Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.875
Filter
Add more filters

Publication year range
1.
Environ Geochem Health ; 46(2): 61, 2024 Jan 28.
Article in English | MEDLINE | ID: mdl-38281271

ABSTRACT

The objective of this study was to investigate the effects of anthracene (Ant) with 3 rings, benzo[a]anthracene (BaA) with 4 rings and benzo[b]fluoranthene (BbF) with 5 rings in fine particulate matter (PM2.5) at different exposure times (4 h and 24 h) and low exposure levels (0 pg/mL, 0.1 pg/mL, 1 pg/mL, 100 pg/mL and 10,000 pg/mL) on RAW264.7 cells. The changes of interleukin-6 (IL-6) and oxidative stress levels in RAW264.7 cells were investigated by methyl-thiazolyl-tetrazolium (MTT) and enzyme-linked immunosorbent assay (ELISA). Pearson correlation analysis was used to analyze the correlation between variables. Ant, BaA and BbF induced the secretion of IL-6 and the occurrence of oxidative stress in RAW264.7 cells. The inflammatory effect and oxidative damage were exacerbated with prolonged exposure time, increasing exposure concentration and increasing number of PAH rings. At the same time, IL-6 was found to have a certain correlation with the levels of ROS, MDA and SOD. Exposure to atmospheric PAHs at low concentrations can also produce toxic effects on cells, IL-6 and oxidative stress work together in cell damage. The study is expected to provide a theoretical and experimental basis for air pollution control and human health promotion.


Subject(s)
Air Pollutants , Polycyclic Aromatic Hydrocarbons , Air Pollutants/toxicity , Anthracenes/toxicity , Interleukin-6 , Macrophages/chemistry , Oxidative Stress , Particulate Matter/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Animals , Mice , RAW 264.7 Cells
2.
Xenobiotica ; 53(12): 653-669, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38014489

ABSTRACT

Occupational exposure to the most widely used monomeric diisocyanate (dNCO), 4,4'-methylene diphenyl diisocyanate (MDI), may lead to the development of occupational asthma (OA). Alveolar macrophages with alternatively activated (M2) phenotype have been implicated in allergic airway responses and the pathogenesis of asthma. Recent in vivo studies demonstrate that M2 macrophage-associated markers and chemokines are induced by MDI-exposure, however, the underlying molecular mechanism(s) by which this proceeds is unclear.Following MDI exposure (in vivo and in vitro) M2 macrophage-associated transcription factors (TFs), markers, and chemokines were determined by RT-qPCR, western blots, and ELISA.Expression of M2 macrophage-associated TFs and markers including Klf4/KLF4, Cd206/CD206, Tgm2/TGM2, Ccl17/CCL17, Ccl22/CCL22, and CCL24 were induced by MDI/MDI-GSH exposure in bronchoalveolar lavage cells (BALCs)/THP-1 macrophages. The expression of CD206, TGM2, CCL17, CCL22, and CCL24 are upregulated by 3.83-, 7.69-, 6.22-, 6.08-, and 1.90-fold in KLF4-overexpressed macrophages, respectively. Endogenous CD206 and TGM2 were downregulated by 1.65-5.17-fold, and 1.15-1.78-fold, whereas CCL17, CCL22, and CCL24 remain unchanged in KLF4-knockdown macrophages. Finally, MDI-glutathione (GSH) conjugate-treated macrophages show increased chemotactic ability to T-cells and eosinophils, which may be attenuated by KLF4 knockdown.Our data suggest that MDI exposure may induce M2 macrophage-associated markers partially through induction of KLF4.


Subject(s)
Asthma, Occupational , Kruppel-Like Factor 4 , Humans , Isocyanates/toxicity , Asthma, Occupational/chemically induced , Macrophages/chemistry , Chemokines/toxicity
3.
PLoS Genet ; 16(5): e1008786, 2020 05.
Article in English | MEDLINE | ID: mdl-32392242

ABSTRACT

Allele-specific expression (ASE) analysis, which quantifies the relative expression of two alleles in a diploid individual, is a powerful tool for identifying cis-regulated gene expression variations that underlie phenotypic differences among individuals. Existing methods for gene-level ASE detection analyze one individual at a time, therefore failing to account for shared information across individuals. Failure to accommodate such shared information not only reduces power, but also makes it difficult to interpret results across individuals. However, when only RNA sequencing (RNA-seq) data are available, ASE detection across individuals is challenging because the data often include individuals that are either heterozygous or homozygous for the unobserved cis-regulatory SNP, leading to sample heterogeneity as only those heterozygous individuals are informative for ASE, whereas those homozygous individuals have balanced expression. To simultaneously model multi-individual information and account for such heterogeneity, we developed ASEP, a mixture model with subject-specific random effect to account for multi-SNP correlations within the same gene. ASEP only requires RNA-seq data, and is able to detect gene-level ASE under one condition and differential ASE between two conditions (e.g., pre- versus post-treatment). Extensive simulations demonstrated the convincing performance of ASEP under a wide range of scenarios. We applied ASEP to a human kidney RNA-seq dataset, identified ASE genes and validated our results with two published eQTL studies. We further applied ASEP to a human macrophage RNA-seq dataset, identified genes showing evidence of differential ASE between M0 and M1 macrophages, and confirmed our findings by results from cardiometabolic trait-relevant genome-wide association studies. To the best of our knowledge, ASEP is the first method for gene-level ASE detection at the population level that only requires the use of RNA-seq data. With the growing adoption of RNA-seq, we believe ASEP will be well-suited for various ASE studies for human diseases.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Quantitative Trait Loci , Sequence Analysis, RNA/methods , Alleles , Female , Gene Expression Regulation , Genetics, Population , Humans , Kidney/chemistry , Macrophages/chemistry , Models, Genetic , Software
4.
Int J Mol Sci ; 24(14)2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37511404

ABSTRACT

Titanium dental implants are one of the modalities to replace missing teeth. The release of titanium particles from the implant's surface may modulate the immune cells, resulting in implant failure. However, little is known about the immune microenvironment that plays a role in peri-implant inflammation as a consequence of titanium particles. In this study, the peri-implant gingival tissues were collected from patients with failed implants, successful implants and no implants, and then a whole transcriptome analysis was performed. The gene set enrichment analysis confirmed that macrophage M1/M2 polarization and lymphocyte proliferation were differentially expressed between the study groups. The functional clustering and pathway analysis of the differentially expressed genes between the failed implants and successful implants versus no implants revealed that the immune response pathways were the most common in both comparisons, implying the critical role of infiltrating immune cells in the peri-implant tissues. The H&E and IHC staining confirmed the presence of titanium particles and immune cells in the tissue samples, with an increase in the infiltration of lymphocytes and macrophages in the failed implant samples. The in vitro validation showed a significant increase in the level of IL-1ß, IL-8 and IL-18 expression by macrophages. Our findings showed evidence that titanium particles modulate lymphocyte and macrophage polarization in peri-implant gingival tissues, which can help in the understanding of the imbalance in osteoblast-osteoclast activity and failure of dental implant osseointegration.


Subject(s)
Dental Implants , Titanium , Humans , Titanium/adverse effects , Titanium/analysis , Gingiva , Lymphocytes/chemistry , Macrophages/chemistry , Inflammation , Dental Implants/adverse effects
5.
Anal Chem ; 94(2): 658-668, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34936345

ABSTRACT

Monitoring the secretion of proteins from single cells can provide important insights into how cells respond to their microenvironment. This is particularly true for immune cells, which can exhibit a large degree of response heterogeneity. Microfabricated well arrays provide a powerful and versatile method to assess the secretion of cytokines, chemokines, and growth factors from single cells, but detection sensitivity has been limited to high levels on the order of 10,000 per cell. Recently, we reported a quantum dot-based immunoassay that lowered the detection limit for the cytokine TNF-α to concentrations to nearly the single-cell level. Here, we adapted this detection method to three additional targets while maintaining high detection sensitivity. Specifically, we detected MCP-1, TGF-ß, IL-10, and TNF-α using quantum dots with different emission spectra, each of which displayed a detection threshold in the range of 1-10 fM or ∼1-2 molecules per well. We then quantified secretion of all four proteins from single macrophage cells that were stimulated toward a pro-inflammatory state with lipopolysaccharide (LPS) or toward a pro-healing state with both LPS and interleukin 4 (IL-4). We found that MCP-1 and TGF-ß were predominantly secreted at high levels only (>10,000 molecules/cell), while a substantial number of cells secreted IL-10 and TNF-α at lower levels that could only be detected using our method. Subsequent principal component and cluster analysis revealed that secretion profiles could be classified as either exclusively pro-inflammatory, including MCP-1 and/or TNF-α, or more subtle responses displaying both pro-healing and pro-inflammatory characters. Our results highlight the heterogeneous and nondiscrete nature of macrophage phenotypes following in vitro stimulation of a cell line. Future work will focus on expanding the multiplexing capacity by extending emission spectra bandwidth and/or spatially barcoding capture antibodies, as well as evaluating the enhanced detection sensitivity capabilities with normal and diseased immune cell populations in vitro and in vivo.


Subject(s)
Cytokines , Tumor Necrosis Factor-alpha , Cytokines/analysis , Immunoassay/methods , Lipopolysaccharides/pharmacology , Macrophages/chemistry , Tumor Necrosis Factor-alpha/analysis
6.
Cell Biol Toxicol ; 38(1): 31-41, 2022 02.
Article in English | MEDLINE | ID: mdl-34021430

ABSTRACT

Anti-inflammatory and proinflammatory responses in macrophages are influenced by cellular metabolism. Macrophages are the primary phagocyte in mucosal environments (i.e., intestinal tract and lungs) acting as first-line defense against microorganisms and environmental pollutants. Given the extensive contamination of our food and water sources with microplastics, we aimed to examine the metabolic response in macrophages to microplastic particles (MPs). Utilizing murine macrophages, we assessed the metabolic response of macrophages after polystyrene MP phagocytosis. The phagocytosis of MP by macrophages induced a metabolic shift toward glycolysis and a reduction in mitochondrial respiration that was associated with an increase of cell surface markers CD80 and CD86 and cytokine gene expression associated with glycolysis. The gastrointestinal consequences of this metabolic switch in the context of an immune response remain uncertain, but the global rise of plastic pollution and MP ingestion potentially poses an unappreciated health risk. Macrophage phagocytosis of microplastics alters cellular metabolism. - Macrophages cannot degrade PS MP. - MP phagocytosis increases glycolysis in murine macrophages. - MP phagocytosis reduces mitochondrial respiration in murine macrophages.


Subject(s)
Microplastics , Water Pollutants, Chemical , Animals , Gastrointestinal Tract , Macrophages/chemistry , Mice , Microplastics/toxicity , Plastics , Polystyrenes/toxicity , Water Pollutants, Chemical/analysis
7.
Mol Ther ; 29(8): 2554-2570, 2021 08 04.
Article in English | MEDLINE | ID: mdl-33887461

ABSTRACT

Mesenchymal stromal cell (MSC) transplantation has been investigated as an advanced treatment of heart failure; however, further improvement of the therapeutic efficacy and mechanistic understanding are needed. Our previous study has reported that epicardial placement of fibrin sealant films incorporating rat amniotic membrane-derived (AM)-MSCs (MSC-dressings) could address limitations of traditional transplantation methods. To progress this finding toward clinical translation, this current study aimed to examine the efficacy of MSC-dressings using human AM-MSCs (hAM-MSCs) and the underpinning mechanism for myocardial repair. Echocardiography demonstrated that cardiac function and structure were improved in a rat ischemic cardiomyopathy model after hAM-MSC-dressing therapy. hAM-MSCs survived well in the rat heart, enhanced myocardial expression of reparative genes, and attenuated adverse remodeling. Copy number analysis by qPCR revealed that upregulated reparative genes originated from endogenous rat cells rather than hAM-MSCs. These results suggest hAM-MSC-dressing therapy stimulates a secondary release of paracrine factors from endogenous cells improving myocardial repair ("secondary paracrine effect"), and cardiac M2-like macrophages were identified as a potential cell source of repair. We demonstrated hAM-MSCs increased M2-like macrophages through not only enhancing M2 polarization but also augmenting their proliferation and migration capabilities via PGE2, CCL2, and TGF-ß1, resulting in enhanced cardiac function after injury.


Subject(s)
Fibrin/chemistry , Heart Failure/therapy , Macrophages/cytology , Mesenchymal Stem Cells/cytology , Animals , Cell Polarity , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Echocardiography , Female , Gene Expression Regulation , Heart Failure/diagnostic imaging , Heart Failure/genetics , Humans , Macrophages/chemistry , Mesenchymal Stem Cell Transplantation , Mice , Rats
8.
Proc Natl Acad Sci U S A ; 116(51): 25649-25658, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31757855

ABSTRACT

Phthiocerol dimycocerosate (DIM) is a major virulence factor of the pathogen Mycobacterium tuberculosis (Mtb). While this lipid promotes the entry of Mtb into macrophages, which occurs via phagocytosis, its molecular mechanism of action is unknown. Here, we combined biophysical, cell biology, and modeling approaches to reveal the molecular mechanism of DIM action on macrophage membranes leading to the first step of Mtb infection. Matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry showed that DIM molecules are transferred from the Mtb envelope to macrophage membranes during infection. Multiscale molecular modeling and 31P-NMR experiments revealed that DIM adopts a conical shape in membranes and aggregates in the stalks formed between 2 opposing lipid bilayers. Infection of macrophages pretreated with lipids of various shapes uncovered a general role for conical lipids in promoting phagocytosis. Taken together, these results reveal how the molecular shape of a mycobacterial lipid can modulate the biological response of macrophages.


Subject(s)
Lipids/chemistry , Macrophages/microbiology , Mycobacterium tuberculosis , Tuberculosis/microbiology , Cell Line , Cell Membrane/chemistry , Cell Membrane/microbiology , Host-Pathogen Interactions/physiology , Humans , Macrophages/chemistry , Molecular Dynamics Simulation , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/physiology , Nuclear Magnetic Resonance, Biomolecular
9.
BMC Biol ; 19(1): 45, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33706790

ABSTRACT

BACKGROUND: Nucleic acids can fold into non-canonical secondary structures named G-quadruplexes (G4s), which consist of guanine-rich sequences stacked into guanine tetrads stabilized by Hoogsteen hydrogen bonding, π-π interactions, and monovalent cations. G4 structure formation and properties are well established in vitro, but potential in vivo functions remain controversial. G4s are evolutionarily enriched at distinct, functional genomic loci, and both genetic and molecular findings indicate that G4s are involved in multiple aspects of cellular homeostasis. In order to gain a deeper understanding of the function of G4 structures and the trigger signals for their formation, robust biochemical methods are needed to detect and quantify G4 structures in living cells. Currently available methods mostly rely on fluorescence microscopy or deep sequencing of immunoprecipitated DNA or RNA using G4-specific antibodies. These methods provide a clear picture of the cellular or genomic localization of G4 structures but are very time-consuming. Here, we assembled a novel protocol that uses the G4-specific antibody BG4 to quantify G4 structures by flow cytometry (BG-flow). RESULTS: We describe and validate a flow cytometry-based protocol for quantifying G4 levels by using the G4-specific antibody BG4 to label standard cultured cells (Hela and THP-1) as well as primary cells obtained from human blood (peripheral blood mononuclear cells (PBMCs)). We additionally determined changes in G4 levels during the cell cycle in immortalized MCF-7 cells, and validated changes previously observed in G4 levels by treating mouse macrophages with the G4-stabilizing agent pyridostatin (PDS). CONCLUSION: We provide mechanistic proof that BG-flow is working in different kinds of cells ranging from mouse to humans. We propose that BG-flow can be combined with additional antibodies for cell surface markers to determine G4 structures in subpopulations of cells, which will be beneficial to address the relevance and consequences of G4 structures in mixed cell populations. This will support ongoing research that discusses G4 structures as a novel diagnostic tool.


Subject(s)
Flow Cytometry/methods , G-Quadruplexes , Leukocytes, Mononuclear/chemistry , Macrophages/chemistry , Animals , HeLa Cells , Humans , Mice , THP-1 Cells
10.
Int J Mol Sci ; 23(3)2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35163544

ABSTRACT

Understanding the interaction between nanoparticles and immune cells is essential for the evaluation of nanotoxicity and development of nanomedicines. However, to date, there is little data on the membrane microstructure and biochemical changes in nanoparticle-loaded immune cells. In this study, we observed the microstructure of nanoparticle-loaded macrophages and changes in lipid droplets using holotomography analysis. Quantitatively analyzing the refractive index distribution of nanoparticle-loaded macrophages, we identified the interactions between nanoparticles and macrophages. The results showed that, when nanoparticles were phagocytized by macrophages, the number of lipid droplets and cell volume increased. The volume and mass of the lipid droplets slightly increased, owing to the absorption of nanoparticles. Meanwhile, the number of lipid droplets increased more conspicuously than the other factors. Furthermore, alveolar macrophages are involved in the development and progression of asthma. Studies have shown that macrophages play an essential role in the maintenance of asthma-related inflammation and tissue damage, suggesting that macrophage cells may be applied to asthma target delivery strategies. Therefore, we investigated the target delivery efficiency of gold nanoparticle-loaded macrophages at the biodistribution level, using an ovalbumin-induced asthma mouse model. Normal and severe asthma models were selected to determine the difference in the level of inflammation in the lung. Consequently, macrophages had increased mobility in models of severe asthma, compared to those of normal asthma disease. In this regard, the detection of observable differences in nanoparticle-loaded macrophages may be of primary interest, as an essential endpoint analysis for investigating nanomedical applications and immunotheragnostic strategies.


Subject(s)
Asthma/diagnostic imaging , Gold/pharmacokinetics , Lipopolysaccharides/adverse effects , Lung/chemistry , Macrophages/transplantation , Ovalbumin/adverse effects , Animals , Asthma/chemically induced , Asthma/metabolism , Disease Models, Animal , Drug Delivery Systems , Feasibility Studies , Female , Lung/diagnostic imaging , Macrophages/chemistry , Macrophages/cytology , Macrophages/drug effects , Metal Nanoparticles , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Tissue Distribution , Tomography
11.
Microbiology (Reading) ; 167(5)2021 05.
Article in English | MEDLINE | ID: mdl-34020726

ABSTRACT

The production of itaconate by macrophages was only discovered in 2011. An increasing number of studies have since revealed essential biological functions for this small molecule, ranging from antimicrobial to immunomodulator. The antibacterial role of itaconate has however been questioned because the estimated concentration of itaconate in macrophages (low-millimolar) is lower than the minimum inhibitory concentration (MIC) of itaconate reported for several bacterial strains (low-to-mid-millimolar). We note that some of these investigations have tended to ignore the high acidity of this small diacid (pKas 3.85 and 5.45), thereby potentially biassing activity measurements. We measured the MIC of itaconate in Escherichia coli (not known to metabolize itaconate) and in Salmonella enterica serovar Typhimurium (known to metabolize itaconate) at varying pH values to probe the effect that pH has on itaconate toxicity. Herein, we demonstrate that the antimicrobial effect of itaconate is dependent upon the pH of the media and that itaconate does have antimicrobial activity at biologically relevant pH and concentrations. Under nutrient-poor conditions, the antimicrobial activity of itaconate in both E. coli and S. Typhimurium increased approximately 200-fold when the pH was dropped by one unit, whereas itaconate was not found to be toxic under nutrient rich conditions. Our results also reveal that the activity of itaconate is synergistic with acidity, yet is not a function of increased permeability with protonation. Similar experiments performed with succinate (a pKa-matched diacid) yielded drastically different results, consistent with a target-based mechanism of action for itaconate. Overall, our work shows the importance of controlling the pH when performing experiments with itaconic acid.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Macrophages/chemistry , Succinates/chemistry , Succinates/pharmacology , Anti-Bacterial Agents/metabolism , Culture Media/chemistry , Culture Media/metabolism , Escherichia coli/drug effects , Hydrogen-Ion Concentration , Macrophages/metabolism , Microbial Sensitivity Tests , Salmonella typhimurium/drug effects , Succinates/metabolism
12.
Biol Chem ; 402(11): 1289-1307, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34390641

ABSTRACT

Nonhealing chronic wounds are among the most common skin disorders with increasing incidence worldwide. However, their treatment is still dissatisfying, that is why novel therapeutic concepts targeting the sustained inflammatory process have emerged. Increasing understanding of chronic wound pathologies has put macrophages in the spotlight of such approaches. Herein, we review current concepts and perspectives of therapeutic macrophage control by ECM-inspired wound dressing materials. We provide an overview of the current understanding of macrophage diversity with particular view on their roles in skin and in physiological and disturbed wound healing processes. Based on this we discuss strategies for their modulation in chronic wounds and how such strategies can be tailored in ECM-inspired wound dressing. The latter utilize and mimic general principles of ECM-mediated cell control, such as binding and delivery of signaling molecules and direct signaling to cells specifically adapted for macrophage regulation in wounds. In this review, we present examples of most recent approaches and discuss ideas for their further development.


Subject(s)
Extracellular Matrix/metabolism , Macrophages/metabolism , Extracellular Matrix/chemistry , Humans , Macrophages/chemistry , Wound Healing
13.
Biol Chem ; 402(11): 1309-1324, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34392640

ABSTRACT

Controlled wound healing requires a temporal and spatial coordination of cellular activities within the surrounding extracellular matrix (ECM). Disruption of cell-cell and cell-matrix communication results in defective repair, like chronic or fibrotic wounds. Activities of macrophages and fibroblasts crucially contribute to the fate of closing wounds. To investigate the influence of the ECM as an active part controlling cellular behavior, coculture models based on fibrillar 3D biopolymers such as collagen have already been successfully used. With well-defined biochemical and biophysical properties such 3D scaffolds enable in vitro studies on cellular processes including infiltration and differentiation in an in vivo like microenvironment. Further, paracrine and autocrine signaling as well as modulation of soluble mediator transport inside the ECM can be modeled using fibrillar 3D scaffolds. Herein, we review the usage of these scaffolds in in vitro coculture models allowing in-depth studies on the crosstalk between macrophages and fibroblasts during different stages of cutaneous wound healing. A more accurate mimicry of the various processes of cellular crosstalk at the different stages of wound healing will contribute to a better understanding of the impact of biochemical and biophysical environmental parameters and help to develop further strategies against diseases such as fibrosis.


Subject(s)
Biopolymers/metabolism , Extracellular Matrix/metabolism , Fibrillar Collagens/metabolism , Macrophages/metabolism , Biopolymers/chemistry , Extracellular Matrix/chemistry , Fibrillar Collagens/chemistry , Humans , Macrophages/chemistry , Wound Healing
14.
Mol Pharm ; 18(7): 2495-2506, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34078087

ABSTRACT

Pancreatic cancer is a lethal malignancy with a dismal prognosis. Gemcitabine is currently used to treat pancreatic cancer, but it is limited by significant toxicity. Clinical trials on the combination of gemcitabine and erlotinib reported unsatisfactory outcomes along with concerns of toxicity. The encapsulation of chemotherapy drugs in polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) can alleviate toxicity through targeted delivery and sustained release. In addition, camouflaging the NPs with a macrophage membrane can evade the immune system and further improve tumor homing. We designed gemcitabine-loaded PLGA NPs with a macrophage membrane coating (MPGNPs) to reduce drug toxicity and increase the accumulation in the tumor. The combination of MPGNPs and erlotinib synergistically inhibited pancreatic cancer cell proliferation in vitro and in vivo by targeting the PI3K/AKT/mTOR and Ras/Raf/MEK/ERK signaling pathways. The MPGNPs were also able to evade phagocytosis and achieve passive targeting to the pancreatic tumors. The combination of MPGNPs and erlotinib showed synergistic anti-tumor efficacy in vitro and in vivo. This study provides a proof-of-concept for treating pancreatic cancer with a combination of MPGNPs and erlotinib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Carriers/chemistry , Drug Liberation , Macrophages/chemistry , Nanoparticles/administration & dosage , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Apoptosis , Cell Proliferation , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Erlotinib Hydrochloride/administration & dosage , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polyesters , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
15.
Circ Res ; 125(12): 1038-1051, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31594470

ABSTRACT

RATIONALE: Macrophages are essential regulators of atherosclerosis. They secrete cytokines, process lipoproteins and cholesterol, and take up apoptotic cells. Multiple subsets of plaque macrophages exist and their differential roles are emerging. OBJECTIVE: Here, we explore macrophage heterogeneity in atherosclerosis plaques using transgenic fluorescent mice in which subsets of macrophages are labeled by GFP (green fluorescent protein), YFP (yellow fluorescent protein), neither, or both. The objective was to define migration patterns of the visible subsets and relate them to their phenotypes and transcriptomes. METHODS AND RESULTS: Apoe-/-Cx3cr1GFPCd11cYFP mice have 4 groups of macrophages in their aortas. The 3 visible subsets show varying movement characteristics. GFP and GFP+YFP+ macrophages extend and retract dendritic processes, dancing on the spot with little net movement while YFP macrophages have a more rounded shape and migrate along the arteries. RNA sequencing of sorted cells revealed significant differences in the gene expression patterns of the 4 subsets defined by GFP and YFP expression, especially concerning chemokine and cytokine expression, matrix remodeling, and cell shape dynamics. Gene set enrichment analysis showed that GFP+ cells have similar transcriptomes to cells found in arteries with tertiary lymphoid organs and regressing plaques while YFP+ cells were associated with progressing and stable plaques. CONCLUSIONS: The combination of quantitative intravital imaging with deep transcriptomes identified 4 subsets of vascular macrophages in atherosclerosis that have unique transcriptomic profiles. Our data link vascular macrophage transcriptomes to their in vivo migratory function. Future work on the functional significance of the change in gene expression and motility characteristics will be needed to fully understand how these subsets contribute to disease progression.


Subject(s)
Atherosclerosis/pathology , Cell Movement/physiology , Macrophages/pathology , Macrophages/physiology , Plaque, Atherosclerotic/pathology , Animals , Atherosclerosis/genetics , Bacterial Proteins/analysis , Female , Green Fluorescent Proteins/analysis , Luminescent Proteins/analysis , Macrophages/chemistry , Male , Mice , Mice, Transgenic , Plaque, Atherosclerotic/genetics
16.
Arterioscler Thromb Vasc Biol ; 40(6): 1491-1509, 2020 06.
Article in English | MEDLINE | ID: mdl-32295421

ABSTRACT

OBJECTIVE: Galectin-3 (formerly known as Mac-2), encoded by the LGALS3 gene, is proposed to regulate macrophage adhesion, chemotaxis, and apoptosis. We investigated the role of galectin-3 in determining the inflammatory profile of macrophages and composition of atherosclerotic plaques. Approach and Results: We observed increased accumulation of galectin-3-negative macrophages within advanced human, rabbit, and mouse plaques compared with early lesions. Interestingly, statin treatment reduced galectin-3-negative macrophage accrual in advanced plaques within hypercholesterolemic (apolipoprotein E deficient) Apoe-/- mice. Accordingly, compared with Lgals3+/+:Apoe-/- mice, Lgals3-/-:Apoe-/- mice displayed altered plaque composition through increased macrophage:smooth muscle cell ratio, reduced collagen content, and increased necrotic core area, characteristics of advanced plaques in humans. Additionally, macrophages from Lgals3-/- mice exhibited increased invasive capacity in vitro and in vivo. Furthermore, loss of galectin-3 in vitro and in vivo was associated with increased expression of proinflammatory genes including MMP (matrix metalloproteinase)-12, CCL2 (chemokine [C-C motif] ligand 2), PTGS2 (prostaglandin-endoperoxide synthase 2), and IL (interleukin)-6, alongside reduced TGF (transforming growth factor)-ß1 expression and consequent SMAD signaling. Moreover, we found that MMP12 cleaves macrophage cell-surface galectin-3 resulting in the appearance of a 22-kDa fragment, whereas plasma levels of galectin-3 were reduced in Mmp12-/-:Apoe-/- mice, highlighting a novel mechanism where MMP12-dependent cleavage of galectin-3 promotes proinflammatory macrophage polarization. Moreover, galectin-3-positive macrophages were more abundant within plaques of Mmp12-/-:Apoe-/- mice compared with Mmp12+/+:Apoe-/- animals. CONCLUSIONS: This study reveals a prominent protective role for galectin-3 in regulating macrophage polarization and invasive capacity and, therefore, delaying plaque progression.


Subject(s)
Atherosclerosis/pathology , Galectin 3/physiology , Macrophages/physiology , Animals , Crosses, Genetic , Female , Galectin 3/analysis , Galectin 3/deficiency , Humans , Inflammation/pathology , Macrophages/chemistry , Macrophages/pathology , Male , Matrix Metalloproteinase 12/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Middle Aged , Plaque, Atherosclerotic/pathology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
17.
Arterioscler Thromb Vasc Biol ; 40(6): e153-e165, 2020 06.
Article in English | MEDLINE | ID: mdl-32295422

ABSTRACT

OBJECTIVE: Macrophages have been described in calcific aortic valve disease, but it is unclear if they promote or counteract calcification. We aimed to determine how macrophages are involved in calcification using the Notch1+/- model of calcific aortic valve disease. Approach and Results: Macrophages in wild-type and Notch1+/- murine aortic valves were characterized by flow cytometry. Macrophages in Notch1+/- aortic valves had increased expression of MHCII (major histocompatibility complex II). We then used bone marrow transplants to test if differences in Notch1+/- macrophages drive disease. Notch1+/- mice had increased valve thickness, macrophage infiltration, and proinflammatory macrophage maturation regardless of transplanted bone marrow genotype. In vitro approaches confirm that Notch1+/- aortic valve cells promote macrophage invasion as quantified by migration index and proinflammatory phenotypes as quantified by Ly6C and CCR2 positivity independent of macrophage genotype. Finally, we found that macrophage interaction with aortic valve cells promotes osteogenic, but not dystrophic, calcification and decreases abundance of the STAT3ß isoform. CONCLUSIONS: This study reveals that Notch1+/- aortic valve disease involves increased macrophage recruitment and maturation driven by altered aortic valve cell secretion, and that increased macrophage recruitment promotes osteogenic calcification and alters STAT3 splicing. Further investigation of STAT3 and macrophage-driven inflammation as therapeutic targets in calcific aortic valve disease is warranted.


Subject(s)
Aortic Valve Stenosis/pathology , Aortic Valve/pathology , Calcinosis/pathology , Macrophages/physiology , STAT3 Transcription Factor/physiology , Animals , Aortic Valve/immunology , Aortic Valve/physiopathology , Aortic Valve Stenosis/immunology , Aortic Valve Stenosis/physiopathology , Bone Marrow Transplantation , Calcinosis/immunology , Calcinosis/physiopathology , Cell Movement , Cyclic S-Oxides/pharmacology , Disease Models, Animal , Gene Expression , Genotype , Humans , Inflammation/pathology , Macrophages/chemistry , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis , Receptor, Notch1/analysis , Receptor, Notch1/genetics , Receptor, Notch1/physiology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics
18.
Arterioscler Thromb Vasc Biol ; 40(6): 1464-1478, 2020 06.
Article in English | MEDLINE | ID: mdl-32268789

ABSTRACT

OBJECTIVE: Despite the current antiatherosclerotic and antithrombotic therapies, the incidence of advanced atherosclerosis-associated clinical events remains high. Whether long noncoding RNAs (lncRNAs) affect the progression of atherosclerosis and whether they are potential targets for the treatment of advanced atherosclerosis are poorly understood. Approach and Results: The progression of atherosclerotic lesions was accompanied by dynamic alterations in lncRNA expression, as revealed by RNA sequencing and quantitative polymerase chain reaction. Among the dynamically changing lncRNAs, we identified a novel lncRNA, lncRNA Associated with the Progression and Intervention of Atherosclerosis (RAPIA), that was highly expressed in advanced atherosclerotic lesions and in macrophages. Inhibition of RAPIA in vivo not only repressed the progression of atherosclerosis but also exerted atheroprotective effects similar to those of atorvastatin on advanced atherosclerotic plaques that had already formed. In vitro assays demonstrated that RAPIA promoted proliferation and reduced apoptosis of macrophages. A molecular sponge interaction between RAPIA and microRNA-183-5p was demonstrated by dual-luciferase reporter and RNA immunoprecipitation assays. Rescue assays indicated that RAPIA functioned at least in part by targeting the microRNA-183-5p/ITGB1 (integrin ß1) pathway in macrophages. In addition, the transcription factor FoxO1 (forkhead box O1) could bind to the RAPIA promoter region and facilitate the expression of RAPIA. CONCLUSIONS: The progression of atherosclerotic lesions was accompanied by dynamic changes in the expression of lncRNAs. Inhibition of the pivotal lncRNA RAPIA may be a novel preventive and therapeutic strategy for advanced atherosclerosis, especially in patients resistant or intolerant to statins.


Subject(s)
Atherosclerosis/therapy , Gene Expression , Macrophages/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , Animals , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Atorvastatin/pharmacology , Cell Proliferation/drug effects , Disease Progression , Forkhead Box Protein O1/metabolism , Humans , Integrin beta1/metabolism , Macrophages/chemistry , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/metabolism , MicroRNAs/pharmacology , Promoter Regions, Genetic/physiology , RAW 264.7 Cells , RNA, Long Noncoding/physiology
19.
Nanomedicine ; 33: 102348, 2021 04.
Article in English | MEDLINE | ID: mdl-33321215

ABSTRACT

Atherosclerosis can lead to most cardiovascular diseases. Although some biomimetic nanomaterials coated by macrophage membranes have been reported in previous studies of atherosclerosis, to our knowledge, no studies regarding the detection of early lesions of atherosclerosis (foam cells) using such a strategy have yet been reported. In the present study, Fe3O4 biomimetic nanoparticles coated with a macrophage membrane (Fe3O4@M) were prepared to investigate the imaging effect on the early lesions of atherosclerosis (foam cells). The results showed that the Fe3O4@M particles are spheres with average diameters of approximately 300 nm. T1 and T2 relaxation values showed that the ratio of r2 to r1 was 26.09. The protein content accounted for approximately 27% of the total weight in Fe3O4@M, and Fe3O4@M nanoparticles exhibited high biosafety. Further testing showed that Fe3O4@M effectively targets early atherosclerotic lesions by the specific recognition of integrin α4ß1 to VCAM-1. Taken together, Fe3O4@M is a promising contrast agent for the diagnosis of early stage atherosclerosis.


Subject(s)
Biomimetic Materials/chemistry , Contrast Media/chemistry , Magnetite Nanoparticles/chemistry , Animals , Atherosclerosis , Cell Membrane Permeability , Cell Survival/drug effects , Humans , Macrophages/chemistry , Macrophages/metabolism , Magnetic Resonance Imaging , Mice , RAW 264.7 Cells , Surface Properties , Vascular Cell Adhesion Molecule-1/metabolism
20.
Nanomedicine ; 37: 102420, 2021 10.
Article in English | MEDLINE | ID: mdl-34182154

ABSTRACT

The treatment of spinal cord injury is still a challenge worldwide; there is still no effective method. Our strategy is to devise a macrophage-mediated degradable gelatin coated mesoporous silica nanoparticles, which could carry pirfenidone and realize spatiotemporal control of pirfenidone release in the lesion site. For the in vivo experiment, three groups of SD rats subjected to spinal cord contusion injury were injected with GNS-PFD, PFD or PBS. Spinal cord functions were observed. In vitro, we investigated the expression of inflammatory and anti-inflammatory factors. Spinal cord function and recovery were better in the GSN-PFD and PFD than the control group. In the in vitro study, the MMPs after SCI in lesion site were lower in the experimental group. Moreover, the expression of anti-inflammatory and inflammatory factors showed better in the experimental group. The inflammatory response of the PFD to time and space can be achieved with the loading of macrophage-mediated degradable gelatin coated mesoporous silica nanoparticles.


Subject(s)
Macrophages/chemistry , Nanoparticles/chemistry , Pyridones/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Gelatin/chemistry , Gelatin/pharmacology , Humans , Pyridones/chemistry , Rats , Rats, Sprague-Dawley , Recovery of Function , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Spinal Cord/drug effects , Spinal Cord/pathology
SELECTION OF CITATIONS
SEARCH DETAIL