Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 768
Filter
1.
Cell Rep ; 43(6): 114352, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38870011

ABSTRACT

Addressing the mononuclear phagocyte system (MPS) and macrophage M1/M2 activation is important in diagnosing hematological disorders and inflammatory pathologies and designing therapeutic tools. CSF1R is a reliable marker to identify all circulating MPS cells and tissue macrophages in humans using a single surface protein. CSF1R permits the quantification and isolation of monocyte and dendritic cell (DC) subsets in conjunction with CD14, CD16, and CD1c and is stable across the lifespan and sexes in the absence of overt pathology. Beyond cell detection, measuring M1/M2 activation in humans poses challenges due to response heterogeneity, transient signaling, and multiple regulation steps for transcripts and proteins. MPS cells respond in a conserved manner to M1/M2 pathways such as interleukin-4 (IL-4), steroids, interferon-γ (IFNγ), and lipopolysaccharide (LPS), for which we propose an ad hoc modular gene expression tool. Signature analysis highlights macrophage activation mosaicism in experimental samples, an emerging concept that points to mixed macrophage activation states in pathology.


Subject(s)
Macrophage Activation , Macrophages , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Macrophage Activation/genetics , Macrophages/metabolism , Macrophages/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interferon-gamma/metabolism , Lipopolysaccharides/pharmacology , Female , Mosaicism , Male , Monocytes/metabolism , Lipopolysaccharide Receptors/metabolism , Interleukin-4/metabolism , Dendritic Cells/metabolism , Dendritic Cells/immunology , Receptors, IgG/metabolism , Receptors, IgG/genetics , Antigens, CD1/metabolism , Antigens, CD1/genetics , Mononuclear Phagocyte System/metabolism , Glycoproteins , Receptor, Macrophage Colony-Stimulating Factor
2.
Nat Commun ; 15(1): 4366, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777821

ABSTRACT

Rapid uptake of nanoparticles by mononuclear phagocyte system (MPS) significantly hampers their therapeutic efficacy. Temporal MPS blockade is one of the few ways to overcome this barrier - the approach rediscovered many times under different names but never extensively used in clinic. Using meta-analysis of the published data we prove the efficacy of this technique for enhancing particle circulation in blood and their delivery to tumours, describe a century of its evolution and potential combined mechanism behind it. Finally, we discuss future directions of the research focusing on the features essential for successful clinical translation of the method.


Subject(s)
Drug Delivery Systems , Mononuclear Phagocyte System , Nanoparticles , Humans , Mononuclear Phagocyte System/metabolism , Nanoparticles/chemistry , Drug Delivery Systems/methods , Neoplasms/drug therapy , Neoplasms/immunology , Animals , Nanoparticle Drug Delivery System/chemistry
3.
Vet Immunol Immunopathol ; 266: 110681, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37992576

ABSTRACT

Macrophage colony-stimulating factor (CSF1) controls the proliferation and differentiation of cells of the mononuclear phagocyte system through binding to the receptor CSF1R. The expression and function of CSF1 has been well-studied in rodents and humans, but knowledge is lacking in other veterinary species. The development of a novel mouse anti-porcine CSF1 monoclonal antibody (mAb) facilitates the characterisation of this growth factor in pigs. Cell surface expression of CSF1 was confirmed on differentiated macrophage populations derived from blood and bone marrow monocytes, and on lung resident macrophages, the first species for this to be confirmed. However, monocytes isolated from blood and bone marrow lacked CSF1 expression. This species-specific mAb delivers the opportunity to further understanding of porcine myeloid cell biology. This is not only vital for the role of pigs as a model for human health, but also as a veterinary species of significant economic and agricultural importance.


Subject(s)
Antibodies, Monoclonal , Macrophage Colony-Stimulating Factor , Swine , Mice , Animals , Humans , Macrophages , Monocytes , Mononuclear Phagocyte System/metabolism
4.
Brain Pathol ; 33(2): e13151, 2023 03.
Article in English | MEDLINE | ID: mdl-36755470

ABSTRACT

Ischemic stroke (IS) is a major cause of disability and death in adults, and the immune response plays an indispensable role in its pathological process. After the onset of IS, an inflammatory storm, with the infiltration and mobilization of the mononuclear phagocyte system (MPS), is triggered in the brain. Microglia are rapidly activated in situ, followed by waves of circulating monocytes into the ischemic area. Activated microglia and monocytes/macrophages are mainly distributed in the peri-infarct area. These cells have similar morphology and functions, such as secreting cytokines and phagocytosis. Previously, the presence of the MPS was considered a marker of an exacerbated inflammatory response that contributes to brain damage. However, recent studies have suggested a rather complicated role of the MPS in IS. Here, we reviewed articles focusing on various functions of the MPS among different phases of IS, including recruitment, polarization, phagocytosis, angiogenesis, and interaction with other types of cells. Moreover, due to the characteristics of the MPS, we also noted clinical research addressing alterations in the MPS as potential biomarkers for IS patients for the purposes of predicting prognosis and developing novel therapeutic strategies.


Subject(s)
Ischemic Stroke , Stroke , Humans , Ischemic Stroke/metabolism , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/pathology , Macrophages/metabolism , Microglia/pathology , Monocytes , Stroke/pathology
5.
Amyloid ; 30(2): 225-238, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36495239

ABSTRACT

Amyloidosis refers to a group of degenerative diseases that are characterized by the deposition of misfolded protein fibrils in various organs. Deposited amyloid may be removed by a phagocyte-dependent innate immune system; however, the precise mechanisms during disease progression remain unclear. We herein investigated the properties of macrophages that contribute to amyloid degradation and disease progression using inducible apolipoprotein A-II amyloidosis model mice. Intravenously injected AApoAII amyloid was efficiently engulfed by reticuloendothelial macrophages in the liver and spleen and disappeared by 24 h. While cultured murine macrophages degraded AApoAII via the endosomal-lysosomal pathway, AApoAII fibrils reduced cell viability and phagocytic capacity. Furthermore, the depletion of reticuloendothelial macrophages before the induction of AApoAII markedly increased hepatic and splenic AApoAII deposition. These results highlight the physiological role of reticuloendothelial macrophages in the early stages of pathogenesis and suggest the maintenance of phagocytic integrity as a therapeutic strategy to inhibit disease progression.


Subject(s)
Amyloidosis , Apolipoprotein A-II , Mice , Animals , Apolipoprotein A-II/metabolism , Amyloidosis/metabolism , Amyloid/metabolism , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/pathology , Macrophages/metabolism , Amyloidogenic Proteins , Disease Progression
6.
J Am Chem Soc ; 144(45): 20653-20660, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36326483

ABSTRACT

Liver sequestration, mainly resulting from the phagocytosis of mononuclear phagocyte system (MPS) cells, is a long-standing barrier in nanoparticle delivery, which severely decreases the disease-targeting ability, leads to nanotoxicity, and inhibits clinical translation. To avoid long-term liver sequestration, we elaborately designed luminescent gold-silver bimetallic nanoparticles that could be rapidly transformed by the hepatic sinusoidal microenvironment rich in glutathione and oxygen, significantly different from monometallic gold nanoparticles that were rapidly sequestrated by Kupffer cells due to the much slower biotransformation. We found that the rapid sinusoidal biotransformation induced by the synergistic reactions of glutathione and oxygen with the reactive silver atoms could help bimetallic nanoparticles to avoid MPS phagocytosis, promote fast release from the liver, prolong blood circulation, enhance renal clearance, and increase disease targeting. With the fast biotransformation in sinusoids, liver sequestration could be turned into a beneficial storage mechanism for nanomedicines to maximize targeting.


Subject(s)
Metal Nanoparticles , Nanoparticles , Gold , Silver , Capillaries/metabolism , Mononuclear Phagocyte System/metabolism , Nanoparticles/metabolism , Biotransformation , Glutathione/metabolism , Oxygen
7.
Adv Mater ; 34(16): e2201095, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35218106

ABSTRACT

Lipid nanoparticles (LNPs) are the leading nonviral technologies for the delivery of exogenous RNA to target cells in vivo. As systemic delivery platforms, these technologies are exemplified by Onpattro, an approved LNP-based RNA interference therapy, administered intravenously and targeted to parenchymal liver cells. The discovery of systemically administered LNP technologies capable of preferential RNA delivery beyond hepatocytes has, however, proven more challenging. Here, preceded by comprehensive mechanistic understanding of in vivo nanoparticle biodistribution and bodily clearance, an LNP-based messenger RNA (mRNA) delivery platform is rationally designed to preferentially target the hepatic reticuloendothelial system (RES). Evaluated in embryonic zebrafish, validated in mice, and directly compared to LNP-mRNA systems based on the lipid composition of Onpattro, RES-targeted LNPs significantly enhance mRNA expression both globally within the liver and specifically within hepatic RES cell types. Hepatic RES targeting requires just a single lipid change within the formulation of Onpattro to switch LNP surface charge from neutral to anionic. This technology not only provides new opportunities to treat liver-specific and systemic diseases in which RES cell types play a key role but, more importantly, exemplifies that rational design of advanced RNA therapies must be preceded by a robust understanding of the dominant nano-biointeractions involved.


Subject(s)
Lipids , Nanoparticles , Animals , Liposomes , Liver/metabolism , Mice , Mononuclear Phagocyte System/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Tissue Distribution , Zebrafish
8.
J Virol ; 96(2): e0087621, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34705561

ABSTRACT

Broad tissue tropism of cytomegaloviruses (CMVs) is facilitated by different glycoprotein entry complexes, which are conserved between human CMV (HCMV) and murine CMV (MCMV). Among the wide array of cell types susceptible to the infection, mononuclear phagocytes (MNPs) play a unique role in the pathogenesis of the infection as they contribute both to the virus spread and immune control. CMVs have dedicated numerous genes for the efficient infection and evasion of macrophages and dendritic cells. In this study, we have characterized the properties and function of M116, a previously poorly described but highly transcribed MCMV gene region that encodes M116.1p, a novel protein necessary for the efficient infection of MNPs and viral spread in vivo. Our study further revealed that M116.1p shares similarities with its positional homologs in HCMV and RCMV, UL116 and R116, respectively, such as late kinetics of expression, N-glycosylation, localization to the virion assembly compartment, and interaction with gH-a member of the CMVs fusion complex. This study, therefore, expands our knowledge about virally encoded glycoproteins that play important roles in viral infectivity and tropism. IMPORTANCE Human cytomegalovirus (HCMV) is a species-specific herpesvirus that causes severe disease in immunocompromised individuals and immunologically immature neonates. Murine cytomegalovirus (MCMV) is biologically similar to HCMV, and it serves as a widely used model for studying the infection, pathogenesis, and immune responses to HCMV. In our previous work, we have identified the M116 ORF as one of the most extensively transcribed regions of the MCMV genome without an assigned function. This study shows that the M116 locus codes for a novel protein, M116.1p, which shares similarities with UL116 and R116 in HCMV and RCMV, respectively, and is required for the efficient infection of mononuclear phagocytes and virus spread in vivo. Furthermore, this study establishes the α-M116 monoclonal antibody and MCMV mutants lacking M116, generated in this work, as valuable tools for studying the role of macrophages and dendritic cells in limiting CMV infection following different MCMV administration routes.


Subject(s)
Mononuclear Phagocyte System/virology , Muromegalovirus/physiology , Viral Envelope Proteins/metabolism , Animals , Fibroblasts/metabolism , Fibroblasts/virology , Glycosylation , Herpesviridae Infections/virology , Membrane Glycoproteins/metabolism , Mice , Mononuclear Phagocyte System/metabolism , Transcription, Genetic , Viral Envelope Proteins/genetics , Virion/metabolism , Virus Assembly , Virus Internalization , Virus Replication
9.
Cell Rep ; 37(5): 109916, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731608

ABSTRACT

Intestinal epithelial cells (IECs) have long been understood to express high levels of major histocompatibility complex class II (MHC class II) molecules but are not considered canonical antigen-presenting cells, and the impact of IEC-MHC class II signaling on gut homeostasis remains enigmatic. As IECs serve as the primary barrier between underlying host immune cells, we reasoned that IEC-intrinsic antigen presentation may play a role in responses toward the microbiota. Mice with an IEC-intrinsic deletion of MHC class II (IECΔMHC class II) are healthy but have fewer microbial-bound IgA, regulatory T cells (Tregs), and immune repertoire selection. This was associated with increased interindividual microbiota variation and altered proportions of two taxa in the ileum where MHC class II on IECs is highest. Intestinal mononuclear phagocytes (MNPs) have similar MHC class II transcription but less surface MHC class II and are capable of acquiring MHC class II from IECs. Thus, epithelial-myeloid interactions mediate development of adaptive responses to microbial antigens within the gastrointestinal tract.


Subject(s)
Adaptive Immunity , Bacteria/immunology , Epithelial Cells/immunology , Gastrointestinal Microbiome , Histocompatibility Antigens Class II/immunology , Ileum/microbiology , Immunity, Mucosal , Mononuclear Phagocyte System/immunology , Myeloid Cells/immunology , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacteria/growth & development , Bacteria/metabolism , Cell Line , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Histocompatibility Antigens Class II/metabolism , Host-Pathogen Interactions , Ileum/immunology , Ileum/metabolism , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/microbiology , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
10.
J Adv Res ; 31: 61-74, 2021 07.
Article in English | MEDLINE | ID: mdl-34194832

ABSTRACT

Background: Even though exosome-based therapy has been shown to be able to control the progression of different pathologies, the data revealed by pharmacokinetic studies warn of the low residence time of exogenous exosomes in circulation that can hinder the clinical translation of therapeutic exosomes. The macrophages related to the organs of the mononuclear phagocytic system are responsible primarily for the rapid clearance and retention of exosomes, which strongly limits the amount of exosomal particles available to reach the target tissue, accumulate in it and release with high efficiency its therapeutic cargo in acceptor target cells to exert the desired biological effect. Aim of review: Endowing exosomes with surface modifications to evade the immune system is a plausible strategy to contribute to the suppression of exosomal clearance and increase the efficiency of their targeted content delivery. Here, we summarize the current evidence about the mechanisms underlying the recognition and sequestration of therapeutic exosomes by phagocytic cells. Also, we propose different strategies to generate 'invisible' exosomes for the immune system, through the incorporation of different anti-phagocytic molecules on the exosomes' surface that allow increasing the circulating half-life of therapeutic exosomes with the purpose to increase their bioavailability to reach the target tissue, transfer their therapeutic molecular cargo and improve their efficacy profile. Key scientific concepts of review: Macrophage-mediated phagocytosis are the main responsible behind the short half-life in circulation of systemically injected exosomes, hindering their therapeutic effect. Exosomes 'Camouflage Cloak' strategy using antiphagocytic molecules can contribute to the inhibition of exosomal clearance, hence, increasing the on-target effect. Some candidate molecules that could exert an antiphagocytic role are CD47, CD24, CD44, CD31, ß2M, PD-L1, App1, and DHMEQ. Pre- and post-isolation methods for exosome engineering are compatible with the loading of therapeutic cargo and the expression of antiphagocytic surface molecules.


Subject(s)
Biological Mimicry , Drug Delivery Systems/methods , Exosomes/metabolism , Phagocytosis , B7-H1 Antigen/metabolism , Biological Availability , CD24 Antigen/metabolism , CD47 Antigen/metabolism , Exosomes/immunology , Humans , Hyaluronan Receptors/metabolism , Immune System , Macrophages/immunology , Macrophages/metabolism , Mononuclear Phagocyte System/metabolism , Phagocytes/immunology , Phagocytes/metabolism
11.
Dis Markers ; 2021: 9911184, 2021.
Article in English | MEDLINE | ID: mdl-34113405

ABSTRACT

BACKGROUND: Osteoarthritis (OA) and rheumatoid arthritis (RA) are well-known cause of joint disability. Although they have shown the analogous clinical features involving chronic synovitis that progresses to cartilage and bone destruction, the pathogenesis that initiates and perpetuates synovial lesions between RA and OA remains elusive. OBJECTIVE: This study is aimed at identifying disease-specific hub genes, exploring immune cell infiltration, and elucidating the underlying mechanisms associated with RA and OA synovial lesion. METHODS: Gene expression profiles (GSE55235, GSE55457, GSE55584, and GSE12021) were selected from Gene Expression Omnibus for analysis. Differentially expressed genes (DEGs) were identified by the "LIMMA" package in Bioconductor. The DEGs were identified by Gene Ontology (GO) and KEGG pathway analysis. A protein-protein interaction network was constructed to identify candidate hub genes by using STRING and Cytoscape. Hub genes were identified by validating from GSE12021. Furthermore, we employed the CIBERSORT website to assess immune cell infiltration between OA and RA. Finally, we explored the correlation between the levels of hub genes and relative proportion of immune cells in OA and RA. RESULTS: We identified 68 DEGs which were mainly enriched in immune response and chemokine signaling pathway. Six hub genes with a cutoff of AUC > 0.80 by ROC analysis and relative expression of P < 0.05 were identified successfully. Compared with OA, the RA synovial tissues consisted of a higher proportion of 7 immune cells, whereas 4 immune cells were found in relatively lower proportion (P < 0.05). In addition, the levels of 6 hub genes were closely associated with relative proportion of 11 immune cells in OA and RA. CONCLUSIONS: We used bioinformatics analysis to identify hub genes and explored immune cell infiltration of immune microenvironment in synovial tissues. Our results should offer insights into the underlying molecular mechanisms of synovial lesion and provide potential target for immune-based therapies of OA and RA.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Osteoarthritis/genetics , Osteoarthritis/immunology , Synovial Membrane/immunology , Transcriptome , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/metabolism , Biomarkers/metabolism , Chemokines/metabolism , Computational Biology , Databases, Genetic , Gene Expression Profiling , Genetic Markers , Humans , Leukocytes/immunology , Leukocytes/metabolism , Mononuclear Phagocyte System/immunology , Mononuclear Phagocyte System/metabolism , Osteoarthritis/diagnosis , Osteoarthritis/metabolism , Protein Interaction Maps/genetics , ROC Curve , Synovial Membrane/metabolism
12.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33397719

ABSTRACT

Exaggerated airway hyperresponsiveness and inflammation are hallmarks of asthma, and lipopolysaccharide (LPS) exposure is linked to the severity of the disease and steroid resistance. To investigate the mechanisms underlying asthma exacerbation, we established a mouse model of LPS-induced steroid-resistant exacerbation on the background of house dust mite (HDM)-induced asthma to profile the immune cells in lung by using single-cell RNA deep sequencing. Twenty immune subsets were identified by their molecular and functional properties. Specific cell clusters of basophils, type 2 innate lymphoid cells (ILC2), and CD8+ memory T cells were the predominant sources of interleukin (IL)-4 and IL-13 transcripts whose expressions were dexamethasone resistant. Production of IL-13 by these cells was validated by IL-13-reporter mice. Neutralization of IL-13 abolished HDM/LPS-induced airway hyperresponsiveness, airway inflammation, and decreased mucus hypersecretion. Furthermore, using Ingenuity Pathway Analysis systems, we identified canonical pathways and upstream regulators that regulate the activation of basophils, ILC2, and CD8+ memory T cells. Our study provides mechanistic insights and an important reference resource for further understanding of the immune landscape during asthma exacerbation.


Subject(s)
Asthma/immunology , Interleukin-13/metabolism , Leukocytes/metabolism , Lung/immunology , Mononuclear Phagocyte System/metabolism , Transcriptome , Animals , Disease Progression , Interleukin-4/metabolism , Lipopolysaccharides , Mice, Inbred BALB C , Pyroglyphidae/immunology , Single-Cell Analysis
13.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33479167

ABSTRACT

Dendritic cells (DCs) and monocytes are crucial mediators of innate and adaptive immune responses during viral infection, but misdirected responses by these cells may contribute to immunopathology. Here, we performed high-dimensional flow cytometry-analysis focusing on mononuclear phagocyte (MNP) lineages in SARS-CoV-2-infected patients with moderate and severe COVID-19. We provide a deep and comprehensive map of the MNP landscape in COVID-19. A redistribution of monocyte subsets toward intermediate monocytes and a general decrease in circulating DCs was observed in response to infection. Severe disease coincided with the appearance of monocytic myeloid-derived suppressor cell-like cells and a higher frequency of pre-DC2. Furthermore, phenotypic alterations in MNPs, and their late precursors, were cell-lineage-specific and associated either with the general response against SARS-CoV-2 or COVID-19 severity. This included an interferon-imprint in DC1s observed in all patients and a decreased expression of the coinhibitory molecule CD200R in pre-DCs, DC2s, and DC3 subsets of severely sick patients. Finally, unsupervised analysis revealed that the MNP profile, alone, pointed to a cluster of COVID-19 nonsurvivors. This study provides a reference for the MNP response to SARS-CoV-2 infection and unravels mononuclear phagocyte dysregulations associated with severe COVID-19.


Subject(s)
COVID-19/immunology , Mononuclear Phagocyte System/immunology , SARS-CoV-2/immunology , Adult , COVID-19/epidemiology , COVID-19/metabolism , COVID-19/virology , Cytokines/immunology , Dendritic Cells/immunology , Female , Humans , Interferons/immunology , Male , Middle Aged , Monocytes/immunology , Mononuclear Phagocyte System/metabolism , Severity of Illness Index , Sweden
15.
J Med Chem ; 63(24): 15621-15638, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33296601

ABSTRACT

Since inception, the magic bullets developed against leishmaniasis traveled a certain path and then dropped down due to either toxicity or the emergence of resistance. The route of administration is also an important concern. We developed a series of water-soluble ferrocenylquinoline derivatives, targeting Leishmania donovani, among which CQFC1 showed the highest efficacy even in comparison to other drugs, in use or used, both in oral and intramuscular routes. It did not induce any toxicity to splenocytes and on hematopoiesis, induced protective cytokines, and did not hamper the drug-metabolizing enzymes in hosts. It acts through the reduction and the inhibition of parasites' survival enzyme trypanothione reductase of replicating amastigotes in hosts' reticuloendothelial tissues. Unlike conventional drugs, this molecule did not induce the resistance-conferring genes in laboratory-maintained resistant L. donovani lines. Experimentally, this easily bioavailable preclinical drug candidate overcame all of the limitations causing the discontinuation of the other conventional antileishmanial drugs.


Subject(s)
Antiprotozoal Agents/chemistry , Leishmania donovani/enzymology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Quinolines/chemistry , Administration, Oral , Animals , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Binding Sites , Disease Models, Animal , Drug Design , Drug Resistance/drug effects , Ferrous Compounds/chemistry , Half-Life , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Metallocenes/chemistry , Mice , Molecular Docking Simulation , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/parasitology , NADH, NADPH Oxidoreductases/metabolism , Protozoan Proteins/metabolism , Quinolines/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , Reactive Oxygen Species/metabolism , Solubility , Structure-Activity Relationship
16.
J Immunol ; 205(11): 3154-3166, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33139489

ABSTRACT

The proliferation, differentiation, and survival of cells of the mononuclear phagocyte system (MPS; progenitors, monocytes, macrophages, and classical dendritic cells) are controlled by signals from the M-CSF receptor (CSF1R). Cells of the MPS lineage have been identified using numerous surface markers and transgenic reporters, but none is both universal and lineage restricted. In this article, we report the development and characterization of a CSF1R reporter mouse. A FusionRed (FRed) cassette was inserted in-frame with the C terminus of CSF1R, separated by a T2A-cleavable linker. The insertion had no effect of CSF1R expression or function. CSF1R-FRed was expressed in monocytes and macrophages and absent from granulocytes and lymphocytes. In bone marrow, CSF1R-FRed was absent in lineage-negative hematopoietic stem cells, arguing against a direct role for CSF1R in myeloid lineage commitment. It was highly expressed in marrow monocytes and common myeloid progenitors but significantly lower in granulocyte-macrophage progenitors. In sections of bone marrow, CSF1R-FRed was also detected in osteoclasts, CD169+ resident macrophages, and, consistent with previous mRNA analysis, in megakaryocytes. In lymphoid tissues, CSF1R-FRed highlighted diverse MPS populations, including classical dendritic cells. Whole mount imaging of nonlymphoid tissues in mice with combined CSF1R-FRed/Csf1r-EGFP confirmed the restriction of CSF1R expression to MPS cells. The two markers highlight the remarkable abundance and regular distribution of tissue MPS cells, including novel macrophage populations within tendon and skeletal muscle and underlying the mesothelial/serosal/capsular surfaces of every major organ. The CSF1R-FRed mouse provides a novel reporter with exquisite specificity for cells of the MPS.


Subject(s)
Biomarkers/metabolism , Mononuclear Phagocyte System/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Cell Differentiation/physiology , Dendritic Cells/metabolism , Hematopoietic Stem Cells/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Tendons/metabolism
17.
J Cardiovasc Pharmacol ; 76(4): 407-413, 2020 10.
Article in English | MEDLINE | ID: mdl-33027195

ABSTRACT

Myocardial infarction (MI) is an irreversible damage of the heart muscle, which often leads to adverse cardiac remodeling and progressive heart failure. After MI, immune cells play a vital role in the clearance of the dying tissue and cardiac remodeling. Post-MI events include the release of danger signals by necrotic cardiomyocytes and the migration of the inflammatory cells, such as dendritic cells, neutrophils, monocytes, and macrophages, into the site of the cardiac injury to digest the cell debris and secrete a variety of inflammatory factors activating the inflammatory response. In this review, we focus on the role of immune cells in the cardiac remodeling after MI and the novel immunotherapies targeting immune cells.


Subject(s)
Leukocytes/immunology , Mononuclear Phagocyte System/immunology , Myocardial Infarction/immunology , Ventricular Remodeling , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunotherapy , Leukocytes/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Mononuclear Phagocyte System/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Neutrophils/immunology , Neutrophils/metabolism , Signal Transduction
18.
Int J Nanomedicine ; 15: 5613-5627, 2020.
Article in English | MEDLINE | ID: mdl-32884257

ABSTRACT

BACKGROUND: Stimuli-responsive gold nano-assemblies have attracted attention as drug delivery systems in the biomedical field. However, there are challenges achieving targeted delivery and controllable drug release for specific diseases. MATERIALS AND METHODS: In this study, a glutathione (GSH)-modified fluorescent gold nanoparticle termed AuLA-GSH was prepared and a Co2+-induced self-assembly drug delivery platform termed AuLA-GSH-Co was constructed. Both the pH-responsive character and drug loading behavior of AuLA-GSH-Co were studied in vitro. Kidney-targeting capability was investigated in vitro and in vivo. Finally, the anti-fibrosis efficiency of AuLA-GSH-Co in a mouse model of unilateral ureteral obstruction (UUO) was explored. RESULTS: AuLA-GSH-Co was sensitive to pH changes and released Co2+ in acidic conditions, allowing it to have controllable drug release abilities. AuLA-GSH-Co was found to improve cellular uptake of Co2+ ions compared to CoCl2 in vitro. AuLA-GSH exhibited specific renal targeting and prolonged renal retention time with low non-specific accumulation in vivo. Moreover, the anti-fibrosis efficiency of AuLA-GSH-Co was higher compared to CoCl2 in a mouse model of unilateral ureteral obstruction (UUO). CONCLUSION: AuLA-GSH-Co could greatly enhance drug delivery efficiency with renal targeting capability and obviously relieve renal fibrosis, providing a promising strategy for renal fibrosis therapy.


Subject(s)
Cobalt/pharmacokinetics , Drug Delivery Systems/methods , Kidney Diseases/drug therapy , Kidney/pathology , Metal Nanoparticles/chemistry , Animals , Cell Line , Cobalt/chemistry , Disease Models, Animal , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Liberation , Fibrosis , Fluorescence , Glutathione/chemistry , Gold/chemistry , Hydrogen-Ion Concentration , Kidney/drug effects , Kidney/metabolism , Kidney Diseases/pathology , Metal Nanoparticles/administration & dosage , Mice, Inbred BALB C , Mononuclear Phagocyte System/drug effects , Mononuclear Phagocyte System/metabolism , Rats , Ureteral Obstruction
19.
Int J Mol Sci ; 21(16)2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32764386

ABSTRACT

Investigation of the potential for nanomaterials to generate immunogenic effects is a key aspect of a robust preclinical evaluation. In combination with physicochemical characterization, such assessments also provide context for how material attributes influence biological outcomes. Furthermore, appropriate models for these assessments allow accurate in vitro to in vivo extrapolation, which is vital for the mechanistic understanding of nanomaterial action. Here we have assessed the immunogenic impact of a small panel of commercially available and in-house prepared nanomaterials on primary human peripheral blood mononuclear cells (PBMCs). A diethylaminoethyl-dextran (DEAE-dex) functionalized superparamagnetic iron oxide nanoparticle (SPION) generated detectable quantities of tumor necrosis factor α (TNFα), interleukin-1ß (IL-1ß), and IL-10, the only tested material to do so. The human leukemia monocytic cell line THP-1 was used to assess the potential for the nanomaterial panel to affect cellular oxidation-reduction (REDOX) via measurement of reactive oxygen species and reduced glutathione. Negatively charged sulfonate-functionalized polystyrene nanoparticles demonstrated a size-related trend for the inhibition of caspase-1, which was not observed for amine-functionalized polystyrene of similar sizes. Silica nanoparticles (310 nm) resulted in a 93% increase in proliferation compared to the untreated control (p < 0.01). No other nanomaterial treatments resulted in significant change from that of unstimulated PBMCs. Responses to the nanomaterials in the assays described demonstrate the utility of primary cells as ex vivo models for nanomaterial biological impact.


Subject(s)
Leukocytes, Mononuclear/drug effects , Metal Nanoparticles/chemistry , Mononuclear Phagocyte System/drug effects , Nanostructures/chemistry , Caspase 1/genetics , Cell Survival/drug effects , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Gene Expression Regulation/drug effects , Humans , Interleukin-10/genetics , Interleukin-1alpha/genetics , Leukocytes, Mononuclear/metabolism , Mononuclear Phagocyte System/metabolism , Oxidation-Reduction/drug effects , Polystyrenes/chemistry , Polystyrenes/pharmacology , Reactive Oxygen Species/metabolism , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Tumor Necrosis Factor-alpha/genetics
20.
Immunol Rev ; 295(1): 54-67, 2020 05.
Article in English | MEDLINE | ID: mdl-32242952

ABSTRACT

We have only recently started to appreciate the extent to which immune cell activation involves significant changes in cellular metabolism. We are now beginning to understand how commitment to specific metabolic pathways influences aspects of cellular biology that are the more usual focus of immunological studies, such as activation-induced changes in gene transcription, post-transcriptional regulation of transcription, post-translational modifications of proteins, cytokine secretion, etc. Here, we focus on metabolic reprogramming in mononuclear phagocytes downstream of stimulation with inflammatory signals (such as LPS and IFNγ) vs alternative activation signals (IL-4), with an emphasis on work on dendritic cells and macrophages from our laboratory, and related studies from others. We cover aspects of glycolysis and its branching pathways (glycogen synthesis, pentose phosphate, serine synthesis, hexose synthesis, and glycerol 3 phosphate shuttle), the tricarboxylic acid pathway, fatty acid synthesis and oxidation, and mitochondrial biology. Although our understanding of the metabolism of mononuclear phagocytes has progressed significantly over the last 10 years, major challenges remain, including understanding the effects of tissue residence on metabolic programming related to cellular activation, and the translatability of findings from mouse to human biology.


Subject(s)
Mononuclear Phagocyte System/immunology , Mononuclear Phagocyte System/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Animals , Energy Metabolism , Humans , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mononuclear Phagocyte System/cytology , Phagocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...