Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Biol Chem ; 299(5): 104627, 2023 05.
Article in English | MEDLINE | ID: mdl-36944399

ABSTRACT

The FimH type-1 fimbrial adhesin allows pathogenic Escherichia coli to adhere to glycoproteins in the epithelial linings of human bladder and intestinal tract, by using multiple fimbriae simultaneously. Pauci- and high-mannose type N-glycans are natural FimH receptors on those glycoproteins. Oligomannose-3 and oligomannose-5 bind with the highest affinity to FimH by using the same Manα1,3Man branch. Oligomannose-6 is generated from oligomannose-5 in the next step of the biogenesis of high-mannose N-glycans, by the transfer of a mannose in α1,2-linkage onto this branch. Using serial crystallography and by measuring the kinetics of binding, we demonstrate that shielding the high-affinity epitope drives the binding of multiple FimH molecules. First, we profiled FimH glycan binding on a microarray containing paucimannosidic N-glycans and in a FimH LEctPROFILE assay. To make the transition to oligomannose-6, we measured the kinetics of FimH binding using paucimannosidic N-glycans, glycoproteins and all four α-dimannosides conjugated to bovine serum albumin. Equimolar mixed interfaces of the dimannosides present in oligomannose-6 and molecular dynamics simulations suggest a positive cooperativity in the bivalent binding of Manα1,3Manα1 and Manα1,6Manα1 dimannosides. The binding of core α1,6-fucosylated oligomannose-3 in cocrystals of FimH is monovalent but interestingly the GlcNAc1-Fuc moiety retains highly flexibility. In cocrystals with oligomannose-6, two FimH bacterial adhesins bind the Manα1,3Manα1 and Manα1,6Manα1 endings of the second trimannose core (A-4'-B). This cooperative switch towards bivalent binding appears sustainable beyond a molar excess of oligomannose-6. Our findings provide important novel structural insights for the design of multivalent FimH antagonists that bind with positive cooperativity.


Subject(s)
Adhesins, Escherichia coli , Mannose Receptor , Models, Molecular , Humans , Adhesins, Escherichia coli/chemistry , Adhesins, Escherichia coli/metabolism , Bacterial Adhesion , Escherichia coli/metabolism , Glycoproteins/metabolism , Mannose/metabolism , Mannose Receptor/chemistry , Mannose Receptor/metabolism , Polysaccharides/metabolism , Protein Binding , Protein Structure, Quaternary , Molecular Docking Simulation
2.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34326259

ABSTRACT

Proinflammatory activation of macrophages in metabolic tissues is critically important in the induction of obesity-induced metaflammation. Here, we demonstrate that the soluble mannose receptor (sMR) plays a direct functional role in both macrophage activation and metaflammation. We show that sMR binds CD45 on macrophages and inhibits its phosphatase activity, leading to an Src/Akt/NF-κB-mediated cellular reprogramming toward an inflammatory phenotype both in vitro and in vivo. Remarkably, increased serum sMR levels were observed in obese mice and humans and directly correlated with body weight. Importantly, enhanced sMR levels increase serum proinflammatory cytokines, activate tissue macrophages, and promote insulin resistance. Altogether, our results reveal sMR as regulator of proinflammatory macrophage activation, which could constitute a therapeutic target for metaflammation and other hyperinflammatory diseases.


Subject(s)
Gene Expression Regulation/drug effects , Macrophage Activation/drug effects , Macrophages/metabolism , Mannose Receptor/chemistry , Membrane Proteins/pharmacology , Animal Feed , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat , Gastrointestinal Microbiome , Inflammation , Macrophage Activation/physiology , Male , Mannose Receptor/metabolism , Mice , Mice, Knockout , Random Allocation
3.
Mol Pharm ; 20(5): 2415-2425, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37014648

ABSTRACT

Tumor-associated macrophages (TAMs) are large phagocytic cells that play numerous roles in cancer biology and are an important component of the relationship between immune system response and tumor progression. The peptide, RP832c, targets the Mannose Receptor (CD206) expressed on M2-like macrophages and is cross-reactive to both human and murine CD206. Additionally, it exhibits therapeutic properties through its ability to shift the population of TAMs from an M2-like (protumor) toward an M1-like phenotype (antitumor) and has demonstrated promise in inhibiting tumor resistance in PD-L1 unresponsive melanoma murine models. In addition, it has shown inhibition in bleomycin-induced pulmonary fibrosis through interactions with CD206 macrophages.1,2 Our work aims to develop a novel CD206 positron emission tomography (PET) imaging probe based on RP832c (Kd = 5.64 µM) as a direct, noninvasive method for the assessment of TAMs in mouse models of cancer. We adapted RP832c to incorporate the chelator DOTA to allow for radiolabeling with the PET isotope 68Ga (t1/2 = 68 min; ß+ = 89%). In vitro stability studies were conducted in mouse serum up to 3 h. The in vitro binding characteristics of [68Ga]RP832c to CD206 were determined by a protein plate binding assay and Surface Plasmon Resonance (SPR). PET imaging and biodistribution studies were conducted in syngeneic tumor models. Stability studies in mouse serum demonstrated that 68Ga remained complexed up to 3 h (less than 1% free 68Ga). Binding affinity studies demonstrated high binding of [68Ga]RP832c to mouse CD206 protein and that the binding of the tracer was able to be blocked significantly when incubated with a blocking solution of native RP832c. PET imaging and biodistribution studies in syngeneic tumor models demonstrated uptake in tumor and CD206 expressing organs of [68Ga]RP832c. A significant correlation was found between the percentage of CD206 present in each tumor imaged with [68Ga]RP832c and PET imaging mean standardized uptake values in a CT26 mouse model of cancer. The data shows that [68Ga]RP832c represents a promising candidate for macrophage imaging in cancer and other diseases.


Subject(s)
Gallium Radioisotopes , Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Gallium Radioisotopes/chemistry , Macrophages/metabolism , Neoplasms/metabolism , Peptides/metabolism , Positron-Emission Tomography/methods , Tissue Distribution , Mannose Receptor/metabolism
4.
Biochem Biophys Res Commun ; 581: 53-59, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34655976

ABSTRACT

Selective laser melting (SLM) titanium (Ti) implants have shown good prospects for personalized clinical application, but further research is necessary to develop stabilized long-term properties. Since surface modification has been proven bioactive for osseointegration, conventional Ti surface treatment technologies, including sandblasting/acid-etching (SLA) and sandblasting/alkali-heating (SAH), were applied to construct micro and micro/nano surfaces. The SAH group with netlike nano-structure topography exhibited appropriate surface roughness and high hydrophilicity, and as expected, the osseointegration capacities in vivo of the three groups were in order of SAH > SLA > SLM. Besides, both in vivo and in vitro studies revealed that the SLA- and SAH-treated SLM Ti implants significantly inhibited osteoclast activity of peri-implants. Considering the close associations between osteoclasts and macrophages, the effects of Ti surface topography on macrophage polarization were detected. The results showed that the SLA- and SAH-treated SLM Ti implants, especially the latter, had the capacity to promote macrophage polarization to the M2 phenotype. Moreover, the cell culture supernatants of M2 macrophages and RAW264.7 cells seeded on SLA- and SAH-treated SLM Ti surfaces had an adverse effect on osteoclastogenesis. Collectively, this study demonstrated that micro/nano topographies of SLM Ti implants were effective for osseointegration promotion, and their inhibition of osteoclastogenesis might be attributed to macrophage polarization. Our findings shed some light on clinical application of SLM Ti implants and also prove a specific association between macrophage polarization and osteoclastogenesis.


Subject(s)
Cell Differentiation/drug effects , Dental Implants , Nanostructures/ultrastructure , Osseointegration/drug effects , Osteogenesis/drug effects , Titanium/pharmacology , Animals , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Biomarkers/metabolism , Cathepsin K/genetics , Cathepsin K/metabolism , Femur/diagnostic imaging , Femur/surgery , Gene Expression , Hydrophobic and Hydrophilic Interactions , Interleukin-10/genetics , Interleukin-10/metabolism , Lasers , Macrophage Activation/drug effects , Male , Mannose Receptor/genetics , Mannose Receptor/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Nanostructures/chemistry , Osseointegration/physiology , RAW 264.7 Cells , Rats, Sprague-Dawley , Surface Properties , Tartrate-Resistant Acid Phosphatase/genetics , Tartrate-Resistant Acid Phosphatase/metabolism , Titanium/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Clin Exp Allergy ; 51(2): 305-317, 2021 02.
Article in English | MEDLINE | ID: mdl-33301598

ABSTRACT

BACKGROUND: Monocytes and macrophages are critical innate immune cells of the airways. Despite their differing functions, few clinical studies discriminate between them and little is known about their regulation in asthma. OBJECTIVE: We aimed to distinguish and quantify macrophages, monocytes and monocyte subsets in induced sputum and blood and examine their relationship with inflammatory and clinical features of asthma. METHODS: We applied flow cytometry to distinguish macrophages, monocytes and subsets in sputum and blood (n = 53; 45 asthma, 8 non-asthma) and a second asthma sputum cohort (n = 26). Monocyte subsets were identified by surface CD14/CD16 (CD14++ CD16- classical, CD14+ CD16+ intermediate and CD14+ CD16++ non-classical monocytes). Surface CD206, a marker of monocyte tissue differentiation, was measured in sputum. Relationship to airway inflammatory phenotype (neutrophilic n = 9, eosinophilic n = 14, paucigranulocytic n = 22) and asthma severity (severe n = 12, non-severe n = 33) was assessed. RESULTS: Flow cytometry- and microscope-quantified sputum differential cell proportions were significantly correlated. Sputum macrophage number was reduced (p = .036), while classical monocyte proportion was increased in asthma vs non-asthma (p = .032). Sputum classical monocyte number was significantly higher in neutrophilic vs paucigranulocytic asthma (p = .013). CD206- monocyte proportion and number were increased in neutrophilic vs eosinophilic asthma (p < .001, p = .013). Increased sputum classical and CD206- monocyte numbers in neutrophilic asthma were confirmed in the second cohort. Blood monocytes did not vary with airway inflammatory phenotype, but blood classical monocyte proportion and number were increased in severe vs non-severe asthma (p = .022, p = .011). CONCLUSION AND CLINICAL RELEVANCE: Flow cytometry allowed distinction of sputum macrophages, monocytes and subsets, revealing compartment-specific dysregulation of monocytes in asthma. We observed an increase in classical and CD206- monocytes in sputum in neutrophilic asthma, suggesting co-recruitment of monocytes and neutrophils to the airways in asthma. Our data suggest further investigation of how airway monocyte dysregulation impacts on asthma-related disease activity is merited.


Subject(s)
Asthma/immunology , Inflammation/immunology , Macrophages, Alveolar/immunology , Monocytes/immunology , Neutrophils/immunology , Adult , Aged , Asthma/blood , Case-Control Studies , Eosinophils/immunology , Female , Flow Cytometry , Humans , Inflammation/blood , Lipopolysaccharide Receptors/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Male , Mannose Receptor/metabolism , Middle Aged , Monocytes/cytology , Monocytes/metabolism , Phenotype , Receptors, IgG/metabolism , Severity of Illness Index , Sputum/cytology
6.
Cytokine ; 143: 155518, 2021 07.
Article in English | MEDLINE | ID: mdl-33840588

ABSTRACT

IL-17A and IL-25 (IL-17 cytokines family) play an important role in the development of asthma, and allergy. Both cytokines act by binding to heterodimeric receptors with IL17RA as a common subunit. This receptor is found on macrophages, and some other cell types. The aim of the study was to determine the expression of IL17RA on asthmatic and control macrophages from induced sputum (IS) with the regard to IL-17/IL-25 background and relation to clinical features of the disease. We found an elevated expression of IL17RA on sputum macrophages in asthma patients vs controls. A characteristic sputum profile of atopic asthmatic was as follows: high CD206 + IL17RA + macrophage percentage, elevated IL-25 level and low CD206 + IL17RA- macrophage percentage. Based on the above results, it seems that CD206 + sputum macrophages are the effector cells that express common subunit of the receptor for IL-17A and IL-25 in asthma. This may be related to the Th2-dependent environment in asthma and increased concentrations of IL-25 and IL-13 as well as eosinophils in the airways. To our knowledge, our study provides the first data on a possible link between immunological reaction orchestrating CD206 + expressing sputum macrophages and IL-25 via IL17RA pathway in the asthmatic airways.


Subject(s)
Asthma/metabolism , Asthma/pathology , Macrophages/metabolism , Receptors, Interleukin-17/metabolism , Sputum/metabolism , Adult , Case-Control Studies , Female , Humans , Male , Mannose Receptor/metabolism , Middle Aged , Sputum/cytology
7.
Cell Rep ; 42(12): 113266, 2023 12 26.
Article in English | MEDLINE | ID: mdl-37979172

ABSTRACT

Chromosome instability (CIN) contributes to resistance to therapies and tumor evolution. Although natural killer (NK) cells can eliminate cells with complex karyotypes, high-CIN human tumors have an immunosuppressive phenotype. To understand which CIN-associated molecular features alter immune recognition during tumor evolution, we overexpress Polo-like kinase 1 (Plk1) in a Her2+ breast cancer model. These high-CIN tumors activate a senescence-associated secretory phenotype (SASP), upregulate PD-L1 and CD206, and induce non-cell-autonomous nuclear factor κB (NF-κß) signaling, facilitating immune evasion. Single-cell RNA sequencing from pre-neoplastic mammary glands unveiled the presence of Arg1+ macrophages, NK cells with reduced effector functions, and increased resting regulatory T cell infiltration. We further show that high PLK1-expressing human breast tumors display gene expression patterns associated with SASP, NF-κß signaling, and immune suppression. These findings underscore the need to understand the immune landscape in CIN tumors to identify more effective therapies, potentially combining immune checkpoint or NF-κß inhibitors with current treatments.


Subject(s)
Breast Neoplasms , Chromosomal Instability , Immune Tolerance , Polo-Like Kinase 1 , Tumor Escape , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Humans , Animals , Mice , Polo-Like Kinase 1/genetics , Polo-Like Kinase 1/metabolism , Cell Line, Tumor , Receptor, ErbB-2/genetics , NF-kappa B/metabolism , B7-H1 Antigen/metabolism , Mannose Receptor/metabolism , Killer Cells, Natural/immunology , Heterografts , MCF-7 Cells , Female
8.
Dev Comp Immunol ; 126: 104257, 2022 01.
Article in English | MEDLINE | ID: mdl-34530040

ABSTRACT

Mannose receptor (MR) as a member of the pattern recognition receptors (PRRs) plays an important role in the immune response. In mammals, the role of MR in the regulation of phagocytosis is clarified; however, its contribution to opsonize phagocytosis remains unclear in bony fish. In this study, the expression pattern of Nile tilapia mannose receptor gene (OnMR) was investigated and its regulation of the phagocytosis of monocytes/macrophages to pathogenic bacteria was identified. The full-length of OnMR open reading frame is 4314 bp, encoding a peptide containing 1437 amino acid residues. The deduced amino acid sequence revealed that OnMR contained a cysteine-rich domain, a fibronectin type II domain, multiple C-type lectin-like domains, a transmembrane domain and a short cytoplasmic domain. Tissue distribution analysis showed the OnMR transcripts was widely distribute in the ten detected tissues, and highly expressed in head kidney, hind kidney, intestine and spleen. After S. agalactiae and A. hydrophila infection, the expression of OnMR in head kidney and spleen increased significantly. Moreover, the expression of OnMR in MO/Mø were also upregulated post the infection of bacteria and mannose solutions in vitro. This suggested that MR, as a mannose receptor on macrophage surface, could respond strongly to the stimulation of pathogenic bacteria. In addition, the (r)OnMR protein could effectively bind and agglutinate S. agalactiae and A. hydrophila, and regulate the phagocytic ability of monocytes/macrophages to pathogenic bacteria. These results suggest that OnMR is involved in response against bacterial infection in Nile tilapia, and this study will help us better understand the function of MR in teleost fish.


Subject(s)
Cichlids , Fish Diseases , Fish Proteins , Mannose Receptor , Streptococcal Infections , Animals , Fish Diseases/microbiology , Fish Proteins/metabolism , Gene Expression Regulation , Mannose Receptor/metabolism , Streptococcal Infections/veterinary , Streptococcus agalactiae
9.
Toxicol Lett ; 355: 116-126, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34863858

ABSTRACT

Microglia is the innate immune cell in central nervous system (CNS) and plays an important role in neuroinflammation. Microglia mediated neuroinflammation is the key factor affecting the development of neurodegenerative diseases. Although there was evidence that paraquat (PQ) could induce inflammatory response, its mechanism was not clear. The present study investigated the mechanisms of PQ-induced inflammatory responses in BV-2 microglia cells, and tried to reveal the role of ROS/Akt1 pathway. The results showed that the cell activation markers (iNOS and CD206) of BV-2 cells were increased after PQ treatment, suggesting that BV-2 microglia were activated. PQ induced the reactive oxygen species (ROS) and inhibited the AKT1 phosphorylation in BV-2 cells. Besides, the M1 markers expression (IL-6, TNF-α and IL-1ß) were significantly increased after PQ treatment, which suggested that PQ induced the increase of M1 phenotype of BV-2 microglia. Pre-treated with NAC (ROS scavenger), the M1 phenotype was decreased while the p-Akt1 was restored compared to PQ stimulation. Furthermore, we built an Akt1(S473E)-overexpression BV-2 cell line. The Akt1 (S473E) partially attenuated the PQ induced increase in M1 phenotype, while ROS did not significantly change. These results indicated that PQ induced BV-2 microglia activation by increased ROS mediated Akt1 activation inhibition, leading to neuroinflammation.


Subject(s)
Herbicides/toxicity , Microglia/drug effects , Paraquat/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Animals , Biomarkers/metabolism , Cell Line , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Mannose Receptor/genetics , Mannose Receptor/metabolism , Mice , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Reactive Oxygen Species
10.
Drug Discov Today ; 26(9): 2124-2137, 2021 09.
Article in English | MEDLINE | ID: mdl-33667654

ABSTRACT

Glycoconjugates are involved in several pathological processes. Glycomimetics that can favorably emulate complex carbohydrate structures, while competing with natural ligands as inhibitors, are gaining considerable attention owing to their improved hydrolytic stability, binding affinity, and pharmacokinetic (PK) properties. Of particular interest are the families of α-d-mannopyranoside analogs, which can be used as inhibitors against adherent invasive Escherichia coli infections. Bacterial resistance to modern antibiotics triggers the search for new alternative antibacterial strategies that are less susceptible to acquiring resistance. In this review, we highlight recent progress in the chemical syntheses of this family of compounds, one of which having reached clinical trials against Crohn's disease (CD).


Subject(s)
Anti-Bacterial Agents , Escherichia coli Infections/drug therapy , Glycoproteins , Nanoparticles , Adhesins, Escherichia coli , Bacterial Adhesion , Biomimetics , Drug Design , Fimbriae Proteins/antagonists & inhibitors , Fimbriae, Bacterial , Humans , Mannose/metabolism , Mannose Receptor/metabolism
11.
Toxins (Basel) ; 13(10)2021 10 10.
Article in English | MEDLINE | ID: mdl-34679010

ABSTRACT

Bee venom phospholipase A2 (bvPLA2) has been reported to have therapeutic effects such as neuroprotection, anti-inflammation, anti-nociception, anti-cancer properties, caused by increasing regulatory T cells (Tregs). The mechanism of Tregs modulation by bvPLA2 has been demonstrated by binding with the mannose receptor, CD206 in experimental models of several diseases. However, it remains unknown whether this mechanism can also be applied in human blood. In this study, we collected peripheral blood samples from healthy donors and analyzed the percentages of monocyte-derived dendritic cells with CD206 (CD206+ DCs) before expansion, the proportion of Tregs, and the subpopulations after expansion treated with bvPLA2 or PBS using flow cytometry and the correlations among them. The percentage of Tregs tended to be higher in the bvPLA2 group than in the control group. There were significant positive correlations between the CD206 population in hPBMC and the proportions of Tregs treated with bvPLA2, especially in the Treg fold change comparing the increase ratio of Tregs in bvPLA2 and in PBS. These findings indicate that bvPLA2 increased the proportion of Tregs in healthy human peripheral blood and the number of CD206+ DCs could be a predictor of the bvPLA2 response of different individuals.


Subject(s)
Bee Venoms/enzymology , Phospholipases A2/pharmacology , T-Lymphocytes, Regulatory/drug effects , Adult , Aged , Bee Venoms/pharmacology , Female , Humans , Leukocytes, Mononuclear/drug effects , Male , Mannose Receptor/metabolism , Middle Aged , Receptors, Cell Surface/metabolism
12.
Theranostics ; 11(18): 8874-8893, 2021.
Article in English | MEDLINE | ID: mdl-34522216

ABSTRACT

Rationale: Inflammation plays a pivotal role in the pathogenesis of the acute coronary syndrome. Detecting plaques with high inflammatory activity and specifically treating those lesions can be crucial to prevent life-threatening cardiovascular events. Methods: Here, we developed a macrophage mannose receptor (MMR)-targeted theranostic nanodrug (mannose-polyethylene glycol-glycol chitosan-deoxycholic acid-cyanine 7-lobeglitazone; MMR-Lobe-Cy) designed to identify inflammatory activity as well as to deliver peroxisome proliferator-activated gamma (PPARγ) agonist, lobeglitazone, specifically to high-risk plaques based on the high mannose receptor specificity. The MMR-Lobe-Cy was intravenously injected into balloon-injured atheromatous rabbits and serial in vivo optical coherence tomography (OCT)-near-infrared fluorescence (NIRF) structural-molecular imaging was performed. Results: One week after MMR-Lobe-Cy administration, the inflammatory NIRF signals in the plaques notably decreased compared to the baseline whereas the signals in saline controls even increased over time. In accordance with in vivo imaging findings, ex vivo NIRF signals on fluorescence reflectance imaging (FRI) and plaque inflammation by immunostainings significantly decreased compared to oral lobeglitazone group or saline controls. The anti-inflammatory effect of MMR-Lobe-Cy was mediated by inhibition of TLR4/NF-κB pathway. Furthermore, acute resolution of inflammation altered the inflamed plaque into a stable phenotype with less macrophages and collagen-rich matrix. Conclusion: Macrophage targeted PPARγ activator labeled with NIRF rapidly stabilized the inflamed plaques in coronary sized artery, which could be quantitatively assessed using intravascular OCT-NIRF imaging. This novel theranostic approach provides a promising theranostic strategy for high-risk coronary plaques.


Subject(s)
Macrophages/physiology , Plaque, Atherosclerotic/diagnosis , Precision Medicine/methods , Acute Coronary Syndrome/diagnosis , Animals , Arteries/metabolism , Atherosclerosis/metabolism , Drug Delivery Systems/methods , Fluorescence , Indocyanine Green/administration & dosage , Inflammation/diagnosis , Macrophages/metabolism , Male , Mannose Receptor/metabolism , Models, Animal , Molecular Imaging/methods , Optical Imaging/methods , PPAR gamma/agonists , PPAR gamma/metabolism , Plaque, Atherosclerotic/pathology , Pyrimidines/therapeutic use , Rabbits , Spectroscopy, Near-Infrared/methods , Thiazolidinediones/therapeutic use , Tomography, Optical Coherence/methods
13.
Adv Mater ; 33(33): e2101228, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34240485

ABSTRACT

Supplement-free induction of cellular differentiation and polarization solely through the topography of materials is an auspicious strategy but has so far significantly lagged behind the efficiency and intensity of media-supplementation-based protocols. Consistent with the idea that 3D structural motifs in the extracellular matrix possess immunomodulatory capacity as part of the natural healing process, it is found in this study that human-monocyte-derived macrophages show a strong M2a-like prohealing polarization when cultured on type I rat-tail collagen fibers but not on collagen I films. Therefore, it is hypothesized that highly aligned nanofibrils also of synthetic polymers, if packed into larger bundles in 3D topographical biomimetic similarity to native collagen I, would induce a localized macrophage polarization. For the automated fabrication of such bundles in a 3D printing manner, the strategy of "melt electrofibrillation" is pioneered by the integration of flow-directed polymer phase separation into melt electrowriting and subsequent selective dissolution of the matrix polymer postprocessing. This process yields nanofiber bundles with a remarkable structural similarity to native collagen I fibers, particularly for medical-grade poly(ε-caprolactone). These biomimetic fibrillar structures indeed induce a pronounced elongation of human-monocyte-derived macrophages and unprecedentedly trigger their M2-like polarization similar in efficacy as interleukin-4 treatment.


Subject(s)
Biomimetic Materials/chemistry , Collagen/chemistry , Cytokines/chemistry , Immunomodulating Agents/chemistry , Polyesters/chemistry , Tissue Scaffolds/chemistry , Animals , Biomimetic Materials/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Line , Collagen/metabolism , Cytokines/genetics , Cytokines/metabolism , Extracellular Matrix/drug effects , Humans , Immunomodulating Agents/metabolism , Immunomodulation , Macrophages/cytology , Mannose Receptor/genetics , Mannose Receptor/metabolism , Nanofibers/chemistry , Polyvinyls/chemistry , Printing, Three-Dimensional , Rats , Tissue Engineering
14.
Front Immunol ; 12: 795554, 2021.
Article in English | MEDLINE | ID: mdl-34975901

ABSTRACT

Increasing evidence suggests that in hosts infected with parasites of the Leishmania donovani complex, transmission of infection to the sand fly vector is linked to parasite repositories in the host skin. However, a detailed understanding of the dispersal (the mechanism of spread) and dispersion (the observed state of spread) of these obligatory-intracellular parasites and their host phagocytes in the skin is lacking. Using endogenously fluorescent parasites as a proxy, we apply image analysis combined with spatial point pattern models borrowed from ecology to characterize dispersion of parasitized myeloid cells (including ManR+ and CD11c+ cells) and predict dispersal mechanisms in a previously described immunodeficient model of L. donovani infection. Our results suggest that after initial seeding of infection in the skin, heavily parasite-infected myeloid cells are found in patches that resemble innate granulomas. Spread of parasites from these initial patches subsequently occurs through infection of recruited myeloid cells, ultimately leading to self-propagating networks of patch clusters. This combination of imaging and ecological pattern analysis to identify mechanisms driving the skin parasite landscape offers new perspectives on myeloid cell behavior following parasitism by L. donovani and may also be applicable to elucidating the behavior of other intracellular tissue-resident pathogens and their host cells.


Subject(s)
Image Processing, Computer-Assisted , Leishmania donovani/pathogenicity , Leishmaniasis, Visceral/parasitology , Microscopy, Confocal , Microscopy, Fluorescence , Myeloid Cells/parasitology , Skin/parasitology , Spatial Analysis , Animals , CD11 Antigens/metabolism , Cluster Analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Host-Parasite Interactions , Insect Vectors/parasitology , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/transmission , Mannose Receptor/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Theoretical , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phlebotomus/parasitology , Skin/immunology , Skin/metabolism
15.
Physiol Res ; 70(2): 213-226, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33676385

ABSTRACT

Pulmonary arterial hypertension (PAH) isa fatal disease characterized by vascular remodeling and chronic inflammation. Macrophages are the key orchestrators of inflammatory and repair responses, and have been demonstrated to be vital in the pathogenesis of PAH. However, specific phenotype of macrophage polarization (M1 & M2 macrophage) in the development of PAH and the underlying mechanisms how they work are still largely unclear. A rat model of monocrotaline (MCT) induced PAH was used. Hemodynamic analysis and histopathological experiments were conducted at day 3, 7, 14, 21 and 28, respectively. In PAH rat lung tissue, confocal microscopic images showed that CD68+NOS2+ M1-like macrophages were remarkably infiltrated on early stage, but dramatically decreased in mid-late stage. Meanwhile, CD68+CD206+ M2-like macrophages in lung tissue accumulated gradually since day 7 to day 28, and the relative ratio of M2/M1 macrophage increased over time. Results detected by western blot and immunohistochemistry were consistent. Further vitro functional studies revealed the possible mechanism involved in this pathophysiological process. By using Transwell co-culture system, it was found that M1 macrophages inducedendothelial cellapoptosis, while M2 macrophages significantly promoted proliferation of both endothelial cell and smooth muscle cell.These data preliminarily demonstrated a temporal dynamic change of macrophage M1/M2 polarization status in the development of experimental PAH. M1 macrophages participated in the initial stage of inflammation by accelerating apoptosis of endothelial cell, while M2 macrophages predominated in the reparative stage of inflammation and the followed stage of aberrant tissue remodeling.


Subject(s)
Macrophages/pathology , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/metabolism , Vascular Remodeling , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apoptosis , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammation Mediators/metabolism , Macrophages/metabolism , Male , Mannose Receptor/metabolism , Monocrotaline , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Nitric Oxide Synthase Type II/metabolism , Phenotype , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Time Factors
16.
ACS Appl Bio Mater ; 4(6): 4789-4799, 2021 06 21.
Article in English | MEDLINE | ID: mdl-35007028

ABSTRACT

Hepatocellular carcinoma (HCC) is the sixth most common neoplasia and the fourth most common cause of cancer-related mortality worldwide. Sorafenib is the first-line molecular therapy for patients in an advanced stage of HCC. However, the recommended clinical dose of Sorafenib is associated with several complications, which derive from its lack of cell specificity and its very low water solubility. To circumvent these drawbacks, in the present study we developed two sugar-coated polydiacetylene-based nanomicelles-Sorafenib carriers targeting mannose and asialoglycoprotein receptors (MR and ASGPR, respectively). The strategies allowed the inducement of apoptosis and reduction of cell proliferation at a nanomolar, instead of micromolar, range in liver cancer cells. The study showed that, contrary to literature data, Sorafenib included into the pMicMan (Man = mannose) vector (targeting MR) is more efficient than pMicGal (Gal = galactose) (targeting ASGPR). Indeed, pMicMan increased the endosomal incorporation with an increased intracellular Sorafenib concentration that induced apoptosis and reduced cell proliferation at a low concentration range (10-20 nM).


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Galactose/administration & dosage , Liver Neoplasms/drug therapy , Mannose/administration & dosage , Nanoparticles/administration & dosage , Polyacetylene Polymer/administration & dosage , Sorafenib/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Asialoglycoprotein Receptor/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Endosomes/metabolism , Galactose/chemistry , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Mannose/chemistry , Mannose Receptor/metabolism , Micelles , Nanoparticles/chemistry , Polyacetylene Polymer/chemistry , Sorafenib/chemistry
17.
Innate Immun ; 27(4): 313-323, 2021 05.
Article in English | MEDLINE | ID: mdl-34013820

ABSTRACT

Trichinella spiralis represents an effective treatment for autoimmune and inflammatory diseases. The effects of recombinant T. spiralis (TS) 53-kDa protein (rTsP53) on acute lung injury (ALI) remain unclear. Here, mice were divided randomly into a control group, LPS group, and rTsP53 + LPS group. ALI was induced in BALB/c mice by LPS (10 mg/kg) injected via the tail vein. rTsP53 (200 µl; 0.4 µg/µl) was injected subcutaneously three times at an interval of 5 d before inducing ALI in the rTsP53+LPS group. Lung pathological score, the ratio and markers of classic activated macrophages (M1) and alternatively activated macrophages (M2), cytokine profiles in alveolar lavage fluid, and pyroptosis protein expression in lung tissue were investigated. RTsP53 decreased lung pathological score. Furthermore, rTsP53 suppressed inflammation by increasing IL-4, IL-10, and IL-13. There was an increase in alveolar M2 macrophage numbers, with an increase in CD206 and arginase-1-positive cells and a decrease in alveolar M1 markers such as CD197 and iNOS. In addition, the polarization of M2 macrophages induced by rTsP53 treatment could alleviate ALI by suppressing lung pyroptosis. RTsP53 was identified as a potential agent for treating LPS-induced ALI via alleviating lung pyroptosis by promoting M2 macrophage polarization.


Subject(s)
Acute Lung Injury/drug therapy , Macrophages/drug effects , Protective Agents/pharmacology , Pyroptosis/drug effects , Trichinella spiralis/chemistry , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid , Cell Count , Cell Polarity/drug effects , Cytokines/chemistry , Interleukins/antagonists & inhibitors , Lipopolysaccharides , Macrophage Activation/drug effects , Male , Mannose Receptor/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/metabolism , Recombinant Proteins/pharmacology
18.
J Tissue Eng Regen Med ; 15(9): 788-797, 2021 09.
Article in English | MEDLINE | ID: mdl-34311512

ABSTRACT

Macrophages, thanks to their extreme plasticity, exert critical roles in wound healing by orchestrating tissue defenses in the early inflammatory phase, and by promoting tissue regeneration and angiogenesis at a later time point. In parallel, platelets release a large number of preformed molecules that could affect immunocyte functions. Platelet-rich plasma and platelet lysate (PL) have been widely used as a therapeutic preside for ulcers, although little is known about the effects of platelet-derived biomolecules on macrophage functions during wound healing. In this study, we analyze the effects of PL on macrophages phenotype and functions. Monocyte-derived macrophages were cultured in the presence of interferon-γ and lipopolysaccharides to induce the M1 polarization and were further exposed to 10% PL. PL treatment reduced CD80, CD86, and PDL-1 and enhanced CD206 and CD200R expression on macrophages analyzed by cytofluorimetry. Additionally, macrophage cultures show reduced TNF-α and CXCL10, while increased arginase protein, PPAR, TGF-ß, and VEGF. TGF-ß secretion was paralleled by the decrease of NFkB and increase of STAT3, STAT6, and SMAD2 and SMAD4. Supernatants of PL-treated macrophages induced a significant increase of type-I collagen and to a lesser extent of type-III collagen production by fibroblasts. Finally, the supernatant of PL-treated macrophages showed significantly reduced capacity to induce the in vitro migration of T lymphocytes. Our results demonstrate that PL dampens the macrophage secretion of pro-inflammatory cytokines and induces the release of arginase, TGF-ß, and VEGF that may affect angiogenesis and tissue regeneration, thus facilitating the wound healing process.


Subject(s)
Blood Platelets/chemistry , Cell Movement , Fibroblasts/metabolism , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , T-Lymphocytes/cytology , Transforming Growth Factor beta/metabolism , Arginase/metabolism , Cell Movement/drug effects , Chemokine CXCL10/metabolism , Collagen Type I/metabolism , Collagen Type III/metabolism , Fibroblasts/drug effects , Humans , Macrophages/drug effects , Mannose Receptor/metabolism , Phenotype , Signal Transduction/drug effects , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL