1
|
Wang X, Liu C, Liang R, Zhou Y, Kong X, Wang W, Wang H, Zhao L, Niu W, Yi C, Jiang F. Elucidating the beneficial impact of exercise on chronic obstructive pulmonary disease and its comorbidities: Integrating proteomic and immunological insights. Br J Pharmacol 2024; 181:5133-5150. [PMID: 39317434 DOI: 10.1111/bph.17328] [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: 04/04/2024] [Revised: 07/02/2024] [Accepted: 07/26/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND AND PURPOSE Physical activity is an effective therapeutic protocol for treating chronic obstructive pulmonary disease (COPD). However, the mechanisms underlying the benefits of physical activity in COPD are not fully elucidated. EXPERIMENTAL APPROACH In a mouse model of COPD, analysis of biological markers and lung proteomics identified the molecular pathways through which exercise ameliorates COPD. KEY RESULTS Exercise improved pulmonary function, emphysema, small airway disease, pulmonary inflammation, glucose metabolic dysregulation, and insulin resistance in COPD mice. Proteomic analysis revealed 430 differentially expressed proteins (DEPs) between the COPD and COPD + Exercise (COPD + Ex) groups. GO analysis indicated that the enriched pathways were predominantly related to the immune response, inflammatory processes, insulin secretion, and glucose metabolic processes. GO analysis revealed IL-33 as a crucial target for the exercise-related amelioration of COPD. KEGG analysis showed that DEPs were significantly enriched in primary immunodeficiency, the intestinal immune network for IgA production, and the NF-κB signalling pathway. Exercise inhibited NF-κB activation by suppressing the CD14/TLR4/MyD88 and TNF-α/TNF-R1/TRAF2/5 pathways in COPD mice. Exercise inhibited expression of BCR, IgM, IgD, IgG, IgE, and IgA by suppressing B-cell receptor signalling. Exercise attenuated glucose metabolic dysregulation and insulin resistance through the suppression of proinflammatory mediators, including MHC I, MHC II, TNF-α, IFN-γ, and IL-1β, while concurrently increasing insulin expression. The qRT-PCR results were consistent with the proteomic results. CONCLUSION AND IMPLICATIONS In a mouse model, exercise improved COPD and its metabolic comorbidities through immune system regulation and inflammation suppression, offering insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Xishuai Wang
- School of Physical Education and Sports Science, Qufu Normal University, Qufu, Shandong, China
- College of Education for the Future, Beijing Normal University, Zhuhai, China
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Cong Liu
- College of Education for the Future, Beijing Normal University, Zhuhai, China
| | - Ruining Liang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yuehui Zhou
- School of Physical Education and Sports Science, Qufu Normal University, Qufu, Shandong, China
| | - Xiliang Kong
- School of Physical Education and Sports Science, Qufu Normal University, Qufu, Shandong, China
| | - Weichao Wang
- Graduate School of Sports Coaching, Kyungil University, Gyeongsan-si, Gyeongsangbuk-do, South Korea
| | - Hongwei Wang
- College of Physical Education, Northwest Normal University, Lanzhou, Gansu, China
| | - Lunan Zhao
- School of Physical Education and Sports Science, Qufu Normal University, Qufu, Shandong, China
| | - Weina Niu
- Basic Department, Qilu Institute of Technology, Qufu, Shandong, China
| | - Chao Yi
- School of Physical Education and Sports Science, Qufu Normal University, Qufu, Shandong, China
| | - Fugao Jiang
- School of Physical Education and Sports Science, Qufu Normal University, Qufu, Shandong, China
| |
Collapse
|
2
|
Kaushik S, Bhargava P, Sharma J, Arava S, Nag TC, Arya DS, Bhatia J. Sesamol attenuates bleomycin-induced pulmonary toxicity and fibrosis in experimental animals. J Biochem Mol Toxicol 2023; 37:e23472. [PMID: 37462223 DOI: 10.1002/jbt.23472] [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] [Received: 01/08/2022] [Revised: 06/29/2023] [Accepted: 07/08/2023] [Indexed: 11/10/2023]
Abstract
Sesamol, a lignan obtained from roasted seeds of Sesamum indicum, has high antioxidant and anti-inflammatory activity. In this study, we have investigated the effect of sesamol on Bleomycin (BLM) induced pulmonary toxicity as well as fibrosis in Wistar rats. Lung toxicity was induced by administration of BLM, 0.015 U/g ip, twice weekly for 28 days whereas lung fibrosis was induced by BLM, 0.015 U/g ip, every 5th day for 49 days. Sesamol administration was started 7 days before first dose of BLM in both the models. It was observed that sesamol 50 mg/kg most effectively attenuated pulmonary toxicity by reducing oxidative stress, inflammation and apoptosis. This dose was further evaluated for its anti-fibrotic effect. It was observed that there was a significant reduction in fibrosis. Lung collagen content was markedly reduced. Furthermore, expression of pro-fibrotic proteins, TGF-β/SMAD and α-SMA, was reduced and that of anti-fibrotic protein, AMPK, was markedly increased. Even though the combination of sesamol with pirfenidone exhibited no additional protection than either drug alone, it is evident from our study that our test drug, sesamol is comparable in efficacy to pirfenidone. Thus, sesamol has promising therapeutic potential in treatment of pulmonary toxicity and fibrosis.
Collapse
Affiliation(s)
- Swati Kaushik
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Poorva Bhargava
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jatin Sharma
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Sudheer Arava
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas C Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir S Arya
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
3
|
Yadav S, Shah D, Dalai P, Agrawal-Rajput R. The tale of antibiotics beyond antimicrobials: Expanding horizons. Cytokine 2023; 169:156285. [PMID: 37393846 DOI: 10.1016/j.cyto.2023.156285] [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] [Received: 02/10/2023] [Revised: 06/02/2023] [Accepted: 06/24/2023] [Indexed: 07/04/2023]
Abstract
Antibiotics had proved to be a godsend for mankind since their discovery. They were once the magical solution to the vexing problem of infection-related deaths. German scientist Paul Ehrlich had termed salvarsan as the silver bullet to treatsyphilis.As time passed, the magic of newly discovered silver bullets got tarnished with raging antibiotic resistance among bacteria and associated side-effects. Still, antibiotics remain the primary line of treatment for bacterial infections. Our understanding of their chemical and biological activities has increased immensely with advancement in the research field. Non-antibacterial effects of antibiotics are studied extensively to optimise their safer, broad-range use. These non-antibacterial effects could be both useful and harmful to us. Various researchers across the globe including our lab are studying the direct/indirect effects and molecular mechanisms behind these non-antibacterial effects of antibiotics. So, it is interesting for us to sum up the available literature. In this review, we have briefed the possible reason behind the non-antibacterial effects of antibiotics, owing to the endosymbiotic origin of host mitochondria. We further discuss the physiological and immunomodulatory effects of antibiotics. We then extend the review to discuss molecular mechanisms behind the plausible use of antibiotics as anticancer agents.
Collapse
Affiliation(s)
- Shivani Yadav
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Dhruvi Shah
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Parmeswar Dalai
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India.
| |
Collapse
|
4
|
Straß S, Geiger J, Cloos N, Späth N, Geiger S, Schwamborn A, De Oliveira da Cunha L, Martorelli M, Guse JH, Sandri TL, Burnet M, Laufer S. Immune cell targeted fumaric esters support a role of GPR109A as a primary target of monomethyl fumarate in vivo. Inflammopharmacology 2023; 31:1223-1239. [PMID: 37004600 DOI: 10.1007/s10787-023-01186-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/19/2023] [Indexed: 04/04/2023]
Abstract
Dimethyl fumarate (DMF) is approved as a treatment for multiple sclerosis (MS), however, its mode of action remains unclear. One hypothesis proposes that Michael addition to thiols by DMF, notably glutathione is immunomodulatory. The alternative proposes that monomethyl fumarate (MMF), the hydrolysis product of DMF, is a ligand to the fatty acid receptor GPR109A found in the lysosomes of immune cells. We prepared esters of MMF and macrolides derived from azithromycin, which were tropic to immune cells by virtue of lysosomal trapping. We tested the effects of these substances in an assay of response to Lipopolysaccharide (LPS) in freshly isolated human peripheral blood mononuclear cells (PBMCs). In this system, we observed that the 4'' ester of MMF (compound 2 and 3) reduced levels of Interleukins (IL)-1β, IL-12 and tumor necrosis factor alpha (TNFα) significantly at a concentration of 1 µM, while DMF required about 25 µM for the same effect. The 2' esters of MMF (compound 1 and 2) were, like MMF itself, inactive in vitro. The 4'' ester formed glutathione conjugates rapidly while the 2' conjugates did not react with thiols but did hydrolyze slowly to release MMF in these cells. We then tested the substances in vivo using the imiquimod/isostearate model of psoriasis where the 2' ester was the most active at 0.06-0.12 mg/kg (approximately 0.1 µmol/kg), improving skin score, body weight and cytokine levels (TNFα, IL-17A, IL-17F, IL-6, IL-1β, NLRP3 and IL-23A). In contrast, the thiol reactive 4'' ester was less active than the 2' ester while DMF was ca. 300-fold less active. The thiol reactive 4'' ester was not easily recovered from either plasma or organs while the 2' ester exhibited conventional uptake and elimination. The 2' ester also reduced levels of IL-6 in acute monosodium urate (MSU) induced inflammation. These data suggest that mechanisms that are relevant in vivo center on the release of MMF. Given that GPR109A is localized to the lysosome, and that lysosomal trapping increases 2' ester activity by > 300 fold, these data suggest that GPR109A may be the main target in vivo. In contrast, the effects associated with glutathione (GSH) conjugation in vitro are unlikely to be as effective in vivo due to the much lower dose in use which cannot titrate the more concentrated thiols. These data support the case for GPR109A modulation in autoimmune diseases.
Collapse
Affiliation(s)
- Simon Straß
- Pharmaceutical Chemistry, Institute for Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
- Synovo GmbH, Tübingen, Germany
| | | | | | | | | | | | | | - Mariella Martorelli
- Pharmaceutical Chemistry, Institute for Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
- Synovo GmbH, Tübingen, Germany
| | | | - Thaisa Lucas Sandri
- Synovo GmbH, Tübingen, Germany
- Institute of Tropical Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | - Stefan Laufer
- Pharmaceutical Chemistry, Institute for Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
The Azithromycin Pro-Drug CSY5669 Boosts Bacterial Killing While Attenuating Lung Inflammation Associated with Pneumonia Caused by Methicillin-Resistant Staphylococcus aureus. Antimicrob Agents Chemother 2022; 66:e0229821. [PMID: 35972289 PMCID: PMC9487537 DOI: 10.1128/aac.02298-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibiotic resistance is a major problem, with methicillin-resistant Staphylococcus aureus (MRSA) being a prototypical example in surgical and community-acquired infections. S. aureus, like many pathogens, is immune evasive and able to multiply within host immune cells. Consequently, compounds that aid host immunity (e.g., by stimulating the host-mediated killing of pathogens) are appealing alternatives or adjuncts to classical antibiotics. Azithromycin is both an antibacterial and an immunomodulatory drug that accumulates in immune cells. We set out to improve the immunomodulatory properties of azithromycin by coupling the immune activators, nitric oxide and acetate, to its core structure. This new compound, designated CSY5669, enhanced the intracellular killing of MRSA by 45% ± 20% in monocyte-derived macrophages and by 55% ± 15% in peripheral blood leukocytes, compared with untreated controls. CSY5669-treated peripheral blood leukocytes produced fewer proinflammatory cytokines, while in both monocyte-derived macrophages and peripheral blood leukocytes, phagocytosis, ROS production, and degranulation were unaffected. In mice with MRSA pneumonia, CSY5669 treatment reduced inflammation, lung pathology and vascular leakage with doses as low as 0.01 μmol/kg p.o. CSY5669 had diminished direct in vitro antibacterial properties compared with azithromycin. Also, CSY5669 was immunomodulatory at concentrations well below 1% of the minimum inhibitory concentration, which would minimize selection for macrolide-resistant bacteria if it were to be used as a host-directed therapy. This study highlights the potential of CSY5669 as a possible adjunctive therapy in pneumonia caused by MRSA, as CSY5669 could enhance bacterial eradication while simultaneously limiting inflammation-associated pathology.
Collapse
|
6
|
Sauer A, Peukert K, Putensen C, Bode C. Antibiotics as immunomodulators: a potential pharmacologic approach for ARDS treatment. Eur Respir Rev 2021; 30:210093. [PMID: 34615700 PMCID: PMC9489085 DOI: 10.1183/16000617.0093-2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/02/2021] [Indexed: 11/05/2022] Open
Abstract
First described in the mid-1960s, acute respiratory distress syndrome (ARDS) is a life-threatening form of respiratory failure with an overall mortality rate of approximately 40%. Despite significant advances in the understanding and treatment of ARDS, no substantive pharmacologic therapy has proven to be beneficial, and current management continues to be primarily supportive. Beyond their antibacterial activity, several antibiotics such as macrolides and tetracyclines exert pleiotropic immunomodulatory effects that might be able to rectify the dysregulated inflammatory response present in patients with ARDS. This review aims to provide an overview of preclinical and clinical studies that describe the immunomodulatory effects of antibiotics in ARDS. Moreover, the underlying mechanisms of their immunomodulatory properties will be discussed. Further studies are necessary to investigate their full therapeutic potential and to identify ARDS phenotypes which are most likely to benefit from their immunomodulatory effects.
Collapse
Affiliation(s)
- Andrea Sauer
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Konrad Peukert
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Putensen
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
7
|
Kricker JA, Page CP, Gardarsson FR, Baldursson O, Gudjonsson T, Parnham MJ. Nonantimicrobial Actions of Macrolides: Overview and Perspectives for Future Development. Pharmacol Rev 2021; 73:233-262. [PMID: 34716226 DOI: 10.1124/pharmrev.121.000300] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Macrolides are among the most widely prescribed broad spectrum antibacterials, particularly for respiratory infections. It is now recognized that these drugs, in particular azithromycin, also exert time-dependent immunomodulatory actions that contribute to their therapeutic benefit in both infectious and other chronic inflammatory diseases. Their increased chronic use in airway inflammation and, more recently, of azithromycin in COVID-19, however, has led to a rise in bacterial resistance. An additional crucial aspect of chronic airway inflammation, such as chronic obstructive pulmonary disease, as well as other inflammatory disorders, is the loss of epithelial barrier protection against pathogens and pollutants. In recent years, azithromycin has been shown with time to enhance the barrier properties of airway epithelial cells, an action that makes an important contribution to its therapeutic efficacy. In this article, we review the background and evidence for various immunomodulatory and time-dependent actions of macrolides on inflammatory processes and on the epithelium and highlight novel nonantibacterial macrolides that are being studied for immunomodulatory and barrier-strengthening properties to circumvent the risk of bacterial resistance that occurs with macrolide antibacterials. We also briefly review the clinical effects of macrolides in respiratory and other inflammatory diseases associated with epithelial injury and propose that the beneficial epithelial effects of nonantibacterial azithromycin derivatives in chronic inflammation, even given prophylactically, are likely to gain increasing attention in the future. SIGNIFICANCE STATEMENT: Based on its immunomodulatory properties and ability to enhance the protective role of the lung epithelium against pathogens, azithromycin has proven superior to other macrolides in treating chronic respiratory inflammation. A nonantibiotic azithromycin derivative is likely to offer prophylactic benefits against inflammation and epithelial damage of differing causes while preserving the use of macrolides as antibiotics.
Collapse
Affiliation(s)
- Jennifer A Kricker
- EpiEndo Pharmaceuticals, Reykjavik, Iceland (J.A.K., C.P.P., F.R.G., O.B., T.G., M.J.P.); Stem Cell Research Unit, Biomedical Center, University of Iceland, Reykjavik, Iceland (J.A.K., T.G.); Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (C.P.P.); Department of Respiratory Medicine (O.B.), Department of Laboratory Hematology (T.G.), Landspitali-University Hospital, Reykjavik, Iceland; Faculty of Biochemistry, Chemistry and Pharmacy, JW Goethe University Frankfurt am Main, Germany (M.J.P.)
| | - Clive P Page
- EpiEndo Pharmaceuticals, Reykjavik, Iceland (J.A.K., C.P.P., F.R.G., O.B., T.G., M.J.P.); Stem Cell Research Unit, Biomedical Center, University of Iceland, Reykjavik, Iceland (J.A.K., T.G.); Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (C.P.P.); Department of Respiratory Medicine (O.B.), Department of Laboratory Hematology (T.G.), Landspitali-University Hospital, Reykjavik, Iceland; Faculty of Biochemistry, Chemistry and Pharmacy, JW Goethe University Frankfurt am Main, Germany (M.J.P.)
| | - Fridrik Runar Gardarsson
- EpiEndo Pharmaceuticals, Reykjavik, Iceland (J.A.K., C.P.P., F.R.G., O.B., T.G., M.J.P.); Stem Cell Research Unit, Biomedical Center, University of Iceland, Reykjavik, Iceland (J.A.K., T.G.); Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (C.P.P.); Department of Respiratory Medicine (O.B.), Department of Laboratory Hematology (T.G.), Landspitali-University Hospital, Reykjavik, Iceland; Faculty of Biochemistry, Chemistry and Pharmacy, JW Goethe University Frankfurt am Main, Germany (M.J.P.)
| | - Olafur Baldursson
- EpiEndo Pharmaceuticals, Reykjavik, Iceland (J.A.K., C.P.P., F.R.G., O.B., T.G., M.J.P.); Stem Cell Research Unit, Biomedical Center, University of Iceland, Reykjavik, Iceland (J.A.K., T.G.); Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (C.P.P.); Department of Respiratory Medicine (O.B.), Department of Laboratory Hematology (T.G.), Landspitali-University Hospital, Reykjavik, Iceland; Faculty of Biochemistry, Chemistry and Pharmacy, JW Goethe University Frankfurt am Main, Germany (M.J.P.)
| | - Thorarinn Gudjonsson
- EpiEndo Pharmaceuticals, Reykjavik, Iceland (J.A.K., C.P.P., F.R.G., O.B., T.G., M.J.P.); Stem Cell Research Unit, Biomedical Center, University of Iceland, Reykjavik, Iceland (J.A.K., T.G.); Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (C.P.P.); Department of Respiratory Medicine (O.B.), Department of Laboratory Hematology (T.G.), Landspitali-University Hospital, Reykjavik, Iceland; Faculty of Biochemistry, Chemistry and Pharmacy, JW Goethe University Frankfurt am Main, Germany (M.J.P.)
| | - Michael J Parnham
- EpiEndo Pharmaceuticals, Reykjavik, Iceland (J.A.K., C.P.P., F.R.G., O.B., T.G., M.J.P.); Stem Cell Research Unit, Biomedical Center, University of Iceland, Reykjavik, Iceland (J.A.K., T.G.); Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (C.P.P.); Department of Respiratory Medicine (O.B.), Department of Laboratory Hematology (T.G.), Landspitali-University Hospital, Reykjavik, Iceland; Faculty of Biochemistry, Chemistry and Pharmacy, JW Goethe University Frankfurt am Main, Germany (M.J.P.)
| |
Collapse
|
8
|
Yadav N, Thakur AK, Shekhar N, Ayushi. Potential of Antibiotics for the Treatment and Management of Parkinson Disease: An Overview. Curr Drug Res Rev 2021; 13:166-171. [PMID: 33719951 DOI: 10.2174/2589977513666210315095133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/04/2020] [Accepted: 01/22/2021] [Indexed: 11/22/2022]
Abstract
Evidences have emerged over the last 2 decades to ascertain the proof of concepts viz. mitochondrial dysfunction, inflammation-derived oxidative damage and cytokine-induced toxicity that play a significant role in Parkinson's disease (PD). The available pharmacotherapies for PD are mainly symptomatic and typically indications of L-DOPA to restrain dopamine deficiency and their consequences. In the 21st century, the role of the antibiotics has emerged at the forefront of medicine in health and human illness. There are several experimental and pre-clinical evidences that supported the potential use of antibiotic as neuroprotective agent. The astonishing effects of antibiotics and their neuroprotective properties against neurodegeneration and neuro-inflammation would be phenomenal for the development of effective therapy against PD. Antibiotics are also testified as useful not only to prevent the formation of alpha-synuclein but also act on mitochondrial dysfunction and neuro-inflammation. Thus, the possible therapy with antibiotics in PD would impact both the pathways leading to neuronal cell death in substantia nigra and pars compacta in midbrain. Moreover, the antibiotic based pharmacotherapy will open a scientific research passageway to add more to the evidence based and rational use of antibiotics for the treatment and management of PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Narayan Yadav
- Neuropharmacology Research Laboratory, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi-110 017. India
| | - Ajit Kumar Thakur
- Neuropharmacology Research Laboratory, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi-110 017. India
| | - Nikhila Shekhar
- Neuropharmacology Research Laboratory, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi-110 017. India
| | - Ayushi
- Neuropharmacology Research Laboratory, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi-110 017. India
| |
Collapse
|
9
|
Reijnders TDY, Saris A, Schultz MJ, van der Poll T. Immunomodulation by macrolides: therapeutic potential for critical care. THE LANCET RESPIRATORY MEDICINE 2020; 8:619-630. [PMID: 32526189 DOI: 10.1016/s2213-2600(20)30080-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 12/17/2022]
Abstract
Critical illness is associated with immune dysregulation, characterised by concurrent hyperinflammation and immune suppression. Hyperinflammation can result in collateral tissue damage and organ failure, whereas immune suppression has been implicated in susceptibility to secondary infections and reactivation of latent viruses. Macrolides are a class of bacteriostatic antibiotics that are used in the intensive care unit to control infections or to alleviate gastrointestinal dysmotility. Yet macrolides also have potent and wide-ranging immunomodulatory properties, which might have the potential to correct immune dysregulation in patients who are critically ill without affecting crucial antimicrobial defences. In this Review, we provide an overview of preclinical and clinical studies that point to the beneficial effects of macrolides in acute diseases relevant to critical care, and we discuss the possible underlying mechanisms of their immunomodulatory effects. Further studies are needed to explore the therapeutic potential of macrolides in critical illness, to identify subgroups of patients who might benefit from treatment, and to develop novel non-antibiotic macrolide derivatives with improved immunomodulatory properties.
Collapse
Affiliation(s)
- Tom D Y Reijnders
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
| | - Anno Saris
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
| | - Marcus J Schultz
- Department of Intensive Care, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands; Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands; Division of Infectious Diseases, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands.
| |
Collapse
|
10
|
Andrada AC, Azuma MM, Furusho H, Hirai K, Xu S, White RR, Sasaki H. Immunomodulation Mediated by Azithromycin in Experimental Periapical Inflammation. J Endod 2020; 46:1648-1654. [PMID: 32763436 DOI: 10.1016/j.joen.2020.07.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The purpose of the present study was to compare the immunomodulatory effect of azithromycin (AZM), ampicillin (AMP), amoxicillin (AMX), and clindamycin (CLI) in vitro and AZM on preexisting periapical lesions compared with AMP. METHODS The susceptibility of 4 common human endodontic pathogens (Parvimonas micra, Streptococcus intermedius, Prevotella intermedia, and Fusobacterium nucleatum) to AZM, AMP, AMX, and CLI was confirmed by agar disk diffusion assay. Preexisting periapical lesions in C57BL/6J mice were treated with AZM, AMP, or phosphate-buffered saline (PBS). Periapical bone healing and the pattern of inflammatory cell infiltration were evaluated after a 10-day treatment by micro-computed tomographic and histology, respectively. Besides, the effect of antibiotics in pathogen-stimulated nuclear factor kappa B activation and the production of interleukin 1 alpha and tumor necrosis factor alpha was assessed in vitro by luciferase assay and enzyme-linked immunosorbent assay. RESULTS All examined endodontic pathogens were susceptible to AZM, AMP, AMX, and CLI. AZM significantly attenuated periapical bone loss versus PBS. PBS resulted in widely diffused infiltration of mixed inflammatory cells. By contrast, AZM brought about localized infiltration of neutrophils and M2 macrophages and advanced fibrosis. Although the effect of AMP on bone was uncertain, inflammatory cell infiltration was considerably milder than PBS. However, most macrophages observed seemed to be M1 macrophages. AZM suppressed pathogen-stimulated nuclear factor kappa B activation and cytokine production, whereas AMP, AMX, and CLI reduced only cytokine production moderately. CONCLUSIONS This study showed that AZM led to the resolution of preexisting experimental periapical inflammation. Our data provide a perspective on host response in antibiotic selection for endodontic treatment. However, well-designed clinical trials are necessary to better elucidate the benefits of AZM as an adjunctive therapy for endodontic treatment when antibiotic therapy is recommended. Although both AZM and AMP were effective on preexisting periapical lesions, AZM led to advanced wound healing, probably depending on its immunomodulatory effect.
Collapse
Affiliation(s)
- Ana Cristina Andrada
- Division of Endodontics, Department of Essentials and Simulation, University of Detroit Mercy School of Dentistry, Detroit, Michigan; Division of Endodontics, Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Mariane Maffei Azuma
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109
| | - Hisako Furusho
- Department of Oral and Maxillofacial Pathobiology, Hiroshima University, Hiroshima, Japan
| | - Kimito Hirai
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109; Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, Massachusetts; Department of Periodontics and Endodontics, Okayama University School of Dentistry, Kitaku, Okayama, Japan
| | - Shuang Xu
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, Massachusetts; Functional Genomics Laboratory, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robert R White
- Division of Endodontics, Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109; Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, Massachusetts; Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts.
| |
Collapse
|
11
|
Pretreatment of nafithromycin attenuates inflammatory response in murine lipopolysaccharide induced acute lung injury. Cytokine 2020; 129:155049. [PMID: 32126500 DOI: 10.1016/j.cyto.2020.155049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 12/11/2022]
Abstract
Acute respiratory distress syndrome following an acute lung injury (ALI) is a life threatening inflammatory condition predominantly characterized by vascular protein leakage, neutrophil recruitment and overexpression of proinflammatory cytokines. Pulmonary and systemic bacterial infections are the major cause of ALI wherein the bacterial cell components play a crucial role. Macrolide/ketolide antibiotics are reported to possess immunomodulatory activity; as a result improved survival has been noted in pneumonia patients. Hence immunomodulatory activity of nafithromycin, a novel lactone ketolide antibacterial agent was assessed in the murine LPS induced ALI model. Vehicle, nafithromycin (100 mg/kg), azithromycin (600 mg/kg) and dexamethasone (20 mg/kg) were administered orally, 1 h prior to LPS challenge and bronchoalveolar lavage (BAL) fluid was collected thereafter at 18, 24 and 48 h to determine the total cell count, total protein, myeloperoxidase (MPO), tumor necrosis factor (TNF)-α and interleukin (IL)-6. Results from the current study showed that pretreatment with nafithromycin significantly reduced the total cell count, total protein, MPO, TNF-α and IL-6 levels in BAL fluid compared to LPS control group. Histopathological evaluations also suggest significant reduction in neutrophil infiltration by nafithromycin. Dexamethasone, a positive reference standard as expected exhibited potent anti-inflammatory activity. The immunomodulatory effect of nafithromycin at dose of 100 mg/kg was comparable to azithromycin dosed at 600 mg/kg. As a result of immunomodulatory activity, nafithromycin is expected to provide additional clinical benefits by resolving the secondary complications associated with severe pneumonia and thereby improving survival in such patients.
Collapse
|
12
|
Lees KA, Orlandi RR, Oakley G, Alt JA. The Role of Macrolides and Doxycycline in Chronic Rhinosinusitis. Immunol Allergy Clin North Am 2020; 40:303-315. [PMID: 32278453 DOI: 10.1016/j.iac.2019.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Antibiotic therapy has become an important adjunct in the management of recalcitrant chronic rhinosinusitis (CRS) because of some antibiotics' immunomodulatory properties even at subtherapeutic antimicrobial levels. Macrolide antibiotics, such as clarithromycin and azithromycin, decrease production of proinflammatory cytokines, impair neutrophil recruitment, inhibit bacterial biofilm formation, and improve mucus quality. Doxycycline, a tetracycline antibiotic, inhibits the activity of matrix metalloproteinases in CRS with nasal polyposis. This article reviews the clinical applications for macrolide and doxycycline use in CRS, considerations for dosing and duration of treatment, and important side effects and drug interactions associated with these medications.
Collapse
Affiliation(s)
- Katherine A Lees
- Rhinology and Anterior Skull Base Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah, 50 North Medical Drive, #3C120, Salt Lake City, UT 84132, USA. https://twitter.com/TheSnotShot
| | - Richard R Orlandi
- Division of Otolaryngology-Head and Neck Surgery, University of Utah, 50 North Medical Drive, #3C120, Salt Lake City, UT 84132, USA
| | - Gretchen Oakley
- Division of Otolaryngology-Head and Neck Surgery, University of Utah, 50 North Medical Drive, #3C120, Salt Lake City, UT 84132, USA
| | - Jeremiah A Alt
- Division of Otolaryngology-Head and Neck Surgery, University of Utah, 50 North Medical Drive, #3C120, Salt Lake City, UT 84132, USA.
| |
Collapse
|
13
|
Parnham MJ, Geisslinger G. Pharmacological plasticity-How do you hit a moving target? Pharmacol Res Perspect 2019; 7:e00532. [PMID: 31768257 PMCID: PMC6868654 DOI: 10.1002/prp2.532] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 09/19/2019] [Accepted: 09/21/2019] [Indexed: 12/23/2022] Open
Abstract
Paul Ehrlich's concept of the magic bullet, by which a single drug induces pharmacological effects by interacting with a single receptor has been a strong driving force in pharmacology for a century. It is continually thwarted, though, by the fact that the treated organism is highly dynamic and the target molecule(s) is (are) never static. In this article, we address some of the factors that modify and cause the mobility and plasticity of drug targets and their interactions with ligands and discuss how these can lead to unexpected (lack of) effects of drugs. These factors include genetic, epigenetic, and phenotypic variability, cellular plasticity, chronobiological rhythms, time, age and disease resolution, sex, drug metabolism, and distribution. We emphasize four existing approaches that can be taken, either singly or in combination, to try to minimize effects of pharmacological plasticity. These are firstly, to enhance specificity using target conditions close to those in diseases, secondly, by simultaneously or thirdly, sequentially aiming at multiple targets, and fourthly, in synchronization with concurrent dietary, psychological, training, and biorhythm-synchronizing procedures to optimize drug therapy.
Collapse
Affiliation(s)
- Michael J. Parnham
- Fraunhofer Institute for Molecular Biology & Applied Ecology IMEBranch for Translational Medicine and Pharmacology TMPFrankfurt am MainGermany
| | - Gerd Geisslinger
- Fraunhofer Institute for Molecular Biology & Applied Ecology IMEBranch for Translational Medicine and Pharmacology TMPFrankfurt am MainGermany
- Institute of Clinical PharmacologyJ.W. Goethe UniversityFrankfurtGermany
| |
Collapse
|
14
|
Amantea D, Petrelli F, Greco R, Tassorelli C, Corasaniti MT, Tonin P, Bagetta G. Azithromycin Affords Neuroprotection in Rat Undergone Transient Focal Cerebral Ischemia. Front Neurosci 2019; 13:1256. [PMID: 31849581 PMCID: PMC6902046 DOI: 10.3389/fnins.2019.01256] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 11/05/2019] [Indexed: 01/04/2023] Open
Abstract
Repurposing existing drugs represents a promising approach for successful development of acute stroke therapies. In this context, the macrolide antibiotic azithromycin has been shown to exert neuroprotection in mice due to its immunomodulatory properties. Here, we have demonstrated that acute administration of a single dose of azithromycin upon reperfusion produces a dose-dependent (ED50 = 1.40 mg/kg; 95% CI = 0.48-4.03) reduction of ischemic brain damage measured 22 h after transient (2 h) middle cerebral artery occlusion (MCAo) in adult male rats. Neuroprotection by azithromycin (150 mg/kg, i.p., upon reperfusion) was associated with a significant elevation of signal transducer and activator of transcription 3 (STAT3) phosphorylation in astrocytes and neurons of the peri-ischemic motor cortex as detected after 2 and 22 h of reperfusion. By contrast, in the core region of the striatum, drug administration resulted in a dramatic elevation of STAT3 phosphorylation only after 22 h of reperfusion, being the signal mainly ascribed to infiltrating leukocytes displaying an M2 phenotype. These early molecular events were associated with a long-lasting neuroprotection, since a single dose of azithromycin reduced brain infarct damage and neurological deficit measured up to 7 days of reperfusion. These data, together with the evidence that azithromycin was effective in a clinically relevant time-window (i.e., when administered after 4.5 h of MCAo), provide robust preclinical evidence to support the importance of developing azithromycin as an effective acute therapy for ischemic stroke.
Collapse
Affiliation(s)
- Diana Amantea
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Francesco Petrelli
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Rosaria Greco
- Headache Science Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Cristina Tassorelli
- Headache Science Center, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | | | - Paolo Tonin
- Regional Center for Serious Brain Injuries, S. Anna Institute, Crotone, Italy
| | - Giacinto Bagetta
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
15
|
van Eeden SF, Hogg JC. Immune-Modulation in Chronic Obstructive Pulmonary Disease: Current Concepts and Future Strategies. Respiration 2019; 99:550-565. [PMID: 31480060 DOI: 10.1159/000502261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 11/19/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is caused by the chronic inhalation of toxic particles and gases that are primarily but not exclusively derived from cigarette smoke that may be either actively or passively inhaled, which initiates a persistent innate and adaptive immune response in the lung. This immune response is associated with an aberrant tissue repair and remodeling process that results in varying degrees of chronic inflammation with excess production of mucus in the central airways and permanent destruction of the smaller conducting airways and gas exchanging surface in the peripheral lung. Currently, the primary aims of treatment in COPD are bronchodilation (inhaled short- and long-acting β-agonist and antimuscarinic therapies), to control symptoms and nonspecific broad-acting anti-inflammatory agents (inhaled and oral corticosteroids, phosphor-di-esterase inhibitors, and macrolides). That provide symptomatic relief but have little or no impact on either disease progression or mortality. As our understanding of the immune pathogenesis of the COPD improves, available immune modulation therapies have the potential to alter or interfere with damaging immune pathways, thereby slowing relentless progression of lung tissue destruction. The purpose of this brief review is to discuss our current understanding of the immune pathogenesis of both the airways and parenchymal injury as well as the dysfunctional tissue repair process to propose immune modulating interventions in an attempt to stabilize or return these pathological changes to their normal state. The ultimate goal of the immune modulation therapy is to improve both morbidity and mortality associated with COPD.
Collapse
Affiliation(s)
- Stephan F van Eeden
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada, .,Pacific Lung Health Centre, St. Paul's Hospital, Vancouver, British Columbia, Canada,
| | - James C Hogg
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Zhang B, Kopper TJ, Liu X, Cui Z, Van Lanen SG, Gensel JC. Macrolide derivatives reduce proinflammatory macrophage activation and macrophage-mediated neurotoxicity. CNS Neurosci Ther 2019; 25:591-600. [PMID: 30677254 PMCID: PMC6488883 DOI: 10.1111/cns.13092] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/07/2018] [Accepted: 11/15/2018] [Indexed: 12/21/2022] Open
Abstract
Introduction Azithromycin (AZM) and other macrolide antibiotics are applied as immunomodulatory treatments for CNS disorders. The immunomodulatory and antibiotic properties of AZM are purportedly independent. Aims To improve the efficacy and reduce antibiotic resistance risk of AZM‐based therapies, we evaluated the immunomodulatory and neuroprotective properties of novel AZM derivatives. We semisynthetically prepared derivatives by altering sugar moieties established as important for inhibiting bacterial protein synthesis. Bone marrow‐derived macrophages (BMDMs) were stimulated in vitro with proinflammatory, M1, stimuli (LPS + INF‐gamma) with and without derivative costimulation. Pro‐ and anti‐inflammatory cytokine production, IL‐12 and IL‐10, respectively, was quantified using ELISA. Neuron culture treatment with BMDM supernatant was used to assess derivative neuroprotective potential. Results Azithromycin and some derivatives increased IL‐10 and reduced IL‐12 production of M1 macrophages. IL‐10/IL‐12 cytokine shifts closely correlated with the ability of AZM and derivatives to mitigate macrophage neurotoxicity. Conclusions Sugar moieties that bind bacterial ribosomal complexes can be modified in a manner that retains AZM immunomodulation and neuroprotection. Since the effects of BMDMs in vitro are predictive of CNS macrophage responses, our results open new therapeutic avenues for managing maladaptive CNS inflammation and support utilization of IL‐10/12 cytokine profiles as indicators of macrophage polarization and neurotoxicity.
Collapse
Affiliation(s)
- Bei Zhang
- Department of Physiology, College of Medicine, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Timothy J Kopper
- Department of Physiology, College of Medicine, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Xiaodong Liu
- Division of Bioorganic, Medicinal, & Computational Chemistry, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - Zheng Cui
- Division of Bioorganic, Medicinal, & Computational Chemistry, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - Steven G Van Lanen
- Division of Bioorganic, Medicinal, & Computational Chemistry, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - John C Gensel
- Department of Physiology, College of Medicine, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
17
|
Perra L, Balloy V, Foussignière T, Moissenet D, Petat H, Mungrue IN, Touqui L, Corvol H, Chignard M, Guillot L. CHAC1 Is Differentially Expressed in Normal and Cystic Fibrosis Bronchial Epithelial Cells and Regulates the Inflammatory Response Induced by Pseudomonas aeruginosa. Front Immunol 2018; 9:2823. [PMID: 30555487 PMCID: PMC6282009 DOI: 10.3389/fimmu.2018.02823] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/15/2018] [Indexed: 12/31/2022] Open
Abstract
In cystic fibrosis (CF), Pseudomonas aeruginosa (Pa) colonizes the lungs, leading to chronic inflammation of the bronchial epithelium. ChaC glutathione-specific γ-glutamylcyclotransferase 1 (CHAC1) mRNA is differentially expressed in primary human airway epithelial cells from bronchi (hAECBs) from patients with CF and healthy patients at baseline and upon infection with Pa. CHAC1 degrades glutathione and is associated with ER stress and apoptosis pathways. In this study, we examined the roles of CHAC1 in the inflammatory response and apoptosis in lung epithelial cells. First, we confirmed by reverse transcription quantitative polymerase chain reaction that CHAC1 mRNA was overexpressed in hAECBs from patients without CF compared with the expression in hAECBs from patients with CF upon Pa (PAK strain) infection. Moreover, the Pa virulence factors LPS and flagellin were shown to induce CHAC1 expression in cells from patients without CF. Using NCI-H292 lung epithelial cells, we found that LPS-induced CHAC1 mRNA expression was PERK-independent and involved ATF4. Additionally, using CHAC1 small interfering RNA, we showed that reduced CHAC1 expression in the context of LPS and flagellin stimulation was associated with modulation of inflammatory markers and alteration of NF-κB signaling. Finally, we showed that Pa was not able to induce apoptosis in NCI-H292 cells. Our results suggest that CHAC1 is involved in the regulation of inflammation in bronchial cells during Pa infection and may explain the excessive inflammation present in the respiratory tracts of patients with CF.
Collapse
Affiliation(s)
- Léa Perra
- Sorbonne Université, Inserm, Centre de recherche Saint-Antoine (CRSA), Paris, France
| | - Viviane Balloy
- Sorbonne Université, Inserm, Centre de recherche Saint-Antoine (CRSA), Paris, France
| | - Tobias Foussignière
- Sorbonne Université, Inserm, Centre de recherche Saint-Antoine (CRSA), Paris, France
| | - Didier Moissenet
- Department of Bacteriology, APHP, Hôpital St-Antoine, Paris, France
| | - Hortense Petat
- Sorbonne Université, Inserm, Centre de recherche Saint-Antoine (CRSA), Paris, France
| | - Imran N Mungrue
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Lhousseine Touqui
- Equipe mixte Institut Pasteur/Paris V "Mucoviscidose et Bronchopathies Chroniques" Institut Pasteur, Paris, France
| | - Harriet Corvol
- Sorbonne Université, Inserm, Centre de recherche Saint-Antoine (CRSA), Paris, France.,Pneumologie Pédiatrique, APHP, Hôpital Trousseau, Paris, France
| | - Michel Chignard
- Sorbonne Université, Inserm, Centre de recherche Saint-Antoine (CRSA), Paris, France
| | - Loic Guillot
- Sorbonne Université, Inserm, Centre de recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|
18
|
Cui Y, Luo L, Li C, Chen P, Chen Y. Long-term macrolide treatment for the prevention of acute exacerbations in COPD: a systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis 2018; 13:3813-3829. [PMID: 30538443 PMCID: PMC6254503 DOI: 10.2147/copd.s181246] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Acute exacerbation of COPD (AECOPD) is associated with an increased hospitalization and mortality. Azithromycin and erythromycin are the recommended drugs to reduce the risk of exacerbations. However, the most suitable duration of therapy and drug-related adverse events are still a matter of debate. The aim of this meta-analysis was to assess the current evidence regarding the efficacy and safety of long-term macrolide treatment for COPD. Materials and methods We comprehensively searched PubMed, Embase, the Cochrane Library, and the Web of Science and performed a systematic review and cumulative meta-analysis of all randomized controlled trials (RCTs) and retrospective studies. Results Eleven RCTs and one retrospective study including a total of 2,151 cases were carried out. Long-term macrolide treatment significantly reduced the total number of cases with one or more exacerbations (OR=0.40; 95% CI=0.24–0.65; P<0.01) and the rate of exacerbations per patient per year (risk ratio [RR]=0.60; 95% CI=0.45–0.78; P<0.01). Subgroup analyses showed that the minimum duration for drug efficacy for both azithromycin and erythromycin therapy was 6 months. In addition, macrolide therapy could improve the St George Respiratory Questionnaire (SGRQ) total score (P<0.01) but did not achieve the level of clinical significance. The frequency of hospitalizations was not significantly different between the treatment and control groups (P=0.50). Moreover, chronic azithromycin treatment was more likely to increase adverse events (P<0.01). Conclusion Prophylactic azithromycin or erythromycin treatment has a significant effect in reducing the frequency of AECOPD in a time-dependent manner. However, long-term macrolide treatment could increase the occurrence of adverse events and macrolide resistance. Future large-scale, well-designed RCTs with extensive follow-up are required to identify patients in whom the benefits outweigh risks.
Collapse
Affiliation(s)
- Yanan Cui
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China,
| | - Lijuan Luo
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China,
| | - Chenbei Li
- Biomedical Clinical Medicine, The Queen Marry University of London of Nanchang University, Jiangxi, China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China,
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China,
| |
Collapse
|
19
|
Jayne JG, Bensman TJ, Schaal JB, Park AYJ, Kimura E, Tran D, Selsted ME, Beringer PM. Rhesus θ-Defensin-1 Attenuates Endotoxin-induced Acute Lung Injury by Inhibiting Proinflammatory Cytokines and Neutrophil Recruitment. Am J Respir Cell Mol Biol 2018; 58:310-319. [PMID: 28954201 DOI: 10.1165/rcmb.2016-0428oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Acute lung injury (ALI) is a clinical syndrome characterized by acute respiratory failure and is associated with substantial morbidity and mortality. Rhesus θ-defensin (RTD)-1 is an antimicrobial peptide with immunomodulatory activity. As airway inflammation and neutrophil recruitment and activation are hallmarks of ALI, we evaluated the therapeutic efficacy of RTD-1 in preclinical models of the disease. We investigated the effect of RTD-1 on neutrophil chemotaxis and macrophage-driven pulmonary inflammation with human peripheral neutrophils and LPS-stimulated murine alveolar macrophage (denoted MH-S) cells. Treatment and prophylactic single escalating doses were administered subcutaneously in a well-established murine model of direct endotoxin-induced ALI. We assessed lung injury by histopathology, pulmonary edema, inflammatory cell recruitment, and inflammatory cytokines/chemokines in the BAL fluid. In vitro studies demonstrated that RTD-1 suppressed CXCL8-induced neutrophil chemotaxis, TNF-mediated neutrophil-endothelial cell adhesion, and proinflammatory cytokine release in activated murine alveolar immortalized macrophages (MH-S) cells. Treatment with RTD-1 significantly inhibited in vivo LPS-induced ALI by reducing pulmonary edema and histopathological changes. Treatment was associated with dose- and time-dependent inhibition of proinflammatory cytokines (TNF, IL-1β, and IL-6), peroxidase activity, and neutrophil recruitment into the airways. Antiinflammatory effects were demonstrated in animals receiving RTD-1 up to 12 hours after LPS challenge. Notably, subcutaneously administered RTD-1 demonstrates good peptide stability as demonstrated by the long in vivo half-life. Taken together, these studies demonstrate that RTD-1 is efficacious in an experimental model of ALI through inhibition of neutrophil chemotaxis and adhesion, and the attenuation of proinflammatory cytokines and gene expression from alveolar macrophages.
Collapse
Affiliation(s)
| | | | - Justin B Schaal
- 2 Keck School of Medicine, University of Southern California, Los Angeles, California; and
| | | | - Elza Kimura
- 3 State University of Maringá, Maringá, Paraná, Brazil
| | - Dat Tran
- 2 Keck School of Medicine, University of Southern California, Los Angeles, California; and
| | - Michael E Selsted
- 2 Keck School of Medicine, University of Southern California, Los Angeles, California; and
| | | |
Collapse
|
20
|
Combining MucilAir™ and Vitrocell ® Powder Chamber for the In Vitro Evaluation of Nasal Ointments in the Context of Aerosolized Pollen. Pharmaceutics 2018; 10:pharmaceutics10020056. [PMID: 29747472 PMCID: PMC6027377 DOI: 10.3390/pharmaceutics10020056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 12/24/2022] Open
Abstract
Hay fever is notoriously triggered when nasal mucosa is exposed to allergenic pollen. One possibility to overcome this pollen exposure may be the application of an ointment with physical protective effects. In this context, we have investigated Bepanthen® Eye and Nose Ointment and the ointment basis petrolatum as reference while using contemporary in vitro techniques. Pollen from false ragweed (Iva xanthiifolia) was used as an allergy-causing model deposited as aerosol using the Vitrocell® Powder Chamber (VPC) on Transwell® inserts, while being coated with either Bepanthen® Eye and Nose Ointment and petrolatum. No pollen penetration into ointments was observed upon confocal scanning laser microscopy during an incubation period of 2 h at 37 °C. The cellular response was further investigated by integrating the MucilAir™ cell system in the VPC and by applying pollen to Bepanthen® Eye and Nose Ointment covered cell cultures. For comparison, MucilAir™ were stimulated by lipopolysaccharides (LPS). No increased cytokine release of IL-6, TNF-α, or IL-8 was found after 4 h of pollen exposure, which demonstrates the safety of such ointments. Since nasal ointments act as a physical barrier against pollen, such preparations might support the prevention and management of hay fever.
Collapse
|
21
|
Scambler T, Holbrook J, Savic S, McDermott MF, Peckham D. Autoinflammatory disease in the lung. Immunology 2018; 154:563-573. [PMID: 29676014 PMCID: PMC6050210 DOI: 10.1111/imm.12937] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 03/21/2018] [Indexed: 01/11/2023] Open
Abstract
Ascertaining the dominant cell type driving an immunological disease is essential to understanding the causal pathology and, therefore, selecting or developing an effective treatment. Classifying immunological diseases in this way has led to successful treatment regimens for many monogenic diseases; however, when the dominant cell type is unclear and there is no obvious causal genetic mutation, then identifying the correct disease classification and appropriate therapy can be challenging. In this review we focus on pulmonary immunological diseases where an innate immune signature has been identified as a predominant aspect of the immunopathology. We describe the molecular pathology of 'autoinflammatory diseases of the lung' and propose that small molecule and biological therapies, including recombinant interleukin-1 receptor antagonist, that target key innate immune pathways, are likely be beneficial in the control of pulmonary and systemic inflammation in these conditions. In addition, the successful use of macrolide antibiotics to treat lung infections in these conditions further confirms that the innate immune system is the key conductor of inflammation in these pulmonary diseases, as there is a strong body of evidence that macrolides are able to modulate the NLRP3 inflammasome and interleukin-1β and interleukin-18 secretion, both of which are central players in the innate immune response. Throughout this review we highlight the published evidence of autoinflammatory disease in chronic obstructive pulmonary disease, bronchiectasis, cystic fibrosis and rheumatoid lung disease and suggest that the fundamental pathology of these diseases places them towards the autoinflammatory pole of the immunological disease continuum.
Collapse
Affiliation(s)
- Thomas Scambler
- Leeds Institute of Rheumatic and Musculoskeletal MedicineSt James's University HospitalLeedsUK
- Cystic Fibrosis Trust Strategic Research CentreSt James's University HospitalLeedsUK
| | - Jonathan Holbrook
- Leeds Institute of Rheumatic and Musculoskeletal MedicineSt James's University HospitalLeedsUK
- Cystic Fibrosis Trust Strategic Research CentreSt James's University HospitalLeedsUK
- Leeds Institute of Biomedical and Clinical SciencesSt James's University HospitalLeedsUK
| | - Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal MedicineSt James's University HospitalLeedsUK
- Cystic Fibrosis Trust Strategic Research CentreSt James's University HospitalLeedsUK
- Department of Clinical Immunology and AllergySt James's University HospitalLeedsUK
| | - Michael F. McDermott
- Leeds Institute of Rheumatic and Musculoskeletal MedicineSt James's University HospitalLeedsUK
- Cystic Fibrosis Trust Strategic Research CentreSt James's University HospitalLeedsUK
| | - Daniel Peckham
- Cystic Fibrosis Trust Strategic Research CentreSt James's University HospitalLeedsUK
- Leeds Institute of Biomedical and Clinical SciencesSt James's University HospitalLeedsUK
- Leeds Centre for Cystic FibrosisSt James's University HospitalLeedsUK
| |
Collapse
|
22
|
Cazzola M, Rogliani P, Puxeddu E, Ora J, Matera MG. An overview of the current management of chronic obstructive pulmonary disease: can we go beyond the GOLD recommendations? Expert Rev Respir Med 2017; 12:43-54. [DOI: 10.1080/17476348.2018.1398086] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Mario Cazzola
- Department of Systems Medicine, Chair of Respiratory Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Paola Rogliani
- Department of Systems Medicine, Chair of Respiratory Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Division of Respiratory Medicine, Department of Internal Medicine, University Hospital “Tor Vergata”, Rome, Italy
| | - Ermanno Puxeddu
- Department of Systems Medicine, Chair of Respiratory Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Division of Respiratory Medicine, Department of Internal Medicine, University Hospital “Tor Vergata”, Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, Department of Internal Medicine, University Hospital “Tor Vergata”, Rome, Italy
| | - Maria Gabriella Matera
- Department of Experimental Medicine, Unit of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
23
|
Ruscitti F, Ravanetti F, Essers J, Ridwan Y, Belenkov S, Vos W, Ferreira F, KleinJan A, van Heijningen P, Van Holsbeke C, Cacchioli A, Villetti G, Stellari FF. Longitudinal assessment of bleomycin-induced lung fibrosis by Micro-CT correlates with histological evaluation in mice. Multidiscip Respir Med 2017; 12:8. [PMID: 28400960 PMCID: PMC5387277 DOI: 10.1186/s40248-017-0089-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/10/2017] [Indexed: 01/15/2023] Open
Abstract
Background The intratracheal instillation of bleomycin in mice induces early damage to alveolar epithelial cells and development of inflammation followed by fibrotic tissue changes and represents the most widely used model of pulmonary fibrosis to investigate human IPF. Histopathology is the gold standard for assessing lung fibrosis in rodents, however it precludes repeated and longitudinal measurements of disease progression and does not provide information on spatial and temporal distribution of tissue damage. Here we investigated the use of the Micro-CT technique to allow the evaluation of disease onset and progression at different time-points in the mouse bleomycin model of lung fibrosis. Micro-CT was throughout coupled with histological analysis for the validation of the imaging results. Methods In bleomycin-instilled and control mice, airways and lung morphology changes were assessed and reconstructed at baseline, 7, 14 and 21 days post-treatment based on Micro-CT images. Ashcroft score, percentage of collagen content and percentage of alveolar air area were detected on lung slides processed by histology and subsequently compared with Micro-CT parameters. Results Extent (%) of fibrosis measured by Micro-CT correlated with Ashcroft score, the percentage of collagen content and the percentage of alveolar air area (r2 = 0.91; 0.77; 0.94, respectively). Distal airway radius also correlated with the Ashcroft score, the collagen content and alveolar air area percentage (r2 = 0.89; 0.78; 0.98, respectively). Conclusions Micro-CT data were in good agreement with histological read-outs as micro-CT was able to quantify effectively and non-invasively disease progression longitudinally and to reduce the variability and number of animals used to assess the damage. This suggests that this technique is a powerful tool for understanding experimental pulmonary fibrosis and that its use could translate into a more efficient drug discovery process, also helping to fill the gap between preclinical setting and clinical practice.
Collapse
Affiliation(s)
| | - Francesca Ravanetti
- Dipartimento di Scienze Medico Veterinarie, Università di Parma, Parma, Italy
| | - Jeroen Essers
- Department of Molecular Genetics, Vascular Surgery, and Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Yanto Ridwan
- Department of Molecular Genetics, Vascular Surgery, and Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Wim Vos
- Fluidda NV, Kontich, Belgium
| | | | - Alex KleinJan
- Department of Pulmonary Medicine Erasmus MC, Rotterdam, The Netherlands
| | - Paula van Heijningen
- Department of Molecular Genetics, Vascular Surgery, and Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Antonio Cacchioli
- Dipartimento di Scienze Medico Veterinarie, Università di Parma, Parma, Italy
| | | | - Franco Fabio Stellari
- Chiesi S.p.A., Pre-Clinical R & D, Parma, Italy.,Chiesi Farmaceutici, Pharmacology & Toxicology Department Corporate Pre-Clinical R & D, Largo Belloli, 11/A, Parma, 43122 Italy
| |
Collapse
|
24
|
Thomson NC. New and developing non-adrenoreceptor small molecule drugs for the treatment of asthma. Expert Opin Pharmacother 2017; 18:283-293. [PMID: 28099820 DOI: 10.1080/14656566.2017.1284794] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Inhaled corticosteroids (ICS) alone or in combination with an inhaled long-acting beta2-agonist (LABA) are the preferred long-term treatment for adults and adolescents with symptomatic asthma. Additional drugs include leukotriene-receptor antagonists, slow-release theophylline and the long-acting muscarinic antagonist (LAMA) tiotropium (approved in 2015). There is a need for more effective therapies, as many patients continue to have poorly controlled asthma. Areas covered: New and developing long-acting non-adrenoreceptor synthetic drugs for the treatment of symptomatic chronic asthma despite treatment with an ICS alone or combined with a LABA. Data was reviewed from studies published up until November 2016. Expert opinion: Tiotropium improves lung function and has a modest effect in reducing exacerbations when added to ICS alone or ICS and LABA. The LAMAs umeclidinium and glycopyrronium are under development in fixed dose combination with ICS and LABA. Novel small molecule drugs, such as CRTH2 receptor antagonists, PDE4 inhibitors, protein kinase inhibitors and nonsteroidal glucocorticoid receptor agonists and 'off-label' use of licensed drugs, such as macrolides and statins are under investigation for asthma, although their effectiveness in clinical practice is not established. To better achieve the goal of developing effective novel small molecule drugs for asthma will require greater understanding of mechanisms of disease and the different phenotypes and endotypes of asthma.
Collapse
Affiliation(s)
- Neil C Thomson
- a Institute of Infection, Immunity & Inflammation , University of Glasgow , Glasgow , UK
| |
Collapse
|
25
|
Thomson NC. New and developing non-adrenoreceptor small molecule drugs for the treatment of asthma. Expert Opin Pharmacother 2017. [DOI: 10.10.1080/14656566.2017.1284794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Neil C Thomson
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
26
|
Cramer CL, Patterson A, Alchakaki A, Soubani AO. Immunomodulatory indications of azithromycin in respiratory disease: a concise review for the clinician. Postgrad Med 2017; 129:493-499. [PMID: 28116959 DOI: 10.1080/00325481.2017.1285677] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Azithromycin has a well-characterized bacteriostatic activity. However, it also has a robust immunomodulatory effect that has proven beneficial in a variety of chronic illnesses. This effect results in decreased production of pro-inflammatory cytokines in the acute phase and promotes resolution of chronic inflammation in the later phases. Specifically, azithromycin has direct activity on airway epithelial cells to maintain their function and reduce mucus secretion. These characteristics have resulted in the use of azithromycin in the management of a variety of chronic lung diseases including chronic obstructive pulmonary disease, cystic fibrosis (CF), non-CF bronchiectasis, bronchiolitis obliterans syndrome, diffuse panbronchiolitis, and asthma. In this review, we present the evidence supporting the role of azithromycin in these conditions with an emphasis on the clinical aspects for the practicing physician.
Collapse
Affiliation(s)
- Cassondra L Cramer
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Allie Patterson
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Abdulrazak Alchakaki
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Ayman O Soubani
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| |
Collapse
|
27
|
Gensel JC, Kopper TJ, Zhang B, Orr MB, Bailey WM. Predictive screening of M1 and M2 macrophages reveals the immunomodulatory effectiveness of post spinal cord injury azithromycin treatment. Sci Rep 2017; 7:40144. [PMID: 28057928 PMCID: PMC5216345 DOI: 10.1038/srep40144] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/01/2016] [Indexed: 11/25/2022] Open
Abstract
Spinal cord injury (SCI) triggers a heterogeneous macrophage response that when experimentally polarized toward alternative forms of activation (M2 macrophages) promotes tissue and functional recovery. There are limited pharmacological therapies that can drive this reparative inflammatory state. In the current study, we used in vitro systems to comprehensively defined markers of macrophages with known pathological (M1) and reparative (M2) properties in SCI. We then used these markers to objectively define the macrophage activation states after SCI in response to delayed azithromycin treatment. Mice were subjected to moderate-severe thoracic contusion SCI. Azithromycin or vehicle was administered beginning 30 minutes post-SCI and then daily for 3 or 7 days post injury (dpi). We detected a dose-dependent polarization toward purportedly protective M2 macrophages with daily AZM treatment. Specifically, AZM doses of 10, 40, or 160 mg/kg decreased M1 macrophage gene expression at 3 dpi while the lowest (10 mg/kg) and highest (160 mg/kg) doses increased M2 macrophage gene expression at 7 dpi. Azithromycin has documented immunomodulatory properties and is commonly prescribed to treat infections in SCI individuals. This work demonstrates the utility of objective, comprehensive macrophage gene profiling for evaluating immunomodulatory SCI therapies and highlights azithromycin as a promising agent for SCI treatment.
Collapse
Affiliation(s)
- John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine University of Kentucky Lexington, Kentucky 40536
| | - Timothy J Kopper
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine University of Kentucky Lexington, Kentucky 40536
| | - Bei Zhang
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine University of Kentucky Lexington, Kentucky 40536
| | - Michael B Orr
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine University of Kentucky Lexington, Kentucky 40536
| | - William M Bailey
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine University of Kentucky Lexington, Kentucky 40536
| |
Collapse
|
28
|
Qiu S, Zhong X. Macrolides: a promising pharmacologic therapy for chronic obstructive pulmonary disease. Ther Adv Respir Dis 2016; 11:147-155. [PMID: 28030992 PMCID: PMC5933650 DOI: 10.1177/1753465816682677] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chronic inflammation plays a central role in the pathogenesis of chronic
obstructive pulmonary disease (COPD). However, there are no effective
anti-inflammatory pharmacologic therapies available for COPD so far. Recent
evidence suggests that an immunologic mechanism has a role in the pathogenesis
of COPD. Macrolides possess anti-inflammatory and immune-modulating effects may
be helpful in the treatment of COPD. Several clinical studies have shown that
long-term use of macrolides reduces the frequency of COPD exacerbations.
However, the subgroups that most effectively respond to long-term treatment of
macrolides still need to be determined. The potential adverse events to
individuals and the microbial resistance in community populations raises great
concern on the long-term use of macrolides. Thus, novel macrolides have
anti-inflammatory and immuno-modulating effects, but without antibiotic effects,
and are promising as an anti-inflammatory agent for the treatment of COPD. In
addition, the combination of macrolides and other anti-inflammatory
pharmacologic agents may be a new strategy for the treatment of COPD.
Collapse
Affiliation(s)
- Shilin Qiu
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoning Zhong
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Shuangyong road, Nanning, Guangxi 530021, China
| |
Collapse
|
29
|
Samson C, Tamalet A, Thien HV, Taytard J, Perisson C, Nathan N, Clement A, Boelle PY, Corvol H. Long-term effects of azithromycin in patients with cystic fibrosis. Respir Med 2016; 117:1-6. [DOI: 10.1016/j.rmed.2016.05.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 05/17/2016] [Accepted: 05/23/2016] [Indexed: 10/21/2022]
|
30
|
Stellari F, Bergamini G, Ruscitti F, Sandri A, Ravanetti F, Donofrio G, Boschi F, Villetti G, Sorio C, Assael BM, Melotti P, Lleo MM. In vivo monitoring of lung inflammation in CFTR-deficient mice. J Transl Med 2016; 14:226. [PMID: 27468800 PMCID: PMC4964274 DOI: 10.1186/s12967-016-0976-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/13/2016] [Indexed: 12/14/2022] Open
Abstract
Background Experimentally, lung inflammation in laboratory animals is usually detected by the presence of inflammatory markers, such as immune cells and cytokines, in the bronchoalveolar lavage fluid (BALF) of sacrificed animals. This method, although extensively used, is time, money and animal life consuming, especially when applied to genetically modified animals. Thus a new and more convenient approach, based on in vivo imaging analysis, has been set up to evaluate the inflammatory response in the lung of CFTR-deficient (CF) mice, a murine model of cystic fibrosis. Methods Wild type (WT) and CF mice were stimulated with P. aeruginosa LPS, TNF-alpha and culture supernatant derived from P. aeruginosa (strain VR1). Lung inflammation was detected by measuring bioluminescence in vivo in mice transiently transgenized with a luciferase reporter gene under the control of a bovine IL-8 gene promoter. Results Differences in bioluminescence (BLI) signal were revealed by comparing the two types of mice after intratracheal challenge with pro-inflammatory stimuli. BLI increased at 4 h after stimulation with TNF-alpha and at 24 h after administration of LPS and VR1 supernatant in CF mice with respect to untreated animals. The BLI signal was significantly more intense and lasted for longer times in CF animals when compared to WT mice. Analysis of BALF markers: leukocytes, cytokines and histology revealed no significant differences between CF and WT mice. Conclusions In vivo gene delivery technology and non-invasive bioluminescent imaging has been successfully adapted to CFTR-deficient mice. Activation of bIL-8 transgene promoter can be monitored by non-invasive BLI imaging in the lung of the same animal and compared longitudinally in both CF or WT mice, after challenge with pro-inflammatory stimuli. The combination of these technologies and the use of CF mice offer the unique opportunity of evaluating the impact of therapies aimed to control inflammation in a CF background. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0976-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fabio Stellari
- Pharmacology & Toxicology Department Corporate Pre-Clinical R&D, Chiesi Farmaceutici, Largo Belloli, 11/A, 43122, Parma, Italy.
| | | | - Francesca Ruscitti
- Dipartimento di Scienze Biomediche, Biotecnologiche e Traslazionali, Università di Parma, Parma, Italy
| | - Angela Sandri
- Dipartimento di Diagnostica e Salute Pubblica, Università di Verona, Verona, Italy
| | - Francesca Ravanetti
- Dipartimento di Scienze Medico Veterinarie, Università di Parma, Parma, Italy
| | - Gaetano Donofrio
- Dipartimento di Scienze Medico Veterinarie, Università di Parma, Parma, Italy
| | - Federico Boschi
- Dipartimento di Informatica, Università di Verona, Verona, Italy
| | - Gino Villetti
- Pharmacology & Toxicology Department Corporate Pre-Clinical R&D, Chiesi Farmaceutici, Largo Belloli, 11/A, 43122, Parma, Italy
| | - Claudio Sorio
- Dipartimento di Medicina, Università di Verona, Verona, Italy
| | - Barouk M Assael
- Centro Fibrosi Cistica, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Paola Melotti
- Centro Fibrosi Cistica, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Maria M Lleo
- Dipartimento di Diagnostica e Salute Pubblica, Università di Verona, Verona, Italy
| |
Collapse
|
31
|
Abstract
Noneosinophilic airway inflammation occurs in approximately 50% of patients with asthma. It is subdivided into neutrophilic or paucigranulocytic inflammation, although the proportion of each subtype is uncertain because of variable cut-off points used to define neutrophilia. This article reviews the evidence for noneosinophilic inflammation being a target for therapy in asthma and assesses clinical trials of licensed drugs, novel small molecules and biologics agents in noneosinophilic inflammation. Current symptoms, rate of exacerbations and decline in lung function are generally less in noneosinophilic asthma than eosinophilic asthma. Noneosinophilic inflammation is associated with corticosteroid insensitivity. Neutrophil activation in the airways and systemic inflammation is reported in neutrophilic asthma. Neutrophilia in asthma may be due to corticosteroids, associated chronic pulmonary infection, altered airway microbiome or delayed neutrophil apoptosis. The cause of poorly controlled noneosinophilic asthma may differ between patients and involve several mechanism including neutrophilic inflammation, T helper 2 (Th2)-low or other subtypes of airway inflammation or corticosteroid insensitivity as well as noninflammatory pathways such as airway hyperreactivity and remodelling. Smoking cessation in asthmatic smokers and removal from exposure to some occupational agents reduces neutrophilic inflammation. Preliminary studies of 'off-label' use of licensed drugs suggest that macrolides show efficacy in nonsmokers with noneosinophilic severe asthma and statins, low-dose theophylline and peroxisome proliferator-activated receptor gamma (PPARγ) agonists may benefit asthmatic smokers with noneosinophilic inflammation. Novel small molecules targeting neutrophilic inflammation, such as chemokine (CXC) receptor 2 (CXCR2) antagonists reduce neutrophils, but do not improve clinical outcomes in studies to date. Inhaled phosphodiesterase (PDE)4 inhibitors, dual PDE3 and PDE4 inhibitors, p38MAPK (mitogen-activated protein kinase) inhibitors, tyrosine kinase inhibitors and PI (phosphoinositide) 3kinase inhibitors are under development and these compounds may be of benefit in noneosinophilic inflammation. The results of clinical trials of biological agents targeting mediators associated with noneosinophilic inflammation, such as interleukin (IL)-17 and tumor necrosis factor (TNF)-α are disappointing. Greater understanding of the mechanisms of noneosinophilic inflammation in asthma should lead to improved therapies.
Collapse
Affiliation(s)
- Neil C Thomson
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 0YN, UK
| |
Collapse
|
32
|
Fuger M, Clair MP, El Ayoun Ibrahim N, L'Excellent S, Nizery L, O'Neill C, Tabone L, Truffinet O, Yakovleff C, de Blic J. [Chronic interstitial lung disease in children: Diagnostic approach and management]. Arch Pediatr 2016; 23:525-31. [PMID: 27021883 DOI: 10.1016/j.arcped.2016.02.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 01/05/2016] [Accepted: 02/23/2016] [Indexed: 02/06/2023]
Abstract
Chronic interstitial lung disease (ILD) in children is a heterogeneous group of rare lung disorders characterized by an inflammatory process of the alveolar wall and the pulmonary interstitium that induces gas exchange disorders. The diagnostic approach to an ILD involves three essential steps: recognizing the ILD, appreciating the impact, and identifying the cause. The spectrum of clinical findings depends to a large extent on age. In the newborn, the beginning is often abrupt (neonatal respiratory distress), whereas there is a more gradual onset in infants (failure to thrive, tachypnea, indrawing of the respiratory muscles). In older children, the onset is insidious and the diagnosis can only be made at an advanced stage of the disease. The diagnosis is based on noninvasive methods (clinical history, respiratory function tests, chest X-ray, and high-resolution CT scan) and invasive techniques (bronchoalveolar lavage, transbronchial biopsy, video-assisted thoracoscopic biopsy, and open lung biopsy). The treatment of interstitial lung disease in children depends on the nature of the underlying pathology. The most common therapeutic approach involves the use of corticosteroids and immunosuppressive agents for their anti-inflammatory and antifibrotic effects. Children with ILD also need support therapy (oxygen therapy, nutritional support, treatment of pulmonary arterial hypertension, vaccination). Lung transplantation is discussed in patients with severe respiratory failure.
Collapse
Affiliation(s)
- M Fuger
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France.
| | - M-P Clair
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| | - N El Ayoun Ibrahim
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| | - S L'Excellent
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| | - L Nizery
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| | - C O'Neill
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| | - L Tabone
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| | - O Truffinet
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| | - C Yakovleff
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| | - J de Blic
- Service de pneumologie pédiatrique, hôpital Necker-Enfants-Malades, AP-HP, université Paris Descartes, 149, rue de Sèvres, 75015 Paris, France
| |
Collapse
|
33
|
Abstract
Corticosteroids are the most effective treatment for asthma, but the therapeutic response varies markedly between individuals, with up to one third of patients showing evidence of insensitivity to corticosteroids. This article summarizes information on genetic, environmental and asthma-related factors as well as demographic and pharmacokinetic variables associated with corticosteroid insensitivity in asthma. Molecular mechanisms proposed to explain corticosteroid insensitivity are reviewed including alterations in glucocorticoid receptor subtype, binding and nuclear translocation, increased proinflammatory transcription factors and defective histone acetylation. Current therapies and future interventions that may restore corticosteroid sensitivity in asthma are discussed, including small molecule drugs and biological agents. In the future, biomarkers may be used in the clinic to predict corticosteroid sensitivity in patients with poorly controlled asthma.
Collapse
Affiliation(s)
- Neil C Thomson
- a Institute of Infection, Immunity & Inflammation , University of Glasgow , Glasgow , UK
| |
Collapse
|
34
|
Sisignano M, Parnham MJ, Geisslinger G. Drug Repurposing for the Development of Novel Analgesics. Trends Pharmacol Sci 2015; 37:172-183. [PMID: 26706620 DOI: 10.1016/j.tips.2015.11.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 01/12/2023]
Abstract
Drug development consumes huge amounts of time and money and the search for novel analgesics, which are urgently required, is particularly difficult, having resulted in many setbacks in the past. Drug repurposing - the identification of new uses for existing drugs - is an alternative approach, which bypasses most of the time- and cost-consuming components of drug development. Recent, unexpected findings suggest a role for several existing drugs, such as minocycline, ceftriaxone, sivelestat, and pioglitazone, as novel analgesics in chronic and neuropathic pain states. Here, we discuss these findings as well as their proposed antihyperalgesic mechanisms and outline the merits of pathway-based repurposing screens, in combination with bioinformatics and novel cellular reprogramming techniques, for the identification of novel analgesics.
Collapse
Affiliation(s)
- Marco Sisignano
- Institute of Clinical Pharmacology, pharmazentrum Frankfurt/ZAFES, University Hospital of Goethe-University, 60590 Frankfurt am Main, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Project Group Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, pharmazentrum Frankfurt/ZAFES, University Hospital of Goethe-University, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Project Group Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
35
|
Barnes PJ. Therapeutic approaches to asthma-chronic obstructive pulmonary disease overlap syndromes. J Allergy Clin Immunol 2015; 136:531-45. [PMID: 26343937 DOI: 10.1016/j.jaci.2015.05.052] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 12/14/2022]
Abstract
The recognition that there are some patients with features of asthma and chronic obstructive pulmonary disease (COPD) has highlighted the need to develop more specific treatments for these clinical phenotypes. Some patients with COPD have predominantly eosinophilic inflammation and might respond to high doses of inhaled corticosteroids and newly developed specific antieosinophil therapies, including blocking antibodies against IL-5, IL-13, IL-33, and thymic stromal lymphopoietin, as well as oral chemoattractant receptor-homologous molecule expressed on TH2 cells antagonists. Other patients have severe asthma or are asthmatic patients who smoke with features of COPD-induced inflammation and might benefit from treatments targeting neutrophils, including macrolides, CXCR2 antagonists, phosphodiesterase 4 inhibitors, p38 mitogen-activating protein kinase inhibitors, and antibodies against IL-1 and IL-17. Other patients appear to have largely fixed obstruction with little inflammation and might respond to long-acting bronchodilators, including long-acting muscarinic antagonists, to reduce hyperinflation. Highly selected patients with severe asthma might benefit from bronchial thermoplasty. Some patients with overlap syndromes can be conveniently treated with triple fixed-dose combination inhaler therapy with an inhaled corticosteroid, long-acting β2-agonist, and long-acting muscarinic antagonist, several of which are now in development. Corticosteroid resistance is a feature of asthma-COPD overlap syndrome, and understanding the various molecular mechanisms of this resistance has identified novel therapeutic targets and presented the prospect of therapies that can restore corticosteroid responsiveness.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom.
| |
Collapse
|
36
|
Past, present and future of macrolide therapy for chronic rhinosinusitis in Japan. Auris Nasus Larynx 2015; 43:131-6. [PMID: 26441370 DOI: 10.1016/j.anl.2015.08.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/17/2015] [Accepted: 08/26/2015] [Indexed: 01/08/2023]
Abstract
In 1984, the effectiveness of low-dose, long-term erythromycin treatment (macrolide therapy) for diffuse panbronchiolitis (DPB) was first reported in Japan. The 5-year survival rate for DPB improved from 62.9 to 91.4% after implementation of macrolide therapy. The usefulness of this treatment has since been demonstrated in patients with other chronic airway diseases, such as chronic bronchitis, cystic fibrosis, bronchiectasis, bronchial asthma, and chronic rhinosinusitis (CRS). The new 14-membered macrolides clarithromycin and roxithromycin and the 15-membered macrolide azithromycin are also effective for treating these inflammatory diseases. The mechanism of action of the 14- and 15-membered macrolides may involve anti-inflammatory rather than anti-bacterial activities. Macrolide therapy is now widely used for the treatment of CRS in Japan; it is particularly effective for treating neutrophil-associated CRS and is useful for suppressing mucus hypersecretion. However, macrolide therapy is not effective for eosinophil-predominant CRS, which is characterized by serum and tissue eosinophilia, high serum IgE levels, multiple polyposis, and bronchial asthma. Recent reports have described the clinical efficacy of macrolides in treating other inflammatory diseases and new biological activities (e.g., anti-viral). New macrolide derivatives exhibiting anti-inflammatory but not anti-bacterial activity thus have therapeutic potential as immunomodulatory drugs. The history, current state, and future perspectives of macrolide therapy for treating CRS in Japan will be discussed in this review.
Collapse
|
37
|
Sonneville F, Ruffin M, Guillot L, Rousselet N, Le Rouzic P, Corvol H, Tabary O. New insights about miRNAs in cystic fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:897-908. [PMID: 25687559 DOI: 10.1016/j.ajpath.2014.12.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 11/26/2014] [Accepted: 12/01/2014] [Indexed: 01/08/2023]
Abstract
The molecular basis of cystic fibrosis (CF) is a mutation-related defect in the epithelial-cell chloride channel called CF transmembrane conductance regulator (CFTR). This defect alters chloride ion transport and impairs water transport across the cell membrane. Marked clinical heterogeneity occurs even among patients carrying the same mutation in the CFTR gene. Recent studies suggest that such heterogeneity could be related to epigenetic factors and/or miRNAs, which are small noncoding RNAs that modulate the expression of various proteins via post-transcriptional inhibition of gene expression. In the respiratory system, it has been shown that the dysregulation of miRNAs could participate in and lead to pathogenicity in several diseases. In CF airways, recent studies have proposed that miRNAs may modulate disease progression by affecting the production of either CFTR or various proteins that are dysregulated in the CF lung. Herein, we provide an overview of studies showing how miRNAs may modulate CF pathology and the efforts to develop miRNA-based treatments and/or to consider miRNAs as biomarkers. The identification of miRNAs involved in CF disease progression opens up new avenues toward treatments targeting selected clinical components of CF, independently from the CFTR mutation.
Collapse
Affiliation(s)
- Florence Sonneville
- Inserm UMR_S938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, Paris, France
| | - Manon Ruffin
- Inserm UMR_S938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, Paris, France
| | - Loïc Guillot
- Inserm UMR_S938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, Paris, France
| | - Nathalie Rousselet
- Inserm UMR_S938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, Paris, France
| | - Philippe Le Rouzic
- Inserm UMR_S938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, Paris, France
| | - Harriet Corvol
- Inserm UMR_S938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, Paris, France; Paediatric Respiratory Department, Hôpital Trousseau, AP-HP, Paris, France
| | - Olivier Tabary
- Inserm UMR_S938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, Paris, France.
| |
Collapse
|
38
|
Ratzinger F, Haslacher H, Poeppl W, Hoermann G, Kovarik JJ, Jutz S, Steinberger P, Burgmann H, Pickl WF, Schmetterer KG. Azithromycin suppresses CD4(+) T-cell activation by direct modulation of mTOR activity. Sci Rep 2014; 4:7438. [PMID: 25500904 PMCID: PMC4262884 DOI: 10.1038/srep07438] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/21/2014] [Indexed: 12/31/2022] Open
Abstract
Advanced macrolides, such as azithromycin (AZM) or clarithromycin (CLM), are antibiotics with immunomodulatory properties. Here we have sought to evaluate their in vitro influence on the activation of CD4(+) T-cells. Isolated CD4(+) T-cells were stimulated with agonistic anti-CD3/anti-CD28 monoclonal antibodies in the presence of 0.6 mg/L, 2.5 mg/L, 10 mg/L or 40 mg/L AZM or CLM. Cell proliferation, cytokine level in supernatants and cell viability was assessed. Intracellular signaling pathways were evaluated using reporter cell lines, FACS analysis, immunoblotting and in vitro kinase assays. AZM inhibited cell proliferation rate and cytokine secretion of CD4(+) T-cells in a dose-dependent manner. Similarly, high concentrations of CLM (40 mg/L) also suppressed these T-cell functions. Analysis of molecular signaling pathways revealed that exposure to AZM reduced the phosphorylation of the S6 ribosomal protein, a downstream target of mTOR. This effect was also observed at 40 mg/L CLM. In vitro kinase studies using recombinant mTOR showed that AZM inhibited mTOR activity. In contrast to rapamycin, this inhibition was independent of FKBP12. We show for the first time that AZM and to a lesser extent CLM act as immunosuppressive agents on CD4(+) T-cells by inhibiting mTOR activity. Our results might have implications for the clinical use of macrolides.
Collapse
Affiliation(s)
- F. Ratzinger
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - H. Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - W. Poeppl
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Austria
| | - G. Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - J. J. Kovarik
- Clinical Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - S. Jutz
- Institute of Immunology, Medical University of Vienna, Austria
| | - P. Steinberger
- Institute of Immunology, Medical University of Vienna, Austria
| | - H. Burgmann
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Austria
| | - W. F. Pickl
- Institute of Immunology, Medical University of Vienna, Austria
| | - K. G. Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| |
Collapse
|