway such as MX-1 and Heme oxygenase 1 (HO-1) and downregulated mitochondrial reactive oxygen species (mito-ROS) and CD147, a host protein that mediates viral entry. 4 F also reduced associated cellular apoptosis and secretion of IL-6 in both SARS-CoV-2 infected Vero-E6 and Calu3 cells. Thus, 4 F attenuates in vitro SARS-CoV-2 replication, associated apoptosis in epithelial cells and secretion of IL-6, a major cytokine related to COVID-19 morbidity. Given established safety of 4 F in humans, clinical studies are warranted to establish 4 F as therapy for COVID-19.
Collapse
Affiliation(s)
- Kelesidis Theodoros
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Madhav Sharma
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Petcherski Anton
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Cristelle Hugo
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - O’Connor Ellen
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, United States
| | - Nan W Hultgren
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Eleni Ritou
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - David S Williams
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Shirihai Orian S
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Reddy Srinivasa T
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, United States
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
13
|
Kaushik P, Ahlawat P, Singh K, Singh R. Chemical constituents, pharmacological activities, and uses of common ayurvedic medicinal plants: a future source of new drugs. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-021-00621-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
14
|
Murdocca M, De Masi C, Pucci S, Mango R, Novelli G, Di Natale C, Sangiuolo F. LOX-1 and cancer: an indissoluble liaison. Cancer Gene Ther 2021; 28:1088-1098. [PMID: 33402733 PMCID: PMC8571092 DOI: 10.1038/s41417-020-00279-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/19/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023]
Abstract
Recently, a strong correlation between metabolic disorders, tumor onset, and progression has been demonstrated, directing new therapeutic strategies on metabolic targets. OLR1 gene encodes the LOX-1 receptor protein, responsible for the recognition, binding, and internalization of ox-LDL. In the past, several studied, aimed to clarify the role of LOX-1 receptor in atherosclerosis, shed light on its role in the stimulation of the expression of adhesion molecules, pro-inflammatory signaling pathways, and pro-angiogenic proteins, including NF-kB and VEGF, in vascular endothelial cells and macrophages. In recent years, LOX-1 upregulation in different tumors evidenced its involvement in cancer onset, progression and metastasis. In this review, we outline the role of LOX-1 in tumor spreading and metastasis, evidencing its function in VEGF induction, HIF-1alpha activation, and MMP-9/MMP-2 expression, pushing up the neoangiogenic and the epithelial-mesenchymal transition process in glioblastoma, osteosarcoma prostate, colon, breast, lung, and pancreatic tumors. Moreover, our studies contributed to evidence its role in interacting with WNT/APC/β-catenin axis, highlighting new pathways in sporadic colon cancer onset. The application of volatilome analysis in high expressing LOX-1 tumor-bearing mice correlates with the tumor evolution, suggesting a closed link between LOX-1 upregulation and metabolic changes in individual volatile compounds and thus providing a viable method for a simple, non-invasive alternative monitoring of tumor progression. These findings underline the role of LOX-1 as regulator of tumor progression, migration, invasion, metastasis formation, and tumor-related neo-angiogenesis, proposing this receptor as a promising therapeutic target and thus enhancing current antineoplastic strategies.
Collapse
Affiliation(s)
- M Murdocca
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy.
| | - C De Masi
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - S Pucci
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - R Mango
- Cardiology Unit, Department of Emergency and Critical Care, Tor Vergata Hospital, Rome, Italy
| | - G Novelli
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - C Di Natale
- Department of Electronic Engineering, Tor Vergata University, Rome, Italy
| | - F Sangiuolo
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
15
|
Multiplex Protein Biomarker Profiling in Patients with Familial Hypercholesterolemia. Genes (Basel) 2021; 12:genes12101599. [PMID: 34680994 PMCID: PMC8535274 DOI: 10.3390/genes12101599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/29/2021] [Accepted: 10/08/2021] [Indexed: 11/17/2022] Open
Abstract
Familial hypercholesterolemia (FH), is an autosomal dominant disorder caused by mutations in the LDLR, APOB, PCSK9, and APOE genes and is characterized by high plasma levels of total and low-density lipoprotein (LDL) cholesterol. Our study aimed to analyze the influences of two different therapies on a wide spectrum of plasma protein biomarkers of cardiovascular diseases. Plasma from FH patients under hypolipidemic therapy (N = 18; men = 8, age 55.4 ± 13.1 years) and patients under combined long-term LDL apheresis/hypolipidemic therapy (N = 14; men = 7; age 58.0 ± 13.6 years) were analyzed in our study. We measured a profile of 184 cardiovascular disease (CVD) associated proteins using a proximity extension assay (PEA). Hypolipidemic therapy significantly (all p < 0.01) influenced 10 plasma proteins (TM, DKK1, CCL3, CD4, PDGF subunit B, AGRP, IL18, THPO, and LOX1 decreased; ST2 increased). Under combined apheresis/hypolipidemic treatment, 18 plasma proteins (LDLR, PCSK9, MMP-3, GDF2, CTRC, SORT1, VEGFD, IL27, CCL24, and KIM1 decreased; OPN, COL1A1, KLK6, IL4RA, PLC, TNFR1, GLO1, and PTX3 increased) were significantly affected (all p < 0.006). Hypolipidemic treatment mainly affected biomarkers involved in vascular endothelial maintenance. Combined therapy influenced proteins that participate in cholesterol metabolism and inflammation.
Collapse
|
16
|
Orlowski S, Mourad JJ, Gallo A, Bruckert E. Coronaviruses, cholesterol and statins: Involvement and application for Covid-19. Biochimie 2021; 189:51-64. [PMID: 34153377 PMCID: PMC8213520 DOI: 10.1016/j.biochi.2021.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/01/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022]
Abstract
The infectious power of coronaviruses is dependent on cholesterol present in the membranes of their target cells. Indeed, the virus enters the infected cell either by fusion or by endocytosis, in both cases involving cholesterol-enriched membrane microdomains. These membrane domains can be disorganized in-vitro by various cholesterol-altering agents, including statins that inhibit cell cholesterol biosynthesis. As a consequence, numerous cell physiology processes, such as signaling cascades, can be compromised. Also, some examples of anti-bacterial and anti-viral effects of statins have been observed for infectious agents known to be cholesterol dependent. In-vivo, besides their widely-reported hypocholesterolemic effect, statins display various pleiotropic effects mediated, at least partially, by perturbation of membrane microdomains as a consequence of the alteration of endogenous cholesterol synthesis. It should thus be worth considering a high, but clinically well-tolerated, dose of statin to treat Covid-19 patients, in the early phase of infection, to inhibit virus entry into the target cells, in order to control the viral charge and hence avoid severe clinical complications. Based on its efficacy and favorable biodisposition, an option would be considering Atorvastatin, but randomized controlled clinical trials are required to test this hypothesis. This new therapeutic proposal takes benefit from being a drug repurposing, applied to a widely-used drug presenting a high efficiency-to-toxicity ratio. Additionally, this therapeutic strategy avoids any risk of drug resistance by viral mutation since it is host-targeted. Noteworthy, the same pharmacological approach could also be proposed to address different animal coronavirus endemic infections that are responsible for heavy economic losses.
Collapse
Affiliation(s)
- Stéphane Orlowski
- Institute for Integrative Biology of the Cell (I2BC), CNRS UMR 9198, and CEA / DRF / Institut des Sciences du Vivant Frédéric-Joliot / SB2SM, and Université Paris-Saclay, 91191, Gif-sur-Yvette, Cedex, France.
| | - Jean-Jacques Mourad
- Department of Internal Medicine and ESH Excellence Centre, Groupe Hospitalier Paris Saint-Joseph, Paris, France.
| | - Antonio Gallo
- Department of Endocrinology and Prevention of Cardiovascular Diseases, Institute of Cardiometabolism and Nutrition (ICAN), La Pitié-Salpêtrière Hospital, AP-HP, Paris, France.
| | - Eric Bruckert
- Department of Endocrinology and Prevention of Cardiovascular Diseases, Institute of Cardiometabolism and Nutrition (ICAN), La Pitié-Salpêtrière Hospital, AP-HP, Paris, France.
| |
Collapse
|
17
|
Akhmedov A, Sawamura T, Chen CH, Kraler S, Vdovenko D, Lüscher TF. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1): a crucial driver of atherosclerotic cardiovascular disease. Eur Heart J 2021; 42:1797-1807. [PMID: 36282110 DOI: 10.1093/eurheartj/ehaa770] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/18/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases (CVDs), specifically lipid-driven atherosclerotic CVDs, remain the number one cause of death worldwide. The lectin-like oxidized low-density lipoprotein (LDL) receptor-1 (LOX-1), a scavenger receptor that promotes endothelial dysfunction by inducing pro-atherogenic signalling and plaque formation via the endothelial uptake of oxidized LDL (oxLDL) and electronegative LDL, contributes to the initiation, progression, and destabilization of atheromatous plaques, eventually leading to the development of myocardial infarction and certain forms of stroke. In addition to its expression in endothelial cells, LOX-1 is expressed in macrophages, cardiomyocytes, fibroblasts, dendritic cells, lymphocytes, and neutrophils, further implicating this receptor in multiple aspects of atherosclerotic plaque formation. LOX-1 holds promise as a novel diagnostic and therapeutic target for certain CVDs; therefore, understanding the molecular structure and function of LOX-1 is of critical importance. In this review, we highlight the latest scientific findings related to LOX-1, its ligands, and their roles in the broad spectrum of CVDs. We describe recent findings from basic research, delineate their translational value, and discuss the potential of LOX-1 as a novel target for the prevention, diagnosis, and treatment of related CVDs.
Collapse
Affiliation(s)
- Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, Schlieren 8952, Switzerland
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Shinshu University 3-1-1, Asahi, Matsumoto 390-8621, Japan
| | - Chu-Huang Chen
- Vascular and Medical Research, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, Schlieren 8952, Switzerland
| | - Daria Vdovenko
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, Schlieren 8952, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, Schlieren 8952, Switzerland.,Royal Brompton and Harefield Hospitals, Sydney Street, London SW3 6NP, UK.,National Heart and Lung Institute, Imperial College, Dovehause Street, London SW3 6LY, UK
| |
Collapse
|
18
|
Groner J, Goepferich A, Breunig M. Atherosclerosis: Conventional intake of cardiovascular drugs versus delivery using nanotechnology - A new chance for causative therapy? J Control Release 2021; 333:536-559. [PMID: 33794270 DOI: 10.1016/j.jconrel.2021.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is the leading cause of death in developed countries. The pathogenetic mechanism relies on a macrophage-based immune reaction to low density lipoprotein (LDL) deposition in blood vessels with dysfunctional endothelia. Thus, atherosclerosis is defined as a chronic inflammatory disease. A plethora of cardiovascular drugs have been developed and are on the market, but the major shortcoming of standard medications is that they do not address the root cause of the disease. Statins and thiazolidinediones that have recently been recognized to exert specific anti-atherosclerotic effects represent a potential breakthrough on the horizon. But their whole potential cannot be realized due to insufficient availability at the pathological site and severe off-target effects. The focus of this review will be to elaborate how both groups of drugs could immensely profit from nanoparticulate carriers. This delivery principle would allow for their accumulation in target macrophages and endothelial cells of the atherosclerotic plaque, increasing bioavailability where it is needed most. Based on the analyzed literature we conclude design criteria for the delivery of statins and thiazolidinediones with nanoparticles for anti-atherosclerotic therapy. Nanoparticles need to be below a diameter of 100 nm to accumulate in the atherosclerotic plaque and should be fabricated using biodegradable materials. Further, the thiazolidinediones or statins must be encapsulated into the particle core, because especially for thiazolidindiones the uptake into cells is prerequisite for their mechanism of action. For optimal uptake into targeted macrophages and endothelial cells, the ideal particle should present ligands on its surface which bind specifically to scavenger receptors. The impact of statins on the lectin-type oxidized LDL receptor 1 (LOX1) seems particularly promising because of its outstanding role in the inflammatory process. Using this pioneering concept, it will be possible to promote the impact of statins and thiazolidinediones on macrophages and endothelial cells and significantly enhance their anti-atherosclerotic therapeutic potential.
Collapse
Affiliation(s)
- Jonas Groner
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany.
| |
Collapse
|
19
|
Barreto J, Karathanasis SK, Remaley A, Sposito AC. Role of LOX-1 (Lectin-Like Oxidized Low-Density Lipoprotein Receptor 1) as a Cardiovascular Risk Predictor: Mechanistic Insight and Potential Clinical Use. Arterioscler Thromb Vasc Biol 2020; 41:153-166. [PMID: 33176449 DOI: 10.1161/atvbaha.120.315421] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Atherosclerosis, the underlying cause of cardiovascular disease (CVD), is a worldwide cause of morbidity and mortality. Reducing ApoB-containing lipoproteins-chiefly, LDL (low-density lipoprotein)-has been the main strategy for reducing CVD risk. Although supported by large randomized clinical trials, the persistence of residual cardiovascular risk after effective LDL reduction has sparked an intense search for other novel CVD biomarkers and therapeutic targets. Recently, Lox-1 (lectin-type oxidized LDL receptor 1), an innate immune scavenger receptor, has emerged as a promising target for early diagnosis and cardiovascular risk prediction and is also being considered as a treatment target. Lox-1 was first described as a 50 kDa transmembrane protein in endothelial cells responsible for oxLDL (oxidized LDL) recognition, triggering downstream pathways that intensify atherosclerosis via endothelial dysfunction, oxLDL uptake, and apoptosis. Lox-1 is also expressed in platelets, where it enhances platelet activation, adhesion to endothelial cells, and ADP-mediated aggregation, thereby favoring thrombus formation. Lox-1 was also identified in cardiomyocytes, where it was implicated in the development of cardiac fibrosis and myocyte apoptosis, the main determinants of cardiac recovery following an ischemic insult. Together, these findings have revealed that Lox-1 is implicated in all the main steps of atherosclerosis and has encouraged the development of immunoassays for measurement of sLox-1 (serum levels of soluble Lox-1) to be used as a potential CVD biomarker. Finally, the recent development of synthetic Lox-1 inhibitors and neutralizing antibodies with promising results in animal models has made Lox-1 a target for drug development. In this review, we discuss the main findings regarding the role of Lox-1 in the development, diagnosis, and therapeutic strategies for CVD prevention and treatment.
Collapse
Affiliation(s)
- Joaquim Barreto
- Atherosclerosis and Vascular Biology Lab (Atherolab), Clinical Research Center, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Brazil (J.B., A.C.S.)
| | - Sotirios K Karathanasis
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD (S.K.K., A.R.)
- NeoProgen, Baltimore, MD (S.K.K.)
| | - Alan Remaley
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD (S.K.K., A.R.)
| | - Andrei C Sposito
- Atherosclerosis and Vascular Biology Lab (Atherolab), Clinical Research Center, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Brazil (J.B., A.C.S.)
| |
Collapse
|
20
|
Luchetti F, Crinelli R, Nasoni MG, Benedetti S, Palma F, Fraternale A, Iuliano L. LDL receptors, caveolae and cholesterol in endothelial dysfunction: oxLDLs accomplices or victims? Br J Pharmacol 2020; 178:3104-3114. [PMID: 32986849 DOI: 10.1111/bph.15272] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/29/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Oxidized LDLs (oxLDLs) and oxysterols play a key role in endothelial dysfunction and the development of atherosclerosis. The loss of vascular endothelium function negatively impacts vasomotion, cell growth, adhesiveness and barrier functions. While for some of these disturbances, a reasonable explanation can be provided from a mechanistic standpoint, for many others, the molecular mediators that are involved are unknown. Caveolae, specific plasma membrane domains, have recently emerged as targets and mediators of oxLDL-induced endothelial dysfunction. Caveolae and their associated protein caveolin-1 (Cav-1) are involved in oxLDLs/LDLs transcytosis, mainly through the scavenger receptor class B type 1 (SR-B1 or SCARB1). In contrast, oxLDLs endocytosis is mediated by the lectin-like oxidized LDL receptor 1 (LOX-1), whose activity depends on an intact caveolae system. In addition, LOX-1 regulates the expression of Cav-1 and vice versa. On the other hand, oxLDLs may affect cholesterol plasma membrane content/distribution thus influencing caveolae architecture, Cav-1 localization and the associated signalling. Overall, the evidence indicate that caveolae have both active and passive roles in oxLDL-induced endothelial cell dysfunction. First, as mediators of lipid uptake and transfer in the subendothelial space and, later, as targets of changes in composition/dynamics of plasma membrane lipids resulting from increased levels of circulating oxLDLs. Gaining a better understanding of how oxLDLs interact with endothelial cells and modulate caveolae-mediated signalling pathways, leading to endothelial dysfunction, is crucial to find new targets for intervention to tackle atherosclerosis and the related clinical entities. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Rita Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Maria Gemma Nasoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Serena Benedetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Francesco Palma
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | | | - Luigi Iuliano
- Department of Medico-Surgical Sciences and Biotechnologies Vascular Biology, Atherothrombosis & Mass Spectrometry, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
21
|
Steroidal glycoalkaloids from Solanum lyratum inhibit the pro-angiogenic activity of A549-derived exosomes. Fitoterapia 2020; 141:104481. [PMID: 31954179 DOI: 10.1016/j.fitote.2020.104481] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/14/2020] [Accepted: 01/14/2020] [Indexed: 01/18/2023]
Abstract
In this study, seven previously undescribed steroidal glycoalkaloids, compounds 1-7, were isolated from Solanum lyratum, along with two known ones (8 and 9). Comprehensive spectroscopy techniques were used to determine their structures. Although 1-8 only showed a weak inhibitory effect on the proliferation of the tumor-derived vascular endothelial cells, however, in a former study we found both total steroidal glycoalkaloids from Solanum lyratum (TSGS) and 9 significantly inhibited tumor angiogenesis and its mechanism was linked to its ability to interfere with cell membrane lipid rafts. Lipid rafts are closely related to the functions of tumor-derived exosomes, a vital factor in cancer progression. Thus, we investigated the impacts of TSGS and 9 on the functions of A549-derived exosomes. Our results indicated that A549-derived exosomes can significantly enhance the angiogenesis abilities of human umbilical vein endothelial cells, whereas the intervention of TSGS or 9 significantly inhibited this activity of A549-derived exosomes. These findings suggest that TSGS and 9 exert anti-tumor angiogenesis by inhibiting the pro-angiogenic activity of A549-derived exosomes.
Collapse
|
22
|
Raniolo S, Croce S, Thomsen RP, Okholm AH, Unida V, Iacovelli F, Manetto A, Kjems J, Desideri A, Biocca S. Cellular uptake of covalent and non-covalent DNA nanostructures with different sizes and geometries. NANOSCALE 2019; 11:10808-10818. [PMID: 31134260 DOI: 10.1039/c9nr02006c] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
DNA nanostructures with different sizes and shapes, assembled through either covalent or non-covalent bonds, namely tetrahedral and octahedral nanocages, rod-shaped chainmails, square box and rectangular DNA origami structures, were compared for their stability in serum, cell surface binding, internalization efficiency, and intracellular degradation rate. For cell internalization a specific cell system, highly expressing the scavenger receptor LOX-1 was used. The results indicate that LOX-1 binds and internalizes a broad family of DNA structures of different sizes that, however, have a different fate and lifetime inside the cells. Covalently linked tetrahedra, octahedra or chainmails are intact inside cells for up to 18 hours whilst the same DNA nanostructures without covalent bonds along with square box and rectangular origami are rapidly degraded. These data suggest that non-covalently linked structures may be useful for fast drug release whilst the covalently-linked structures could be appropriate vehicles for slow release of molecules.
Collapse
Affiliation(s)
- Sofia Raniolo
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Roma, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kattoor AJ, Kanuri SH, Mehta JL. Role of Ox-LDL and LOX-1 in Atherogenesis. Curr Med Chem 2019; 26:1693-1700. [DOI: 10.2174/0929867325666180508100950] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/01/2017] [Accepted: 12/01/2017] [Indexed: 02/02/2023]
Abstract
Oxidized LDL (ox-LDL) plays a central role in atherosclerosis by acting on multiple
cells such as endothelial cells, macrophages, platelets, fibroblasts and smooth muscle cells
through LOX-1. LOX-1 is a 50 kDa transmembrane glycoprotein that serves as receptor for
ox-LDL, modified lipoproteins, activated platelets and advance glycation end-products. Ox-
LDL through LOX-1, in endothelial cells, causes increase in leukocyte adhesion molecules,
activates pathways of apoptosis, increases reactive oxygen species and cause endothelial dysfunction.
In vascular smooth muscle cells and fibroblasts, they stimulate proliferation, migration
and collagen synthesis. LOX-1 expressed on macrophages inhibit macrophage migration
and stimulate foam cell formation. They also stimulate generation of metalloproteinases and
contribute to plaque instability and thrombosis. Drugs that modulate LOX-1 are desirable targets
against atherosclerosis. Many naturally occurring compounds have been shown to modulate
LOX-1 expression and atherosclerosis. Currently, novel drug design techniques are used
to identify molecules that can bind to LOX-1 and inhibit its activation by ox-LDL. In addition,
techniques using RNA interference and monoclonal antibody against LOX-1 are currently
being investigated for clinical use.
Collapse
Affiliation(s)
- Ajoe John Kattoor
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Sri Harsha Kanuri
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Jawahar L. Mehta
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| |
Collapse
|
24
|
Pucci S, Polidoro C, Greggi C, Amati F, Morini E, Murdocca M, Biancolella M, Orlandi A, Sangiuolo F, Novelli G. Pro-oncogenic action of LOX-1 and its splice variant LOX-1Δ4 in breast cancer phenotypes. Cell Death Dis 2019; 10:53. [PMID: 30718451 PMCID: PMC6362207 DOI: 10.1038/s41419-018-1279-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022]
Abstract
The identification of new predictive biomarkers and therapeutic target for tailored therapy in breast cancer onset and progression is an interesting challenge. OLR-1 gene encodes the cell membrane receptor LOX-1 (lectin-like oxidized low-density lipoprotein receptor). We have recently identified a novel alternative OLR-1 isoform, LOX-1Δ4, whose expression and functions are still not clarified. In the present paper, we demonstrated that LOX-1 is overexpressed in 70% of human breast cancer (n = 47) and positively correlated to the tumor stage and grade (p < 0.01). Observations on LOX-1 and its splice variant Δ4 pointed out a different expression pattern correlated to breast cancer phenotypes. Overexpressing LOX-1 and LOX-1Δ4 in vitro, we obtained a strong enhancement of proliferative rate and a downregulation of cell death-related proteins. In addition, we observed a strong modulation of histone H4 acetylation and Ku70, the limiting factor of DNA double-strand breaks repair machinery implied in apoptosis inhibition and drug resistance acquisition. Moreover, LOX-1Δ4 overexpression is able to increase proliferation in a non-tumorigenic epithelial cell line, MCF12-F, acting as an oncogene. Altogether, these results suggest that LOX-1 may acts as a molecular link among metabolism, inflammation and cancer, indicating its potential role as biomarker and new molecular target, representing an attractive and concrete opportunity to improve current strategies for breast cancer tailored therapy.
Collapse
Affiliation(s)
- Sabina Pucci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.
| | - Chiara Polidoro
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Chiara Greggi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Francesca Amati
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Elena Morini
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Michela Murdocca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Michela Biancolella
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.,Neuromed, I.R.C.C.S, Pozzilli, Italy
| |
Collapse
|
25
|
Raniolo S, Vindigni G, Unida V, Ottaviani A, Romano E, Desideri A, Biocca S. Entry, fate and degradation of DNA nanocages in mammalian cells: a matter of receptors. NANOSCALE 2018; 10:12078-12086. [PMID: 29911715 DOI: 10.1039/c8nr02411a] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
DNA has been used to build nanostructures with potential biomedical applications. However, their use is limited by the lack of information on the mechanism of entry, intracellular fate and degradation rate of nanostructures inside cells. We generated octahedral DNA nanocages functionalized with folic acid and investigated the cellular uptake mediated by two distinctive internalization pathways, using two cellular systems expressing the oxidized low-density lipoprotein receptor-1 (LOX-1) and the α isoform of the folate receptor (αFR), respectively. Here, we report that DNA nanocages are very efficiently and selectively internalized by both receptors with an efficiency at least 30 times higher than that observed in cells not expressing the receptors. When internalized by LOX-1, nanocages traffic to lysosomes within 4 hours and are rapidly degraded. When the uptake is mediated by αFR, DNA nanocages are highly stable (>48 hours) and accumulate inside cells in a time-dependent way. These data demonstrate that the selection of the cellular receptor is crucial for targeting specific sub-cellular compartments and for modulating the DNA nanocage intracellular half-life, indicating that vitamin-mediated uptake may constitute a protected pathway for intracellular drug delivery.
Collapse
Affiliation(s)
- Sofia Raniolo
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
26
|
da Silva ED, Cancela M, Monteiro KM, Ferreira HB, Zaha A. Antigen B from Echinococcus granulosus enters mammalian cells by endocytic pathways. PLoS Negl Trop Dis 2018; 12:e0006473. [PMID: 29727452 PMCID: PMC5955594 DOI: 10.1371/journal.pntd.0006473] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 05/16/2018] [Accepted: 04/23/2018] [Indexed: 01/18/2023] Open
Abstract
Background Cystic hydatid disease is a zoonosis caused by the larval stage (hydatid) of Echinococcus granulosus (Cestoda, Taeniidae). The hydatid develops in the viscera of intermediate host as a unilocular structure filled by the hydatid fluid, which contains parasitic excretory/secretory products. The lipoprotein Antigen B (AgB) is the major component of E. granulosus metacestode hydatid fluid. Functionally, AgB has been implicated in immunomodulation and lipid transport. However, the mechanisms underlying AgB functions are not completely known. Methodology/Principal findings In this study, we investigated AgB interactions with different mammalian cell types and the pathways involved in its internalization. AgB uptake was observed in four different cell lines, NIH-3T3, A549, J774 and RH. Inhibition of caveolae/raft-mediated endocytosis causes about 50 and 69% decrease in AgB internalization by RH and A549 cells, respectively. Interestingly, AgB colocalized with the raft endocytic marker, but also showed a partial colocalization with the clathrin endocytic marker. Finally, AgB colocalized with an endolysosomal tracker, providing evidence for a possible AgB destination after endocytosis. Conclusions/Significance The results indicate that caveolae/raft-mediated endocytosis is the main route to AgB internalization, and that a clathrin-mediated entry may also occur at a lower frequency. A possible fate for AgB after endocytosis seems to be the endolysosomal system. Cellular internalization and further access to subcellular compartments could be a requirement for AgB functions as a lipid carrier and/or immunomodulatory molecule, contributing to create a more permissive microenvironment to metacestode development and survival. Antigen B (AgB) is an oligomeric lipoprotein highly abundant in Echinococcus granulosus hydatid fluid. AgB has already been characterized as an immunomodulatory protein, capable of inducing a permissive immune response to parasite development. Also, an important role in lipid acquisition is attributed to AgB, because it has been found associated to different classes of host lipids. However, the mechanisms of interaction employed by AgB to perform its functions remain undetermined. In this study, we demonstrate that mammalian cells are able to internalize E. granulosus AgB in culture and found that specific mechanisms of endocytosis are involved. Our results extend the understanding of AgB biological role indicating cellular internalization as a mechanism of interaction, which in turn, may represent a target to intervention.
Collapse
Affiliation(s)
- Edileuza Danieli da Silva
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Martin Cancela
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Karina Mariante Monteiro
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Henrique Bunselmeyer Ferreira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Arnaldo Zaha
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- * E-mail:
| |
Collapse
|
27
|
Balzan S, Lubrano V. LOX-1 receptor: A potential link in atherosclerosis and cancer. Life Sci 2018; 198:79-86. [PMID: 29462603 DOI: 10.1016/j.lfs.2018.02.024] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/07/2018] [Accepted: 02/16/2018] [Indexed: 12/19/2022]
Abstract
Altered production of reactive oxygen species (ROS), causing lipid peroxidation and DNA damage, contributes to the progression of atherosclerosis and cancer. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a lectin-like receptor for oxidized low-density lipoproteins (ox-LDL) primarily expressed in endothelial cells and vasculature-rich organs. LOX-1 receptors is a marker for atherosclerosis, and once activated by ox-LDL or other ligands, stimulates the expression of adhesion molecules, pro-inflammatory signaling pathways and proangiogenic proteins, including NF-kB and VEGF, in vascular endothelial cells and macrophages. Several different types of cancer reported LOX-1 gene upregulation, and numerous interplays exist concerning LOX-1 in atherosclerosis, metabolic diseases and cancer. One of them involves NF-kB, an oncogenic protein that regulates the transcription of several inflammatory genes response. In a model of cellular transformation, the MCF10A ER-Src, inhibition of LOX-1 gene reduces NF-kB activation and the inflammatory and hypoxia pathways, suggesting a mechanistic connection between cellular transformation and atherosclerosis. The remodeling proteins MMP-2 and MMP-9 have been found increased in angiogenesis in atherosclerotic plaque and also in human prostate cancer cells. In this review, we outlined the role of LOX-1 in atherogenesis and tumorigenesis as a potential link in these diseases, suggesting that LOX-1 inhibition could represent a promising strategy in the treatment of atherosclerosis and tumors.
Collapse
Affiliation(s)
- Silvana Balzan
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, Pisa 56124, Italy.
| | - Valter Lubrano
- Fondazione CNR/Regione Toscana G. Monasterio, Via Moruzzi 1, Pisa 56124, Italy
| |
Collapse
|
28
|
Murdocca M, Mango R, Pucci S, Biocca S, Testa B, Capuano R, Paolesse R, Sanchez M, Orlandi A, di Natale C, Novelli G, Sangiuolo F. The lectin-like oxidized LDL receptor-1: a new potential molecular target in colorectal cancer. Oncotarget 2018; 7:14765-80. [PMID: 26895376 PMCID: PMC4924750 DOI: 10.18632/oncotarget.7430] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/29/2016] [Indexed: 01/01/2023] Open
Abstract
The identification of new biomarkers and targets for tailored therapy in human colorectal cancer (CRC) onset and progression is an interesting challenge. CRC tissue produces an excess of ox-LDL, suggesting a close correlation between lipid dysfunction and malignant transformation. Lectin-like oxidized LDL receptor-1 (LOX-1) is involved in several mechanisms closely linked to tumorigenesis. Here we report a tumor specific LOX-1 overexpression in human colon cancers: LOX-1 results strongly increased in the 72% of carcinomas (P<0.001), and strongly overexpressed in 90% of highly aggressive and metastatic tumours (P<0.001), as compared to normal mucosa. Moreover LOX-1 results modulated since the early stage of the disease (adenomas vs normal mucosa; P<0.001) suggesting an involvement in tumor insurgence and progression. The in vitro knockdown of LOX-1 in DLD-1 and HCT-8 colon cancer cells by siRNA and anti-LOX-1 antibody triggers to an impaired proliferation rate and affects the maintenance of cell growth and tumorigenicity. The wound-healing assay reveals an evident impairment in closing the scratch. Lastly knockdown of LOX-1 delineates a specific pattern of volatile compounds characterized by the presence of a butyrate derivative, suggesting a potential role of LOX-1 in tumor-specific epigenetic regulation in neoplastic cells. The role of LOX-1 as a novel biomarker and molecular target represents a concrete opportunity to improve current therapeutic strategies for CRC. In addition, the innovative application of a technology focused to the identification of LOX-1 driven volatiles specific to colorectal cancer provides a promising diagnostic tool for CRC screening and for monitoring the response to therapy.
Collapse
Affiliation(s)
- Michela Murdocca
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Ruggiero Mango
- Department of Emergency and Critical Care, Section of Cardiology, Policlinic of Tor Vergata, Rome, Italy
| | - Sabina Pucci
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Silvia Biocca
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Barbara Testa
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Rosamaria Capuano
- Department of Electronic Engineering, Tor Vergata University, Rome, Italy
| | - Roberto Paolesse
- Department of Chemical Science and Technology, Tor Vergata University, Rome, Italy
| | - Massimo Sanchez
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Corrado di Natale
- Department of Electronic Engineering, Tor Vergata University, Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
29
|
Griffin JI, Wang G, Smith WJ, Vu VP, Scheinman R, Stitch D, Moldovan R, Moghimi SM, Simberg D. Revealing Dynamics of Accumulation of Systemically Injected Liposomes in the Skin by Intravital Microscopy. ACS NANO 2017; 11:11584-11593. [PMID: 29045127 PMCID: PMC5770233 DOI: 10.1021/acsnano.7b06524] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Accumulation of intravenously injected cytotoxic liposomes in the skin induces serious toxicity. We used single time point and longitudinal intravital microscopy to understand skin accumulation dynamics of non-PEGylated and PEGylated liposomes after systemic injection into mice. Non-PEGylated egg phosphatidylcholine (PC) liposomes showed short circulation half-life (1.3 h) and immediate aggregation in the blood, with some aggregates lodging in skin microvasculature soon after the injection. At 24 h, and more prominently at 48 h postinjection, liposomes appeared in dermal and subdermal cells. PEGylated egg PC liposomes showed long circulation half-life (22 h) and no aggregation in the blood. PEGylated liposomes started to accumulate in the skin microvasculature as soon as 5 min after the injection. Within 3 h postinjection, PEGylated liposomes accumulated in extravascular cells in the dermis and subdermis. Liposomes were present in the skin for at least 7 days postinjection. A regulatory approved PEGylated liposomal doxorubicin (LipoDox) and empty liposomes of the same composition as LipoDox showed similar skin distribution as PEGylated egg PC liposomes, suggesting that this phenomenon is relevant to liposomes of different lipid composition. Decorating liposomes with shorter PEGs (350 or 700) in addition to PEG 2000 did not decrease the deposition. Outside the capillaries, liposomes partially colocalized with CD45-, F4/80+ cells. The accumulation of liposomes was not due to prior neutrophil/platelet binding and transport across endothelium. Moreover, our studies have excluded a role of complement in the skin accumulation of liposomes. Further understanding of mechanisms of this important phenomenon can improve the safety of liposomal nanocarriers.
Collapse
Affiliation(s)
- James I. Griffin
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
| | - Weston J. Smith
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
| | - Vivian P. Vu
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
| | - Robert Scheinman
- Colorado Center for Nanomedicine and Nanosafety, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
| | - Dominik Stitch
- Advanced Light Microscopy Core Facility, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Radu Moldovan
- Advanced Light Microscopy Core Facility, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Seyed Moein Moghimi
- Colorado Center for Nanomedicine and Nanosafety, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- School of Pharmacy, The Faculty of Medical Sciences, Newcastle University, King George VI Building, Newcastle upon Tyne NE1 7RU, U.K
- Division of Stratified Medicine, Biomarkers & Therapeutics, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE1 7RU, U.K
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, United States
- Corresponding Author
| |
Collapse
|
30
|
Membrane cholesterol depletion in cortical neurons highlights altered NMDA receptor functionality in a mouse model of amyotrophic lateral sclerosis. Biochim Biophys Acta Mol Basis Dis 2017; 1864:509-519. [PMID: 29154925 DOI: 10.1016/j.bbadis.2017.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/16/2017] [Accepted: 11/13/2017] [Indexed: 12/25/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a chronic neurodegenerative disease affecting upper and lower motor neurons, with unknown aetiology. Lipid rafts, cholesterol enriched microdomains of the plasma membrane, have been linked to neurodegenerative disorders like ALS. The NMDA-receptor subcellular localization in lipid rafts is known to play many roles, from modulating memory strength to neurotoxicity. In this study, performed on the widely used G93A mouse model of ALS, we have shown an equal content of total membrane cholesterol in Control and G93A cortical cultures. Moreover, by electrophysiological studies, we have recorded NMDA- and AMPA-evoked currents which were not significantly different between the two neuronal populations. To study the role of membrane cholesterol on glutamate receptor functionality, we have analysed NMDA and AMPA receptors following cholesterol membrane depletion by methyl-β-cyclodextrin (MβCD). Interestingly, MβCD chronic treatment has provoked a significant reduction of NMDA-evoked currents in both cellular populations which was dose- and time-dependent but significantly higher in ALS neurons compared to Control. The different MβCD effect on NMDA-evoked currents was not due to a different membrane receptor subunit composition but seemed to cause in both neuronal populations a NMDA receptor membrane redistribution. MβCD treatment effect was receptor-specific since no alterations in the two neuronal populations were detected on AMPA receptors. These results lead us to speculate for an altered proteomic composition of lipid rafts in cortical mutated neurons and suggest the need for further studies on the lipid rafts composition and on their interaction with membrane receptors in ALS cortices.
Collapse
|
31
|
Hofmann A, Brunssen C, Morawietz H. Contribution of lectin-like oxidized low-density lipoprotein receptor-1 and LOX-1 modulating compounds to vascular diseases. Vascul Pharmacol 2017; 107:S1537-1891(17)30171-4. [PMID: 29056472 DOI: 10.1016/j.vph.2017.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/29/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022]
Abstract
The lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is the major receptor for binding and uptake of oxidized low-density lipoprotein (oxLDL) in endothelial cells. LOX-1 is also expressed in macrophages, smooth muscle cells and platelets. Following internalization of oxLDL, LOX-1 initiates a vicious cycle from activation of pro-inflammatory signaling pathways, thus promoting an increased reactive oxygen species formation and secretion of pro-inflammatory cytokines. LOX-1 plays a pivotal role in the development of endothelial dysfunction, foam cell and advanced lesions formation as well as in myocardial ischemia. Furthermore, it is known that LOX-1 plays a pivotal role in mitochondrial DNA damage, vascular cell apoptosis, and autophagy. A large number of studies provide evidence of a LOX-1's role in endothelial dysfunction, hypertension, diabetes, and obesity. In addition, novel insights into LOX-1 ligands and the activated signaling pathways have been gained. Recent studies have shown an interaction of LOX-1 with microRNA's, thus providing novel tools to regulate LOX-1 function. Because LOX-1 is increased in atherosclerotic plaques and contributes to endothelial dysfunction, several compounds were tested in vivo and in vitro to modulate the LOX-1 expression in therapeutic approaches.
Collapse
Affiliation(s)
- Anja Hofmann
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
32
|
Statins do not inhibit the FGFR signaling in chondrocytes. Osteoarthritis Cartilage 2017; 25:1522-1530. [PMID: 28583899 DOI: 10.1016/j.joca.2017.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 05/19/2017] [Accepted: 05/25/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Statins are widely used drugs for cholesterol lowering, which were recently found to counteract the effects of aberrant fibroblast growth factor receptor (FGFR3) signaling in cell and animal models of FGFR3-related chondrodysplasia. This opened an intriguing therapeutic possibility for human dwarfing conditions caused by gain-of-function mutations in FGFR3, although the mechanism of statin action on FGFR3 remains unclear. Here, we determine the effect of statins on FGFR signaling in chondrocytes. DESIGN Cultured chondrocyte cell lines, mouse embryonic tibia cultures and limb bud micromasses were treated with FGF2 to activate FGFR signaling. The effects of atorvastatin, fluvastatin, lovastatin and pravastatin on FGFR3 protein stability and on FGFR-mediated chondrocyte growth-arrest, loss of extracellular matrix (ECM), induction of premature senescence and hypertrophic differentiation were evaluated. RESULTS Statins did not alter the level of FGFR3 protein expression nor produce any effect on FGFR-mediated inhibition of chondrocyte proliferation and hypertrophic differentiation in cultured chondrocyte cell lines, mouse tibia cultures or limb bud micromasses. CONCLUSION We conclude that statins do not inhibit the FGFR signaling in chondrocytes. Therefore the statin-mediated rescue of FGFR3-related chondrodysplasia, described before, is likely not intrinsic to the growth plate cartilage.
Collapse
|
33
|
Desai AJ, Miller LJ. Changes in the plasma membrane in metabolic disease: impact of the membrane environment on G protein-coupled receptor structure and function. Br J Pharmacol 2017; 175:4009-4025. [PMID: 28691227 DOI: 10.1111/bph.13943] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/08/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022] Open
Abstract
Drug development targeting GPCRs often utilizes model heterologous cell expression systems, reflecting an implicit assumption that the membrane environment has little functional impact on these receptors or on their responsiveness to drugs. However, much recent data have illustrated that membrane components can have an important functional impact on intrinsic membrane proteins. This review is directed toward gaining a better understanding of the structure of the plasma membrane in health and disease, and how this organelle can influence GPCR structure, function and regulation. It is important to recognize that the membrane provides a potential mode of lateral allosteric regulation of GPCRs and can affect the effectiveness of drugs and their biological responses in various disease states, which can even vary among individuals across the population. The type 1 cholecystokinin receptor is reviewed as an exemplar of a class A GPCR that is affected in this way by changes in the plasma membrane. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
34
|
Batais MA, Khan AR, Bin Abdulhak AA. The Use of Statins and Risk of Community-Acquired Pneumonia. Curr Infect Dis Rep 2017; 19:26. [PMID: 28639080 DOI: 10.1007/s11908-017-0581-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF THE REVIEW Community-acquired pneumonia (CAP) is still associated with a large burden and causes significant morbidity and mortality. Besides universal vaccination and antibiotic treatment, statins as adjunctive therapy may also have a beneficial role in the prevention and treatment of CAP. Our goal from this review is to discuss the epidemiology of CAP, and role of statins as adjunctive therapy in the development of CAP. RECENT FINDINGS Statins are lipid-lowering medications characterized by their ability to control hypercholesterolemia in addition to other pleiotropic effects that could explain their role in the pathogenesis of CAP. While most observational studies have shown that statins reduce risk of pneumonia in the general population, patients with diabetes, and recently in patients with myocardial infarction, no randomized controlled trial (RCT) to date has been conducted to assess the efficacy of statins to prevent development of CAP. Given the paucity of robust randomized evidence to assess statin use and the development of CAP, and considering conflicting results of the observational studies, we are not in favor of initiation of statins for either the prevention or treatment of CAP.
Collapse
Affiliation(s)
- Mohammed A Batais
- College of Medicine, King Saud University, Riyadh, 29391, Saudi Arabia.
| | - Abdur Rahman Khan
- Department of Medicine, Division of Cardiovascular Diseases, University of Louisville, Louisville, KY, USA
| | - Aref A Bin Abdulhak
- Department of Medicine, Division of Cardiovascular Diseases, University of Iowa Hospitals and Clinics, 200 Hawkins Dr., Int. Med. E315 GH, Iowa City, IA, 52242, USA
| |
Collapse
|
35
|
Iacovelli F, Tucci FG, Macari G, Falconi M. Multiple molecular dynamics simulations of human LOX-1 and Trp150Ala mutant reveal the structural determinants causing the full deactivation of the receptor. Proteins 2017; 85:1902-1912. [DOI: 10.1002/prot.25344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 11/06/2022]
Affiliation(s)
| | | | - Gabriele Macari
- Department of Biology; University of Rome “Tor Vergata”; Rome Italy
| | - Mattia Falconi
- Department of Biology; University of Rome “Tor Vergata”; Rome Italy
| |
Collapse
|
36
|
Caliceti C, Rizzo P, Ferrari R, Fortini F, Aquila G, Leoncini E, Zambonin L, Rizzo B, Calabria D, Simoni P, Mirasoli M, Guardigli M, Hrelia S, Roda A, Cicero AFG. Novel role of the nutraceutical bioactive compound berberine in lectin-like OxLDL receptor 1-mediated endothelial dysfunction in comparison to lovastatin. Nutr Metab Cardiovasc Dis 2017; 27:552-563. [PMID: 28511903 DOI: 10.1016/j.numecd.2017.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/31/2017] [Accepted: 04/10/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Oxidized LDL (oxLDL) or pro-inflammatory stimuli lead to increased oxidative stress linked to endothelial dysfunction and atherosclerosis. The oxLDL receptor-1 (LOX1) is elevated within atheromas and cholesterol-lowering statins inhibit LOX1 expression. Berberine (BBR), an alkaloid extracted from plants of gender Berberis, has lipid-lowering and anti-inflammatory activity. However, its role in regulating LOX1-mediated signaling is still unknown. The aim of this study was to investigate the effect of BBR on oxLDL- and TNFα-induced endothelial dysfunction in human umbilical vein endothelial cells (HUVECs) and to compare it with that of lovastatin (LOVA). METHODS AND RESULTS Cytotoxicity was determined by lactate dehydrogenase assay. Antioxidant capacity was measured with chemiluminescent and fluorescent method and intracellular ROS levels through a fluorescent dye. Gene and protein expression levels were assayed by qRT-PCR and western blot, respectively. HUVECs exposure to oxLDL (30 μg/ml) or TNFα (10 ng/ml) for 24 h led to a significant increase in LOX1 expression, effect abrogated by BBR (5 μM) and LOVA (5 μM). BBR but not LOVA treatment abolished the TNFα-induced cytotoxicity and restored the activation of Akt signaling. In spite of a low direct antioxidant capacity, both compounds reduced intracellular ROS levels generated by treatment of TNFα but only BBR inhibited NOX2 expression, MAPK/Erk1/2 signaling and subsequent NF-κB target genes VCAM and ICAM expression, induced by TNFα. CONCLUSIONS These findings demonstrated for the first time that BBR could prevent the oxLDL and TNFα - induced LOX1 expression and oxidative stress, key events that lead to NOX, MAPK/Erk1/2 and NF-κB activation linked to endothelial dysfunction. CHEMICAL COMPOUNDS STUDIED IN THIS ARTICLE Berberine (PubChem CID: 2353); Lovastatin (PubChem CID: 53232).
Collapse
Affiliation(s)
- C Caliceti
- Department of Chemistry "Giacomo Ciamician" - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Centro Interdipartimentale di Ricerca Industriale Energia e Ambiente (CIRI EA) - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy.
| | - P Rizzo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy; Maria Cecilia Hospital, GVM Care&Research, E.S: Health Science Foundation, Cotignola, Italy
| | - R Ferrari
- Department of Medical Sciences, Cardiology and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy; Maria Cecilia Hospital, GVM Care&Research, E.S: Health Science Foundation, Cotignola, Italy
| | - F Fortini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - G Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - E Leoncini
- Department for Life Quality Studies - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - L Zambonin
- Department of Pharmacy and Biotechnology - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - B Rizzo
- Department for Life Quality Studies - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - D Calabria
- Centro Interdipartimentale di Ricerca Industriale Energia e Ambiente (CIRI EA) - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy
| | - P Simoni
- Department of Medical and Surgical Sciences-DIMEC, Sant'Orsola Malpighi Hospital, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - M Mirasoli
- Department of Chemistry "Giacomo Ciamician" - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Centro Interdipartimentale di Ricerca Industriale Energia e Ambiente (CIRI EA) - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy
| | - M Guardigli
- Department of Chemistry "Giacomo Ciamician" - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy
| | - S Hrelia
- Department for Life Quality Studies - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - A Roda
- Department of Chemistry "Giacomo Ciamician" - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Centro Interdipartimentale di Ricerca Industriale Energia e Ambiente (CIRI EA) - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy
| | - A F G Cicero
- Department of Medical and Surgical Sciences-DIMEC, Sant'Orsola Malpighi Hospital, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
37
|
Bruno V, Rizzacasa B, Pietropolli A, Capogna MV, Massoud R, Ticconi C, Piccione E, Cortese C, Novelli G, Amati F. OLR1 and Loxin Expression in PBMCs of Women with a History of Unexplained Recurrent Miscarriage: A Pilot Study. Genet Test Mol Biomarkers 2017; 21:363-372. [PMID: 28409654 DOI: 10.1089/gtmb.2016.0331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
AIMS The aim of this study was to evaluate the expression of OLR1 and its alternative splicing isoform Loxin in unexplained recurrent miscarriage (uRM). METHODS Sixty-three women of reproductive age were recruited and were divided into four groups: 18 pregnant and 23 non-pregnant women with uRM, and 12 pregnant and 10 non-pregnant women with physiological pregnancies. Complementary DNA derived from peripheral blood mononuclear cells (PBMCs) was analyzed by quantitative real-time PCR to evaluate the expression of OLR1 and Loxin. Oxidized low-density lipoproteins (ox-LDLs) were assayed from serum by a commercially available kit. RESULTS Pregnant uRM women presented with a higher, though not significant, OLR1/Loxin ratio and a higher ox-LDLs serum level (p ≤ 0.05) compared with pregnant control women. OLR1 and Loxin levels were significantly decreased in non-pregnant uRM women compared with the control (OLR1: 0.00018 vs. 0.00043, p ≤ 0.005; Loxin: 0.00018 vs. 0.00060, p ≤ 0.005, respectively). Loxin expression decreased by about two-thirds (p ≤ 0.005) in pregnant women compared with non-pregnant control women. A higher expression of OLR1 in pregnant women compared with non-pregnant women with uRM (p ≤ 0.05) was observed, but no variation in Loxin expression was observed. CONCLUSIONS The results of this study show an association of peripheral OLR1 and Loxin expression levels in uRM women, and they suggest the possible existence of an uncontrolled oxidative stress in these women in the first trimester of pregnancy.
Collapse
Affiliation(s)
- Valentina Bruno
- 1 Academic Department of Biomedicine and Prevention, Section of Gynecology and Obstetrics, University of Rome Tor Vergata and Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital , Rome, Italy .,2 Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital, Rome , Italy
| | - Barbara Rizzacasa
- 3 Department of Biomedicine and Prevention, Section of Genetics, University of Rome Tor Vergata , Rome, Italy
| | - Adalgisa Pietropolli
- 1 Academic Department of Biomedicine and Prevention, Section of Gynecology and Obstetrics, University of Rome Tor Vergata and Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital , Rome, Italy .,2 Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital, Rome , Italy
| | - Maria Vittoria Capogna
- 1 Academic Department of Biomedicine and Prevention, Section of Gynecology and Obstetrics, University of Rome Tor Vergata and Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital , Rome, Italy .,2 Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital, Rome , Italy
| | - Renato Massoud
- 4 Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Tor Vergata University Hospital , Rome, Italy
| | - Carlo Ticconi
- 1 Academic Department of Biomedicine and Prevention, Section of Gynecology and Obstetrics, University of Rome Tor Vergata and Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital , Rome, Italy .,2 Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital, Rome , Italy
| | - Emilio Piccione
- 1 Academic Department of Biomedicine and Prevention, Section of Gynecology and Obstetrics, University of Rome Tor Vergata and Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital , Rome, Italy .,2 Department of Surgery, Section of Gynecology and Obstetrics, Tor Vergata University Hospital, Rome , Italy
| | - Claudio Cortese
- 4 Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Tor Vergata University Hospital , Rome, Italy
| | - Giuseppe Novelli
- 3 Department of Biomedicine and Prevention, Section of Genetics, University of Rome Tor Vergata , Rome, Italy
| | - Francesca Amati
- 3 Department of Biomedicine and Prevention, Section of Genetics, University of Rome Tor Vergata , Rome, Italy
| |
Collapse
|
38
|
Chistiakov DA, Melnichenko AA, Orekhov AN, Bobryshev YV. How do macrophages sense modified low-density lipoproteins? Int J Cardiol 2017; 230:232-240. [DOI: 10.1016/j.ijcard.2016.12.164] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/19/2016] [Accepted: 12/25/2016] [Indexed: 01/18/2023]
|
39
|
Halls ML, Cooper DMF. Adenylyl cyclase signalling complexes - Pharmacological challenges and opportunities. Pharmacol Ther 2017; 172:171-180. [PMID: 28132906 DOI: 10.1016/j.pharmthera.2017.01.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signalling pathways involving the vital second messanger, cAMP, impact on most significant physiological processes. Unsurprisingly therefore, the activation and regulation of cAMP signalling is tightly controlled within the cell by processes including phosphorylation, the scaffolding of protein signalling complexes and sub-cellular compartmentalisation. This inherent complexity, along with the highly conserved structure of the catalytic sites among the nine membrane-bound adenylyl cyclases, presents significant challenges for efficient inhibition of cAMP signalling. Here, we will describe the biochemistry and cell biology of the family of membrane-bound adenylyl cyclases, their organisation within the cell, and the nature of the cAMP signals that they produce, as a prelude to considering how cAMP signalling might be perturbed. We describe the limitations associated with direct inhibition of adenylyl cyclase activity, and evaluate alternative strategies for more specific targeting of adenylyl cyclase signalling. The inherent complexity in the activation and organisation of adenylyl cyclase activity may actually provide unique opportunities for selectively targeting discrete adenylyl cyclase functions in disease.
Collapse
Affiliation(s)
- Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, Victoria, Australia
| | - Dermot M F Cooper
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| |
Collapse
|
40
|
Overexpression of LOXIN Protects Endothelial Progenitor Cells From Apoptosis Induced by Oxidized Low Density Lipoprotein. J Cardiovasc Pharmacol 2017; 67:326-35. [PMID: 26771151 DOI: 10.1097/fjc.0000000000000358] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human endothelial progenitor cells (hEPC) are adult stem cells located in the bone marrow and peripheral blood. Studies have indicated that hEPC play an important role in the recovery and repair of injured endothelium, however, their quantity and functional capacity is reduced in several diseases including hypercholesterolemia. Recently, it has been demonstrated that hEPC express lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) and its activation by oxidized low-density lipoprotein (ox-LDL) induces cellular dysfunction and apoptosis. This study aimed to investigate whether overexpression of LOXIN, a truncated isoform of LOX-1 that acts as a dominant negative, plays a protective role against ox-LDL-induced apoptosis in hEPC. Human endothelial progenitor cells exposed to ox-LDL showed a significant increase in LOX-1 expression, and apoptosis began at ox-LDL concentrations above 50 μg/mL. All hEPC apoptosed at 200 μg/mL ox-LDL. High LOXIN expression was generated using adenoviral systems in hEPC and SiHa cells transduced with 100 colony-forming units per cell. Transduced LOXIN localized to the plasma membrane and blocked ox-LDL uptake mediated by LOX-1. Overexpression of LOXIN protected hEPC from ox-LDL-induced apoptosis, and therefore maybe a novel way of improving hEPC function and quantity. These results suggest that adenoviral vectors of LOXIN may provide a possible treatment for diseases related to ox-LDL and vascular endothelium dysfunction, including atherosclerosis.
Collapse
|
41
|
Panes O, González C, Hidalgo P, Valderas JP, Acevedo M, Contreras S, Sánchez X, Pereira J, Rigotti A, Mezzano D. Platelet tissue factor activity and membrane cholesterol are increased in hypercholesterolemia and normalized by rosuvastatin, but not by atorvastatin. Atherosclerosis 2016; 257:164-171. [PMID: 28142075 DOI: 10.1016/j.atherosclerosis.2016.12.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/04/2016] [Accepted: 12/15/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND AIMS High plasma LDL-cholesterol (LDL-C) and platelet responses have major pathogenic roles in atherothrombosis. Thus, statins and anti-platelet drugs constitute mainstays in cardiovascular prevention/treatment. However, the role of platelet tissue factor-dependent procoagulant activity (TF-PCA) has remained unexplored in hypercholesterolemia. We aimed to study platelet TF-PCA and its relationship with membrane cholesterol in vitro and in 45 hypercholesterolemic patients (HC-patients) (LDL-C >3.37 mmol/L, 130 mg/dL) and 37 control subjects (LDL-C <3.37 mmol/L). The effect of 1-month administration of 80 mg/day atorvastatin (n = 21) and 20 mg/day rosuvastatin (n = 24) was compared. METHODS Platelet TF-PCA was induced by GPIbα activation with VWF-ristocetin. RESULTS Cholesterol-enriched platelets in vitro had augmented aggregation/secretion and platelet FXa generation (1.65-fold increase, p = 0.01). HC-patients had 1.5-, 2.3- and 2.5-fold increases in platelet cholesterol, TF protein and activity, respectively; their platelets had neither hyper-aggregation nor endogenous thrombin generation (ETP). Rosuvastatin, but not atorvastatin, normalized platelet cholesterol, TF protein and FXa generation. It also increased slightly the plasma HDL-C levels, which correlated negatively with TF-PCA. CONCLUSIONS Platelets from HC-patients were not hyper-responsive to low concentrations of classical agonists and had normal PRP-ETP, before and after statin administration. However, washed platelets from HC-patients had increased membrane cholesterol, TF protein and TF-PCA. The platelet TF-dependent PCA was specifically expressed after VWF-induced GPIbα activation. Rosuvastatin, but not atorvastatin treatment, normalized the membrane cholesterol, TF protein and TF-PCA in HC-patients, possibly unveiling a new pleiotropic effect of rosuvastatin. Modulation of platelet TF-PCA may become a novel target to prevent/treat atherothrombosis without increasing bleeding risks.
Collapse
Affiliation(s)
- Olga Panes
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - César González
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Hidalgo
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan P Valderas
- Faculty of Medicine and Odontology, University of Antofagasta, Chile
| | - Mónica Acevedo
- Department of Cardiology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susana Contreras
- Department of Nutrition and Metabolic Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ximena Sánchez
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jaime Pereira
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Attilio Rigotti
- Department of Nutrition and Metabolic Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diego Mezzano
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
42
|
Chu YC, Huang KT. CRP/oxLDL co-incubates impair endothelial functions through CD32, LOX-1, and keratin 1 with dependence on their ratio. J Taiwan Inst Chem Eng 2016. [DOI: 10.1016/j.jtice.2016.03.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
43
|
Vindigni G, Raniolo S, Ottaviani A, Falconi M, Franch O, Knudsen BR, Desideri A, Biocca S. Receptor-Mediated Entry of Pristine Octahedral DNA Nanocages in Mammalian Cells. ACS NANO 2016; 10:5971-9. [PMID: 27214742 DOI: 10.1021/acsnano.6b01402] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
DNA offers excellent programming properties for the generation of nanometer-scaled polyhedral structures with a broad variety of potential applications. Translation to biomedical applications requires improving stability in biological fluids, efficient and selective cell binding, and/or internalization of the assembled DNA nanostructures. Here, we report an investigation on the selective mechanism of cellular uptake of pristine DNA nanocages in cells expressing the receptor "oxidized low-density lipoprotein receptor-1" (LOX-1), a scavenger receptor associated with cardiovascular diseases and, more recently, identified as a tumor marker. For this purpose a truncated octahedral DNA nanocage functionalized with a single biotin molecule, which allows DNA cage detection through the biotin-streptavidin assays, was constructed. The results indicate that DNA nanocages are stable in biological fluids, including human serum, and are selectively bound and very efficiently internalized in vesicles only in LOX-1-expressing cells. The amount of internalized cages is 30 times higher in LOX-1-expressing cells than in normal fibroblasts, indicating that the receptor-mediated uptake of pristine DNA nanocages can be pursued for a selective cellular internalization. These results open the route for a therapeutic use of pristine DNA cages targeting LOX-1-overexpressing tumor cells.
Collapse
Affiliation(s)
- Giulia Vindigni
- Department of Systems Medicine, University of Rome Tor Vergata , Via Montpellier 1, 00133, Rome, Italy
| | - Sofia Raniolo
- Department of Systems Medicine, University of Rome Tor Vergata , Via Montpellier 1, 00133, Rome, Italy
| | - Alessio Ottaviani
- Department of Biology, Interuniversity Consortium, National Institute Biostructure and Biosystem (INBB), University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Mattia Falconi
- Department of Biology, Interuniversity Consortium, National Institute Biostructure and Biosystem (INBB), University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | | | | | - Alessandro Desideri
- Department of Biology, Interuniversity Consortium, National Institute Biostructure and Biosystem (INBB), University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Silvia Biocca
- Department of Systems Medicine, University of Rome Tor Vergata , Via Montpellier 1, 00133, Rome, Italy
| |
Collapse
|
44
|
Terabe K, Takahashi N, Takemoto T, Knudson W, Ishiguro N, Kojima T. Simvastatin inhibits CD44 fragmentation in chondrocytes. Arch Biochem Biophys 2016; 604:1-10. [PMID: 27242325 DOI: 10.1016/j.abb.2016.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 11/17/2022]
Abstract
In human osteoarthritic chondrocytes, the hyaluronan receptor CD44 undergoes proteolytic cleavage at the cell surface. CD44 cleavage is thought to require transit of CD44 into cholesterol-rich lipid rafts. The purpose of this study was to investigate whether statins exert a protective effect on articular chondrocytes due to diminution of cholesterol. Three model systems of chondrocytes were examined including human HCS-2/8 chondrosarcoma cells, human osteoarthritic chondrocytes and normal bovine articular chondrocytes. Treatment with IL-1β + Oncostatin M resulted in a substantial increase in CD44 fragmentation in each of the three chondrocyte models. Pre-incubation with simvastatin prior to treatment with IL-1β + Oncostatin M decreased the level of CD44 fragmentation, decreased the proportion of CD44 that transits into the lipid raft fractions, decreased ADAM10 activity and diminished the interaction between CD44 and ADAM10. In HCS-2/8 cells and bovine articular chondrocytes, fragmentation of CD44 was blocked by the knockdown of ADAM10. Inhibition of CD44 fragmentation by simvastatin also resulted in improved retention of pericellular matrix. Addition of cholesterol and farnesyl-pyrophosphate reversed the protective effects of simvastatin. Thus, the addition of simvastatin exerts positive effects on chondrocytes including reduced CD44 fragmentation and enhanced the retention of pericellular matrix.
Collapse
Affiliation(s)
- Kenya Terabe
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC 27834, USA
| | - Nobunori Takahashi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Toki Takemoto
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Warren Knudson
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC 27834, USA
| | - Naoki Ishiguro
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Toshihisa Kojima
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
45
|
Compartmentalization of GPCR signalling controls unique cellular responses. Biochem Soc Trans 2016; 44:562-7. [DOI: 10.1042/bst20150236] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Indexed: 02/08/2023]
Abstract
With >800 members, G protein-coupled receptors (GPCRs) are the largest class of cell-surface signalling proteins, and their activation mediates diverse physiological processes. GPCRs are ubiquitously distributed across all cell types, involved in many diseases and are major drug targets. However, GPCR drug discovery is still characterized by very high attrition rates. New avenues for GPCR drug discovery may be provided by a recent shift away from the traditional view of signal transduction as a simple chain of events initiated from the plasma membrane. It is now apparent that GPCR signalling is restricted to highly organized compartments within the cell, and that GPCRs activate distinct signalling pathways once internalized. A high-resolution understanding of how compartmentalized signalling is controlled will probably provide unique opportunities to selectively and therapeutically target GPCRs.
Collapse
|
46
|
Biocca S, Iacovelli F, Matarazzo S, Vindigni G, Oteri F, Desideri A, Falconi M. Molecular mechanism of statin-mediated LOX-1 inhibition. Cell Cycle 2016; 14:1583-95. [PMID: 25950192 DOI: 10.1080/15384101.2015.1026486] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Statins are largely used in clinics in the treatment of patients with cardiovascular diseases for their effect on lowering circulating cholesterol. Lectin-like oxidized low-density lipoprotein (LOX-1), the primary receptor for ox-LDL, plays a central role in the pathogenesis of atherosclerosis and cardiovascular disorders. We have recently shown that chronic exposure of cells to lovastatin disrupts LOX-1 receptor cluster distribution in plasma membranes, leading to a marked loss of LOX-1 function. Here we investigated the molecular mechanism of statin-mediated LOX-1 inhibition and we demonstrate that all tested statins are able to displace the binding of fluorescent ox-LDL to LOX-1 by a direct interaction with LOX-1 receptors in a cell-based binding assay. Molecular docking simulations confirm the interaction and indicate that statins completely fill the hydrophobic tunnel that crosses the C-type lectin-like (CTLD) recognition domain of LOX-1. Classical molecular dynamics simulation technique applied to the LOX-1 CTLD, considered in the entire receptor structure with or without a statin ligand inside the tunnel, indicates that the presence of a ligand largely increases the dimer stability. Electrophoretic separation and western blot confirm that different statins binding stabilize the dimer assembly of LOX-1 receptors in vivo. The simulative and experimental results allow us to propose a CTLD clamp motion, which enables the receptor-substrate coupling. These findings reveal a novel and significant functional effect of statins.
Collapse
Key Words
- Ato, atorvastatin
- CTLD, C-type lectin-like domain
- Cav-1, caveolin-1
- DMEM, Dulbecco's modified Eagle's medium
- DiI, 1,1′-dioctadecyl-3,3,3′,3′-tetramethyllindocarbocyanine perchlorate
- Flu, fluvastatin
- HEK, human embryonic kidney
- HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A
- LDL, low-density lipoprotein
- LDL-C, low-density lipoprotein-cholesterol
- LOX-1 receptor
- LOX-1, lectin-like oxidized low-density lipoprotein receptor-1
- Lov, lovastatin
- Mab, monoclonal antibody
- Pra, pravastatin
- molecular docking
- molecular dynamics simulation
- monomer-dimer ratio
- ox-LDL, oxidized low-density lipoprotein
- statin
- substrate recognition
Collapse
Affiliation(s)
- Silvia Biocca
- a Department of Systems Medicine and Center of Biostatistics and Bioinformatics ; University of Rome Tor Vergata ; Rome , Italy
| | | | | | | | | | | | | |
Collapse
|
47
|
Fechtner T, Galle JN, Hegemann JH. The novel chlamydial adhesin CPn0473 mediates the lipid raft-dependent uptake of Chlamydia pneumoniae. Cell Microbiol 2016; 18:1094-105. [PMID: 26780295 PMCID: PMC5067637 DOI: 10.1111/cmi.12569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/21/2015] [Accepted: 01/04/2016] [Indexed: 01/23/2023]
Abstract
Chlamydiae are Gram‐negative, obligate intracellular pathogens that pose a serious threat to public health worldwide. Chlamydial surface molecules are essential for host cell invasion. The first interaction with the host cell is thereby accomplished by the Outer membrane complex protein B (OmcB) binding to heparan sulfate moieties on the host cell surface, followed by the interaction of the chlamydial polymorphic membrane proteins (Pmps) with host cell receptors. Specifically, the interaction of the Pmp21 adhesin and invasin with its human interaction partner, the epidermal growth factor receptor, results in receptor activation, down‐stream signalling and finally internalization of the bacteria. Blocking both, the OmcB and Pmp21 adhesion pathways, did not completely abolish infection, suggesting the presence of additional factors relevant for host cell invasion. Here, we show that the novel surface protein CPn0473 of Chlamydia pneumoniae contributes to the binding and invasion of infectious chlamydial particles. CPn0473 is expressed late in the infection cycle and located on the infectious chlamydial cell surface. Soluble recombinant CPn0473 as well as rCPn0473‐coupled fluorescent latex beads adhere to human epithelial HEp‐2 cells. Interestingly, in classical infection blocking experiments pretreatment of HEp‐2 cells with rCPn0473 does not attenuate adhesion but promotes dose‐dependently internalization by C. pneumoniae suggesting an unusual mode of action for this adhesin. This CPn0473‐dependent promotion of infection by C. pneumoniae depends on two different domains within the protein and requires intact lipid rafts. Thus, inhibition of the interaction of CPn0473 with the host cell could provide a way to reduce the virulence of C. pneumoniae.
Collapse
Affiliation(s)
- Tim Fechtner
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Jan N Galle
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Johannes H Hegemann
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| |
Collapse
|
48
|
Kilbride P, Woodward HJ, Tan KB, Thanh NTK, Chu KME, Minogue S, Waugh MG. Modeling the effects of cyclodextrin on intracellular membrane vesicles from Cos-7 cells prepared by sonication and carbonate treatment. PeerJ 2015; 3:e1351. [PMID: 26528413 PMCID: PMC4627923 DOI: 10.7717/peerj.1351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/05/2015] [Indexed: 01/24/2023] Open
Abstract
Cholesterol has important functions in the organization of membrane structure and this may be mediated via the formation of cholesterol-rich, liquid-ordered membrane microdomains often referred to as lipid rafts. Methyl-beta-cyclodextrin (cyclodextrin) is commonly used in cell biology studies to extract cholesterol and therefore disrupt lipid rafts. However, in this study we reassessed this experimental strategy and investigated the effects of cyclodextrin on the physical properties of sonicated and carbonate-treated intracellular membrane vesicles isolated from Cos-7 fibroblasts. We treated these membranes, which mainly originate from the trans-Golgi network and endosomes, with cyclodextrin and measured the effects on their equilibrium buoyant density, protein content, represented by the palmitoylated protein phosphatidylinositol 4-kinase type IIα, and cholesterol. Despite the reduction in mass stemming from cholesterol removal, the vesicles became denser, indicating a possible large volumetric decrease, and this was confirmed by measurements of hydrodynamic vesicle size. Subsequent mathematical analyses demonstrated that only half of this change in membrane size was attributable to cholesterol loss. Hence, the non-selective desorption properties of cyclodextrin are also involved in membrane size and density changes. These findings may have implications for preceding studies that interpreted cyclodextrin-induced changes to membrane biochemistry in the context of lipid raft disruption without taking into account our finding that cyclodextrin treatment also reduces membrane size.
Collapse
Affiliation(s)
- Peter Kilbride
- UCL Institute for Liver & Digestive Health, University College London , London , United Kingdom
| | - Holly J Woodward
- UCL Institute for Liver & Digestive Health, University College London , London , United Kingdom
| | - Kuan Boone Tan
- Biophysics Group, Department of Physics & Astronomy, University College London , London , United Kingdom
| | - Nguyễn T K Thanh
- Biophysics Group, Department of Physics & Astronomy, University College London , London , United Kingdom
| | - K M Emily Chu
- UCL Institute for Liver & Digestive Health, University College London , London , United Kingdom
| | - Shane Minogue
- UCL Institute for Liver & Digestive Health, University College London , London , United Kingdom
| | - Mark G Waugh
- UCL Institute for Liver & Digestive Health, University College London , London , United Kingdom
| |
Collapse
|
49
|
Gioia M, Vindigni G, Testa B, Raniolo S, Fasciglione GF, Coletta M, Biocca S. Membrane Cholesterol Modulates LOX-1 Shedding in Endothelial Cells. PLoS One 2015; 10:e0141270. [PMID: 26495844 PMCID: PMC4619672 DOI: 10.1371/journal.pone.0141270] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/05/2015] [Indexed: 11/19/2022] Open
Abstract
The lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a scavenger receptor responsible for ox-LDL recognition, binding and internalization, which is up-regulated during atherogenesis. Its activation triggers endothelium dysfunction and induces inflammation. A soluble form of LOX-1 has been identified in the human blood and its presence considered a biomarker of cardiovascular diseases. We recently showed that cholesterol-lowering drugs inhibit ox-LDL binding and internalization, rescuing the ox-LDL induced apoptotic phenotype in primary endothelial cells. Here we have investigated the molecular bases of human LOX-1 shedding by metalloproteinases and the role of cell membrane cholesterol on the regulation of this event by modulating its level with MβCD and statins. We report that membrane cholesterol affects the release of different forms of LOX-1 in cells transiently and stably expressing human LOX-1 and in a human endothelial cell line (EA.hy926). In particular, our data show that i) cholesterol depletion triggers the release of LOX-1 in exosomes as a full-length transmembrane isoform and as a truncated ectodomain soluble fragment (sLOX-1); ii) endothelial cells secrete a soluble metalloproteinase which induces LOX-1 ectodomain shedding and iii) long term statins treatment enhances sLOX-1 proteolytic shedding.
Collapse
Affiliation(s)
- Magda Gioia
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Giulia Vindigni
- Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133, Rome, Italy
| | - Barbara Testa
- Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133, Rome, Italy
| | - Sofia Raniolo
- Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133, Rome, Italy
| | - Giovanni Francesco Fasciglione
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Massimiliano Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Silvia Biocca
- Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133, Rome, Italy
| |
Collapse
|
50
|
|