1
|
Chen J, Shi S, Li X, Gao F, Zhu X, Feng R, Hu K, Li Y, Chen S, Zhang R, Wang X, Ding C, Liu G, Chen T, Liang W. CCL7 promotes macrophage polarization and synovitis to exacerbate rheumatoid arthritis. iScience 2025; 28:112177. [PMID: 40224025 PMCID: PMC11987677 DOI: 10.1016/j.isci.2025.112177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/12/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Chemokine C-C motif ligand 7 (CCL7) is implicated in various immune and inflammatory processes; however, its role in rheumatoid arthritis (RA) remains unclear. In this study, we observed that CCL7 expression was upregulated in synovial M1-polarized macrophages and in the serum of RA mice and patients. CCL7 was found to promote macrophage polarization toward the M1 phenotype while inhibiting M2 differentiation in vitro. Furthermore, intra-articular injection of recombinant CCL7 protein in mice resulted in enhanced M1 polarization, increased inflammation, and fibrosis within synovial tissues, which exacerbated arthritis-associated pain. These effects were partially mitigated by treatment with a CCL7 neutralizing antibody. Mechanistically, we identified a CCL7 autocrine positive feedback loop that amplifies inflammation via the CCL7-CCR1-JAK2/STAT1 pathway. Collectively, our findings reveal a previously unrecognized CCL7-mediated autocrine inflammatory amplification loop that modulates macrophage polarization and exacerbates RA progression, positioning CCL7 as a potential therapeutic target for RA.
Collapse
Affiliation(s)
- Jun Chen
- Department of Rehabilitation Therapy, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan 442099, China
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shuo Shi
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xiaojia Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Feng Gao
- Department of Physical Therapy, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan 442099, China
| | - Xu Zhu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ru Feng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Ke Hu
- Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yicheng Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Shuiyuan Chen
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Rongkai Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Xiaoshuai Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tianyu Chen
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Wenquan Liang
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
- Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
2
|
Luksch H, Schulze F, Geißler-Lösch D, Sprott D, Höfs L, Szegö EM, Tonnus W, Winkler S, Günther C, Linkermann A, Behrendt R, Teichmann LL, Falkenburger BH, Rösen-Wolff A. Tissue inflammation induced by constitutively active STING is mediated by enhanced TNF signaling. eLife 2025; 14:e101350. [PMID: 40111902 PMCID: PMC11996172 DOI: 10.7554/elife.101350] [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: 07/08/2024] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
Constitutive activation of STING by gain-of-function mutations triggers manifestation of the systemic autoinflammatory disease STING-associated vasculopathy with onset in infancy (SAVI). In order to investigate the role of signaling by tumor necrosis factor (TNF) in SAVI, we used genetic inactivation of TNF receptors 1 and 2 in murine SAVI, which is characterized by T cell lymphopenia, inflammatory lung disease, and neurodegeneration. Genetic inactivation of TNFR1 and TNFR2, however, rescued the loss of thymocytes, reduced interstitial lung disease, and neurodegeneration. Furthermore, genetic inactivation of TNFR1 and TNFR2 blunted transcription of cytokines, chemokines, and adhesions proteins, which result from chronic STING activation in SAVI mice. In addition, increased transendothelial migration of neutrophils was ameliorated. Taken together, our results demonstrate a pivotal role of TNFR signaling in the pathogenesis of SAVI in mice and suggest that available TNFR antagonists could ameliorate SAVI in patients.
Collapse
Affiliation(s)
- Hella Luksch
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Felix Schulze
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - David Geißler-Lösch
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - David Sprott
- Department of Physiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Lennart Höfs
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Eva M Szegö
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, Faculty of Medicine and University Hospital Carl GustavDresdenGermany
| | - Stefan Winkler
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Claudia Günther
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, Faculty of Medicine and University Hospital Carl GustavDresdenGermany
| | - Rayk Behrendt
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital BonnBonnGermany
| | | | - Björn H Falkenburger
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
- Deutsches Zentrum für Neurodegenerative ErkrankungenDresdenGermany
| | - Angela Rösen-Wolff
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| |
Collapse
|
3
|
Zhu P, Pfrender EM, Steffeck AWT, Reczek CR, Zhou Y, Thakkar AV, Gupta NR, Kupai A, Willbanks A, Lieber RL, Roy I, Chandel NS, Peek CB. Immunomodulatory role of the stem cell circadian clock in muscle repair. SCIENCE ADVANCES 2025; 11:eadq8538. [PMID: 40043110 PMCID: PMC11881903 DOI: 10.1126/sciadv.adq8538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/30/2025] [Indexed: 03/09/2025]
Abstract
Circadian rhythms orchestrate physiological processes such as metabolism, immune function, and tissue regeneration, aligning them with the optimal time of day (TOD). This study identifies an interplay between the circadian clock within muscle stem cells (SCs) and their capacity to modulate the immune microenvironment during muscle regeneration. We reveal that the SC clock triggers TOD-dependent inflammatory gene transcription after injury, particularly genes related to neutrophil activity and chemotaxis. These responses are driven by cytosolic regeneration of the signaling metabolite nicotinamide adenine dinucleotide (oxidized form) (NAD+), as enhancing cytosolic NAD+ regeneration in SCs is sufficient to induce inflammatory responses that influence muscle regeneration. Mononuclear single-cell sequencing of the regenerating muscle niche further implicates the cytokine CCL2 in mediating SC-neutrophil cross-talk in a TOD-dependent manner. Our findings highlight the intersection between SC metabolic shifts and immune responses within the muscle microenvironment, dictated by circadian rhythms, and underscore the potential for targeting circadian and metabolic pathways to enhance tissue regeneration.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eric M. Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam W. T. Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Colleen R. Reczek
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yalu Zhou
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Feinberg Cardiovascular and Renal Research Institute, Chicago, IL, USA
| | - Abhishek Vijay Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Neha R. Gupta
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ariana Kupai
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Amber Willbanks
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
| | - Richard L. Lieber
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, USA
- Hines VA Hospital, Maywood, IL, USA
| | - Ishan Roy
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Navdeep S. Chandel
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Clara B. Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
4
|
Ueland T, Astrup E, Otterdal K, Lekva T, Janardhanan J, Michelsen AE, Aukrust P, Varghese GM, Damås JK. C-C Motif Ligand 7 and C-C Motif Chemokine Receptor 3 Dysregulation in Patients With Scrub Typhus and Association With Mortality. J Infect Dis 2025; 231:e59-e67. [PMID: 39190223 PMCID: PMC11793035 DOI: 10.1093/infdis/jiae401] [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: 01/09/2024] [Revised: 06/28/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Scrub typhus, caused by Orientia tsutsugamushi, involves infiltration of a mixture of perivascular lymphocytes and macrophages into affected organs. We investigated if this is characterized by chemokine dysregulation. METHODS mRNA expression of chemokines and receptors was screened in whole blood by cDNA microarray in a subgroup of patients and controls. Regulated transcripts were analyzed in plasma by enzyme immunoassays (chemokines) and in whole blood by quantitative polymerase chain reaction (receptors) from patients with scrub typhus (n = 129), patients with similar febrile illness without O tsutsugamushi infection (n = 31), and healthy controls (n = 31). RESULTS cDNA microarray identified dysregulation of the chemokines CCL18 and CCL23 and the receptor CCR3 in severe scrub typhus. Plasma CCL7 (a ligand for CCR3), CCL18, and CCL23 were higher in patients with scrub typhus, with a decline during follow-up. Conversely, mRNA levels of CCR3 and CCR8 (the receptor for CCL18) were decreased in whole blood at hospital admission, followed by an increase during follow-up. CCL7 was independently associated with disease severity. Admission CCL7 levels were associated with short-time mortality. CONCLUSIONS Our findings suggest that CCL7 could represent a hitherto unknown pathogenic mediator in O tsutsugamushi infection, contributing to local and systemic inflammation.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet
- Faculty of Medicine, University of Oslo
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø
| | - Elisabeth Astrup
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet
- Institute of Clinical Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Kari Otterdal
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet
| | - Tove Lekva
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet
| | - Jeshina Janardhanan
- Department of Medicine and Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Annika E Michelsen
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet
- Faculty of Medicine, University of Oslo
| | - Pål Aukrust
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet
- Faculty of Medicine, University of Oslo
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet
| | - George M Varghese
- Department of Medicine and Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Jan K Damås
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim
- Department of Infectious Diseases, St Olavs Hospital, Trondheim, Norway
| |
Collapse
|
5
|
Sarkar S, Han JX, Azzopardi K, Dhar P, Saeed MA, Day S, Ranganathan S, Sutton P. Protease-activated receptor 1 in the pathogenesis of cystic fibrosis. BMJ Open Respir Res 2025; 12:e002960. [PMID: 39832889 PMCID: PMC12004468 DOI: 10.1136/bmjresp-2024-002960] [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: 10/16/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The most common cause of death in those with cystic fibrosis (CF) is respiratory failure due to bronchiectasis resulting from repeated cycles of respiratory infection and inflammation. Protease-activated receptor 1 (PAR1) is a cell surface receptor activated by serine proteases including neutrophil elastase, which is recognised as a potent modulator of inflammation. While PAR1 is known to play an important role in regulating inflammation, nothing is known about any potential role of this receptor in CF pathogenesis. METHODS PAR1 (PAR1-/- ) and intestinal-corrected CFTR (Cftr-/- ) deficient mice were crossed to generate double knock-out (DKO) mutants lacking both PAR1 and CFTR, as well as matching sibling single mutant and wildtype (WT) littermate controls. Mice were weighed weekly to 15 weeks of age; then, the lungs and intestines were examined. RESULTS Cftr-deficient mice gained body weight at a significantly slower rate than WT controls and presented with no lung inflammation, but had increased weights of their ilea and proximal colons. DKO mice (lacking both CFTR and PAR1) gained body weight at a similar rate to Cftr-/- mice but only gained weight in their proximal colons. Weight gain in the ilea of Cftr-/- but not DKO mice was associated with increased ileal levels in the pro-inflammatory cytokine interleukin (IL)-6. CONCLUSIONS This study provides the first evidence of PAR1 contributing to the pathological effects of Cftr deficiency in the intestine and suggests a possible effect of PAR1 on the regulation of IL-6 in CF pathogenesis.
Collapse
Affiliation(s)
- Sohinee Sarkar
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Jia-Xi Han
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Kristy Azzopardi
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Poshmaal Dhar
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Muhammad A Saeed
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Sophie Day
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Sarath Ranganathan
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Philip Sutton
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
6
|
Wilson AL, Moffitt LR, Doran BR, Basri B, Do J, Jobling TW, Plebanski M, Stephens AN, Bilandzic M. Leader cells promote immunosuppression to drive ovarian cancer progression in vivo. Cell Rep 2024; 43:114979. [PMID: 39541210 DOI: 10.1016/j.celrep.2024.114979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/10/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Over 75% of patients with ovarian cancer present with late-stage disease, often accompanied by extensive metastasis. The metastatic cascade is driven by a sub-population of transcriptionally plastic cells known as "leader cells" (LCs), which play a critical role in collective invasion yet remain poorly understood. LCs are marked by the expression of keratin-14 (KRT14), which determines their migratory and invasive capacity in ovarian cancer. This study demonstrates that KRT14+ LCs promote tumor progression through immunosuppression and immune privilege in vivo. In the ID8 syngeneic epithelial ovarian cancer mouse model, tumor-specific loss of KRT14+ LCs impairs tumor progression and metastatic spread without affecting cellular proliferation. Immune profiling shows reduced immunosuppressive regulatory T cells (Tregs) and M2 macrophages and improved CD8+ T cell/Treg ratios in LC knockout (LCKO) mice. Conversely, forced LC overexpression accelerates metastasis and increases the secretion of immunosuppressive chemokines, such as CCL22 and CCL5, highlighting the role of KRT14+ LCs in immune suppression and metastatic progression.
Collapse
Affiliation(s)
- Amy L Wilson
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Laura R Moffitt
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Brittany R Doran
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Bashira Basri
- Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Jennie Do
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Thomas W Jobling
- Monash Medical Centre, Department of Gynecology Oncology, Monash Health, 823-865 Centre Rd, Bentleigh East, VIC 3165, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Andrew N Stephens
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
7
|
Hufnagel M, Rademaekers A, Weisert A, Häberlein H, Franken S. Pharmacological profile of dicaffeoylquinic acids and their role in the treatment of respiratory diseases. Front Pharmacol 2024; 15:1371613. [PMID: 39239645 PMCID: PMC11374715 DOI: 10.3389/fphar.2024.1371613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Dicaffeoylquinic acids (DCQAs) are polyphenolic compounds found in various medicinal plants such as Echinacea species and Hedera helix, whose multi-constituent extracts are used worldwide to treat respiratory diseases. Besides triterpenes, saponins, alkamides, and other constituents, DCQAs are an important group of substances for the pharmacological activity of plant-derived extracts. Therefore, the pharmacological properties of DCQAs have been studied over the last decades, suggesting antioxidative, anti-inflammatory, antimicrobial, hypoglycaemic, cardiovascular protective, neuroprotective, and hepatoprotective effects. However, the beneficial pharmacological profile of DCQAs has not yet been linked to their use in treating respiratory diseases such as acute or even chronic bronchitis. The aim of this review was to assess the potential of DCQAs for respiratory indications based on published in vitro and in vivo pharmacological and pre-clinical data, with particular focus on antioxidative, anti-inflammatory, and respiratory-related effects such as antitussive or antispasmodic properties. A respective literature search revealed a large number of publications on the six DCQA isoforms. Based on this search, a focus was placed on 1,3-, 3,4-, 3,5-, and 4,5-DCQA, as the publications focused mainly on these isomers. Based on the available pre-clinical data, DCQAs trigger cellular mechanisms that are important in the treatment of respiratory diseases such as decreasing NF-κB activation, reducing oxidative stress, or activating the Nrf2 pathway. Taken together, these data suggest an essential role for DCQAs within herbal medicines used for the treatment of respiratory diseases and highlights the need for the identifications of DCQAs as lead substances within such extracts.
Collapse
Affiliation(s)
| | | | - Anika Weisert
- Engelhard Arzneimittel GmbH & Co. KG, Niederdorfelden, Germany
| | - Hanns Häberlein
- Medical Faculty, Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | - Sebastian Franken
- Medical Faculty, Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
8
|
Huang G, Geng Y, Kulur V, Liu N, Liu X, Taghavifar F, Liang J, Noble PW, Jiang D. Arrestin beta 1 Regulates Alveolar Progenitor Renewal and Lung Fibrosis. JOURNAL OF RESPIRATORY BIOLOGY AND TRANSLATIONAL MEDICINE 2024; 1:10006. [PMID: 38736470 PMCID: PMC11087074 DOI: 10.35534/jrbtm.2024.10006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
The molecular mechanisms that regulate progressive pulmonary fibrosis remain poorly understood. Type 2 alveolar epithelial cells (AEC2s) function as adult stem cells in the lung. We previously showed that there is a loss of AEC2s and a failure of AEC2 renewal in the lungs of idiopathic pulmonary fibrosis (IPF) patients. We also reported that beta-arrestins are the key regulators of fibroblast invasion, and beta-arrestin 1 and 2 deficient mice exhibit decreased mortality, decreased matrix deposition, and increased lung function in bleomycin-induced lung fibrosis. However, the role of beta-arrestins in AEC2 regeneration is unclear. In this study, we investigated the role and mechanism of Arrestin beta 1 (ARRB1) in AEC2 renewal and in lung fibrosis. We used conventional deletion as well as cell type-specific deletion of ARRB1 in mice and found that Arrb1 deficiency in fibroblasts protects mice from lung fibrosis, and the knockout mice exhibit enhanced AEC2 regeneration in vivo, suggesting a role of fibroblast-derived ARRB1 in AEC2 renewal. We further found that Arrb1-deficient fibroblasts promotes AEC2 renewal in 3D organoid assays. Mechanistically, we found that CCL7 is among the top downregulated cytokines in Arrb1 deficient fibroblasts and CCL7 inhibits AEC2 regeneration in 3D organoid experiments. Therefore, fibroblast ARRB1 mediates AEC2 renewal, possibly by releasing chemokine CCL7, leading to fibrosis in the lung.
Collapse
Affiliation(s)
- Guanling Huang
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Current Address: GH, Sanofi, 500 Kendall Street, Cambridge, MA 02142, USA
| | - Yan Geng
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Current Address: YG, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
| | - Vrishika Kulur
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ningshan Liu
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xue Liu
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Forough Taghavifar
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jiurong Liang
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W. Noble
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
9
|
Cheetham CJ, McKelvey MC, McAuley DF, Taggart CC. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. Int J Mol Sci 2024; 25:5492. [PMID: 38791530 PMCID: PMC11122108 DOI: 10.3390/ijms25105492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.
Collapse
Affiliation(s)
- Coby J. Cheetham
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK;
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| |
Collapse
|
10
|
Trivedi A, Lu TM, Summers B, Kim K, Rhee AJ, Houghton S, Byers DE, Lis R, Reed HO. Lung lymphatic endothelial cells undergo inflammatory and prothrombotic changes in a model of chronic obstructive pulmonary disease. Front Cell Dev Biol 2024; 12:1344070. [PMID: 38440076 PMCID: PMC10910060 DOI: 10.3389/fcell.2024.1344070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
The lymphatic vasculature regulates lung homeostasis through drainage of fluid and trafficking of immune cells and plays a key role in the response to lung injury in several disease states. We have previously shown that lymphatic dysfunction occurs early in the pathogenesis of chronic obstructive pulmonary disease (COPD) caused by cigarette smoke (CS) and that this is associated with increased thrombin and fibrin clots in lung lymph. However, the direct effects of CS and thrombin on lymphatic endothelial cells (LECs) in COPD are not entirely clear. Studies of the blood vasculature have shown that COPD is associated with increased thrombin after CS exposure that causes endothelial dysfunction characterized by changes in the expression of coagulation factors and leukocyte adhesion proteins. Here, we determined whether similar changes occur in LECs. We used an in vitro cell culture system and treated human lung microvascular lymphatic endothelial cells with cigarette smoke extract (CSE) and/or thrombin. We found that CSE treatment led to decreased fibrinolytic activity in LECs, which was associated with increased expression of plasminogen activator inhibitor 1 (PAI-1). LECs treated with both CSE and thrombin together had a decreased expression of tissue factor pathway inhibitor (TFPI) and increased expression of adhesion molecules. RNA sequencing of lung LECs isolated from mice exposed to CS also showed upregulation of prothrombotic and inflammatory pathways at both acute and chronic exposure time points. Analysis of publicly available single-cell RNA sequencing of LECs as well as immunohistochemical staining of lung tissue from COPD patients supported these data and showed increased expression of inflammatory markers in LECs from COPD patients compared to those from controls. These studies suggest that in parallel with blood vessels, the lymphatic endothelium undergoes inflammatory changes associated with CS exposure and increased thrombin in COPD. Further research is needed to unravel the mechanisms by which these changes affect lymphatic function and drive tissue injury in COPD.
Collapse
Affiliation(s)
- Anjali Trivedi
- Department of Medicine, Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, United States
| | - Tyler M. Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, United States
- Molecular and Cellular Biology Program, SUNY Downstate School of Graduate Studies, Brooklyn, NY, United States
| | - Barbara Summers
- Department of Medicine, Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, United States
| | - Kihwan Kim
- Department of Medicine, Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, United States
| | - Alexander J. Rhee
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Sean Houghton
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Derek E. Byers
- Department of Medicine, Division of Pulmonary and Critical Care, Washington University School of Medicine, St. Louis, MO, United States
| | - Raphaël Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Hasina Outtz Reed
- Department of Medicine, Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, United States
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
11
|
Wohnhaas CT, Baßler K, Watson CK, Shen Y, Leparc GG, Tilp C, Heinemann F, Kind D, Stierstorfer B, Delić D, Brunner T, Gantner F, Schultze JL, Viollet C, Baum P. Monocyte-derived alveolar macrophages are key drivers of smoke-induced lung inflammation and tissue remodeling. Front Immunol 2024; 15:1325090. [PMID: 38348034 PMCID: PMC10859862 DOI: 10.3389/fimmu.2024.1325090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Smoking is a leading risk factor of chronic obstructive pulmonary disease (COPD), that is characterized by chronic lung inflammation, tissue remodeling and emphysema. Although inflammation is critical to COPD pathogenesis, the cellular and molecular basis underlying smoking-induced lung inflammation and pathology remains unclear. Using murine smoke models and single-cell RNA-sequencing, we show that smoking establishes a self-amplifying inflammatory loop characterized by an influx of molecularly heterogeneous neutrophil subsets and excessive recruitment of monocyte-derived alveolar macrophages (MoAM). In contrast to tissue-resident AM, MoAM are absent in homeostasis and characterized by a pro-inflammatory gene signature. Moreover, MoAM represent 46% of AM in emphysematous mice and express markers causally linked to emphysema. We also demonstrate the presence of pro-inflammatory and tissue remodeling associated MoAM orthologs in humans that are significantly increased in emphysematous COPD patients. Inhibition of the IRAK4 kinase depletes a rare inflammatory neutrophil subset, diminishes MoAM recruitment, and alleviates inflammation in the lung of cigarette smoke-exposed mice. This study extends our understanding of the molecular signaling circuits and cellular dynamics in smoking-induced lung inflammation and pathology, highlights the functional consequence of monocyte and neutrophil recruitment, identifies MoAM as key drivers of the inflammatory process, and supports their contribution to pathological tissue remodeling.
Collapse
Affiliation(s)
- Christian T. Wohnhaas
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Kevin Baßler
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Carolin K. Watson
- Immunology & Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Yang Shen
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Germán G. Leparc
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Cornelia Tilp
- Immunology & Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Fabian Heinemann
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David Kind
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Birgit Stierstorfer
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Denis Delić
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Brunner
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Florian Gantner
- Department of Biology, University of Konstanz, Konstanz, Germany
- Translational Medicine & Clinical Pharmacology, C. H. Boehringer Sohn AG & Co. KG, Biberach, Germany
| | - Joachim L. Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and University of Bonn, Bonn, Germany
| | - Coralie Viollet
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Patrick Baum
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
12
|
Farahnak K, Bai YZ, Yokoyama Y, Morkan DB, Liu Z, Amrute JM, De Filippis Falcon A, Terada Y, Liao F, Li W, Shepherd HM, Hachem RR, Puri V, Lavine KJ, Gelman AE, Bharat A, Kreisel D, Nava RG. B cells mediate lung ischemia/reperfusion injury by recruiting classical monocytes via synergistic B cell receptor/TLR4 signaling. J Clin Invest 2024; 134:e170118. [PMID: 38488011 PMCID: PMC10940088 DOI: 10.1172/jci170118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 01/17/2024] [Indexed: 03/18/2024] Open
Abstract
Ischemia/reperfusion injury-mediated (IRI-mediated) primary graft dysfunction (PGD) adversely affects both short- and long-term outcomes after lung transplantation, a procedure that remains the only treatment option for patients suffering from end-stage respiratory failure. While B cells are known to regulate adaptive immune responses, their role in lung IRI is not well understood. Here, we demonstrated by intravital imaging that B cells are rapidly recruited to injured lungs, where they extravasate into the parenchyma. Using hilar clamping and transplant models, we observed that lung-infiltrating B cells produce the monocyte chemokine CCL7 in a TLR4-TRIF-dependent fashion, a critical step contributing to classical monocyte (CM) recruitment and subsequent neutrophil extravasation, resulting in worse lung function. We found that synergistic BCR-TLR4 activation on B cells is required for the recruitment of CMs to the injured lung. Finally, we corroborated our findings in reperfused human lungs, in which we observed a correlation between B cell infiltration and CM recruitment after transplantation. This study describes a role for B cells as critical orchestrators of lung IRI. As B cells can be depleted with currently available agents, our study provides a rationale for clinical trials investigating B cell-targeting therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew E. Gelman
- Department of Surgery
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ankit Bharat
- Department of Surgery, Northwestern University, Chicago, Illinois, USA
| | - Daniel Kreisel
- Department of Surgery
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | |
Collapse
|
13
|
Li J, Fang Z, Xu S, Rao H, Liu J, Lei K, Yang L, Wang C, Zeng Z. The link between neutrophils, NETs, and NLRP3 inflammasomes: The dual effect of CD177 and its therapeutic potential in acute respiratory distress syndrome/acute lung injury. BIOMOLECULES & BIOMEDICINE 2024; 24:798-812. [PMID: 38226808 PMCID: PMC11293216 DOI: 10.17305/bb.2023.10101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 01/17/2024]
Abstract
Neutrophils are important inflammatory effector cells that protect against foreign invasion but also cause self-harm. Numerous neutrophils infiltrate the lungs in acute respiratory distress syndrome/acute lung injury (ARDS/ALI) patients. However, the exact impact of neutrophil infiltration on ARDS's onset and progression remains unclear. To investigate this, we analyzed two ARDS-related datasets from the Gene Expression Omnibus public database and discovered an association between CD177, a neutrophil-specific surface protein, and ARDS progression. We used quantitative flow cytometry to assess CD177+ neutrophils in the peripheral blood of clinical ARDS patients vs healthy controls, finding a significant increase in CD177+ neutrophils percentage among total neutrophils in ARDS patients. This finding was further confirmed in ALI mouse models. Subsequent animal experiments showed that anti-CD177 effectively reduces pulmonary edema, neutrophil infiltration, and inflammatory cytokine release, along with a decrease in reactive oxygen species (ROS) and myeloperoxidase (MPO) levels. We also established an in vitro co-culture system to mimic neutrophil and lung epithelial cell interactions. In the anti-CD177 group, we observed decreased expression of NLRP3, caspase 1, peptidyl arginine deiminase (PAD4), MPO, and ROS, along with a reduction in certain inflammatory cytokines. These results indicate a crucial role for the CD177 gene in ARDS's development and progression. Inhibiting CD177 may help mitigate excessive activation of NLRP3 inflammasomes, ROS, and neutrophil extracellular traps (NETs), thus alleviating ARDS.
Collapse
Affiliation(s)
- Jingying Li
- Department of Critical Care Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Zhansheng Fang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Shumin Xu
- Department of Operating Room, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Haiwei Rao
- Department of Critical Care Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Junzhe Liu
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Kunjian Lei
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Lufei Yang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Chong Wang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Zhenguo Zeng
- Department of Critical Care Medicine, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| |
Collapse
|
14
|
Kim YT, Huh JW, Choi YH, Yoon HK, Nguyen TT, Chun E, Jeong G, Park S, Ahn S, Lee WK, Noh YW, Lee KS, Ahn HS, Lee C, Lee SM, Kim KS, Suh GJ, Jeon K, Kim S, Jin M. Highly secreted tryptophanyl tRNA synthetase 1 as a potential theranostic target for hypercytokinemic severe sepsis. EMBO Mol Med 2024; 16:40-63. [PMID: 38177528 PMCID: PMC10883277 DOI: 10.1038/s44321-023-00004-y] [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: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 01/06/2024] Open
Abstract
Despite intensive clinical and scientific efforts, the mortality rate of sepsis remains high due to the lack of precise biomarkers for patient stratification and therapeutic guidance. Secreted human tryptophanyl-tRNA synthetase 1 (WARS1), an endogenous ligand for Toll-like receptor (TLR) 2 and TLR4 against infection, activates the genes that signify the hyperinflammatory sepsis phenotype. High plasma WARS1 levels stratified the early death of critically ill patients with sepsis, along with elevated levels of cytokines, chemokines, and lactate, as well as increased numbers of absolute neutrophils and monocytes, and higher Sequential Organ Failure Assessment (SOFA) scores. These symptoms were recapitulated in severely ill septic mice with hypercytokinemia. Further, injection of WARS1 into mildly septic mice worsened morbidity and mortality. We created an anti-human WARS1-neutralizing antibody that suppresses proinflammatory cytokine expression in marmosets with endotoxemia. Administration of this antibody into severe septic mice attenuated cytokine storm, organ failure, and early mortality. With antibiotics, the antibody almost completely prevented fatalities. These data imply that blood-circulating WARS1-guided anti-WARS1 therapy may provide a novel theranostic strategy for life-threatening systemic hyperinflammatory sepsis.
Collapse
Affiliation(s)
- Yoon Tae Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Jin Won Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yun Hui Choi
- R&D Center, MirimGENE, Incheon, Republic of Korea
| | | | | | - Eunho Chun
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Geunyeol Jeong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Sunyoung Park
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Sungwoo Ahn
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Won-Kyu Lee
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Young-Woock Noh
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Kyoung Sun Lee
- Non-Clinical Evaluation Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Hee-Sung Ahn
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Cheolju Lee
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Sang Min Lee
- Department of Internal Medicine, Gil Medical Center, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Kyung Su Kim
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Gil Joon Suh
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyeongman Jeon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, Institute for Artificial Intelligence and Biomedical Research, The interdisciplinary graduate program in integrative biotechnology, College of Pharmacy & College of Medicine, Gangnam Severance Hospital, Yonsei University, Incheon, Republic of Korea
| | - Mirim Jin
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
- R&D Center, MirimGENE, Incheon, Republic of Korea.
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.
- Department of Microbiology, College of Medicine, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
15
|
Stumpff JP, Kim SY, McFadden MI, Nishida A, Shirazi R, Steuerman Y, Gat-Viks I, Forero A, Nair MG, Morrison J. Pleural macrophages translocate to the lung during infection to promote improved influenza outcomes. Proc Natl Acad Sci U S A 2023; 120:e2300474120. [PMID: 38100417 PMCID: PMC10743374 DOI: 10.1073/pnas.2300474120] [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: 01/10/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Seasonal influenza results in 3 to 5 million cases of severe disease and 250,000 to 500,000 deaths annually. Macrophages have been implicated in both the resolution and progression of the disease, but the drivers of these outcomes are poorly understood. We probed mouse lung transcriptomic datasets using the Digital Cell Quantifier algorithm to predict immune cell subsets that correlated with mild or severe influenza A virus (IAV) infection outcomes. We identified a unique lung macrophage population that transcriptionally resembled small serosal cavity macrophages and whose presence correlated with mild disease. Until now, the study of serosal macrophage translocation in the context of viral infections has been neglected. Here, we show that pleural macrophages (PMs) migrate from the pleural cavity to the lung after infection with IAV. We found that the depletion of PMs increased morbidity and pulmonary inflammation. There were increased proinflammatory cytokines in the pleural cavity and an influx of neutrophils within the lung. Our results show that PMs are recruited to the lung during IAV infection and contribute to recovery from influenza. This study expands our knowledge of PM plasticity and identifies a source of lung macrophages independent of monocyte recruitment and local proliferation.
Collapse
Affiliation(s)
- James P. Stumpff
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| | - Sang Yong Kim
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA92521
| | - Matthew I. McFadden
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH43210
- Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, WA98109
| | - Roksana Shirazi
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| | - Yael Steuerman
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Irit Gat-Viks
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Adriana Forero
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH43210
- Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Meera G. Nair
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA92521
| | - Juliet Morrison
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| |
Collapse
|
16
|
Fan H, Liu J, Sun J, Feng G, Li J. Advances in the study of B cells in renal ischemia-reperfusion injury. Front Immunol 2023; 14:1216094. [PMID: 38022595 PMCID: PMC10646530 DOI: 10.3389/fimmu.2023.1216094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a non-negligible clinical challenge for clinicians in surgeries such as renal transplantation. Functional loss of renal tubular epithelial cell (TEC) in IRI leads to the development of acute kidney injury, delayed graft function (DGF), and allograft rejection. The available evidence indicates that cellular oxidative stress, cell death, microvascular dysfunction, and immune response play an important role in the pathogenesis of IRI. A variety of immune cells, including macrophages and T cells, are actively involved in the progression of IRI in the immune response. The role of B cells in IRI has been relatively less studied, but there is a growing body of evidence for the involvement of B cells, which involve in the development of IRI through innate immune responses, adaptive immune responses, and negative immune regulation. Therefore, therapies targeting B cells may be a potential direction to mitigate IRI. In this review, we summarize the current state of research on the role of B cells in IRI, explore the potential effects of different B cell subsets in the pathogenesis of IRI, and discuss possible targets of B cells for therapeutic aim in renal IRI.
Collapse
Affiliation(s)
- Hongzhao Fan
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia Liu
- Dietetics Teaching and Research Section, Henan Medical College, Xinzheng, China
| | - Jiajia Sun
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guiwen Feng
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinfeng Li
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Desai RW, Demir K, Tsolakos N, Moir-Savitz TR, Gaworski CL, Weil R, Oldham MJ, Lalonde G. Comparison of the toxicological potential of two JUUL ENDS products to reference cigarette 3R4F and filtered air in a 90-day nose-only inhalation toxicity study. Food Chem Toxicol 2023; 179:113917. [PMID: 37451597 DOI: 10.1016/j.fct.2023.113917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/13/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023]
Abstract
Electronic nicotine delivery systems (ENDS) are generally recognized as less harmful alternatives for those who would otherwise continue to smoke cigarettes. The potential toxicity of aerosols generated from JUUL Device and Virginia Tobacco (VT3) or Menthol (ME3) JUULpods at 3.0% nicotine concentration was assessed in rats exposed at target aerosol concentrations of 1400 μg/L for up to 6 h/day on a 5 day/week basis for at least 90 days (general accordance with OECD 413). 3R4F reference cigarette smoke (250 μg/L) and Filtered Air were used as comparators. JUUL ENDS product aerosol exposures at >5x the 3R4F cigarette smoke level resulted in greater plasma nicotine and cotinine levels (up to 2x). Notable cigarette smoke related effects included pronounced body weight reductions in male rats, pulmonary inflammation evidenced by elevated lactate dehydrogenase, pro-inflammatory cytokines and neutrophils in bronchoalveolar lavage fluid, increased heart and lung weights, and minimal to marked respiratory tract histopathology. In contrast, ENDS aerosol exposed animals had minimal body weight changes, no measurable inflammatory changes and minimal to mild laryngeal squamous metaplasia. Despite the higher exposure levels, VT3 and ME3 did not result in significant toxicity or appreciable respiratory histopathology relative to 3R4F cigarette smoke following 90 days administration.
Collapse
Affiliation(s)
| | | | - Nikos Tsolakos
- Protatonce Ltd, National Centre of Scientific Research Demokritos, Patriarchou Grigoriou E' & Neapoleos 27, Technological Park Lefkippos, Bldg 27, 15341, Ag. Paraskevi, Attiki, Greece
| | - Tessa R Moir-Savitz
- AmplifyBio, 1425 NE Plain City-Georgesville Rd, West Jefferson, OH, 43162, USA
| | | | | | | | | |
Collapse
|
18
|
Chang LA, Choi A, Rathnasinghe R, Warang P, Noureddine M, Jangra S, Chen Y, De Geest BG, Schotsaert M. Influenza breakthrough infection in vaccinated mice is characterized by non-pathological lung eosinophilia. Front Immunol 2023; 14:1217181. [PMID: 37600776 PMCID: PMC10437116 DOI: 10.3389/fimmu.2023.1217181] [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: 05/04/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Eosinophils are important mediators of mucosal tissue homeostasis, anti-helminth responses, and allergy. Lung eosinophilia has previously been linked to aberrant Type 2-skewed T cell responses to respiratory viral infection and may also be a consequence of vaccine-associated enhanced respiratory disease (VAERD), particularly in the case of respiratory syncytial virus (RSV) and the formalin-inactivated RSV vaccine. We previously reported a dose-dependent recruitment of eosinophils to the lungs of mice vaccinated with alum-adjuvanted trivalent inactivated influenza vaccine (TIV) following a sublethal, vaccine-matched H1N1 (A/New Caledonia/20/1999; NC99) influenza challenge. Given the differential role of eosinophil subset on immune function, we conducted the investigations herein to phenotype the lung eosinophils observed in our model of influenza breakthrough infection. Here, we demonstrate that eosinophil influx into the lungs of vaccinated mice is adjuvant- and sex-independent, and only present after vaccine-matched sublethal influenza challenge but not in mock-challenged mice. Furthermore, vaccinated and challenged mice had a compositional shift towards more inflammatory eosinophils (iEos) compared to resident eosinophils (rEos), resembling the shift observed in ovalbumin (OVA)-sensitized allergic control mice, however without any evidence of enhanced morbidity or aberrant inflammation in lung cytokine/chemokine signatures. Furthermore, we saw a lung eosinophil influx in the context of a vaccine-mismatched challenge. Additional layers of heterogeneity in the eosinophil compartment were observed via unsupervised clustering analysis of flow cytometry data. Our collective findings are a starting point for more in-depth phenotypic and functional characterization of lung eosinophil subsets in the context of vaccine- and infection-induced immunity.
Collapse
Affiliation(s)
- Lauren A. Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Angela Choi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Raveen Rathnasinghe
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Moataz Noureddine
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | | | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
19
|
Kramer ED, Tzetzo SL, Colligan SH, Hensen ML, Brackett CM, Clausen BE, Taketo MM, Abrams SI. β-Catenin signaling in alveolar macrophages enhances lung metastasis through a TNF-dependent mechanism. JCI Insight 2023; 8:e160978. [PMID: 37092550 PMCID: PMC10243816 DOI: 10.1172/jci.insight.160978] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 03/08/2023] [Indexed: 04/25/2023] Open
Abstract
The main cause of malignancy-related mortality is metastasis. Although metastatic progression is driven by diverse tumor-intrinsic mechanisms, there is a growing appreciation for the contribution of tumor-extrinsic elements of the tumor microenvironment, especially macrophages, which correlate with poor clinical outcomes. Macrophages consist of bone marrow-derived and tissue-resident populations. In contrast to bone marrow-derived macrophages, the transcriptional pathways that govern the pro-metastatic activities of tissue-resident macrophages (TRMs) remain less clear. Alveolar macrophages (AMs) are a TRM population with critical roles in tissue homeostasis and metastasis. Wnt/β-catenin signaling is a hallmark of cancer and has been identified as a pathologic regulator of AMs in infection. We tested the hypothesis that β-catenin expression in AMs enhances metastasis in solid tumor models. Using a genetic β-catenin gain-of-function approach, we demonstrated that (a) enhanced β-catenin in AMs heightened lung metastasis; (b) β-catenin activity in AMs drove a dysregulated inflammatory program strongly associated with Tnf expression; and (c) localized TNF-α blockade abrogated this metastatic outcome. Last, β-catenin gene CTNNB1 and TNF expression levels were positively correlated in AMs of patients with lung cancer. Overall, our findings revealed a Wnt/β-catenin/TNF-α pro-metastatic axis in AMs with potential therapeutic implications against tumors refractory to the antineoplastic actions of TNF-α.
Collapse
Affiliation(s)
| | | | | | | | - Craig M. Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Björn E. Clausen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Makoto M. Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
20
|
Amadeo F, Hanson V, Liptrott NJ, Wilm B, Murray P, Taylor A. Fate of intravenously administered umbilical cord mesenchymal stromal cells and interactions with the host's immune system. Biomed Pharmacother 2023; 159:114191. [PMID: 36623449 DOI: 10.1016/j.biopha.2022.114191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells showing promise in pre-clinical studies and currently used in many clinical trials. The regenerative potential of MSCs is mediated, at least in part, by direct and indirect immunomodulatory processes. However, the mechanism of action is not fully understood yet, and there are still concerns about possible undesired negative effects associated with the administration of living cells. In this study, we (i) compare the long-term fate and safety of umbilical cord (UC-)MSCs administered to immunocompetent and immunocompromised (severe combined immunodeficient (SCID) and non-obese diabetic (NOD)/SCID) animals, and (ii) investigate the immunological response of the host to the administered cells. Intravenous administration of firefly luciferase expressing UC-MSCs revealed that the cells get trapped in the lungs of both immunocompetent and immunocompromised animals, with > 95% of the cells disappearing within 72 h after administration. In 27% of the SCID and 45% of the NOD/SCID, a small fraction of the cells lived up to day 14 but in most cases they all disappeared earlier. One NOD/SCID mouse showed a weak signal up to day 31. Immunocompetent mice displayed elevated percentages of neutrophils in the lungs, the blood, and the spleen 2 h after the administration of the cells. The concentration of neutrophil chemoattractants (MCP1, CCL7, Gro-α and IP-10) were also increased in the plasma of the animals 2 h after the administration of the MSCs. Our results suggest that although the UC-MSCs are short-lived in mice, they still result in an immunological response that might contribute to a therapeutic effect.
Collapse
Affiliation(s)
- Francesco Amadeo
- Cellular Therapies Laboratory, NHS Blood and Transplant, Liverpool, UK; Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool, UK; Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Vivien Hanson
- Cellular Therapies Laboratory, NHS Blood and Transplant, Liverpool, UK
| | - Neill J Liptrott
- Immunocompatibility Group, Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool, UK; Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool, UK; Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Arthur Taylor
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool, UK; Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK.
| |
Collapse
|
21
|
Zhang H, Yang K, Chen F, Liu Q, Ni J, Cao W, Hua Y, He F, Liu Z, Li L, Fan G. Role of the CCL2-CCR2 axis in cardiovascular disease: Pathogenesis and clinical implications. Front Immunol 2022; 13:975367. [PMID: 36110847 PMCID: PMC9470149 DOI: 10.3389/fimmu.2022.975367] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
The CCL2-CCR2 axis is one of the major chemokine signaling pathways that has received special attention because of its function in the development and progression of cardiovascular disease. Numerous investigations have been performed over the past decades to explore the function of the CCL2-CCR2 signaling axis in cardiovascular disease. Laboratory data on the CCL2-CCR2 axis for cardiovascular disease have shown satisfactory outcomes, yet its clinical translation remains challenging. In this article, we describe the mechanisms of action of the CCL2-CCR2 axis in the development and evolution of cardiovascular diseases including heart failure, atherosclerosis and coronary atherosclerotic heart disease, hypertension and myocardial disease. Laboratory and clinical data on the use of the CCL2-CCR2 pathway as a targeted therapy for cardiovascular diseases are summarized. The potential of the CCL2-CCR2 axis in the treatment of cardiovascular diseases is explored.
Collapse
Affiliation(s)
- Haixia Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Ke Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qianqian Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Weilong Cao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng He
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, China
| | - Zhihao Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Lan Li, ; Guanwei Fan,
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, China
- *Correspondence: Lan Li, ; Guanwei Fan,
| |
Collapse
|
22
|
Biryukov SS, Cote CK, Klimko CP, Dankmeyer JL, Rill NO, Shoe JL, Hunter M, Shamsuddin Z, Velez I, Hedrick ZM, Rosario-Acevedo R, Talyansky Y, Schmidt LK, Orne CE, Fetterer DP, Burtnick MN, Brett PJ, Welkos SL, DeShazer D. Evaluation of two different vaccine platforms for immunization against melioidosis and glanders. Front Microbiol 2022; 13:965518. [PMID: 36060742 PMCID: PMC9428723 DOI: 10.3389/fmicb.2022.965518] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Burkholderia pseudomallei and the closely related species, Burkholderia mallei, produce similar multifaceted diseases which range from rapidly fatal to protracted and chronic, and are a major cause of mortality in endemic regions. Besides causing natural infections, both microbes are Tier 1 potential biothreat agents. Antibiotic treatment is prolonged with variable results, hence effective vaccines are urgently needed. The purpose of our studies was to compare candidate vaccines that target both melioidosis and glanders to identify the most efficacious one(s) and define residual requirements for their transition to the non-human primate aerosol model. Studies were conducted in the C57BL/6 mouse model to evaluate the humoral and cell-mediated immune response and protective efficacy of three Burkholderia vaccine candidates against lethal aerosol challenges with B. pseudomallei K96243, B. pseudomallei MSHR5855, and B. mallei FMH. The recombinant vaccines generated significant immune responses to the vaccine antigens, and the live attenuated vaccine generated a greater immune response to OPS and the whole bacterial cells. Regardless of the candidate vaccine evaluated, the protection of mice was associated with a dampened cytokine response within the lungs after exposure to aerosolized bacteria. Despite being delivered by two different platforms and generating distinct immune responses, two experimental vaccines, a capsule conjugate + Hcp1 subunit vaccine and the live B. pseudomallei 668 ΔilvI strain, provided significant protection and were down-selected for further investigation and advanced development.
Collapse
Affiliation(s)
- Sergei S. Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
- *Correspondence: Christopher K. Cote
| | - Christopher P. Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Jennifer L. Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Nathaniel O. Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Jennifer L. Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Zain Shamsuddin
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Ivan Velez
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Zander M. Hedrick
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Raysa Rosario-Acevedo
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Yuli Talyansky
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Lindsey K. Schmidt
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - Caitlyn E. Orne
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - David P. Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Mary N. Burtnick
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Paul J. Brett
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Susan L. Welkos
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - David DeShazer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
- David DeShazer
| |
Collapse
|
23
|
Talsma AD, Niemi JP, Pachter JS, Zigmond RE. The primary macrophage chemokine, CCL2, is not necessary after a peripheral nerve injury for macrophage recruitment and activation or for conditioning lesion enhanced peripheral regeneration. J Neuroinflammation 2022; 19:179. [PMID: 35820932 PMCID: PMC9277969 DOI: 10.1186/s12974-022-02497-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/23/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Peripheral nerve injuries stimulate the regenerative capacity of injured neurons through a neuroimmune phenomenon termed the conditioning lesion (CL) response. This response depends on macrophage accumulation in affected dorsal root ganglia (DRGs) and peripheral nerves. The macrophage chemokine CCL2 is upregulated after injury and is allegedly required for stimulating macrophage recruitment and pro-regenerative signaling through its receptor, CCR2. In these tissues, CCL2 is putatively produced by neurons in the DRG and Schwann cells in the distal nerve. METHODS Ccl2fl/fl mice were crossed with Advillin-Cre, P0-Cre, or both to create conditional Ccl2 knockouts (CKOs) in sensory neurons, Schwann cells, or both to hypothetically remove CCL2 and macrophages from DRGs, nerves or both. CCL2 was localized using Ccl2-RFPfl/fl mice. CCL2-CCR2 signaling was further examined using global Ccl2 KOs and Ccr2gfp knock-in/knock-outs. Unilateral sciatic nerve transection was used as the injury model, and at various timepoints, chemokine expression, macrophage accumulation and function, and in vivo regeneration were examined using qPCR, immunohistochemistry, and luxol fast blue staining. RESULTS Surprisingly, in all CKOs, DRG Ccl2 gene expression was decreased, while nerve Ccl2 was not. CCL2-RFP reporter mice revealed CCL2 expression in several cell types beyond the expected neurons and Schwann cells. Furthermore, macrophage accumulation, myelin clearance, and in vivo regeneration were unaffected in all CKOs, suggesting CCL2 may not be necessary for the CL response. Indeed, Ccl2 global knockout mice showed normal macrophage accumulation, myelin clearance, and in vivo regeneration, indicating these responses do not require CCL2. CCR2 ligands, Ccl7 and Ccl12, were upregulated after nerve injury and perhaps could compensate for the absence of Ccl2. Finally, Ccr2gfp knock-in/knock-out animals were used to differentiate resident and recruited macrophages in the injured tissues. Ccr2gfp/gfp KOs showed a 50% decrease in macrophages in the distal nerve compared to controls with a relative increase in resident macrophages. In the DRG there was a small but insignificant decrease in macrophages. CONCLUSIONS CCL2 is not necessary for macrophage accumulation, myelin clearance, and axon regeneration in the peripheral nervous system. Without CCL2, other CCR2 chemokines, resident macrophage proliferation, and CCR2-independent monocyte recruitment can compensate and allow for normal macrophage accumulation.
Collapse
Affiliation(s)
- Aaron D Talsma
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Jon P Niemi
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Joel S Pachter
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, 06030-6125, USA
| | - Richard E Zigmond
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA.
| |
Collapse
|
24
|
McKelvey MC, Abladey AA, Small DM, Doherty DF, Williams R, Scott A, Spek CA, Borensztajn KS, Holsinger L, Booth R, O'Kane CM, McAuley DF, Taggart CC, Weldon S. Cathepsin S Contributes to Lung Inflammation in Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med 2022; 205:769-782. [PMID: 35073247 DOI: 10.1164/rccm.202107-1631oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Although the cysteine protease cathepsin S has been implicated in the pathogenesis of several inflammatory lung diseases, its role has not been examined in the context of acute respiratory distress syndrome, a condition that still lacks specific and effective pharmacological treatments. Objectives: To characterize the status of cathepsin S in acute lung inflammation and examine the role of cathepsin S in disease pathogenesis. Methods: Human and mouse model BAL fluid samples were analyzed for the presence and activity of cathepsin S and its endogenous inhibitors. Recombinant cathepsin S was instilled directly into the lungs of mice. The effects of cathepsin S knockout and pharmacological inhibition were examined in two models of acute lung injury. Protease-activated receptor-1 antagonism was used to test a possible mechanism for cathepsin S-mediated inflammation. Measurements and Main Results: Pulmonary cathepsin S concentrations and activity were elevated in acute respiratory distress syndrome, a phenotype possibly exacerbated by the loss of the endogenous antiprotease cystatin SN. Direct cathepsin S instillation into the lungs induced key pathologies of acute respiratory distress syndrome, including neutrophilia and alveolar leakage. Conversely, in murine models of acute lung injury, genetic knockdown and prophylactic or therapeutic inhibition of cathepsin S reduced neutrophil recruitment and protein leakage. Cathepsin S may partly mediate its pathogenic effects via protease-activated receptor-1, because antagonism of this receptor abrogated cathepsin S-induced airway inflammation. Conclusions: Cathepsin S contributes to acute lung injury and may represent a novel therapeutic target for acute respiratory distress syndrome.
Collapse
Affiliation(s)
| | | | | | | | - Richard Williams
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Aaron Scott
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, England, United Kingdom
| | - C Arnold Spek
- Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Keren S Borensztajn
- INSERM UMRS_933, Université Pierre et Marie Curie, Hôpital Trousseau, Paris, France; and
| | | | | | | | | | | | | |
Collapse
|
25
|
Martynova E, Hamza S, Markelova M, Garanina E, Davidyuk Y, Shakirova V, Kaushal N, Baranwal M, Stott-Marshall RJ, Foster TL, Rizvanov A, Khaiboullina S. Immunogenic SARS-CoV-2 S and N Protein Peptide and Cytokine Combinations as Biomarkers for Early Prediction of Fatal COVID-19. Front Immunol 2022; 13:830715. [PMID: 35386707 PMCID: PMC8979210 DOI: 10.3389/fimmu.2022.830715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/14/2022] [Indexed: 01/05/2023] Open
Abstract
Early indications of the likelihood of severe coronavirus disease 2019 COVID-19 can influence treatments and could improve clinical outcomes. However, knowledge on the prediction markers of COVID-19 fatality risks remains limited. Here, we analyzed and quantified the reactivity of serum samples from acute (non-fatal and fatal) and convalescent COVID-19 patients with the spike surface glycoprotein (S protein) and nucleocapsid phosphoprotein (N protein) SARS-CoV-2 peptide libraries. Cytokine activation was also analyzed. We demonstrated that IgM from fatal COVID-19 serum reacted with several N protein peptides. In contrast, IgM from non-fatal serum reacted more with S protein peptides. Further, higher levels of pro-inflammatory cytokines were found in fatal COVID-19 serum compared to non-fatal. Many of these cytokines were pro-inflammatory and chemokines. Differences in IgG reactivity from fatal and non-fatal COVID-19 sera were also demonstrated. Additionally, the longitudinal analysis of IgG reactivity with SARS-CoV-2 S and N protein identified peptides with the highest longevity in humoral immune response. Finally, using IgM antibody reactivity with S and N SARS-CoV-2 peptides and selected cytokines, we have identified a panel of biomarkers specific to patients with a higher risk of fatal COVID-19 compared with that of patients who survive. This panel could be used for the early prediction of COVID-19 fatality risk.
Collapse
Affiliation(s)
- Ekaterina Martynova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Shaimaa Hamza
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Maria Markelova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Ekaterina Garanina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Yuriy Davidyuk
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Venera Shakirova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan, Russia
| | - Neha Kaushal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Robert J. Stott-Marshall
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, Wolfson Centre for Global Virus Research, University of Nottingham, Loughborough, United Kingdom
| | - Toshana L. Foster
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, Wolfson Centre for Global Virus Research, University of Nottingham, Loughborough, United Kingdom
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Svetlana Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
26
|
She S, Ren L, Chen P, Wang M, Chen D, Wang Y, Chen H. Functional Roles of Chemokine Receptor CCR2 and Its Ligands in Liver Disease. Front Immunol 2022; 13:812431. [PMID: 35281057 PMCID: PMC8913720 DOI: 10.3389/fimmu.2022.812431] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a family of cytokines that orchestrate the migration and positioning of immune cells within tissues and are critical for the function of the immune system. CCR2 participates in liver pathology, including acute liver injury, chronic hepatitis, fibrosis/cirrhosis, and tumor progression, by mediating the recruitment of immune cells to inflammation and tumor sites. Although a variety of chemokines have been well studied in various diseases, there is no comprehensive review presenting the roles of all known chemokine ligands of CCR2 (CCL2, CCL7, CCL8, CCL12, CCL13, CCL16, and PSMP) in liver disease, and this review aims to fill this gap. The introduction of each chemokine includes its discovery, its corresponding chemotactic receptors, physiological functions and roles in inflammation and tumors, and its impact on different immune cell subgroups.
Collapse
Affiliation(s)
- Shaoping She
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People’s Hospital, Beijing, China
| | - Liying Ren
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Pu Chen
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People’s Hospital, Beijing, China
| | - Mingyang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Dongbo Chen
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People’s Hospital, Beijing, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Hongsong Chen
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People’s Hospital, Beijing, China
- *Correspondence: Hongsong Chen,
| |
Collapse
|
27
|
Tirunavalli SK, Gourishetti K, Kotipalli RSS, Kuncha M, Kathirvel M, Kaur R, Jerald MK, Sistla R, Andugulapati SB. Dehydrozingerone ameliorates Lipopolysaccharide induced acute respiratory distress syndrome by inhibiting cytokine storm, oxidative stress via modulating the MAPK/NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153729. [PMID: 34517257 PMCID: PMC8390101 DOI: 10.1016/j.phymed.2021.153729] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 06/01/2023]
Abstract
BACKGROUND Inflammation-mediated lung injury is a major cause of health problems in many countries and has been the leading cause of morbidity/mortality in intensive care units. In the current COVID-19 pandemic, the majority of the patients experienced serious pneumonia resulting from inflammation (Acute respiratory distress syndrome/ARDS). Pathogenic infections cause cytokine release syndrome (CRS) by hyperactivation of immune cells, which in turn release excessive cytokines causing ARDS. Currently, there are no standard therapies for viral, bacterial or pathogen-mediated CRS. PURPOSE This study aimed to investigate and validate the protective effects of Dehydrozingerone (DHZ) against LPS induced lung cell injury by in-vitro and in-vivo models and to gain insights into the molecular mechanisms that mediate these therapeutic effects. METHODS The therapeutic activity of DHZ was determined in in-vitro models by pre-treating the cells with DHZ and exposed to LPS to stimulate the inflammatory cascade of events. We analysed the effect of DHZ on LPS induced inflammatory cytokines, chemokines and cell damage markers expression/levels using various cell lines. We performed gene expression, ELISA, and western blot analysis to elucidate the effect of DHZ on inflammation and its modulation of MAPK and NF-κB pathways. Further, the prophylactic and therapeutic effect of DHZ was evaluated against the LPS induced ARDS model in rats. RESULTS DHZ significantly (p < 0.01) attenuated the LPS induced ROS, inflammatory cytokine, chemokine gene expression and protein release in macrophages. Similarly, DHZ treatment protected the lung epithelial and endothelial cells by mitigating the LPS induced inflammatory events in a dose-dependent manner. In vivo analysis showed that DHZ treatment significantly (p < 0.001) mitigated the LPS induced ARDS pathophysiology of increase in the inflammatory cells in BALF, inflammatory cytokine and chemokines in lung tissues. LPS stimulated neutrophil-mediated events, apoptosis, alveolar wall thickening and alveolar inflammation were profoundly reduced by DHZ treatment in a rat model. CONCLUSION This study demonstrates for the first time that DHZ has the potential to ameliorate LPS induced ARDS by inhibiting cytokine storm and oxidative through modulating the MAPK and NF-κB pathways. This data provides pre-clinical support to develop DHZ as a potential therapeutic agent against ARDS.
Collapse
Affiliation(s)
- Satya Krishna Tirunavalli
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Karthik Gourishetti
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | | | - Madusudhana Kuncha
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | | | - Rajwinder Kaur
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | - Mahesh Kumar Jerald
- CSIR - Centre for Cellular & Molecular Biology (CCMB), Hyderabad 500 007, India
| | - Ramakrishna Sistla
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Sai Balaji Andugulapati
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India.
| |
Collapse
|
28
|
Bhalerao A, Raut S, Noorani B, Mancuso S, Cucullo L. Molecular Mechanisms of Multi-Organ Failure in COVID-19 and Potential of Stem Cell Therapy. Cells 2021; 10:2878. [PMID: 34831101 PMCID: PMC8616204 DOI: 10.3390/cells10112878] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 01/08/2023] Open
Abstract
As the number of confirmed cases and deaths occurring from Coronavirus disease 2019 (COVID-19) surges worldwide, health experts are striving hard to fully comprehend the extent of damage caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although COVID-19 primarily manifests itself in the form of severe respiratory distress, it is also known to cause systemic damage to almost all major organs and organ systems within the body. In this review, we discuss the molecular mechanisms leading to multi-organ failure seen in COVID-19 patients. We also examine the potential of stem cell therapy in treating COVID-19 multi-organ failure cases.
Collapse
Affiliation(s)
- Aditya Bhalerao
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA
| | - Snehal Raut
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Salvatore Mancuso
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
29
|
Chu SJ, Tang SE, Pao HP, Wu SY, Liao WI. Protease-Activated Receptor-1 Antagonist Protects Against Lung Ischemia/Reperfusion Injury. Front Pharmacol 2021; 12:752507. [PMID: 34658893 PMCID: PMC8514687 DOI: 10.3389/fphar.2021.752507] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/20/2021] [Indexed: 01/14/2023] Open
Abstract
Protease-activated receptor (PAR)-1 is a thrombin-activated receptor that plays an essential role in ischemia/reperfusion (IR)-induced acute inflammation. PAR-1 antagonists have been shown to alleviate injuries in various IR models. However, the effect of PAR-1 antagonists on IR-induced acute lung injury (ALI) has not yet been elucidated. This study aimed to investigate whether PAR-1 inhibition could attenuate lung IR injury. Lung IR was induced in an isolated perfused rat lung model. Male rats were treated with the specific PAR-1 antagonist SCH530348 (vorapaxar) or vehicle, followed by ischemia for 40 min and reperfusion for 60 min. To examine the role of PAR-1 and the mechanism of SCH530348 in lung IR injury, western blotting and immunohistochemical analysis of lung tissue were performed. In vitro, mouse lung epithelial cells (MLE-12) were treated with SCH530348 or vehicle and subjected to hypoxia-reoxygenation (HR). We found that SCH530348 decreased lung edema and neutrophil infiltration, attenuated thrombin production, reduced inflammatory factors, including cytokine-induced neutrophil chemoattractant-1, interleukin-6 and tumor necrosis factor-α, mitigated lung cell apoptosis, and downregulated the phosphoinositide 3-kinase (PI3K), nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in IR-injured lungs. In addition, SCH530348 prevented HR-induced NF-κB activation and inflammatory chemokine production in MLE12 cells. Our results demonstrate that SCH530348 exerts protective effects by blocking PAR-1 expression and modulating the downstream PI3K, NF-κB and MAPK pathways. These findings indicate that the PAR-1 antagonist protects against IR-induced ALI and is a potential therapeutic candidate for lung protection following IR injury.
Collapse
Affiliation(s)
- Shi-Jye Chu
- Department of Internal Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Shih-En Tang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan.,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| |
Collapse
|
30
|
Kong M, Dong W, Zhu Y, Fan Z, Miao X, Guo Y, Li C, Duan Y, Lu Y, Li Z, Xu Y. Redox-sensitive activation of CCL7 by BRG1 in hepatocytes during liver injury. Redox Biol 2021; 46:102079. [PMID: 34454163 PMCID: PMC8406035 DOI: 10.1016/j.redox.2021.102079] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/04/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Liver injuries induced by various stimuli share in common an acute inflammatory response, in which circulating macrophages home to the liver parenchyma to participate in the regulation of repair, regeneration, and fibrosis. In the present study we investigated the role of hepatocyte-derived C-C motif ligand 7 (CCL7) in macrophage migration during liver injury focusing on its transcriptional regulation. We report that CCL7 expression was up-regulated in the liver by lipopolysaccharide (LPS) injection (acute liver injury) or methionine-and-choline-deficient (MCD) diet feeding (chronic liver injury) paralleling increased macrophage infiltration. CCL7 expression was also inducible in hepatocytes, but not in hepatic stellate cells or in Kupffer cells, by LPS treatment or exposure to palmitate in vitro. Hepatocyte-specific deletion of Brahma-related gene 1 (BRG1), a chromatin remodeling protein, resulted in a concomitant loss of CCL7 induction and macrophage infiltration in the murine livers. Of interest, BRG1-induced CCL7 transcription and macrophage migration was completely blocked by the antioxidant N-acetylcystine. Further analyses revealed that BRG1 interacted with activator protein 1 (AP-1) to regulate CCL7 transcription in hepatocytes in a redox-sensitive manner mediated in part by casein kinase 2 (CK2)-catalyzed phosphorylation of BRG1. Importantly, a positive correlation between BRG1/CCL7 expression and macrophage infiltration was identified in human liver biopsy specimens. In conclusion, our data unveil a novel role for BRG1 as a redox-sensitive activator of CCL7 transcription.
Collapse
Affiliation(s)
- Ming Kong
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Wenhui Dong
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yuwen Zhu
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Zhiwen Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Xiulian Miao
- College of Life Sciences and Institute of Biomedical Research, Liaocheng University, China
| | - Yan Guo
- College of Life Sciences and Institute of Biomedical Research, Liaocheng University, China
| | - Chengping Li
- College of Life Sciences and Institute of Biomedical Research, Liaocheng University, China
| | - Yunfei Duan
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Changzhou, The Third Hospital Affiliated to Soochow University, Changzhou, China
| | - Yunjie Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Changzhou, The Third Hospital Affiliated to Soochow University, Changzhou, China.
| | - Zilong Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.
| | - Yong Xu
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; College of Life Sciences and Institute of Biomedical Research, Liaocheng University, China.
| |
Collapse
|
31
|
Wang F, Hou W, Xiao C, Hao Y, Su N, Deng Y, Wang J, Yu L, Xie JM, Xiong JW, Luo Y. Endothelial cell membrane-based biosurface for targeted delivery to acute injury: analysis of leukocyte-mediated nanoparticle transportation. NANOSCALE 2021; 13:14636-14643. [PMID: 34558568 DOI: 10.1039/d1nr04181a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Mimicking and leveraging biological structures and materials provide important approaches to develop functional vehicles for drug delivery. Taking advantage of the affinity and adhesion between the activated endothelial cells and innate immune cells during inflammatory responses, hybrid polyester nanoparticles coated with endothelial cell membranes (EM-P) containing adhesion molecules were fabricated and their capability as vehicles to travel to the acute injury sites through leukocyte-mediated processes was investigated. The in vivo studies and quantitative analyses performed through the lung-inflammation mouse models demonstrated that the EM-Ps preferentially interacted with the neutrophils and monocytes in the circulation and the cellular membrane-based biosurface improved the nanoparticle transportation to the inflamed lung possibly via the motility of neutrophils. Utilizing the transgenic zebrafish model, the leukocyte-mediated transportation and biodistribution of EM-Ps were further visualized in real time at the whole-organism level. Endothelial membranes provided a new biosurface for developing biomimetic vehicles to allow the immune cell-mediated transportation and may enable advanced systems for active and highly efficient drug delivery.
Collapse
Affiliation(s)
- Fang Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Wenda Hou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Chenglu Xiao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Yaoyao Hao
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Ni Su
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Yu Deng
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jieting Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Luying Yu
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jing-Ming Xie
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America 02155
| |
Collapse
|
32
|
Xue J, Zhang Y, Zhang J, Zhu Z, Lv Q, Su J. Astrocyte-derived CCL7 promotes microglia-mediated inflammation following traumatic brain injury. Int Immunopharmacol 2021; 99:107975. [PMID: 34293712 DOI: 10.1016/j.intimp.2021.107975] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 11/18/2022]
Abstract
Microglia are immune cells of the central nervous system that mediate neuroinflammation. It is widely known that microglia-mediated inflammation in the brain contribute to the widespread tissue damage and neurological deficits in traumatic brain injury (TBI). However, the mechanisms responsible for this inflammatory response remain elusive. Here, we investigated the role of astrocyte-derived chemokine (C-C motif) ligand 7 (CCL7) in microglial-controlled inflammation following TBI. Our results demonstrated that astrocyte-derived CCL7 induced microglial activation and the release of proinflammatory mediators in the cortex and serum of rats that underwent experimental TBI. Furthermore, CCL7 knockout improved microglia-controlled inflammation, brain morphology and neurological dysfunction following TBI. In vitro, CCL7-siRNA attenuated the LPS-induced expression of pro-inflammatory markers in the co-culture of microglia and astrocytes. Collectively, our findings uncover an important role for astrocyte-derived CCL7 in promoting microglia-mediated inflammation after TBI and suggests CCL7 could serve as a potential therapeutic strategy for attenuating TBI by inhibiting microglial activation.
Collapse
Affiliation(s)
- Jianqin Xue
- Department of Rehabilitation Medicine, Jintan Hospital affiliated to Jiangsu University, Changzhou 213200, China
| | - Yu Zhang
- Department of Rehabilitation Medicine, Jintan Hospital affiliated to Jiangsu University, Changzhou 213200, China
| | - Junhua Zhang
- Neurology Department, Jintan Hospital affiliated to Jiangsu University, Changzhou 213200, China
| | - Zhujun Zhu
- Department of Rehabilitation Medicine, Jintan Hospital affiliated to Jiangsu University, Changzhou 213200, China
| | - Qi Lv
- Department of Rehabilitation Medicine, Jintan Hospital affiliated to Jiangsu University, Changzhou 213200, China
| | - Jianhua Su
- Neurology Department, Jintan Hospital affiliated to Jiangsu University, Changzhou 213200, China.
| |
Collapse
|
33
|
McKelvey MC, Brown R, Ryan S, Mall MA, Weldon S, Taggart CC. Proteases, Mucus, and Mucosal Immunity in Chronic Lung Disease. Int J Mol Sci 2021; 22:5018. [PMID: 34065111 PMCID: PMC8125985 DOI: 10.3390/ijms22095018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Dysregulated protease activity has long been implicated in the pathogenesis of chronic lung diseases and especially in conditions that display mucus obstruction, such as chronic obstructive pulmonary disease, cystic fibrosis, and non-cystic fibrosis bronchiectasis. However, our appreciation of the roles of proteases in various aspects of such diseases continues to grow. Patients with muco-obstructive lung disease experience progressive spirals of inflammation, mucostasis, airway infection and lung function decline. Some therapies exist for the treatment of these symptoms, but they are unable to halt disease progression and patients may benefit from novel adjunct therapies. In this review, we highlight how proteases act as multifunctional enzymes that are vital for normal airway homeostasis but, when their activity becomes immoderate, also directly contribute to airway dysfunction, and impair the processes that could resolve disease. We focus on how proteases regulate the state of mucus at the airway surface, impair mucociliary clearance and ultimately, promote mucostasis. We discuss how, in parallel, proteases are able to promote an inflammatory environment in the airways by mediating proinflammatory signalling, compromising host defence mechanisms and perpetuating their own proteolytic activity causing structural lung damage. Finally, we discuss some possible reasons for the clinical inefficacy of protease inhibitors to date and propose that, especially in a combination therapy approach, proteases represent attractive therapeutic targets for muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Ryan Brown
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Sinéad Ryan
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| |
Collapse
|
34
|
Savla SR, Prabhavalkar KS, Bhatt LK. Cytokine storm associated coagulation complications in COVID-19 patients: Pathogenesis and Management. Expert Rev Anti Infect Ther 2021; 19:1397-1413. [PMID: 33832398 PMCID: PMC8074652 DOI: 10.1080/14787210.2021.1915129] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction SARS-CoV-2, the causative agent of COVID-19, attacks the immune system causing an exaggerated and uncontrolled release of pro-inflammatory mediators (cytokine storm). Recent studies propose an active role of coagulation disorders in disease progression. This hypercoagulability has been displayed by marked increase in D-dimer in hospitalized patients. Areas Covered This review summarizes the pathogenesis of SARS-CoV-2 infection, generation of cytokine storm, the interdependence between inflammation and coagulation, its consequences and the possible management options for coagulation complications like venous thromboembolism (VTE), microthrombosis, disseminated intravascular coagulation (DIC), and systemic and local coagulopathy. We searched PubMed, Scopus, and Google Scholar for relevant reports using COVID-19, cytokine storm, and coagulation as keywords. Expert Opinion A prophylactic dose of 5000–7500 units of low molecular weight heparin (LMWH) has been recommended for hospitalized COVID-19 patients in order to prevent VTE. Treatment dose of LMWH, based on disease severity, is being contemplated for patients showing a marked rise in levels of D-dimer due to possible pulmonary thrombi. Additionally, targeting PAR-1, thrombin, coagulation factor Xa and the complement system may be potentially useful in reducing SARS-CoV-2 infection induced lung injury, microvascular thrombosis, VTE and related outcomes like DIC and multi-organ failure.
Collapse
Affiliation(s)
- Shreya R Savla
- Department of Pharmacology, Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Lokesh K Bhatt
- Department of Pharmacology, Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| |
Collapse
|
35
|
Chiang CC, Korinek M, Cheng WJ, Hwang TL. Targeting Neutrophils to Treat Acute Respiratory Distress Syndrome in Coronavirus Disease. Front Pharmacol 2020; 11:572009. [PMID: 33162887 PMCID: PMC7583590 DOI: 10.3389/fphar.2020.572009] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/11/2020] [Indexed: 01/08/2023] Open
Abstract
This review describes targeting neutrophils as a potential therapeutic strategy for acute respiratory distress syndrome (ARDS) associated with coronavirus disease 2019 (COVID-19), a pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Neutrophil counts are significantly elevated in patients with COVID-19 and significantly correlated with disease severity. The neutrophil-to-lymphocyte ratio can serve as a clinical marker for predicting fatal complications related to ARDS in patients with COVID-19. Neutrophil-associated inflammation plays a critical pathogenic role in ARDS. The effector functions of neutrophils, acting as respiratory burst oxidants, granule proteases, and neutrophil extracellular traps, are linked to the pathogenesis of ARDS. Hence, neutrophils can not only be used as pathogenic markers but also as candidate drug targets for COVID-19 associated ARDS.
Collapse
Affiliation(s)
- Chih-Chao Chiang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Puxin Fengze Chinese Medicine Clinic, Taoyuan, Taiwan
| | - Michal Korinek
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Jen Cheng
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
36
|
Cui TX, Brady AE, Fulton CT, Zhang YJ, Rosenbloom LM, Goldsmith AM, Moore BB, Popova AP. CCR2 Mediates Chronic LPS-Induced Pulmonary Inflammation and Hypoalveolarization in a Murine Model of Bronchopulmonary Dysplasia. Front Immunol 2020; 11:579628. [PMID: 33117383 PMCID: PMC7573800 DOI: 10.3389/fimmu.2020.579628] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 11/28/2022] Open
Abstract
The histopathology of bronchopulmonary dysplasia (BPD) includes hypoalveolarization and interstitial thickening due to abnormal myofibroblast accumulation. Chorioamnionitis and sepsis are major risk factors for BPD development. The cellular mechanisms leading to these lung structural abnormalities are poorly understood. We used an animal model with repeated lipopolysaccharide (LPS) administration into the airways of immature mice to simulate prolonged airway exposure to gram-negative bacteria, focusing on the role of C-C chemokine receptor type 2-positive (CCR2+) exudative macrophages (ExMf). Repetitive LPS exposure of immature mice induced persistent hypoalveolarization observed at 4 and 18 days after the last LPS administration. LPS upregulated the expression of lung pro-inflammatory cytokines (TNF-α, IL-17a, IL-6, IL-1β) and chemokines (CCL2, CCL7, CXCL1, and CXCL2), while the expression of genes involved in lung alveolar and mesenchymal cell development (PDGFR-α, FGF7, FGF10, and SPRY1) was decreased. LPS induced recruitment of ExMf, including CCR2+ ExMf, as well as other myeloid cells like DCs and neutrophils. Lungs of LPS-exposed CCR2−/− mice showed preserved alveolar structure and normal patterns of α-actin and PDGFRα expression at the tips of the secondary alveolar crests. Compared to wild type mice, a significantly lower number of ExMf, including TNF-α+ ExMf were recruited to the lungs of CCR2−/− mice following repetitive LPS exposure. Further, pharmacological inhibition of TLR4 with TAK-242 also blocked the effect of LPS on alveolarization, α-SMA and PDGFRα expression. TNF-α and IL-17a induced α-smooth muscle actin expression in the distal airspaces of E16 fetal mouse lung explants. In human preterm lung mesenchymal stromal cells, TNF-α reduced mRNA and protein expression of PDGFR-α and decreased mRNA expression of WNT2, FOXF2, and SPRY1. Collectively, our findings demonstrate that in immature mice repetitive LPS exposure, through TLR4 signaling increases lung inflammation and impairs lung alveolar growth in a CCR2-dependent manner.
Collapse
Affiliation(s)
- Tracy X Cui
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Alexander E Brady
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Christina T Fulton
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ying-Jian Zhang
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Liza M Rosenbloom
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Adam M Goldsmith
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States
| | - Antonia P Popova
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
37
|
José RJ, Williams A, Manuel A, Brown JS, Chambers RC. Targeting coagulation activation in severe COVID-19 pneumonia: lessons from bacterial pneumonia and sepsis. Eur Respir Rev 2020; 29:29/157/200240. [PMID: 33004529 PMCID: PMC7537941 DOI: 10.1183/16000617.0240-2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 08/20/2020] [Indexed: 12/15/2022] Open
Abstract
Novel coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), has rapidly spread throughout the world, resulting in a pandemic with high mortality. There are no effective treatments for the management of severe COVID-19 and current therapeutic trials are focused on antiviral therapy and attenuation of hyper-inflammation with anti-cytokine therapy. Severe COVID-19 pneumonia shares some pathological similarities with severe bacterial pneumonia and sepsis. In particular, it disrupts the haemostatic balance, which results in a procoagulant state locally in the lungs and systemically. This culminates in the formation of microthrombi, disseminated intravascular coagulation and multi-organ failure. The deleterious effects of exaggerated inflammatory responses and activation of coagulation have been investigated in bacterial pneumonia and sepsis and there is recognition that although these pathways are important for the host immune response to pathogens, they can lead to bystander tissue injury and are negatively associated with survival. In the past two decades, evidence from preclinical studies has led to the emergence of potential anticoagulant therapeutic strategies for the treatment of patients with pneumonia, sepsis and acute respiratory distress syndrome, and some of these anticoagulant approaches have been trialled in humans. Here, we review the evidence from preclinical studies and clinical trials of anticoagulant treatment strategies in bacterial pneumonia and sepsis, and discuss the importance of these findings in the context of COVID-19.
Collapse
Affiliation(s)
- Ricardo J José
- Centre for Inflammation and Tissue Repair, University College London, London, UK .,Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Andrew Williams
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Ari Manuel
- University Hospital Aintree, Liverpool, UK
| | - Jeremy S Brown
- Centre for Inflammation and Tissue Repair, University College London, London, UK.,Dept of Thoracic Medicine, University College London Hospital, London, UK
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| |
Collapse
|
38
|
Mao K, Geng W, Liao Y, Luo P, Zhong H, Ma P, Xu J, Zhang S, Tan Q, Jin Y. Identification of robust genetic signatures associated with lipopolysaccharide-induced acute lung injury onset and astaxanthin therapeutic effects by integrative analysis of RNA sequencing data and GEO datasets. Aging (Albany NY) 2020; 12:18716-18740. [PMID: 32969837 PMCID: PMC7585091 DOI: 10.18632/aging.104042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/19/2020] [Indexed: 01/24/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening clinical conditions predominantly arising from uncontrolled inflammatory reactions. It has been found that the administration of astaxanthin (AST) can exert protective effects against lipopolysaccharide (LPS)-induced ALI; however, the robust genetic signatures underlying LPS induction and AST treatment remain obscure. Here we performed a statistical meta-analysis of five publicly available gene expression datasets from LPS-induced ALI mouse models, conducted RNA-sequencing (RNA-seq) to screen differentially expressed genes (DEGs) in response to LPS administration and AST treatment, and integrative analysis to determine robust genetic signatures associated with LPS-induced ALI onset and AST administration. Both the meta-analyses and our experimental data identified a total of 198 DEGs in response to LPS administration, and 11 core DEGs (Timp1, Ly6i, Cxcl13, Irf7, Cxcl5, Ccl7, Isg15, Saa3, Saa1, Tgtp1, and Gbp11) were identified to be associated with AST therapeutic effects. Further, the 11 core DEGs were verified by quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC), and functional enrichment analysis revealed that these genes are primarily associated with neutrophils and chemokines. Collectively, these findings unearthed the robust genetic signatures underlying LPS administration and the molecular targets of AST for ameliorating ALI/ARDS which provide directions for further research.
Collapse
Affiliation(s)
- Kaimin Mao
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Wei Geng
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yuhan Liao
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Ping Luo
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Hua Zhong
- College of Life Sciences, Wuhan University, Hubei Province, Wuhan, 430072, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Juanjuan Xu
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Shuai Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| |
Collapse
|
39
|
Inaba A, Tuong ZK, Riding AM, Mathews RJ, Martin JL, Saeb-Parsy K, Clatworthy MR. B Lymphocyte-Derived CCL7 Augments Neutrophil and Monocyte Recruitment, Exacerbating Acute Kidney Injury. THE JOURNAL OF IMMUNOLOGY 2020; 205:1376-1384. [PMID: 32737150 PMCID: PMC7444279 DOI: 10.4049/jimmunol.2000454] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/01/2020] [Indexed: 12/21/2022]
Abstract
AKI is a serious condition affecting one fifth of hospital patients. In AKI, B cells produce CCL7 and facilitate neutrophil and monocyte recruitment. CCL7 blockade in mice reduces myeloid cell infiltration and ameliorates AKI.
Acute kidney injury (AKI) is a serious condition affecting one fifth of hospital inpatients. B lymphocytes have immunological functions beyond Ab production and may produce cytokines and chemokines that modulate inflammation. In this study, we investigated leukocyte responses in a mouse model of AKI and observed an increase in circulating and kidney B cells, particularly a B220low subset, following AKI. We found that B cells produce the chemokine CCL7, with the potential to facilitate neutrophil and monocyte recruitment to the injured kidney. Siglec-G–deficient mice, which have increased numbers of B220low innate B cells and a lower B cell activation threshold, had increased Ccl7 transcripts, increased neutrophil and monocyte numbers in the kidney, and more severe AKI. CCL7 blockade in mice reduced myeloid cell infiltration into the kidney and ameliorated AKI. In two independent cohorts of human patients with AKI, we observed significantly higher CCL7 transcripts compared with controls, and in a third cohort, we observed an increase in urinary CCL7 levels in AKI, supporting the clinical importance of this pathway. Together, our data suggest that B cells contribute to early sterile inflammation in AKI via the production of leukocyte-recruiting chemokines.
Collapse
Affiliation(s)
- Akimichi Inaba
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom; .,Cambridge University Hospitals National Health Service Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Zewen K Tuong
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom.,Cellular Genetics, Wellcome Trust Sanger Institute, Hinxton CB10 1RQ, United Kingdom
| | - Alexandra M Riding
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom
| | - Rebeccah J Mathews
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom
| | - Jack L Martin
- Cambridge University Hospitals National Health Service Foundation Trust, Cambridge CB2 0QQ, United Kingdom.,Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; and
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; and.,National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, United Kingdom
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom.,Cambridge University Hospitals National Health Service Foundation Trust, Cambridge CB2 0QQ, United Kingdom.,Cellular Genetics, Wellcome Trust Sanger Institute, Hinxton CB10 1RQ, United Kingdom.,National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
40
|
Rayees S, Joshi JC, Tauseef M, Anwar M, Baweja S, Rochford I, Joshi B, Hollenberg MD, Reddy SP, Mehta D. PAR2-Mediated cAMP Generation Suppresses TRPV4-Dependent Ca 2+ Signaling in Alveolar Macrophages to Resolve TLR4-Induced Inflammation. Cell Rep 2020; 27:793-805.e4. [PMID: 30995477 DOI: 10.1016/j.celrep.2019.03.053] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/18/2018] [Accepted: 03/13/2019] [Indexed: 12/19/2022] Open
Abstract
Alveolar macrophages (AMs), upon sensing pathogens, trigger host defense by activating toll-like receptor 4 (TLR4), but the counterbalancing mechanisms that deactivate AM inflammatory signaling and prevent lethal edema, the hallmark of acute lung injury (ALI), remain unknown. Here, we demonstrate the essential role of AM protease-activating receptor 2 (PAR2) in rapidly suppressing inflammation to prevent long-lasting injury. We show that thrombin, released during TLR4-induced lung injury, directly activates PAR2 to generate cAMP, which abolishes Ca2+ entry through the TRPV4 channel. Deletion of PAR2 and thus the accompanying cAMP generation augments Ca2+ entry via TRPV4, causing sustained activation of the transcription factor NFAT to produce long-lasting TLR4-mediated inflammatory lung injury. Rescuing thrombin-sensitive PAR2 expression or blocking TRPV4 activity in PAR2-null AMs restores their capacity to resolve inflammation and reverse lung injury. Thus, activation of the thrombin-induced PAR2-cAMP cascade in AMs suppresses TLR4 inflammatory signaling to reinstate tissue integrity.
Collapse
Affiliation(s)
- Sheikh Rayees
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Jagdish Chandra Joshi
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Mohammad Tauseef
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA; Department of Pharmaceutical Sciences, College of Pharmacy, Chicago State University, Chicago, IL 60628, USA
| | - Mumtaz Anwar
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Sukriti Baweja
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Ian Rochford
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Bhagwati Joshi
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Morley D Hollenberg
- Department of Physiology and Pharmacology and Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Sekhar P Reddy
- Department of Pediatrics, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Dolly Mehta
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA.
| |
Collapse
|
41
|
Gupta N, Liu R, Shin S, Sinha R, Pogliano J, Pogliano K, Griffin JH, Nizet V, Corriden R. SCH79797 improves outcomes in experimental bacterial pneumonia by boosting neutrophil killing and direct antibiotic activity. J Antimicrob Chemother 2019. [PMID: 29514266 DOI: 10.1093/jac/dky033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objectives The role of protease-activated receptor 1 (PAR1) in the pathogenesis of pneumonia and sepsis is ambiguous given the existing literature. As PAR1 is classically activated by the coagulation-based protease thrombin and leads to vascular leakage, our hypothesis was that PAR1 blockade with SCH79797 would be therapeutically beneficial in an experimental model of murine Gram-negative pneumonia. Methods In this study, we administered SCH79797 via the intrapulmonary route 6 h after the establishment of Escherichia coli pneumonia and observed a significant improvement in survival, lung injury, bacterial clearance and inflammation. We focused on neutrophils as a potential target of the PAR1 antagonist, since they are the predominant inflammatory cell type in the infected lung. Results Neutrophils appear to express PAR1 at low levels and the PAR1 antagonist SCH79797 enhanced neutrophil killing. Part of this effect may be explained by alterations in the generation of reactive oxygen species (ROS). SCH79797 also led to robust neutrophil extracellular trap (NET) generation and cathelicidin-related antimicrobial peptide (CRAMP) release by neutrophils. Surprisingly, SCH79797 also had a potent, direct antibiotic effect with disruption of the E. coli cell membrane. However, the newer-generation PAR1 antagonist, vorapaxar (SCH530348), had no appreciable effect on neutrophil activity or direct bacterial killing, which suggests the effects seen with SCH79797 may be PAR1 independent. Conclusions In summary, we observed that intrapulmonary treatment with SCH79797 has significant therapeutic effects in a model of E. coli pneumonia that appear to be due, in part, to both neutrophil-stimulating and direct antibacterial effects of SCH79797.
Collapse
Affiliation(s)
- Naveen Gupta
- University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA.,Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Roland Liu
- University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Stephanie Shin
- University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Ranjeet Sinha
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Joseph Pogliano
- University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kit Pogliano
- University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - John H Griffin
- University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA.,Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Victor Nizet
- University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Ross Corriden
- University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
42
|
Xing D, Wells JM, Giordano SS, Feng W, Gaggar A, Yan J, Hage FG, Li L, Chen YF, Oparil S. Induced pluripotent stem cell-derived endothelial cells attenuate lipopolysaccharide-induced acute lung injury. J Appl Physiol (1985) 2019; 127:444-456. [PMID: 31295064 PMCID: PMC6732441 DOI: 10.1152/japplphysiol.00587.2018] [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: 07/02/2018] [Revised: 06/11/2019] [Accepted: 06/27/2019] [Indexed: 02/08/2023] Open
Abstract
The chemokine receptors CXCR1/2 and CCR2/5 play a critical role in neutrophil and monocyte recruitment to sites of injury and/or inflammation. Neutrophil-mediated inflammation and endothelial cell (EC) injury are unifying factors in the pathogenesis of the acute respiratory distress syndrome. This study tested the hypothesis that systemic administration of rat-induced pluripotent stem cell (iPS)-derived ECs (iPS-ECs) overexpressing CXCR1/2 or CCR2/5 attenuates lipopolysaccharide (LPS)-induced acute lung injury. Rat iPS-ECs were transduced with adenovirus containing cDNA of CXCR1/2 or CCR2/5. Ovariectomized Sprague-Dawley rats (10 wk old) received intraperitoneal injection of LPS and intravenous infusion of 1) saline vehicle, 2) AdNull-iPS-ECs (iPS-ECs transduced with empty adenoviral vector), 3) CXCR1/2-iPS-ECs (iPS-ECs overexpressing CXCR1/2), or 4) CCR2/5-iPS-ECs (iPS-ECs overexpressing CCR2/5) at 2 h post-LPS. Rats receiving intraperitoneal injection of saline served as sham controls. Later (4 h), proinflammatory cytokine/chemokine mRNA and protein levels were measured in total lung homogenates by real-time RT-PCR and Luminex multiplex assays, and neutrophil and macrophage infiltration in alveoli was measured by immunohistochemical staining. Pulmonary microvascular permeability was assessed by the Evans blue technique, and pulmonary edema was estimated by wet-to-dry lung weight ratios. Albumin levels and neutrophil counts were assessed in bronchoalveolar lavage fluid at 24 h post-LPS. Both CXCR1/2-iPS-ECs and CCR2/5-iPS-ECs significantly reduced LPS-induced proinflammatory mediator expression, neutrophil and macrophage infiltration, pulmonary edema, and vascular permeability compared with controls. These provocative findings provide strong evidence that targeted delivery of iPS-ECs overexpressing CXCR1/2 or CCR2/5 prevents LPS-induced acute lung injury.NEW & NOTEWORTHY We have developed a novel approach to address neutrophil-mediated inflammation and endothelial damage by targeted delivery of rat-induced pluripotent stem cell (iPS)-derived endothelial cell (ECs)overexpressing chemokine receptors CXCR1/2 and CCR2/5 in injured lung tissue in a model of acute lung injury. We have demonstrated that intravenously transfused CXCR1/2-iPS-ECs and CCR2/5-iPS-ECs are recruited to lipopolysaccharide-injured lungs and attenuate lipopolysaccharide-induced parenchymal lung injury responses, including inflammatory mediator expression, inflammatory cell infiltration, and vascular leakage compared with controls.
Collapse
Affiliation(s)
- Dongqi Xing
- Division of Pulmonary, Allergy & Critical Care Medicine, Lung Health Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - J Michael Wells
- Division of Pulmonary, Allergy & Critical Care Medicine, Lung Health Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Samantha S Giordano
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wenguang Feng
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amit Gaggar
- Division of Pulmonary, Allergy & Critical Care Medicine, Lung Health Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Jie Yan
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico
| | - Fadi G Hage
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Li Li
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Physiology, School of Medicine, Shihezi University, Xinjiang, China
| | - Yiu-Fai Chen
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
43
|
Lou J, Hu Y, Wu MD, Che LQ, Wu YF, Zhao Y, Tian BP, Bao ZQ, Zhu C, Wu YP, He LL, Bai CX, Zhou J, Ying SM, Li W, Chen ZH, Chen DX, Dorling A, Shen HH. Endothelial cell-specific anticoagulation reduces inflammation in a mouse model of acute lung injury. Acta Pharmacol Sin 2019; 40:769-780. [PMID: 30446733 DOI: 10.1038/s41401-018-0175-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 09/24/2018] [Indexed: 01/11/2023]
Abstract
Tissue factor (TF)-dependent coagulation contributes to lung inflammation and the pathogenesis of acute lung injury (ALI). In this study, we explored the roles of targeted endothelial anticoagulation in ALI using two strains of transgenic mice expressing either a membrane-tethered human tissue factor pathway inhibitor (hTFPI) or hirudin fusion protein on CD31+ cells, including vascular endothelial cells (ECs). ALI was induced by intratracheal injection of LPS, and after 24 h the expression of TF and protease-activated receptors (PARs) on EC in lungs were assessed, alongside the extent of inflammation and injury. The expression of TF and PARs on the EC in lungs was upregulated after ALI. In the two strains of transgenic mice, expression of either of hTFPI or hirudin by EC was associated with significant reduction of inflammation, as assessed by the extent of leukocyte infiltration or the levels of proinflammatory cytokines, and promoted survival after LPS-induced ALI. The beneficial outcomes were associated with inhibition of the expression of chemokine CCL2 in lung tissues. The protection observed in the CD31-TFPI-transgenic strain was abolished by injection of an anti-hTFPI antibody, but not by prior engraftment of the transgenic strains with WT bone marrow, confirming that the changes observed were a specific transgenic expression of anticoagulants by EC. These results demonstrate that the inflammation in ALI is TF and thrombin dependent, and that expression of anticoagulants by EC significantly inhibits the development of ALI via repression of leukocyte infiltration, most likely via inhibition of chemokine gradients. These data enhance our understanding of the pathology of ALI and suggest a novel therapeutic strategy for treatment.
Collapse
|
44
|
Wiesemann A, Ketteler J, Slama A, Wirsdörfer F, Hager T, Röck K, Engel DR, Fischer JW, Aigner C, Jendrossek V, Klein D. Inhibition of Radiation-Induced Ccl2 Signaling Protects Lungs from Vascular Dysfunction and Endothelial Cell Loss. Antioxid Redox Signal 2019; 30:213-231. [PMID: 29463096 DOI: 10.1089/ars.2017.7458] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aims: Radiation-induced normal tissue toxicity often precludes the application of curative radiation doses. Here we investigated the therapeutic potential of chemokine C-C motif ligand 2 (Ccl2) signaling inhibition to protect normal lung tissue from radiotherapy (RT)-induced injury. Results: RT-induced vascular dysfunction and associated adverse effects can be efficiently antagonized by inhibition of Ccl2 signaling using either the selective Ccl2 inhibitor bindarit (BIN) or mice deficient for the main Ccl2 receptor CCR2 (KO). BIN-treatment efficiently counteracted the RT-induced expression of Ccl2, normalized endothelial cell (EC) morphology and vascular function, and limited lung inflammation and metastasis early after irradiation (acute effects). A similar protection of the vascular compartment was detected by loss of Ccl2 signaling in lungs of CCR2-KO mice. Long-term Ccl2 signaling inhibition also significantly limited EC loss and accompanied fibrosis progression as adverse late effect. With respect to the human situation, we further confirmed that Ccl2 secreted by RT-induced senescent epithelial cells resulted in the activation of normally quiescent but DNA-damaged EC finally leading to EC loss in ex vivo cultured human normal lung tissue. Innovation: Abrogation of certain aspects of the secretome of irradiated resident lung cells, in particular signaling inhibition of the senescence-associated secretory phenotype-factor Ccl2 secreted predominantly by RT-induced senescent epithelial cells, resulted in protection of the endothelial compartment. Conclusions: Radioprotection of the normal tissue via Ccl2 signaling inhibition without simultaneous protection or preferable radiosensitization of tumor tissue might improve local tumor control and survival, because higher doses of radiation could be used.
Collapse
Affiliation(s)
- Alina Wiesemann
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Julia Ketteler
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Alexis Slama
- 2 Department of Thoracic Surgery and Surgical Endoscopy, Ruhrlandklinik-University Clinic Essen , Essen, Germany
| | - Florian Wirsdörfer
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Thomas Hager
- 3 Institute of Pathology, University Clinic Essen, University of Duisburg-Essen , Essen, Germany
| | - Katharina Röck
- 4 Institute for Pharmacology, University Hospital, Heinrich-Heine-University , Düsseldorf, Germany
| | - Daniel R Engel
- 5 Department Immunodynamics, Institute of Experimental Immunology and Imaging, University Duisburg-Essen, University Hospital Essen , Essen, Germany
| | - Jens W Fischer
- 4 Institute for Pharmacology, University Hospital, Heinrich-Heine-University , Düsseldorf, Germany
| | - Clemens Aigner
- 2 Department of Thoracic Surgery and Surgical Endoscopy, Ruhrlandklinik-University Clinic Essen , Essen, Germany
| | - Verena Jendrossek
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Diana Klein
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| |
Collapse
|
45
|
Ford J, Hughson A, Lim K, Bardina SV, Lu W, Charo IF, Lim JK, Fowell DJ. CCL7 Is a Negative Regulator of Cutaneous Inflammation Following Leishmania major Infection. Front Immunol 2019; 9:3063. [PMID: 30671055 PMCID: PMC6331479 DOI: 10.3389/fimmu.2018.03063] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022] Open
Abstract
The chemokine CCL7 (MCP3) is known to promote the recruitment of many innate immune cell types including monocytes and neutrophils to sites of bacterial and viral infection and eosinophils and basophils to sites of allergic inflammation. CCL7 upregulation has been associated with many inflammatory settings including infection, cardiovascular disease, and the tumor microenvironment. CCL7's pleotropic effects are due in part to its ability to bind numerous chemokine receptors, namely CCR1, CCR2, CCR3, CCR5, and CCR10. CCL7-blockade or CCL7-deficiency is often marked by decreased inflammation and poor pathogen control. In the context of Leishmania major infection, CCL7 is specifically upregulated in the skin one-2 weeks after infection but its role in L. major control is unclear. To determine CCL7's impact on the response to L. major we infected WT and CCL7-/- C57BL/6 mice. L. major infection of CCL7-deficient mice led to an unexpected increase in inflammation in the infected skin 2 weeks post-infection. A broad increase in immune cell subsets was observed but was dominated by enhanced neutrophilic infiltration. Increased neutrophil recruitment was associated with an enhanced IL-17 gene profile in the infected skin. CCL7 was shown to directly antagonize neutrophil migration in vitro and CCL7 add-back in vivo specifically reduced neutrophil influx into the infected skin revealing an unexpected role for CCL7 in limiting neutrophil recruitment during L. major infection. Enhanced neutrophilic infiltration in CCL7-deficient mice changed the balance of L. major infected host cells with an increase in the ratio of infected neutrophils over monocytes/macrophages. To determine the consequence of CCL7 deficiency on L. major control we analyzed parasite load cutaneously at the site of infection and viscerally in the draining LN and spleen. The CCL7-/- mice supported robust cutaneous parasite control similar to their WT C57BL/6 counterparts. In contrast, CCL7-deficiency led to greater parasite dissemination and poor parasite control in the spleen. Our studies reveal a novel role for CCL7 in negatively regulating cutaneous inflammation, specifically neutrophils, early during L. major infection. We propose that CCL7-mediated dampening of the early immune response in the skin may limit the ability of the parasite to disseminate without compromising cutaneous control.
Collapse
Affiliation(s)
- Jill Ford
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, United States
| | - Angela Hughson
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, United States
| | - Kihong Lim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, United States
| | - Susana V Bardina
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Wuyuan Lu
- Department of Biochemistry and Molecular Biology, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Israel F Charo
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Deborah J Fowell
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, United States
| |
Collapse
|
46
|
Liu Y, Cai Y, Liu L, Wu Y, Xiong X. Crucial biological functions of CCL7 in cancer. PeerJ 2018; 6:e4928. [PMID: 29915688 PMCID: PMC6004300 DOI: 10.7717/peerj.4928] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
Chemokine (C-C motif) ligand 7 (CCL7), a CC chemokine, is a chemotactic factor and attractant for various kinds of leukocytes, including monocytes and neutrophils. CCL7 is widely expressed in multiple cell types and can participate in anti-inflammatory responses through binding to its receptors to mediate the recruitment of immune cells. Abnormal CCL7 expression is associated with certain immune diseases. Furthermore, CCL7 plays a pivotal role in tumorigenesis. CCL7 promotes tumor progression by supporting the formation of the tumor microenvironment and facilitating tumor invasion and metastasis, although some studies have suggested that CCL7 has tumor suppressor effects. In this review, we summarize the currently available information regarding the influence of CCL7 on tumors.
Collapse
Affiliation(s)
- Yangyang Liu
- First Clinical Medical College, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Yadi Cai
- First Clinical Medical College, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Li Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, People's Republic of China
| | - Yudong Wu
- Department of Breast Surgery, Jiangxi Provincial Cancer Hospital, Nanchang, People's Republic of China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
47
|
Signaling Crosstalk of TGF-β/ALK5 and PAR2/PAR1: A Complex Regulatory Network Controlling Fibrosis and Cancer. Int J Mol Sci 2018; 19:ijms19061568. [PMID: 29795022 PMCID: PMC6032192 DOI: 10.3390/ijms19061568] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/09/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Both signaling by transforming growth factor-β (TGF-β) and agonists of the G Protein-coupled receptors proteinase-activated receptor-1 (PAR1) and -2 (PAR2) have been linked to tissue fibrosis and cancer. Intriguingly, TGF-β and PAR signaling either converge on the regulation of certain matrix genes overexpressed in these pathologies or display mutual regulation of their signaling components, which is mediated in part through sphingosine kinases and sphingosine-1-phosphate and indicative of an intimate signaling crosstalk between the two pathways. In the first part of this review, we summarize the various regulatory interactions that have been discovered so far according to the organ/tissue in which they were described. In the second part, we highlight the types of signaling crosstalk between TGF-β on the one hand and PAR2/PAR1 on the other hand. Both ligand–receptor systems interact at various levels and by several mechanisms including mutual regulation of ligand–ligand, ligand–receptor, and receptor–receptor at the transcriptional, post-transcriptional, and receptor transactivation levels. These mutual interactions between PAR2/PAR1 and TGF-β signaling components eventually result in feed-forward loops/vicious cycles of matrix deposition and malignant traits that exacerbate fibrosis and oncogenesis, respectively. Given the crucial role of PAR2 and PAR1 in controlling TGF-β receptor activation, signaling, TGF-β synthesis and bioactivation, combining PAR inhibitors with TGF-β blocking agents may turn out to be more efficient than targeting TGF-β alone in alleviating unwanted TGF-β-dependent responses but retaining the beneficial ones.
Collapse
|
48
|
Salisbury ML, Myers JL, Belloli EA, Kazerooni EA, Martinez FJ, Flaherty KR. Diagnosis and Treatment of Fibrotic Hypersensitivity Pneumonia. Where We Stand and Where We Need to Go. Am J Respir Crit Care Med 2017; 196:690-699. [PMID: 28002680 DOI: 10.1164/rccm.201608-1675pp] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
| | | | | | - Ella A Kazerooni
- 3 Department of Radiology, University of Michigan, Ann Arbor, Michigan; and
| | - Fernando J Martinez
- 4 Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cornell Medical College, New York, New York
| | - Kevin R Flaherty
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine
| |
Collapse
|
49
|
Costa PM, Gosens I, Williams A, Farcal L, Pantano D, Brown DM, Stone V, Cassee FR, Halappanavar S, Fadeel B. Transcriptional profiling reveals gene expression changes associated with inflammation and cell proliferation following short-term inhalation exposure to copper oxide nanoparticles. J Appl Toxicol 2017; 38:385-397. [PMID: 29094763 DOI: 10.1002/jat.3548] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/20/2017] [Accepted: 09/05/2017] [Indexed: 01/09/2023]
Abstract
Our recent studies revealed a dose-dependent proinflammatory response to copper oxide nanoparticles (CuO NPs) in rats following short-term inhalation exposure for five consecutive days. Here transcriptomics approaches were applied using the same model to assess global gene expression in lung tissues obtained 1 day post-exposure and after a recovery period of 22 days from rats exposed to clean air or 6 hour equivalent doses of 3.3 mg m-3 (low dose) and 13.2 mg m-3 (high dose). Microarray analyses yielded about 1000 differentially expressed genes in the high-dose group and 200 in low-dose compared to the clean air control group, and less than 20 after the recovery period. Pathway analysis indicated cell proliferation/survival and inflammation as the main processes triggered by exposure to CuO NPs. We did not find significant perturbations of pathways related to oxidative stress. Upregulation of epithelial cell transforming protein 2 (Ect2), a known oncogene, was noted and ECT2 protein was upregulated in the lungs of exposed animals. Proliferation of alveolar epithelial cells was demonstrated based on Ki67 expression. The gene encoding monocyte chemoattractant protein 1 (or CCL2) was also upregulated and this was confirmed by immunohistochemistry. However, no aberrant DNA methylation of inflammation-associated genes was observed. In conclusion, we have found that inhalation of CuO NPs in rats causes upregulation of the oncoprotein ECT2 and the chemokine CCL2 and other proinflammatory markers as well as proliferation in bronchoalveolar epithelium after a short-term inhalation exposure. Thus, pathways known to be associated with neoplastic processes and inflammation were affected in this model.
Collapse
Affiliation(s)
- Pedro M Costa
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ilse Gosens
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Lucian Farcal
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Daniele Pantano
- School of Life Sciences, Heriot-Watt University, Edinburgh, UK
| | - David M Brown
- School of Life Sciences, Heriot-Watt University, Edinburgh, UK
| | - Vicki Stone
- School of Life Sciences, Heriot-Watt University, Edinburgh, UK
| | - Flemming R Cassee
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands.,Institute for Risk Assessment Studies, Utrecht University, Utrecht, The Netherlands
| | - Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Bengt Fadeel
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Schauer AE, Klassert TE, von Lachner C, Riebold D, Schneeweiß A, Stock M, Müller MM, Hammerschmidt S, Bufler P, Seifert U, Dietert K, Dinarello CA, Nold MF, Gruber AD, Nold-Petry CA, Slevogt H. IL-37 Causes Excessive Inflammation and Tissue Damage in Murine Pneumococcal Pneumonia. J Innate Immun 2017; 9:403-418. [PMID: 28601872 PMCID: PMC6738772 DOI: 10.1159/000469661] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pneumoniae infections can lead to severe complications with excessive immune activation and tissue damage. Interleukin-37 (IL-37) has gained importance as a suppressor of innate and acquired immunity, and its effects have been therapeutic as they prevent tissue damage in autoimmune and inflammatory diseases. By using RAW macrophages, stably transfected with human IL-37, we showed a 70% decrease in the cytokine levels of IL-6, TNF-α, and IL-1β, and a 2.2-fold reduction of the intracellular killing capacity of internalized pneumococci in response to pneumococcal infection. In a murine model of infection with S. pneumoniae, using mice transgenic for human IL-37b (IL-37tg), we observed an initial decrease in cytokine expression of IL-6, TNF-α, and IL-1β in the lungs, followed by a late-phase enhancement of pneumococcal burden and subsequent increase of proinflammatory cytokine levels. Additionally, a marked increase in recruitment of alveolar macrophages and neutrophils was noted, while TRAIL mRNA was reduced 3-fold in lungs of IL-37tg mice, resulting in necrotizing pneumonia with augmented death of infiltrating neutrophils, enhanced bacteremic spread, and increased mortality. In conclusion, we have identified that IL-37 modulates several core components of a successful inflammatory response to pneumococcal pneumonia, which lead to increased inflammation, tissue damage, and mortality.
Collapse
Affiliation(s)
- Anja E. Schauer
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | | | | | - Diana Riebold
- InfectoGnostics Research Campus Jena, Centre for Applied Research Jena, Jena, Germany
| | - Anne Schneeweiß
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Magdalena Stock
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Mario M. Müller
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Sven Hammerschmidt
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Greifswald, Germany
| | - Philip Bufler
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Ulrike Seifert
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Kristina Dietert
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel F. Nold
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Achim D. Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Claudia A. Nold-Petry
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Hortense Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany
| |
Collapse
|