1
|
Sivakumar P, Saul M, Robinson D, King LE, Amin NB. SomaLogic proteomics reveals new biomarkers and provides mechanistic, clinical insights into Acetyl coA Carboxylase (ACC) inhibition in Non-alcoholic Steatohepatitis (NASH). Sci Rep 2024; 14:17072. [PMID: 39048608 PMCID: PMC11269579 DOI: 10.1038/s41598-024-67843-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
Non-alcoholic Fatty Liver Disease (NAFLD) and Non-alcoholic Steatohepatitis (NASH) are major metabolic diseases with increasing global prevalence and no approved therapies. There is a mounting need to develop biomarkers of diagnosis, prognosis and treatment response that can effectively replace current requirements for liver biopsies, which are invasive, error-prone and expensive. We performed SomaLogic serum proteome profiling with baseline (n = 231) and on-treatment (n = 72, Weeks 12 and 16, Placebo and 25 mg PF-05221304) samples from a Phase 2a trial (NCT03248882) with Clesacostat (PF-05221304), an acetyl coA carboxylase inhibitor (ACCi) in patients with NAFLD/NASH. SomaSignal NASH probability scores and expression data for 7000+ analytes were analyzed to identify potential biomarkers associated with baseline clinical measures of NAFLD/NASH [Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF), alanine aminotransferase (ALT) and aspartate aminotransferase (AST)] as well as biomarkers of treatment response to ACCi. SomaSignal NASH probability scores identified biopsy-proven/clinically defined NIT-based (Presumed) NASH classification of the cohort with > 70% agreement. Clesacostat-induced reduction in steatosis probability scores aligned with observed clinical reduction in hepatic steatosis based on MRI-PDFF. We identify a set of 69 analytes that robustly correlate with clinical measures of hepatic inflammation and steatosis (MRI-PDFF, ALT and AST), 27 of which were significantly reversed with ACC inhibition. Clesacostat treatment dramatically upregulated Wnt5a protein and Apolipoproteins C3 and E, with drug-induced changes significantly correlating to changes on MRI-PDFF. Our data demonstrate the utility of SomaLogic- analyte panel for diagnosis and treatment response in NAFLD/NASH and provide potential new mechanistic insights into liver steatosis reduction, inflammation and serum triglyceride elevation with ACC inhibition. (Clinical Trial Identifier: NCT03248882).
Collapse
Affiliation(s)
- Pitchumani Sivakumar
- Translational Clinical Sciences, Pfizer Research and Development, 500 Arcola Road, Collegeville, PA, 19426, USA.
| | - Michelle Saul
- Translational Biomarker Statistics, Pfizer Research and Development, San Diego, USA
| | - Douglas Robinson
- Translational Biomarker Statistics, Pfizer Research and Development, San Diego, USA
| | - Lindsay E King
- Clinical Bioanalytics, Pfizer Research and Development, Cambridge, USA
| | - Neeta B Amin
- Internal Medicine, Pfizer Research and Development, Cambridge, USA
| |
Collapse
|
2
|
Wang J, Parajuli N, Wang Q, Khalasawi N, Peng H, Zhang J, Yin C, Mi QS, Zhou L. MiR-23a Regulates Skin Langerhans Cell Phagocytosis and Inflammation-Induced Langerhans Cell Repopulation. BIOLOGY 2023; 12:925. [PMID: 37508356 PMCID: PMC10376168 DOI: 10.3390/biology12070925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/04/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023]
Abstract
Langerhans cells (LCs) are skin-resident macrophage that act similarly to dendritic cells for controlling adaptive immunity and immune tolerance in the skin, and they are key players in the development of numerous skin diseases. While TGF-β and related downstream signaling pathways are known to control numerous aspects of LC biology, little is known about the epigenetic signals that coordinate cell signaling during LC ontogeny, maintenance, and function. Our previous studies in a total miRNA deletion mouse model showed that miRNAs are critically involved in embryonic LC development and postnatal LC homeostasis; however, the specific miRNA(s) that regulate LCs remain unknown. miR-23a is the first member of the miR-23a-27a-24-2 cluster, a direct downstream target of PU.1 and TGF-b, which regulate the determination of myeloid versus lymphoid fates. Therefore, we used a myeloid-specific miR-23a deletion mouse model to explore whether and how miR-23a affects LC ontogeny and function in the skin. We observed the indispensable role of miR-23a in LC antigen uptake and inflammation-induced LC epidermal repopulation; however, embryonic LC development and postnatal homeostasis were not affected by cells lacking miR23a. Our results suggest that miR-23a controls LC phagocytosis by targeting molecules that regulate efferocytosis and endocytosis, whereas miR-23a promotes homeostasis in bone marrow-derived LCs that repopulate the skin after inflammatory insult by targeting Fas and Bcl-2 family proapoptotic molecules. Collectively, the context-dependent regulatory role of miR-23a in LCs represents an extra-epigenetic layer that incorporates TGF-b- and PU.1-mediated regulation during steady-state and inflammation-induced repopulation.
Collapse
Affiliation(s)
- Jie Wang
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Nirmal Parajuli
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Qiyan Wang
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Namir Khalasawi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Hongmei Peng
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Jun Zhang
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Congcong Yin
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Biochemistry, Microbiology and Immunology, School of Medicine, Wayne State University, Detroit, MI 48202, USA
- Department of Internal Medicine, Henry Ford Health, Detroit, MI 48202, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (J.W.); (N.P.); (Q.W.); (C.Y.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Biochemistry, Microbiology and Immunology, School of Medicine, Wayne State University, Detroit, MI 48202, USA
- Department of Internal Medicine, Henry Ford Health, Detroit, MI 48202, USA
| |
Collapse
|
3
|
Wu TY, Tien N, Lin CL, Cheah YC, Hsu CY, Tsai FJ, Fang YJ, Lim YP. Influence of antipsychotic medications on hyperlipidemia risk in patients with schizophrenia: evidence from a population-based cohort study and in vitro hepatic lipid homeostasis gene expression. Front Med (Lausanne) 2023; 10:1137977. [PMID: 37425327 PMCID: PMC10324036 DOI: 10.3389/fmed.2023.1137977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/02/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Schizophrenia increases the risk of mortality and cardiovascular disease (CVD) risk. However, the correlation between antipsychotics (APs) and CVD remains controversial. Hyperlipidemia is a significant risk factor for CVD. Methods We conducted a nationwide population-based retrospective cohort study to investigate the effects of APs on the risk of hyperlipidemia and lipid homeostasis gene expression. We used data from the Longitudinal Health Insurance Database of Taiwan on new-onset schizophrenia patients and a comparison cohort without schizophrenia. We used a Cox proportional hazards regression model to analyze the differences in hyperlipidemia development between the two cohorts. Furthermore, we examined the effects of APs on the hepatic expression of lipid homeostasis-related genes. Results After adjusting for potential interrelated confounding factors, the case group (N = 4,533) was found to have a higher hyperlipidemia risk than the control cohort (N = 4,533) [adjusted hazard ratio (aHR), 1.30, p < 0.001]. Patients with schizophrenia without APs had a significantly higher risk of hyperlipidemia (aHR, 2.16; p < 0.001). However, patients receiving APs had a significantly lower risk of hyperlipidemia than patients not receiving APs (all aHR ≤ 0.42, p < 0.001). First-generation antipsychotics (FGAs) induce the expression of hepatic lipid catabolism genes in an in vitro model. Discussion Patients with schizophrenia had a higher risk of hyperlipidemia than controls; however, compared with non-treated patients, AP users had a lower risk of hyperlipidemia. Early diagnosis and management of hyperlipidemia may help prevent CVD.
Collapse
Affiliation(s)
- Tien-Yuan Wu
- Graduate Institute of Clinical Pharmacy, College of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Pharmacy, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Ni Tien
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Cun Cheah
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chung Y. Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Division of Medical Genetics, China Medical University Children's Hospital, Taichung, Taiwan
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Yi-Jen Fang
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Ph.D. Program in Environmental and Occupational Medicine, College of Medicine, Kaohsiung Medical University and National Health Research Institutes, Kaohsiung, Taiwan
- Department of Environmental Health, Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung-Hsing University, Taichung, Taiwan
- Digestive Disease Center, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Yun-Ping Lim
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Mahdinia E, Shokri N, Taheri AT, Asgharzadeh S, Elahimanesh M, Najafi M. Cellular crosstalk in atherosclerotic plaque microenvironment. Cell Commun Signal 2023; 21:125. [PMID: 37254185 DOI: 10.1186/s12964-023-01153-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/28/2023] [Indexed: 06/01/2023] Open
Abstract
Atherosclerosis is an underlying pathology of many vascular diseases as a result of cellular, structural and molecular dysfunctions within the sub-endothelial space. This review deals with the events involved in the formation, growth and remodeling of plaque, including the cell recruitment, cell polarization, and cell fat droplets. It also describes cross talking between endothelial cells, macrophages, and vascular smooth muscle cells, as well as the cellular pathways involved in plaque development in the plaque microenvironment. Finally, it describes the plaque structural components and the role of factors involved in the rupture and erosion of plaques in the vessel. Video Abstract.
Collapse
Affiliation(s)
- Elmira Mahdinia
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Shokri
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolkarim Talebi Taheri
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Asgharzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Ghazvin University of Medical Sciences, Ghazvin, Iran
| | - Mohammad Elahimanesh
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Microbial Biotechnology Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Norda S, Papadantonaki R. Regulation of cells of the arterial wall by hypoxia and its role in the development of atherosclerosis. VASA 2023; 52:6-21. [PMID: 36484144 DOI: 10.1024/0301-1526/a001044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cell's response to hypoxia depends on stabilization of the hypoxia-inducible factor 1 complex and transactivation of nuclear factor kappa-B (NF-κB). HIF target gene transcription in cells resident to atherosclerotic lesions adjoins a complex interplay of cytokines and mediators of inflammation affecting cholesterol uptake, migration, and inflammation. Maladaptive activation of the HIF-pathway and transactivation of nuclear factor kappa-B causes monocytes to invade early atherosclerotic lesions, maintaining inflammation and aggravating a low-oxygen environment. Meanwhile HIF-dependent upregulation of the ATP-binding cassette transporter ABCA1 causes attenuation of cholesterol efflux and ultimately macrophages becoming foam cells. Hypoxia facilitates neovascularization by upregulation of vascular endothelial growth factor (VEGF) secreted by endothelial cells and vascular smooth muscle cells lining the arterial wall destabilizing the plaque. HIF-knockout animal models and inhibitor studies were able to show beneficial effects on atherogenesis by counteracting the HIF-pathway in the cell wall. In this review the authors elaborate on the up-to-date literature on regulation of cells of the arterial wall through activation of HIF-1α and its effect on atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Stephen Norda
- Department of Cardiovascular Medicine, University Hospital Münster, Germany
| | - Rosa Papadantonaki
- Emergency Department, West Middlesex University Hospital, Chelsea and Westminster NHS Trust, London, United Kingdom
| |
Collapse
|
6
|
Estrada-Valencia R, de Lima ME, Colonnello A, Rangel-López E, Saraiva NR, de Ávila DS, Aschner M, Santamaría A. The Endocannabinoid System in Caenorhabditis elegans. Rev Physiol Biochem Pharmacol 2023; 184:1-31. [PMID: 34401955 PMCID: PMC8850531 DOI: 10.1007/112_2021_64] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The existence of a formal Endocannabinoid System in C. elegans has been questioned due to data showing the absence of typical cannabinoid receptors in the worm; however, the presence of a full metabolism for endocannabinoids, alternative ligands, and receptors for these agents and a considerable number of orthologous and homologous genes regulating physiological cannabinoid-like signals and responses - several of which are similar to those of mammals - demonstrates a well-structured and functional complex system in nematodes. In this review, we describe and compare similarities and differences between the Endocannabinoid System in mammals and nematodes, highlighting the basis for the integral study of this novel system in the worm.
Collapse
Affiliation(s)
| | - María Eduarda de Lima
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Aline Colonnello
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Edgar Rangel-López
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Nariani Rocha Saraiva
- Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans, UNIPAMPA, Uruguaiana, Brazil
| | - Daiana Silva de Ávila
- Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans, UNIPAMPA, Uruguaiana, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico.
| |
Collapse
|
7
|
Baraniecki Ł, Tokarz-Deptuła B, Syrenicz A, Deptuła W. Macrophage efferocytosis in atherosclerosis. Scand J Immunol 2022; 97:e13251. [PMID: 36583598 DOI: 10.1111/sji.13251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/17/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022]
Abstract
This paper presents the role of macrophage efferocytosis, the process of elimination of apoptotic bodies-elements formed during vascular atherosclerosis. The mechanisms of macrophage efferocytosis are presented, introducing the specific signals of this process, that is, 'find me', 'eat me' and 'don't eat me'. The role of the process of efferocytosis in the formation of vascular atherosclerosis is also presented, including the factors and mechanisms that determine it, as well as the factors that determine the maintenance of homeostasis in the vessels, including the formation of vascular atherosclerosis.
Collapse
Affiliation(s)
| | | | - Anhelli Syrenicz
- Department of Endocrinology, Metabolic Diseases and Internal Diseases, Pomeranian Medical University, Szczecin, Poland
| | - Wiesław Deptuła
- Faculty of Biological and Veterinary Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
8
|
Meszaros M, Bikov A. Obstructive Sleep Apnoea and Lipid Metabolism: The Summary of Evidence and Future Perspectives in the Pathophysiology of OSA-Associated Dyslipidaemia. Biomedicines 2022; 10:2754. [PMID: 36359273 PMCID: PMC9687681 DOI: 10.3390/biomedicines10112754] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
Obstructive sleep apnoea (OSA) is associated with cardiovascular and metabolic comorbidities, including hypertension, dyslipidaemia, insulin resistance and atherosclerosis. Strong evidence suggests that OSA is associated with an altered lipid profile including elevated levels of triglyceride-rich lipoproteins and decreased levels of high-density lipoprotein (HDL). Intermittent hypoxia; sleep fragmentation; and consequential surges in the sympathetic activity, enhanced oxidative stress and systemic inflammation are the postulated mechanisms leading to metabolic alterations in OSA. Although the exact mechanisms of OSA-associated dyslipidaemia have not been fully elucidated, three main points have been found to be impaired: activated lipolysis in the adipose tissue, decreased lipid clearance from the circulation and accelerated de novo lipid synthesis. This is further complicated by the oxidisation of atherogenic lipoproteins, adipose tissue dysfunction, hormonal changes, and the reduced function of HDL particles in OSA. In this comprehensive review, we summarise and critically evaluate the current evidence about the possible mechanisms involved in OSA-associated dyslipidaemia.
Collapse
Affiliation(s)
- Martina Meszaros
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary
| | - Andras Bikov
- North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester M13 9MT, UK
| |
Collapse
|
9
|
Warde KM, Lim YJ, Ribes Martinez E, Beuschlein F, O'Shea P, Hantel C, Dennedy MC. Mitotane Targets Lipid Droplets to Induce Lipolysis in Adrenocortical Carcinoma. Endocrinology 2022; 163:6633639. [PMID: 35797592 PMCID: PMC9342684 DOI: 10.1210/endocr/bqac102] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Adrenocortical carcinoma (ACC) is a rare aggressive cancer with low overall survival. Adjuvant mitotane improves survival but is limited by poor response rates and resistance. Mitotane's efficacy is attributed to the accumulation of toxic free cholesterol, predominantly through cholesterol storage inhibition. However, targeting this pathway has proven unsuccessful. We hypothesize that mitotane-induced free-cholesterol accumulation is also mediated through enhanced breakdown of lipid droplets. METHODOLOGY ATCC-H295R (mitotane-sensitive) and MUC-1 (mitotane-resistant) ACC cells were evaluated for lipid content using specific BODIPY dyes. Protein expression was evaluated by immunoblotting and flow cytometry. Cell viability was measured by quantifying propidium iodide-positive cells following mitotane treatment and pharmacological inhibitors of lipolysis. RESULTS H295R and MUC-1 cells demonstrated similar neutral lipid droplet numbers at baseline. However, evaluation of lipid machinery demonstrated distinct profiles in each model. Analysis of intracellular lipid droplet content showed H295R cells preferentially store cholesteryl esters, whereas MUC-1 cells store triacylglycerol. Decreased lipid droplets were associated with increased lipolysis in H295R and in MUC-1 at toxic mitotane concentrations. Pharmacological inhibition of lipolysis attenuated mitotane-induced toxicity in both models. CONCLUSION We highlight that lipid droplet breakdown and activation of lipolysis represent a putative additional mechanism for mitotane-induced cytotoxicity in ACC. Further understanding of cholesterol and lipids in ACC offers potential novel therapeutic exploitation, especially in mitotane-resistant disease.
Collapse
Affiliation(s)
- Kate M Warde
- Discipline of Pharmacology and Therapeutics, National University of Ireland, Galway, H91 TK33, Ireland
| | - Yi Jan Lim
- Discipline of Pharmacology and Therapeutics, National University of Ireland, Galway, H91 TK33, Ireland
| | - Eduardo Ribes Martinez
- Discipline of Pharmacology and Therapeutics, National University of Ireland, Galway, H91 TK33, Ireland
| | - Felix Beuschlein
- Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich, 81377, Germany
- Department of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, Zurich 8091, Switzerland
| | - Paula O'Shea
- Department of Clinical Biochemistry, Galway University Hospitals, Saolta Hospitals Group, Newcastle Road, Galway, H91 RW28, Ireland
| | - Constanze Hantel
- Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich, 81377, Germany
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Germany
| | - Michael Conall Dennedy
- Discipline of Pharmacology and Therapeutics, National University of Ireland, Galway, H91 TK33, Ireland
| |
Collapse
|
10
|
Garcia-Arcos I, Park SS, Mai M, Alvarez-Buve R, Chow L, Cai H, Baumlin-Schmid N, Agudelo CW, Martinez J, Kim MD, Dabo AJ, Salathe M, Goldberg IJ, Foronjy RF. LRP1 loss in airway epithelium exacerbates smoke-induced oxidative damage and airway remodeling. J Lipid Res 2022; 63:100185. [PMID: 35202607 PMCID: PMC8953659 DOI: 10.1016/j.jlr.2022.100185] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
The LDL receptor-related protein 1 (LRP1) partakes in metabolic and signaling events regulated in a tissue-specific manner. The function of LRP1 in airways has not been studied. We aimed to study the function of LRP1 in smoke-induced disease. We found that bronchial epithelium of patients with chronic obstructive pulmonary disease and airway epithelium of mice exposed to smoke had increased LRP1 expression. We then knocked out LRP1 in human bronchial epithelial cells in vitro and in airway epithelial club cells in mice. In vitro, LRP1 knockdown decreased cell migration and increased transforming growth factor β activation. Tamoxifen-inducible airway-specific LRP1 knockout mice (club Lrp1-/-) induced after complete lung development had increased inflammation in the bronchoalveolar space and lung parenchyma at baseline. After 6 months of smoke exposure, club Lrp1-/- mice showed a combined restrictive and obstructive phenotype, with lower compliance, inspiratory capacity, and forced expiratory volume0.05/forced vital capacity than WT smoke-exposed mice. This was associated with increased values of Ashcroft fibrotic index. Proteomic analysis of room air exposed-club Lrp1-/- mice showed significantly decreased levels of proteins involved in cytoskeleton signaling and xenobiotic detoxification as well as decreased levels of glutathione. The proteome fingerprint created by smoke eclipsed many of the original differences, but club Lrp1-/- mice continued to have decreased lung glutathione levels and increased protein oxidative damage and airway cell proliferation. Therefore, LRP1 deficiency leads to greater lung inflammation and damage and exacerbates smoke-induced lung disease.
Collapse
Affiliation(s)
- Itsaso Garcia-Arcos
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA.
| | - Sangmi S Park
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Michelle Mai
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Roger Alvarez-Buve
- Respiratory Department, Hospital University Arnau de Vilanova and Santa Maria, IRB Lleida, University of Lleida, Lleida, Catalonia, Spain
| | - Lillian Chow
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Huchong Cai
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | | | - Christina W Agudelo
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Jennifer Martinez
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Michael D Kim
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Abdoulaye J Dabo
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Matthias Salathe
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ira J Goldberg
- Department of Medicine, NYU Langone School of Medicine, New York, NY, USA
| | - Robert F Foronjy
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| |
Collapse
|
11
|
Control of Cholesterol Metabolism Using a Systems Approach. BIOLOGY 2022; 11:biology11030430. [PMID: 35336806 PMCID: PMC8945167 DOI: 10.3390/biology11030430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 11/25/2022]
Abstract
Simple Summary Cholesterol is the main sterol in mammals that is essential for healthy cell functionining. It plays a key role in metabolic regulation and signaling, it is a precursor molecule of bile acids, oxysterols, and all steroid hormones. It also contributes to the structural makeup of the membranes. Its homeostasis is tightly controlled since it can harm the body if it is allowed to reach abnormal blood concentrations. One of the diseases associated with elevated cholesterol levels being the major cause of morbidities and mortalities worldwide, is atherosclerosis. In this study, we have developed a model of the cholesterol metabolism taking into account local inflammation and oxidative stress. The aim was to investigate the impact of the interplay of those processes and cholesterol metabolism disturbances on the atherosclerosis development and progression. We have also analyzed the effect of combining different classes of drugs targeting selected components of cholesterol metabolism. Abstract Cholesterol is an essential component of mammalian cells and is involved in many fundamental physiological processes; hence, its homeostasis in the body is tightly controlled, and any disturbance has serious consequences. Disruption of the cellular metabolism of cholesterol, accompanied by inflammation and oxidative stress, promotes the formation of atherosclerotic plaques and, consequently, is one of the leading causes of death in the Western world. Therefore, new drugs to regulate disturbed cholesterol metabolism are used and developed, which help to control cholesterol homeostasis but still do not entirely cure atherosclerosis. In this study, a Petri net-based model of human cholesterol metabolism affected by a local inflammation and oxidative stress, has been created and analyzed. The use of knockout of selected pathways allowed us to observe and study the effect of various combinations of commonly used drugs on atherosclerosis. The analysis results led to the conclusion that combination therapy, targeting multiple pathways, may be a fundamental concept in the development of more effective strategies for the treatment and prevention of atherosclerosis.
Collapse
|
12
|
Xie B, Njoroge W, Dowling LM, Sulé-Suso J, Cinque G, Yang Y. Detection of lipid efflux from foam cell models using a label-free infrared method. Analyst 2022; 147:5372-5385. [DOI: 10.1039/d2an01041k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Synchrotron-based microFTIR spectroscopy was used to study the process of lipid efflux in a foam cell model. The anti-atherosclerotic drug, atorvastatin, removed low-density lipoprotein from the foam cells in a dose, and time dependent manner.
Collapse
Affiliation(s)
- Bowen Xie
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Wanjiku Njoroge
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Lewis M. Dowling
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Josep Sulé-Suso
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
- Oncology Department, Cancer Centre, University Hospitals of North Midlands, Stoke-on-Trent, ST4 6QG, UK
| | - Gianfelice Cinque
- MIRIAM beamline B22, Diamond Light Source, Harwell Science and Innovation Campus, Chilton-Didcot OX11 0DE, UK
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
| |
Collapse
|
13
|
Biomediators in Polycystic Ovary Syndrome and Cardiovascular Risk. Biomolecules 2021; 11:biom11091350. [PMID: 34572562 PMCID: PMC8467803 DOI: 10.3390/biom11091350] [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: 07/29/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is extremely heterogeneous in terms of clinical manifestations. The variability of the syndrome's phenotype is derived from the genetic and molecular heterogeneity, with a great deal of environmental factors that may have long-term health consequences, such as metabolic and cardiovascular (CV) diseases. There is no doubt that women with PCOS suffer from metabolic complications more than their age-matched counterparts in the general population and at an earlier age. Obesity, low steroid hormone-binding globulin (SHBG), hyperandrogenemia, insulin resistance, and compensatory hyperinsulinemia are biomediators and early predictors of metabolic complications in PCOS. Doubts remain about the real risk of CV diseases in PCOS and the molecular mechanisms at the basis of CV complications. Based on that assumption, this review will present the available evidence on the potential implications of some biomediators, in particular, hyperandrogenism, estrogen-progesterone imbalance, insulin resistance, and low SHBG, in the processes leading to CV disease in PCOS, with the final aim to propose a more accurate CV risk assessment.
Collapse
|
14
|
Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: A pathological or physiological phenomenon? Acta Neuropathol Commun 2021; 9:149. [PMID: 34503576 PMCID: PMC8428049 DOI: 10.1186/s40478-021-01246-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss the synaptic aspects of Tau pathology occurring during Alzheimer's disease (AD) and how this may relate to memory impairment, a major hallmark of AD. Whilst the clinical diagnosis of AD patients is a loss of working memory and long-term declarative memory, the histological diagnosis is the presence of neurofibrillary tangles of hyperphosphorylated Tau and Amyloid-beta plaques. Tau pathology spreads through synaptically connected neurons to impair synaptic function preceding the formation of neurofibrillary tangles, synaptic loss, axonal retraction and cell death. Alongside synaptic pathology, recent data suggest that Tau has physiological roles in the pre- or post- synaptic compartments. Thus, we have seen a shift in the research focus from Tau as a microtubule-stabilising protein in axons, to Tau as a synaptic protein with roles in accelerating spine formation, dendritic elongation, and in synaptic plasticity coordinating memory pathways. We collate here the myriad of emerging interactions and physiological roles of synaptic Tau, and discuss the current evidence that synaptic Tau contributes to pathology in AD.
Collapse
|
15
|
Lakpa KL, Khan N, Afghah Z, Chen X, Geiger JD. Lysosomal Stress Response (LSR): Physiological Importance and Pathological Relevance. J Neuroimmune Pharmacol 2021; 16:219-237. [PMID: 33751445 PMCID: PMC8099033 DOI: 10.1007/s11481-021-09990-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/08/2021] [Indexed: 02/08/2023]
Abstract
Extensive work has characterized endoplasmic reticulum (ER) and mitochondrial stress responses. In contrast, very little has been published about stress responses in lysosomes; subcellular acidic organelles that are physiologically important and are of pathological relevance. The greater lysosomal system is dynamic and is comprised of endosomes, lysosomes, multivesicular bodies, autophagosomes, and autophagolysosomes. They are important regulators of cellular physiology, they represent about 5% of the total cellular volume, they are heterogeneous in their sizes and distribution patterns, they are electron dense, and their subcellular positioning within cells varies in response to stimuli, insults and pH. These organelles are also integral to the pathogenesis of lysosomal storage diseases and it is increasingly recognized that lysosomes play important roles in the pathogenesis of such diverse conditions as neurodegenerative disorders and cancer. The purpose of this review is to focus attention on lysosomal stress responses (LSR), compare LSR with better characterized stress responses in ER and mitochondria, and form a framework for future characterizations of LSR. We synthesized data into the concept of LSR and present it here such that the definition of LSR can be modified as new knowledge is added and specific therapeutics are developed.
Collapse
Affiliation(s)
- Koffi L Lakpa
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA
| | - Nabab Khan
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA
| | - Zahra Afghah
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA
| | - Jonathan D Geiger
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA.
| |
Collapse
|
16
|
Chen J, Su Y, Pi S, Hu B, Mao L. The Dual Role of Low-Density Lipoprotein Receptor-Related Protein 1 in Atherosclerosis. Front Cardiovasc Med 2021; 8:682389. [PMID: 34124208 PMCID: PMC8192809 DOI: 10.3389/fcvm.2021.682389] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
Low-density lipoprotein receptor–related protein-1 (LRP1) is a large endocytic and signaling receptor belonging to the LDL receptor (LDLR) gene family and that is widely expressed in several tissues. LRP1 comprises a large extracellular domain (ECD; 515 kDa, α chain) and a small intracellular domain (ICD; 85 kDa, β chain). The deletion of LRP1 leads to embryonic lethality in mice, revealing a crucial but yet undefined role in embryogenesis and development. LRP1 has been postulated to participate in numerous diverse physiological and pathological processes ranging from plasma lipoprotein homeostasis, atherosclerosis, tumor evolution, and fibrinolysis to neuronal regeneration and survival. Many studies using cultured cells and in vivo animal models have revealed the important roles of LRP1 in vascular remodeling, foam cell biology, inflammation and atherosclerosis. However, its role in atherosclerosis remains controversial. LRP1 not only participates in the removal of atherogenic lipoproteins and proatherogenic ligands in the liver but also mediates the uptake of aggregated LDL to promote the formation of macrophage- and vascular smooth muscle cell (VSMC)-derived foam cells, which causes a prothrombotic transformation of the vascular wall. The dual and opposing roles of LRP1 may also represent an interesting target for atherosclerosis therapeutics. This review highlights the influence of LRP1 during atherosclerosis development, focusing on its dual role in vascular cells and immune cells.
Collapse
Affiliation(s)
- Jiefang Chen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Su
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shulan Pi
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
He Z, Wang G, Wu J, Tang Z, Luo M. The molecular mechanism of LRP1 in physiological vascular homeostasis and signal transduction pathways. Biomed Pharmacother 2021; 139:111667. [PMID: 34243608 DOI: 10.1016/j.biopha.2021.111667] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023] Open
Abstract
Interactions between vascular smooth muscle cells (VSMCs), endothelial cells (ECs), pericytes (PCs) and macrophages (MФ), the major components of blood vessels, play a crucial role in maintaining vascular structural and functional homeostasis. Low-density lipoprotein (LDL) receptor-related protein-1 (LRP1), a transmembrane receptor protein belonging to the LDL receptor family, plays multifunctional roles in maintaining endocytosis, homeostasis, and signal transduction. Accumulating evidence suggests that LRP1 modulates vascular homeostasis mainly by regulating vasoactive substances and specific intracellular signaling pathways, including the plasminogen activator inhibitor 1 (PAI-1) signaling pathway, platelet-derived growth factor (PDGF) signaling pathway, transforming growth factor-β (TGF-β) signaling pathway and vascular endothelial growth factor (VEGF) signaling pathway. The aim of the present review is to focus on recent advances in the discovery and mechanism of vascular homeostasis regulated by LRP1-dependent signaling pathways. These recent discoveries expand our understanding of the mechanisms controlling LRP1 as a target for studies on vascular complications.
Collapse
Affiliation(s)
- Zhaohui He
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Wang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Zonghao Tang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
18
|
Lin P, Ji HH, Li YJ, Guo SD. Macrophage Plasticity and Atherosclerosis Therapy. Front Mol Biosci 2021; 8:679797. [PMID: 34026849 PMCID: PMC8138136 DOI: 10.3389/fmolb.2021.679797] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a chronic disease starting with the entry of monocytes into the subendothelium and the subsequent differentiation into macrophages. Macrophages are the major immune cells in atherosclerotic plaques and are involved in the dynamic progression of atherosclerotic plaques. The biological properties of atherosclerotic plaque macrophages determine lesion size, composition, and stability. The heterogenicity and plasticity of atherosclerotic macrophages have been a hotspot in recent years. Studies demonstrated that lipids, cytokines, chemokines, and other molecules in the atherosclerotic plaque microenvironment regulate macrophage phenotype, contributing to the switch of macrophages toward a pro- or anti-atherosclerosis state. Of note, M1/M2 classification is oversimplified and only represent two extreme states of macrophages. Moreover, M2 macrophages in atherosclerosis are not always protective. Understanding the phenotypic diversity and functions of macrophages can disclose their roles in atherosclerotic plaques. Given that lipid-lowering therapy cannot completely retard the progression of atherosclerosis, macrophages with high heterogeneity and plasticity raise the hope for atherosclerosis regression. This review will focus on the macrophage phenotypic diversity, its role in the progression of the dynamic atherosclerotic plaque, and finally discuss the possibility of treating atherosclerosis by targeting macrophage microenvironment.
Collapse
Affiliation(s)
- Ping Lin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Hong-Hai Ji
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Yan-Jie Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
19
|
Padro T, Muñoz-Garcia N, Badimon L. The role of triglycerides in the origin and progression of atherosclerosis. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2021; 33 Suppl 2:20-28. [PMID: 34006350 DOI: 10.1016/j.arteri.2021.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/13/2021] [Indexed: 01/02/2023]
Abstract
Hypertriglyceridaemia has been associated with cardiovascular disease risk in humans for several decades. However, only recently, data from basic research, as well as from genetic and observational studies, have suggested triglyceride-rich lipoproteins (TRLs) as causal factors for atherosclerotic cardiovascular disease. Novel findings highlighting the relevance of TRL-derived lipolytic products (remnant lipoprotein particles "RLPs"), rather than plasma triglycerides or TRL themselves, as the true mediators in atherosclerosis, have contributed to explain a causal relationship through a number of direct and indirect mechanisms. Thus, experimental studies in animal models and in vitro cell culture methods reveal that RLPs, having sizes below 70-80nm, enter the arterial wall and accumulate within the sub-endothelial space. They then become involved in the cholesterol deposition of cholesterol in the intima in addition to several pro-inflammatory and pro-apoptotic pathways. In this review, a summary is presented of current understanding of the pathophysiological mechanisms by which TRLs and their lipolytic derived RLP induce the formation and progression of atherosclerotic lesions, and actively contribute to cardiovascular disease.
Collapse
Affiliation(s)
- Teresa Padro
- Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBERCV Instituto de Salud Carlos III, Barcelona, Spain.
| | - Natalia Muñoz-Garcia
- Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBERCV Instituto de Salud Carlos III, Barcelona, Spain; Cardiovascular Research Chair, UAB, Barcelona, Spain
| |
Collapse
|
20
|
Min H, Xu L, Parrott R, Overall CC, Lillich M, Rabjohns EM, Rampersad RR, Tarrant TK, Meadows N, Fernandez-Castaneda A, Gaultier A, Kurtzberg J, Filiano AJ. Mesenchymal stromal cells reprogram monocytes and macrophages with processing bodies. STEM CELLS (DAYTON, OHIO) 2020; 39:115-128. [PMID: 33166420 DOI: 10.1002/stem.3292] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/28/2020] [Indexed: 11/11/2022]
Abstract
Mesenchymal stromal cells (MSCs) are widely used in clinical trials because of their ability to modulate inflammation. The success of MSCs has been variable over 25 years, most likely due to an incomplete understanding of their mechanism. After MSCs are injected, they traffic to the lungs and other tissues where they are rapidly cleared. Despite being cleared, MSCs suppress the inflammatory response in the long term. Using human cord tissue-derived MSCs (hCT-MSCs), we demonstrated that hCT-MSCs directly interact and reprogram monocytes and macrophages. After engaging hCT-MSCs, monocytes and macrophages engulfed cytoplasmic components of live hCT-MSCs, then downregulated gene programs for antigen presentation and costimulation, and functionally suppressed the activation of helper T cells. We determined that low-density lipoprotein receptor-related proteins on monocytes and macrophages mediated the engulfment of hCT-MSCs. Since a large amount of cellular information can be packaged in cytoplasmic RNA processing bodies (p-bodies), we generated p-body deficient hCT-MSCs and confirmed that they failed to reprogram monocytes and macrophages in vitro and in vivo. hCT-MSCs suppressed an inflammatory response caused by a nasal lipopolysaccharide challenge. Although both control and p-body deficient hCT-MSCs were engulfed by infiltrating lung monocytes and macrophages, p-body deficient hCT-MSCs failed to suppress inflammation and downregulate MHC-II. Overall, we identified a novel mechanism by which hCT-MSCs indirectly suppressed a T-cell response by directly interacting and reprogramming monocytes and macrophages via p-bodies. The results of this study suggest a novel mechanism for how MSCs can reprogram the inflammatory response and have long-term effects to suppress inflammation.
Collapse
Affiliation(s)
- Hyunjung Min
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA.,Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Li Xu
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Roberta Parrott
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Christopher C Overall
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Melina Lillich
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Emily M Rabjohns
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Rishi R Rampersad
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Teresa K Tarrant
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Norin Meadows
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Anthony Fernandez-Castaneda
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Alban Gaultier
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA.,Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Anthony J Filiano
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA.,Department of Neurosurgery, Duke University, Durham, North Carolina, USA.,Department of Immunology, Duke University, Durham, North Carolina, USA.,Department of Pathology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
21
|
Liu G, Zeng M, Li X, Rong Y, Hu Z, Zhang H, Liu X. Expression and analysis of ESR1, IGF-1, FSH, VLDLR, LRP, LH, PRLR genes in Pekin duck and Black Muscovy duck. Gene 2020; 769:145183. [PMID: 33007371 DOI: 10.1016/j.gene.2020.145183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/12/2020] [Accepted: 09/23/2020] [Indexed: 12/27/2022]
Abstract
In order to explore the influence of egg-laying regulatory genes on egg production in ducks at different laying stages, Pekin duck and Black Muscovy duck were used in this study, including early laying stage (20-30 weeks old), peak laying period (31-48 weeks old) and late laying stage (49-66 weeks old). Relative quantitative RT-PCR was used to detect the mRNA transcription level of selected egg-laying regulatory genes in the ovary tissues of ducks at different laying stages. Study shows: during the laying period of Pekin duck, ESR1, LRP1, IGF-1 and LHR were involved in the regulation of egg-laying, and the high expression of LRP1 in the late stage could inhibit egg production. Still, the expression products of the other three genes showed promoting effect. During the laying period of Black Muscovy duck, FSH, VLDLR, IGF-1, PRLR, LHR and LRP1 participated in the regulation of egg-laying, in which the expression products of the first five genes could promote egg production, while LRP1 showed inhibitory effect. Through our experiments, these data will provide strong theoretical support for the breeding of Pekin duck and Black Muscovy duck.
Collapse
Affiliation(s)
- Guangyu Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Mingfei Zeng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Xingxing Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Yu Rong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Zhigang Hu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Huilin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Xiaolin Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China.
| |
Collapse
|
22
|
Yang P, Pi X, Marion TN, Wang J, Wang G, Xie Y, Xie D, Liu Y. Gout inheritance in an extended Chinese family analyzed by whole-exome sequencing: A case-report. Medicine (Baltimore) 2020; 99:e20057. [PMID: 32569156 PMCID: PMC7310917 DOI: 10.1097/md.0000000000020057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 02/06/2020] [Accepted: 03/26/2020] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Gout is a worldwide chronic disease generally caused by high serum levels of uric acid. Using whole exome sequencing, we aimed to explore genetic alterations in hereditary gout. PATIENTS' CONCERNS There were 9 direct descendants diagnosed with gout in total in this family. The patients concerned about the high incidence and inheritance of gout. DIAGNOSIS The youngest propositus was diagnosed as gout in our hospital. Diagnoses of other patients in this family were made on the foundation of history and clinical tests. INTERVENTIONS Six direct descendants and 3 healthy spouses in 1 family were recruited in our study. Whole-exome sequencing was conducted in all participants. OUTCOMES Whole-exome sequencing and genetic analysis revealed 2 putative rare inherited deleterious variants, which were detected only in direct descendants. Twelve gout and uric acid (UC)-related nucleotide sequence variants previously reported by GWAS were detected among all subjects. CONCLUSIONS In the case of this family, the GWAS identified gout and UC-related nucleotide sequence variants may increase the risk of developing gout, but penetrance was not complete. The rare sequence variants in low-density lipoprotein receptor-related protein 1 (LRP1) and oncoprotein induced transcript 3 (OIT3) may have contributed to inheritance of gout within the 5 generations of family members in this study.
Collapse
Affiliation(s)
| | - Xuenan Pi
- Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Tony N. Marion
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee
| | | | - Gang Wang
- Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | | | - Dan Xie
- Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yi Liu
- Department of Rheumatology
| |
Collapse
|
23
|
Yang Y, Wu LN, Chen JF, Wu X, Xia JH, Meng ZN, Liu XC, Lin HR. Whole-genome sequencing of leopard coral grouper ( Plectropomus leopardus) and exploration of regulation mechanism of skin color and adaptive evolution. Zool Res 2020; 41:328-340. [PMID: 32212431 PMCID: PMC7231471 DOI: 10.24272/j.issn.2095-8137.2020.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/25/2020] [Indexed: 01/09/2023] Open
Abstract
Leopard coral groupers belong to the Plectropomus genus of the Epinephelidae family and are important fish for coral reef ecosystems and the marine aquaculture industry. To promote future research of this species, a high-quality chromosome-level genome was assembled using PacBio sequencing and Hi-C technology. A 787.06 Mb genome was assembled, with 99.7% (784.57 Mb) of bases anchored to 24 chromosomes. The leopard coral grouper genome size was smaller than that of other groupers, which may be related to its ancient status among grouper species. A total of 22 317 protein-coding genes were predicted. This high-quality genome of the leopard coral grouper is the first genomic resource for Plectropomus and should provide a pivotal genetic foundation for further research. Phylogenetic analysis of the leopard coral grouper and 12 other fish species showed that this fish is closely related to the brown-marbled grouper. Expanded genes in the leopard coral grouper genome were mainly associated with immune response and movement ability, which may be related to the adaptive evolution of this species to its habitat. In addition, we also identified differentially expressed genes (DEGs) associated with carotenoid metabolism between red and brown-colored leopard coral groupers. These genes may play roles in skin color decision by regulating carotenoid content in these groupers.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Life Sciences School, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Li-Na Wu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Life Sciences School, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Jing-Fang Chen
- State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Xi Wu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Life Sciences School, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Jun-Hong Xia
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Life Sciences School, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
- Southern Laboratory of Ocean Science and Engineering, Zhuhai, Guangdong 519000, China
| | - Zi-Ning Meng
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Life Sciences School, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
- Southern Laboratory of Ocean Science and Engineering, Zhuhai, Guangdong 519000, China. E-mail:
| | - Xiao-Chun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Life Sciences School, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
- Southern Laboratory of Ocean Science and Engineering, Zhuhai, Guangdong 519000, China. E-mail:
| | - Hao-Ran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Life Sciences School, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
- Southern Laboratory of Ocean Science and Engineering, Zhuhai, Guangdong 519000, China
| |
Collapse
|
24
|
Puig N, Montolio L, Camps-Renom P, Navarra L, Jiménez-Altayó F, Jiménez-Xarrié E, Sánchez-Quesada JL, Benitez S. Electronegative LDL Promotes Inflammation and Triglyceride Accumulation in Macrophages. Cells 2020; 9:cells9030583. [PMID: 32121518 PMCID: PMC7140452 DOI: 10.3390/cells9030583] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022] Open
Abstract
Electronegative low-density lipoprotein (LDL) (LDL(−)), a modified LDL that is present in blood and exerts atherogenic effects on endothelial cells and monocytes. This study aimed to determine the action of LDL(−) on monocytes differentiated into macrophages. LDL(−) and in vitro-modified LDLs (oxidized, aggregated, and acetylated) were added to macrophages derived from THP1 monocytes over-expressing CD14 (THP1-CD14). Then, cytokine release, cell differentiation, lipid accumulation, and gene expression were measured by ELISA, flow cytometry, thin-layer chromatography, and real-time PCR, respectively. LDL(−) induced more cytokine release in THP1-CD14 macrophages than other modified LDLs. LDL(−) also promoted morphological changes ascribed to differentiated macrophages. The addition of high-density lipoprotein (HDL) and anti-TLR4 counteracted these effects. LDL(−) was highly internalized by macrophages, and it was the major inductor of intracellular lipid accumulation in triglyceride-enriched lipid droplets. In contrast to inflammation, the addition of anti-TLR4 had no effect on lipid accumulation, thus suggesting an uptake pathway alternative to TLR4. In this regard, LDL(−) upregulated the expression of the scavenger receptors CD36 and LOX-1, as well as several genes involved in triglyceride (TG) accumulation. The importance and novelty of the current study is that LDL(−), a physiologically modified LDL, exerted atherogenic effects in macrophages by promoting differentiation, inflammation, and triglyceride-enriched lipid droplets formation in THP1-CD14 macrophages, probably through different receptors.
Collapse
Affiliation(s)
- Núria Puig
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain; (N.P.); (L.M.); (L.N.)
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Building M, Universitat Autònoma de Barcelona (UAB), 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Lara Montolio
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain; (N.P.); (L.M.); (L.N.)
| | - Pol Camps-Renom
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, and IIB-Sant Pau, 08041 Barcelona, Spain;
| | - Laia Navarra
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain; (N.P.); (L.M.); (L.N.)
| | - Francesc Jiménez-Altayó
- Departament of Pharmacology. Neuroscience Institute. Faculty of Medicine, UAB, 08193 Cerdanyola del Vallès, Barcelona, Spain;
| | - Elena Jiménez-Xarrié
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, and IIB-Sant Pau, 08041 Barcelona, Spain;
- Correspondence: (E.J.-X.); (J.L.S.-Q.); (S.B.); Tel.: +34-93-553-7595 (S.B.)
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain; (N.P.); (L.M.); (L.N.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain
- Correspondence: (E.J.-X.); (J.L.S.-Q.); (S.B.); Tel.: +34-93-553-7595 (S.B.)
| | - Sonia Benitez
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain; (N.P.); (L.M.); (L.N.)
- Correspondence: (E.J.-X.); (J.L.S.-Q.); (S.B.); Tel.: +34-93-553-7595 (S.B.)
| |
Collapse
|
25
|
Lv C, Niu S, Yan S, Bai C, Yu X, Hou J, Gao W, Zhang J, Zhao Z, Yang C, Zhang Y. Low-density lipoprotein receptor-related protein 1 regulates muscle fiber development in cooperation with related genes to affect meat quality. Poult Sci 2019; 98:3418-3425. [PMID: 30982888 DOI: 10.3382/ps/pez168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/13/2019] [Indexed: 12/30/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is an important signal protein that is widely involved in physiological processes, such as lipid metabolism, cell movement, and disease processes. However, the relationship between LRP1 and meat quality remains unknown in chickens. The present study aimed to investigate the correlation between LRP1 and meat quality that builds on our preliminary research, as well as to reveal the underlying molecular mechanism of LRP1 on meat-quality traits. The results showed that LRP1 was significantly correlated with shear force (P < 0.05). Several key genes involved in muscle growth and development, including IGF-1, IGFBP-5, IGF-1R, IGF-2, and MyoD, were down-regulated significantly (P < 0.05 or P < 0.01), and MSTN was up-regulated significantly (P < 0.01) in the presence of LRP1 interference. Cell proliferation- or apoptosis-related genes, including PMP22, CDKN2C, and p53, increased significantly (P < 0.05 or P < 0.01), whereas Bcl-x decreased significantly (P < 0.05) in the RNAi group. We conclude that LRP1 regulates muscle fiber development in cooperation with related genes that affect myoblast proliferation and apoptosis, thereby impacting shear force in chickens. This study will provide a valuable resource for biological investigations of muscle growth and meat-quality-related genes in chickens. The results could be useful in identifying candidate genes that could be used for selective breeding to improve meat quality.
Collapse
Affiliation(s)
- Chao Lv
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| | - Shuling Niu
- College of Animal Science, Jilin University, Changchun 130062, P. R. China.,Department of Animal Science and Technology, Changchun Sci-Tech University, Changchun 130600, P. R. China
| | - Shouqing Yan
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| | - Chunyan Bai
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| | - Xi Yu
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| | - Jiani Hou
- Department of Animal Science and Technology, Changchun Sci-Tech University, Changchun 130600, P. R. China
| | - Wenjing Gao
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| | - Jinyu Zhang
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| | - Zhihui Zhao
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| | - Caini Yang
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| | - Yonghong Zhang
- College of Animal Science, Jilin University, Changchun 130062, P. R. China
| |
Collapse
|
26
|
Vascular Macrophages in Atherosclerosis. J Immunol Res 2019; 2019:4354786. [PMID: 31886303 PMCID: PMC6914912 DOI: 10.1155/2019/4354786] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/19/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is the main pathological basis for the occurrence of most cardiovascular diseases, the leading global health threat, and a great burden for society. It has been well established that atherosclerosis is not only a metabolic disorder but also a chronic, sterile, and maladaptive inflammatory process encompassing both innate and adaptive immunity. Macrophages, the major immune cell population in atherosclerotic lesions, have been shown to play critical roles in all stages of atherosclerosis, including the initiation and progression of advanced atherosclerosis. Macrophages have emerged as a novel potential target for antiatherosclerosis therapy. In addition, the macrophage phenotype is greatly influenced by microenvironmental stimuli in the plaques and presents complex heterogeneity. This article reviews the functions of macrophages in different stages of atherosclerosis, as well as the phenotypes and functions of macrophage subsets. New treatment strategies based on macrophage-related inflammation are also discussed.
Collapse
|
27
|
Ghazaryan A, Landfester K, Mailänder V. Protein deglycosylation can drastically affect the cellular uptake. NANOSCALE 2019; 11:10727-10737. [PMID: 31120044 DOI: 10.1039/c8nr08305c] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Targeted drug delivery mediated by nanocarriers is a major issue in modern-day medicine. Upon coming in contact with biological fluids (e.g. blood), nanocarriers are rapidly covered by biomolecules (proteins, lipids, etc.) which results in the formation of a surface layer, widely known as the biomolecular corona. The biomolecular corona subsequently confers a certain biological identity to the corona-covered nanocarriers which can be crucial during their subsequent interactions with cells or other biological entities. In contrast to the proteins of the corona, little is known about the impact of the non-protein constituents of the corona, such as sugars. Here, we investigate the role of protein glycosylation of the corona in cellular uptake. We show that deglycosylation of clusterin (CLU) and apolipoprotein AI (Apo AI) significantly changes (increases and decreases, respectively) the cellular uptake of nanocarriers covered with these proteins.
Collapse
Affiliation(s)
- Artur Ghazaryan
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | | | |
Collapse
|
28
|
Tree MO, Londono-Renteria B, Troupin A, Clark KM, Colpitts TM, Conway MJ. Dengue virus reduces expression of low-density lipoprotein receptor-related protein 1 to facilitate replication in Aedes aegypti. Sci Rep 2019; 9:6352. [PMID: 31015516 PMCID: PMC6478881 DOI: 10.1038/s41598-019-42803-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 03/28/2019] [Indexed: 01/09/2023] Open
Abstract
Aedes aegypti is the primary vector of a number of viruses pathogenic to humans including dengue virus (DENV). DENV infection leads to widespread transcriptomic and proteomic alterations in mosquito cells. Here we identified alterations to the mosquito cell secretome during DENV infection by performing liquid chromatography tandem mass spectrometry. We found that an extracellular fragment of low-density lipoprotein receptor-related protein 1 (LRP-1) was present during infection. Previous literature suggests that LRP-1 regulates cholesterol homeostasis. Therefore, we hypothesized that DENV modifies LRP-1 protein expression to maintain host-derived intracellular cholesterol, which would facilitate virus replication within membrane-associated replication compartments. Accordingly, stimuli that are present during flavivirus infection reduced LRP-1 protein expression. We also found that dsRNA knockdown of LRP-1 increased intracellular cholesterol and DENV viral RNA. Further, depletion of intracellular lipids reduced infection. Together, these data suggest that DENV reduces LRP-1 protein expression, possibly through regulated intramembrane proteolysis (RIP), to increase intracellular cholesterol and facilitate replication in Ae. aegypti.
Collapse
Affiliation(s)
- Maya O Tree
- Foundational Sciences, Central Michigan University, College of Medicine, Mount Pleasant, MI, United States of America
| | - Berlin Londono-Renteria
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America.,Department of Entomology, Kansas State University, Manhattan, Kansas, United States of America
| | - Andrea Troupin
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Kellie M Clark
- Foundational Sciences, Central Michigan University, College of Medicine, Mount Pleasant, MI, United States of America
| | - Tonya M Colpitts
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America.,Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA, United States of America
| | - Michael J Conway
- Foundational Sciences, Central Michigan University, College of Medicine, Mount Pleasant, MI, United States of America.
| |
Collapse
|
29
|
Shroff N, Ander BP, Zhan X, Stamova B, Liu D, Hull H, Hamade FR, Dykstra-Aiello C, Ng K, Sharp FR, Jickling GC. HDAC9 Polymorphism Alters Blood Gene Expression in Patients with Large Vessel Atherosclerotic Stroke. Transl Stroke Res 2019; 10:19-25. [PMID: 29651704 PMCID: PMC6186202 DOI: 10.1007/s12975-018-0619-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/06/2018] [Accepted: 03/06/2018] [Indexed: 12/20/2022]
Abstract
The histone deacetylase 9 (HDAC9) polymorphism rs2107595 is associated with an increased risk for large vessel atherosclerotic stroke (LVAS). In humans, there remains a need to better understand this HDAC9 polymorphism's contribution to large vessel stroke. In this pilot study, we evaluated whether the HDAC9 polymorphism rs2107595 is associated with differences in leukocyte gene expression in patients with LVAS. HDAC9 SNP rs2107595 was genotyped in 155 patients (43 LVAS and 112 vascular risk factor controls). RNA isolated from blood was processed on whole genome microarrays. Gene expression was compared between HDAC9 risk allele-positive and risk allele-negative LVAS patients and controls. Functional analysis identified canonical pathways and molecular functions associated with rs2107595 in LVAS. In HDAC9 SNP rs2107595 risk allele-positive LVAS patients, there were 155 genes differentially expressed compared to risk allele-negative patients (fold change > |1.2|, p < 0.05). The 155 genes separated the risk allele-positive and risk allele-negative LVAS patients on a principal component analysis. Pathways associated with HDAC9 risk allele-positive status involved IL-6 signaling, cholesterol efflux, and platelet aggregation. These preliminary data suggest an association with the HDAC9 rs2107595 risk allele and peripheral immune, lipid, and clotting systems in LVAS. Further study is required to evaluate whether these differences are related to large vessel atherosclerosis and stroke risk.
Collapse
Affiliation(s)
- Natasha Shroff
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA.
- MIND Institute Wet Labs, Room 2415, 2805 50th Street, Sacramento, CA, 95817, USA.
| | - Bradley P Ander
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Xinhua Zhan
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Boryana Stamova
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - DaZhi Liu
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Heather Hull
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Farah R Hamade
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Cheryl Dykstra-Aiello
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Kwan Ng
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Frank R Sharp
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Glen C Jickling
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| |
Collapse
|
30
|
Schubert K, Collins LE, Green P, Nagase H, Troeberg L. LRP1 Controls TNF Release via the TIMP-3/ADAM17 Axis in Endotoxin-Activated Macrophages. THE JOURNAL OF IMMUNOLOGY 2019; 202:1501-1509. [PMID: 30659107 DOI: 10.4049/jimmunol.1800834] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/16/2018] [Indexed: 11/19/2022]
Abstract
The metalloproteinase ADAM17 plays a pivotal role in initiating inflammation by releasing TNF from its precursor. Prolonged TNF release causes many chronic inflammatory diseases, indicating that tight regulation of ADAM17 activity is essential for resolution of inflammation. In this study, we report that the endogenous ADAM17 inhibitor TIMP-3 inhibits ADAM17 activity only when it is bound to the cell surface and that cell surface levels of TIMP-3 in endotoxin-activated human macrophages are dynamically controlled by the endocytic receptor LRP1. Pharmacological blockade of LRP1 inhibited endocytic clearance of TIMP-3, leading to an increase in cell surface levels of the inhibitor that blocked TNF release. Following LPS stimulation, TIMP-3 levels on the surface of macrophages increased 4-fold within 4 h and continued to accumulate at 6 h, before a return to baseline levels at 8 h. This dynamic regulation of cell surface TIMP-3 levels was independent of changes in TIMP-3 mRNA levels, but correlated with shedding of LRP1. These results shed light on the basic mechanisms that maintain a regulated inflammatory response and ensure its timely resolution.
Collapse
Affiliation(s)
- Kristin Schubert
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Laura E Collins
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Patricia Green
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Hideaki Nagase
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Linda Troeberg
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
31
|
Au DT, Migliorini M, Strickland DK, Muratoglu SC. Macrophage LRP1 Promotes Diet-Induced Hepatic Inflammation and Metabolic Dysfunction by Modulating Wnt Signaling. Mediators Inflamm 2018; 2018:7902841. [PMID: 30524198 PMCID: PMC6247401 DOI: 10.1155/2018/7902841] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/22/2018] [Accepted: 09/03/2018] [Indexed: 12/20/2022] Open
Abstract
Hepatic inflammation is associated with the development of insulin resistance, which can perpetuate the disease state and may increase the risk of metabolic syndrome and diabetes. Despite recent advances, mechanisms linking hepatic inflammation and insulin resistance are still unclear. The low-density lipoprotein receptor-related protein 1 (LRP1) is a large endocytic and signaling receptor that is highly expressed in macrophages, adipocytes, hepatocytes, and vascular smooth muscle cells. To investigate the potential role of macrophage LRP1 in hepatic inflammation and insulin resistance, we conducted experiments using macrophage-specific LRP1-deficient mice (macLRP1-/- ) generated on a low-density lipoprotein receptor knockout (LDLR-/- ) background and fed a Western diet. LDLR-/-; macLRP1-/- mice gained less body weight and had improved glucose tolerance compared to LDLR-/- mice. Livers from LDLR-/-; macLRP1-/- mice displayed lower levels of gene expression for several inflammatory cytokines, including Ccl3, Ccl4, Ccl8, Ccr1, Ccr2, Cxcl9, and Tnf, and reduced phosphorylation of GSK3α and p38 MAPK proteins. Furthermore, LRP1-deficient peritoneal macrophages displayed altered cholesterol metabolism. Finally, circulating levels of sFRP-5, a potent anti-inflammatory adipokine that functions as a decoy receptor for Wnt5a, were elevated in LDLR-/-; macLRP1-/- mice. Surface plasmon resonance experiments revealed that sFRP-5 is a novel high affinity ligand for LRP1, revealing that LRP1 regulates levels of this inhibitor of Wnt5a-mediated signaling. Collectively, our results suggest that LRP1 expression in macrophages promotes hepatic inflammation and the development of glucose intolerance and insulin resistance by modulating Wnt signaling.
Collapse
Affiliation(s)
- Dianaly T. Au
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mary Migliorini
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Selen C. Muratoglu
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
32
|
Mueller PA, Zhu L, Tavori H, Huynh K, Giunzioni I, Stafford JM, Linton MF, Fazio S. Deletion of Macrophage Low-Density Lipoprotein Receptor-Related Protein 1 (LRP1) Accelerates Atherosclerosis Regression and Increases C-C Chemokine Receptor Type 7 (CCR7) Expression in Plaque Macrophages. Circulation 2018; 138:1850-1863. [PMID: 29794082 PMCID: PMC6343494 DOI: 10.1161/circulationaha.117.031702] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 05/14/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND We previously showed that mice lacking MΦLRP1-/- (low-density lipoprotein receptor-related protein 1 in macrophages) undergo accelerated atherosclerotic plaque formation due to changes in macrophages including increased apoptosis, decreased efferocytosis, and exaggerated transition to the inflammatory M1 phenotype. Here we sought to explore the role of macrophage low-density lipoprotein receptor-related protein 1 during regression of atherosclerosis since regressing plaques are characterized by transitioning of macrophages to M2 status as inflammation resolves. METHODS Apolipoprotein E-/- mice on a high-fat diet for 12 weeks were reconstituted with bone marrow from apolipoprotein E-producing wild-type or MΦLRP1-/- mice, and then placed on a chow diet for 10 weeks (n=9 to 11 mice/group). A cohort of apolipoprotein E-/- mice reconstituted with apolipoprotein E-/- bone marrow served as baseline controls (n=9). RESULTS Plaques of both wild-type and MΦLRP1-/- bone marrow recipients regressed compared with controls (11% and 22%, respectively; P<0.05), and plaques of MΦLRP1-/- recipients were 13% smaller than those of wild-type recipients ( P<0.05). Recipients of MΦLRP1-/- marrow had 36% fewer M1 macrophages ( P<0.01) and 2.5-fold more CCR7 (C-C chemokine receptor type 7)-positive macrophages in the plaque relative to wild-type mice ( P<0.01). Additionally, in vivo studies of cellular egress showed a 4.6-fold increase in 5-ethynyl-2´-deoxyuridine-labeled CCR7+ macrophages in mediastinal lymph nodes. Finally, in vivo studies of reverse cholesterol transport showed a 1.4-fold higher reverse cholesterol transport in MΦLRP1-/- recipient mice ( P<0.01). CONCLUSIONS Absence of macrophage low-density lipoprotein receptor-related protein 1 unexpectedly accelerates atherosclerosis regression, enhances reverse cholesterol transport, and increases expression of the motility receptor CCR7, which drives macrophage egress from lesions.
Collapse
Affiliation(s)
- Paul A. Mueller
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Lin Zhu
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN
- Division of Diabetes, Metabolism and Endocrinology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Tennessee Valley Healthcare System, Nashville, TN
| | - Hagai Tavori
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Katherine Huynh
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Ilaria Giunzioni
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - John M. Stafford
- Division of Diabetes, Metabolism and Endocrinology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Tennessee Valley Healthcare System, Nashville, TN
| | - MacRae F. Linton
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| |
Collapse
|
33
|
Actis Dato V, Chiabrando GA. The Role of Low-Density Lipoprotein Receptor-Related Protein 1 in Lipid Metabolism, Glucose Homeostasis and Inflammation. Int J Mol Sci 2018; 19:ijms19061780. [PMID: 29914093 PMCID: PMC6032055 DOI: 10.3390/ijms19061780] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 12/30/2022] Open
Abstract
Metabolic syndrome (MetS) is a highly prevalent disorder which can be used to identify individuals with a higher risk for cardiovascular disease and type 2 diabetes. This metabolic syndrome is characterized by a combination of physiological, metabolic, and molecular alterations such as insulin resistance, dyslipidemia, and central obesity. The low-density lipoprotein receptor-related protein 1 (LRP1—A member of the LDL receptor family) is an endocytic and signaling receptor that is expressed in several tissues. It is involved in the clearance of chylomicron remnants from circulation, and has been demonstrated to play a key role in the lipid metabolism at the hepatic level. Recent studies have shown that LRP1 is involved in insulin receptor (IR) trafficking and intracellular signaling activity, which have an impact on the regulation of glucose homeostasis in adipocytes, muscle cells, and brain. In addition, LRP1 has the potential to inhibit or sustain inflammation in macrophages, depending on its cellular expression, as well as the presence of particular types of ligands in the extracellular microenvironment. In this review, we summarize existing perspectives and the latest innovations concerning the role of tissue-specific LRP1 in lipoprotein and glucose metabolism, and examine its ability to mediate inflammatory processes related to MetS and atherosclerosis.
Collapse
Affiliation(s)
- Virginia Actis Dato
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba X5000HUA, Argentina.
| | - Gustavo Alberto Chiabrando
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba X5000HUA, Argentina.
| |
Collapse
|
34
|
Fava M, Barallobre-Barreiro J, Mayr U, Lu R, Didangelos A, Baig F, Lynch M, Catibog N, Joshi A, Barwari T, Yin X, Jahangiri M, Mayr M. Role of ADAMTS-5 in Aortic Dilatation and Extracellular Matrix Remodeling. Arterioscler Thromb Vasc Biol 2018; 38:1537-1548. [PMID: 29622560 PMCID: PMC6026471 DOI: 10.1161/atvbaha.117.310562] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 03/19/2018] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Thoracic aortic aneurysm (TAA), a degenerative disease of the aortic wall, is accompanied by changes in the structure and composition of the aortic ECM (extracellular matrix). The ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of proteases has recently been implicated in TAA formation. This study aimed to investigate the contribution of ADAMTS-5 to TAA development. Approach and Results— A model of aortic dilatation by AngII (angiotensin II) infusion was adopted in mice lacking the catalytic domain of ADAMTS-5 (Adamts5Δcat). Adamts5Δcat mice showed an attenuated rise in blood pressure while displaying increased dilatation of the ascending aorta (AsAo). Interestingly, a proteomic comparison of the aortic ECM from AngII-treated wild-type and Adamts5Δcat mice revealed versican as the most upregulated ECM protein in Adamts5Δcat mice. This was accompanied by a marked reduction of ADAMTS-specific versican cleavage products (versikine) and a decrease of LRP1 (low-density lipoprotein-related protein 1). Silencing LRP1 expression in human aortic smooth muscle cells reduced the expression of ADAMTS5, attenuated the generation of versikine, but increased soluble ADAMTS-1. A similar increase in ADAMTS-1 was observed in aortas of AngII-treated Adamts5Δcat mice but was not sufficient to maintain versican processing and prevent aortic dilatation. Conclusions— Our results support the emerging role of ADAMTS proteases in TAA. ADAMTS-5 rather than ADAMTS-1 is the key protease for versican regulation in murine aortas. Further studies are needed to define the ECM substrates of the different ADAMTS proteases and their contribution to TAA formation.
Collapse
MESH Headings
- ADAMTS1 Protein/metabolism
- ADAMTS5 Protein/deficiency
- ADAMTS5 Protein/genetics
- ADAMTS5 Protein/metabolism
- Angiotensin II
- Animals
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Cells, Cultured
- Dilatation, Pathologic
- Disease Models, Animal
- Extracellular Matrix/enzymology
- Extracellular Matrix/pathology
- Humans
- Low Density Lipoprotein Receptor-Related Protein-1/genetics
- Low Density Lipoprotein Receptor-Related Protein-1/metabolism
- Male
- Mice, Knockout
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle
- Receptors, LDL/metabolism
- Tumor Suppressor Proteins/metabolism
- Vascular Remodeling
- Versicans/metabolism
Collapse
Affiliation(s)
- Marika Fava
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
- St George's University of London, NHS Trust, United Kingdom (M.F., M.J.)
- Cardiovascular Institute, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (M.F., M.M.)
| | - Javier Barallobre-Barreiro
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ursula Mayr
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ruifang Lu
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Athanasios Didangelos
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ferheen Baig
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Marc Lynch
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Norman Catibog
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Abhishek Joshi
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Temo Barwari
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Xiaoke Yin
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Marjan Jahangiri
- St George's University of London, NHS Trust, United Kingdom (M.F., M.J.)
| | - Manuel Mayr
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
- Cardiovascular Institute, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (M.F., M.M.)
| |
Collapse
|
35
|
El Khoury P, Roussel R, Fumeron F, Abou-Khalil Y, Velho G, Mohammedi K, Jacob MP, Steg PG, Potier L, Ghaleb Y, Elbitar S, Ragot S, Andreata F, Caligiuri G, Hadjadj S, Boileau C, Marre M, Abifadel M, Varret M, Hansel B. Plasma proprotein-convertase-subtilisin/kexin type 9 (PCSK9) and cardiovascular events in type 2 diabetes. Diabetes Obes Metab 2018; 20:943-953. [PMID: 29205760 DOI: 10.1111/dom.13181] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/13/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022]
Abstract
AIM To investigate whether plasma concentrations of proprotein-convertase-subtilisin/kexin type 9 (PCSK9) were associated with cardiovascular (CV) events in two cohorts of patients with type 2 diabetes mellitus. METHODS We considered patients from the DIABHYCAR (n = 3137) and the SURDIAGENE (n = 1468) studies. Baseline plasma PCSK9 concentration was measured using an immunofluorescence assay. In post hoc, but preplanned, analyses we assessed the relationship between PCSK9 and the following endpoints: (1) a combined endpoint of major CV events: CV death, non-fatal myocardial infarction (MI), stroke and heart failure-related hospital admission; (2) a composite of all CV events: MI, stroke, heart failure-related hospital admission, coronary/peripheral angioplasty or bypass, CV death; (3) MI; (4) stroke/transient ischaemic attack (TIA); and (5) CV death. RESULTS In the DIABHYCAR study, plasma PCSK9 tertiles were associated with the incidence of MI, all CV events and stroke/TIA (P for trend <.05). In adjusted Cox analysis, plasma PCSK9 was associated, independently of classic risk factors, with the incidence of major CV events (hazard ratio [HR] for 1-unit increase of log[PCSK9] 1.28 [95% confidence interval {CI} 1.06-1.55]), the incidence of MI (HR 1.66 [95% CI 1.05-2.63]), and the incidence of all CV events (HR 1.22 [95% CI 1.04-1.44]), but not with CV death. Plasma PCSK9 was not associated with the incidence of CV disease in the participants of the SURDIAGENE study with high CV risk treated with statins and insulin. CONCLUSIONS We found that PCSK9 was inconsistently associated with CV events in populations with type 2 diabetes. The association may depend on the level of CV risk and the background treatment.
Collapse
Affiliation(s)
- Petra El Khoury
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Ronan Roussel
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Frederic Fumeron
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Yara Abou-Khalil
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Gilberto Velho
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Kamel Mohammedi
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Marie-Paule Jacob
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Philippe Gabriel Steg
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- FACT, HUPNVS, Département de Cardiologie, AP-HP, CHU Xavier Bichat, Paris, France
- NHLI, Imperial College, Royal Brompton Hospital, London, UK
| | - Louis Potier
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Youmna Ghaleb
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Sandy Elbitar
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Stephanie Ragot
- Centre Investigation Clinique 1402, University of Poitiers, Poitiers, France
- Centre Investigation Clinique, CHU Poitiers, Poitiers, France
- Centre Investigation Clinique CIC1402, INSERM, Poitiers, France
| | - Francesco Andreata
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Giusepinna Caligiuri
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Samy Hadjadj
- Centre Investigation Clinique 1402, University of Poitiers, Poitiers, France
- Centre Investigation Clinique, CHU Poitiers, Poitiers, France
- Centre Investigation Clinique CIC1402, INSERM, Poitiers, France
- Pole DUNE, CHU Poitiers, Poitiers, France
- U1082, INSERM, Poitiers, France
| | - Catherine Boileau
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Département de génétique, AP-HP, CHU Xavier Bichat, Paris, France
| | - Michel Marre
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Marianne Abifadel
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Mathilde Varret
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Boris Hansel
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| |
Collapse
|
36
|
Dai X, Zhang D, Wang C, Wu Z, Liang C. The Pivotal Role of Thymus in Atherosclerosis Mediated by Immune and Inflammatory Response. Int J Med Sci 2018; 15:1555-1563. [PMID: 30443178 PMCID: PMC6216065 DOI: 10.7150/ijms.27238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/06/2018] [Indexed: 12/26/2022] Open
Abstract
Atherosclerosis is one kind of chronic inflammatory disease, in which multiple types of immune cells or factors are involved. Data from experimental and clinical studies on atherosclerosis have confirmed the key roles of immune cells and inflammation in such process. The thymus as a key organ in T lymphocyte ontogenesis has an important role in optimizing immune system function throughout the life, and dysfunction of thymus has been proved to be associated with severity of atherosclerosis. Based on previous research, we begin with the hypothesis that low density lipoprotein or cholesterol reduces the expression of the thymus transcription factor Foxn1 via low density lipoprotein receptors on the membrane surface and low density lipoprotein receptor related proteins on the cell surface, which cause the thymus function decline or degradation. The imbalance of T cell subgroups and the decrease of naive T cells due to thymus dysfunction cause the increase or decrease in the secretion of various inflammatory factors, which in turn aggravates or inhibits atherosclerosis progression and cardiovascular events. Hence, thymus may be the pivotal role in coronary heart disease mediated by atherosclerosis and cardiovascular events and it can imply a novel treatment strategy for the clinical management of patients with atherosclerosis in addition to different commercial drugs. Modulation of immune system by inducing thymus function may be a therapeutic approach for the prevention of atherosclerosis. Purpose of this review is to summarize and discuss the recent advances about the impact of thymus function on atherosclerosis by the data from animal or human studies and the potential mechanisms.
Collapse
Affiliation(s)
- Xianliang Dai
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Cardiology, 101 Hospital of PLA, Wuxi, Jiangsu province 214041, China
| | - Danfeng Zhang
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Chaoqun Wang
- Department of Endocrinology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai 200003, China
| | - Zonggui Wu
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| |
Collapse
|
37
|
Xian X, Ding Y, Dieckmann M, Zhou L, Plattner F, Liu M, Parks JS, Hammer RE, Boucher P, Tsai S, Herz J. LRP1 integrates murine macrophage cholesterol homeostasis and inflammatory responses in atherosclerosis. eLife 2017; 6:e29292. [PMID: 29144234 PMCID: PMC5690284 DOI: 10.7554/elife.29292] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 10/22/2017] [Indexed: 12/11/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional cell surface receptor with diverse physiological roles, ranging from cellular uptake of lipoproteins and other cargo by endocytosis to sensor of the extracellular environment and integrator of a wide range of signaling mechanisms. As a chylomicron remnant receptor, LRP1 controls systemic lipid metabolism in concert with the LDL receptor in the liver, whereas in smooth muscle cells (SMC) LRP1 functions as a co-receptor for TGFβ and PDGFRβ in reverse cholesterol transport and the maintenance of vascular wall integrity. Here we used a knockin mouse model to uncover a novel atheroprotective role for LRP1 in macrophages where tyrosine phosphorylation of an NPxY motif in its intracellular domain initiates a signaling cascade along an LRP1/SHC1/PI3K/AKT/PPARγ/LXR axis to regulate and integrate cellular cholesterol homeostasis through the expression of the major cholesterol exporter ABCA1 with apoptotic cell removal and inflammatory responses.
Collapse
Affiliation(s)
- Xunde Xian
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Yinyuan Ding
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
- Key Laboratory of Medical Electrophysiology, Ministry of Education of ChinaInstitute of Cardiovascular Research, Southwest Medical UniversityLuzhouChina
| | - Marco Dieckmann
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Li Zhou
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Florian Plattner
- Department of PsychiatryUniversity of Texas Southwestern Medical CenterDallasUnited States
- Center for Translational Neurodegeneration ResearchUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Mingxia Liu
- Section on Molecular Medicine, Department of Internal MedicineWake Forest School of MedicineWinston-SalemNorth Carolina
| | - John S Parks
- Section on Molecular Medicine, Department of Internal MedicineWake Forest School of MedicineWinston-SalemNorth Carolina
| | - Robert E Hammer
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasUnited States
| | | | - Shirling Tsai
- Department of SurgeryUT Southwestern Medical CenterDallasUnited States
- Dallas VA Medical CenterDallasUnited States
| | - Joachim Herz
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
- Center for Translational Neurodegeneration ResearchUniversity of Texas Southwestern Medical CenterDallasUnited States
- Department of NeuroscienceUT SouthwesternDallasUnited States
- Department of Neurology and NeurotherapeuticsUT SouthwesternDallasUnited States
| |
Collapse
|
38
|
Theret L, Jeanne A, Langlois B, Hachet C, David M, Khrestchatisky M, Devy J, Hervé E, Almagro S, Dedieu S. Identification of LRP-1 as an endocytosis and recycling receptor for β1-integrin in thyroid cancer cells. Oncotarget 2017; 8:78614-78632. [PMID: 29108253 PMCID: PMC5667986 DOI: 10.18632/oncotarget.20201] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/25/2017] [Indexed: 12/14/2022] Open
Abstract
LRP-1 is a large endocytic receptor mediating the clearance of various molecules from the extracellular matrix. LRP-1 was reported to control focal adhesion turnover to optimize the adhesion-deadhesion balance to support invasion. To better understand how LRP-1 coordinates cell-extracellular matrix interface, we explored its ability to regulate cell surface integrins in thyroid carcinomas. Using an antibody approach, we demonstrated that β1-integrin levels were increased at the plasma membrane under LRP1 silencing or upon RAP treatment, used as LRP-1 antagonist. Our data revealed that LRP-1 binds with both inactive and active β1-integrin conformations and identified the extracellular ligand-binding domains II or IV of LRP-1 as sufficient to bind β1-integrin. Using a recombinant β1-integrin, we demonstrated that LRP-1 acts as a regulator of β1-integrin intracellular traffic. Moreover, RAP or LRP-1 blocking antibodies decreased up to 36% the number of β1-integrin-containing endosomes. LRP-1 blockade did not significantly affect the levels of β1-integrin-containing lysosomes while decreasing localization of β1-integrin within Rab-11 positive vesicles. Overall, we identified an original molecular process in which LRP-1 acts as a main regulator of β1-integrin internalization and recycling in thyroid cancer cells.
Collapse
Affiliation(s)
- Louis Theret
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Albin Jeanne
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France.,SATT Nord, Lille, France
| | - Benoit Langlois
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Cathy Hachet
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Marion David
- VECT-HORUS SAS, Faculté de Médecine Secteur Nord, Marseille, France
| | | | - Jérôme Devy
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Emonard Hervé
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Sébastien Almagro
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Stéphane Dedieu
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| |
Collapse
|
39
|
Luchetti F, Crinelli R, Cesarini E, Canonico B, Guidi L, Zerbinati C, Di Sario G, Zamai L, Magnani M, Papa S, Iuliano L. Endothelial cells, endoplasmic reticulum stress and oxysterols. Redox Biol 2017; 13:581-587. [PMID: 28783588 PMCID: PMC5545768 DOI: 10.1016/j.redox.2017.07.014] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/21/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
Oxysterols are bioactive lipids that act as regulators of lipid metabolism, inflammation, cell viability and are involved in several diseases, including atherosclerosis. Mounting evidence linked the atherosclerosis to endothelium dysfunction; in fact, the endothelium regulates the vascular system with roles in processes such as hemostasis, cell cholesterol, hormone trafficking, signal transduction and inflammation. Several papers shed light the ability of oxysterols to induce apoptosis in different cell lines including endothelial cells. Apoptotic endothelial cell and endothelial denudation may constitute a critical step in the transition to plaque erosion and vessel thrombosis, so preventing the endothelial damaged has garnered considerable attention as a novel means of treating atherosclerosis. Endoplasmic reticulum (ER) is the site where the proteins are synthetized and folded and is necessary for most cellular activity; perturbations of ER homeostasis leads to a condition known as endoplasmic reticulum stress. This condition evokes the unfolded protein response (UPR) an adaptive pathway that aims to restore ER homeostasis. Mounting evidence suggests that chronic activation of UPR leads to cell dysfunction and death and recently has been implicated in pathogenesis of endothelial dysfunction. Autophagy is an essential catabolic mechanism that delivers misfolded proteins and damaged organelles to the lysosome for degradation, maintaining basal levels of autophagic activity it is critical for cell survival. Several evidence suggests that persistent ER stress often results in stimulation of autophagic activities, likely as a compensatory mechanism to relieve ER stress and consequently cell death. In this review, we summarize evidence for the effect of oxysterols on endothelial cells, especially focusing on oxysterols-mediated induction of endoplasmic reticulum stress. Endothelial cells dysfunction is critical in the process of atherothrombosis. Endoplasmic reticulum stress is a key component in endothelial cell dysfunction. Oxysterols are oxidation products of cholesterol found in atherosclerosis lesions. Oxysterols are potential modulators of endoplasmic reticulum stress.
Collapse
Affiliation(s)
- F Luchetti
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - R Crinelli
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - E Cesarini
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - B Canonico
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - L Guidi
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - C Zerbinati
- Department of Medico-Surgical Sciences and Biotechnologies Vascular Biology, Atherothrombosis & Mass Spectrometry, Sapienza University of Rome, Latina, Italy
| | - G Di Sario
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - L Zamai
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - M Magnani
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - S Papa
- Departments of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - L Iuliano
- Department of Medico-Surgical Sciences and Biotechnologies Vascular Biology, Atherothrombosis & Mass Spectrometry, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
40
|
Macrophages in vascular inflammation and atherosclerosis. Pflugers Arch 2017; 469:485-499. [PMID: 28168325 DOI: 10.1007/s00424-017-1941-y] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is characterized by lipid accumulation and chronic inflammation of the arterial wall, and its main complications-myocardial infarction and ischemic stroke-together constitute the first cause of death worldwide. Accumulation of lipid-laden macrophage foam cells in the intima of inflamed arteries has long been recognized as a hallmark of atherosclerosis. However, in recent years, an unexpected complexity in the mechanisms of macrophage accumulation in lesions, in the protective and pathogenic functions performed by macrophages and how they are regulated has been uncovered. Here, we provide an overview of the latest developments regarding the various mechanisms of macrophage accumulation in lesion, the major functional features of lesion macrophages, and how the plaque microenvironment may affect macrophage phenotype. Finally, we discuss how best to apprehend the heterogeneous ontogeny and functionality of atherosclerotic plaque macrophages and argue that moving away from a rigid nomenclature of arbitrarily defined macrophage subsets would be beneficial for research in the field.
Collapse
|
41
|
Chronic Moderate Alcohol Intakes Accelerate SR-B1 Mediated Reverse Cholesterol Transport. Sci Rep 2016; 6:33032. [PMID: 27618957 PMCID: PMC5020497 DOI: 10.1038/srep33032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/18/2016] [Indexed: 11/17/2022] Open
Abstract
Cholesterol is essential for all animal life. However, a high level of cholesterol in the body is strongly associated with the progression of various severe diseases. In our study, the potential involvement of alcohol in the regulation of high density lipoprotein (HDL) receptor scavenger receptor class B and type I (SR-B1)-mediated reverse cholesterol transport was investigated. We separated male C57BL/6 mice into four diets: control, alcohol, Control + HC and alcohol + HC. The SR-B1 level and 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate- high- density lipoprotein (DiI-HDL) uptake were also measured in AML12 cells and HL7702 cells treated with alcohol. The control + HC diet led to increased hepatic triglyceride and cholesterol levels while alcohol + HC led no significant change. Compared with that of the control group, the SR-B1 mRNA level was elevated by 27.1% (P < 0.05), 123.8% (P < 0.001) and 343.6% (P < 0.001) in the alcohol, control + HC and alcohol + HC groups, respectively. In AML12 and HL7702 cells, SR-B1 level and DiI-HDL uptake were repressed by SR-B1 siRNA or GW9662. However, these effects were reversed through alcohol treatment. These data suggest that a moderate amount of alcohol plays a novel role in reverse cholesterol transport, mainly mediated by PPARγ and SR-B1.
Collapse
|
42
|
Boukais K, Bayles R, Borges LDF, Louedec L, Boulaftali Y, Ho-Tin-Noé B, Arocas V, Bouton MC, Michel JB. Uptake of Plasmin-PN-1 Complexes in Early Human Atheroma. Front Physiol 2016; 7:273. [PMID: 27445860 PMCID: PMC4927630 DOI: 10.3389/fphys.2016.00273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/16/2016] [Indexed: 12/16/2022] Open
Abstract
Zymogens are delivered to the arterial wall by radial transmural convection. Plasminogen can be activated within the arterial wall to produce plasmin, which is involved in evolution of the atherosclerotic plaque. Vascular smooth muscle cells (vSMCs) protect the vessels from proteolytic injury due to atherosclerosis development by highly expressing endocytic LDL receptor-related protein-1 (LRP-1), and by producing anti-proteases, such as Protease Nexin-1 (PN-1). PN-1 is able to form covalent complexes with plasmin. We hypothesized that plasmin-PN-1 complexes could be internalized via LRP-1 by vSMCs during the early stages of human atheroma. LRP-1 is also responsible for the capture of aggregated LDL in human atheroma. Plasmin activity and immunohistochemical analyses of early human atheroma showed that the plasminergic system is activated within the arterial wall, where intimal foam cells, including vSMCs and platelets, are the major sites of PN-1 accumulation. Both PN-1 and LRP-1 are overexpressed in early atheroma at both messenger and protein levels. Cell biology studies demonstrated an increased expression of PN-1 and tissue plasminogen activator by vSMCs in response to LDL. Plasmin-PN-1 complexes are internalized via LRP-1 in vSMCs, whereas plasmin alone is not. Tissue PN-1 interacts with plasmin in early human atheroma via two complementary mechanisms: plasmin inhibition and tissue uptake of plasmin-PN-1 complexes via LRP-1 in vSMCs. Despite this potential protective effect, plasminogen activation by vSMCs remains abnormally elevated in the intima in early stages of human atheroma.
Collapse
Affiliation(s)
- Kamel Boukais
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Richard Bayles
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Department of Physiology and Pharmacology, Oregon Health and Science UniversityPortland, OR, USA
| | - Luciano de Figueiredo Borges
- Departement of Biological Science, Federal University of São PauloSão Paulo, Brazil; Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão Paulo, Brazil
| | - Liliane Louedec
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Yacine Boulaftali
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Benoit Ho-Tin-Noé
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Véronique Arocas
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Marie-Christine Bouton
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| | - Jean-Baptiste Michel
- UMR 1148, Laboratory for Vascular Translational Science, Institut National de la Santé et de la Recherche MédicaleParis, France; Paris7 Denis Diderot UniversityParis, France
| |
Collapse
|
43
|
Abstract
The remarkable plasticity and plethora of biological functions performed by macrophages have enticed scientists to study these cells in relation to atherosclerosis for >50 years, and major discoveries continue to be made today. It is now understood that macrophages play important roles in all stages of atherosclerosis, from initiation of lesions and lesion expansion, to necrosis leading to rupture and the clinical manifestations of atherosclerosis, to resolution and regression of atherosclerotic lesions. Lesional macrophages are derived primarily from blood monocytes, although recent research has shown that lesional macrophage-like cells can also be derived from smooth muscle cells. Lesional macrophages take on different phenotypes depending on their environment and which intracellular signaling pathways are activated. Rather than a few distinct populations of macrophages, the phenotype of the lesional macrophage is more complex and likely changes during the different phases of atherosclerosis and with the extent of lipid and cholesterol loading, activation by a plethora of receptors, and metabolic state of the cells. These different phenotypes allow the macrophage to engulf lipids, dead cells, and other substances perceived as danger signals; efflux cholesterol to high-density lipoprotein; proliferate and migrate; undergo apoptosis and death; and secrete a large number of inflammatory and proresolving molecules. This review article, part of the Compendium on Atherosclerosis, discusses recent advances in our understanding of lesional macrophage phenotype and function in different stages of atherosclerosis. With the increasing understanding of the roles of lesional macrophages, new research areas and treatment strategies are beginning to emerge.
Collapse
Affiliation(s)
- Ira Tabas
- From the Departments of Medicine (I.T.), Anatomy and Cell Biology (I.T.), and Physiology and Cellular Biophysics (I.T.), Columbia University, New York; and the Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (K.E.B.) and Department of Pathology (K.E.B.), UW Diabetes Institute, University of Washington School of Medicine, Seattle
| | - Karin E Bornfeldt
- From the Departments of Medicine (I.T.), Anatomy and Cell Biology (I.T.), and Physiology and Cellular Biophysics (I.T.), Columbia University, New York; and the Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (K.E.B.) and Department of Pathology (K.E.B.), UW Diabetes Institute, University of Washington School of Medicine, Seattle.
| |
Collapse
|
44
|
Lillis AP, Muratoglu SC, Au DT, Migliorini M, Lee MJ, Fried SK, Mikhailenko I, Strickland DK. Correction: LDL Receptor-Related Protein-1 (LRP1) Regulates Cholesterol Accumulation in Macrophages. PLoS One 2016; 11:e0147457. [PMID: 26797092 PMCID: PMC4721597 DOI: 10.1371/journal.pone.0147457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
45
|
Lan G, Xie W, Li L, Zhang M, Liu D, Tan YL, Cheng HP, Gong D, Huang C, Zheng XL, Yin WD, Tang CK. MicroRNA-134 actives lipoprotein lipase-mediated lipid accumulation and inflammatory response by targeting angiopoietin-like 4 in THP-1 macrophages. Biochem Biophys Res Commun 2015; 472:410-7. [PMID: 26546816 DOI: 10.1016/j.bbrc.2015.10.158] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 10/30/2015] [Indexed: 12/22/2022]
Abstract
Angiopoietin-like 4 (Angptl4), a secreted protein, is an important regulator to irreversibly inhibit lipoprotein lipase (LPL) activity. Macrophage LPL contributes to foam cell formation via a so-called"molecular bridge" between lipoproteins and receptors on cell surface. It has been reported that macrophage ANGPTL4 suppresses LPL activity, foam cell formation and inflammatory gene expression to reduce atherosclerosis development. Recently, some studies demonstrated that microRNA-134 is upregulated in atherosclerotic macrophages. Here we demonstrate that miR-134 directly binds to 3'UTR of ANGPTL4 mRNA to suppression the expression of ANGPTL4. To investigate the potential roles of macrophage miR-134, THP-1 macrophages were transfected with miR-134 mimics or inhibitors. Our results showed that LPL activity and protein were dramatically increased. We also found that miR-134 activated LPL-mediated lipid accumulation. Collectively, our findings indicate that miR-134 may regulate lipid accumulation and proinfiammatory cytokine secretion in macrophages by targeting the ANGPTL4 gene. Our results have also suggested a promising and potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Gang Lan
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Wei Xie
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Liang Li
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Min Zhang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Dan Liu
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Yu-Lin Tan
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Hai-Peng Cheng
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Duo Gong
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Chong Huang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Wei-Dong Yin
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|