1
|
Öz HH, Braga CL, Gudneppanavar R, Di Pietro C, Huang PH, Zhang PX, Krause DS, Egan ME, Murray TS, Bruscia EM. CCR2+ monocytes are dispensable to resolve acute pulmonary Pseudomonas aeruginosa infections in WT and cystic fibrosis mice. J Leukoc Biol 2025; 117:qiae218. [PMID: 39365279 PMCID: PMC11953069 DOI: 10.1093/jleuko/qiae218] [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: 08/06/2024] [Revised: 09/04/2024] [Accepted: 10/01/2024] [Indexed: 10/05/2024] Open
Abstract
Extravasation of CCR2-positive monocytes into tissue and to the site of injury is a fundamental immunological response to infections. Nevertheless, exuberant recruitment and/or activity of these monocytes and monocyte-derived macrophages can propagate tissue damage, especially in chronic inflammatory disease conditions. We have previously shown that inhibiting the recruitment of CCR2-positive monocytes ameliorates lung tissue damage caused by chronic neutrophilic inflammation in cystic fibrosis mouse models. A potential concern with targeting monocyte recruitment for therapeutic benefit in cystic fibrosis, however, is whether they are essential for eradicating infections such as Pseudomonas aeruginosa, a pathogen that commonly colonizes and damages the lungs of patients with cystic fibrosis. In this study, we investigated the role of CCR2-positive monocytes in the immune response to acute pulmonary P. aeruginosa infection. Our data show that the altered host immune response caused by the lack of monocyte recruitment to the lungs does not impact P. aeruginosa lung colonization, clearance, and the severity of the infection. These results also hold up in a cystic fibrosis mouse background, which has a hyperinflammatory immune response yet exhibits reduced bactericidal activity. Thus, we lay the groundwork for future studies to investigate the use of CCR2 inhibitors as a potential therapy to ameliorate lung tissue damage in cystic fibrosis. This could be given alone or as an adjunct therapy with CFTR modulators that significantly improve clinical outcomes for eligible patients but do not completely resolve the persistent infection and inflammation that drive lung tissue damage.
Collapse
Affiliation(s)
- Hasan H Öz
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Cassia L Braga
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Ravindra Gudneppanavar
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Caterina Di Pietro
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Pamela H Huang
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Ping-Xia Zhang
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
- Departments of Laboratory Medicine, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Diane S Krause
- Departments of Laboratory Medicine, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
- Departments of Cell Biology, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Marie E Egan
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Thomas S Murray
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
- Departments of Laboratory Medicine, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| | - Emanuela M Bruscia
- Departments of Pediatrics, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, United States
| |
Collapse
|
2
|
Hampton TH, Barnaby R, Roche C, Nymon A, Fukutani KF, MacKenzie TA, Charpentier LA, Stanton BA. Gene expression responses of CF airway epithelial cells exposed to elexacaftor/tezacaftor/ivacaftor suggest benefits beyond improved CFTR channel function. Am J Physiol Lung Cell Mol Physiol 2024; 327:L905-L916. [PMID: 39437760 PMCID: PMC11684945 DOI: 10.1152/ajplung.00272.2024] [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: 08/19/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
The combination of elexacaftor/tezacaftor/ivacaftor (ETI, Trikafta) reverses the primary defect in cystic fibrosis (CF) by improving CFTR-mediated Cl- and HCO3- secretion by airway epithelial cells (AECs), leading to improved lung function and less frequent exacerbations and hospitalizations. However, studies have shown that CFTR modulators like ivacaftor, a component of ETI, have numerous effects on CF cells beyond improved CFTR channel function. Because little is known about the effect of ETI on CF AEC gene expression, we exposed primary human AEC to ETI for 48 h and interrogated the transcriptome by RNA-seq and qPCR. ETI increased CFTR Cl- secretion, and defensin gene expression (DEFB1), an observation consistent with reports of decreased bacterial burden in the lungs of people with CF (pwCF). ETI decreased MMP10 and MMP12 gene expression, suggesting that ETI may reduce proteolytic-induced lung destruction in CF. ETI also reduced the expression of the stress response gene heme oxygenase (HMOX1). qPCR analysis confirmed DEFB1, HMOX1, MMP10, and MMP12 gene expression results observed by RNA-seq. Gene pathway analysis revealed that ETI decreased inflammatory signaling, cellular proliferation, and MHC class II antigen presentation. Collectively, these findings suggest that the clinical observation that ETI reduces lung infections in pwCF is related in part to drug-induced increases in DEFB1 and that ETI may reduce lung damage by reducing MMP10 and MMP12 gene expression. Moreover, pathway analysis also identified several other genes responsible for the ETI-induced reduction in inflammation observed in pwCF.NEW & NOTEWORTHY Gene expression responses by CF AECs exposed to ETI suggest that in addition to improving CFTR channel function, ETI is likely to enhance resistance to bacterial infection by increasing levels of beta-defensin 1 (hBD-1). ETI may also reduce lung damage by suppressing MMP10 and MMP12 and reduce airway inflammation by repressing proinflammatory cytokine secretion by CF AECs.
Collapse
Affiliation(s)
- Thomas H Hampton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Roxanna Barnaby
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Carolyn Roche
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Amanda Nymon
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Kiyoshi Ferreira Fukutani
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Todd A MacKenzie
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States
| | - Lily A Charpentier
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Bruce A Stanton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| |
Collapse
|
3
|
Hampton TH, Barnaby R, Roche C, Nymon A, Fukutani KF, MacKenzie TA, Stanton BA. Gene expression responses of CF airway epithelial cells exposed to elexacaftor/tezacaftor/ivacaftor (ETI) suggest benefits beyond improved CFTR channel function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610162. [PMID: 39257747 PMCID: PMC11383677 DOI: 10.1101/2024.08.28.610162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The combination of elexacaftor/tezacaftor/ivacaftor (ETI, Trikafta) reverses the primary defect in Cystic Fibrosis (CF) by improving CFTR mediated Cl - and HCO 3 - secretion by airway epithelial cells (AEC), leading to improved lung function and less frequent exacerbations and hospitalizations. However, studies have shown that CFTR modulators like ivacaftor, a component of ETI, has numerous effects on CF cells beyond improved CFTR channel function. Because little is known about the effect of ETI on CF AEC gene expression we exposed primary human AEC to ETI for 48 hours and interrogated the transcriptome by RNA-seq and qPCR. ETI increased defensin gene expression ( DEFB1 ) an observation consistent with reports of decreased bacterial burden in the lungs of people with CF (pwCF). ETI also decreased MMP10 and MMP12 gene expression, suggesting that ETI may reduce proteolytic induced lung destruction in CF. ETI also reduced the expression of the stress response gene heme oxygenase ( HMOX1 ). qPCR analysis confirmed DEFB1, HMOX1, MMP10 and MMP12 gene expression results observed by RNA-seq. Gene pathway analysis revealed that ETI decreased inflammatory signaling, cellular proliferation and MHC Class II antigen presentation. Collectively, these findings suggest that the clinical observation that ETI reduces lung infections in pwCF is related in part to drug induced increases in DEFB1 , and that ETI may reduce lung damage by reducing MMP10 and MMP12 gene expression, which is predicted to reduce matrix metalloprotease activity. Moreover, pathway analysis also identified several genes responsible for the ETI induced reduction in inflammation observed in people with CF. New and Noteworthy Gene expression responses by CF AEC exposed to ETI suggest that in addition to improving CFTR channel function, ETI is likely to increase resistance to bacterial infection by increasing levels of beta defensin 1 (hBD-1). ETI may also reduce lung damage by suppressing MMP10, and reduce airway inflammation by repressing proinflammatory cytokine secretion by AEC cells.
Collapse
|
4
|
Vaillancourt M, Galdino ACM, Limsuwannarot SP, Celedonio D, Dimitrova E, Broerman M, Bresee C, Doi Y, Lee JS, Parks WC, Jorth P. A compensatory RNase E variation increases Iron Piracy and Virulence in multidrug-resistant Pseudomonas aeruginosa during Macrophage infection. PLoS Pathog 2023; 19:e1010942. [PMID: 37027441 PMCID: PMC10115287 DOI: 10.1371/journal.ppat.1010942] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 04/19/2023] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
During chronic cystic fibrosis (CF) infections, evolved Pseudomonas aeruginosa antibiotic resistance is linked to increased pulmonary exacerbations, decreased lung function, and hospitalizations. However, the virulence mechanisms underlying worse outcomes caused by antibiotic resistant infections are poorly understood. Here, we investigated evolved aztreonam resistant P. aeruginosa virulence mechanisms. Using a macrophage infection model combined with genomic and transcriptomic analyses, we show that a compensatory mutation in the rne gene, encoding RNase E, increased pyoverdine and pyochelin siderophore gene expression, causing macrophage ferroptosis and lysis. We show that iron-bound pyochelin was sufficient to cause macrophage ferroptosis and lysis, however, apo-pyochelin, iron-bound pyoverdine, or apo-pyoverdine were insufficient to kill macrophages. Macrophage killing could be eliminated by treatment with the iron mimetic gallium. RNase E variants were abundant in clinical isolates, and CF sputum gene expression data show that clinical isolates phenocopied RNase E variant functions during macrophage infection. Together these data show how P. aeruginosa RNase E variants can cause host damage via increased siderophore production and host cell ferroptosis but may also be targets for gallium precision therapy.
Collapse
Affiliation(s)
- Mylene Vaillancourt
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Anna Clara Milesi Galdino
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Sam P. Limsuwannarot
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Diana Celedonio
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Elizabeth Dimitrova
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Matthew Broerman
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Catherine Bresee
- Biostatistics Core, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Yohei Doi
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Janet S. Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - William C. Parks
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Peter Jorth
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| |
Collapse
|
5
|
The Role of Matrix Metalloproteinase in Inflammation with a Focus on Infectious Diseases. Int J Mol Sci 2022; 23:ijms231810546. [PMID: 36142454 PMCID: PMC9500641 DOI: 10.3390/ijms231810546] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are involved in extracellular matrix remodeling through the degradation of extracellular matrix components and are also involved in the inflammatory response by regulating the pro-inflammatory cytokines TNF-α and IL-1β. Dysregulation in the inflammatory response and changes in the extracellular matrix by MMPs are related to the development of various diseases including lung and cardiovascular diseases. Therefore, numerous studies have been conducted to understand the role of MMPs in disease pathogenesis. MMPs are involved in the pathogenesis of infectious diseases through a dysregulation of the activity and expression of MMPs. In this review, we discuss the role of MMPs in infectious diseases and inflammatory responses. Furthermore, we present the potential of MMPs as therapeutic targets in infectious diseases.
Collapse
|
6
|
de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol Rev 2022; 74:712-768. [PMID: 35738680 DOI: 10.1124/pharmrev.121.000349] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The first matrix metalloproteinase (MMP) was discovered in 1962 from the tail of a tadpole by its ability to degrade collagen. As their name suggests, matrix metalloproteinases are proteases capable of remodeling the extracellular matrix. More recently, MMPs have been demonstrated to play numerous additional biologic roles in cell signaling, immune regulation, and transcriptional control, all of which are unrelated to the degradation of the extracellular matrix. In this review, we will present milestones and major discoveries of MMP research, including various clinical trials for the use of MMP inhibitors. We will discuss the reasons behind the failures of most MMP inhibitors for the treatment of cancer and inflammatory diseases. There are still misconceptions about the pathophysiological roles of MMPs and the best strategies to inhibit their detrimental functions. This review aims to discuss MMPs in preclinical models and human pathologies. We will discuss new biochemical tools to track their proteolytic activity in vivo and ex vivo, in addition to future pharmacological alternatives to inhibit their detrimental functions in diseases. SIGNIFICANCE STATEMENT: Matrix metalloproteinases (MMPs) have been implicated in most inflammatory, autoimmune, cancers, and pathogen-mediated diseases. Initially overlooked, MMP contributions can be both beneficial and detrimental in disease progression and resolution. Thousands of MMP substrates have been suggested, and a few hundred have been validated. After more than 60 years of MMP research, there remain intriguing enigmas to solve regarding their biological functions in diseases.
Collapse
Affiliation(s)
- Luiz G N de Almeida
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Hayley Thode
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Yekta Eslambolchi
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sameeksha Chopra
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Daniel Young
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sean Gill
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Laurent Devel
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Antoine Dufour
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| |
Collapse
|
7
|
Chuliá-Peris L, Carreres-Rey C, Gabasa M, Alcaraz J, Carretero J, Pereda J. Matrix Metalloproteinases and Their Inhibitors in Pulmonary Fibrosis: EMMPRIN/CD147 Comes into Play. Int J Mol Sci 2022; 23:ijms23136894. [PMID: 35805895 PMCID: PMC9267107 DOI: 10.3390/ijms23136894] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 02/06/2023] Open
Abstract
Pulmonary fibrosis (PF) is characterized by aberrant extracellular matrix (ECM) deposition, activation of fibroblasts to myofibroblasts and parenchymal disorganization, which have an impact on the biomechanical traits of the lung. In this context, the balance between matrix metalloproteinases (MMPs) and their tissue inhibitors of metalloproteinases (TIMPs) is lost. Interestingly, several MMPs are overexpressed during PF and exhibit a clear profibrotic role (MMP-2, -3, -8, -11, -12 and -28), but a few are antifibrotic (MMP-19), have both profibrotic and antifibrotic capacity (MMP7), or execute an unclear (MMP-1, -9, -10, -13, -14) or unknown function. TIMPs are also overexpressed in PF; hence, the modulation and function of MMPs and TIMP are more complex than expected. EMMPRIN/CD147 (also known as basigin) is a transmembrane glycoprotein from the immunoglobulin superfamily (IgSF) that was first described to induce MMP activity in fibroblasts. It also interacts with other molecules to execute non-related MMP aactions well-described in cancer progression, migration, and invasion. Emerging evidence strongly suggests that CD147 plays a key role in PF not only by MMP induction but also by stimulating fibroblast myofibroblast transition. In this review, we study the structure and function of MMPs, TIMPs and CD147 in PF and their complex crosstalk between them.
Collapse
Affiliation(s)
- Lourdes Chuliá-Peris
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain; (L.C.-P.); (C.C.-R.); (J.C.)
| | - Cristina Carreres-Rey
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain; (L.C.-P.); (C.C.-R.); (J.C.)
| | - Marta Gabasa
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (M.G.); (J.A.)
| | - Jordi Alcaraz
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (M.G.); (J.A.)
- Thoracic Oncology Unit, Hospital Clinic Barcelona, 08036 Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Spain
| | - Julián Carretero
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain; (L.C.-P.); (C.C.-R.); (J.C.)
| | - Javier Pereda
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain; (L.C.-P.); (C.C.-R.); (J.C.)
- Correspondence:
| |
Collapse
|
8
|
Liao L, Dang W, Lin T, Yu J, Liu T, Li W, Xiao S, Feng L, Huang J, Fu R, Li J, Liu L, Wang M, Tao H, Jiang H, Chen K, Diao X, Zhou B, Shen X, Luo C. A potent PGK1 antagonist reveals PGK1 regulates the production of IL-1β and IL-6. Acta Pharm Sin B 2022; 12:4180-4192. [PMID: 36386479 PMCID: PMC9643279 DOI: 10.1016/j.apsb.2022.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/02/2022] Open
Abstract
Glycolytic metabolism enzymes have been implicated in the immunometabolism field through changes in metabolic status. PGK1 is a catalytic enzyme in the glycolytic pathway. Here, we set up a high-throughput screen platform to identify PGK1 inhibitors. DC-PGKI is an ATP-competitive inhibitor of PGK1 with an affinity of Kd = 99.08 nmol/L. DC-PGKI stabilizes PGK1 in vitro and in vivo, and suppresses both glycolytic activity and the kinase function of PGK1. In addition, DC-PGKI unveils that PGK1 regulates production of IL-1β and IL-6 in LPS-stimulated macrophages. Mechanistically, inhibition of PGK1 with DC-PGKI results in NRF2 (nuclear factor-erythroid factor 2-related factor 2, NFE2L2) accumulation, then NRF2 translocates to the nucleus and binds to the proximity region of Il-1β and Il-6 genes, and inhibits LPS-induced expression of these genes. DC-PGKI ameliorates colitis in the dextran sulfate sodium (DSS)-induced colitis mouse model. These data support PGK1 as a regulator of macrophages and suggest potential utility of PGK1 inhibitors in the treatment of inflammatory bowel disease.
Collapse
|
9
|
Raeeszadeh-Sarmazdeh M, Coban M, Mahajan S, Hockla A, Sankaran B, Downey GP, Radisky DC, Radisky ES. Engineering of tissue inhibitor of metalloproteinases TIMP-1 for fine discrimination between closely-related stromelysins MMP-3 and MMP-10. J Biol Chem 2022; 298:101654. [PMID: 35101440 PMCID: PMC8902619 DOI: 10.1016/j.jbc.2022.101654] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 12/03/2022] Open
Abstract
Matrix metalloproteinases (MMPs) have long been known as key drivers in the development and progression of diseases, including cancer and neurodegenerative, cardiovascular, and many other inflammatory and degenerative diseases, making them attractive potential drug targets. Engineering selective inhibitors based upon tissue inhibitors of metalloproteinases (TIMPs), endogenous human proteins that tightly yet nonspecifically bind to the family of MMPs, represents a promising new avenue for therapeutic development. Here, we used a counter-selective screening strategy for directed evolution of yeast-displayed human TIMP-1 to obtain TIMP-1 variants highly selective for the inhibition of MMP-3 in preference over MMP-10. As MMP-3 and MMP-10 are the most similar MMPs in sequence, structure, and function, our results thus clearly demonstrate the capability for engineering full-length TIMP proteins to be highly selective MMP inhibitors. We show using protein crystal structures and models of MMP-3-selective TIMP-1 variants bound to MMP-3 and counter-target MMP-10 how structural alterations within the N-terminal and C-terminal TIMP-1 domains create new favorable and selective interactions with MMP-3 and disrupt unique interactions with MMP-10. While our MMP-3-selective inhibitors may be of interest for future investigation in diseases where this enzyme drives pathology, our platform and screening strategy can be employed for developing selective inhibitors of additional MMPs implicated as therapeutic targets in disease.
Collapse
Affiliation(s)
| | - Mathew Coban
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224
| | - Shivansh Mahajan
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224
| | - Alexandra Hockla
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging, Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Gregory P Downey
- Departments of Medicine, Pediatrics, and Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado 80206; Departments of Medicine, and Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224.
| |
Collapse
|
10
|
Shukla P, Mandalla A, Elrick MJ, Venkatesan A. Clinical Manifestations and Pathogenesis of Acute Necrotizing Encephalopathy: The Interface Between Systemic Infection and Neurologic Injury. Front Neurol 2022; 12:628811. [PMID: 35058867 PMCID: PMC8764155 DOI: 10.3389/fneur.2021.628811] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Acute necrotizing encephalopathy (ANE) is a devastating neurologic condition that can arise following a variety of systemic infections, including influenza and SARS-CoV-2. Affected individuals typically present with rapid changes in consciousness, focal neurological deficits, and seizures. Neuroimaging reveals symmetric, bilateral deep gray matter lesions, often involving the thalami, with evidence of necrosis and/or hemorrhage. The clinical and radiologic picture must be distinguished from direct infection of the central nervous system by some viruses, and from metabolic and mitochondrial disorders. Outcomes following ANE are poor overall and worse in those with brainstem involvement. Specific management is often directed toward modulating immune responses given the potential role of systemic inflammation and cytokine storm in potentiating neurologic injury in ANE, though benefits of such approaches remain unclear. The finding that many patients have mutations in the nucleoporin gene RANBP2, which encodes a multifunctional protein that plays a key role in nucleocytoplasmic transport, may allow for the development of disease models that provide insights into pathogenic mechanisms and novel therapeutic approaches.
Collapse
Affiliation(s)
- Priya Shukla
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Abby Mandalla
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Matthew J Elrick
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Arun Venkatesan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
11
|
Swaminathan G, Citron M, Xiao J, Norton JE, Reens AL, Topçuoğlu BD, Maritz JM, Lee KJ, Freed DC, Weber TM, White CH, Kadam M, Spofford E, Bryant-Hall E, Salituro G, Kommineni S, Liang X, Danilchanka O, Fontenot JA, Woelk CH, Gutierrez DA, Hazuda DJ, Hannigan GD. Vaccine Hyporesponse Induced by Individual Antibiotic Treatment in Mice and Non-Human Primates Is Diminished upon Recovery of the Gut Microbiome. Vaccines (Basel) 2021; 9:vaccines9111340. [PMID: 34835271 PMCID: PMC8619314 DOI: 10.3390/vaccines9111340] [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] [Received: 08/31/2021] [Revised: 10/19/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
Emerging evidence demonstrates a connection between microbiome composition and suboptimal response to vaccines (vaccine hyporesponse). Harnessing the interaction between microbes and the immune system could provide novel therapeutic strategies for improving vaccine response. Currently we do not fully understand the mechanisms and dynamics by which the microbiome influences vaccine response. Using both mouse and non-human primate models, we report that short-term oral treatment with a single antibiotic (vancomycin) results in the disruption of the gut microbiome and this correlates with a decrease in systemic levels of antigen-specific IgG upon subsequent parenteral vaccination. We further show that recovery of microbial diversity before vaccination prevents antibiotic-induced vaccine hyporesponse, and that the antigen specific IgG response correlates with the recovery of microbiome diversity. RNA sequencing analysis of small intestine, spleen, whole blood, and secondary lymphoid organs from antibiotic treated mice revealed a dramatic impact on the immune system, and a muted inflammatory signature is correlated with loss of bacteria from Lachnospiraceae, Ruminococcaceae, and Clostridiaceae. These results suggest that microbially modulated immune pathways may be leveraged to promote vaccine response and will inform future vaccine design and development strategies.
Collapse
Affiliation(s)
- Gokul Swaminathan
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
- Correspondence: (G.S.); (G.D.H.)
| | - Michael Citron
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - Jianying Xiao
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - James E. Norton
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Abigail L. Reens
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Begüm D. Topçuoğlu
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Julia M. Maritz
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Keun-Joong Lee
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, MRL, Merck & Co. Inc., Rahway, NJ 07065, USA; (K.-J.L.); (G.S.)
| | - Daniel C. Freed
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - Teresa M. Weber
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - Cory H. White
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Mahika Kadam
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Erin Spofford
- Safety Assessment and Laboratory Animal Research, MRL, Merck & Co. Inc., Boston, MA 02115, USA; (E.S.); (E.B.-H.)
| | - Erin Bryant-Hall
- Safety Assessment and Laboratory Animal Research, MRL, Merck & Co. Inc., Boston, MA 02115, USA; (E.S.); (E.B.-H.)
| | - Gino Salituro
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, MRL, Merck & Co. Inc., Rahway, NJ 07065, USA; (K.-J.L.); (G.S.)
| | - Sushma Kommineni
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Xue Liang
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Olga Danilchanka
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Jane A. Fontenot
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, LA 70503, USA;
| | - Christopher H. Woelk
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Dario A. Gutierrez
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Daria J. Hazuda
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - Geoffrey D. Hannigan
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
- Correspondence: (G.S.); (G.D.H.)
| |
Collapse
|
12
|
Guizani I, Fourti N, Zidi W, Feki M, Allal-Elasmi M. SARS-CoV-2 and pathological matrix remodeling mediators. Inflamm Res 2021; 70:847-858. [PMID: 34286362 PMCID: PMC8294315 DOI: 10.1007/s00011-021-01487-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Recognizing only sharp elevation in a short period of time, the COVID-19 SARS-CoV-2 propagation is more and more marked in the whole world. Induced inflammation afterwards infection engenders a high infiltration of immune cells and cytokines that triggers matrix metalloproteinases (MMPs) activation. These endopeptidases are mediators of the lung extracellular matrix (ECM), a basic element for alveoli structure and gas exchange. METHODS When immune cells, MMPs, secreted cytokines and several other mediators are gathered a pathological matrix remodeling occurs. This phenomenon tends to tissue destruction in the first place and a pulmonary hypertrophy and fibrosis in the second place. FINDINGS After pathological matrix remodeling establishment, pathological diseases take place even after infection state. Since post COVID-19 pulmonary fibrosis is an emerging complication of the disease, there is an urge to better understand and characterize the implication of ECM remodeling during SARS-CoV-2 infection. CONCLUSION Targeting MMPs and their inhibitors could be a probable solution for occurred events since there are many cured patients that remain with severe sequels even after the end of infection.
Collapse
Affiliation(s)
- Imen Guizani
- LR99ES11, Laboratory of Biochemistry, Department of Biochemistry, Faculty of Medicine, La Rabta Hospital, University of Tunis El Manar, Jebbari, 1007, Tunis, Tunisia
- Faculty of Mathematics, Physics and Natural Sciences, University of Tunis El Manar, Tunis, Tunisia
| | - Nesrine Fourti
- LR99ES11, Laboratory of Biochemistry, Department of Biochemistry, Faculty of Medicine, La Rabta Hospital, University of Tunis El Manar, Jebbari, 1007, Tunis, Tunisia
- Faculty of Mathematics, Physics and Natural Sciences, University of Tunis El Manar, Tunis, Tunisia
| | - Wiem Zidi
- LR99ES11, Laboratory of Biochemistry, Department of Biochemistry, Faculty of Medicine, La Rabta Hospital, University of Tunis El Manar, Jebbari, 1007, Tunis, Tunisia
| | - Moncef Feki
- LR99ES11, Laboratory of Biochemistry, Department of Biochemistry, Faculty of Medicine, La Rabta Hospital, University of Tunis El Manar, Jebbari, 1007, Tunis, Tunisia
| | - Monia Allal-Elasmi
- LR99ES11, Laboratory of Biochemistry, Department of Biochemistry, Faculty of Medicine, La Rabta Hospital, University of Tunis El Manar, Jebbari, 1007, Tunis, Tunisia.
| |
Collapse
|
13
|
Gerber A, Goldklang M, Stearns K, Ma X, Xiao R, Zelonina T, D'Armiento J. Attenuation of pulmonary injury by an inhaled MMP inhibitor in the endotoxin lung injury model. Am J Physiol Lung Cell Mol Physiol 2020; 319:L1036-L1047. [PMID: 33026238 DOI: 10.1152/ajplung.00420.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by pulmonary edema and poor gas exchange resulting from severe inflammatory lung injury. Neutrophilic infiltration and increased pulmonary vascular permeability are hallmarks of early ARDS and precipitate a self-perpetuating cascade of inflammatory signaling. The biochemical processes initiating these events remain unclear. Typically associated with extracellular matrix degradation, recent data suggest matrix metalloproteinases (MMPs) are regulators of pulmonary inflammation. To demonstrate that inhalation of a broad MMP inhibitor attenuates LPS induced pulmonary inflammation. Nebulized CGS27023AM (CGS) was administered to LPS-injured mice. Pulmonary CGS levels were examined by mass spectroscopy. Inflammatory scoring of hematoxylin-eosin sections, examination of vascular integrity via lung wet/dry and bronchoalveolar lvage/serum FITC-albumin ratios were performed. Cleaved caspase-3 levels were also assessed. Differential cell counts and pulse-chase labeling were utilized to determine the effects of CGS on neutrophil migration. The effects of CGS on human neutrophil migration and viability were examined using Boyden chambers and MTT assays. Nebulization successfully delivered CGS to the lungs. Treatment decreased pulmonary inflammatory scores, edema, and apoptosis in LPS treated animals. Neutrophil chemotaxis was reduced by CGS treatment, with inhalation causing significant reductions in both the total number and newly produced bromodeoxyuridine-positive cells infiltrating the lung. Mechanistic studies on cells isolated from humans demonstrate that CGS-treated neutrophils exhibit decreased chemotaxis. The protective effect observed following treatment with a nonspecific MMP inhibitor indicates that one or more MMPs mediate the development of pulmonary edema and neutrophil infiltration in response to LPS injury. In accordance with this, inhaled MMP inhibitors warrant further study as a potential new therapeutic avenue for treatment of acute lung injury.
Collapse
Affiliation(s)
- Adam Gerber
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York
| | - Monica Goldklang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York
| | - Kyle Stearns
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York
| | - Xinran Ma
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York
| | - Rui Xiao
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York
| | - Tina Zelonina
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York
| | - Jeanine D'Armiento
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
14
|
Kaczorowska A, Miękus N, Stefanowicz J, Adamkiewicz-Drożyńska E. Selected Matrix Metalloproteinases (MMP-2, MMP-7) and Their Inhibitor (TIMP-2) in Adult and Pediatric Cancer. Diagnostics (Basel) 2020; 10:diagnostics10080547. [PMID: 32751899 PMCID: PMC7460349 DOI: 10.3390/diagnostics10080547] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) consists of numerous biologically relevant elements. One of the most important components of the TME is the extracellular matrix (ECM). The compounds of the ECM create a network that provides structural and biochemical support to surrounding cells. The most important substances involved in the regulation of the ECM degradation process are matrix metalloproteinases (MMPs) and their endogenous inhibitors (tissue inhibitors of metalloproteinases, TIMPs). The disruption of the physiological balance between MMP activation and deactivation could lead to progression of various diseases such as cardiovascular disease, cancer, fibrosis arthritis, chronic tissue ulcers, pathologies of the nervous system (such as stroke and Alzheimer's disease), periodontitis, and atheroma. MMP-TIMP imbalance results in matrix proteolysis associated with various pathological processes such as tumor invasion. The present review discusses the involvement of two MMPs, MMP-2 and MMP-7, in cancer pathogenesis. These two MMPs have been proven in several studies, conducted mostly on adults, to make an important contribution to cancer development and progression. In the current review, several studies that indicate the importance of MMP-TIMP balance determination for the pediatric population are also highlighted. The authors of this review believe that carrying out biochemical and clinical studies focused on metalloproteinases and their inhibitors in tumors in children will be of great relevance for future patient diagnosis, determination of a prognosis, and monitoring of therapy.
Collapse
Affiliation(s)
- Aleksandra Kaczorowska
- Department of Pediatrics, Hematology and Oncology, Faculty of Medicine, Medical University of Gdańsk, 7 Dębinki Street, 80-952 Gdańsk, Poland; (A.K.); (E.A.-D.)
- University Clinical Centre, 7 Debinki Street, 80-952 Gdansk, Poland
| | - Natalia Miękus
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland;
| | - Joanna Stefanowicz
- Department of Pediatrics, Hematology and Oncology, Faculty of Medicine, Medical University of Gdańsk, 7 Dębinki Street, 80-952 Gdańsk, Poland; (A.K.); (E.A.-D.)
- University Clinical Centre, 7 Debinki Street, 80-952 Gdansk, Poland
- Faculty of Health Sciences, Medical University of Gdańsk, Maria Sklodowska-Curie Street 3a, 80-210 Gdańsk, Poland
- Correspondence: ; Tel.: +48-58-349-28-08
| | - Elżbieta Adamkiewicz-Drożyńska
- Department of Pediatrics, Hematology and Oncology, Faculty of Medicine, Medical University of Gdańsk, 7 Dębinki Street, 80-952 Gdańsk, Poland; (A.K.); (E.A.-D.)
- University Clinical Centre, 7 Debinki Street, 80-952 Gdansk, Poland
| |
Collapse
|
15
|
BIG1 controls macrophage pro-inflammatory responses through ARF3-mediated PI(4,5)P2 synthesis. Cell Death Dis 2020; 11:374. [PMID: 32415087 PMCID: PMC7229175 DOI: 10.1038/s41419-020-2590-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/29/2020] [Indexed: 12/29/2022]
Abstract
Sepsis is caused by a dysregulated host inflammatory response to serious infections resulting in life-threatening organ dysfunction. The high morbidity and mortality make sepsis still a major clinical problem. Here, we investigated the roles of Brefeldin A-inhibited guanine nucleotide-exchange factor 1 (BIG1) in the pathogenesis process of sepsis and the underlying mechanisms. We found myeloid cell-specific BIG1 knockout (BIG1 cKO) significantly reduced the mortality and organ damage in LPS-induced and CLP-induced polymicrobial sepsis mouse model. The serum concentration and mRNA expression of pro-inflammatory cytokines including TNF-α, IL-6, IL-1β, and IL-12 were obviously decreased in BIG1 cKO mice. In bone marrow-derived macrophages or THP-1 cells, BIG1 deficiency caused an inhibited ARF3 activation, which reduced PI(4,5)P2 synthesis and the recruitment of TIRAP to the plasma membrane through inhibiting the activation of PIP5K induced by LPS, and eventually resulted in the inhibitory activity of TLR4-MyD88 signaling pathway. These results reveal a crucial new role of BIG1 in regulating macrophage inflammation responses, and provide evidence for BIG1 as a potential promising therapeutic target in sepsis.
Collapse
|
16
|
Morrell ED, Mikacenic C, Gong KQ, Kosamo S, Wurfel MM, Manicone AM. Alveolar MMP28 is associated with clinical outcomes and measures of lung injury in acute respiratory distress syndrome. Crit Care 2020; 24:141. [PMID: 32268921 PMCID: PMC7144344 DOI: 10.1186/s13054-020-02847-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/23/2020] [Indexed: 01/08/2023] Open
Affiliation(s)
- Eric D Morrell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Lung Biology, University of Washington, 850 Republican St, Seattle, WA, 98109, USA
| | - Carmen Mikacenic
- Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Lung Biology, University of Washington, 850 Republican St, Seattle, WA, 98109, USA
| | - Ke-Qin Gong
- Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Lung Biology, University of Washington, 850 Republican St, Seattle, WA, 98109, USA
| | - Susanna Kosamo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Lung Biology, University of Washington, 850 Republican St, Seattle, WA, 98109, USA
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Lung Biology, University of Washington, 850 Republican St, Seattle, WA, 98109, USA
| | - Anne M Manicone
- Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Lung Biology, University of Washington, 850 Republican St, Seattle, WA, 98109, USA.
| |
Collapse
|
17
|
de Jong JM, Wang P, Oomkens M, Baron W. Remodeling of the interstitial extracellular matrix in white matter multiple sclerosis lesions: Implications for remyelination (failure). J Neurosci Res 2020; 98:1370-1397. [PMID: 31965607 DOI: 10.1002/jnr.24582] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/29/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) provides protection, rigidity, and structure toward cells. It consists, among others, of a wide variety of glycoproteins and proteoglycans, which act together to produce a complex and dynamic environment, most relevant in transmembrane events. In the brain, the ECM occupies a notable proportion of its volume and maintains the homeostasis of central nervous system (CNS). In addition, remodeling of the ECM, that is transient changes in ECM proteins regulated by matrix metalloproteinases (MMPs), is an important process that modulates cell behavior upon injury, thereby facilitating recovery. Failure of ECM remodeling plays an important role in the pathogenesis of multiple sclerosis (MS), a neurodegenerative demyelinating disease of the CNS with an inflammatory response against protective myelin sheaths that surround axons. Remyelination of denuded axons improves the neuropathological conditions of MS, but this regeneration process fails over time, leading to chronic disease progression. In this review, we uncover abnormal ECM remodeling in MS lesions by discussing ECM remodeling in experimental demyelination models, that is when remyelination is successful, and compare alterations in ECM components to the ECM composition and MMP expression in the parenchyma of demyelinated MS lesions, that is when remyelination fails. Inter- and intralesional differences in ECM remodeling in the distinct white matter MS lesions are discussed in terms of consequences for oligodendrocyte behavior and remyelination (failure). Hence, the review will aid to understand how abnormal ECM remodeling contributes to remyelination failure in MS lesions and assists in developing therapeutic strategies to promote remyelination.
Collapse
Affiliation(s)
- Jody M de Jong
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peng Wang
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michelle Oomkens
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wia Baron
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
18
|
Rosowski EE. Determining macrophage versus neutrophil contributions to innate immunity using larval zebrafish. Dis Model Mech 2020; 13:13/1/dmm041889. [PMID: 31932292 PMCID: PMC6994940 DOI: 10.1242/dmm.041889] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The specific roles of the two major innate immune cell types – neutrophils and macrophages – in response to infection and sterile inflammation are areas of great interest. The larval zebrafish model of innate immunity, and the imaging capabilities it provides, is a source of new research and discoveries in this field. Multiple methods have been developed in larval zebrafish to specifically deplete functional macrophages or neutrophils. Each of these has pros and cons, as well as caveats, that often make it difficult to directly compare results from different studies. The purpose of this Review is to (1) explore the pros, cons and caveats of each of these immune cell-depleted models; (2) highlight and place into a broader context recent key findings on the specific functions of innate immune cells using these models; and (3) explore future directions in which immune cell depletion methods are being expanded. Summary: Macrophages and neutrophils are distinct innate immune cells with diverse roles in diverse inflammatory contexts. Recent research in larval zebrafish using cell-specific depletion methods has revealed new insights into these cells' functions.
Collapse
Affiliation(s)
- Emily E Rosowski
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
19
|
Cho KI, Yoon D, Yu M, Peachey NS, Ferreira PA. Microglial activation in an amyotrophic lateral sclerosis-like model caused by Ranbp2 loss and nucleocytoplasmic transport impairment in retinal ganglion neurons. Cell Mol Life Sci 2019; 76:3407-3432. [PMID: 30944974 PMCID: PMC6698218 DOI: 10.1007/s00018-019-03078-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/21/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022]
Abstract
Nucleocytoplasmic transport is dysregulated in sporadic and familial amyotrophic lateral sclerosis (ALS) and retinal ganglion neurons (RGNs) are purportedly involved in ALS. The Ran-binding protein 2 (Ranbp2) controls rate-limiting steps of nucleocytoplasmic transport. Mice with Ranbp2 loss in Thy1+-motoneurons develop cardinal ALS-like motor traits, but the impairments in RGNs and the degree of dysfunctional consonance between RGNs and motoneurons caused by Ranbp2 loss are unknown. This will help to understand the role of nucleocytoplasmic transport in the differential vulnerability of neuronal cell types to ALS and to uncover non-motor endophenotypes with pathognomonic signs of ALS. Here, we ascertain Ranbp2's function and endophenotypes in RGNs of an ALS-like mouse model lacking Ranbp2 in motoneurons and RGNs. Thy1+-RGNs lacking Ranbp2 shared with motoneurons the dysregulation of nucleocytoplasmic transport. RGN abnormalities were comprised morphologically by soma hypertrophy and optic nerve axonopathy and physiologically by a delay of the visual pathway's evoked potentials. Whole-transcriptome analysis showed restricted transcriptional changes in optic nerves that were distinct from those found in sciatic nerves. Specifically, the level and nucleocytoplasmic partition of the anti-apoptotic and novel substrate of Ranbp2, Pttg1/securin, were dysregulated. Further, acetyl-CoA carboxylase 1, which modulates de novo synthesis of fatty acids and T-cell immunity, showed the highest up-regulation (35-fold). This effect was reflected by the activation of ramified CD11b+ and CD45+-microglia, increase of F4\80+-microglia and a shift from pseudopodial/lamellipodial to amoeboidal F4\80+-microglia intermingled between RGNs of naive mice. Further, there was the intracellular sequestration in RGNs of metalloproteinase-28, which regulates macrophage recruitment and polarization in inflammation. Hence, Ranbp2 genetic insults in RGNs and motoneurons trigger distinct paracrine signaling likely by the dysregulation of nucleocytoplasmic transport of neuronal-type selective substrates. Immune-modulators underpinning RGN-to-microglial signaling are regulated by Ranbp2, and this neuronal-glial system manifests endophenotypes that are likely useful in the prognosis and diagnosis of motoneuron diseases, such as ALS.
Collapse
Affiliation(s)
- Kyoung-In Cho
- Department of Ophthalmology, Duke University Medical Center, DUEC 3802, 2351 Erwin Road, Durham, NC, 27710, USA
| | - Dosuk Yoon
- Department of Ophthalmology, Duke University Medical Center, DUEC 3802, 2351 Erwin Road, Durham, NC, 27710, USA
| | - Minzhong Yu
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Neal S Peachey
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
- Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Paulo A Ferreira
- Department of Ophthalmology, Duke University Medical Center, DUEC 3802, 2351 Erwin Road, Durham, NC, 27710, USA.
| |
Collapse
|
20
|
Maldonado M, Salgado-Aguayo A, Herrera I, Cabrera S, Ortíz-Quintero B, Staab-Weijnitz CA, Eickelberg O, Ramírez R, Manicone AM, Selman M, Pardo A. Upregulation and Nuclear Location of MMP28 in Alveolar Epithelium of Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2019; 59:77-86. [PMID: 29373068 DOI: 10.1165/rcmb.2017-0223oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive aging-associated disease of unknown etiology. A growing body of evidence indicates that aberrant activated alveolar epithelial cells induce the expansion and activation of the fibroblast population, leading to the destruction of the lung architecture. Some matrix metalloproteinases (MMPs) are upregulated in IPF, indicating that they may be important in the pathogenesis and/or progression of IPF. In the present study, we examined the expression of MMP28 in this disease and evaluated its functional effects in two alveolar epithelial cell lines and in human primary bronchial epithelial cells. We found that the enzyme is expressed in bronchial (apical and cytoplasmic localization) and alveolar (cytoplasmic and nuclear localization) epithelial cells in two different groups of patients with IPF. In vitro MMP28 epithelial silencing decreased the proliferation rate and delayed wound closing, whereas overexpression showed opposite effects, protecting from apoptosis and enhanced epithelial-mesenchymal transition. Our findings demonstrate that MMP28 is upregulated in epithelial cells from IPF lungs, where it may play a role in increasing the proliferative and migratory phenotype in a catalysis-dependent manner.
Collapse
Affiliation(s)
- Mariel Maldonado
- 1 Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Alfonso Salgado-Aguayo
- 2 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Iliana Herrera
- 2 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Sandra Cabrera
- 1 Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Blanca Ortíz-Quintero
- 2 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Claudia A Staab-Weijnitz
- 3 Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Oliver Eickelberg
- 3 Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany.,4 Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, Colorado; and
| | - Remedios Ramírez
- 1 Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Anne M Manicone
- 5 Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Moisés Selman
- 2 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Annie Pardo
- 1 Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
21
|
Li Y, Sun Q, Jiang M, Li S, Zhang J, Xu Z, Guo D, Gu T, Wang B, Xiao L, Zhou T, Zhuo W. KLF9 suppresses gastric cancer cell invasion and metastasis through transcriptional inhibition of MMP28. FASEB J 2019; 33:7915-7928. [PMID: 30913394 DOI: 10.1096/fj.201802531r] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Yang Li
- College of Animal ScienceZhejiang University Hangzhou China
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Qiang Sun
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Mingchun Jiang
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Shuang Li
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Jiayu Zhang
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Zhangqi Xu
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Dongyang Guo
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Tianning Gu
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Boya Wang
- Department of PharmacySir Run Run Shaw HospitalZhejiang University School of Medicine Hangzhou China
| | - Lei Xiao
- College of Animal ScienceZhejiang University Hangzhou China
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Tianhua Zhou
- Institute of GastroenterologyZhejiang University Hangzhou China
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases Hangzhou China
| | - Wei Zhuo
- Institute of GastroenterologyZhejiang University Hangzhou China
- Department of Cell BiologyCancer Institute of the Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| |
Collapse
|
22
|
Liang X, Gupta K, Quintero JR, Cernadas M, Kobzik L, Christou H, Pier GB, Owen CA, Çataltepe S. Macrophage FABP4 is required for neutrophil recruitment and bacterial clearance in Pseudomonas aeruginosa pneumonia. FASEB J 2019; 33:3562-3574. [PMID: 30462529 PMCID: PMC6988858 DOI: 10.1096/fj.201802002r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/15/2018] [Indexed: 01/29/2023]
Abstract
Fatty acid binding protein 4 (FABP4), an intracellular lipid chaperone and adipokine, is expressed by lung macrophages, but the function of macrophage-FABP4 remains elusive. We investigated the role of FABP4 in host defense in a murine model of Pseudomonas aeruginosa pneumonia. Compared with wild-type (WT) mice, FABP4-deficient (FABP4-/-) mice exhibited decreased bacterial clearance and increased mortality when challenged intranasally with P. aeruginosa. These findings in FABP4-/- mice were associated with a delayed neutrophil recruitment into the lungs and were followed by greater acute lung injury and inflammation. Among leukocytes, only macrophages expressed FABP4 in WT mice with P. aeruginosa pneumonia. Chimeric FABP4-/- mice with WT bone marrow were protected from increased mortality seen in chimeric WT mice with FABP4-/- bone marrow during P. aeruginosa pneumonia, thus confirming the role of macrophages as the main source of protective FABP4 against that infection. There was less production of C-X-C motif chemokine ligand 1 (CXCL1) in FABP4-/- alveolar macrophages and lower airway CXCL1 levels in FABP4-/- mice. Delivering recombinant CXCL1 to the airways protected FABP4-/- mice from increased susceptibility to P. aeruginosa pneumonia. Thus, macrophage-FABP4 has a novel role in pulmonary host defense against P. aeruginosa infection by facilitating crosstalk between macrophages and neutrophils via regulation of macrophage CXCL1 production.-Liang, X., Gupta, K., Rojas Quintero, J., Cernadas, M., Kobzik, L., Christou, H., Pier, G. B., Owen, C. A., Çataltepe, S. Macrophage FABP4 is required for neutrophil recruitment and bacterial clearance in Pseudomonas aeruginosa pneumonia.
Collapse
Affiliation(s)
- Xiaoliang Liang
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kushagra Gupta
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joselyn Rojas Quintero
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manuela Cernadas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lester Kobzik
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gerald B. Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA; and
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | - Sule Çataltepe
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Rohani MG, Dimitrova E, Beppu A, Wang Y, Jefferies CA, Parks WC. Macrophage MMP10 Regulates TLR7-Mediated Tolerance. Front Immunol 2018; 9:2817. [PMID: 30564235 PMCID: PMC6288447 DOI: 10.3389/fimmu.2018.02817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023] Open
Abstract
Using an in vivo model of tolerance to TLR7-induced skin inflammation, we found a critical role for macrophage-derived MMP10 in mediating immune hypo-responsiveness. Cutaneous exposure to Imiquimod (IMQ), a TLR7 agonist, induced acute expression of pro-inflammatory factors (IL1β, IL6, CXCL1) and neutrophil influx equally in both wildtype and Mmp10 -/- mice. However, whereas subsequent exposure (11 and 12 days later) to IMQ led to marked abrogation of pro-inflammatory factor expression in wildtype mice, Mmp10 -/- mice responded similarly as they did to the first application. In addition, the second exposure led to increased expression of negative regulators of TLR signaling (TNFAIP3, IRAK3) and immunosuppressive cytokines (IL10, TGFβ1) in wildtype mice but not in Mmp10 -/- mice. In vitro studies demonstrated that prior exposure of IMQ to bone marrow-derived macrophages (BMDM) made wildtype cells refractory to subsequent stimulation but did not for Mmp10 -/- macrophages. These findings expand the critical roles MMP10 plays in controlling macrophage activation to indicate that the development of immune tolerance to TLR7 ligand is dependent on this macrophage-derived proteinase.
Collapse
Affiliation(s)
- Maryam G Rohani
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Elizabeth Dimitrova
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Andrew Beppu
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ying Wang
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Caroline A Jefferies
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - William C Parks
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
24
|
Transcriptional and functional diversity of human macrophage repolarization. J Allergy Clin Immunol 2018; 143:1536-1548. [PMID: 30445062 DOI: 10.1016/j.jaci.2018.10.046] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/17/2018] [Accepted: 10/10/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Macrophage plasticity allows cells to adopt different phenotypes, a property with important implications in disorders such as cystic fibrosis (CF) and asthma. OBJECTIVE We sought to examine the transcriptional and functional significance of macrophage repolarization from an M1 to an M2 phenotype and assess the role of a common human genetic disorder (CF) and a prototypical allergic disease (asthma) in this transformation. METHODS Monocyte-derived macrophages were collected from healthy subjects and patients with CF and polarized to an M2 state by using IL-4, IL-10, glucocorticoids, apoptotic PMNs, or azithromycin. We performed transcriptional profiling and pathway analysis for each stimulus. We assessed the ability of M2-repolarized macrophages to respond to LPS rechallenge and clear apoptotic neutrophils and used murine models to determine conserved functional responses to IL-4 and IL-10. We investigated whether M2 signatures were associated with alveolar macrophage phenotypes in asthmatic patients. RESULTS We found that macrophages exhibit highly diverse responses to distinct M2-polarizing stimuli. Specifically, IL-10 activated proinflammatory pathways and abrogated LPS tolerance, allowing rapid restoration of LPS responsiveness. In contrast, IL-4 enhanced LPS tolerance, dampening proinflammatory responses after repeat LPS challenge. A common theme observed across all M2 stimuli was suppression of interferon-associated pathways. We found that CF macrophages had intact reparative and transcriptional responses, suggesting that macrophage contributions to CF-related lung disease are primarily shaped by their environment. Finally, we leveraged in vitro-derived signatures to show that allergen provocation induces distinct M2 state transcriptional patterns in alveolar macrophages. CONCLUSION Our findings highlight the diversity of macrophage polarization, attribute functional consequences to different M2 stimuli, and provide a framework to phenotype macrophages in disease states.
Collapse
|
25
|
Tarique AA, Sly PD, Cardenas DG, Luo L, Stow JL, Bell SC, Wainwright CE, Fantino E. Differential expression of genes and receptors in monocytes from patients with cystic fibrosis. J Cyst Fibros 2018; 18:342-348. [PMID: 30177416 DOI: 10.1016/j.jcf.2018.07.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/29/2018] [Accepted: 07/30/2018] [Indexed: 11/30/2022]
Abstract
INTRODUCTION We previously reported defective alternative polarization (M2) of macrophages and early expression of classically polarized (M1) macrophage markers in unpolarized monocyte-derived macrophages (MDMs) in patients with cystic fibrosis (CF). The present study assessed whether the mechanism(s) underlying defective macrophage polarization resided in circulating monocytes. METHODS Monocyte subsets (classical, intermediate and non-classical), markers for monocyte activation (CD163) and recruitment (CD195), receptors/genes associated with macrophage differentiation and polarization were analyzed in CF and compared with healthy individuals. RESULTS No differences were observed in the monocyte subsets or in the expression of CD163 or CD195. Expression of the M-CSF receptor, TLR4, γC, IL-4Rα, IL-13Rα1, TIMP-1 and Cox-2 were higher in CF monocytes, albeit at low levels, whereas, LRP1, MMP9, MMP28 were downregulated compared to mooncytes from healthy individuals. CONCLUSIONS Our data suggest that differences in CF monocytes may contribute to the reported CFTR-dependent defect in macrophage differentiation, polarization and function.
Collapse
Affiliation(s)
- Abdullah A Tarique
- Child Health Research Centre (CHRC), The University of Queensland, Australia
| | - Peter D Sly
- Child Health Research Centre (CHRC), The University of Queensland, Australia; Department of Respiratory and Sleep Medicine, Children's Health Queensland, Brisbane, Australia.
| | - Diana G Cardenas
- Child Health Research Centre (CHRC), The University of Queensland, Australia
| | - Lin Luo
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, Australia
| | - Jennifer L Stow
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, Australia
| | - Scott C Bell
- QIMR Berghofer Medical Research Institute, Brisbane, Australia; Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| | - Claire E Wainwright
- Child Health Research Centre (CHRC), The University of Queensland, Australia; Department of Respiratory and Sleep Medicine, Children's Health Queensland, Brisbane, Australia
| | - Emmanuelle Fantino
- Child Health Research Centre (CHRC), The University of Queensland, Australia
| |
Collapse
|
26
|
Long ME, Gong KQ, Volk JS, Eddy WE, Chang MY, Frevert CW, Altemeier WA, Gale M, Liles WC, Manicone AM. Matrix metalloproteinase 28 is regulated by TRIF- and type I IFN-dependent signaling in macrophages. Innate Immun 2018; 24:357-365. [PMID: 30068264 PMCID: PMC6545921 DOI: 10.1177/1753425918791024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/19/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are transcriptionally regulated proteases that have multiple roles in modifying the extracellular matrix (ECM) and inflammatory response. Our previous work identified Mmp28 as a key regulator of inflammation and macrophage polarization during experimental models of pulmonary infection, fibrosis, and chronic smoke exposure. However, the signaling pathways responsible for regulation of macrophage Mmp28 expression remain undefined. This study utilized murine macrophages obtained from wild type, Tlr2-/-, Tlr4-/-, MyD88-/-, Ticam1 Lps2 ( Trifmutant), and Ifnar1-/- mice to test the hypothesis that macrophage Mmp28 expression was dependent on TRIF and type I IFN. Our results support the hypothesis, demonstrating that increased macrophage Mmp28 expression was dependent on type I IFN after LPS and poly(I:C) stimulation. To gain further insight into the function of MMP28, we explored the inflammatory response of macrophages derived from wild type or Mmp28-/- mice to stimulation with poly(I:C). Our data support a role for MMP28 in regulating the macrophage inflammatory response to poly(I:C) because expression of Ccl2, Ccl4, Cxcl10, and Il6 were increased in Mmp28-/- macrophages. Together, these data support a model in which macrophages integrate TRIF- and type I IFN-dependent signaling to coordinate regulation of proteins with the capacity to modify the ECM.
Collapse
Affiliation(s)
- Matthew E. Long
- Center for Lung Biology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA,
USA
| | - Ke-Qin Gong
- Center for Lung Biology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA,
USA
| | - Joseph S. Volk
- Center for Lung Biology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA,
USA
| | - William E. Eddy
- Center for Lung Biology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA,
USA
| | - Mary Y. Chang
- Department of Comparative Medicine,
University of Washington, Seattle, WA, USA
| | - Charles W. Frevert
- Center for Lung Biology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA,
USA
- Department of Comparative Medicine,
University of Washington, Seattle, WA, USA
| | - William A. Altemeier
- Center for Lung Biology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA,
USA
| | - Michael Gale
- Department of Immunology and the Center
for Innate Immunity and Immune Disease, University of Washington, Seattle, WA,
USA
| | - W. Conrad Liles
- Center for Lung Biology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA,
USA
| | - Anne M. Manicone
- Center for Lung Biology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA,
USA
| |
Collapse
|
27
|
Gharib SA, Manicone AM, Parks WC. Matrix metalloproteinases in emphysema. Matrix Biol 2018; 73:34-51. [PMID: 29406250 DOI: 10.1016/j.matbio.2018.01.018] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/14/2017] [Accepted: 01/24/2018] [Indexed: 02/07/2023]
Abstract
Several studies have implicated a causative role for specific matrix metalloproteinases (MMPs) in the development and progression of cigarette smoke-induced chronic obstructive pulmonary disease (COPD) and its severe sequela, emphysema. However, the precise function of any given MMP in emphysema remains an unanswered question. Emphysema results from the degradation of alveolar elastin - among other possible mechanisms - a process that is often thought to be caused by elastolytic proteinases made by macrophages. In this article, we discuss the data suggesting, supporting, or refuting causative roles of macrophage-derived MMPs, with a focus on MMPs-7, -9, -10, -12, and 28, in both the human disease and mouse models of emphysema. Findings from experimental models suggest that some MMPs, such as MMP-12, may directly breakdown elastin, whereas others, particularly MMP-10 and MMP-28, promote the development of emphysema by influencing the proteolytic and inflammatory activities of macrophages.
Collapse
Affiliation(s)
- Sina A Gharib
- Center for Lung Biology, University of Washington, Seattle, WA, USA
| | - Anne M Manicone
- Center for Lung Biology, University of Washington, Seattle, WA, USA
| | - William C Parks
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
28
|
Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 2018. [PMID: 29524630 DOI: 10.1016/j.matbio.2018.03.005] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| | - Jeffrey C Horowitz
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Alexandra Naba
- Department of Physiology & Biophysics, University of Illinois at Chicago, United States.
| | | | - Kamran Atabai
- Lung Biology Center, University of California, San Francisco, United States.
| | | | | | - Richard A Corley
- Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, United States.
| | - Bi-Sen Ding
- Weill Cornell Medical College, United States.
| | - Adam J Engler
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, United States.
| | - Kirk C Hansen
- Biochemistry & Molecular Genetics, University of Colorado Denver, United States.
| | - James S Hagood
- Pediatric Respiratory Medicine, University of California San Diego, United States.
| | - Farrah Kheradmand
- Division of Pulmonary and Critical Care, Baylor College of Medicine, United States.
| | - Qing S Lin
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, United States.
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, United States.
| | - Laura Niklason
- Department of Anesthesiology, Yale University, United States.
| | - Luis A Ortiz
- Division of Environmental and Occupational Health, University of Pittsburgh, United States.
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, United States.
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, United States.
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Harold A Chapman
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, United States.
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| |
Collapse
|
29
|
Weihbrecht K, Goar WA, Carter CS, Sheffield VC, Seo S. Genotypic and phenotypic characterization of the Sdccag8Tn(sb-Tyr)2161B.CA1C2Ove mouse model. PLoS One 2018; 13:e0192755. [PMID: 29444170 PMCID: PMC5812623 DOI: 10.1371/journal.pone.0192755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/30/2018] [Indexed: 12/13/2022] Open
Abstract
Nephronophthisis-related ciliopathies (NPHP-RC) are a group of disorders that present with end-stage renal failure in childhood/adolescence, kidney cysts, retinal degeneration, and cerebellar hypoplasia. One disorder that shares clinical features with NPHP-RC is Bardet-Biedl Syndrome (BBS). Serologically defined colon cancer antigen 8 (SDCCAG8; also known as NPHP10 and BBS16) is an NPHP gene that is also associated with BBS. To better understand the patho-mechanisms of NPHP and BBS caused by loss of SDCCAG8 function, we characterized an SDCCAG8 mouse model (Sdccag8Tn(sb-Tyr)2161B.CA1C2Ove) generated by Sleeping Beauty Transposon (SBT)-mediated insertion mutagenesis. Consistent with the previously reported, independent SDCCAG8 mouse models, our mutant mice display pre-axial polydactyly in their hind limbs. In addition, we report patterning defects in the secondary palate, brain abnormalities, as well as neonatal lethality associated with developmental defects in the lung in our mouse model. The neonatal lethality phenotype is genetic background dependent and rescued by introducing 129S6/SvEvTac background. Genetic modifier(s) responsible for this effect were mapped to a region between SNPs rs3714172 and rs3141832 on chromosome 11. While determining the precise genetic lesion in our mouse model, we found that SBT insertion resulted in a deletion of multiple exons from both Sdccag8 and its neighboring gene Akt3. We ascribe the patterning defects in the limb and the secondary palate as well as lung abnormalities to loss of SDCCAG8, while the developmental defects in the brain are likely due to the loss of AKT3. This mouse model may be useful to study features not observed in other SDCCAG8 models but cautions are needed in interpreting data.
Collapse
Affiliation(s)
- Katie Weihbrecht
- Department of Pediatrics, University of Iowa, Iowa City, lowa, United States of America
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, lowa, United States of America
- Institute for Vision Research, University of Iowa, lowa, United States of America
| | - Wesley A. Goar
- Department of Pediatrics, University of Iowa, Iowa City, lowa, United States of America
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, lowa, United States of America
- Institute for Vision Research, University of Iowa, lowa, United States of America
| | - Calvin S. Carter
- Department of Pediatrics, University of Iowa, Iowa City, lowa, United States of America
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, lowa, United States of America
- Institute for Vision Research, University of Iowa, lowa, United States of America
| | - Val C. Sheffield
- Department of Pediatrics, University of Iowa, Iowa City, lowa, United States of America
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, lowa, United States of America
- Institute for Vision Research, University of Iowa, lowa, United States of America
| | - Seongjin Seo
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, lowa, United States of America
- Institute for Vision Research, University of Iowa, lowa, United States of America
- * E-mail:
| |
Collapse
|
30
|
Matrix metalloproteinases as regulators of inflammatory processes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2036-2042. [DOI: 10.1016/j.bbamcr.2017.05.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/06/2017] [Accepted: 05/09/2017] [Indexed: 12/26/2022]
|
31
|
Zou S, Luo Q, Song Z, Zhang L, Xia Y, Xu H, Xiang Y, Yin Y, Cao J. Contribution of Progranulin to Protective Lung Immunity During Bacterial Pneumonia. J Infect Dis 2017; 215:1764-1773. [PMID: 28595330 DOI: 10.1093/infdis/jix197] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/20/2017] [Indexed: 12/21/2022] Open
Abstract
Background Progranulin (PGRN) is an important immunomodulatory factor in a variety of inflammatory diseases. However, its role in pulmonary immunity against bacterial infection remains unknown. Methods Pneumonia was induced in PGRN-deficient and normal wild-type mice using Pseudomonas aeruginosa or Staphylococcus aureus, and we assessed the effects of PGRN on survival, bacterial burden, cytokine and chemokine production, and pulmonary leukocyte recruitment after bacterial pneumonia. Results Patients with community-acquired pneumonia displayed elevated PGRN levels. Likewise, mice with Gram-negative and Gram-positive pneumonia had increased PGRN production in the lung and circulation. Progranulin deficiency led to increased bacterial growth and dissemination accompanied by enhanced lung injury and mortality in bacterial pneumonia, which was associated with impaired recruitment of macrophages and neutrophils in the lung. The reduced number of pulmonary macrophages and neutrophils observed in PGRN-deficient mice was related to a reduction of CCL2 and CXCL1 in the lungs after bacterial pneumonia. Importantly, therapeutic administration of PGRN improved mortality in severe bacterial pneumonia. Conclusions This study supports a novel role for PGRN in pulmonary immunity and suggests that treatment with PGRN may be a viable therapy for bacterial pneumonia.
Collapse
Affiliation(s)
- Shan Zou
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University.,Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, China
| | - Qin Luo
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University.,Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, China
| | - Zhixin Song
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, China
| | - Liping Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University
| | - Yun Xia
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University
| | - Huajian Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University
| | - Yu Xiang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University
| | - Yibing Yin
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, China
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University
| |
Collapse
|
32
|
Long ME, Gong KQ, Eddy WE, Liles WC, Manicone AM. Pharmacologic inhibition of MEK1/2 reduces lung inflammation without impairing bacterial clearance in experimental Pseudomonas aeruginosa pneumonia. Pneumonia (Nathan) 2017; 9:13. [PMID: 28879065 PMCID: PMC5583963 DOI: 10.1186/s41479-017-0037-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023] Open
Abstract
This study was designed to test the therapeutic potential of a MEK1/2 inhibitor (MEKi) in an experimental model of Pseudomonas aeruginosa pneumonia. The study found that treatment with MEKi reduced alveolar neutrophilic inflammation and led to faster recovery of weight compared to carrier-treated mice, without impairing bacterial clearance. Alveolar macrophages isolated from MEKi-treated mice also had increased M2 gene and protein expression, supporting the concept that MEKi modulates in vivo macrophage inflammatory responses. In summary, this report demonstrates the potential of MEKi to promote the resolution of inflammation in vivo during a primary lung infection without impairing bacterial clearance.
Collapse
Affiliation(s)
- Matthew E. Long
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, 850 Republican St, Seattle, WA 98109 USA
| | - Ke-Qin Gong
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, 850 Republican St, Seattle, WA 98109 USA
| | - William E. Eddy
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, 850 Republican St, Seattle, WA 98109 USA
| | - W. Conrad Liles
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, 850 Republican St, Seattle, WA 98109 USA
| | - Anne M. Manicone
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, 850 Republican St, Seattle, WA 98109 USA
| |
Collapse
|
33
|
Valproic acid mitigates the inflammatory response and prevents acute respiratory distress syndrome in a murine model of Escherichia coli pneumonia at the expense of bacterial clearance. J Trauma Acute Care Surg 2017; 82:758-765. [PMID: 28099388 DOI: 10.1097/ta.0000000000001389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Histone deacetylase inhibitors (HDACI) are members of a family of epigenetic modifying agents with broad anti-inflammatory properties. These anti-inflammatory properties may have important therapeutic implications in acute respiratory distress syndrome (ARDS). However, administration of HDACI may create an immunosuppressive environment conducive to bacterial growth. Accordingly, the aim of the current study is to investigate the effect of HDACI valproic acid (VPA) on host inflammatory response and bacterial burden in a murine model of Escherichia coli pneumonia-induced ARDS. METHODS ARDS was induced in male C57BL6 mice (n = 24) by endotracheal instillation of 3 × 10 E. coli. VPA (250 mg/kg) was administered 30 minutes after E. coli instillation in the intervention group. Blood samples were collected at 3 and 6 hours, and animals were sacrificed at 6 hours. Bronchoalveolar lavage (BAL) was performed, and tissue specimens were harvested. Cytokine levels were measured in blood and BAL, and so was transalveolar protein transit. Cell counts and colony forming units were quantified in BAL fluid. RESULTS VPA reduced neutrophil influx into the lungs and local tissue destruction through decreased myeloperoxidase activity. It also ameliorated the pulmonary and systemic inflammatory response. This led to greater bacterial proliferation in the pulmonary parenchyma. CONCLUSION Administration of VPA in a clinically relevant bacterial model of murine ARDS mitigates the host inflammatory response, essentially preventing ARDS, but creates an immunosuppressive environment that favors bacterial overgrowth.
Collapse
|
34
|
Gill SE, Nadler ST, Li Q, Frevert CW, Park PW, Chen P, Parks WC. Shedding of Syndecan-1/CXCL1 Complexes by Matrix Metalloproteinase 7 Functions as an Epithelial Checkpoint of Neutrophil Activation. Am J Respir Cell Mol Biol 2017; 55:243-51. [PMID: 26934670 DOI: 10.1165/rcmb.2015-0193oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Although neutrophils play critical roles in innate immunity, in excess these cells cause severe tissue damage. Thus, neutrophil activation must be tightly regulated to prevent indiscriminant damage. Previously, we reported that mice lacking matrix metalloproteinase (MMP) 7 are protected from lung injury owing to markedly impaired neutrophil movement from the interstitium into mucosal lumenal spaces. This phenotype resulted from a lack of MMP7 shedding of syndecan-1, a heparan sulfate proteoglycan that carries the neutrophil chemokine CXCL1 as cargo. Here, we assessed if shedding syndecan-1/CXCL1 complexes affects neutrophil activation. Whereas injured monolayers of wild-type alveolar type II cells potently stimulated neutrophil activation, as gauged by release of myeloperoxidase, cells from Mmp7(-/-) or syndecan-1-null (Sdc1(-/-)) mice or human cells with MMP7 knockdown did not. In vivo, we observed reduced myeloperoxidase release relative to neutrophil numbers in bleomycin-injured Mmp7(-/-) and Sdc1(-/-) mice. Furthermore, we determined that soluble syndecan-1 directly stimulated neutrophil activation in the absence of cellular damage. These data indicate that MMP7 shedding of syndecan-1/CXCL1 complexes functions as a checkpoint that restricts neutrophil activation at sites of epithelial injury.
Collapse
Affiliation(s)
- Sean E Gill
- 1 Center for Lung Biology and Department of.,2 Centre for Critical Illness Research, Western University, London, Ontario, Canada
| | | | | | - Charles W Frevert
- 1 Center for Lung Biology and Department of.,3 Comparative Medicine, University of Washington, Seattle, Washington
| | - Pyong Woo Park
- 4 Department of Medicine, Children's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Peter Chen
- 1 Center for Lung Biology and Department of.,5 Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - William C Parks
- 1 Center for Lung Biology and Department of.,5 Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
35
|
Brilha S, Sathyamoorthy T, Stuttaford LH, Walker NF, Wilkinson RJ, Singh S, Moores RC, Elkington PT, Friedland JS. Early Secretory Antigenic Target-6 Drives Matrix Metalloproteinase-10 Gene Expression and Secretion in Tuberculosis. Am J Respir Cell Mol Biol 2017; 56:223-232. [PMID: 27654284 DOI: 10.1165/rcmb.2016-0162oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Tuberculosis (TB) causes disease worldwide, and multidrug resistance is an increasing problem. Matrix metalloproteinases (MMPs), particularly the collagenase MMP-1, cause lung extracellular matrix destruction, which drives disease transmission and morbidity. The role in such tissue damage of the stromelysin MMP-10, a key activator of the collagenase MMP-1, was investigated in direct Mycobacterium tuberculosis (Mtb)-infected macrophages and in conditioned medium from Mtb-infected monocyte-stimulated cells. Mtb infection increased MMP-10 secretion from primary human macrophages 29-fold, whereas Mtb-infected monocytes increased secretion by 4.5-fold from pulmonary epithelial cells and 10.5-fold from fibroblasts. Inhibition of MMP-10 activity decreased collagen breakdown. In two independent cohorts of patients with TB from different continents, MMP-10 was increased in both induced sputum and bronchoalveolar lavage fluid compared with control subjects and patients with other respiratory diseases (both P < 0.05). Mtb drove 3.5-fold greater MMP-10 secretion from human macrophages than the vaccine strain bacillus Calmette-Guerin (P < 0.001), whereas both mycobacteria up-regulated TNF-α secretion equally. Using overlapping, short, linear peptides covering the sequence of early secretory antigenic target-6, a virulence factor secreted by Mtb, but not bacillus Calmette-Guerin, we found that stimulation of human macrophages with a single specific 15-amino acid peptide sequence drove threefold greater MMP-10 secretion than any other peptide (P < 0.001). Mtb-driven MMP-10 secretion was inhibited in a dose-dependent manner by p38 and extracellular signal-related kinase mitogen-activated protein kinase blockade (P < 0.001 and P < 0.01 respectively), but it was not affected by inhibition of NF-κB. In summary, Mtb activates inflammatory and stromal cells to secrete MMP-10, and this is partly driven by the virulence factor early secretory antigenic target-6, implicating it in TB-associated tissue destruction.
Collapse
Affiliation(s)
- Sara Brilha
- 1 Infectious Diseases and Immunity, Imperial College London, London, United Kingdom.,2 Centre for Inflammation and Tissue Repair, Respiratory Medicine, University College London, London, United Kingdom
| | | | - Laura H Stuttaford
- 1 Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Naomi F Walker
- 1 Infectious Diseases and Immunity, Imperial College London, London, United Kingdom.,3 Clinical Infectious Diseases Research Initiative, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,4 Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Robert J Wilkinson
- 3 Clinical Infectious Diseases Research Initiative, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,5 Department of Medicine, Imperial College London, London, United Kingdom.,6 The Francis Crick Institute, London, United Kingdom.,7 Wellcome Trust Imperial College Centre for Global Health, Imperial College London, London, United Kingdom; and
| | - Shivani Singh
- 1 Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Rachel C Moores
- 1 Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Paul T Elkington
- 1 Infectious Diseases and Immunity, Imperial College London, London, United Kingdom.,8 National Institute of Health Research Respiratory Biomedical Research Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jon S Friedland
- 1 Infectious Diseases and Immunity, Imperial College London, London, United Kingdom.,7 Wellcome Trust Imperial College Centre for Global Health, Imperial College London, London, United Kingdom; and
| |
Collapse
|
36
|
Hendrix AY, Kheradmand F. The Role of Matrix Metalloproteinases in Development, Repair, and Destruction of the Lungs. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:1-29. [PMID: 28662821 DOI: 10.1016/bs.pmbts.2017.04.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Normal gas exchange after birth requires functional lung alveolar units that are lined with epithelial cells, parts of which are intricately fused with microvascular capillaries. A significant phase of alveolar lung development occurs in the perinatal period, continues throughout early stages in life, and requires activation of matrix-remodeling enzymes. Failure to achieve an optimum number of alveoli during lung maturation can cause several untoward medical consequences including disabling obstructive and/or restrictive lung diseases that limit physiological endurance and increase mortality. Several members of the matrix metalloproteinase (MMP) family are critical in lung remodeling before and after birth; however, their resurgence in response to environmental factors, infection, and injury can also compromise lung function. Therefore, temporal expression, regulation, and function of MMPs play key roles in developing and maintaining adequate oxygenation under steady state, as well as in diseased conditions. Broadly, with the exception of MMP2 and MMP14, most deletional mutations of MMPs fail to perturb lung development; however, their individual absence can alter the pathophysiology of respiratory diseases. Specifically, under stressed conditions such as acute respiratory infection and allergic inflammation, MMP2 and MMP9 can play a protective role through bacterial clearance and production of chemotactic gradient, while loss of MMP12 can protect mice from smoke-induced lung disease. Therefore, better understanding of the expression and function of MMPs under normal lung development and their resurgence in response respiratory diseases could provide new therapeutic options in the future.
Collapse
Affiliation(s)
- Amanda Y Hendrix
- Section of Pulmonary and Critical Care, and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Farrah Kheradmand
- Section of Pulmonary and Critical Care, and Immunology, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
37
|
Matrix Metalloproteinase-28 Is a Key Contributor to Emphysema Pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1288-1300. [PMID: 28399390 DOI: 10.1016/j.ajpath.2017.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 02/16/2017] [Indexed: 10/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) comprises chronic bronchitis and emphysema, and is a leading cause of morbidity and mortality. Because tissue destruction is the prominent characteristic of emphysema, extracellular proteinases, particularly those with elastolytic ability, are often considered to be key drivers in this disease. Several human and mouse studies have implicated roles for matrix metalloproteinases (MMPs), particularly macrophage-derived proteinases, in COPD pathogenesis. MMP-28 is expressed by the pulmonary epithelium and macrophage, and we have found that it regulates macrophage recruitment and polarization. We hypothesized that MMP-28 has contributory roles in emphysema via alteration of macrophage numbers and activation. Because of the established association of emphysema pathogenesis to macrophage influx, we evaluated the inflammatory changes and lung histology of Mmp28-/- mice exposed to 3 and 6 months of cigarette smoke. At earlier time points, we found altered macrophage polarization in the smoke-exposed Mmp28-/- lung consistent with other published findings that MMP-28 regulates macrophage activation. At both 3 and 6 months, Mmp28-/- mice had blunted inflammatory responses more closely resembling nonsmoked mice, with a reduction in neutrophil recruitment and CXCL1 chemokine expression. By 6 months, Mmp28-/- mice were protected from emphysema. These results highlight a previously unrecognized role for MMP-28 in promoting chronic lung inflammation and tissue remodeling induced by cigarette smoke and highlight another potential target to modulate COPD.
Collapse
|
38
|
Smigiel KS, Parks WC. Matrix Metalloproteinases and Leukocyte Activation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:167-195. [PMID: 28413028 DOI: 10.1016/bs.pmbts.2017.01.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As their name implies, matrix metalloproteinases (MMPs) are thought to degrade extracellular matrix proteins, a function that is indeed performed by some members. However, regardless of their cell source, matrix degradation is not the only function of these enzymes. Rather, individual MMPs have been shown to regulate specific immune processes, such as leukocyte influx and migration, antimicrobial activity, macrophage activation, and restoration of barrier function, typically by processing a range of nonmatrix protein substrates. Indeed, MMP expression is low under steady-state conditions but is markedly induced during inflammatory processes including infection, wound healing, and cancer. Increasing research is showing that MMPs are not just a downstream consequence of a generalized inflammatory process, but rather are critical factors in the overall regulation of the pattern, type, and duration of immune responses. This chapter outlines the role of leukocytes in tissue remodeling and describes recent progress in our understanding of how MMPs alter leukocyte activity.
Collapse
Affiliation(s)
- Kate S Smigiel
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - William C Parks
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
39
|
Vandivort TC, Birkland TP, Domiciano TP, Mitra S, Kavanagh TJ, Parks WC. Stromelysin-2 (MMP-10) facilitates clearance and moderates inflammation and cell death following lung exposure to long multiwalled carbon nanotubes. Int J Nanomedicine 2017; 12:1019-1031. [PMID: 28223796 PMCID: PMC5304974 DOI: 10.2147/ijn.s123484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Multiwalled carbon nanotubes (MWCNTs) are nanomaterials composed of multiple layers of graphene cylinders with unique properties that make them valuable for a number of industries. However, rising global production has led to concerns regarding potential occupational exposures to them as raw materials during handling. This is especially true for long MWCNT fibers, whose aspect ratio has been posited to initiate pathology similar to that of asbestos. Matrix metalloproteinases (MMPs) are a class of extracellular endopeptidases that control various processes related to tissue repair, inflammation, and more. Stromelysin-2 (MMP-10) has roles in modulating macrophage activation and function, and hence, we used an MMP-10 null (Mmp10−/−) mouse model to assess its role in controlling lung responses to inhaled long MWCNTs. Oropharyngeal aspiration of long MWCNTs (80 µg/mouse) by wild-type mice induced expression of Mmp10 mRNA, which was accompanied by a robust inflammatory response characterized by elevated expression of Tnfa, Il6, and Il1b. In Mmp10−/− mice, we found that absence of MMP-10 led to impaired pulmonary clearance of MWCNTs and reduced macrophage cell survival. Exposure of wild-type bone marrow-derived macrophages (BMDMs) and alveolar macrophages to MWCNTs caused a rapid, dose-dependent upregulation of Mmp10 mRNA expression, which was accompanied by expression of pro-inflammatory products (Il6 and Il1b). These products were further enhanced in Mmp10−/− macrophages, resulting in increased caspase-3-dependent cell death compared with wild-type cells. These findings indicate that MMP-10 facilitates the clearance of MWCNTs and moderates the pro-inflammatory response of exposed alveolar and infiltrated macrophages.
Collapse
Affiliation(s)
- Tyler C Vandivort
- Cedars-Sinai Medical Center, Women's Guild Lung Institute, Los Angeles, CA; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Timothy P Birkland
- Cedars-Sinai Medical Center, Women's Guild Lung Institute, Los Angeles, CA
| | | | - Somenath Mitra
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ, USA
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - William C Parks
- Cedars-Sinai Medical Center, Women's Guild Lung Institute, Los Angeles, CA
| |
Collapse
|
40
|
Wight TN, Frevert CW, Debley JS, Reeves SR, Parks WC, Ziegler SF. Interplay of extracellular matrix and leukocytes in lung inflammation. Cell Immunol 2017; 312:1-14. [PMID: 28077237 PMCID: PMC5290208 DOI: 10.1016/j.cellimm.2016.12.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022]
Abstract
During inflammation, leukocytes influx into lung compartments and interact with extracellular matrix (ECM). Two ECM components, versican and hyaluronan, increase in a range of lung diseases. The interaction of leukocytes with these ECM components controls leukocyte retention and accumulation, proliferation, migration, differentiation, and activation as part of the inflammatory phase of lung disease. In addition, bronchial epithelial cells from asthmatic children co-cultured with human lung fibroblasts generate an ECM that is adherent for monocytes/macrophages. Macrophages are present in both early and late lung inflammation. Matrix metalloproteinase 10 (MMP10) is induced in alveolar macrophages with injury and infection and modulates macrophage phenotype and their ability to degrade collagenous ECM components. Collectively, studies outlined in this review highlight the importance of specific ECM components in the regulation of inflammatory events in lung disease. The widespread involvement of these ECM components in the pathogenesis of lung inflammation make them attractive candidates for therapeutic intervention.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - Charles W Frevert
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Jason S Debley
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, and Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Stephen R Reeves
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, and Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
41
|
Long ME, Eddy WE, Gong KQ, Lovelace-Macon LL, McMahan RS, Charron J, Liles WC, Manicone AM. MEK1/2 Inhibition Promotes Macrophage Reparative Properties. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:862-872. [PMID: 28003382 PMCID: PMC5224968 DOI: 10.4049/jimmunol.1601059] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/16/2016] [Indexed: 12/18/2022]
Abstract
Macrophages have important functional roles in regulating the timely promotion and resolution of inflammation. Although many of the intracellular signaling pathways involved in the proinflammatory responses of macrophages are well characterized, the components that regulate macrophage reparative properties are less well understood. We identified the MEK1/2 pathway as a key regulator of macrophage reparative properties. Pharmacological inhibition of the MEK1/2 pathway by a MEK1/2 inhibitor (MEKi) significantly increased expression of IL-4/IL-13 (M2)-responsive genes in murine bone marrow-derived and alveolar macrophages. Deletion of the MEK1 gene using LysMCre+/+Mek1fl/fl macrophages as an alternate approach yielded similar results. MEKi enhanced STAT6 phosphorylation, and MEKi-induced changes in M2 polarization were dependent on STAT6. In addition, MEKi treatment significantly increased murine and human macrophage efferocytosis of apoptotic cells, independent of macrophage polarization and STAT6. These phenotypes were associated with increased gene and protein expression of Mertk, Tyro3, and Abca1, three proteins that promote macrophage efferocytosis. We also studied the effects of MEKi on in vivo macrophage efferocytosis and polarization. MEKi-treated mice had increased efferocytosis of apoptotic polymorphonuclear leukocytes instilled into the peritoneum. Furthermore, administration of MEKi after LPS-induced lung injury led to improved recovery of weight, fewer neutrophils in the alveolar compartment, and greater macrophage M2 polarization. Collectively, these results show that MEK1/2 inhibition is capable of promoting the reparative properties of murine and human macrophages. These studies suggest that the MEK1/2 pathway may be a therapeutic target to promote the resolution of inflammation via modulation of macrophage functions.
Collapse
Affiliation(s)
- Matthew E Long
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109
| | - William E Eddy
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109
| | - Ke-Qin Gong
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109
| | - Lara L Lovelace-Macon
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109
| | - Ryan S McMahan
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109
| | - Jean Charron
- Centre de Recherche sur le Cancer de l'Université Laval, Centre de Recherche du Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, Quebec G1R 3S3, Canada; and
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec G1V 0A6, Canada
| | - W Conrad Liles
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109
| | - Anne M Manicone
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109;
| |
Collapse
|
42
|
Filamentous Bacteriophage Produced by Pseudomonas aeruginosa Alters the Inflammatory Response and Promotes Noninvasive Infection In Vivo. Infect Immun 2016; 85:IAI.00648-16. [PMID: 27795361 DOI: 10.1128/iai.00648-16] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/07/2016] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa is an important opportunistic human pathogen that lives in biofilm-like cell aggregates at sites of chronic infection, such as those that occur in the lungs of patients with cystic fibrosis and nonhealing ulcers. During growth in a biofilm, P. aeruginosa dramatically increases the production of filamentous Pf bacteriophage (Pf phage). Previous work indicated that when in vivo Pf phage production was inhibited, P. aeruginosa was less virulent. However, it is not clear how the production of abundant quantities of Pf phage similar to those produced by biofilms under in vitro conditions affects pathogenesis. Here, using a murine pneumonia model, we show that the production of biofilm-relevant amounts of Pf phage prevents the dissemination of P. aeruginosa from the lung. Furthermore, filamentous phage promoted bacterial adhesion to mucin and inhibited bacterial invasion of airway epithelial cultures, suggesting that Pf phage traps P. aeruginosa within the lung. The in vivo production of Pf phage was also associated with reduced lung injury, reduced neutrophil recruitment, and lower cytokine levels. Additionally, when producing Pf phage, P. aeruginosa was less prone to phagocytosis by macrophages than bacteria not producing Pf phage. Collectively, these data suggest that filamentous Pf phage alters the progression of the inflammatory response and promotes phenotypes typically associated with chronic infection.
Collapse
|
43
|
Puntorieri V, McCaig LA, Howlett CJ, Yao LJ, Lewis JF, Yamashita CM, Veldhuizen RAW. Lack of matrix metalloproteinase 3 in mouse models of lung injury ameliorates the pulmonary inflammatory response in female but not in male mice. Exp Lung Res 2016; 42:365-379. [PMID: 27676418 DOI: 10.1080/01902148.2016.1231243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND The acute respiratory distress syndrome (ARDS) is a complex pulmonary disorder in which the local release of cytokines and chemokines appears central to the pathophysiology. OBJECTIVE Based on the known role of matrix metalloproteinase-3 (MMP3) in inflammatory processes, the objective was to examine the role of MMP3 in the pathogenesis of ARDS through the modulation of pulmonary inflammation. MATERIALS AND METHODS Female and male, wild type (MMP3+/+) and knock out (MMP3-/-) mice were exposed to two, clinically relevant models of ARDS including (i) lipopolysaccharide (LPS)-induced lung injury, and (ii) hydrochloric acid-induced lung injury. Parameters of lung injury and inflammation were assessed through measurements in lung lavage including total protein content, inflammatory cell influx, and concentrations of mediators such as TNF-α, IL-6, G-CSF, CXCL1, CXCL2, and CCL2. Lung histology and compliance were also evaluated in the LPS model of injury. RESULTS Following intra-tracheal LPS instillation, all mice developed lung injury, as measured by an increase in lavage neutrophils, and decrease in lung compliance, with no overall effect of genotype observed. Increased concentrations of lavage inflammatory cytokines and chemokines were also observed following LPS injury, however, LPS-instilled female MMP3-/- mice had lower levels of inflammatory mediators compared to LPS-instilled female MMP3+/+ mice. This effect of the genotype was not observed in male mice. Similar findings, including the MMP3-related sex differences, were also observed after acid-induced lung injury. CONCLUSION MMP3 contributes to the pathogenesis of ARDS, by affecting the pulmonary inflammatory response in female mice in relevant models of lung injury.
Collapse
Affiliation(s)
- Valeria Puntorieri
- a Department of Physiology and Pharmacology , Lawson Health Research Institute, Western University , London , Ontario , Canada
| | - Lynda A McCaig
- a Department of Physiology and Pharmacology , Lawson Health Research Institute, Western University , London , Ontario , Canada
| | - Christopher J Howlett
- b Department of Pathology and Laboratory Medicine , Western University , London , Ontario , Canada
| | - Li-Juan Yao
- c Department of Medicine , Western University , London , Ontario , Canada
| | - James F Lewis
- c Department of Medicine , Western University , London , Ontario , Canada
| | - Cory M Yamashita
- a Department of Physiology and Pharmacology , Lawson Health Research Institute, Western University , London , Ontario , Canada.,c Department of Medicine , Western University , London , Ontario , Canada
| | - Ruud A W Veldhuizen
- a Department of Physiology and Pharmacology , Lawson Health Research Institute, Western University , London , Ontario , Canada.,c Department of Medicine , Western University , London , Ontario , Canada
| |
Collapse
|
44
|
McMahan RS, Birkland TP, Smigiel KS, Vandivort TC, Rohani MG, Manicone AM, McGuire JK, Gharib SA, Parks WC. Stromelysin-2 (MMP10) Moderates Inflammation by Controlling Macrophage Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:899-909. [PMID: 27316687 PMCID: PMC4955757 DOI: 10.4049/jimmunol.1600502] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/24/2016] [Indexed: 02/06/2023]
Abstract
Several members of the matrix metalloproteinase (MMP) family control a range of immune processes, such as leukocyte influx and chemokine activity. Stromelysin-2 (MMP10) is expressed by macrophages in numerous tissues after injury; however, little is known of its function. In this study, we report that MMP10 is expressed by macrophages in human lungs from patients with cystic fibrosis and induced in mouse macrophages in response to Pseudomonas aeruginosa infection both in vivo and by isolated resident alveolar and bone marrow-derived macrophages (BMDM). Our data indicates that macrophage MMP10 serves a beneficial function in response to acute infection. Whereas wild-type mice survived infection with minimal morbidity, 50% of Mmp10(-/-) mice died and all showed sustained weight loss (morbidity). Although bacterial clearance and neutrophil influx did not differ between genotypes, macrophage numbers were ∼3-fold greater in infected Mmp10(-/-) lungs than in wild-types. Adoptive transfer of wild-type BMDM normalized infection-induced morbidity in Mmp10(-/-) recipients to wild-type levels, demonstrating that the protective effect of MMP10 was due to its production by macrophages. Both in vivo and in cultured alveolar macrophages and BMDM, expression of several M1 macrophage markers was elevated, whereas M2 markers were reduced in Mmp10(-/-) tissue and cells. Global gene expression analysis revealed that infection-mediated transcriptional changes persisted in Mmp10(-/-) BMDM long after they were downregulated in wild-type cells. These results indicate that MMP10 serves a beneficial role in response to acute infection by moderating the proinflammatory response of resident and infiltrating macrophages.
Collapse
Affiliation(s)
- Ryan S McMahan
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105
| | - Timothy P Birkland
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195
| | - Kate S Smigiel
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Tyler C Vandivort
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105; Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Maryam G Rohani
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195; Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Anne M Manicone
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195
| | - John K McGuire
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Pediatrics, University of Washington, Seattle, WA 98195
| | - Sina A Gharib
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195
| | - William C Parks
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195; Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| |
Collapse
|
45
|
Sorting nexin 10 acting as a novel regulator of macrophage polarization mediates inflammatory response in experimental mouse colitis. Sci Rep 2016; 6:20630. [PMID: 26856241 PMCID: PMC4746623 DOI: 10.1038/srep20630] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/06/2016] [Indexed: 02/07/2023] Open
Abstract
In response to changes in microenvironment, macrophages polarize into functionally distinct phenotypes, playing a crucial role in the pathogenesis of inflammatory bowel disease (IBD). Here, we investigated the effects of sorting nexin 10 (SNX10), a protein involved in endosomal trafficking and osteoclast maturation, on regulation of macrophage polarization and progression of mouse colitis. Our results revealed that SNX10 deficiency increased the population of M2-type monocytes/macrophages, and protected against colonic inflammation and pathological damage induced by dextran sulfate sodium (DSS). By in vitro study, we showed that deficiency of SNX10 polarized macrophages derived from mouse bone marrow or human peripheral blood mononuclear cells (PBMCs) towards an anti-inflammatory M2 phenotype, which partially reversed by SNX10 plasmid transfection. Adoptive transfer of SNX10−/− macrophages ameliorated colitis in WT mice. However, transfer of WT macrophages exacerbated colitis in SNX10−/− mice. Our data disclose a crucial role and novel function for SNX10 in macrophage polarization. Loss of SNX10 function may be a potential promising therapeutic strategy for IBD.
Collapse
|
46
|
Altara R, Manca M, Sabra R, Eid AA, Booz GW, Zouein FA. Temporal cardiac remodeling post-myocardial infarction: dynamics and prognostic implications in personalized medicine. Heart Fail Rev 2015; 21:25-47. [PMID: 26498937 DOI: 10.1007/s10741-015-9513-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite dramatic improvements in short-term mortality rates following myocardial infarction (MI), long-term survival for MI patients who progress to heart failure remains poor. MI occurs when the left ventricle (LV) is deprived of oxygen for a sufficient period of time to induce irreversible necrosis of the myocardium. The LV response to MI involves significant tissue, cellular, and molecular level modifications, as well as substantial hemodynamic changes that feedback negatively to amplify the response. Inflammation to remove necrotic myocytes and fibroblast activation to form a scar are key wound healing responses that are highly variable across individuals. Few biomarkers of early remodeling stages are currently clinically adopted. The discovery of underlying pathophysiological mechanisms and associated novel biomarkers has the potential of improving prognostic capability and therapeutic monitoring. Combining these biomarkers with other prominent ones could constitute a powerful diagnostic and prognostic tool that directly reflects the pathophysiological remodeling of the LV. Understanding temporal remodeling at the tissue, cellular, and molecular level and its link to a well-defined set of biomarkers at early stages post-MI is a prerequisite for improving personalized care and devising more successful therapeutic interventions. Here we summarize the integral mechanisms that occur during early cardiac remodeling in the post-MI setting and highlight the most prominent biomarkers for assessing disease progression.
Collapse
Affiliation(s)
- Raffaele Altara
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Marco Manca
- DG-DI, Medical Applications, CERN, Geneva, Switzerland
| | - Ramzi Sabra
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA. .,Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
47
|
Zhu GF, Guo HJ, Huang Y, Wu CT, Zhang XF. Eriodictyol, a plant flavonoid, attenuates LPS-induced acute lung injury through its antioxidative and anti-inflammatory activity. Exp Ther Med 2015; 10:2259-2266. [PMID: 26668626 DOI: 10.3892/etm.2015.2827] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 05/15/2015] [Indexed: 02/07/2023] Open
Abstract
Acute lung injury (ALI) is characterized by excessive inflammatory responses and oxidative injury in the lung tissue. It has been suggested that anti-inflammatory or antioxidative agents could have therapeutic effects in ALI, and eriodictyol has been reported to exhibit antioxidative and anti-inflammatory activity in vitro. The aim of the present study was to investigate the effect of eriodictyol on lipopolysaccharide (LPS)-induced ALI in a mouse model. The mice were divided into four groups: Phosphate-buffered saline-treated healthy control, LPS-induced ALI, vehicle-treated ALI (LPS + vehicle) and eriodictyol-treated ALI (LPS + eriodictyol). Eriodictyol (30 mg/kg) was administered orally once, 2 days before the induction of ALI. The data showed that eriodictyol pretreatment attenuated LPS-induced ALI through its antioxidative and anti-inflammatory activity. Furthermore, the eriodictyol pretreatment activated the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway in the ALI mouse model, which attenuated the oxidative injury and inhibited the inflammatory cytokine expression in macrophages. In combination, the results of the present study demonstrated that eriodictyol could alleviate the LPS-induced lung injury in mice by regulating the Nrf2 pathway and inhibiting the expression of inflammatory cytokines in macrophages, suggesting that eriodictyol could be used as a potential drug for the treatment of LPS-induced lung injury.
Collapse
Affiliation(s)
- Guang-Fa Zhu
- Department of Infectious Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, P.R. China
| | - Hong-Juan Guo
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, P.R. China
| | - Yan Huang
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, P.R. China
| | - Chun-Ting Wu
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, P.R. China
| | - Xiang-Feng Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, P.R. China
| |
Collapse
|
48
|
Rohani MG, McMahan RS, Razumova MV, Hertz AL, Cieslewicz M, Pun SH, Regnier M, Wang Y, Birkland TP, Parks WC. MMP-10 Regulates Collagenolytic Activity of Alternatively Activated Resident Macrophages. J Invest Dermatol 2015; 135:2377-2384. [PMID: 25927164 PMCID: PMC4567949 DOI: 10.1038/jid.2015.167] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 04/08/2015] [Accepted: 04/20/2015] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinase-10 (MMP-10) is expressed by macrophages and epithelium in response to injury, but its functions in wound repair are unknown. We observed increased collagen deposition and skin stiffness in Mmp10(-/-) wounds, with no difference in collagen expression or reepithelialization. Increased collagen deposition in Mmp10(-/-) wounds was accompanied by less collagenolytic activity and reduced expression of specific metallocollagenases, particularly MMP-8 and MMP-13, where MMP-13 was the key collagenase. Ablation and adoptive transfer approaches and cell-based models demonstrated that the MMP-10-dependent collagenolytic activity was a product of alternatively activated (M2) resident macrophages. These data demonstrate a critical role for macrophage MMP-10 in controlling the tissue remodeling activity of macrophages and moderating scar formation during wound repair.
Collapse
Affiliation(s)
- Maryam G Rohani
- Center for Lung Biology, University of Washington, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA; Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Ryan S McMahan
- Center for Lung Biology, University of Washington, Seattle, Washington, USA; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Maria V Razumova
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Angie L Hertz
- Center for Lung Biology, University of Washington, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Maryelise Cieslewicz
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Suzie H Pun
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Ying Wang
- Center for Lung Biology, University of Washington, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA; Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Timothy P Birkland
- Center for Lung Biology, University of Washington, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| | - William C Parks
- Center for Lung Biology, University of Washington, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA; Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
49
|
McClelland KS, Bell K, Larney C, Harley VR, Sinclair AH, Oshlack A, Koopman P, Bowles J. Purification and Transcriptomic Analysis of Mouse Fetal Leydig Cells Reveals Candidate Genes for Specification of Gonadal Steroidogenic Cells1. Biol Reprod 2015; 92:145. [DOI: 10.1095/biolreprod.115.128918] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/02/2015] [Indexed: 01/12/2023] Open
|
50
|
Cao J, Xu F, Lin S, Song Z, Zhang L, Luo P, Xu H, Li D, Zheng K, Ren G, Yin Y. IL-27 controls sepsis-induced impairment of lung antibacterial host defence. Thorax 2014; 69:926-37. [PMID: 25074706 DOI: 10.1136/thoraxjnl-2014-205777] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Interleukin 27 (IL-27) is an important cytokine regulating host immune responses. However, its role in sepsis-induced immunosuppression remains unclear. AIM To investigate the role of IL-27 in modulating sepsis-induced immunosuppression using a murine model of caecal ligation and puncture (CLP)-induced sepsis followed by secondary challenge with Pseudomonas aeruginosa. METHODS CLP or sham surgery was performed in wild-type (WT) and IL-27 receptor (IL-27R)/WSX-1 knockout (KO) mice, and then mice were infected with intratracheal P aeruginosa. RESULTS IL-27 was upregulated in patients with sepsis and septic mice. Following sepsis and secondary intrapulmonary bacterial challenge, IL-27R KO mice had higher survival rates and improved bacterial clearance from lung and blood compared with WT mice, which was associated with early increased pulmonary cytokine/chemokine production, as well as enhanced neutrophil recruitment to airspaces. Neutralisation of IL-27 in septic mice significantly improved survival and clearance of bacteria from the lungs of septic mice infected with P aeruginosa, and direct application of recombinant IL-27 could increase susceptibility to P aeruginosa infection. The resistance of septic IL-27R KO mice to secondary P aeruginosa infection was abrogated by depletion of alveolar macrophages (AMs) and neutrophils. AMs from septic IL-27R KO mice had higher bacterial uptake and killing capacities, enhanced cytokine/chemokine production, and increased expression of costimulatory molecules compared with those from WT mice, while neutrophils from septic IL-27R KO mice had increased bacterial killing ability and higher expression of adhesion molecule Mac-1 compared with WT neutrophils. CONCLUSIONS IL-27 is an important mediator of sepsis-induced impairment of lung antibacterial host defence.
Collapse
Affiliation(s)
- Ju Cao
- Department of Laboratory Medicine, The Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Emergency and Intensive Care Unit, The Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Shihui Lin
- Department of Emergency and Intensive Care Unit, The Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Zhixin Song
- Key Laboratory of Diagnostic Medicine designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lipin Zhang
- Department of Laboratory Medicine, The Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Peng Luo
- Department of Laboratory Medicine, The Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Huajian Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Dairong Li
- Department of Respiratory Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Zheng
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yibing Yin
- Key Laboratory of Diagnostic Medicine designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|