1
|
Hosseini E, Afradiasbagharani P, Mohammadian M, Amjadi F, Tabatabaei M, Tanhaye Kalate Sabz F, Zandieh Z. Granulocyte-Macrophage Colony-Stimulating Factor Cytokine Addition After the Freeze-Thawing Process Improves Human Sperm Motility and Vitality in Asthenoteratozoospermia Patients. Biopreserv Biobank 2024; 22:38-45. [PMID: 37801668 DOI: 10.1089/bio.2022.0174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023] Open
Abstract
The cryopreservation-thawing process of spermatozoa cells has negative impacts on their structure, function, and fertility parameters, which are known as cryoinjury. Asthenozoospermia patients are more susceptible to cryoinjury. Granulocyte-macrophage colony-stimulating factor (GM-CSF) increases sperm glucose uptake via the induction of glucose transporters, resulting in increased sperm motility. This study aimed to investigate the efficiency of GM-CSF supplementation of the cryopreservation media for semen samples of asthenoteratozoospermia patients. The study was carried out on 20 semen samples from infertile men referred to diagnosing semen analysis. To avoid subjective bias, two main sperm motility parameters, including velocity along the curvilinear path and velocity along the straight-line path were considered by the computer-assisted sperm analysis system. Afterward, each semen sample was divided into three equal aliquots and randomly assigned to one of the following groups: group I (control, freezing media only), group II (+GM-CSF, freezing medium supplemented with 2 μL/mL GM-CSF), or group III (GM-CSF added after thawing and washing). Following semen thawing, standard parameters, mitochondrial membrane potential (MMP), and the DNA Fragmentation Index were analyzed. Total sperm motility (progressive and non-progressive) improved significantly in group III samples after a 30-minute incubation with GM-CSF compared with the control group (26.5% ± 3.1% vs. 17.51% ± 2.59%). However, no differences in progressive motility or sperm morphology were found among the three thawed samples. The percentage of vitality was significantly higher in group III compared with the other two groups (28.38% ± 3.4% vs. 22.4% ± 3.08% and 22.14% ± 2.77%, respectively) (p < 0.05). JC-1 levels (a marker of MMP) were not significantly different between the examined groups (44.95% ± 8.26% vs. 36.61% ± 6.95% vs. 46.67% ± 7.7%, for control, group II, and group III, respectively) (p > 0.05). GM-CSF may be advantageous as an additive after freezing, improving total motility and viability after 30 minutes of post-thaw incubation; however, when supplied to the freezing media before cryopreservation, it is unable to protect against cryoinjury.
Collapse
Affiliation(s)
- Elham Hosseini
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Obstetrics and Gynecology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Parivash Afradiasbagharani
- Department of Obstetrics and Gynecology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Urology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Masoud Mohammadian
- Mousavi Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Microbiology, Biology Research Center, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - FatemehSadat Amjadi
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Science, Tehran, Iran
- Department of Anatomical Science, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Tabatabaei
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Science, Tehran, Iran
| | - Fateme Tanhaye Kalate Sabz
- Department of Anatomical Sciences and Pathology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zahra Zandieh
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Science, Tehran, Iran
- Department of Anatomical Science, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Saha R, Patkar S, Pillai MM, Tayalia P. Bilayered skin substitute incorporating rutin nanoparticles for antioxidant, anti-inflammatory, and anti-fibrotic effect. BIOMATERIALS ADVANCES 2023; 150:213432. [PMID: 37119696 DOI: 10.1016/j.bioadv.2023.213432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/20/2023] [Accepted: 04/17/2023] [Indexed: 05/01/2023]
Abstract
Hypertrophic scarring in large burns and delayed healing in chronic wounds are consequences of prolonged and aggravated inflammation, sustained infiltration of immune cells, free radical generation, and abundance of inflammatory mediators. Therefore, it is imperative to curb hyperinflammation to expedite wound healing. In this study, rutin nanoparticles (RNPs) were synthesized without an encapsulant and incorporated into eggshell membrane powder-crosslinked gelatin-chitosan cryogels to impart antioxidant and anti-inflammatory properties for treating hyperinflammation. The resultant nanoparticles were found to be 17.53 ± 4.03 nm in size and were stable at room temperature for a month with no visible sedimentation. RNPs were found to be non-cytotoxic and exhibited anti-inflammatory (by increasing IL-10 levels) and antioxidant properties (by controlling the generation of reactive oxygen species and enhancing catalase production in human macrophages). Additionally, RNPs were found to reduce α-SMA expression in fibroblasts, thereby demonstrating their anti-scarring effect. In vivo studies with a bilayered skin substitute constituting an RNP-incorporated cryogel proved that it is biocompatible, does not induce renal toxicity, aids wound healing, and induces better re-epithelialization than the control groups at the initial stages. Thus, RNP-incorporated cryogels containing bilayered skin substitutes are an advanced and novel alternative to commercial dermo-epidermal substitutes that lack anti-inflammatory or anti-scarring properties.
Collapse
Affiliation(s)
- Rituparna Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Shivali Patkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Mamatha M Pillai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Prakriti Tayalia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
3
|
Seif M, Kakoschke TK, Ebel F, Bellet MM, Trinks N, Renga G, Pariano M, Romani L, Tappe B, Espie D, Donnadieu E, Hünniger K, Häder A, Sauer M, Damotte D, Alifano M, White PL, Backx M, Nerreter T, Machwirth M, Kurzai O, Prommersberger S, Einsele H, Hudecek M, Löffler J. CAR T cells targeting Aspergillus fumigatus are effective at treating invasive pulmonary aspergillosis in preclinical models. Sci Transl Med 2022; 14:eabh1209. [PMID: 36170447 DOI: 10.1126/scitranslmed.abh1209] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Aspergillus fumigatus is a ubiquitous mold that can cause severe infections in immunocompromised patients, typically manifesting as invasive pulmonary aspergillosis (IPA). Adaptive and innate immune cells that respond to A. fumigatus are present in the endogenous repertoire of patients with IPA but are infrequent and cannot be consistently isolated and expanded for adoptive immunotherapy. Therefore, we gene-engineered A. fumigatus-specific chimeric antigen receptor (Af-CAR) T cells and demonstrate their ability to confer antifungal reactivity in preclinical models in vitro and in vivo. We generated a CAR targeting domain AB90-E8 that recognizes a conserved protein antigen in the cell wall of A. fumigatus hyphae. T cells expressing the Af-CAR recognized A. fumigatus strains and clinical isolates and exerted a direct antifungal effect against A. fumigatus hyphae. In particular, CD8+ Af-CAR T cells released perforin and granzyme B and damaged A. fumigatus hyphae. CD8+ and CD4+ Af-CAR T cells produced cytokines that activated macrophages to potentiate the antifungal effect. In an in vivo model of IPA in immunodeficient mice, CD8+ Af-CAR T cells localized to the site of infection, engaged innate immune cells, and reduced fungal burden in the lung. Adoptive transfer of CD8+ Af-CAR T cells conferred greater antifungal efficacy compared to CD4+ Af-CAR T cells and an improvement in overall survival. Together, our study illustrates the potential of gene-engineered T cells to treat aggressive infectious diseases that are difficult to control with conventional antimicrobial therapy and support the clinical development of Af-CAR T cell therapy to treat IPA.
Collapse
Affiliation(s)
- Michelle Seif
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Tamara Katharina Kakoschke
- Klinik und Poliklinik für Mund-, Kiefer- und Gesichtschirurgie, Klinikum der Universität München, LMU, 80337 München, Germany.,Institut für Infektionsmedizin und Zoonosen, Medizinische Fakultät, LMU, 80539 München, Germany
| | - Frank Ebel
- Institut für Infektionsmedizin und Zoonosen, Medizinische Fakultät, LMU, 80539 München, Germany
| | - Marina Maria Bellet
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Nora Trinks
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum und RVZ - Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Giorgia Renga
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Marilena Pariano
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Luigina Romani
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Beeke Tappe
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - David Espie
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014 Paris, France.,CAR-T Cells Department, Invectys, 75013 Paris, France
| | - Emmanuel Donnadieu
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014 Paris, France.,Equipe labellisée Ligue Contre le Cancer, 75014 Paris, France
| | - Kerstin Hünniger
- Institut für Hygiene und Mikrobiologie, Julius-Maximilians-Universität Würzburg, 97080 Würzburg, Germany.,Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Antje Häder
- Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Markus Sauer
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum und RVZ - Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Diane Damotte
- Department of Pathology, Paris Centre University Hospitals, AP-HP, 75014 Paris, France.,INSERM U1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Pierre and Marie Curie, 75006 Paris, France
| | - Marco Alifano
- Department of Thoracic Surgery, Paris Centre University Hospitals, AP-HP, Paris, France; University Paris Descartes, 75014 Paris, France
| | - P Lewis White
- Public Health Wales, Microbiology Cardiff, UHW, CF14 4XW Cardiff, UK
| | - Matthijs Backx
- Public Health Wales, Microbiology Cardiff, UHW, CF14 4XW Cardiff, UK
| | - Thomas Nerreter
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Markus Machwirth
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Oliver Kurzai
- Institut für Hygiene und Mikrobiologie, Julius-Maximilians-Universität Würzburg, 97080 Würzburg, Germany.,Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Sabrina Prommersberger
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Jürgen Löffler
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
4
|
Membrane-Tethered Mucin 1 Is Stimulated by Interferon and Virus Infection in Multiple Cell Types and Inhibits Influenza A Virus Infection in Human Airway Epithelium. mBio 2022; 13:e0105522. [PMID: 35699372 PMCID: PMC9426523 DOI: 10.1128/mbio.01055-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Influenza A virus (IAV) causes significant morbidity and mortality in the human population. Tethered mucin 1 (MUC1) is highly expressed in airway epithelium, the primary site of IAV replication, and also by other cell types that influence IAV infection, including macrophages. MUC1 has the potential to influence infection dynamics through physical interactions and/or signaling activity, yet MUC1 modulation and its impact during viral pathogenesis remain unclear. Thus, we investigated MUC1-IAV interactions in an in vitro model of human airway epithelium (HAE). Our data indicate that a recombinant IAV hemagglutinin (H3) and H3N2 virus can bind endogenous HAE MUC1. Notably, infection of HAE with H1N1 or H3N2 IAV strains does not trigger MUC1 shedding but instead stimulates an increase in cell-associated MUC1 protein. We observed a similar increase after type I or III interferon (IFN) stimulation; however, inhibition of IFN signaling during H1N1 infection only partially abrogated this increase, indicating that multiple soluble factors contribute to MUC1 upregulation during the antiviral response. In addition to HAE, primary human monocyte-derived macrophages also upregulated MUC1 protein in response to IFN treatment and conditioned media from IAV-infected HAE. Then, to determine the impact of MUC1 on IAV pathogenesis, we developed HAE genetically depleted of MUC1 and found that MUC1 knockout cultures exhibited enhanced viral growth compared to control cultures for several IAV strains. Together, our data support a model whereby MUC1 inhibits productive uptake of IAV in HAE. Infection then stimulates MUC1 expression on multiple cell types through IFN-dependent and -independent mechanisms that further impact infection dynamics.
Collapse
|
5
|
COVID-19: Proposing a Ketone-Based Metabolic Therapy as a Treatment to Blunt the Cytokine Storm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6401341. [PMID: 33014275 PMCID: PMC7519203 DOI: 10.1155/2020/6401341] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Human SARS-CoV-2 infection is characterized by a high mortality rate due to some patients developing a large innate immune response associated with a cytokine storm and acute respiratory distress syndrome (ARDS). This is characterized at the molecular level by decreased energy metabolism, altered redox state, oxidative damage, and cell death. Therapies that increase levels of (R)-beta-hydroxybutyrate (R-BHB), such as the ketogenic diet or consuming exogenous ketones, should restore altered energy metabolism and redox state. R-BHB activates anti-inflammatory GPR109A signaling and inhibits the NLRP3 inflammasome and histone deacetylases, while a ketogenic diet has been shown to protect mice from influenza virus infection through a protective γδ T cell response and by increasing electron transport chain gene expression to restore energy metabolism. During a virus-induced cytokine storm, metabolic flexibility is compromised due to increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) that damage, downregulate, or inactivate many enzymes of central metabolism including the pyruvate dehydrogenase complex (PDC). This leads to an energy and redox crisis that decreases B and T cell proliferation and results in increased cytokine production and cell death. It is hypothesized that a moderately high-fat diet together with exogenous ketone supplementation at the first signs of respiratory distress will increase mitochondrial metabolism by bypassing the block at PDC. R-BHB-mediated restoration of nucleotide coenzyme ratios and redox state should decrease ROS and RNS to blunt the innate immune response and the associated cytokine storm, allowing the proliferation of cells responsible for adaptive immunity. Limitations of the proposed therapy include the following: it is unknown if human immune and lung cell functions are enhanced by ketosis, the risk of ketoacidosis must be assessed prior to initiating treatment, and permissive dietary fat and carbohydrate levels for exogenous ketones to boost immune function are not yet established. The third limitation could be addressed by studies with influenza-infected mice. A clinical study is warranted where COVID-19 patients consume a permissive diet combined with ketone ester to raise blood ketone levels to 1 to 2 mM with measured outcomes of symptom severity, length of infection, and case fatality rate.
Collapse
|
6
|
Zellnitz S, Zellnitz L, Müller M, Meindl C, Schröttner H, Fröhlich E. Impact of drug particle shape on permeability and cellular uptake in the lung. Eur J Pharm Sci 2019; 139:105065. [DOI: 10.1016/j.ejps.2019.105065] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/24/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023]
|
7
|
Layhadi JA, Turner J, Crossman D, Fountain SJ. ATP Evokes Ca 2+ Responses and CXCL5 Secretion via P2X 4 Receptor Activation in Human Monocyte-Derived Macrophages. THE JOURNAL OF IMMUNOLOGY 2017; 200:1159-1168. [PMID: 29255078 DOI: 10.4049/jimmunol.1700965] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/16/2017] [Indexed: 01/08/2023]
Abstract
Leukocytes sense extracellular ATP, a danger-associated molecular pattern, released during cellular stress and death, via activation of cell surface P2X and P2Y receptors. Here, we investigate P2 receptor expression in primary human monocyte-derived macrophages and receptors that mediate ATP-evoked intracellular [Ca2+]i signals and cytokine production in response to ATP concentrations that exclude P2X7 receptor activation. Expression of P2X1, P2X4, P2X5, P2X7, P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, and P2Y13 was confirmed by quantitative RT-PCR and immunocytochemistry. ATP elicited intracellular Ca2+ responses in a concentration-dependent fashion (EC50 = 11.4 ± 2.9 μM, n = 3). P2Y11 and P2Y13 activations mediated the amplitude of [Ca2+]i response, whereas P2X4 activation, but not P2X1 or P2X7, determined the duration of Ca2+ response during a sustained phase. ATP mediated gene induction of CXCL5, a proinflammatory chemokine. P2X4 antagonism (PSB-12062 or BX430) inhibited ATP-mediated induction of CXCL5 gene expression and secretion of CXCL5 by primary macrophage. Inhibition of CXCL5 secretion by P2X4 antagonists was lost in the absence of extracellular Ca2+ Reciprocally, positive allosteric modulation of P2X4 (ivermectin) augmented ATP-mediated CXCL5 secretion. P2X7, P2Y11, or P2Y13 receptor did not contribute to CXCL5 secretion. Together, the data reveals a role for P2X4 in determining the duration of ATP-evoked Ca2+ responses and CXCL5 secretion in human primary macrophage.
Collapse
Affiliation(s)
- Janice A Layhadi
- School of Biological Sciences, University of East Anglia, Norwich, Norfolk NR4 7TJ, United Kingdom
| | - Jeremy Turner
- Elsie Bertram Diabetes Centre, Norfolk and Norwich University Hospital, Norwich NR4 7UY, United Kingdom; and
| | - David Crossman
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, United Kingdom
| | - Samuel J Fountain
- School of Biological Sciences, University of East Anglia, Norwich, Norfolk NR4 7TJ, United Kingdom;
| |
Collapse
|
8
|
Victoni T, Salvator H, Abrial C, Brollo M, Porto LCS, Lagente V, Naline E, Grassin-Delyle S, Devillier P. Human lung and monocyte-derived macrophages differ with regard to the effects of β 2-adrenoceptor agonists on cytokine release. Respir Res 2017. [PMID: 28637505 PMCID: PMC5480184 DOI: 10.1186/s12931-017-0613-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background β2-adrenoceptor agonists have been shown to reduce the lipopolysaccharide (LPS)-induced cytokine release by human monocyte-derived macrophages (MDMs). We compare the expression of β2-adrenoceptors and the inhibitory effect of formoterol and salmeterol on the LPS-induced release of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6 and a range of chemokines (CCL2, 3, 4, and IL-8) by human lung macrophages (LMs) and MDMs. Methods LMs were isolated from patients undergoing resection and MDMs were obtained from blood monocytes in the presence of GM-CSF. LMs and MDMs were incubated in the absence or presence of formoterol or salmeterol prior to stimulation with LPS. The effects of formoterol were also assessed in the presence of the phosphodiesterase inhibitor roflumilast. Results LPS-induced cytokine production was higher in LMs than in MDMs. Salmeterol and formoterol exerted an inhibitory effect on the LPS-induced production of TNF-α, IL-6, CCL2, CCL3, and CCL4 in MDMs. In contrast, the β2-adrenoceptor agonists were devoid of any effect on LMs - even in the presence of roflumilast. The expression of β2-adrenergic receptors was detected on Western blots in MDMs but not in LMs. Conclusions Concentrations of β2-adrenoceptor agonists that cause relaxation of the human bronchus can inhibit cytokine production by LPS-stimulated MDMs but not by LMs. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0613-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tatiana Victoni
- Laboratory of Histocompatibility and Cryopresevation, Laboratory of Tissue Repair, Rio de Janeiro, Brazil.,Laboratory of Research in Respiratory Pharmacology-UPRES EA220, UFR Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin, Université Paris-Saclay, 11, rue Guillaume Lenoir, F-92150, Suresnes, France
| | - Hélène Salvator
- Laboratory of Research in Respiratory Pharmacology-UPRES EA220, UFR Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin, Université Paris-Saclay, 11, rue Guillaume Lenoir, F-92150, Suresnes, France.,Department of Airway Diseases, Foch Hospital, Suresnes, France
| | - Charlotte Abrial
- Laboratory of Research in Respiratory Pharmacology-UPRES EA220, UFR Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin, Université Paris-Saclay, 11, rue Guillaume Lenoir, F-92150, Suresnes, France
| | - Marion Brollo
- Laboratory of Research in Respiratory Pharmacology-UPRES EA220, UFR Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin, Université Paris-Saclay, 11, rue Guillaume Lenoir, F-92150, Suresnes, France
| | | | - Vincent Lagente
- Nutrition Metabolisms and Cancer, INSERM, INRA, Université Rennes 1, Université Bretagne Loire, Rennes, France
| | - Emmanuel Naline
- Laboratory of Research in Respiratory Pharmacology-UPRES EA220, UFR Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin, Université Paris-Saclay, 11, rue Guillaume Lenoir, F-92150, Suresnes, France.,Department of Airway Diseases, Foch Hospital, Suresnes, France
| | - Stanislas Grassin-Delyle
- Department of Airway Diseases, Foch Hospital, Suresnes, France.,INSERM UMR1173 & Mass Spectrometry Facility, UFR Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin, Université Paris-Saclay, Montigny-le-Bretonneux, France
| | - Philippe Devillier
- Laboratory of Research in Respiratory Pharmacology-UPRES EA220, UFR Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin, Université Paris-Saclay, 11, rue Guillaume Lenoir, F-92150, Suresnes, France. .,Department of Airway Diseases, Foch Hospital, Suresnes, France.
| |
Collapse
|
9
|
Short JD, Downs K, Tavakoli S, Asmis R. Protein Thiol Redox Signaling in Monocytes and Macrophages. Antioxid Redox Signal 2016; 25:816-835. [PMID: 27288099 PMCID: PMC5107717 DOI: 10.1089/ars.2016.6697] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Monocyte and macrophage dysfunction plays a critical role in a wide range of inflammatory disease processes, including obesity, impaired wound healing diabetic complications, and atherosclerosis. Emerging evidence suggests that the earliest events in monocyte or macrophage dysregulation include elevated reactive oxygen species production, thiol modifications, and disruption of redox-sensitive signaling pathways. This review focuses on the current state of research in thiol redox signaling in monocytes and macrophages, including (i) the molecular mechanisms by which reversible protein-S-glutathionylation occurs, (ii) the identification of bona fide S-glutathionylated proteins that occur under physiological conditions, and (iii) how disruptions of thiol redox signaling affect monocyte and macrophage functions and contribute to atherosclerosis. Recent Advances: Recent advances in redox biochemistry and biology as well as redox proteomic techniques have led to the identification of many new thiol redox-regulated proteins and pathways. In addition, major advances have been made in expanding the list of S-glutathionylated proteins and assessing the role that protein-S-glutathionylation and S-glutathionylation-regulating enzymes play in monocyte and macrophage functions, including monocyte transmigration, macrophage polarization, foam cell formation, and macrophage cell death. CRITICAL ISSUES Protein-S-glutathionylation/deglutathionylation in monocytes and macrophages has emerged as a new and important signaling paradigm, which provides a molecular basis for the well-established relationship between metabolic disorders, oxidative stress, and cardiovascular diseases. FUTURE DIRECTIONS The identification of specific S-glutathionylated proteins as well as the mechanisms that control this post-translational protein modification in monocytes and macrophages will facilitate the development of new preventive and therapeutic strategies to combat atherosclerosis and other metabolic diseases. Antioxid. Redox Signal. 25, 816-835.
Collapse
Affiliation(s)
- John D Short
- 1 Department of Pharmacology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Kevin Downs
- 2 Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Sina Tavakoli
- 3 Department of Radiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Reto Asmis
- 4 Department of Clinical Laboratory Sciences, University of Texas Health Science Center at San Antonio , San Antonio, Texas.,5 Department of Biochemistry, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| |
Collapse
|
10
|
Differential induction of inflammatory cytokines and reactive oxygen species in murine peritoneal macrophages and resident fresh bone marrow cells by acute staphylococcus aureus infection: contribution of toll-like receptor 2 (TLR2). Inflammation 2015; 38:224-44. [PMID: 25266881 DOI: 10.1007/s10753-014-0026-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Among the known Toll-like receptors (TLRs), Toll-like receptor 2 (TLR2) is a key sensor for detecting Staphylococcus aureus invasion. But the function of TLR2 during S. aureus infection in different cell populations is unclear. Two different cell subtypes were chosen to study the interaction of S. aureus with TLR2 because macrophages are extremely different from one compartment to another and their capacity to respond to live bacteria or bacterial products differs from one site to another. The contribution of TLR2 to the host innate response against acute live S. aureus infection and heat-killed S. aureus (HKSA) using anti-TLR2 antibody in murine peritoneal macrophages and resident fresh bone marrow cells has been investigated here. TLR2 blocking before infection induces the release of interleukin (IL)-10 by macrophages thereby inhibiting excessive production of oxidants by activating antioxidant enzymes. TLR2-blocked peritoneal macrophages showed impaired release of tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ) and IL-6 in response to both live and heat-killed S. aureus infection except bone marrow cells. TLR2-mediated free radical production and killing of S. aureus were modulated by TLR2 blocking in peritoneal macrophages and resident bone marrow cells. This study supported that S. aureus persists in resident bone marrow cells in a state of quiescence.
Collapse
|
11
|
Lindsay CD, Griffiths GD. Addressing bioterrorism concerns: options for investigating the mechanism of action of Staphylococcus aureus enterotoxin B. Hum Exp Toxicol 2013; 32:606-19. [PMID: 23023027 DOI: 10.1177/0960327112458941] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Staphylococcal enterotoxin B (SEB) is of concern to military and civilian populations as a bioterrorism threat agent. It is a highly potent toxin produced by Staphylococcus aureus and is stable in storage and under aerosolisation; it is able to produce prolonged highly incapacitating illness at very low-inhaled doses and death at elevated doses. Concerns regarding SEB are compounded by the lack of effective medical countermeasures for mass treatment of affected populations. This article considers the mechanism of action of SEB, the availability of appropriate experimental models for evaluating the efficacy of candidate medical countermeasures with particular reference to the need to realistically model SEB responses in man and the availability of candidate countermeasures (with an emphasis on commercial off-the-shelf options). The proposed in vitro approaches would be in keeping with Dstl’s commitment to reduction, refinement and replacement of animal models in biomedical research, particularly in relation to identifying valid alternatives to the use of nonhuman primates in experimental studies.
Collapse
Affiliation(s)
- C D Lindsay
- Biomedical Sciences Department, Dstl Porton Down, Salisbury, Wiltshire, UK.
| | | |
Collapse
|
12
|
Hoskins C, Wang L, Cheng WP, Cuschieri A. Dilemmas in the reliable estimation of the in-vitro cell viability in magnetic nanoparticle engineering: which tests and what protocols? NANOSCALE RESEARCH LETTERS 2012; 7:77. [PMID: 22247975 PMCID: PMC3275459 DOI: 10.1186/1556-276x-7-77] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 01/16/2012] [Indexed: 05/31/2023]
Abstract
Magnetic nanoparticles [MNPs] made from iron oxides have many applications in biomedicine. Full understanding of the interactions between MNPs and mammalian cells is a critical issue for their applications. In this study, MNPs were coated with poly(ethylenimine) [MNP-PEI] and poly(ethylene glycol) [MNP-PEI-PEG] to provide a subtle difference in their surface charge and their cytotoxicity which were analysed by three standard cell viability assays: 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium [MTS], CellTiter-Blue and CellTiter-Glo (Promega, Southampton, UK) in SH-SY5Y and RAW 264.7 cells The data were validated by traditional trypan blue exclusion. In comparison to trypan blue manual counting, the MTS and Titer-Blue assays appeared to have consistently overestimated the viability. The Titer-Glo also experienced a small overestimation. We hypothesise that interactions were occurring between the assay systems and the nanoparticles, resulting in incorrect cell viability evaluation. To further understand the cytotoxic effect of the nanoparticles on these cells, reactive oxygen species production, lipid peroxidation and cell membrane integrity were investigated. After pegylation, the MNP-PEI-PEG possessed a lower positive surface charge and exhibited much improved biocompatibility compared to MNP-PEI, as demonstrated not only by a higher cell viability, but also by a markedly reduced oxidative stress and cell membrane damage. These findings highlight the importance of assay selection and of dissection of different cellular responses in in-vitro characterisation of nanostructures.
Collapse
Affiliation(s)
- Clare Hoskins
- Institute for Medical Science and Technology (IMSaT), Wilson House, 1 Wurzburg Loan, University of Dundee, Dundee, DD2 1FD, UK.
| | | | | | | |
Collapse
|
13
|
Bacillus anthracis edema toxin suppresses human macrophage phagocytosis and cytoskeletal remodeling via the protein kinase A and exchange protein activated by cyclic AMP pathways. Infect Immun 2009; 77:2530-43. [PMID: 19307216 DOI: 10.1128/iai.00905-08] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Bacillus anthracis, the etiological agent of anthrax, is a gram-positive spore-forming bacterium. It produces edema toxin (EdTx), a powerful adenylate cyclase that increases cyclic AMP (cAMP) levels in host cells. Because other cAMP-increasing agents inhibit key macrophage (MPhi) functions, such as phagocytosis, it was hypothesized that EdTx would exhibit similar suppressive activities. Our previous GeneChip data showed that EdTx downregulated MPhi genes involved in actin cytoskeleton remodeling, including protein kinase A (PKA). To further examine the role of EdTx during anthrax pathogenesis, we explored the hypothesis that EdTx treatment leads to deregulation of the cAMP-dependent PKA system, resulting in impaired cytoskeletal functions essential for MPhi activity. Our data revealed that EdTx significantly suppressed human MPhi phagocytosis of Ames spores. Cytoskeletal changes, such as decreased cell spreading and lowered F-actin content, were also observed for toxin-treated MPhis. Further, EdTx altered the protein levels and activity of PKA and exchange protein activated by cAMP (Epac), a recently identified cAMP-binding molecule. By using PKA- and Epac-selective cAMP analogs, we confirmed the involvement of both pathways in the inhibition of MPhi functions elicited by EdTx-generated cAMP. These results suggested that EdTx weakened the host immune response by increasing cAMP levels, which then signaled via PKA and Epac to cripple MPhi phagocytosis and interfered with cytoskeletal remodeling.
Collapse
|
14
|
Kazawa T, Kawasaki T, Sakamoto A, Imamura M, Ohashi R, Jiang S, Tanaka T, Iwanari H, Hamakubo T, Sakai J, Kodama T, Naito M. Expression of liver X receptor α and lipid metabolism in granulocyte-macrophage colony-stimulating factor-induced human monocyte-derived macrophage. Pathol Int 2009; 59:152-60. [DOI: 10.1111/j.1440-1827.2009.02343.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
15
|
Global effects of inorganic arsenic on gene expression profile in human macrophages. Mol Immunol 2009; 46:649-56. [DOI: 10.1016/j.molimm.2008.08.268] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 07/30/2008] [Accepted: 08/12/2008] [Indexed: 11/19/2022]
|
16
|
Monick MM, Powers LS, Barrett CW, Hinde S, Ashare A, Groskreutz DJ, Nyunoya T, Coleman M, Spitz DR, Hunninghake GW. Constitutive ERK MAPK activity regulates macrophage ATP production and mitochondrial integrity. THE JOURNAL OF IMMUNOLOGY 2008; 180:7485-96. [PMID: 18490749 DOI: 10.4049/jimmunol.180.11.7485] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A unique feature of human alveolar macrophages is their prolonged survival in the face of a stressful environment. We have shown previously that the ERK MAPK is constitutively active in these cells and is important in prolonging cell survival. This study examines the role of the ERK pathway in maintaining mitochondrial energy production. The data demonstrate that ATP levels in alveolar macrophages depend on intact mitochondria and optimal functioning of the electron transport chain. Significant levels of MEK and ERK localize to the mitochondria and inhibition of ERK activity induces an early and profound depletion in cellular ATP coincident with a loss of mitochondrial transmembrane potential. The effect of ERK suppression on ATP levels was specific, since it did not occur with PI3K/Akt, p38, or JNK suppression. ERK inhibition led to cytosolic release of mitochondrial proteins and caspase activation. Both ERK inhibition and mitochondrial blockers induced loss of plasma membrane permeability and cell death. The cell death induced by ERK inhibition had hallmarks of both apoptotic (caspase activation) and necrotic (ATP loss) cell death. By blocking ERK inhibition-induced reactive oxygen species, caspase activation was prevented, although necrotic pathways continued to induce cell death. This suggests that mitochondrial dysfunction caused by ERK inhibition generates both apoptotic and necrotic cell death-inducing pathways. As a composite, these data demonstrate a novel mitochondrial role for ERK in maintaining mitochondrial membrane potential and ATP production in human alveolar macrophages.
Collapse
Affiliation(s)
- Martha M Monick
- Department of Medicine, University of Iowa Carver College of Medicine and Veterans Administration Medical Center, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Yokota S, Ohara N, Kobayashi T. The effects of organic extract of diesel exhaust particles on ischemia/reperfusion-related arrhythmia and on pulmonary inflammation. J Toxicol Sci 2008; 33:1-10. [PMID: 18303179 DOI: 10.2131/jts.33.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since our previous study demonstrated the exacerbation of acute myocardial ischemia/reperfusion (AMIR)-related arrhythmia by intratracheal instillation (IT) of diesel exhaust particles (DEP), the influence of IT with extracts of DEP in organic solvents on AMIR-related arrhythmia was examined in rats. Oxidative activity in a non-biological assay system and proinflammatory activity in mice of DEP extracts were examined. The dichloromethane-soluble fraction (DMSF) of DEP was further fractionated into n-hexane-soluble (n-HSF) and n-hexane-insoluble (n-HISF) fractions. The oxidative activities of the fractions evaluated by dithiothreitol assay were ranked as follows: n-HISF>DMSF>n-HSF. Twenty-one to 34 hr after IT, the AMIR experiment was performed. Exacerbation of AMIR-related arrhythmia and increased reperfusion-related mortality were observed only in rats treated with DMSF. In fact, n-HSF and n-HISF did not affect arrhythmia up to 5 mg/kg. Twelve hr after IT, a significant increase in neutrophil count was observed only with DMSF. The levels of granulocyte colony-stimulating factor and interleukin-6 in bronchoalveolar lavage fluid were significantly elevated in the group treated with DMSF, while neither, n-HSF nor n-HISF, affected the level of cytokines up to 5 mg/kg. In fact, tumor necrosis factor-alpha, IL-10 and granulocyte-macrophage colony-stimulating factor were unchanged with any of the fractions. In conclusion, exacerbation of AMIR-related arrhythmia by DMSF suggests the contribution of non-particle components of DEP to arrhythmia while the component contributed to the effects did not become clear. Furthermore, it is confirmed that exacerbation of AMIR-related arrhythmia is accompanied by an increased neutrophil count in the circulatory blood.
Collapse
Affiliation(s)
- Syunji Yokota
- Laboratory of Toxicology, Hatano Research Institute, Food and Drug Safety Center, Hadano, Kanagawa, Japan.
| | | | | |
Collapse
|
18
|
Hervé-Grépinet V, Veillard F, Godat E, Heuzé-Vourc'h N, Lecaille F, Lalmanach G. Extracellular catalase activity protects cysteine cathepsins from inactivation by hydrogen peroxide. FEBS Lett 2008; 582:1307-12. [PMID: 18343226 DOI: 10.1016/j.febslet.2008.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 02/08/2008] [Accepted: 03/05/2008] [Indexed: 11/15/2022]
Abstract
The resistance of secreted cysteine cathepsins to peroxide inactivation was evaluated using as model THP-1 cells. Differentiated cells released mostly cathepsin B, but also cathepsins H, K, and L, with a maximum of endopeptidase activity at day 6. Addition of non-cytotoxic concentrations of H(2)O(2) did not affect mRNA expression levels and activity of cathepsins, while the catalase activity remained also unchanged, consistently with RT-PCR analysis. Conversely inhibition of extracellular catalase led to a striking inactivation of secreted cysteine cathepsins by H(2)O(2). This report suggests that catalase may participate in the protection of extracellular cysteine proteases against peroxidation.
Collapse
Affiliation(s)
- Virginie Hervé-Grépinet
- INSERM U 618, Protéases et Vectorisation Pulmonaires, Equipe Protéases et Pathologies Pulmonaires, Tours F-37000, France
| | | | | | | | | | | |
Collapse
|
19
|
Tephly LA, Carter AB. Constitutive NADPH oxidase and increased mitochondrial respiratory chain activity regulate chemokine gene expression. Am J Physiol Lung Cell Mol Physiol 2007; 293:L1143-55. [PMID: 17704189 DOI: 10.1152/ajplung.00114.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alveolar macrophages, which generate high levels of reactive oxygen species, especially O(2)(*-), are involved in the recruitment of neutrophils to sites of inflammation and injury in the lung, and the generation of chemotactic proteins triggers this cellular recruitment. In this study, we asked whether O(2)(*-) generation in alveolar macrophages had a role in the expression of chemokines. Specifically, we hypothesized that O(2)(*-) generation is necessary for chemokine expression in alveolar macrophages after TNF-alpha stimulation. We found that alveolar macrophages have high constitutive NADPH oxidase activity that was not increased by TNF-alpha, but TNF-alpha increased the activity of the mitochondrial respiratory chain. In addition, the mitochondrial respiratory chain increased O(2)(*-) generation if the NADPH oxidase was inhibited. O(2)(*-) generation was necessary for macrophage inflammatory protein-2 (MIP-2) gene expression, because inhibition of NADPH oxidase or the mitochondrial respiratory chain or overexpression of Cu,Zn-superoxide dismutase significantly inhibited expression of MIP-2. TNF-alpha activated the ERK MAP kinase, and ERK activity was essential for chemokine gene expression. In addition, overexpression of the MEK1-->ERK pathway significantly increased IL-8 expression, and a small interfering RNA to the NADPH oxidase inhibited ERK- and TNF-alpha-induced chemokine expression. Collectively, these results suggest that in alveolar macrophages, O(2)(*-) generation mediates chemokine expression after TNF-alpha stimulation in an ERK-dependent manner.
Collapse
Affiliation(s)
- Linda A Tephly
- Division of Pulmonary, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | | |
Collapse
|
20
|
Ohgimoto K, Ohgimoto S, Ihara T, Mizuta H, Ishido S, Ayata M, Ogura H, Hotta H. Difference in production of infectious wild-type measles and vaccine viruses in monocyte-derived dendritic cells. Virus Res 2006; 123:1-8. [PMID: 16959355 DOI: 10.1016/j.virusres.2006.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2006] [Revised: 07/01/2006] [Accepted: 07/12/2006] [Indexed: 12/21/2022]
Abstract
Macrophages (Mø) and dendritic cells (DC) are thought to be targets of measles virus (MeV) at the early stage of infection. We compared the growth of Edmonston-derived vaccine strains and fresh clinical isolates of MeV in monocytes, monocyte-derived granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced Mø (GM-Mø) and in monocyte-derived DC (Mo-DC). Neither vaccine strains nor fresh isolates thrived in monocytes and GM-Mø and no differences were evident among them. On the other hand, infectious virus production was robust in Mo-DC infected with fresh isolates, but below the limits of detection in those infected with vaccine strains. Although the vaccine strains infected Mo-DC and replicated comparably with the fresh isolates, they accumulated far less matrix (M) protein. This was attributed to a difference in the stability of M protein produced in Mo-DC between the strains. Impaired production of infectious viruses in DC may be one cause of vaccine strain attenuation.
Collapse
Affiliation(s)
- Kaori Ohgimoto
- Division of Microbiology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Monick MM, Powers LS, Gross TJ, Flaherty DM, Barrett CW, Hunninghake GW. Active ERK contributes to protein translation by preventing JNK-dependent inhibition of protein phosphatase 1. THE JOURNAL OF IMMUNOLOGY 2006; 177:1636-45. [PMID: 16849472 DOI: 10.4049/jimmunol.177.3.1636] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human alveolar macrophages, central to immune responses in the lung, are unique in that they have an extended life span in contrast to precursor monocytes. We have shown previously that the ERK MAPK (ERK) pathway is constitutively active in human alveolar macrophages and contributes to the prolonged survival of these cells. We hypothesized that ERK maintains survival, in part, by positively regulating protein translation. In support of this hypothesis, we have found novel links among ERK, JNK, protein phosphatase 1 (PP1), and the eukaryotic initiation factor (eIF) 2alpha. eIF2alpha is active when hypophosphorylated and is essential for initiation of protein translation (delivery of initiator tRNA charged with methionine to the ribosome). Using [(35)S]methionine labeling, we found that ERK inhibition significantly decreased protein translation rates in alveolar macrophages. Decreased protein translation resulted from phosphorylation (and inactivation) of eIF2alpha. We found that ERK inhibition increased JNK activity. JNK in turn inactivated (via phosphorylation) PP1, the phosphatase responsible for maintaining the hypophosphorylated state of eIF2alpha. As a composite, our data demonstrate that in human alveolar macrophages, constitutive ERK activity positively regulates protein translation via the following novel pathway: active ERK inhibits JNK, leading to activation of PP1alpha, eIF2alpha dephosphorylation, and translation initiation. This new role for ERK in alveolar macrophage homeostasis may help to explain the survival characteristic of these cells within their unique high oxygen and stress microenvironment.
Collapse
Affiliation(s)
- Martha M Monick
- University of Iowa Carver College of Medicine, Veterans Administration Medical Center, Iowa City, IA 52242, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Väänänen AJ, Salmenperä P, Hukkanen M, Rauhala P, Kankuri E. Cathepsin B is a differentiation-resistant target for nitroxyl (HNO) in THP-1 monocyte/macrophages. Free Radic Biol Med 2006; 41:120-31. [PMID: 16781460 DOI: 10.1016/j.freeradbiomed.2006.03.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Revised: 02/08/2006] [Accepted: 03/21/2006] [Indexed: 12/31/2022]
Abstract
We previously showed that the one-electron reduction product of nitric oxide (NO), nitroxyl (HNO), irreversibly inhibits the proteolytic activity of the model cysteine protease papain. This result led us to investigate the differential effects of the nitrogen oxides, such as nitroxyl (HNO), NO, and in situ-generated peroxynitrite on cysteine modification-sensitive cellular proteolytic enzymes. We used Angeli's salt, diethylaminenonoate (DEA/NO), and 3-morpholinosydnoniminehydrochloride (SIN-1), as donors of HNO, NO, and peroxynitrite, respectively. In this study we evaluated their inhibitory activities on the lysosomal mammalian papain homologue cathepsin B and on the cytosolic 26S proteasome in THP-1 monocyte/macrophages after LPS activation or TPA differentiation. HNO-generating Angeli's salt caused a concentration-dependent (62 +/- 4% at 316 muM) inhibition of the 26S proteasome activity, resulting in accumulation of protein-bound polyubiquitinylated proteins in LPS-activated cells, whereas neither DEA/NO nor SIN-1 showed any effect. Angeli's salt, but not DEA/NO or SIN-1, also caused (94 +/- 2% at 316 muM) inhibition of lysosomal cathepsin B activity in LPS-activated cells. Induction of macrophage differentiation did not significantly alter the inhibitory effect of HNO on lysosomal cathepsin B activity, but protected the proteasome from HNO-induced inhibition. The protection awarded by macrophage differentiation was associated with induction of the GSH synthesis rate-limiting enzyme gamma-glutamylcysteine synthetase, as well as with increased intracellular GSH. In conclusion, HNO abrogates both lysosomal and cytosolic proteolysis in THP-1 cells. Macrophage differentiation, associated with upregulation of antioxidant defenses such as increased cellular GSH, does not protect the lysosomal cysteine protease cathepsin B from inhibition.
Collapse
Affiliation(s)
- Antti J Väänänen
- Department of Pharmacology, Institute of Biomedicine, Biomedicum Helsinki, P.O. Box 63, University of Helsinki 00014, Finland
| | | | | | | | | |
Collapse
|
23
|
Akagawa KS, Komuro I, Kanazawa H, Yamazaki T, Mochida K, Kishi F. Functional heterogeneity of colony-stimulating factor-induced human monocyte-derived macrophages. Respirology 2006; 11 Suppl:S32-6. [PMID: 16423268 DOI: 10.1111/j.1440-1843.2006.00805.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Macrophages (Mphis) have various functions and play a critical role in host defense and the maintenance of homeostasis. Mphis exist in every tissue in the body, but Mphis from different tissues exhibit a wide range of phenotypes with regard to their morphology, cell surface antigen expression and function, and are called by different names. However, the precise mechanism of the generation of macrophage heterogeneity is not known. In the present study, the authors examined the functional heterogeneity of Mphis generated from human monocytes under the influence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and macrophage-CSF (M-CSF). METHODOLOGY CD14 positive human monocytes (Mos) were incubated with M-CSF and GM-CSF for 6-7 days to stimulate the generation of M-CSF-induced monocyte-derived Mphis (M-Mphis) and GM-CSF-induced monocyte-derived Mphis (GM-Mphis), respectively. The expression of cell surface antigens and several functions such as antigen presenting cell activity, susceptibility to oxidant stress, and the susceptibility to HIV-1 and mycobacterium tuberculosis infection were examined. RESULTS GM-Mphis and M-Mphis are distinct in their morphology, cell surface antigen expression, and functions examined. The phenotype of GM-Mphis closely resembles that of human Alveolar-Mphis (A-Mphis), indicating that CSF-induced human monocyte-derived Mphis are useful to clarify the molecular mechanism of heterogeneity of human Mphis, and GM-Mphis will become a model of human A-Mphis.
Collapse
Affiliation(s)
- Kiyoko S Akagawa
- Department of Immunology, National Institute of Infectious Diseases, Toyama, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
24
|
Yoshioka Y, Kitao T, Kishino T, Yamamuro A, Maeda S. Nitric oxide protects macrophages from hydrogen peroxide-induced apoptosis by inducing the formation of catalase. THE JOURNAL OF IMMUNOLOGY 2006; 176:4675-81. [PMID: 16585560 DOI: 10.4049/jimmunol.176.8.4675] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We investigated the cytoprotective effect of NO on H2O2-induced cell death in mouse macrophage-like cell line RAW264. H2O2-treated cells showed apoptotic features, such as activation of caspase-9 and caspase-3, nuclear fragmentation, and DNA fragmentation. These apoptotic features were significantly inhibited by pretreatment for 24 h with NO donors, sodium nitroprusside and 1-hydroxy-2-oxo-3,3-bis-(2-aminoethyl)-1-triazene, at a low nontoxic concentration. The cytoprotective effect of NO was abrogated by the catalase inhibitor 3-amino-1,2,4-triazole but was not affected by a glutathione synthesis inhibitor, L-buthionine-(S,R)-sulfoximine. NO donors increased the level of catalase and its activity in a concentration-dependent manner. Cycloheximide, a protein synthesis inhibitor, inhibited both the NO-induced increase in the catalase level and the cytoprotective effect of NO. These results indicate that NO at a low concentration protects macrophages from H2O2-induced apoptosis by inducing the production of catalase.
Collapse
Affiliation(s)
- Yasuhiro Yoshioka
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, Japan.
| | | | | | | | | |
Collapse
|
25
|
Sakurai T, Ohta T, Tomita N, Kojima C, Hariya Y, Mizukami A, Fujiwara K. Evaluation of immunotoxic and immunodisruptive effects of inorganic arsenite on human monocytes/macrophages. Int Immunopharmacol 2006; 6:304-15. [PMID: 16459422 DOI: 10.1016/j.intimp.2005.06.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A trivalent inorganic arsenic, arsenite, has been causing chronic inflammation in humans through the consumption of contaminated well water. The total peripheral blood arsenic concentrations of chronic arsenic-exposed patients, who had inflammatory-like immune responses, are less than 1 microM, thus, nM concentrations may be very important regarding the chronic inflammatory effects by arsenite. However, there are few reports about the biological effects of low concentrations of arsenite in mammalian cells, especially in normal immune effector cells. In this study, we examined whether arsenite has any biological and/or toxicological effects on the differentiation of human peripheral blood monocytes into macrophages using the colony-stimulating factor (CSF) in vitro compared with that of other metallic compounds, and found that arsenite sensitively inhibited the CSF-induced in vitro maturation of monocytes into macrophages at nM levels, and it also induced small, nonadhesive and CD14-positive abnormal macrophage generation from monocytes with granulocyte-macrophage CSF (GM-CSF) at 50-500 nM without cell death. The addition of other metallic compounds, including chromium, selenium, mercury, cadmium, nickel, copper, zinc, cobalt, manganese and other human pentavalent arsenic metabolites, such as inorganic arsenate, monomethylarsonic acid and dimethylarsinic acid, could not induce the same abnormal cell generation from monocytes with CSFs at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at n-mM levels accompanied by cell death. This work may have implications in the arsenic-induced chronic inflammation in humans.
Collapse
Affiliation(s)
- Teruaki Sakurai
- Laboratory of Environmental Chemistry, School of Life Science, Tokyo University of Pharmacy and Life Science, Horinouchi 1432-1, Hachioji, Tokyo 192-0392, Japan.
| | | | | | | | | | | | | |
Collapse
|
26
|
Yasui K, Kobayashi N, Yamazaki T, Agematsu K, Matsuzaki S, Ito S, Nakata S, Baba A, Koike K. Superoxide dismutase (SOD) as a potential inhibitory mediator of inflammation via neutrophil apoptosis. Free Radic Res 2005; 39:755-62. [PMID: 16036355 DOI: 10.1080/10715760500104066] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Superoxide dismutase (SOD) is supposed to be an effective agent for neutrophil-mediated inflammation in the area of critical medicine. We investigated the involvement of SOD in the regulation of neutrophil apoptosis. Exogenously added SOD effectively induced neutrophil apoptosis, and the fluorescence patterns determined using annexin-V and the 7-AAD were similar to those seen in Fas-mediated neutrophil apoptosis. Neutrophils are short-lived leukocytes that need to be removed safely by apoptosis. The clearance of apoptotic neutrophils from sites of inflammation is a crucial determinant of the resolution of inflammation. Catalase inhibited the neutrophil apoptosis and caspase-3 activation. Spontaneous apoptosis, hydrogen peroxide and anti-Fas antibody-induced apoptosis of neutrophils were accelerated in Down's syndrome patients, in whom the SOD gene is overexpressed. Hydrogen peroxide was thought to be a possible major mediator of ROS-induced neutrophil apoptosis in caspase-dependent manner. Neutrophil apoptosis represents a crucial step in the mechanism governing the resolution of inflammation and has been suggested as a possible target for the control of neutrophil-mediated tissue injury. SOD may be a potential inhibitory mediator of neutrophil-mediated inflammation.
Collapse
Affiliation(s)
- Kozo Yasui
- Department of Pediatrics, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto, 390-8621, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Komuro I, Yasuda T, Iwamoto A, Akagawa KS. Catalase plays a critical role in the CSF-independent survival of human macrophages via regulation of the expression of BCL-2 family. J Biol Chem 2005; 280:41137-45. [PMID: 16204228 DOI: 10.1074/jbc.m509793200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
M-colony-stimulating factor (M-CSF)-induced monocyte-derived macrophages (M-Mphi) required continuous presence of M-CSF for their survival, and depletion of M-CSF from the culture induced apoptosis, whereas human alveolar macrophages (A-Mphi) and granulocyte-macrophage (GM)-CSF-induced monocyte-derived macrophages (GM-Mphi) survived even in the absence of CSF. The expression of BCL-2 was higher in M-Mphi, and M-CSF withdrawal down-regulated the expression. The expression of BCL-X(L) was higher in A-Mphi and GM-Mphi, and the expression was CSF-independent. The expression of MCL-1 and BAX were not different between M-Mphi and GM-Mphi and were CSF-independent. Down-regulation of the expression of BCL-2 and BCL-X(L) by RNA interference showed the important role of BCL-2 and BCL-X(L) in the survival of M-Mphi and GM-Mphi, respectively. Human erythrocyte catalase (HEC) and conditioned medium obtained from GM-Mphi or A-Mphi cultured in the absence of GM-CSF prevented the M-Mphi from apoptosis and restored the expression of BCL-2. The activity of the conditioned medium was abrogated by pretreatment with anti-HEC antibody. Anti-HEC antibody also induced the apoptosis of M-Mphi cultured in the presence of M-CSF and GM-Mphi and A-Mphi cultured in the presence or absence of GM-CSF and down-regulated the expression of BCL-2 and BCL-X(L) in these Mphis. GM-Mphi and A-Mphi, but not M-Mphi, can produce both extracellular catalase and cell-associated catalase in a CSF-independent manner. Intracellular glutathione levels were kept equivalent in these Mphis, both in the presence or absence of CSF. These results indicate a critical role of extracellular catalase in the survival of human macrophages via regulation of the expression of BCL-2 family genes.
Collapse
Affiliation(s)
- Iwao Komuro
- Department of Immunology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | |
Collapse
|
28
|
Sakurai T, Ohta T, Tomita N, Kojima C, Hariya Y, Mizukami A, Fujiwara K. Evaluation of immunotoxic and immunodisruptive effects of inorganic arsenite on human monocytes/macrophages. Int Immunopharmacol 2005; 4:1661-73. [PMID: 15454118 DOI: 10.1016/j.intimp.2004.07.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Revised: 06/23/2004] [Accepted: 07/26/2004] [Indexed: 10/26/2022]
Abstract
A trivalent inorganic arsenic, arsenite, has been causing chronic inflammation in humans through the consumption of contaminated well water. The total peripheral blood arsenic concentrations of chronic arsenic-exposed patients, who had inflammatory-like immune responses, are less than 1 microM, thus, nM concentrations may be very important regarding the chronic inflammatory effects by arsenite. However, there are few reports about the biological effects of low concentrations of arsenite in mammalian cells, especially in normal immune effector cells. In this study, we examined whether arsenite has any biological and/or toxicological effects on the differentiation of human peripheral blood monocytes into macrophages using the colony-stimulating factor (CSF) in vitro compared with that of other metallic compounds, and found that arsenite sensitively inhibited the CSF-induced in vitro maturation of monocytes into macrophages at nM levels, and it also induced small, nonadhesive and CD14-positive abnormal macrophage generation from monocytes with granulocyte-macrophage CSF (GM-CSF) at 50-500 nM without cell death. The addition of other metallic compounds, including chromium, selenium, mercury, cadmium, nickel, copper, zinc, cobalt, manganese and other human pentavalent arsenic metabolites, such as inorganic arsenate, monomethylarsonic acid and dimethylarsinic acid, could not induce the same abnormal cell generation from monocytes with CSFs at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at n-mM levels accompanied by cell death. This work may have implications in the arsenic-induced chronic inflammation in humans.
Collapse
Affiliation(s)
- Teruaki Sakurai
- Laboratory of Environmental Chemistry, School of Life Science, Tokyo University of Pharmacy and Life Science, Horinouchi 1432-1, Hachioji, Tokyo 192-0392, Japan.
| | | | | | | | | | | | | |
Collapse
|
29
|
Kawamura K, Iyonaga K, Ichiyasu H, Nagano J, Suga M, Sasaki Y. Differentiation, maturation, and survival of dendritic cells by osteopontin regulation. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2005; 12:206-12. [PMID: 15643009 PMCID: PMC540203 DOI: 10.1128/cdli.12.1.206-212.2005] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dendritic cells (DCs) are antigen-presenting cells with the ability to induce primary immune responses necessary in innate immunity and adaptive immunity. Osteopontin (OPN) is a secreted acidic phosphoprotein containing an arginine-glycine-aspartate sequence and has been suggested to play an important role in early cellular immune responses. The interaction between DCs and OPN has not been clarified. We hypothesized that there is an important interaction between DCs and OPN, which is an indispensable extracellular matrix component in early cellular immune responses. Human monocyte-derived DCs synthesized OPN especially during the differentiation from monocytes to immature DCs. By blocking of OPN with anti-OPN antibody, cultured DCs became smaller and expressed lower levels of costimulatory molecules and major histocompatibility complex class II antigens than untreated DCs. Furthermore, DCs treated with anti-OPN antibody easily underwent apoptosis. These results suggest that human DCs can produce OPN and that OPN may play a role in the differentiation, maturation, and survival of DCs by autocrine and/or paracrine pathways.
Collapse
Affiliation(s)
- Kodai Kawamura
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Sakurai T, Ohta T, Fujiwara K. Inorganic arsenite alters macrophage generation from human peripheral blood monocytes. Toxicol Appl Pharmacol 2005; 203:145-53. [PMID: 15710175 DOI: 10.1016/j.taap.2004.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2004] [Accepted: 08/09/2004] [Indexed: 10/26/2022]
Abstract
Inorganic arsenite has caused severe inflammatory chronic poisoning in humans through the consumption of contaminated well water. In this study, we examined the effects of arsenite at nanomolar concentrations on the in vitro differentiation of human macrophages from peripheral blood monocytes. While arsenite was found to induce cell death in a culture system containing macrophage colony stimulating factor (M-CSF), macrophages induced by granulocyte-macrophage CSF (GM-CSF) survived the treatment, but were morphologically, phenotypically, and functionally altered. In particular, arsenite-induced cells expressed higher levels of a major histocompatibility complex (MHC) class II antigen, HLA-DR, and CD14. They were more effective at inducing allogeneic or autologous T cell responses and responded more strongly to bacterial lipopolysaccharide (LPS) by inflammatory cytokine release as compared to cells induced by GM-CSF alone. On the other hand, arsenite-induced cells expressed lower levels of CD11b and CD54 and phagocytosed latex beads or zymosan particles less efficiently. We also demonstrated that the optimum amount of cellular reactive oxygen species (ROS) induced by nM arsenite might play an important role in this abnormal monocyte differentiation. This work may have implications in chronic arsenic poisoning because the total peripheral blood arsenic concentrations of these patients are at nM levels.
Collapse
Affiliation(s)
- Teruaki Sakurai
- Laboratory of Environmental Chemistry, School of Life Science, Tokyo University of Pharmacy and Life Science, Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | | | | |
Collapse
|
31
|
Hultén LM, Ullström C, Krettek A, van Reyk D, Marklund SL, Dahlgren C, Wiklund O. Human macrophages limit oxidation products in low density lipoprotein. Lipids Health Dis 2005; 4:6. [PMID: 15745457 PMCID: PMC555960 DOI: 10.1186/1476-511x-4-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Accepted: 03/04/2005] [Indexed: 01/24/2023] Open
Abstract
This study tested the hypothesis that human macrophages have the ability to modify oxidation products in LDL and oxidized LDL (oxLDL) via a cellular antioxidant defence system. While many studies have focused on macrophage LDL oxidation in atherosclerosis development, less attention has been given to the cellular antioxidant capacity of these cells. Compared to cell-free controls (6.2 ± 0.7 nmol/mg LDL), macrophages reduced TBARS to 4.42 ± 0.4 nmol/mg LDL after 24 h incubation with LDL (P = 0.022). After 2 h incubation with oxLDL, TBARS were 3.69 ± 0.5 nmol/mg LDL in cell-free media, and 2.48 ± 0.9 nmol/mg LDL in the presence of macrophages (P = 0.034). A reduction of lipid peroxides in LDL (33.7 ± 6.6 nmol/mg LDL) was found in the presence of cells after 24 h compared to cell-free incubation (105.0 ± 14.1 nmol/mg LDL) (P = 0.005). The levels of lipid peroxides in oxLDL were 137.9 ± 59.9 nmol/mg LDL and in cell-free media 242 ± 60.0 nmol/mg LDL (P = 0.012). Similar results were obtained for hydrogen peroxide. Reactive oxygen species were detected in LDL, acetylated LDL, and oxLDL by isoluminol-enhanced chemiluminescence (CL). Interestingly, oxLDL alone gives a high CL signal. Macrophages reduced the CL response in oxLDL by 45% (P = 0.0016). The increased levels of glutathione in oxLDL-treated macrophages were accompanied by enhanced catalase and glutathione peroxidase activities. Our results suggest that macrophages respond to oxidative stress by endogenous antioxidant activity, which is sufficient to decrease reactive oxygen species both in LDL and oxLDL. This may suggest that the antioxidant activity is insufficient during atherosclerosis development. Thus, macrophages may play a dual role in atherogenesis, i.e. both by promoting and limiting LDL-oxidation.
Collapse
Affiliation(s)
- Lillemor Mattsson Hultén
- Wallenberg Laboratory for Cardiovascular Research, Sahlgrenska University Hospital, SE-413 45 Göteborg, Sweden
| | - Christina Ullström
- Wallenberg Laboratory for Cardiovascular Research, Sahlgrenska University Hospital, SE-413 45 Göteborg, Sweden
| | - Alexandra Krettek
- Wallenberg Laboratory for Cardiovascular Research, Sahlgrenska University Hospital, SE-413 45 Göteborg, Sweden
| | - David van Reyk
- Department of Health Sciences, University of Technology, Sydney, N.S.W. 2007, Australia
| | - Stefan L Marklund
- Medical Biosciences, Clinical Chemistry, Umeå University Hospital, SE-901 85 Umeå, Sweden
| | - Claes Dahlgren
- Phagocyte Research Laboratory, Department of Rheumatology and Inflammation Research, University of Göteborg, SE-413 46 Göteborg, Sweden
| | - Olov Wiklund
- Wallenberg Laboratory for Cardiovascular Research, Sahlgrenska University Hospital, SE-413 45 Göteborg, Sweden
| |
Collapse
|
32
|
Pelletier M, Bouchard A, Girard D. In vivo and in vitro roles of IL-21 in inflammation. THE JOURNAL OF IMMUNOLOGY 2005; 173:7521-30. [PMID: 15585879 DOI: 10.4049/jimmunol.173.12.7521] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-21 is a cytokine known to mediate its biological action via the IL-21R, composed of a specific chain, IL-21Ralpha, and the common gamma-chain (CD132). Recent data suggest that IL-21 possesses proinflammatory properties. However, there is no clear evidence that IL-21 induces inflammation in vivo and, curiously, the interaction between IL-21 and neutrophils has never been investigated, despite the fact that these cells express CD132 and respond to other CD132-dependent cytokines involved in inflammatory disorders. Using the murine air pouch model, we found that IL-21 induced inflammation in vivo, based on recruitment of neutrophil and monocyte populations. In contrast to LPS, administration of IL-21 into the air pouch did not significantly increase the concentration of IL-6, CCL5, CCL3, and CXCL2. We demonstrated that HL-60 cells expressed IL-21Ralpha, which is down-regulated during their differentiation toward neutrophils, and that IL-21Ralpha is not detected in neutrophils. Concomitant with this, IL-21 induced Erk-1/2 phosphorylation in HL-60 cells, but not in neutrophils. To eliminate the possibility that IL-21 could activate neutrophils even in the absence of IL-21Ralpha, we demonstrated that IL-21 did not modulate several neutrophil functions. IL-21-induced Erk-1/2 phosphorylation was not associated with proliferation or differentiation of HL-60 toward neutrophils, monocytes, or macrophages. IL-21Ralpha was detected in human monocytes and monocyte-derived macrophages, but IL-21 increased CXCL8 production only in monocyte-derived macrophages. We conclude that IL-21 is a proinflammatory cytokine, but not a neutrophil agonist. We propose that IL-21 attracts neutrophils indirectly in vivo via a mechanism independent of IL-6, CCL3, CCL5, and CXCL2 production.
Collapse
Affiliation(s)
- Martin Pelletier
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier, Université du Québec, Pointe-Claire, Canada
| | | | | |
Collapse
|
33
|
Abstract
HIV-1, like the other lentiviruses, has evolved the ability to infect nondividing cells including macrophages. HIV-1 replication in monocytes/macrophages entails peculiar features and differs in many respects from that in CD4 T lymphocytes. HIV-1 exhibits different tropism for CD4 T cells and macrophages. The virus can enter macrophages via several routes. Mitosis is not required for nuclear import of viral DNA or for its integration into the host cell genome. Specific cellular factors are required for HIV-1 transcription in macrophages. The assembly and budding of viral particles in macrophages take place in late endosomal compartments. Viral particles can use the exosome pathway to exit cells. Given their functions in host defence against pathogens and the regulation of the immune response plus their permissivity to HIV-1 infection, monocytes/macrophages exert a dual role in HIV infection. They contribute to the establishment and persistence of HIV-1 infection, and may activate surrounding T cells favouring their infection. Furthermore, monocytes/macrophages act as a Trojan horse to transmit HIV-1 to the central nervous system. They also exhibit antiviral activity and express many molecules that inhibit HIV-1 replication. Activated microglia and macrophages may also exert a neurotrophic and neuroprotective effect on infected brain regulating glutamate metabolism or by secretion of neurotrophins. This review will discuss specific aspects of viral replication in monocytes/macrophages and the role of their interactions with the cellular environment in HIV-1 infection swinging between protection and pathogenesis.
Collapse
Affiliation(s)
- Alessia Verani
- Human Virology Unit, DIBIT, San Raffaele Scientific Institute, Milan, Italy
| | | | | |
Collapse
|
34
|
Shoda E, Kawano E, Aoki K, Akagawa KS. Capacity of Monocytes from Patients with Chronic Myelomonocytic Leukemia to Differentiate into Macrophages and Multinucleated Giant Cells. J Clin Exp Hematop 2005. [DOI: 10.3960/jslrt.45.89] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
35
|
Monick MM, Mallampalli RK, Bradford M, McCoy D, Gross TJ, Flaherty DM, Powers LS, Cameron K, Kelly S, Merrill AH, Hunninghake GW. Cooperative prosurvival activity by ERK and Akt in human alveolar macrophages is dependent on high levels of acid ceramidase activity. THE JOURNAL OF IMMUNOLOGY 2004; 173:123-35. [PMID: 15210766 DOI: 10.4049/jimmunol.173.1.123] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human alveolar macrophages are unique in that they have an extended life span in contrast to precursor monocytes. In evaluating the role of sphingolipids in alveolar macrophage survival, we found high levels of sphingosine, but not sphingosine-1-phosphate. Sphingosine is generated by the action of ceramidase(s) on ceramide, and alveolar macrophages have high constitutive levels of acid ceramidase mRNA, protein, and activity. The high levels of acid ceramidase were specific to alveolar macrophages, because there was little ceramidase protein or activity (or sphingosine) in monocytes from matching donors. In evaluating prolonged survival of alveolar macrophages, we observed a requirement for constitutive activity of ERK MAPK and the PI3K downstream effector Akt. Blocking acid ceramidase but not sphingosine kinase activity in alveolar macrophages led to decreased ERK and Akt activity and induction of cell death. These studies suggest an important role for sphingolipids in prolonging survival of human alveolar macrophages via distinct survival pathways.
Collapse
Affiliation(s)
- Martha M Monick
- Division of Pulmonary, Critical Care, and Occupational Medicine, Roy J. and Lucille A. Carver College of Medicine, Eckstein Medical Research Building Room 100, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Carter AB, Tephly LA, Venkataraman S, Oberley LW, Zhang Y, Buettner GR, Spitz DR, Hunninghake GW. High Levels of Catalase and Glutathione Peroxidase Activity Dampen H2O2Signaling in Human Alveolar Macrophages. Am J Respir Cell Mol Biol 2004; 31:43-53. [PMID: 14962975 DOI: 10.1165/rcmb.2003-0377oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Results are presented which support the hypothesis that adequate steady-state levels of hydrogen peroxide (H2O2) are required to overcome the effects of high catalase and glutathione peroxidase (GPx) expression for p38 mitogen-activated protein (MAP) kinase activation and tumor necrosis factor (TNF)-alpha gene expression in human alveolar macrophages stimulated with asbestos. We found significant differences in the types and amounts of reactive oxygen species generated in human blood monocytes compared with human alveolar macrophages. This difference in reactive oxygen species production is related, in part, to the differences in antioxidant enzyme expression and activity. Most importantly, catalase and GPx activities were significantly increased in alveolar macrophages compared with blood monocytes. Asbestos activated the p38 MAP kinase and induced TNF-alpha gene expression only in blood monocytes. Increasing the steady-state levels of H2O2 by using polyethylene glycol superoxide dismutase, an antioxidant that crosses the cell membrane, or aminotriazole, an irreversible inhibitor of catalase, allowed the p38 MAP kinase to be activated in alveolar macrophages. In addition, asbestos-stimulated macrophages cultured with polyethylene glycol superoxide dismutase had a significant increase in gene expression mediated by the TNF-alpha promoter. These results demonstrate that high catalase and GPx activity in human alveolar macrophages limits the effectiveness of H2O2 to act as a mediator of inflammatory gene expression.
Collapse
MESH Headings
- Adolescent
- Adult
- Amitrole/pharmacology
- Asbestos
- Catalase/metabolism
- Cells, Cultured
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/genetics
- Glutathione Peroxidase/metabolism
- Humans
- Hydrogen Peroxide/metabolism
- Lung/enzymology
- Lung/metabolism
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/enzymology
- Macrophages, Alveolar/metabolism
- Mitogen-Activated Protein Kinases/genetics
- Monocytes/drug effects
- Monocytes/enzymology
- Oxidative Stress/drug effects
- Oxidative Stress/physiology
- Pneumonia/enzymology
- Pneumonia/metabolism
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Superoxide Dismutase/metabolism
- Superoxide Dismutase/pharmacology
- Tumor Necrosis Factor-alpha/genetics
- Up-Regulation/drug effects
- Up-Regulation/physiology
- p38 Mitogen-Activated Protein Kinases
Collapse
Affiliation(s)
- A Brent Carter
- Department of Medicine, University of Iowa Roy J. and Lucille A Carver College of Medicine, Iowa City Veterans Administration medical Center, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Kumar AP, Piedrafita FJ, Reynolds WF. Peroxisome proliferator-activated receptor gamma ligands regulate myeloperoxidase expression in macrophages by an estrogen-dependent mechanism involving the -463GA promoter polymorphism. J Biol Chem 2003; 279:8300-15. [PMID: 14668325 DOI: 10.1074/jbc.m311625200] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A functional myeloperoxidase (MPO) promoter polymorphism, -463GA, has been associated with incidence or severity of inflammatory diseases, including atherosclerosis and Alzheimer's disease, and some cancers. The polymorphism is within an Alu element encoding four hexamer repeats recognized by nuclear receptors (AluRRE). Here we show that peroxisome proliferator-activated receptor gamma (PPARgamma) agonists strongly regulate MPO gene expression through the AluRRE. Opposite effects were observed in granulocyte/macrophage colony-stimulating factor (GMCSF)- versus macrophage colony-stimulating factor (MCSF)-derived macrophages (Mphi): Expression was markedly up-regulated (mean 26-fold) in MCSF-Mphi and down-regulated (34-fold) in GMCSF-Mphi. This was observed with rosiglitazone and three other PPARgamma ligands of the thiazolidinedione class, as well as the natural prostaglandin metabolite 15-deoxy-Delta(12,14) prostaglandin J(2). The selective PPARgamma antagonist, GW9662, blocked both the positive and negative effects on MPO expression. Gel retardation assays showed PPARgamma bound hexamers 3/4, and estrogen receptor-alpha bound hexamers 1/2, with -463A in hexamer 1 enhancing binding. Estrogen blocked PPARgamma effects on MPO expression, especially for the A allele. Charcoal filtration of fetal calf serum eliminated the block of PPARgamma, whereas replenishing the medium with 17beta-estradiol reinstated the block. These findings suggest a model in which estrogen receptor binds the AluRRE, preventing PPARgamma binding to the adjacent site. The positive and negative regulation by PPARgamma ligands, and the block by estrogen, was also observed in transgenic mice expressing the G and A alleles. The mouse MPO gene, which lacks the primate-specific AluRRE, was unresponsive to PPARgamma ligands, suggesting the human MPO transgenes will enhance the utility of mouse models for diseases involving MPO, such as atherosclerosis and Alzheimer's.
Collapse
MESH Headings
- Alu Elements/genetics
- Animals
- Arteriosclerosis/enzymology
- Binding Sites
- Bone Marrow Cells/enzymology
- Cells, Cultured
- Estradiol/pharmacology
- Estrogen Receptor alpha
- Estrogens/pharmacology
- Gene Expression Regulation, Enzymologic/drug effects
- Genotype
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- Humans
- Macrophage Colony-Stimulating Factor/pharmacology
- Macrophages/enzymology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Peroxidase/analysis
- Peroxidase/genetics
- Polymorphism, Genetic
- Promoter Regions, Genetic/genetics
- Prostaglandin D2/analogs & derivatives
- Prostaglandin D2/pharmacology
- RNA, Messenger/analysis
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Estrogen/metabolism
- Receptors, LDL/deficiency
- Receptors, Retinoic Acid/metabolism
- Retinoid X Receptors
- Rosiglitazone
- Thiazolidinediones/pharmacology
- Transcription Factors/agonists
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Alan P Kumar
- Sidney Kimmel Cancer Center, San Diego, California 92121, USA
| | | | | |
Collapse
|
38
|
Komuro I, Yokota Y, Yasuda S, Iwamoto A, Kagawa KS. CSF-induced and HIV-1-mediated distinct regulation of Hck and C/EBPbeta represent a heterogeneous susceptibility of monocyte-derived macrophages to M-tropic HIV-1 infection. J Exp Med 2003; 198:443-53. [PMID: 12900520 PMCID: PMC2194092 DOI: 10.1084/jem.20022018] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2002] [Revised: 06/13/2003] [Accepted: 06/13/2003] [Indexed: 11/17/2022] Open
Abstract
Granulocyte/macrophage colony-stimulating factor (GM-CSF)-induced monocyte-derived macrophages (GM-MPhi) are permissive to M-tropic HIV-1 entry, but inhibit viral replication at posttranscriptional and translational levels, whereas M-CSF-induced macrophages (M-MPhi) produce a large amount of HIV-1. M-MPhi express a high level of Hck and a large isoform of C/EBPbeta, and HIV-1 infection increases the expression of Hck but not of C/EBPbeta. GM-MPhi express a high level of C/EBPbeta and a low level of Hck, and HIV-1 infection drastically increases the expression of a short isoform of C/EBPbeta but decreases that of Hck. Treatment of M-MPhi with antisense oligonucleotide for Hck (AS-Hck) not only suppresses the expression of Hck, but also stimulates the induction of the short isoform of C/EBPbeta and inhibits the viral replication. Treatment of GM-MPhi with a moderate amount of AS-C/EBPbeta not only inhibits the expression of the small isoform of C/EBPbeta preferentially, but also stimulates the induction of Hck and stimulates the virus production at a high rate. These results suggest that CSF-induced and HIV-1-mediated distinct regulation of Hck and small isoform of C/EBPbeta represent the heterogeneous susceptibility of tissue MPhi to HIV-1 infection, and the regulation of Hck and C/EBPbeta are closely related and these two molecules affect one another.
Collapse
Affiliation(s)
- Iwao Komuro
- Department of Immunology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | |
Collapse
|
39
|
Akagawa KS. Functional heterogeneity of colony-stimulating factor-induced human monocyte-derived macrophages. Int J Hematol 2002; 76:27-34. [PMID: 12138892 DOI: 10.1007/bf02982715] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Macrophages have various functions and play a critical role in host defense and the maintenance of homeostasis. However, macrophages are heterogeneous and exhibit a wide range of phenotypes with regard to their morphology, cell surface antigen expression, and function. When blood monocytes are cultured in medium alone in vitro, monocytes die, and colony-stimulating factors (CSFs) such as macrophage (M)-CSF or granulocyte-macrophage (GM)-CSF are necessary for their survival and differentiation into macrophages. However, M-CSF-induced monocyte-derived macrophages (M-Mphi) and GM-CSF-induced monocyte-derived macrophages (GM-Mphi) are distinct in their morphology, cell surface antigen expression, and functions, including Fcgamma receptor mediated-phagocytosis, H2O2 production, H2O2 sensitivity, catalase activity, susceptibility to human immunodeficiency virus type 1 and Mycobacterium tuberculosis, and suppressor activity. The characteristics of GM-Mphi resemble those of human alveolar macrophages.
Collapse
Affiliation(s)
- Kiyoko S Akagawa
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|