Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Publication year range
1.
Immunity ; 50(4): 941-954, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995508

ABSTRACT

Arterial inflammation is a hallmark of atherosclerosis, and appropriate management of this inflammation represents a major unmet therapeutic need for cardiovascular disease patients. Here, we review the diverse contributions of immune cells to atherosclerosis, the mechanisms of immune cell activation in this context, and the cytokine circuits that underlie disease progression. We discuss the recent application of these insights in the form of immunotherapy to treat cardiovascular disease and highlight how studies on the cardiovascular co-morbidity that arises in autoimmunity might reveal additional roles for cytokines in atherosclerosis. Currently, data point to interleukin-1ß (IL-1ß), tumor necrosis factor (TNF), and IL-17 as cytokines that, at least in some settings, are effective targets to reduce cardiovascular disease progression.


Subject(s)
Cardiovascular Diseases/immunology , Cytokines/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Atherosclerosis/drug therapy , Atherosclerosis/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Cardiovascular Diseases/drug therapy , Cholesterol/metabolism , Clinical Trials as Topic , Cytokines/antagonists & inhibitors , Cytokines/therapeutic use , Disease Progression , Foam Cells/immunology , Foam Cells/metabolism , Gastrointestinal Microbiome , Humans , Inflammasomes/immunology , Inflammation/drug therapy , Inflammation/immunology , Interleukin-1beta/antagonists & inhibitors , Mice, Knockout , Models, Immunological , Muscle, Smooth, Vascular/immunology , Phagocytes/immunology , Phagocytes/metabolism , Signal Transduction , Swine , Translational Research, Biomedical
2.
Nature ; 556(7702): 501-504, 2018 04.
Article in English | MEDLINE | ID: mdl-29670287

ABSTRACT

Metabolic regulation has been recognized as a powerful principle guiding immune responses. Inflammatory macrophages undergo extensive metabolic rewiring 1 marked by the production of substantial amounts of itaconate, which has recently been described as an immunoregulatory metabolite 2 . Itaconate and its membrane-permeable derivative dimethyl itaconate (DI) selectively inhibit a subset of cytokines 2 , including IL-6 and IL-12 but not TNF. The major effects of itaconate on cellular metabolism during macrophage activation have been attributed to the inhibition of succinate dehydrogenase2,3, yet this inhibition alone is not sufficient to account for the pronounced immunoregulatory effects observed in the case of DI. Furthermore, the regulatory pathway responsible for such selective effects of itaconate and DI on the inflammatory program has not been defined. Here we show that itaconate and DI induce electrophilic stress, react with glutathione and subsequently induce both Nrf2 (also known as NFE2L2)-dependent and -independent responses. We find that electrophilic stress can selectively regulate secondary, but not primary, transcriptional responses to toll-like receptor stimulation via inhibition of IκBζ protein induction. The regulation of IκBζ is independent of Nrf2, and we identify ATF3 as its key mediator. The inhibitory effect is conserved across species and cell types, and the in vivo administration of DI can ameliorate IL-17-IκBζ-driven skin pathology in a mouse model of psoriasis, highlighting the therapeutic potential of this regulatory pathway. Our results demonstrate that targeting the DI-IκBζ regulatory axis could be an important new strategy for the treatment of IL-17-IκBζ-mediated autoimmune diseases.


Subject(s)
Activating Transcription Factor 3/metabolism , I-kappa B Proteins/metabolism , Succinates/metabolism , Animals , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Glutathione/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-6/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Psoriasis/drug therapy , Psoriasis/pathology , Stress, Physiological/drug effects , Succinates/administration & dosage , Succinates/chemistry , Succinates/pharmacology , Succinates/therapeutic use , Toll-Like Receptors/immunology
3.
Eur Heart J ; 44(29): 2730-2742, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37377160

ABSTRACT

AIMS: Excess dietary sodium intake and retention lead to hypertension. Impaired dermal lymphangiogenesis and lymphatic dysfunction-mediated sodium and fluid imbalance are pathological mechanisms. The adenosine A2A receptor (A2AR) is expressed in lymphatic endothelial cells (LECs), while the roles and mechanisms of LEC-A2AR in skin lymphangiogenesis during salt-induced hypertension are not clear. METHODS AND RESULTS: The expression of LEC-A2AR correlated with lymphatic vessel density in both high-salt diet (HSD)-induced hypertensive mice and hypertensive patients. Lymphatic endothelial cell-specific A2AR knockout mice fed HSD exhibited 17 ± 2% increase in blood pressure and 17 ± 3% increase in Na+ content associated with decreased lymphatic density (-19 ± 2%) compared with HSD-WT mice. A2AR activation by agonist CGS21680 increased lymphatic capillary density and decreased blood pressure in HSD-WT mice. Furthermore, this A2AR agonist activated MSK1 directly to promote VEGFR2 activation and endocytosis independently of VEGF as assessed by phosphoprotein profiling and immunoprecipitation assays in LECs. VEGFR2 kinase activity inhibitor fruquintinib or VEGFR2 knockout in LECs but not VEGF-neutralizing antibody bevacizumab suppressed A2AR activation-mediated decrease in blood pressure. Immunostaining revealed phosphorylated VEGFR2 and MSK1 expression in the LECs were positively correlated with skin lymphatic vessel density and A2AR level in hypertensive patients. CONCLUSION: The study highlights a novel A2AR-mediated VEGF-independent activation of VEGFR2 signaling in dermal lymphangiogenesis and sodium balance, which might be a potential therapeutic target in salt-sensitive hypertension.


Subject(s)
Hypertension , Lymphangiogenesis , Mice , Animals , Receptor, Adenosine A2A/metabolism , Endothelial Cells/metabolism , Protein Kinase Inhibitors , Sodium/metabolism
4.
Anal Chem ; 95(20): 8011-8019, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37154434

ABSTRACT

Traditional lateral flow assay (LFA) is restricted to providing qualitative or semi-quantitative results and often requires special equipment for obtaining quantitative results. Herein, we proposed a naked-eye readout distance quantitative lateral flow assay based on the permeability changes in enzyme-catalyzed hydrogelation, which not only has the advantages of being simple, immediate, of high efficiency and low cost, and accurate in quantification but also avoids the use of special equipment. The developed LFA method includes three principal components of a nitrocellulose (NC) membrane containing a control line (C line) loading goat anti-rabbit (GAR) antibodies and a test line (T line) loading specific antibodies, alginate-tyramine conjugates forming a hydrogel in the presence of hydrogen peroxide (H2O2) and horseradish peroxidase (HRP), and the HRP-AuNPs-Ab probe only labeling targets captured on the T line. Hemoglobin A1c (HbA1c) was chosen as a representative example to demonstrate the feasibility of our method. Under the optimal conditions, the developed LFA method shows excellent performance in standard samples and real human blood samples where the results of real human blood samples show a high linear correlation with the clinical data obtained by ion exchange chromatography (R2 = 0.9929) and the margin of recovery is only 3.8%. All results demonstrated that our developed LFA method not only has enormous potential in the quantitative detection of HbA1c in clinical complex samples but also can serve as a versatile method for highly efficient detection of other target biomolecules due to the fungibility of antibodies.


Subject(s)
Hydrogen Peroxide , Metal Nanoparticles , Animals , Humans , Rabbits , Glycated Hemoglobin , Hydrogen Peroxide/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Antibodies , Permeability , Catalysis
5.
Anal Chem ; 95(27): 10231-10240, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37322584

ABSTRACT

Due to the highly heterogeneous nature of hepatocellular carcinoma (HCC), the accurate diagnosis of HCC during the early phase of development is still a challenging task. Therefore, the further development of novel diagnostic methods by discovering new biomarkers is required to improve the rate of HCC diagnosis in the early phase. In this work, an oxygen-modified three-dimensional interconnected porous carbon probe is designed and fabricated to profile the differences of N-glycans in human serum from health controls (H) and patients with hepatic dysfunction (HD) and HCC for the discovery of new biomarkers with HCC development. Excitingly, we discovered that the expression levels of 12 serum N-glycans were gradually increased from H to patients with HD and eventually to patients with HCC. Moreover, two machine learning models established based on these 12 serum N-glycans reached a satisfactory accuracy for predicting HCC development where the receiver operating characteristic curve arrived above 0.95 for distinguishing healthy controls and patients with liver diseases (HD or HCC) and the ROC curve arrived at 0.85 for distinguishing HD and HCC. Our work not only developed a new method for the large-scale characterization of serum N-glycans but also provided valuable guidance for accurate and highly sensitive diagnosis of early liver cancer development in a non-invasive manner.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Liver Neoplasms/diagnosis , Porosity , Biomarkers, Tumor , Biomarkers , ROC Curve , Polysaccharides
6.
Environ Sci Technol ; 57(49): 20460-20469, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38019752

ABSTRACT

Biodiesel, derived from alkyl esters of vegetable oils or animal fats, has gained prominence as a greener alternative to diesel due to its reduced particle mass. However, it remains debatable whether biodiesel exposure has more severe health issues than diesel. This study performed high-resolution mass spectrometry to examine the detailed particle chemical compositions and lipidomics analysis of human lung epithelial cells treated with emissions from biodiesel and diesel fuels. Results show the presence of the peak substances of CHO compounds in biodiesel combustion that contain a phthalate ester (PAEs) structure (e.g., n-amyl isoamyl phthalate and diisobutyl phthalate). PAEs have emerged as persistent organic pollutants across various environmental media and are known to possess endocrine-disrupting properties in the environment. We further observed that biodiesel prevents triglyceride storage compared to diesel and inhibits triglycerides from becoming phospholipids, particularly with increased phosphatidylglycerols (PGs) and phosphatidylethanolamines (PEs), which potentially could lead to a higher probability of cancer metastasis.


Subject(s)
Air Pollutants , Vehicle Emissions , Animals , Humans , Vehicle Emissions/analysis , Biofuels/analysis , Lipid Metabolism , Gasoline/analysis , Air Pollutants/analysis
7.
Mikrochim Acta ; 191(1): 11, 2023 12 06.
Article in English | MEDLINE | ID: mdl-38055058

ABSTRACT

A hydrophilic Al-MOFs composite was prepared using cheap and available reagents in water via a suitable large-scale production, an economical and environment-friendly method for capturing N-glycopeptides. The prepared Al-MOFs composite with high hydrolytically stable and hydrophilic 1D channels exhibits an ultralow detection limit (0.5 fmol/µL), and excellent reusability (at least 10 cycles) in the capture of N-glycopeptides from standard bio-samples. Interestingly, the Al-MOFs composite also shows remarkable performance in practical applications, where 300 N-glycopeptides ascribed to 124 glycoproteins were identified in 1 µL human serum and were successfully applied in profiling the differences of N-glycopeptides during diabetes progression. Moreover, 12 specific glycoproteins used as biomarkers to accurately distinguish the progression of diabetes are identified. The present work provides a potential commercial method for large-scale glycoproteomics research in complex clinical samples while offering new guidance for the precise diagnosis of diabetes progression.


Subject(s)
Diabetes Mellitus , Metal-Organic Frameworks , Humans , Diabetes Mellitus/diagnosis , Mass Spectrometry , Glycopeptides , Water , Glycoproteins
8.
Anal Chem ; 94(43): 15076-15084, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36265544

ABSTRACT

Efficient isolation and downstream bioinformation analysis of circulating tumor cells (CTCs) in whole blood contribute to the early diagnosis of cancer and investigation of cancer metastasis. However, the separation and release of CTCs remain a great challenge due to the extreme rarity of CTCs and severe interference from other cells in complex clinical samples. Herein, we developed a low-cost and easy-to-fabricate aptamer-functionalized wafer with a three-dimensional (3D) interconnected porous structure by grafting polydopamine (PDA), poly(ethylene glycol) (PEG), and aptamer in sequence (Ni@PDA-PEG-Apt) for the capture and release of CTCs. The Ni@PDA-PEG-Apt wafer integrated the features of Ni foam with a 3D interconnected porous structure offering enough tunnels for cells to flow through and enhancing aptamer-cell contact frequency, the spacer PEG with flexible and high hydrophilic property increasing anti-interference ability and providing the wafer with more binding sites for aptamer, which result in an enhanced capture specificity and efficiency for CTCs. Because of these advantages, the Ni@PDA-PEG-Apt wafer achieved a high capture efficiency of 78.25%. The captured cancer cells were mildly released by endonuclease with up to 61.85% efficiency and good proliferation. Furthermore, tumor cells were injected into mice and experienced circulation in vivo. In blood samples after circulation, 65% of target tumor cells can be efficiently captured by the wafer, followed by released and recultured cells with high viability. Further downstream metabolomics analysis showed that target cancer cells remained with high biological activity and can be well separated from MCF-10A cells based on metabolic profiles by the PCA analysis, indicating the great potential of our strategy for further research on the progression of cancer metastasis. Notably, not only is the wafer cheap with a cost of only 3.58 U.S. dollars and easily prepared by environmental-friendly reagents but also the process of capturing and releasing tumor cells can be completed within an hour, which is beneficial for large-scale clinical use in the future.


Subject(s)
Neoplastic Cells, Circulating , Mice , Animals , Neoplastic Cells, Circulating/pathology , Porosity , Cell Count , Polyethylene Glycols/chemistry , Cell Separation/methods , Cell Line, Tumor
9.
Arterioscler Thromb Vasc Biol ; 41(2): 822-836, 2021 02.
Article in English | MEDLINE | ID: mdl-33327748

ABSTRACT

OBJECTIVE: vMIP-II (viral macrophage inflammatory protein 2)/vCCL2 (viral chemotactic cytokine ligand 2) binds to multiple chemokine receptors, and vMIP-II-based positron emission tomography tracer (64Cu-DOTA-vMIP-II: vMIP-II tracer) accumulates at atherosclerotic lesions in mice. Given that it would be expected to react with multiple chemokine receptors on monocytes and macrophages, we wondered if its accumulation in atherosclerosis lesion-bearing mice might correlate with overall macrophage burden or, alternatively, the pace of monocyte recruitment. Approach and Results: We employed a mouse model of atherosclerosis regression involving adenoassociated virus 8 vector encoding murine Apoe (AAV-mApoE) treatment of Apoe-/- mice where the pace of monocyte recruitment slows before macrophage burden subsequently declines. Accumulation of 64Cu-DOTA-vMIP-II at Apoe-/- plaque sites was strong but declined with AAV-mApoE-induced decline in monocyte recruitment, before macrophage burden reduced. Monocyte depletion indicated that monocytes and macrophages themselves were not the only target of the 64Cu-DOTA-vMIP-II tracer. Using fluorescence-tagged vMIP-II tracer, competitive receptor blocking with CXCR4 antagonists, endothelial-specific Cre-mediated deletion of CXCR4, CXCR4-specific tracer 64Cu-DOTA-FC131, and CXCR4 staining during disease progression and regression, we show endothelial cell expression of CXCR4 is a key target of 64Cu-DOTA-vMIP-II imaging. Expression of CXCR4 was low in nonplaque areas but strongly detected on endothelium of progressing plaques, especially on proliferating endothelium, where vascular permeability was increased and monocyte recruitment was the strongest. CONCLUSIONS: Endothelial injury status of plaques is marked by CXCR4 expression and this injury correlates with the tendency of such plaques to recruit monocytes. Furthermore, our findings suggest positron emission tomography tracers that mark CXCR4 can be used translationally to monitor the state of plaque injury and monocyte recruitment.


Subject(s)
Aorta, Thoracic/diagnostic imaging , Atherosclerosis/diagnostic imaging , Chemokines/administration & dosage , Endothelium, Vascular/diagnostic imaging , Molecular Imaging , Monocytes/metabolism , Organometallic Compounds/administration & dosage , Positron-Emission Tomography , Radiopharmaceuticals/administration & dosage , Receptors, CXCR4/metabolism , Animals , Aorta, Thoracic/immunology , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers/metabolism , Cell Line , Chemokines/pharmacokinetics , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Injections, Intravenous , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Monocytes/immunology , Monocytes/pathology , Organometallic Compounds/pharmacokinetics , Plaque, Atherosclerotic , Predictive Value of Tests , Radiopharmaceuticals/pharmacokinetics , Receptors, CXCR4/genetics
10.
Analyst ; 147(22): 4954-4961, 2022 Nov 07.
Article in English | MEDLINE | ID: mdl-36226526

ABSTRACT

The comprehensive characterization of N-glycans is of significant importance for the discovery of potential biomarkers and the diagnosis and therapy of diseases. Herein, we designed and fabricated a porous graphitized carbon biomaterial (CS-900-1C) using cheap and available chitosan as the carbon source via a facile pyrolysis process and a post-oxidation strategy for the effective capture of N-glycans. Thanks to its large surface area (2846 m2 g-1), high graphitization degree, suitable oxidation degree and unique porous structure, the CS-900-1C biomaterial exhibits an ultralow detection limit (1 ng µL-1), an excellent size-exclusion effect (OVA digest : BSA protein : OVA protein, 1 : 1000 : 1000, w/w/w) and satisfactory reusability (at least 8 cycles) in the capture of standard N-glycans. Moreover, CS-900-1C has successfully been applied in profiling the difference of N-glycans during diabetes progression (obesity, impaired glucose tolerance, diabetes patients and healthy control) where we discovered that the expression levels of five N-glycans show a gradually increasing trend as diabetes progresses. Remarkably, the five specific N-glycans could be considered as biomarkers to accurately diagnose the progression of diabetes. Our work not only developed a novel porous graphitized carbon biomaterial for the large-scale characterization of N-glycans but also provided new guidance for the precise therapy of diabetes.


Subject(s)
Carbon , Chitosan , Humans , Porosity , Carbon/chemistry , Biocompatible Materials , Polysaccharides/chemistry
11.
Mol Pharm ; 18(3): 1386-1396, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33591187

ABSTRACT

Chemokines and chemokine receptors play an important role in the initiation and progression of atherosclerosis by mediating the trafficking of inflammatory cells. Chemokine receptor 5 (CCR5) has major implications in promoting the development of plaques to advanced stage and related vulnerability. CCR5 antagonist has demonstrated the effective inhibition of atherosclerotic progression in mice, making it a potential biomarker for atherosclerosis management. To accurately determine CCR5 in vivo, we synthesized CCR5 targeted Comb nanoparticles through a modular design and construction strategy with control over the physiochemical properties and functionalization of CCR5 targeting peptide d-Ala-peptide T-amide (DAPTA-Comb). In vivo pharmacokinetic evaluation through 64Cu radiolabeling showed extended blood circulation of 64Cu-DAPTA-Combs conjugated with 10%, 25%, and 40% DAPTA. The different organ distribution profiles of the three nanoparticles demonstrated the effect of DAPTA on not only physicochemical properties but also targeting efficiency. In vivo positron emission tomography/computed tomography (PET/CT) imaging in an apolipoprotein E knockout mouse atherosclerosis model (ApoE-/-) showed that the three 64Cu-DAPTA-Combs could sensitively and specifically detect CCR5 along the progression of atherosclerotic lesions. In an ApoE-encoding adenoviral vector (AAV) induced plaque regression ApoE-/- mouse model, decreased monocyte recruitment, CD68+ macrophages, CCR5 expression, and plaque size were all associated with reduced PET signals, which not only further confirmed the targeting efficiency of 64Cu-DAPTA-Combs but also highlighted the potential of these targeted nanoparticles for atherosclerosis imaging. Moreover, the up-regulation of CCR5 and colocalization with CD68+ macrophages in the necrotic core of ex vivo human plaque specimens warrant further investigation for atherosclerosis prognosis.


Subject(s)
Atherosclerosis/diagnostic imaging , Atherosclerosis/metabolism , Nanoparticles/administration & dosage , Receptors, CCR5/metabolism , Alanine/metabolism , Animals , Apolipoproteins E/metabolism , Chemokines/metabolism , Copper Radioisotopes/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/metabolism , Positron Emission Tomography Computed Tomography/methods , Radiopharmaceuticals/metabolism
12.
J Biol Chem ; 294(43): 15836-15849, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31495784

ABSTRACT

Cholesterol esters are a key ingredient of foamy cells in atherosclerotic lesions; their formation is catalyzed by two enzymes: acyl-CoA:cholesterol acyltransferases (ACATs; also called sterol O-acyltransferases, or SOATs) ACAT1 and ACAT2. ACAT1 is present in all body cells and is the major isoenzyme in macrophages. Whether blocking ACAT1 benefits atherosclerosis has been under debate for more than a decade. Previously, our laboratory developed a myeloid-specific Acat1 knockout (KO) mouse (Acat1-M/-M), devoid of ACAT1 only in macrophages, microglia, and neutrophils. In previous work using the ApoE KO (ApoE-/-) mouse model for early lesions, Acat1-M/-M significantly reduced lesion macrophage content and suppressed atherosclerosis progression. In advanced lesions, cholesterol crystals become a prominent feature. Here we evaluated the effects of Acat1-M/-M in the ApoE KO mouse model for more advanced lesions and found that mice lacking myeloid Acat1 had significantly reduced lesion cholesterol crystal contents. Acat1-M/-M also significantly reduced lesion size and macrophage content without increasing apoptotic cell death. Cell culture studies showed that inhibiting ACAT1 in macrophages caused cells to produce less proinflammatory responses upon cholesterol loading by acetyl low-density lipoprotein. In advanced lesions, Acat1-M/-M reduced but did not eliminate foamy cells. In advanced plaques isolated from ApoE-/- mice, immunostainings showed that both ACAT1 and ACAT2 are present. In cell culture, both enzymes are present in macrophages and smooth muscle cells and contribute to cholesterol ester biosynthesis. Overall, our results support the notion that targeting ACAT1 or targeting both ACAT1 and ACAT2 in macrophages is a novel strategy to treat advanced lesions.


Subject(s)
Atherosclerosis/enzymology , Atherosclerosis/prevention & control , Inflammation/pathology , Macrophages, Peritoneal/enzymology , Myeloid Cells/enzymology , Sterol O-Acyltransferase/metabolism , Animals , Apolipoproteins E , Apoptosis , Atherosclerosis/pathology , Cholesterol/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Gene Deletion , Gene Silencing , Hydroxycholesterols/metabolism , Lipoproteins, LDL/metabolism , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myeloid Cells/pathology , Myocytes, Smooth Muscle/metabolism , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells
13.
Int J Mol Sci ; 21(3)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31979310

ABSTRACT

Epidemiological results revealed that there is an inverse correlation between high-density lipoprotein (HDL) cholesterol levels and risks of atherosclerotic cardiovascular disease (ASCVD). Mounting evidence supports that HDLs are atheroprotective, therefore, many therapeutic approaches have been developed to increase HDL cholesterol (HDL-C) levels. Nevertheless, HDL-raising therapies, such as cholesteryl ester transfer protein (CETP) inhibitors, failed to ameliorate cardiovascular outcomes in clinical trials, thereby casting doubt on the treatment of cardiovascular disease (CVD) by increasing HDL-C levels. Therefore, HDL-targeted interventional studies were shifted to increasing the number of HDL particles capable of promoting ATP-binding cassette transporter A1 (ABCA1)-mediated cholesterol efflux. One such approach was the development of reconstituted HDL (rHDL) particles that promote ABCA1-mediated cholesterol efflux from lipid-enriched macrophages. Here, we explore the manipulation of rHDL nanoparticles as a strategy for the treatment of CVD. In addition, we discuss technological capabilities and the challenge of relating preclinical in vivo mice research to clinical studies. Finally, by drawing lessons from developing rHDL nanoparticles, we also incorporate the viabilities and advantages of the development of a molecular imaging probe with HDL nanoparticles when applied to ASCVD, as well as gaps in technology and knowledge required for putting the HDL-targeted therapeutics into full gear.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Atherosclerosis/drug therapy , Drug Discovery , Lipoproteins, HDL/metabolism , Nanoparticles , Animals , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Atherosclerosis/diagnostic imaging , Atherosclerosis/metabolism , Biomimetics , Humans , Macrophages/metabolism , Mice , Molecular Imaging , Nanoparticles/chemistry , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism
14.
Arterioscler Thromb Vasc Biol ; 38(8): 1702-1710, 2018 08.
Article in English | MEDLINE | ID: mdl-29903736

ABSTRACT

Objective- Macrophages play important roles in the pathogenesis of atherosclerosis, but their dynamics within plaques remain obscure. We aimed to quantify macrophage positional dynamics within progressing and regressing atherosclerotic plaques. Approach and Results- In a stable intravital preparation, large asymmetrical foamy macrophages in the intima of carotid artery plaques were sessile, but smaller rounded cells nearer plaque margins, possibly newly recruited monocytes, mobilized laterally along plaque borders. Thus, to test macrophage dynamics in plaques over a longer period of time in progressing and regressing disease, we quantified displacement of nondegradable phagocytic particles within macrophages for up to 6 weeks. In progressing plaques, macrophage-associated particles appeared to mobilize to deeper layers in plaque, whereas in regressing plaques, the label was persistently located near the lumen. By measuring the distance of the particles from the floor of the plaque, we discovered that particles remained at the same distance from the floor regardless of plaque progression or regression. The apparent deeper penetration of labeled cells in progressing conditions could be attributed to monocyte recruitment that generated new superficial layers of macrophages over the labeled phagocytes. Conclusions- Although there may be individual exceptions, as a population, newly differentiated macrophages fail to penetrate significantly deeper than the limited depth they reside on initial entry, regardless of plaque progression, or regression. These limited dynamics may prevent macrophages from escaping areas with unfavorable conditions (such as hypoxia) and pose a challenge for newly recruited macrophages to clear debris through efferocytosis deep within plaque.


Subject(s)
Aorta/pathology , Aortic Diseases/pathology , Atherosclerosis/pathology , Carotid Arteries/pathology , Carotid Artery Diseases/pathology , Macrophages/pathology , Plaque, Atherosclerotic , Animals , Aorta/metabolism , Aortic Diseases/genetics , Aortic Diseases/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Carotid Arteries/metabolism , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Cell Differentiation , Cell Movement , Disease Models, Animal , Disease Progression , Female , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phagocytosis , Phenotype , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Time Factors
15.
Am J Physiol Endocrinol Metab ; 315(3): E340-E356, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29533741

ABSTRACT

Macrophages are phagocytes that play important roles in health and diseases. Acyl-CoA:cholesterol acyltransferase 1 (ACAT1) converts cellular cholesterol to cholesteryl esters and is expressed in many cell types. Unlike global Acat1 knockout (KO), myeloid-specific Acat1 KO ( Acat1-) does not cause overt abnormalities in mice. Here, we performed analyses in age- and sex-matched Acat1-M/-M and wild-type mice on chow or Western diet and discovered that Acat1-M/-M mice exhibit resistance to Western diet-induced obesity. On both chow and Western diets, Acat1-M/-M mice display decreased adipocyte size and increased insulin sensitivity. When fed with Western diet, Acat1-M/-M mice contain fewer infiltrating macrophages in white adipose tissue (WAT), with significantly diminished inflammatory phenotype. Without Acat1, the Ly6Chi monocytes express reduced levels of integrin-ß1, which plays a key role in the interaction between monocytes and the inflamed endothelium. Adoptive transfer experiment showed that the appearance of leukocytes from Acat1-M/-M mice to the inflamed WAT of wild-type mice is significantly diminished. Under Western diet, Acat1-M/-M causes suppression of multiple proinflammatory genes in WAT. Cell culture experiments show that in RAW 264.7 macrophages, inhibiting ACAT1 with a small-molecule ACAT1-specific inhibitor reduces inflammatory responses to lipopolysaccharide. We conclude that under Western diet, blocking ACAT1 in macrophages attenuates inflammation in WAT. Other results show that Acat1-M/-M does not compromise antiviral immune response. Our work reveals that blocking ACAT1 suppresses diet-induced obesity in part by slowing down monocyte infiltration to WAT as well as by reducing the inflammatory responses of adipose tissue macrophages.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/physiology , Diet , Inflammation/genetics , Inflammation/pathology , Insulin Resistance/genetics , Macrophages/pathology , Obesity/genetics , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/physiology , Adipocytes/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Cell Size , Female , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Inflammation/immunology , Integrin beta1/metabolism , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/physiopathology , RAW 264.7 Cells
16.
J Biol Chem ; 291(12): 6232-44, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26801614

ABSTRACT

Acyl-CoA:cholesterol acyltransferase 1 (Acat1) converts cellular cholesterol to cholesteryl esters and is considered a drug target for treating atherosclerosis. However, in mouse models for atherosclerosis, global Acat1 knockout (Acat1(-/-)) did not prevent lesion development. Acat1(-/-) increased apoptosis within lesions and led to several additional undesirable phenotypes, including hair loss, dry eye, leukocytosis, xanthomatosis, and a reduced life span. To determine the roles of Acat1 in monocytes/macrophages in atherosclerosis, we produced a myeloid-specific Acat1 knockout (Acat1(-M/-M)) mouse and showed that, in the Apoe knockout (Apoe(-/-)) mouse model for atherosclerosis, Acat1(-M/-M) decreased the plaque area and reduced lesion size without causing leukocytosis, dry eye, hair loss, or a reduced life span. Acat1(-M/-M) enhanced xanthomatosis in apoe(-/-) mice, a skin disease that is not associated with diet-induced atherosclerosis in humans. Analyses of atherosclerotic lesions showed that Acat1(-M/-M) reduced macrophage numbers and diminished the cholesterol and cholesteryl ester load without causing detectable apoptotic cell death. Leukocyte migration analysis in vivo showed that Acat1(-M/-M) caused much fewer leukocytes to appear at the activated endothelium. Studies in inflammatory (Ly6C(hi)-positive) monocytes and in cultured macrophages showed that inhibiting ACAT1 by gene knockout or by pharmacological inhibition caused a significant decrease in integrin ß 1 (CD29) expression in activated monocytes/macrophages. The sparse presence of lesion macrophages without Acat1 can therefore, in part, be attributed to decreased interaction between inflammatory monocytes/macrophages lacking Acat1 and the activated endothelium. We conclude that targeting ACAT1 in a myeloid cell lineage suppresses atherosclerosis progression while avoiding many of the undesirable side effects caused by global Acat1 inhibition.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Atherosclerosis/immunology , Macrophages/immunology , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , B-Lymphocytes/metabolism , Bone Marrow/pathology , Cell Movement , Cell Proliferation , Diet, High-Fat/adverse effects , Disease Progression , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Female , Hematopoietic Stem Cells/physiology , Leukocytosis/genetics , Leukocytosis/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/enzymology
18.
J Virol ; 89(4): 2405-14, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505074

ABSTRACT

UNLABELLED: Viral infection results in the generation of massive numbers of activated effector CD8(+) T cells that recognize viral components. Most of these are short-lived effector T cells (SLECs) that die after clearance of the virus. However, a small proportion of this population survives and forms antigen-specific memory precursor effector cells (MPECs), which ultimately develop into memory cells. These can participate in a recall response upon reexposure to antigen even at protracted times postinfection. Here, antiapoptotic myeloid cell leukemia 1 (MCL1) was found to prolong survival upon T cell stimulation, and mice expressing human MCL1 as a transgene exhibited a skewing in the proportion of CD8(+) T cells, away from SLECs toward MPECs, during the acute phase of vaccinia virus infection. A higher frequency and total number of antigen-specific CD8(+) T cells were observed in MCL1 transgenic mice. These findings show that MCL1 can shape the makeup of the CD8(+) T cell response, promoting the formation of long-term memory. IMPORTANCE: During an immune response to a virus, CD8(+) T cells kill cells infected by the virus, and most die when the infection resolves. However, a small proportion of cells survives and differentiates into long-lived memory cells that confer protection from reinfection by the same virus. This report shows that transgenic expression of an MCL1 protein enhances survival of memory CD8(+) T cells following infection with vaccinia virus. This is important because it shows that MCL1 expression may be an important determinant of the formation of long-term CD8(+) T cell memory.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Vaccinia virus/immunology , Vaccinia/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Survival , Disease Models, Animal , Humans , Immunologic Memory , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/genetics
19.
J Neurosci ; 34(43): 14484-501, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25339759

ABSTRACT

Acyl-CoA:cholesterol acyltransferase 1 (ACAT1) is a resident endoplasmic reticulum enzyme that prevents the buildup of cholesterol in membranes by converting it to cholesterol esters. Blocking ACAT1 pharmacologically or by Acat1 gene knock-out (KO) decreases amyloidopathy in mouse models for Alzheimer's disease. However, the beneficial actions of ACAT1 blockage to treat Alzheimer's disease remained not well understood. Microglia play essential roles in the proteolytic clearance of amyloid ß (Aß) peptides. Here we show that Acat1 gene KO in mouse increases phagocytic uptake of oligomeric Aß1-42 and stimulates lysosomal Aß1-42 degradation in cultured microglia and in vivo. Additional results show that Acat1 gene KO or a specific ACAT1 inhibitor K604 stimulates autophagosome formation and transcription factor EB-mediated lysosomal proteolysis. Surprisingly, the effect of ACAT1 blockage does not alter mTOR signaling or endoplasmic reticulum stress response but can be modulated by agents that disrupt cholesterol biosynthesis. To our knowledge, our current study provides the first example that a small molecule (K604) can promote autophagy in an mTOR-independent manner to activate the coordinated lysosomal expression and regulation network. Autophagy is needed to degrade misfolded proteins/peptides. Our results implicate that blocking ACAT1 may provide a new way to benefit multiple neurodegenerative diseases.


Subject(s)
Amyloid beta-Peptides/metabolism , Autophagy/physiology , Lysosomes/metabolism , Microglia/metabolism , Peptide Fragments/metabolism , Proteolysis , Sterol O-Acyltransferase/deficiency , Animals , Animals, Newborn , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Transport/physiology
20.
Arterioscler Thromb Vasc Biol ; 33(9): 2081-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23846496

ABSTRACT

OBJECTIVE: To investigate the role of acyl-CoA:cholesterol acyltransferase 1 (ACAT1) in hematopoiesis. APPROACH AND RESULTS: ACAT1 converts cellular cholesterol to cholesteryl esters for storage in multiple cell types and is a potential drug target for human diseases. In mouse models for atherosclerosis, global Acat1 knockout causes increased lesion size; bone marrow transplantation experiments suggest that the increased lesion size might be caused by ACAT1 deficiency in macrophages. However, bone marrow contains hematopoietic stem cells that give rise to cells in myeloid and lymphoid lineages; these cell types affect atherosclerosis at various stages. Here, we test the hypothesis that global Acat1(-/-) may affect hematopoiesis, rather than affecting macrophage function only, and show that Acat1(-/-) mice contain significantly higher numbers of myeloid cells and other cells than wild-type mice. Detailed analysis of bone marrow cells demonstrated that Acat1(-/-) causes a higher proportion of the stem cell-enriched Lin(-)Sca-1(+)c-Kit(+) population to proliferate, resulting in higher numbers of myeloid progenitor cells. In addition, we show that Acat1(-/-) causes higher monocytosis in Apoe(-/-) mouse during atherosclerosis development. CONCLUSIONS: ACAT1 plays important roles in hematopoiesis in normal mouse and in Apoe(-/-) mouse during atherosclerosis development.


Subject(s)
Acetyl-CoA C-Acetyltransferase/deficiency , Atherosclerosis/enzymology , Bone Marrow Cells/enzymology , Cell Proliferation , Hematopoiesis , Hematopoietic Stem Cells/enzymology , Leukocytosis/enzymology , Acetyl-CoA C-Acetyltransferase/genetics , Animals , Antigens, Ly/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/immunology , Biomarkers/metabolism , Cell Lineage , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genotype , Leukocytosis/genetics , Leukocytosis/immunology , MAP Kinase Signaling System , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Precursor Cells, B-Lymphoid/enzymology , Proto-Oncogene Proteins c-kit/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL