1
|
Jatczak-Pawlik I, Ewiak-Paszyńska A, Domowicz M, Jurewicz A, Stasiołek M. Intracellular Accumulation and Secretion of YKL-40 (CHI3L1) in the Course of DMSO-Induced HL-60 Cell Differentiation. Pharmaceuticals (Basel) 2024; 17:443. [PMID: 38675403 PMCID: PMC11053806 DOI: 10.3390/ph17040443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/04/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
YKL-40 (CHI3L1) is a matrix glycoprotein stored in human neutrophil-specific granules and released upon activation. While it is implicated in inflammation, cancer progression, and cell differentiation, its exact physiological role remains unclear. This study investigated the intracellular expression and secretion of YKL-40 by untreated and DMSO-treated HL-60 cells in association with surface expression of CD11b and CD66b throughout the differentiation process (up to 120 h). Secreted YKL-40 protein and mRNA levels of YKL-40, CD66b, and CD11b were measured by ELISA and quantitative RT-PCR, respectively. The intracellular YKL-40 and surface CD11b and CD66b expression were assessed by flow cytometry. A significant increase in CD11b expression confirmed DMSO-induced differentiation of HL-60 cells. Upon DMSO stimulation, YKL-40 mRNA expression increased in a time-dependent manner, unlike CD66b. The lack of CD66b (a granulocyte maturation and activation marker) on the surface of HL-60 cells might suggest that DMSO treatment did not induce full maturation or activation. The intracellular YKL-40 protein expression was increasing up to 96 h of DMSO treatment and then declined. YKL-40 secretion into the culture medium was detectable only at later time points (96 and 120 h), which was correlated with a decreased proliferation of DMSO-treated HL-60 cells. These findings suggest sequential changes in YKL-40 production and secretion during DMSO-induced differentiation of HL-60 cells and might contribute to a better understanding of YKL-40's involvement in both physiological processes and disease development, including multiple sclerosis.
Collapse
Affiliation(s)
| | | | | | | | - Mariusz Stasiołek
- Department of Neurology, Medical University of Lodz, Kosciuszki Street 4, 90-419 Lodz, Poland; (I.J.-P.); (A.E.-P.); (M.D.); (A.J.)
| |
Collapse
|
2
|
Hachani A, Giulieri SG, Guérillot R, Walsh CJ, Herisse M, Soe YM, Baines SL, Thomas DR, Cheung SD, Hayes AS, Cho E, Newton HJ, Pidot S, Massey RC, Howden BP, Stinear TP. A high-throughput cytotoxicity screening platform reveals agr-independent mutations in bacteraemia-associated Staphylococcus aureus that promote intracellular persistence. eLife 2023; 12:e84778. [PMID: 37289634 PMCID: PMC10259494 DOI: 10.7554/elife.84778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/23/2023] [Indexed: 06/10/2023] Open
Abstract
Staphylococcus aureus infections are associated with high mortality rates. Often considered an extracellular pathogen, S. aureus can persist and replicate within host cells, evading immune responses, and causing host cell death. Classical methods for assessing S. aureus cytotoxicity are limited by testing culture supernatants and endpoint measurements that do not capture the phenotypic diversity of intracellular bacteria. Using a well-established epithelial cell line model, we have developed a platform called InToxSa (intracellular toxicity of S. aureus) to quantify intracellular cytotoxic S. aureus phenotypes. Studying a panel of 387 S. aureus bacteraemia isolates, and combined with comparative, statistical, and functional genomics, our platform identified mutations in S. aureus clinical isolates that reduced bacterial cytotoxicity and promoted intracellular persistence. In addition to numerous convergent mutations in the Agr quorum sensing system, our approach detected mutations in other loci that also impacted cytotoxicity and intracellular persistence. We discovered that clinical mutations in ausA, encoding the aureusimine non-ribosomal peptide synthetase, reduced S. aureus cytotoxicity, and increased intracellular persistence. InToxSa is a versatile, high-throughput cell-based phenomics platform and we showcase its utility by identifying clinically relevant S. aureus pathoadaptive mutations that promote intracellular residency.
Collapse
Affiliation(s)
- Abderrahman Hachani
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Stefano G Giulieri
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Romain Guérillot
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Calum J Walsh
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Marion Herisse
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ye Mon Soe
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Sarah L Baines
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - David R Thomas
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Infection and Immunity Program, Department of Microbiology and Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Shane Doris Cheung
- Biological Optical Microscopy Platform, University of MelbourneMelbourneAustralia
| | - Ashleigh S Hayes
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ellie Cho
- Biological Optical Microscopy Platform, University of MelbourneMelbourneAustralia
| | - Hayley J Newton
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Infection and Immunity Program, Department of Microbiology and Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Sacha Pidot
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ruth C Massey
- School of Microbiology, University College CorkCorkIreland
- School of Medicine, University College CorkCorkIreland
- APC Microbiome Ireland, University College CorkCorkIreland
- School of Cellular and Molecular Medicine, University of BristolBristolUnited Kingdom
| | - Benjamin P Howden
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| |
Collapse
|
3
|
Wrighton S, Ahnlide VK, André O, Bahnan W, Nordenfelt P. Group A streptococci induce stronger M protein-fibronectin interaction when specific human antibodies are bound. Front Microbiol 2023; 14:1069789. [PMID: 36778879 PMCID: PMC9909010 DOI: 10.3389/fmicb.2023.1069789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023] Open
Abstract
Group A streptococcus (GAS) is a highly adapted, human-specific pathogen that is known to manipulate the immune system through various mechanisms. GAS' M protein constitutes a primary target of the immune system due to its spatial configuration and dominance on the bacterial surface. Antibody responses targeting the M protein have been shown to favor the conserved C region. Such antibodies (Abs) circumvent antigenic escape and efficiently bind to various M types. The ability of GAS to bind to fibronectin (Fn), a high molecular weight glycoprotein of the extracellular matrix, has long been known to be essential for the pathogen's evolutionary success and fitness. However, some strains lack the ability to efficiently bind Fn. Instead, they have been found to additionally bind Fn via the A-B domains of their M proteins. Here, we show that human Abs can induce increased Fn-binding affinity in M proteins, likely by enhancing the weak A-B domain binding. We found that this enhanced Fn binding leads to a reduction in Ab-mediated phagocytosis, indicating that this constitutes a GAS immune escape mechanism. We could show that the Fc domain of Abs is necessary to trigger this phenomenon and that Ab flexibility may also play a key role. We, moreover, saw that our Abs could enhance Fn binding in 3 out of 5 emm type strains tested, belonging to different clades, making it likely that this is a more generalizable phenomenon. Together our results suggest a novel synergistic interplay of GAS and host proteins which ultimately benefits the bacterium.
Collapse
|
4
|
Abstract
Phagocytosis is relevant for many research fields and is often measured as a functional outcome. However, accurate quantification can be challenging, and many researchers find it difficult to study in a robust manner. There are many ways to measure phagocytosis, but what is often overlooked is the importance of experimental design and how the analysis is planned and performed. Experimental factors like reaction volume, time, and phagocyte-prey concentrations often have a large impact on the outcome, as is the choice of detection strategy with different fluorescent or colorimetric labels of prey and phagocyte. By using dose-response curve principles for both experimental design and analysis, it is possible to increase the sensitivity and robustness, leading to accurate quantification of phagocytosis that is comparable across experiments and systems.Here, we describe how to quantify phagocytosis using flow cytometry with a robust, high-throughput, and easy-to-use approach. The prey is first fluorescently double stained, followed by optional opsonization before being introduced to the phagocyte in a wide range of ratios. After incubation, data is acquired through flow cytometry. It can be assessed on both the population and single-cell level of the phagocytes, separating adhesion and internalization. As an example, we provide an experimental protocol for studying phagocytosis of opsonized Streptococcus pyogenes using the THP-1 cell line. This approach is easily incorporated into most existing phagocytosis assays and allows for reproducible results with high sensitivity.
Collapse
Affiliation(s)
- Therese de Neergaard
- Department of Clinical Sciences Lund, Faculty of Medicine, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Department of Clinical Sciences Lund, Faculty of Medicine, Division of Infection Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
5
|
Georgakis S, Gkirtzimanaki K, Papadaki G, Gakiopoulou H, Drakos E, Eloranta ML, Makridakis M, Kontostathi G, Zoidakis J, Baira E, Rönnblom L, Boumpas DT, Sidiropoulos P, Verginis P, Bertsias G. NETs decorated with bioactive IL-33 infiltrate inflamed tissues and induce IFN-α production in patients with SLE. JCI Insight 2021; 6:147671. [PMID: 34554930 PMCID: PMC8663547 DOI: 10.1172/jci.insight.147671] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 09/22/2021] [Indexed: 12/28/2022] Open
Abstract
IL-33, a nuclear alarmin released during cell death, exerts context-specific effects on adaptive and innate immune cells, eliciting potent inflammatory responses. We screened blood, skin, and kidney tissues from patients with systemic lupus erythematosus (SLE), a systemic autoimmune disease driven by unabated type I IFN production, and found increased amounts of extracellular IL-33 complexed with neutrophil extracellular traps (NETs), correlating with severe, active disease. Using a combination of molecular, imaging, and proteomic approaches, we show that SLE neutrophils, activated by disease immunocomplexes, release IL-33–decorated NETs that stimulate robust IFN-α synthesis by plasmacytoid DCs in a manner dependent on the IL-33 receptor ST2L. IL33-silenced neutrophil-like cells cultured under lupus-inducing conditions generated NETs with diminished interferogenic effect. Importantly, NETs derived from patients with SLE are enriched in mature bioactive isoforms of IL-33 processed by the neutrophil proteases elastase and cathepsin G. Pharmacological inhibition of these proteases neutralized IL-33–dependent IFN-α production elicited by NETs. We believe these data demonstrate a novel role for cleaved IL-33 alarmin decorating NETs in human SLE, linking neutrophil activation, type I IFN production, and end-organ inflammation, with skin pathology mirroring that observed in the kidneys.
Collapse
Affiliation(s)
- Spiros Georgakis
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Iraklio, Greece.,Infections and Immunity, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Iraklio, Greece
| | - Katerina Gkirtzimanaki
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Iraklio, Greece.,Infections and Immunity, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Iraklio, Greece
| | - Garyfalia Papadaki
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Iraklio, Greece.,Infections and Immunity, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Iraklio, Greece
| | - Hariklia Gakiopoulou
- 1st Department of Pathology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Elias Drakos
- Department of Pathology, University of Crete, Medical School, Iraklio, Greece
| | - Maija-Leena Eloranta
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Manousos Makridakis
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Georgia Kontostathi
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Jerome Zoidakis
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Eirini Baira
- Laboratory of Toxicological Assessment of Pesticides, Scientific Directorate of Pesticides Assessment and Phytopharmacy, Benaki Phytopathological Institute, Athens, Greece
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Dimitrios T Boumpas
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece.,Joint Rheumatology Program and 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Prodromos Sidiropoulos
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Iraklio, Greece.,Infections and Immunity, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Iraklio, Greece
| | - Panayotis Verginis
- Infections and Immunity, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Iraklio, Greece.,Laboratory of Immune Regulation and Tolerance, University of Crete, Medical School, Iraklio, Greece
| | - George Bertsias
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Iraklio, Greece.,Infections and Immunity, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Iraklio, Greece
| |
Collapse
|
6
|
Abstract
M and M-like proteins are major virulence factors of the widespread and potentially deadly bacterial pathogen Streptococcus pyogenes. These proteins confer resistance against innate and adaptive immune responses by recruiting specific human proteins to the streptococcal surface. Nonimmune recruitment of immunoglobulins G (IgG) and A (IgA) through their fragment crystallizable (Fc) domains by M and M-like proteins was described almost 40 years ago, but its impact on virulence remains unresolved. These interactions have been suggested to be consequential under immune conditions at mucosal surfaces and in secretions but not in plasma, while other evidence suggests importance in evading phagocytic killing in nonimmune blood. Recently, an indirect effect of Fc-binding through ligand-induced stabilization of an M-like protein was shown to increase virulence. Nonimmune recruitment has also been seen to contribute to tissue damage in animal models of autoimmune diseases triggered by S. pyogenes infection. The damage was treatable by targeting Fc-binding. This and other potential therapeutic applications warrant renewed attention to Fc-binding by M and M-like proteins.
Collapse
Affiliation(s)
- Jori O. Mills
- Department of Chemistry & Biochemistry, La Jolla, California, United States of America
| | - Partho Ghosh
- Department of Chemistry & Biochemistry, La Jolla, California, United States of America
| |
Collapse
|
7
|
Blanter M, Gouwy M, Struyf S. Studying Neutrophil Function in vitro: Cell Models and Environmental Factors. J Inflamm Res 2021; 14:141-162. [PMID: 33505167 PMCID: PMC7829132 DOI: 10.2147/jir.s284941] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/04/2020] [Indexed: 01/21/2023] Open
Abstract
Neutrophils are the most abundant immune cell type in the blood and constitute the first line of defense against invading pathogens. Despite their important role in many diseases, they are challenging to study due to their short life span and the inability to cryopreserve or expand them in vitro. Thus, research into neutrophils has to rely on cells freshly isolated from peripheral blood of human donors, introducing donor-dependent variation in the experimental data. To counteract these problems, researchers tried to develop adequate cell models, such as cell lines. For those functional studies that cannot rely on cell models, a standardization of protocols regarding neutrophil purification and culturing could be a solution. In this review, we provide an overview of the most commonly used models for neutrophil function (HL-60, PLB-985, NB4, Kasumi-1 and induced pluripotent stem cells). In addition, we describe the effects of glucose concentration, pH, oxygen tension and temperature on neutrophil function.
Collapse
Affiliation(s)
- Marfa Blanter
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium
| |
Collapse
|
8
|
de Neergaard T, Sundwall M, Wrighton S, Nordenfelt P. High-Sensitivity Assessment of Phagocytosis by Persistent Association-Based Normalization. THE JOURNAL OF IMMUNOLOGY 2020; 206:214-224. [PMID: 33268484 DOI: 10.4049/jimmunol.2000032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 10/29/2020] [Indexed: 01/04/2023]
Abstract
Phagocytosis is measured as a functional outcome in many research fields, but accurate quantification can be challenging, with no robust method available for cross-laboratory reproducibility. In this study, we identified a simple, measurable parameter, persistent prey-phagocyte association, to use for normalization and dose-response analysis. We apply this in a straightforward analytical method, persistent association-based normalization, in which the multiplicity of prey (MOP) ratio needed to elicit half of the phagocytes to associate persistently (MOP50) is determined first. MOP50 is then applied to normalize for experimental factors, separately analyzing association and internalization. We use reference human phagocyte THP-1 cells with different prey and opsonization conditions to compare the persistent association-based normalization method to standard ways of assessing phagocytosis and find it to perform better, exhibiting increased robustness, sensitivity, and reproducibility. The approach is easily incorporated into most existing phagocytosis assays and allows for reproducible results with high sensitivity.
Collapse
Affiliation(s)
- Therese de Neergaard
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Martin Sundwall
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Sebastian Wrighton
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
9
|
Zha H, Matsunami E, Blazon-Brown N, Koutsogiannaki S, Hou L, Bu W, Babazada H, Odegard KC, Liu R, Eckenhoff RG, Yuki K. Volatile anesthetics affect macrophage phagocytosis. PLoS One 2019; 14:e0216163. [PMID: 31071106 PMCID: PMC6508649 DOI: 10.1371/journal.pone.0216163] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/15/2019] [Indexed: 12/18/2022] Open
Abstract
Background Perioperative infections, particularly surgical site infections pose significant morbidity and mortality. Phagocytosis is a critical step for microbial eradication. We examined the effect of commonly used anesthetics on macrophage phagocytosis and its mechanism. Methods The effect of anesthetics (isoflurane, sevoflurane, propofol) on macrophage phagocytosis was tested using RAW264.7 mouse cells, mouse peritoneal macrophages, and THP-1 human cells. Either opsonized sheep erythrocytes or fluorescent labeled Escherichia coli were used as phagocytic objects. The activation of Rap1, a critical protein in phagocytosis was assessed using the active Rap1 pull-down and detection kit. To examine anesthetic binding site(s) on Rap1, photolabeling experiments were performed using azi-isoflurane and azi-sevoflurane. The alanine scanning mutagenesis of Rap1 was performed to assess the role of anesthetic binding site in Rap1 activation and phagocytosis. Results Macrophage phagocytosis was significantly attenuated by the exposure of isoflurane (50% reduction by 1% isoflurane) and sevoflurane (50% reduction by 1.5% sevoflurane), but not by propofol. Photolabeling experiments showed that sevoflurane directly bound to Rap1. Mutagenesis analysis demonstrated that the sevoflurane binding site affected Rap1 activation and macrophage phagocytosis. Conclusions We showed that isoflurane and sevoflurane attenuated macrophage phagocytosis, but propofol did not. Our study showed for the first time that sevoflurane served as a novel small GTPase Rap1 inhibitor. The finding will further enrich our understanding of yet-to-be determined mechanism of volatile anesthetics and their off-target effects. The sevoflurane binding site was located outside the known Rap1 functional sites, indicating the discovery of a new functional site on Rap1 and this site would serve as a pocket for the development of novel Rap1 inhibitors.
Collapse
Affiliation(s)
- Hui Zha
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pediatrics, Union Hospital, Tonji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Erika Matsunami
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Anesthesia, Kawasaki Saiwai Hospital, Kawasaki, Kanagawa, Japan
| | - Nathan Blazon-Brown
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Sophia Koutsogiannaki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lifei Hou
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Weiming Bu
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Hasan Babazada
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kirsten C. Odegard
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Renyu Liu
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Roderic G. Eckenhoff
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
10
|
Oliveira JSSD, Santos GDS, Moraes JA, Saliba AM, Barja-Fidalgo TC, Mattos-Guaraldi AL, Nagao PE. Reactive oxygen species generation mediated by NADPH oxidase and PI3K/Akt pathways contribute to invasion of Streptococcus agalactiae in human endothelial cells. Mem Inst Oswaldo Cruz 2018; 113:e140421. [PMID: 29641644 PMCID: PMC5887998 DOI: 10.1590/0074-02760170421] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 03/05/2018] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Streptococcus agalactiae can causes sepsis, pneumonia, and meningitis in neonates, the elderly, and immunocompromised patients. Although the virulence properties of S. agalactiae have been partially elucidated, the molecular mechanisms related to reactive oxygen species (ROS) generation in infected human endothelial cells need further investigation. OBJECTIVES This study aimed to evaluate the influence of oxidative stress in human umbilical vein endothelial cells (HUVECs) during S. agalactiae infection. METHODS ROS production during S. agalactiae-HUVEC infection was detected using the probe CM-H2DCFDA. Microfilaments labelled with phalloidin-FITC and p47phox-Alexa 546 conjugated were analysed by immunofluorescence. mRNA levels of p47phox (NADPH oxidase subunit) were assessed using Real Time qRT-PCR. The adherence and intracellular viability of S. agalactiae in HUVECs with or without pre-treatment of DPI, apocynin (NADPH oxidase inhibitors), and LY294002 (PI3K inhibitor) were evaluated by penicillin/gentamicin exclusion. Phosphorylation of p47phox and Akt activation by S. agalactiae were evaluated by immunoblotting analysis. FINDINGS Data showed increased ROS production 15 min after HUVEC infection. Real-Time qRT-PCR and western blotting performed in HUVEC infected with S. agalactiae detected alterations in mRNA levels and activation of p47phox. Pre-treatment of endothelial cells with NADPH oxidase (DPI and apocynin) and PI3K/Akt pathway (LY294002) inhibitors reduced ROS production, bacterial intracellular viability, and generation of actin stress fibres in HUVECs infected with S. agalactiae. CONCLUSIONS ROS generation via the NADPH oxidase pathway contributes to invasion of S. agalactiae in human endothelial cells accompanied by cytoskeletal reorganisation through the PI3K/Akt pathway, which provides novel evidence for the involvement of oxidative stress in S. agalactiae pathogenesis.
Collapse
Affiliation(s)
- Jessica Silva Santos de Oliveira
- Departamento de Biologia Celular, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Gabriela da Silva Santos
- Departamento de Biologia Celular, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - João Alfredo Moraes
- Departamento de Biologia Celular, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Alessandra Mattos Saliba
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Thereza Christina Barja-Fidalgo
- Departamento de Biologia Celular, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | | | - Prescilla Emy Nagao
- Departamento de Biologia Celular, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
11
|
Zhu LF, Xiao M, Chen YQ, Wang LY, Luo XF, Yuan XH, Ren JH, Chen ZZ, Hu JD, Yang T. In vitro effects of reprogramming factors on the expressions of pluripotent genes and CD 34 gene in human acute promyelocytic leukemia HL-60 cells. Genomics 2017; 109:331-335. [PMID: 28533192 DOI: 10.1016/j.ygeno.2017.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/28/2017] [Accepted: 05/18/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Our study aims to explore the in vitro effects of reprogramming factors on the expressions of pluripotent genes and CD34 gene in HL-60 cells. METHODS According to the construction of lentiviral vector LV-OSCK of reprogramming factors (Oct-4, Sox2, Klf4, c-Myc), 293T cells were transfected to detect virus titer. The endogenous pluripotent genes (Oct4, SOX2, c-Myc and Klf4) and CD34 mRNA and protein expressions were detected by AP staining, immunofluorescence staining, qRT-PCR and flow cytometry. RESULTS Expressions of Oct4, SOX2, c-Myc and Klf4 were 0.220±0.013, 0.186±0.009, 0.287±0.015 and 0.153±0.007. These levels were significantly higher in the experimental group than the control and blank groups. CD34 protein expression in the experimental group was also discovered to be significantly higher than the other two groups. CONCLUSION The reprogramming factors could increase the expressions of pluripotent genes and CD34 gene in HL-60 cells.
Collapse
Affiliation(s)
- Liang-Fang Zhu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Min Xiao
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Yong-Quan Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Ling-Yan Wang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Xiao-Feng Luo
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Xiao-Hong Yuan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Jin-Hua Ren
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Zhi-Zhe Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Jian-Da Hu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Ting Yang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China.
| |
Collapse
|
12
|
Chen S, Zhang JQ, Chen JZ, Chen HX, Qiu FN, Yan ML, Chen YL, Peng CH, Tian YF, Wang YD. The over expression of long non-coding RNA ANRIL promotes epithelial-mesenchymal transition by activating the ATM-E2F1 signaling pathway in pancreatic cancer: An in vivo and in vitro study. Int J Biol Macromol 2017; 102:718-728. [PMID: 28344092 DOI: 10.1016/j.ijbiomac.2017.03.123] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 03/07/2017] [Accepted: 03/21/2017] [Indexed: 11/27/2022]
Abstract
This study aims to investigate the roles of lncRNA ANRIL in epithelial-mesenchymal transition (EMT) by regulating the ATM-E2F1 signaling pathway in pancreatic cancer (PC). PC rat models were established and ANRIL overexpression and interference plasmids were transfected. The expression of ANRIL, EMT markers (E-cadherin, N-cadherin and Vimentin) and ATM-E2F1 signaling pathway-related proteins (ATM, E2F1, INK4A, INK4B and ARF) were detected. Small molecule drugs were applied to activate and inhibit the ATM-E2F1 signaling pathway. Transwell assay and the scratch test were adopted to detect cell invasion and migration abilities. ANRIL expression in the PC cells was higher than in normal pancreatic duct epithelial cells. In the PC rat models and PC cells, ANRIL interference promoted the expressions of INK4B, INK4A, ARF and E-cadherin, while reduced N-cadherin and Vimentin expression. Over-expressed ANRIL decreased the expression of INK4B, INK4A, ARF and E-cadherin, but raised N-cadherin and Vimentin expressions. By inhibiting the ATM-E2F1 signaling pathway in PC cells, E-cadherin expression increased but N-cadherin and Vimentin expressions decreased. After ANRIL was silenced or the ATM-E2F1 signaling pathway inhibited, PC cell migration and invasion abilities were decreased. In conclusion, over-expression of lncRNA ANRIL can promote EMT of PC cells by activating the ATM-E2F1 signaling pathway.
Collapse
Affiliation(s)
- Shi Chen
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China; Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jia-Qiang Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiang-Zhi Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Hepatobiliary Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Hui-Xing Chen
- Department of Hepatobiliary Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Fu-Nan Qiu
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China
| | - Mao-Lin Yan
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China
| | - Yan-Ling Chen
- Department of Hepatobiliary Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Cheng-Hong Peng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi-Feng Tian
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China.
| | - Yao-Dong Wang
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China.
| |
Collapse
|
13
|
Yaseen R, Blodkamp S, Lüthje P, Reuner F, Völlger L, Naim HY, von Köckritz-Blickwede M. Antimicrobial activity of HL-60 cells compared to primary blood-derived neutrophils against Staphylococcus aureus. J Negat Results Biomed 2017; 16:2. [PMID: 28214466 PMCID: PMC5316427 DOI: 10.1186/s12952-017-0067-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 01/26/2017] [Indexed: 01/06/2023] Open
Abstract
Background The human leukemia cell line HL-60 is considered an alternative cell culture model to study neutrophil differentiation and migration. The aim of this study was to characterize the suitability of HL-60 cells differentiated to neutrophil-like cells (nHL-60) as substitute for blood-derived human neutrophils to investigate the interaction of neutrophils with Staphylococcus aureus. Methods For this purpose, antimicrobial activity, bacterial uptake, production of reactive oxygen species and the release of neutrophil extracellular traps (NETs) by nHL-60 cells were analyzed and compared to primary blood-derived neutrophils using Staphylococcus aureus as important human and animal pathogen. Results Overall, the antimicrobial activities of nHL-60 cells were distinctly lower compared to blood-derived neutrophils. Furthermore, production of reactive oxygen species as well as NET formation was clearly impaired in nHL-60 cells. Conclusion This study indicates that HL-60 cells are of limited usage as an alternative model to study antimicrobial functions of neutrophils against Staphylococcus aureus. Electronic supplementary material The online version of this article (doi:10.1186/s12952-017-0067-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ragheda Yaseen
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30599, Hannover, Germany
| | - Stefanie Blodkamp
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30599, Hannover, Germany
| | - Petra Lüthje
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30599, Hannover, Germany.,Division of Clinical Microbiology, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Friederike Reuner
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30599, Hannover, Germany
| | - Lena Völlger
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30599, Hannover, Germany
| | - Hassan Y Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30599, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30599, Hannover, Germany. .,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Buenteweg 17, 30599, Hannover, Germany.
| |
Collapse
|
14
|
Shrestha S, Kim SY, Yun YJ, Kim JK, Lee JM, Shin M, Song DK, Hong CW. Retinoic acid induces hypersegmentation and enhances cytotoxicity of neutrophils against cancer cells. Immunol Lett 2017; 182:24-29. [PMID: 28065603 DOI: 10.1016/j.imlet.2017.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/02/2017] [Indexed: 02/08/2023]
Abstract
Hypersegmentation of nuclei is considered a distinct characteristic of the antitumoral phenotype of neutrophils. Retinoic acid, a metabolite of retinol, reorganizes and induces segmentation of the nucleus during the differentiation of neutrophils. However, the role of retinoic acid in the phenotype polarization of neutrophils has not been fully established. Here, we investigated the effect of retinoic acid on phenotype polarization of neutrophils. Retinoic acid-induced the hypersegmentation of human neutrophils via retinoic acid receptors and mTOR pathways. Retinoic acid-induced hypersegmented neutrophils enhanced neutrophil extracellular traps (NETs) formation in response to phorbol-12-myristate 13-acetate (PMA) and fMLP (N-Formylmethionine-leucyl-phenylalanine) stimulation, and increased cytotoxicity against various tumor cells. Moreover, retinoic acid treatment attenuated tumor growth in a murine model of tumor. Taken together, these results suggests that retinoic acid induces the phenotype polarization of neutrophils to exert antitumor effects.
Collapse
Affiliation(s)
- Sanjeeb Shrestha
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Shin-Yeong Kim
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Young-Jin Yun
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jun-Kyu Kim
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jae Man Lee
- Department of Biochemistry & Cell Biology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Minsang Shin
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Dong-Keun Song
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Chang-Won Hong
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
15
|
Involvement of NADH Oxidase in Competition and Endocarditis Virulence in Streptococcus sanguinis. Infect Immun 2016; 84:1470-1477. [PMID: 26930704 PMCID: PMC4862721 DOI: 10.1128/iai.01203-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/20/2016] [Indexed: 11/20/2022] Open
Abstract
Here, we report for the first time that the Streptococcus sanguinis nox gene encoding NADH oxidase is involved in both competition with Streptococcus mutans and virulence for infective endocarditis. An S. sanguinis nox mutant was found to fail to inhibit the growth of Streptococcus mutans under microaerobic conditions. In the presence of oxygen, the recombinant Nox protein of S. sanguinis could reduce oxygen to water and oxidize NADH to NAD(+) The oxidation of NADH to NAD(+) was diminished in the nox mutant. The nox mutant exhibited decreased levels of extracellular H2O2; however, the intracellular level of H2O2 in the mutant was increased. Furthermore, the virulence of the nox mutant was attenuated in a rabbit endocarditis model. The nox mutant also was shown to be more sensitive to blood killing, oxidative and acid stresses, and reduced growth in serum. Thus, NADH oxidase contributes to multiple phenotypes related to competitiveness in the oral cavity and systemic virulence.
Collapse
|
16
|
Chen ML, Wu S, Tsai TC, Wang LK, Tsai FM. Regulation of neutrophil phagocytosis of Escherichia coli by antipsychotic drugs. Int Immunopharmacol 2014; 23:550-7. [PMID: 25448498 DOI: 10.1016/j.intimp.2014.09.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/24/2014] [Accepted: 09/26/2014] [Indexed: 01/21/2023]
Abstract
Antipsychotic drugs (APDs) have been used to ease the symptoms of schizophrenia. APDs have recently been reported to regulate the immune response. Our previous studies revealed that the atypical APDs risperidone and clozapine and the typical APD haloperidol can inhibit the phagocytic ability of macrophages. Our research next determined the effects of APDs on the phagocytic ability of neutrophils, which are the most abundant type of white blood cells in mammals. Here we provide evidence that clozapine and haloperidol can induce increased phagocytic uptake of Escherichia coli by differentiated HL-60 cells and by purified human neutrophils. Furthermore, clozapine and haloperidol can increase the myeloperoxidase activity and IL-8 production in neutrophils. Our results also show that clozapine can inhibit E. coli survival within differentiated HL-60 cells. Furthermore, clozapine and haloperidol are shown to enhance cell surface Mac-1 expression and the activated AKT signaling pathway in purified neutrophils exposed to E. coli. These results indicate that clozapine and haloperidol can increase the phagocytic ability of neutrophils by increasing AKT activation when cells are exposed to bacteria.
Collapse
Affiliation(s)
- Mao-Liang Chen
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, Taiwan
| | - Semon Wu
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, Taiwan; Department of Life Science, Chinese Culture University, Shih Lin, Taipei, Taiwan
| | - Tzung-Chieh Tsai
- Department of Microbiology, Immunology and Biopharmaceuticals, National Chiayi University, Chiayi, Taiwan
| | - Lu-Kai Wang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, Taiwan.
| |
Collapse
|
17
|
Chen ML, Wu S, Tsai TC, Wang LK, Chou WM, Tsai FM. The caffeic acid in aqueous extract of Tournefortia sarmentosa enhances neutrophil phagocytosis of Escherichia coli. Immunopharmacol Immunotoxicol 2014; 36:390-6. [PMID: 25311172 DOI: 10.3109/08923973.2014.956753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tournefortia sarmentosa, a Chinese herbal medicine, is considered an antioxidant or a detoxicating agent. Recently T. sarmentosa has received attention for its effects on the immune response. Here we provide evidence that aqueous extract of T. sarmentosa can induce increased phagocytic uptake of Escherichia coli by differentiated HL-60 cells and by neutrophils. Our results also revealed that T. sarmentosa can inhibit E. coli survival within differentiated HL-60 cells. Furthermore, aqueous extract of T. sarmentosa has been shown to enhance cell surface Mac-1 expression and the activated AKT signaling pathway in E. coli-stimulated neutrophils. We also examined the effect of each constituents in aqueous extract of T. sarmentosa on phagocytic uptake of E. coli by differentiated HL-60 cells or neutrophils. Bacterial survival, cell surface Mac-1 expression, and AKT activation of neutrophils were also examined. Our results showed that caffeic acid is an important constituent in mediating aqueous extract of T. sarmentosa-induced phagocytic uptake. Taken together, these results suggest that aqueous extract of T. sarmentosa exerts effects that enhance inflammatory responses by improving phagocytic capability, inhibiting bacterial survival within cells, and increasing Mac-1 expression of neutrophils.
Collapse
Affiliation(s)
- Mao-Liang Chen
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation , New Taipei City, Taiwan , ROC
| | | | | | | | | | | |
Collapse
|
18
|
Martínez-Zárate AD, Martínez-Vieyra I, Alonso-Rangel L, Cisneros B, Winder SJ, Cerecedo D. Dystroglycan depletion inhibits the functions of differentiated HL-60 cells. Biochem Biophys Res Commun 2014; 448:274-80. [DOI: 10.1016/j.bbrc.2014.04.110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 04/22/2014] [Indexed: 12/16/2022]
|
19
|
Abstract
Neutrophils have an incredible ability to find and eradicate intruders such as bacteria and fungi. They do this largely through the process of phagocytosis, where the target is internalized into a phagosome, and eventually destroyed by the hostile phagosomal environment. It is important to study phagocytosis in order to understand how neutrophils interact with various pathogens and how they respond to different stimuli. Here, I describe a method to study neutrophil phagocytosis of bacteria using flow cytometry. The bacteria are fluorescently labeled before being introduced to neutrophils. After phagocytosis, both any remaining extracellular bacteria and neutrophils are labeled using one-step staining before three-color analysis. To assess phagocytosis, first the average time it takes for the neutrophils to internalize all bound bacteria is determined. Experiments are then performed using that time point while varying the bacteria-to-neutrophil ratio for full control of the analysis. Due to the ease with which multiple samples can be analyzed, and the quantitative nature of flow cytometry, this approach is both reproducible and sensitive.
Collapse
|
20
|
Nordenfelt P, Björck L. IgG-binding bacterial proteins and pathogenesis. Future Microbiol 2013; 8:299-301. [DOI: 10.2217/fmb.13.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Pontus Nordenfelt
- Department of Clinical Sciences, Lund University, 22184 Lund, Sweden.
| | - Lars Björck
- Department of Clinical Sciences, Lund University, 22184 Lund, Sweden
| |
Collapse
|
21
|
Derochette S, Franck T, Mouithys-Mickalad A, Deby-Dupont G, Neven P, Serteyn D. Intra- and extracellular antioxidant capacities of the new water-soluble form of curcumin (NDS27) on stimulated neutrophils and HL-60 cells. Chem Biol Interact 2013; 201:49-57. [PMID: 23291280 DOI: 10.1016/j.cbi.2012.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/19/2012] [Accepted: 12/26/2012] [Indexed: 12/14/2022]
Abstract
Phagocytic cells, especially neutrophils (PMNs) are specialized in the production of reactive oxygen species (ROS) to kill pathogenic agents, but an excessive ROS production is associated with tissue damages and inflammatory diseases. Phagocytes are thus prime therapeutic targets to control inflammatory events associated to ROS production. Nowadays, there is a growing interest for the use of polyphenols to modulate the inflammatory response. The aim of this work was to study the antioxidant effect of NDS27, a highly water-soluble form of the polyphenolic molecule curcumin, on in vitro stimulated equine PMNs and human promyelocytic leukemia cells (HL-60). NDS27 was either pre-incubated with cells and eliminated before their activation (intracellular effect) or let in the medium (extracellular effect). Our results indicate that NDS27 significantly and dose-dependently (10(-6) M-10(-4) M) inhibited the ROS production in both cell types without affecting their viability. NDS27 was able to cross and interact with cell membrane, especially for HL-60 cells, while we observed a better intracellular antioxidant effect with PMNs. The activity of myeloperoxidase (MPO) released by PMNs and HL-60 cells, was decreased by NDS27, but more efficiently for PMNs. These results suggested that the greater efficiency of NDS27 in PMNs is due to an inhibitory effect on cells which are more mature for ROS production, probably by targeting the enzymes implied in respiratory burst like MPO. The modulatory effect of NDS27 on the oxidant activity of cells involved in immune and inflammatory responses opens perspectives for a therapeutic control of pathologies with excessive inflammatory reactions.
Collapse
Affiliation(s)
- Sandrine Derochette
- Centre for Oxygen, R&D (CORD), Institute of Chemistry, B6a, University of Liège, Allée de la Chimie 3, B-4000 Liège, Belgium.
| | | | | | | | | | | |
Collapse
|
22
|
Nordenfelt P, Waldemarson S, Linder A, Mörgelin M, Karlsson C, Malmström J, Björck L. Antibody orientation at bacterial surfaces is related to invasive infection. ACTA ACUST UNITED AC 2012; 209:2367-81. [PMID: 23230002 PMCID: PMC3526361 DOI: 10.1084/jem.20120325] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Several of the most significant bacterial pathogens in humans, including Streptococcus pyogenes, express surface proteins that bind IgG antibodies via their fragment crystallizable (Fc) region, and the dogma is that this protects the bacteria against phagocytic killing in blood. However, analysis of samples from a patient with invasive S. pyogenes infection revealed dramatic differences in the presence and orientation of IgG antibodies at the surface of bacteria from different sites. In the throat, IgG was mostly bound to the bacterial surface via Fc, whereas in the blood IgG was mostly bound via fragment antigen-binding (Fab). In infected and necrotic tissue, the Fc-binding proteins were removed from the bacterial surface. Further investigation showed that efficient bacterial IgGFc-binding occurs only in IgG-poor environments, such as saliva. As a consequence, the bacteria are protected against phagocytic killing, whereas in blood plasma where the concentration of IgG is high, the antibodies preferentially bind via Fab, facilitating opsonization and bacterial killing. IgG-poor environments represent the natural habitat for IgGFc-binding bacteria, and IgGFc-binding proteins may have evolved to execute their function in such environments. The lack of protection in plasma also helps to explain why cases of severe invasive infections with IgGFc-binding bacteria are so rare compared with superficial and uncomplicated infections.
Collapse
Affiliation(s)
- Pontus Nordenfelt
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
23
|
Lu T, Kobayashi SD, Quinn MT, Deleo FR. A NET Outcome. Front Immunol 2012; 3:365. [PMID: 23227026 PMCID: PMC3514450 DOI: 10.3389/fimmu.2012.00365] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 11/16/2012] [Indexed: 01/11/2023] Open
Abstract
Neutrophils constitute a critical part of innate immunity and are well known for their ability to phagocytose and kill invading microorganisms. The microbicidal processes employed by neutrophils are highly effective at killing most ingested bacteria and fungi. However, an alternative non-phagocytic antimicrobial mechanism of neutrophils has been proposed whereby microorganisms are eliminated by neutrophil extracellular traps (NETs). NETs are comprised of DNA, histones, and antimicrobial proteins extruded by neutrophils during NETosis, a cell death pathway reported to be distinct from apoptosis, phagocytosis-induced cell death, and necrosis. Although multiple laboratories have reported NETs using various stimuli in vitro, the molecular mechanisms involved in this process have yet to be definitively elucidated, and many questions regarding the formation and putative role or function of NETs in innate host defense remain unanswered. It is with these questions in mind that we provide some reflection and perspective on NETs and NETosis.
Collapse
Affiliation(s)
- Thea Lu
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton, MT, USA
| | | | | | | |
Collapse
|
24
|
Nordenfelt P, Grinstein S, Björck L, Tapper H. V-ATPase-mediated phagosomal acidification is impaired by Streptococcus pyogenes through Mga-regulated surface proteins. Microbes Infect 2012; 14:1319-29. [PMID: 22981599 DOI: 10.1016/j.micinf.2012.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 08/10/2012] [Accepted: 08/13/2012] [Indexed: 12/22/2022]
Abstract
Streptococcus pyogenes, a significant bacterial pathogen in humans, interferes with the membrane traffic of human neutrophils and survives following phagocytosis. The mechanism(s) behind this property is not known, but in contrast to wild-type bacteria, mutant bacteria lacking virulence factors regulated by the transcriptional regulator Mga, are phagocytosed and killed. In the present work we investigated whether differences in phagosomal acidification may contribute to this difference. Phagosomal pH in neutrophil-differentiated HL-60 cells was studied by fluorescence ratio imaging, and phagosomes containing wild-type S. pyogenes bacteria of the M1 serotype exhibited little or no acidification, whereas Mga mutant bacteria were found in more acidic phagosomes. With phagosomes containing these bacteria, proton delivery was inhibited by adding folimycin, a vacuolar-type adenosine triphosphatase (V-ATPase) inhibitor. This inhibitor had no effect on phagosomes containing wild-type bacteria, indicating either inactivation or removal of V-ATPases by the bacteria. Analysis of isolated bacteria-containing phagosomes confirmed the latter scenario and showed a more efficient delivery of V-ATPases to phagosomes containing Mga mutant bacteria. The results demonstrate that V-ATPase-mediated phagosomal proton delivery is reduced during phagocytosis of wild-type S. pyogenes, leading to impaired acidification, and that surface proteins of the mga regulon are responsible for this effect.
Collapse
Affiliation(s)
- Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Sciences, BMC, B14, Lund University, SE-221 84 Lund, Sweden.
| | | | | | | |
Collapse
|
25
|
Malmström L, Nordenfelt P, Malmström J. Business intelligence strategies enables rapid analysis of quantitative proteomics data. ACTA ACUST UNITED AC 2012. [DOI: 10.7243/2050-2273-1-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
26
|
Nordenfelt P, Tapper H. Phagosome dynamics during phagocytosis by neutrophils. J Leukoc Biol 2011; 90:271-84. [PMID: 21504950 DOI: 10.1189/jlb.0810457] [Citation(s) in RCA: 210] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The neutrophil is a key player in immunity, and its activities are essential for the resolution of infections. Neutrophil-pathogen interactions usually trigger a large arsenal of antimicrobial measures that leads to the highly efficient killing of pathogens. In neutrophils, the phagocytic process, including the formation and maturation of the phagosome, is in many respects very different from that in other phagocytes. Although the complex mechanisms that coordinate the membrane traffic, oxidative burst, and release of granule contents required for the microbicidal activities of neutrophils are not completely understood, it is evident that they are unique and differ from those in macrophages. Neutrophils exhibit more rapid rates of phagocytosis and higher intensity of oxidative respiratory response than do macrophages. The phagosome maturation pathway in macrophages, which is linked to the endocytic pathway, is replaced in neutrophils by the rapid delivery of preformed granules to nonacidic phagosomes. This review describes the plasticity and dynamics of the phagocytic process with a special focus on neutrophil phagosome maturation.
Collapse
Affiliation(s)
- Pontus Nordenfelt
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden.
| | | |
Collapse
|