1
|
Chung KP, Cheng CN, Chen YJ, Hsu CL, Huang YL, Hsieh MS, Kuo HC, Lin YT, Juan YH, Nakahira K, Chen YF, Liu WL, Ruan SY, Chien JY, Plataki M, Cloonan SM, Carmeliet P, Choi AMK, Kuo CH, Yu CJ. Alveolar epithelial cells mitigate neutrophilic inflammation in lung injury through regulating mitochondrial fatty acid oxidation. Nat Commun 2024; 15:7241. [PMID: 39174557 PMCID: PMC11341863 DOI: 10.1038/s41467-024-51683-1] [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: 10/01/2023] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Type 2 alveolar epithelial (AT2) cells of the lung are fundamental in regulating alveolar inflammation in response to injury. Impaired mitochondrial long-chain fatty acid β-oxidation (mtLCFAO) in AT2 cells is assumed to aggravate alveolar inflammation in acute lung injury (ALI), yet the importance of mtLCFAO to AT2 cell function needs to be defined. Here we show that expression of carnitine palmitoyltransferase 1a (CPT1a), a mtLCFAO rate limiting enzyme, in AT2 cells is significantly decreased in acute respiratory distress syndrome (ARDS). In mice, Cpt1a deletion in AT2 cells impairs mtLCFAO without reducing ATP production and alters surfactant phospholipid abundance in the alveoli. Impairing mtLCFAO in AT2 cells via deleting either Cpt1a or Acadl (acyl-CoA dehydrogenase long chain) restricts alveolar inflammation in ALI by hindering the production of the neutrophilic chemokine CXCL2 from AT2 cells. This study thus highlights mtLCFAO as immunometabolism to injury in AT2 cells and suggests impaired mtLCFAO in AT2 cells as an anti-inflammatory response in ARDS.
Collapse
Grants
- K08 HL157728 NHLBI NIH HHS
- 109-O04, 110-O07, 110-S4872, 111-S0075, 113-S0079 National Taiwan University Hospital (NTUH)
- NTUCDP-112L7745, NTUCDP-112L7746, 110T099, NTU-NFG-110L7422 National Taiwan University (NTU)
- National Science and Technology Council (Taiwan) (MOST-108-2628-B-002-017 [K.P.C.], MOST-109-2628-B-002-044 [K.P.C.], MOST-110-2628-B-002-029 [K.P.C.], MOST-110-2628-B-002-045-MY3 [K.P.C.], MOST-111-2628-B-002-030-MY3 [K.P.C.])
- National Science and Technology Council (Taiwan), MOST 107-2314-B-002-235-MY3
- National Science and Technology Council (Taiwan), MOST 110-2314-B-002-262
- National Taiwan University School of Pharmacy Endowment Fund in support of the Platform for Clinical Mass Spectrometry and NMR Structure Elucidation
- Research funding provided by Mr. Barry Lam, the chairman of Quanta Computer Inc
Collapse
Affiliation(s)
- Kuei-Pin Chung
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chih-Ning Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Jung Chen
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Lin Huang
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Chun Kuo
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Ting Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsiu Juan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kiichi Nakahira
- Department of Pharmacology, Nara Medical University, Kashihara, Nara, Japan
| | - Yen-Fu Chen
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
- Department of Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan
| | - Sheng-Yuan Ruan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jung-Yien Chien
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan.
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
2
|
Mao Y, Alarfaj AA, Hussein-Al-Ali SH, Ma H. Diterpene Coronarin Attenuates Lipopolysaccharide-Induced Acute Lung Injury in Both In Vivo and In Vitro Models. Appl Biochem Biotechnol 2024; 196:4140-4155. [PMID: 37906408 DOI: 10.1007/s12010-023-04711-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Acute lung injury (ALI) is a clinical condition occurs due to severe systemic inflammatory response for clinical stimulus like pneumonia, sepsis, trauma, aspiration, inhalation of toxic gases, and pancreatitis. Disruption of alveolar barriers, activation of macrophages, infiltration of neutrophils, and proinflammatory cytokines are the vital events occurs during ALI. The drugs which inhibit these inflammatory response can protect lungs from inflammatory insults. In this study, we examined the potency of phytochemical coronarin, a diterpene which have been proven to possess anti-inflammatory, antioxidant, antiangiogenic, and antitumor activities. Healthy BALB/c mice were induced to acute lung injury with intra-tracheal administration of LPS and then treated with 5 and 10 mg/kg concentration of coronarin. The wet/dry lung weight of mice were estimated to assess the induction of pulmonary edema. BALF fluid was analyzed for protein concentrations and immune cells count. Myeloperoxidase activity and levels of chemokines MCP-2 and MIP-2, iNOS, COX-2, and PGE-2 were quantified to assess the immunomodulatory effect of coronarin against LPS-induced ALI. The levels of proinflammatory cytokines was measured to examine the anti-inflammatory property of coronarin, and it was confirmed with histopathological analysis of the lung tissue. Murine RAW 264.7 cells were utilized for the in vitro analysis. Cell cytoxicity and cytoprotective property of coronarin was assessed with MTT assay in LPS-treated Murine RAW 264.7. The anti-inflammatory property of coronarin was further confirmed in in vitro condition by estimating the levels of pro-inflammatory cytokines in coronarin-treated and untreated LPS-induced cells. Overall, our in vivo and in vitro results confirm coronarin significantly inhibited the infiltration of neutrophils prevented immunodulatory activity and synthesis of proinflammatory cytokines and alleviated the acute lung injury induced by LPS. Coronarin is a potent anti-inflammatory drug which can be subjected to further research to be prescribed as drug for ALI.
Collapse
Affiliation(s)
- Ya Mao
- Department of Cardiothoracic Surgery, Yantai Mountain Hospital, Yantai, 264001, China
| | - Abdullah A Alarfaj
- Department of Respiratory II, Yantai Mountain Hospital, Yantai, 264001, China
| | - Samer Hasan Hussein-Al-Ali
- Faculty of Pharmacy, PO Box 33 and 22 Isra University Office 11622 by Queen Alia International Airport south of the capital, Amman, Jordan
| | - Hongxia Ma
- Department of thoracic surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250000, China.
| |
Collapse
|
3
|
Toya S, Struyf S, Huerta L, Morris P, Gavioli E, Minnella EM, Cesta MC, Allegretti M, Proost P. A narrative review of chemokine receptors CXCR1 and CXCR2 and their role in acute respiratory distress syndrome. Eur Respir Rev 2024; 33:230172. [PMID: 39048127 PMCID: PMC11267298 DOI: 10.1183/16000617.0172-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/15/2024] [Indexed: 07/27/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe form of acute respiratory failure characterised by extensive inflammatory injury to the alveolocapillary barrier leading to alveolar oedema, impaired gas exchange and, ultimately, hypoxaemia necessitating the use of supplemental oxygen combined with some degree of positive airway pressure. Although much heterogeneity exists regarding the aetiology, localisation and endotypic characterisation of ARDS, what remains largely undisputed is the role of the innate immune system, and in particular of neutrophils, in precipitating and propagating lung injury. Activated neutrophils, recruited to the lung through chemokine gradients, promote injury by releasing oxidants, proteases and neutrophil extracellular traps, which ultimately cause platelet aggregation, microvascular thrombosis and cellular death. Among various neutrophilic chemoattractants, interleukin-8/C-X-C motif ligand 8 and related chemokines, collectively called ELR+ chemokines, acting on neutrophils through the G protein-coupled receptors CXCR1 and CXCR2, are pivotal in orchestrating the neutrophil activation status and chemotaxis in the inflamed lung. This allows efficient elimination of infectious agents while at the same time minimising collateral damage to host tissue. Therefore, understanding how CXCR1 and CXCR2 receptors are regulated is important if we hope to effectively target them for therapeutic use in ARDS. In the following narrative review, we provide an overview of the role of ELR+ chemokines in acute lung injury (ALI) and ARDS, we summarise the relevant regulatory pathways of their cognisant receptors CXCR1/2 and highlight current preclinical and clinical evidence on the therapeutic role of CXCR1 and CXCR2 inhibition in animal models of ALI, as well as in ARDS patients.
Collapse
Affiliation(s)
| | - Sofie Struyf
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Luis Huerta
- Keck School of Medicine of USC, Department of Medicine, Pulmonary and Critical Care Medicine, Los Angeles, CA, USA
| | - Peter Morris
- The University of Alabama at Birmingham, Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine, Birmingham, AL, USA
| | | | | | | | | | - Paul Proost
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| |
Collapse
|
4
|
Simmons SR, Herring SE, Tchalla EYI, Lenhard AP, Bhalla M, Bou Ghanem EN. Activating A1 adenosine receptor signaling boosts early pulmonary neutrophil recruitment in aged mice in response to Streptococcus pneumoniae infection. Immun Ageing 2024; 21:34. [PMID: 38840213 PMCID: PMC11151497 DOI: 10.1186/s12979-024-00442-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Streptococcus pneumoniae (pneumococcus) is a leading cause of pneumonia in older adults. Successful control of pneumococci requires robust pulmonary neutrophil influx early in infection. However, aging is associated with aberrant neutrophil recruitment and the mechanisms behind that are not understood. Here we explored how neutrophil recruitment following pneumococcal infection changes with age and the host pathways regulating this. RESULTS Following pneumococcal infection there was a significant delay in early neutrophil recruitment to the lungs of aged mice. Neutrophils from aged mice showed defects in trans-endothelial migration in vitro compared to young controls. To understand the pathways involved, we examined immune modulatory extracellular adenosine (EAD) signaling, that is activated upon cellular damage. Signaling through the lower affinity A2A and A2B adenosine receptors had no effect on neutrophil recruitment to infected lungs. In contrast, inhibition of the high affinity A1 receptor in young mice blunted neutrophil recruitment to the lungs following infection. A1 receptor inhibition decreased expression of CXCR2 on circulating neutrophils, which is required for trans-endothelial migration. Indeed, A1 receptor signaling on neutrophils was required for their ability to migrate across endothelial cells in response to infection. Aging was not associated with defects in EAD production or receptor expression on neutrophils. However, agonism of A1 receptor in aged mice rescued the early defect in neutrophil migration to the lungs and improved control of bacterial burden. CONCLUSIONS This study suggests age-driven defects in EAD damage signaling can be targeted to rescue the delay in pulmonary neutrophil migration in response to bacterial pneumonia.
Collapse
Affiliation(s)
- Shaunna R Simmons
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Sydney E Herring
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Essi Y I Tchalla
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Alexsandra P Lenhard
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Elsa N Bou Ghanem
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
5
|
Simmons SR, Herring SE, Tchalla EYI, Lenhard AP, Bhalla M, Bou Ghanem EN. Activating A1 adenosine receptor signaling boosts early pulmonary neutrophil recruitment in aged mice in response to Streptococcus pneumoniae infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574741. [PMID: 38260350 PMCID: PMC10802397 DOI: 10.1101/2024.01.08.574741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Streptococcus pneumoniae (pneumococcus) is a leading cause of pneumonia in older adults. Successful control of pneumococci requires robust pulmonary neutrophil influx early in infection. However, aging is associated with aberrant neutrophil recruitment and the mechanisms behind that are not understood. Here we explored how neutrophil recruitment following pneumococcal infection changes with age and the host pathways regulating this. Results Following pneumococcal infection there was a significant delay in early neutrophil recruitment to the lungs of aged mice. Neutrophils from aged mice showed defects in trans-endothelial migration in vitro compared to young controls. To understand the pathways involved, we examined immune modulatory extracellular adenosine (EAD) signaling, that is activated upon cellular damage. Signaling through the lower affinity A2A and A2B adenosine receptors had no effect on neutrophil recruitment to infected lungs. In contrast, inhibition of the high affinity A1 receptor in young mice blunted neutrophil recruitment to the lungs following infection. A1 receptor inhibition decreased expression of CXCR2 on circulating neutrophils, which is required for transendothelial migration. Indeed, A1 receptor signaling on neutrophils was required for their ability to migrate across endothelial cells in response to infection. Aging was not associated with defects in EAD production or receptor expression on neutrophils. However, agonism of A1 receptor in aged mice rescued the early defect in neutrophil migration to the lungs and improved control of bacterial burden. Conclusions This study suggests age-driven defects in EAD damage signaling can be targeted to rescue the delay in pulmonary neutrophil migration in response to bacterial pneumonia.
Collapse
|
6
|
Cantor J. Maximizing the Therapeutic Effect of Endothelin Receptor Antagonists in Pulmonary Fibrosis: A Paradigm for Treating the Disease. Int J Mol Sci 2024; 25:4184. [PMID: 38673771 PMCID: PMC11050024 DOI: 10.3390/ijms25084184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/12/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Using a lipopolysaccharide model of acute lung injury, we previously showed that endothelin-1 (ET-1), a potent mediator of vasoconstriction, may act as a "gatekeeper" for the influx of inflammatory cells into the lung. These studies provided a rationale for testing the effect of HJP272, an endothelin receptor antagonist (ERA), in hamster models of pulmonary fibrosis induced by intratracheal instillation of either bleomycin (BLM) or amiodarone (AM). To determine the temporal effects of blocking ET-1 activity, animals were given HJP272 either 1 h before initiation of lung injury or 24 h afterward. The results indicated that pretreatment with this agent caused significant reductions in various inflammatory parameters, whereas post-treatment was ineffective. This finding suggests that ERAs are only effective at a very early stage of pulmonary fibrosis and explains their lack of success in clinical trials involving patients with this disease. Nevertheless, ERAs could serve as prophylactic agents when combined with drugs that may induce pulmonary fibrosis. Furthermore, developing a biomarker for the initial changes in the lung extracellular matrix could increase the efficacy of ERAs and other therapeutic agents in preventing the progression of the disease. While no such biomarker currently exists, we propose the ratio of free to peptide-bound desmosine, a unique crosslink of elastin, as a potential candidate for detecting the earliest modifications in lung microarchitecture associated with pulmonary fibrosis.
Collapse
Affiliation(s)
- Jerome Cantor
- School of Pharmacy and Health Sciences, Queens, NY 11439, USA
| |
Collapse
|
7
|
Holloman BL, Wilson K, Cannon A, Nagarkatti M, Nagarkatti PS. Indole-3-carbinol attenuates lipopolysaccharide-induced acute respiratory distress syndrome through activation of AhR: role of CCR2+ monocyte activation and recruitment in the regulation of CXCR2+ neutrophils in the lungs. Front Immunol 2024; 15:1330373. [PMID: 38596679 PMCID: PMC11002125 DOI: 10.3389/fimmu.2024.1330373] [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: 10/30/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction Indole-3-carbinol (I3C) is found in cruciferous vegetables and used as a dietary supplement. It is known to act as a ligand for aryl hydrocarbon receptor (AhR). In the current study, we investigated the role of AhR and the ability of I3C to attenuate LPS-induced Acute Respiratory Distress Syndrome (ARDS). Methods To that end, we induced ARDS in wild-type C57BL/6 mice, Ccr2gfp/gfp KI/KO mice (mice deficient in the CCR2 receptor), and LyZcreAhRfl/fl mice (mice deficient in the AhR on myeloid linage cells). Additionally, mice were treated with I3C (65 mg/kg) or vehicle to investigate its efficacy to treat ARDS. Results I3C decreased the neutrophils expressing CXCR2, a receptor associated with neutrophil recruitment in the lungs. In addition, LPS-exposed mice treated with I3C revealed downregulation of CCR2+ monocytes in the lungs and lowered CCL2 (MCP-1) protein levels in serum and bronchoalveolar lavage fluid. Loss of CCR2 on monocytes blocked the recruitment of CXCR2+ neutrophils and decreased the total number of immune cells in the lungs during ARDS. In addition, loss of the AhR on myeloid linage cells ablated I3C-mediated attenuation of CXCR2+ neutrophils and CCR2+ monocytes in the lungs from ARDS animals. Interestingly, scRNASeq showed that in macrophage/monocyte cell clusters of LPS-exposed mice, I3C reduced the expression of CXCL2 and CXCL3, which bind to CXCR2 and are involved in neutrophil recruitment to the disease site. Discussion These findings suggest that CCR2+ monocytes are involved in the migration and recruitment of CXCR2+ neutrophils during ARDS, and the AhR ligand, I3C, can suppress ARDS through the regulation of immune cell trafficking.
Collapse
Affiliation(s)
| | | | | | | | - Prakash S. Nagarkatti
- Nagarkatti Laboratory, University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC, United States
| |
Collapse
|
8
|
Murphy DM, Walsh A, Stein L, Petrasca A, Cox DJ, Brown K, Duffin E, Jameson G, Connolly SA, O'Connell F, O'Sullivan J, Basdeo SA, Keane J, Phelan JJ. Human Macrophages Activate Bystander Neutrophils' Metabolism and Effector Functions When Challenged with Mycobacterium tuberculosis. Int J Mol Sci 2024; 25:2898. [PMID: 38474145 DOI: 10.3390/ijms25052898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Neutrophils are dynamic cells, playing a critical role in pathogen clearance; however, neutrophil infiltration into the tissue can act as a double-edged sword. They are one of the primary sources of excessive inflammation during infection, which has been observed in many infectious diseases including pneumonia and active tuberculosis (TB). Neutrophil function is influenced by interactions with other immune cells within the inflammatory lung milieu; however, how these interactions affect neutrophil function is unclear. Our study examined the macrophage-neutrophil axis by assessing the effects of conditioned medium (MΦ-CM) from primary human monocyte-derived macrophages (hMDMs) stimulated with LPS or a whole bacterium (Mycobacterium tuberculosis) on neutrophil function. Stimulated hMDM-derived MΦ-CM boosts neutrophil activation, heightening oxidative and glycolytic metabolism, but diminishes migratory potential. These neutrophils exhibit increased ROS production, elevated NET formation, and heightened CXCL8, IL-13, and IL-6 compared to untreated or unstimulated hMDM-treated neutrophils. Collectively, these data show that MΦ-CM from stimulated hMDMs activates neutrophils, bolsters their energetic profile, increase effector and inflammatory functions, and sequester them at sites of infection by decreasing their migratory capacity. These data may aid in the design of novel immunotherapies for severe pneumonia, active tuberculosis and other diseases driven by pathological inflammation mediated by the macrophage-neutrophil axis.
Collapse
Affiliation(s)
- Dearbhla M Murphy
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Anastasija Walsh
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Laura Stein
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Andreea Petrasca
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, The University of Dublin, D02 R590 Dublin, Ireland
| | - Donal J Cox
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Kevin Brown
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Emily Duffin
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Gráinne Jameson
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Sarah A Connolly
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Fiona O'Connell
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute (TTMI), St. James's Hospital, Dublin 8, D08 W9RT Dublin, Ireland
| | - Jacintha O'Sullivan
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute (TTMI), St. James's Hospital, Dublin 8, D08 W9RT Dublin, Ireland
| | - Sharee A Basdeo
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| | - James J Phelan
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, D08 W9RT Dublin, Ireland
| |
Collapse
|
9
|
Zhu H, Ding D, Fan X, Yang Q, Wang Y, Xue H, Kang C. The occurrence and development of vertebral osteoporosis regulated by IL-8. Medicine (Baltimore) 2023; 102:e35680. [PMID: 37933016 PMCID: PMC10627673 DOI: 10.1097/md.0000000000035680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 11/08/2023] Open
Abstract
Vertebral osteoporotic fracture is a common type of fracture, and the incidence is higher in the elderly. However, the relationship between vertebral osteoporotic fractures and interleukin-8 (IL-8) remains unclear. A total of 163 patients with osteoporotic vertebral fractures were recruited. Clinical and follow-up data were recorded, and the expression levels of IL1, MMP9, IL-8, and C-reactive protein in blood were measured. Pearson Chi-square test and Spearman correlation coefficient were used to analyze the relationship between vertebral osteoporotic fractures and related parameters. Univariate and multivariate logistic regression and univariate and multivariate Cox proportional hazards regression were used for further analysis. Pearson chi-square test, Spearman correlation coefficient and Logistic regression analysis showed that IL1 and IL-8 were significantly associated with vertebral osteoporotic fractures. Univariate Cox regression analysis showed that age and IL-8 expression level were significantly associated with maintenance time from recovery to recurrence of vertebral osteoporotic fractures. Multivariate Cox regression analysis showed that IL-8 expression level was significantly associated with maintenance time from recovery to recurrence of vertebral osteoporotic fractures. The higher the expression level of IL-8, the more likely it is to develop vertebral osteoporotic fracture, and the more likely it is to relapse in a short time.
Collapse
Affiliation(s)
- Hao Zhu
- Department of Orthopedics, The Second Central Hospital of Baoding, Zhuozhou City, Hebei Province, P.R. China
| | - Danyang Ding
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Badachu Xixia Zhuang, Shijingshan District, Beijing, P.R. China
| | - Xingyu Fan
- Rehabilitation Center, Lianyungang First People’s Hospital, Lianyungang City, Jiangsu Province, P.R. China
| | - Qian Yang
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Badachu Xixia Zhuang, Shijingshan District, Beijing, P.R. China
| | - Ye Wang
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Badachu Xixia Zhuang, Shijingshan District, Beijing, P.R. China
| | - Hui Xue
- Department of Orthopedics, The Second Central Hospital of Baoding, Zhuozhou City, Hebei Province, P.R. China
| | - Chunbo Kang
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Badachu Xixia Zhuang, Shijingshan District, Beijing, P.R. China
| |
Collapse
|
10
|
Du Y, Chen Y, Li F, Mao Z, Ding Y, Wang W. Genetically Engineered Cellular Nanovesicle as Targeted DNase I Delivery System for the Clearance of Neutrophil Extracellular Traps in Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303053. [PMID: 37759381 PMCID: PMC10646266 DOI: 10.1002/advs.202303053] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/18/2023] [Indexed: 09/29/2023]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) are prevalent critical illnesses with a high mortality rate among patients in intensive care units. Neutrophil extracellular traps (NETs) are implicated in the pathogenesis of ALI/ARDS and represent a promising therapeutic target. However, the clinical application of deoxyribonuclease I (DNase I), the only drug currently available to clear NETs, is limited due to the lack of precise and efficient delivery strategies. Therefore, targeted delivery of DNase I to the inflamed lung remains a critical issue to be addressed. Herein, a novel biomimetic DNase I delivery system is developed (DCNV) that employs genetically and bioorthogonally engineered cellular nanovesicles for pulmonary NETs clearance. The CXC motif chemokine receptor 2 overexpressed cellular nanovesicles can mimic the inflammatory chemotaxis of neutrophils in ALI/ARDS, leading to enhanced lung accumulation. Furthermore, DNase I immobilized through bioorthogonal chemistry exhibits remarkable enzymatic activity in NETs degradation, thus restraining inflammation and safeguarding lung tissue in the lipopolysaccharide-induced ALI murine model. Collectively, the findings present a groundbreaking proof-of-concept in the utilization of biomimetic cellular nanovesicles to deliver DNase I for treating ALI/ARDS. This innovative strategy may usher in a new era in the development of pharmacological interventions for various inflammation-related diseases.
Collapse
Affiliation(s)
- Yang Du
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouZhejiang310009China
- National Innovation Center for Fundamental Research on Cancer MedicineHangzhouZhejiang310009China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- ZJU‐Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic DiseaseHangzhouZhejiang310058China
| | - Yining Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouZhejiang310009China
- National Innovation Center for Fundamental Research on Cancer MedicineHangzhouZhejiang310009China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- ZJU‐Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic DiseaseHangzhouZhejiang310058China
| | - Fangyuan Li
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouZhejiang310009China
- National Innovation Center for Fundamental Research on Cancer MedicineHangzhouZhejiang310009China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- ZJU‐Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic DiseaseHangzhouZhejiang310058China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouZhejiang310009China
- National Innovation Center for Fundamental Research on Cancer MedicineHangzhouZhejiang310009China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- ZJU‐Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic DiseaseHangzhouZhejiang310058China
| |
Collapse
|
11
|
Gunalp S, Helvaci DG, Oner A, Bursalı A, Conforte A, Güner H, Karakülah G, Szegezdi E, Sag D. TRAIL promotes the polarization of human macrophages toward a proinflammatory M1 phenotype and is associated with increased survival in cancer patients with high tumor macrophage content. Front Immunol 2023; 14:1209249. [PMID: 37809073 PMCID: PMC10551148 DOI: 10.3389/fimmu.2023.1209249] [Citation(s) in RCA: 2] [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: 04/20/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Background TNF-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can either induce cell death or activate survival pathways after binding to death receptors (DRs) DR4 or DR5. TRAIL is investigated as a therapeutic agent in clinical trials due to its selective toxicity to transformed cells. Macrophages can be polarized into pro-inflammatory/tumor-fighting M1 macrophages or anti-inflammatory/tumor-supportive M2 macrophages and an imbalance between M1 and M2 macrophages can promote diseases. Therefore, identifying modulators that regulate macrophage polarization is important to design effective macrophage-targeted immunotherapies. The impact of TRAIL on macrophage polarization is not known. Methods Primary human monocyte-derived macrophages were pre-treated with either TRAIL or with DR4 or DR5-specific ligands and then polarized into M1, M2a, or M2c phenotypes in vitro. The expression of M1 and M2 markers in macrophage subtypes was analyzed by RNA sequencing, qPCR, ELISA, and flow cytometry. Furthermore, the cytotoxicity of the macrophages against U937 AML tumor targets was assessed by flow cytometry. TCGA datasets were also analyzed to correlate TRAIL with M1/M2 markers, and the overall survival of cancer patients. Results TRAIL increased the expression of M1 markers at both mRNA and protein levels while decreasing the expression of M2 markers at the mRNA level in human macrophages. TRAIL also shifted M2 macrophages towards an M1 phenotype. Our data showed that both DR4 and DR5 death receptors play a role in macrophage polarization. Furthermore, TRAIL enhanced the cytotoxicity of macrophages against the AML cancer cells in vitro. Finally, TRAIL expression was positively correlated with increased expression of M1 markers in the tumors from ovarian and sarcoma cancer patients and longer overall survival in cases with high, but not low, tumor macrophage content. Conclusions TRAIL promotes the polarization of human macrophages toward a proinflammatory M1 phenotype via both DR4 and DR5. Our study defines TRAIL as a new regulator of macrophage polarization and suggests that targeting DRs can enhance the anti-tumorigenic response of macrophages in the tumor microenvironment by increasing M1 polarization.
Collapse
Affiliation(s)
- Sinem Gunalp
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Derya Goksu Helvaci
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Aysenur Oner
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | | | - Alessandra Conforte
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Hüseyin Güner
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Science, Abdullah Gül University, Kayseri, Türkiye
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Eva Szegezdi
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Duygu Sag
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| |
Collapse
|
12
|
Xie W, Huang T, Guo Y, Zhang Y, Chen W, Li Y, Chen C, Li P. Neutrophil-derived cathelicidin promotes cerebral angiogenesis after ischemic stroke. J Cereb Blood Flow Metab 2023; 43:1503-1518. [PMID: 37194247 PMCID: PMC10414012 DOI: 10.1177/0271678x231175190] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/10/2023] [Accepted: 04/09/2023] [Indexed: 05/18/2023]
Abstract
Neutrophils play critical roles in the evolving of brain injuries following ischemic stroke. However, how they impact the brain repair in the late phase after stroke remain uncertain. Using a prospective clinical stroke patient cohort, we found significantly increased cathelicidin antimicrobial peptide (CAMP) in the peripheral blood of stroke patients compared to that of healthy controls. While in the mouse stroke model, CAMP was present in the peripheral blood, brain ischemic core and significantly increased at day 1, 3, 7, 14 after middle cerebral artery occlusion (MCAO). CAMP-/- mice exhibited significantly increased infarct volume, exacerbated neurological outcome, reduced cerebral endothelial cell proliferation and vascular density at 7 and 14 days after MCAO. Using bEND3 cells subjected to oxygen-glucose deprivation (OGD), we found significantly increased angiogenesis-related gene expression with the treatment of recombinant CAMP peptide (rCAMP) after reoxygenation. Intracerebroventricular injection (ICV) of AZD-5069, the antagonist of CAMP receptor CXCR2, or knockdown of CXCR2 by shCXCR2 recombinant adeno-associated virus (rAAV) impeded angiogenesis and neurological recovery after MCAO. Administration of rCAMP promoted endothelial proliferation and angiogenesis and attenuated neurological deficits 14 days after MCAO. In conclusion, neutrophil derived CAMP represents an important mediator that could promote post-stroke angiogenesis and neurological recovery in the late phase after stroke.
Collapse
Affiliation(s)
| | | | | | - Yueman Zhang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijie Chen
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Saemann L, Naujoks P, Hartrumpf L, Pohl S, Simm A, Szabó G. Sex-Specific Protection of Endothelial Function after Vascular Ischemia/Reperfusion Injury by the Senomorphic Agent Ruxolitinib. Int J Mol Sci 2023; 24:11727. [PMID: 37511486 PMCID: PMC10381013 DOI: 10.3390/ijms241411727] [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: 06/26/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Ischemia/reperfusion (I/R)-induced endothelial dysfunction occurs in various cardiovascular disorders. I/R injury is partially driven by the release of cytokines. Known for its use in senotherapy, the JAK inhibitor ruxolitinib is able to block the release of cytokines. We investigated the effect of ruxolitinib on the cytokine release and endothelial-dependent vasorelaxation in an in vitro model of I/R. Aortic segments of C57BL/6J mice (N = 12/group) were divided into three groups: control, in vitro I/R (I/R group), and in vitro I/R with ruxolitinib during ischemic incubation (I/R+Ruxo group). We determined cytokine expression. In organ bath chambers, we investigated the maximal endothelial-dependent relaxation to acetylcholine (RmaxACh) and maximal endothelial-independent relaxation to sodium-nitroprusside (RmaxSNP). RmaxACh was decreased in I/R compared to the control (83.6 ± 2.4 vs. 48.6 ± 3.4%; p < 0.05) and I/R+Ruxo (74.4 ± 2.6 vs. 48.6 ± 3.4%; p < 0.05). RmaxSNP was comparable between all groups. IL-10 was detectable only in I/R+Ruxo. CXCL5, CCL2, CCL3, CCL8, CCL11, ICAM-1, IL-1α, IL-7, TNF-α, and G-CSF were decreased or not detectable in I/R+Ruxo. In I/R+Ruxo, ICAM-1 was reduced in rings only from male mice. Treatment of the aorta from mice during in vitro ischemia with the senomorphic agent ruxolitinib reduces cytokine release and protects the endothelium from I/R-mediated dysfunction.
Collapse
Affiliation(s)
- Lars Saemann
- Department of Cardiac Surgery, University Hospital Halle, 06120 Halle, Germany
| | - Paula Naujoks
- Department of Cardiac Surgery, University Hospital Halle, 06120 Halle, Germany
| | - Lotta Hartrumpf
- Department of Cardiac Surgery, University Hospital Halle, 06120 Halle, Germany
| | - Sabine Pohl
- Department of Cardiac Surgery, University Hospital Halle, 06120 Halle, Germany
| | - Andreas Simm
- Department of Cardiac Surgery, University Hospital Halle, 06120 Halle, Germany
| | - Gábor Szabó
- Department of Cardiac Surgery, University Hospital Halle, 06120 Halle, Germany
- Department of Cardiac Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| |
Collapse
|
14
|
Maus KD, Stephenson DJ, Macknight HP, Vu NT, Hoeferlin LA, Kim M, Diegelmann RF, Xie X, Chalfant CE. Skewing cPLA 2α activity toward oxoeicosanoid production promotes neutrophil N2 polarization, wound healing, and the response to sepsis. Sci Signal 2023; 16:eadd6527. [PMID: 37433004 PMCID: PMC10565596 DOI: 10.1126/scisignal.add6527] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 06/16/2023] [Indexed: 07/13/2023]
Abstract
Uncontrolled inflammation is linked to poor outcomes in sepsis and wound healing, both of which proceed through distinct inflammatory and resolution phases. Eicosanoids are a class of bioactive lipids that recruit neutrophils and other innate immune cells. The interaction of ceramide 1-phosphate (C1P) with the eicosanoid biosynthetic enzyme cytosolic phospholipase A2 (cPLA2) reduces the production of a subtype of eicosanoids called oxoeicosanoids. We investigated the effect of shifting the balance in eicosanoid biosynthesis on neutrophil polarization and function. Knockin mice expressing a cPLA2 mutant lacking the C1P binding site (cPLA2αKI/KI mice) showed enhanced and sustained neutrophil infiltration into wounds and the peritoneum during the inflammatory phase of wound healing and sepsis, respectively. The mice exhibited improved wound healing and reduced susceptibility to sepsis, which was associated with an increase in anti-inflammatory N2-type neutrophils demonstrating proresolution behaviors and a decrease in proinflammatory N1-type neutrophils. The N2 polarization of cPLA2αKI/KI neutrophils resulted from increased oxoeicosanoid biosynthesis and autocrine signaling through the oxoeicosanoid receptor OXER1 and partially depended on OXER1-dependent inhibition of the pentose phosphate pathway (PPP). Thus, C1P binding to cPLA2α suppresses neutrophil N2 polarization, thereby impairing wound healing and the response to sepsis.
Collapse
Affiliation(s)
- Kenneth D Maus
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Daniel J Stephenson
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - H Patrick Macknight
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - Ngoc T Vu
- Department of Applied Biochemistry, School of Biotechnology, International University-VNU HCM, Ho Chi Minh City, Vietnam
| | - L Alexis Hoeferlin
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University-School of Medicine, Richmond VA 23298, USA
| | - Minjung Kim
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Robert F Diegelmann
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University-School of Medicine, Richmond VA 23298, USA
| | - Xiujie Xie
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - Charles E Chalfant
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA 22903, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22903, USA
- Program in Cancer Biology, University of Virginia Cancer Center, Charlottesville, VA 22903, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond VA, 23298, USA
| |
Collapse
|
15
|
Chatterjee T, Lewis TL, Arora I, Gryshyna AE, Underwood L, Masjoan Juncos JX, Aggarwal S. Sex-Based Disparities in Leukocyte Migration and Activation in Response to Inhalation Lung Injury: Role of SDF-1/CXCR4 Signaling. Cells 2023; 12:1719. [PMID: 37443753 PMCID: PMC10340292 DOI: 10.3390/cells12131719] [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: 05/25/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of the study was to determine whether sex-related differences exist in immune response to inhalation lung injury. C57BL/6 mice were exposed to Cl2 gas (500 ppm for 15, 20, or 30 min). Results showed that male mice have higher rates of mortality and lung injury than females. The binding of the chemokine ligand C-X-C motif chemokine 12 (CXCL12), also called stromal-derived-factor-1 (SDF-1), to the C-X-C chemokine receptor type 4 (CXCR4) on lung cells promotes the migration of leukocytes from circulation to lungs. Therefore, the hypothesis was that elevated SDF-1/CXCR4 signaling mediates exaggerated immune response in males. Plasma, blood leukocytes, and lung cells were collected from mice post-Cl2 exposure. Plasma levels of SDF-1 and peripheral levels of CXCR4 in lung cells were higher in male vs. female mice post-Cl2 exposure. Myeloperoxidase (MPO) and elastase activity was significantly increased in leukocytes of male mice exposed to Cl2. Lung cells were then ex vivo treated with SDF-1 (100 ng/mL) in the presence or absence of the CXCR4 inhibitor, AMD3100 (100 nM). SDF-1 significantly increased migration, MPO, and elastase activity in cells obtained from male vs. female mice post-Cl2 exposure. AMD3100 attenuated these effects, suggesting that differential SDF-1/CXCR4 signaling may be responsible for sex-based disparities in the immune response to inhalation lung injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205-3703, USA; (T.C.); (T.L.L.); (I.A.); (A.E.G.); (L.U.); (J.X.M.J.)
| |
Collapse
|
16
|
Fadanni GP, Calixto JB. Recent progress and prospects for anti-cytokine therapy in preclinical and clinical acute lung injury. Cytokine Growth Factor Rev 2023; 71-72:13-25. [PMID: 37481378 DOI: 10.1016/j.cytogfr.2023.07.002] [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: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/24/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a heterogeneous cause of respiratory failure that has a rapid onset, a high mortality rate, and for which there is no effective pharmacological treatment. Current evidence supports a critical role of excessive inflammation in ARDS, resulting in several cytokines, cytokine receptors, and proteins within their downstream signalling pathways being putative therapeutic targets. However, unsuccessful trials of anti-inflammatory drugs have thus far hindered progress in the field. In recent years, the prospects of precision medicine and therapeutic targeting of cytokines coevolving into effective treatments have gained notoriety. There is an optimistic and growing understanding of ARDS subphenotypes as well as advances in treatment strategies and clinical trial design. Furthermore, large trials of anti-cytokine drugs in patients with COVID-19 have provided an unprecedented amount of information that could pave the way for therapeutic breakthroughs. While current clinical and nonclinical ARDS research suggest relatively limited potential in monotherapy with anti-cytokine drugs, combination therapy has emerged as an appealing strategy and may provide new perspectives on finding safe and effective treatments. Accurate evaluation of these drugs, however, also relies on well-founded experimental research and the implementation of biomarker-guided stratification in future trials. In this review, we provide an overview of anti-cytokine therapy for acute lung injury and ARDS, highlighting the current preclinical and clinical evidence for targeting the main cytokines individually and the therapeutic prospects for combination therapy.
Collapse
Affiliation(s)
- Guilherme Pasetto Fadanni
- Centre of Innovation and Preclinical Studies (CIEnP), Florianópolis, Santa Catarina, Brazil; Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil.
| | - João Batista Calixto
- Centre of Innovation and Preclinical Studies (CIEnP), Florianópolis, Santa Catarina, Brazil; Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
17
|
Wang SW, Zhang Q, Lu D, Fang YC, Yan XC, Chen J, Xia ZK, Yuan QT, Chen LH, Zhang YM, Nan FJ, Xie X. GPR84 regulates pulmonary inflammation by modulating neutrophil functions. Acta Pharmacol Sin 2023:10.1038/s41401-023-01080-z. [PMID: 37016043 PMCID: PMC10072043 DOI: 10.1038/s41401-023-01080-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/14/2023] [Indexed: 04/06/2023] Open
Abstract
Acute lung injury (ALI) is an acute, progressive hypoxic respiratory failure that could develop into acute respiratory distress syndrome (ARDS) with very high mortality rate. ALI is believed to be caused by uncontrolled inflammation, and multiple types of immune cells, especially neutrophils, are critically involved in the development of ALI. The treatment for ALI/ARDS is very limited, a better understanding of the pathogenesis and new therapies are urgently needed. Here we discover that GPR84, a medium chain fatty acid receptor, plays critical roles in ALI development by regulating neutrophil functions. GPR84 is highly upregulated in the cells isolated from the bronchoalveolar lavage fluid of LPS-induced ALI mice. GPR84 deficiency or blockage significantly ameliorated ALI mice lung inflammation by reducing neutrophils infiltration and oxidative stress. Further studies reveal that activation of GPR84 strongly induced reactive oxygen species production from neutrophils by stimulating Lyn, AKT and ERK1/2 activation and the assembly of the NADPH oxidase. These results reveal an important role of GPR84 in neutrophil functions and lung inflammation and strongly suggest that GPR84 is a potential drug target for ALI.
Collapse
Affiliation(s)
- Si-Wei Wang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing Zhang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Dan Lu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - You-Chen Fang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Ci Yan
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Jing Chen
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhi-Kan Xia
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian-Ting Yuan
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lin-Hai Chen
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | | | - Fa-Jun Nan
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Xin Xie
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
18
|
Gong HH, Worley MJ, Carver KA, Goldstein DR, Deng JC. Neutrophils drive pulmonary vascular leakage in MHV-1 infection of susceptible A/J mice. Front Immunol 2023; 13:1089064. [PMID: 36685578 PMCID: PMC9853883 DOI: 10.3389/fimmu.2022.1089064] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023] Open
Abstract
Background Lung inflammation, neutrophil infiltration, and pulmonary vascular leakage are pathological hallmarks of acute respiratory distress syndrome (ARDS) which can lethally complicate respiratory viral infections. Despite similar comorbidities, however, infections in some patients may be asymptomatic while others develop ARDS as seen with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections for example. Methods In this study, we infected resistant C57BL/6 and susceptible A/J strains of mice with pulmonary administration of murine hepatitis virus strain 1 (MHV-1) to determine mechanisms underlying susceptibility to pulmonary vascular leakage in a respiratory coronavirus infection model. Results A/J animals displayed increased lung injury parameters, pulmonary neutrophil influx, and deficient recruitment of other leukocytes early in the infection. Moreover, under basal conditions, A/J neutrophils overexpressed primary granule protein genes for myeloperoxidase and multiple serine proteases. During infection, myeloperoxidase and elastase protein were released in the bronchoalveolar spaces at higher concentrations compared to C57BL/6 mice. In contrast, genes from other granule types were not differentially expressed between these 2 strains. We found that depletion of neutrophils led to mitigation of lung injury in infected A/J mice while having no effect in the C57BL/6 mice, demonstrating that an altered neutrophil phenotype and recruitment profile is a major driver of lung immunopathology in susceptible mice. Conclusions These results suggest that host susceptibility to pulmonary coronaviral infections may be governed in part by underlying differences in neutrophil phenotypes, which can vary between mice strains, through mechanisms involving primary granule proteins as mediators of neutrophil-driven lung injury.
Collapse
Affiliation(s)
- Henry H. Gong
- University of Michigan, Ann Arbor, MI, United States
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Matthew J. Worley
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Kyle A. Carver
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Daniel R. Goldstein
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Jane C. Deng
- University of Michigan, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Medicine Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
| |
Collapse
|
19
|
Matsushima K, Shichino S, Ueha S. Thirty-five years since the discovery of chemotactic cytokines, interleukin-8 and MCAF: A historical overview. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2023; 99:213-226. [PMID: 37518010 DOI: 10.2183/pjab.99.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Inflammation is a host defense response to various invading stimuli, but an excessive and persistent inflammatory response can cause tissue injury, which can lead to irreversible organ damage and dysfunction. Excessive inflammatory responses are believed to link to most human diseases. A specific type of leukocyte infiltration into invaded tissues is required for inflammation. Historically, the underlying molecular mechanisms of this process during inflammation were an enigma, compromising research in the fields of inflammation, immunology, and pathology. However, the pioneering discovery of chemotactic cytokines (chemokines), monocyte-derived neutrophil chemotactic factor (MDNCF; interleukin [IL]-8, CXCL8) and monocyte chemotactic and activating factor (MCAF; monocyte chemotactic factor 1 [MCP-1], CCL2) in the late 1980s finally enabled us to address this issue. In this review, we provide a historical overview of chemokine research over the last 35 years.
Collapse
Affiliation(s)
- Kouji Matsushima
- Division of Molecular Regulation of Inflammation and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammation and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammation and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science
| |
Collapse
|
20
|
Wang W, Zhang Z, Liu Y, Kong L, Li W, Hu W, Wang Y, Liu X. Nano-integrated cascade antioxidases opsonized by albumin bypass the blood-brain barrier for treatment of ischemia-reperfusion injury. Biomater Sci 2022; 10:7103-7116. [PMID: 36341569 DOI: 10.1039/d2bm01401g] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Potent antioxidative drugs are urgently needed to treat ischemia-reperfusion (I/R) induced reactive oxygen species (ROS)-mediated cerebrovascular and neural injury during ischemia strokes. However, current antioxidative agents have limited application in such disease due to low blood-brain barrier (BBB) penetration. We herein designed a "neutrophil piggybacking" strategy based on albumin opsonized nanoparticles co-encapsulated with antioxidases catalase (CAT) and superoxide dismutase 1 (SOD1). The system utilized the natural potential of neutrophils to target inflamed tissues to deliver antioxidases to injured sites in the brain. In addition, the system was integrated with a selenium (Se)-containing crosslinker to inhibit ferroptosis. We showed that the nanoparticles opsonized in the hybrid form rather than with an albumin-shell structure exhibited enhanced neutrophil targeting and efficient BBB penetration in vitro and in vivo. We further showed that the neutrophil-mediated delivery of antioxidases effectively reduced oxidative damage and apoptosis of neurons in brain tissue in a transient middle cerebral artery occlusion (tMCAO) mouse model. Moreover, the successful delivery of Se with the nanoparticles increased the expression of glutathione peroxidase 4 (GPX4) and effectively inhibited neuronal ferroptosis, achieving a satisfactory neuroprotective effect in I/R injury mice. Our study demonstrated that the rationally designed nanomedicines using the "neutrophil piggybacking" strategy can efficiently penetrate the BBB, greatly expanding the application of nanomedicines in the treatment of central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Wuxuan Wang
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China.
| | - Zheng Zhang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences University of Science and Technology of China, Hefei, Anhui 230027, China. .,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yi Liu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Lingqi Kong
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China.
| | - Wenyu Li
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China.
| | - Wei Hu
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China.
| | - Yucai Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Xinfeng Liu
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China.
| |
Collapse
|
21
|
Liu N, Bauer M, Press AT. The immunological function of CXCR2 in the liver during sepsis. J Inflamm (Lond) 2022; 19:23. [DOI: 10.1186/s12950-022-00321-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 11/15/2022] [Indexed: 12/02/2022] Open
Abstract
Abstract
Background
The chemokine receptor CXCR2 and its ligands, especially CXCL8, are crucial mediators for the progression of liver inflammation and liver failure in sepsis. Neutrophils have the highest CXCR2 expression in mice and humans, and their activation via CXCL8 facilitates their migration to the inflamed liver for the clearance of the pathogens and, in turn, the inflammation.
Main body
In sepsis, the inflammatory insult causes extensive neutrophil migration to the liver that overwhelms the immune response. To compensate for the strong receptor activation, CXCR2 desensitizes, incapacitating the immune cells to efficiently clear pathogens, causing further life-threatening liver damage and uncontrolled pathogen spread.
Conclusion
CXCR2 function during infection strongly depends on the expressing cell type. It signals pro- and anti-inflammatory effects that may prompt novel cell-type-specific CXCR2-directed therapeutics.
Collapse
|
22
|
Immunological and Pathological Peculiarity of Severe Acute Respiratory Syndrome Coronavirus 2 Beta Variant. Microbiol Spectr 2022; 10:e0237122. [PMID: 36005818 PMCID: PMC9602775 DOI: 10.1128/spectrum.02371-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Diverse severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have emerged since the beginning of the COVID-19 pandemic. We investigated the immunological and pathological peculiarity of the SARS-CoV-2 beta variant of concern (VoC) compared to the ancestral strain. Comparative analysis of phenotype and pathology revealed that the beta VoC induces slower disease progression and a prolonged presymptomatic period in the early stages of SARS-CoV-2 infection but ultimately causes sudden death in the late stages of infection in the K18-hACE2 mouse model. The beta VoC induced enhanced activation of CXCL1/2-CXCR2-NLRP3-IL-1β signal cascade accelerating neutrophil recruitment and lung pathology in beta variant-infected mice, as evidenced by multiple analyses of SARS-CoV-2-induced inflammatory cytokines and transcriptomes. CCL2 was one of the most highly secreted cytokines in the early stages of infection. Its blockade reduced virus-induced weight loss and delayed mortality. Our study provides a better understanding of the variant characteristics and need for treatment. IMPORTANCE Since the outbreak of COVID-19, diverse SARS-CoV-2 variants have been identified. These variants have different infectivity and transmissibility from the ancestral strains. However, underlying molecular mechanisms have not yet been fully elucidated. In our study, the beta variant showed distinct pathological conditions and cytokine release kinetics from an ancestral strain in a mouse model. It was associated with higher neutrophil recruitment by increased levels of CXCL1/2, CXCR2, and interleukin 1β (IL-1β) at a later stage of viral infection. Our study will provide a better understanding of SARS-CoV-2 pathogenesis.
Collapse
|
23
|
Komolafe K, Pacurari M. CXC Chemokines in the Pathogenesis of Pulmonary Disease and Pharmacological Relevance. Int J Inflam 2022; 2022:4558159. [PMID: 36164329 PMCID: PMC9509283 DOI: 10.1155/2022/4558159] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Chemokines and their receptors play important roles in the pathophysiology of many diseases by regulating the cellular migration of major inflammatory and immune players. The CXC motif chemokine subfamily is the second largest family, and it is further subdivided into ELR motif CXC (ELR+) and non-ELR motif (ELR-) CXC chemokines, which are effective chemoattractants for neutrophils and lymphocytes/monocytes, respectively. These chemokines and their receptors are expected to have a significant impact on a wide range of lung diseases, many of which have inflammatory or immunological underpinnings. As a result, manipulations of this subfamily of chemokines and their receptors using small molecular agents and other means have been explored for potential therapeutic benefit in the setting of several lung pathologies. Furthermore, encouraging preclinical data has necessitated the progression of a few of these drugs into clinical trials in order to make the most effective use of interventions in the development of viable targeted therapeutics. The current review presents the understanding of the roles of CXC ligands (CXCLs) and their cognate receptors (CXCRs) in the pathogenesis of several lung diseases such as allergic rhinitis, COPD, lung fibrosis, lung cancer, pneumonia, and tuberculosis. The potential therapeutic benefits of pharmacological or other CXCL/CXCR axis manipulations are also discussed.
Collapse
Affiliation(s)
- Kayode Komolafe
- RCMI Center for Health Disparities Research, Jackson State University, Jackson, MS 39217, USA
| | - Maricica Pacurari
- RCMI Center for Health Disparities Research, Jackson State University, Jackson, MS 39217, USA
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, Jackson, MS 39217, USA
| |
Collapse
|
24
|
The Renshen Chishao Decoction Could Ameliorate the Acute Lung Injury but Could Not Reduce the Neutrophil Extracellular Traps Formation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7784148. [PMID: 36072401 PMCID: PMC9444383 DOI: 10.1155/2022/7784148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/18/2022] [Accepted: 08/01/2022] [Indexed: 11/25/2022]
Abstract
The acute lung injury (ALI) causes severe pulmonary diseases, leading to a high mortality rate. The Renshen and Chishao have protective and anti-inflammatory effects against the ALI. To explore the protective effects of the Renshen Chishao (RC) decoction against the ALI, we established the lipopolysaccharide-indued ALI model and randomly divided the mice into seven groups: control group, ALI group, high-dose RC group, middle-dose RC group, low-dose RC group, middle-dose RC group + CXCR2 antagonist group, and ALI + CXCR2 antagonist group. We estimated the lung injury by the hematoxylin and eosin staining, the neutrophil extracellular traps (NETs) formations by the immunofluorescence colocalization and enzyme-linked immunosorbent assay (ELISA), and the CXCR2/CXCL2 pathway by the flow cytometry, ELISA, and real-time polymerase chain reaction. We conducted the high-throughput sequencing and enrichment analyses to explore the potential mechanisms. The results showed that the RC decoction pathologically ameliorated the lipopolysaccharide-induced lung injury and inflammatory response but failed to reduce the circulating and lung tissue NETs formation and the blood neutrophil percent. The high-dose RC decoction increased the plasma CXCL2 level, but the RC decoction had no effects on the neutrophilic CXCR2 levels. Under the inhibition of the CXCR2, the middle-dose RC decoction still decreased the lung injury score but as yet had unobvious influence on the NETs formation. Other potential mechanisms of the RC decoction against the ALI involved the pathways of ribosome and coronavirus disease 2019 (COVID-19); the target genes of inflammatory factors, such as Ccl17, Cxcl17, Cd163, Cxcr5, and Il31ra, and lncRNAs; and the regulations of the respiratory cilia. In conclusion, the RC decoction pathologically ameliorated the lipopolysaccharide-induced lung inflammatory injury via upregulating the CXCL2/CXCR2 pathway but could not reduce the circulating or lung tissue NETs formation.
Collapse
|
25
|
Lee HY, You DJ, Taylor-Just AJ, Linder KE, Atkins HM, Ralph LM, De la Cruz G, Bonner JC. Pulmonary exposure of mice to ammonium perfluoro(2-methyl-3-oxahexanoate) (GenX) suppresses the innate immune response to carbon black nanoparticles and stimulates lung cell proliferation. Inhal Toxicol 2022; 34:244-259. [PMID: 35704474 DOI: 10.1080/08958378.2022.2086651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) have been associated with respiratory diseases in humans, yet the mechanisms through which PFAS cause susceptibility to inhaled agents is unknown. Herein, we investigated the effects of ammonium perfluoro(2-methyl-3-oxahexanoate) (GenX), an emerging PFAS, on the pulmonary immune response of mice to carbon black nanoparticles (CBNP). We hypothesized that pulmonary exposure to GenX would increase susceptibility to CBNP through suppression of innate immunity. METHODS Male C57BL/6 mice were exposed to vehicle, 4 mg/kg CBNP, 10 mg/kg GenX, or CBNP and GenX by oropharyngeal aspiration. Bronchoalveolar lavage fluid (BALF) was collected at 1 and 14 days postexposure for cytokines and total protein. Lung tissue was harvested for histopathology, immunohistochemistry (Ki67 and phosphorylated (p)-STAT3), western blotting (p-STAT3 and p-NF-κB), and qRT-PCR for cytokine mRNAs. RESULTS CBNP increased CXCL-1 and neutrophils in BALF at both time points evaluated. However, GenX/CBNP co-exposure reduced CBNP-induced CXCL-1 and neutrophils in BALF. Moreover, CXCL-1, CXCL-2 and IL-1β mRNAs were increased by CBNP in lung tissue but reduced by GenX. Western blotting showed that CBNP induced p-NF-κB in lung tissue, while the GenX/CBNP co-exposed group displayed decreased p-NF-κB. Furthermore, mice exposed to GenX or GenX/CBNP displayed increased numbers of BALF macrophages undergoing mitosis and increased Ki67 immunostaining. This was correlated with increased p-STAT3 by western blotting and immunohistochemistry in lung tissue from mice co-exposed to GenX/CBNP. CONCLUSIONS Pulmonary exposure to GenX suppressed CBNP-induced innate immune response in the lungs of mice yet promoted the proliferation of macrophages and lung epithelial cells.
Collapse
Affiliation(s)
- Ho Young Lee
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Dorothy J You
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Alexia J Taylor-Just
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Keith E Linder
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA
| | - Hannah M Atkins
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Lauren M Ralph
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - James C Bonner
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
26
|
Kim S, Shukla RK, Kim E, Cressman SG, Yu H, Baek A, Choi H, Kim A, Sharma A, Wang Z, Huang CA, Reneau JC, Boyaka PN, Liyanage NPM, Kim S. Comparison of CD3e Antibody and CD3e-sZAP Immunotoxin Treatment in Mice Identifies sZAP as the Main Driver of Vascular Leakage. Biomedicines 2022; 10:1221. [PMID: 35740248 PMCID: PMC9220018 DOI: 10.3390/biomedicines10061221] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 02/03/2023] Open
Abstract
Anti-CD3-epsilon (CD3e) monoclonal antibodies (mAbs) and CD3e immunotoxins (ITs) are promising targeted therapy options for various T-cell disorders. Despite significant advances in mAb and IT engineering, vascular leakage syndrome (VLS) remains a major dose-limiting toxicity for ITs and has been poorly characterized for recent "engineered" mAbs. This study undertakes a direct comparison of non-mitogenic CD3e-mAb (145-2C11 with Fc-silentTM murine IgG1: S-CD3e-mAb) and a new murine-version CD3e-IT (saporin-streptavidin (sZAP) conjugated with S-CD3e-mAb: S-CD3e-IT) and identifies their distinct toxicity profiles in mice. As expected, the two agents showed different modes of action on T cells, with S-CD3e-mAb inducing nearly complete modulation of CD3e on the cell surface, while S-CD3e-IT depleted the cells. S-CD3e-IT significantly increased the infiltration of polymorphonuclear leukocytes (PMNs) into the tissue parenchyma of the spleen and lungs, a sign of increased vascular permeability. By contrast, S-CD3e-mAbs-treated mice showed no notable signs of vascular leakage. Treatment with control ITs (sZAP conjugated with Fc-silent isotype antibodies) induced significant vascular leakage without causing T-cell deaths. These results demonstrate that the toxin portion of S-CD3e-IT, not the CD3e-binding portion (S-CD3e-mAb), is the main driver of vascular leakage, thus clarifying the molecular target for improving safety profiles in CD3e-IT therapy.
Collapse
Affiliation(s)
- Shihyoung Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Rajni Kant Shukla
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Sophie G. Cressman
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Hannah Yu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Alice Baek
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Hyewon Choi
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Alan Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Amit Sharma
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
- Department of Microbial Immunity and Infection, The Ohio State University, Columbus, OH 43210, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Zhirui Wang
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Division of Plastic & Reconstructive Surgery, 12700 East 19th Avenue, Aurora, CO 80045, USA; (Z.W.); (C.A.H.)
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Division of Transplant Surgery, 12700 East 19th Avenue, Aurora, CO 80045, USA
| | - Christene A. Huang
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Division of Plastic & Reconstructive Surgery, 12700 East 19th Avenue, Aurora, CO 80045, USA; (Z.W.); (C.A.H.)
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Division of Transplant Surgery, 12700 East 19th Avenue, Aurora, CO 80045, USA
| | - John C. Reneau
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA;
| | - Prosper N. Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
| | - Namal P. M. Liyanage
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
- Department of Microbial Immunity and Infection, The Ohio State University, Columbus, OH 43210, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Sanggu Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (R.K.S.); (E.K.); (S.G.C.); (H.Y.); (A.B.); (H.C.); (A.K.); (A.S.); (P.N.B.); (N.P.M.L.)
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
27
|
Zenobia C, Darveau RP. Does Oral Endotoxin Contribute to Systemic Inflammation? FRONTIERS IN ORAL HEALTH 2022; 3:911420. [PMID: 35677024 PMCID: PMC9169450 DOI: 10.3389/froh.2022.911420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/29/2022] [Indexed: 12/27/2022] Open
Abstract
The oral microbiome, with a unique emphasis on Porphyromonas gingivalis has been associated with a constellation of inflammatory diseases such as cardiovascular disease, rheumatoid arthritis, Alzheimer's disease, type II diabetes, and non-alcoholic associated fatty liver disease. Periodontal disease has also been shown to induce "leaky gut" leading to metabolic endotoxemia. Several recent studies investigating the habitants of the blood microbiome have found the majority of species appear to be derived from oral and skin bacterial communities in otherwise healthy individuals. Many of the same pathologies associated with perturbations of oral health, such as cardiovascular disease, show alterations to the composition of the blood microbiome as well as circulating neutrophil phenotypes. Gingival inflammation is associated with activated blood neutrophil phenotypes that can exacerbate a distal inflammatory insult which may explain the connection between oral and systemic inflammatory conditions. While in the oral cavity, neutrophils encounter oral microbes that are adept in manipulating neutrophil activity which can re-enter the vasculature thereafter. Endotoxin from oral microbes can differ significantly depending on bacterial community and state of oral health to alter cellular LPS tolerance mechanisms which may contribute to the primed neutrophil phenotype seen in periodontitis and provide a mechanism by which the oral-microbes can affect systemic health outcomes. This review synthesizes the studies between inflammatory diseases and oral health with emphasis on microbiome and corresponding lipopolysaccharides in immune tolerance and activation.
Collapse
Affiliation(s)
| | - Richard P. Darveau
- Departments of Periodontology and Microbiology, University of Washington, Seattle, WA, United States
| |
Collapse
|
28
|
Fan Z, Pitmon E, Wen L, Miller J, Ehinger E, Herro R, Liu W, Chen J, Mikulski Z, Conrad DJ, Marki A, Orecchioni M, Kumari P, Zhu YP, Marcovecchio PM, Hedrick CC, Hodges CA, Rathinam VA, Wang K, Ley K. Bone Marrow Transplantation Rescues Monocyte Recruitment Defect and Improves Cystic Fibrosis in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:745-752. [PMID: 35031577 PMCID: PMC8855460 DOI: 10.4049/jimmunol.1901171] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/19/2021] [Indexed: 02/03/2023]
Abstract
Cystic fibrosis (CF) is an inherited life-threatening disease accompanied by repeated lung infections and multiorgan inflammation that affects tens of thousands of people worldwide. The causative gene, cystic fibrosis transmembrane conductance regulator (CFTR), is mutated in CF patients. CFTR functions in epithelial cells have traditionally been thought to cause the disease symptoms. Recent work has shown an additional defect: monocytes from CF patients show a deficiency in integrin activation and adhesion. Because monocytes play critical roles in controlling infections, defective monocyte function may contribute to CF progression. In this study, we demonstrate that monocytes from CFTRΔF508 mice (CF mice) show defective adhesion under flow. Transplanting CF mice with wild-type (WT) bone marrow after sublethal irradiation replaced most (60-80%) CF monocytes with WT monocytes, significantly improved survival, and reduced inflammation. WT/CF mixed bone marrow chimeras directly demonstrated defective CF monocyte recruitment to the bronchoalveolar lavage and the intestinal lamina propria in vivo. WT mice reconstituted with CF bone marrow also show lethality, suggesting that the CF defect in monocytes is not only necessary but also sufficient to cause disease. We also show that monocyte-specific knockout of CFTR retards weight gains and exacerbates dextran sulfate sodium-induced colitis. Our findings show that providing WT monocytes by bone marrow transfer rescues mortality in CF mice, suggesting that similar approaches may mitigate disease in CF patients.
Collapse
Affiliation(s)
- Zhichao Fan
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Elise Pitmon
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Lai Wen
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
| | - Jacqueline Miller
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
| | - Erik Ehinger
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
| | - Rana Herro
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Wei Liu
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Ju Chen
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Zbigniew Mikulski
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, CA
| | - Douglas J Conrad
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Alex Marki
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
| | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
| | - Puja Kumari
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Yanfang Peipei Zhu
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
| | - Paola M Marcovecchio
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA
| | - Craig A Hodges
- Department of Genetics and Genome Sciences, Cystic Fibrosis Mouse Models Core, School of Medicine, Case Western Reserve University, Cleveland, OH; and
| | - Vijay A Rathinam
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Kepeng Wang
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA;
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| |
Collapse
|
29
|
Brune JE, Chang MY, Altemeier WA, Frevert CW. Type I Interferon Signaling Increases Versican Expression and Synthesis in Lung Stromal Cells During Influenza Infection. J Histochem Cytochem 2021; 69:691-709. [PMID: 34666527 PMCID: PMC8554580 DOI: 10.1369/00221554211054447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Versican, a chondroitin sulfate proteoglycan, is an essential component of the extracellular matrix (ECM) in inflammatory lung disease. Versican's potential as an immunomodulatory molecule makes it a promising therapeutic target for controlling host immune responses in the lungs. To establish changes to versican expression and accumulation during influenza A viral pneumonia, we document the temporal and spatial changes to versican mRNA and protein in concert with pulmonary inflammatory cell infiltration. These studies were performed in the lungs of wild-type C57BL6/J mice on days 3, 6, 9, and 12 post-infection with influenza A virus using immunohistochemistry, in situ hybridization, and quantitative digital pathology. Using duplex in situ hybridization, we demonstrate that type I interferon signaling contributes significantly to versican expression in lung stromal cells. Our findings show that versican is a type I interferon-stimulated gene in pulmonary fibroblasts and pericytes in the context of viral pneumonia. These data also provide a guide for future studies to determine the role of versican in the pulmonary immune response to influenza infection.
Collapse
Affiliation(s)
- Jourdan E. Brune
- Center for Lung Biology, University of Washington, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Mary Y. Chang
- Center for Lung Biology, University of Washington, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - William A. Altemeier
- Center for Lung Biology, University of Washington, Seattle, Washington
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Charles W. Frevert
- Center for Lung Biology, University of Washington, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
30
|
Nintedanib Regulates GRK2 and CXCR2 to Reduce Neutrophil Recruitment in Endotoxin-Induced Lung Injury. Int J Mol Sci 2021; 22:ijms22189898. [PMID: 34576061 PMCID: PMC8464681 DOI: 10.3390/ijms22189898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/25/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
The role of nintedanib, a multiple tyrosine kinase inhibitor, in the treatment of sepsis-induced acute lung injury (ALI) remains unclear. Lipopolysaccharide (LPS), also known as endotoxin, has been used to induce ALI. The goal of this study was to assess the effect of nintedanib in attenuating the histopathological changes of LPS-induced ALI. Nintedanib was administered via oral gavage to male C57BL/6 mice 24 h and 10 min before intratracheal endotoxin instillation. Lung histopathological characteristics, adhesion molecule expression, and the regulatory signaling pathways of neutrophil chemotaxis were analyzed after 24 h. We found that nintedanib significantly reduced histopathological changes and neutrophil recruitment in LPS-induced ALI. The number of neutrophils in bronchoalveolar lavage fluid (BALF) was reduced in nintedanib-treated relative to untreated mice with ALI. Nintedanib mediated the downregulation of the chemotactic response to LPS by reducing the expression of adhesion molecules and the phosphorylated p38:total p38 mitogen-activated protein kinase (MAPK) ratio in the lungs of mice with ALI. Nintedanib also reduced the expression of lymphocyte antigen 6 complex locus G6D (Ly6G) and very late antigen 4 (VLA-4) in BALF neutrophils and mediated the downregulation of chemokine (C-X-C motif) receptor 2 (CXCR2) and upregulation of G protein-coupled receptor kinase 2 (GRK2) activity in peripheral blood neutrophils in mice with LPS-induced ALI. Nintedanib improved the histopathological changes of LPS-induced ALI by reducing neutrophil chemotaxis. These effects were mediated by the inhibition of adhesion molecules via the activation of GRK2 and the inhibition of p38 MAPK and CXCR2.
Collapse
|
31
|
Pleiotropic and Potentially Beneficial Effects of Reactive Oxygen Species on the Intracellular Signaling Pathways in Endothelial Cells. Antioxidants (Basel) 2021; 10:antiox10060904. [PMID: 34205032 PMCID: PMC8229098 DOI: 10.3390/antiox10060904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) are exposed to molecular dioxygen and its derivative reactive oxygen species (ROS). ROS are now well established as important signaling messengers. Excessive production of ROS, however, results in oxidative stress, a significant contributor to the development of numerous diseases. Here, we analyze the experimental data and theoretical concepts concerning positive pro-survival effects of ROS on signaling pathways in endothelial cells (ECs). Our analysis of the available experimental data suggests possible positive roles of ROS in induction of pro-survival pathways, downstream of the Gi-protein-coupled receptors, which mimics insulin signaling and prevention or improvement of the endothelial dysfunction. It is, however, doubtful, whether ROS can contribute to the stabilization of the endothelial barrier.
Collapse
|
32
|
Khawaja A, Bromage DI. The innate immune response in myocarditis. Int J Biochem Cell Biol 2021; 134:105973. [PMID: 33831592 DOI: 10.1016/j.biocel.2021.105973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022]
Abstract
Acute myocarditis is an inflammatory condition of the heart characterised by cellular injury and the influx of leucocytes, including neutrophils, monocytes, macrophages and lymphocytes. While this response is vital for tissue repair, excessive scar deposition and maladaptive ventricular remodelling can result in a legacy of heart failure. It is increasingly recognised as a clinical phenomenon due, in part, to increased availability of cardiac magnetic resonance imaging in patients presenting with chest pain in the absence of significant coronary artery disease. Emerging epidemiological evidence has associated myocarditis with poor outcomes in the context of left ventricular impairment, and even when the left ventricle is preserved outcomes are less benign than once thought. Despite this, our understanding of the contribution of the inflammatory response to the pathophysiology of acute myocarditis lags behind that of acute myocardial infarction, which is the vanguard cardiovascular condition for inflammation research. We recently reviewed monocyte and macrophage phenotype and function in acute myocardial infarction, concluding that their plasticity and heterogeneity might account for conflicting evidence from attempts to target specific leucocyte subpopulations. Here, we revise our understanding of myocardial inflammation, which is predominantly derived from myocardial infarction research, review experimental evidence for the immune response in acute myocarditis, focusing on innate immunity, and discuss potential future directions for immunotherapy research in acute myocarditis.
Collapse
Affiliation(s)
- Abdullah Khawaja
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Daniel I Bromage
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
33
|
Browne JA, NandyMazumdar M, Paranjapye A, Leir SH, Harris A. The Bromodomain Containing 8 (BRD8) transcriptional network in human lung epithelial cells. Mol Cell Endocrinol 2021; 524:111169. [PMID: 33476703 PMCID: PMC8035426 DOI: 10.1016/j.mce.2021.111169] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022]
Abstract
Mechanisms regulating gene expression in the airway epithelium underlie its response to the environment. A network of transcription factors (TFs) and architectural proteins, modulate chromatin accessibility and recruit activating or repressive signals. Bromodomain-containing proteins function as TFs or by engaging methyltransferase or acetyltransferase activity to induce chromatin modifications. Here we investigate the role of Bromodomain Containing 8 (BRD8) in coordinating lung epithelial function. Sites of BRD8 occupancy genome-wide were mapped in human lung epithelial cell lines (Calu-3 and 16HBE14o-). CCCTC-Binding Factor (CTCF) was identified as a predicted co-factor of BRD8, based upon motif over-representation under BRD8 ChIP-seq peaks. Following siRNA-mediated depletion of BRD8, differentially expressed genes with nearby peaks of BRD8 occupancy were subject to gene ontology process enrichment analysis. BRD8 targets are enriched for genes involved in the innate immune response and the cell cycle. Depletion of BRD8 increased the secretion of the antimicrobial peptide beta-defensin 1 and multiple chemokines, and reduced cell proliferation.
Collapse
Affiliation(s)
- James A Browne
- Department of Genetics and Genome Sciences, Cleveland, OH, USA
| | | | | | - Shih-Hsing Leir
- Department of Genetics and Genome Sciences, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ann Harris
- Department of Genetics and Genome Sciences, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
34
|
Saunders RA, Michniacki TF, Hames C, Moale HA, Wilke C, Kuo ME, Nguyen J, Hartlerode AJ, Moore BB, Sekiguchi JM. Elevated inflammatory responses and targeted therapeutic intervention in a preclinical mouse model of ataxia-telangiectasia lung disease. Sci Rep 2021; 11:4268. [PMID: 33608602 PMCID: PMC7895952 DOI: 10.1038/s41598-021-83531-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Ataxia-telangiectasia (A-T) is an autosomal recessive, multisystem disorder characterized by cerebellar degeneration, cancer predisposition, and immune system defects. A major cause of mortality in A-T patients is severe pulmonary disease; however, the underlying causes of the lung complications are poorly understood, and there are currently no curative therapeutic interventions. In this study, we examined the lung phenotypes caused by ATM-deficient immune cells using a mouse model of A-T pulmonary disease. In response to acute lung injury, ATM-deficiency causes decreased survival, reduced blood oxygen saturation, elevated neutrophil recruitment, exaggerated and prolonged inflammatory responses and excessive lung injury compared to controls. We found that ATM null bone marrow adoptively transferred to WT recipients induces similar phenotypes that culminate in impaired lung function. Moreover, we demonstrated that activated ATM-deficient macrophages exhibit significantly elevated production of harmful reactive oxygen and nitrogen species and pro-inflammatory cytokines. These findings indicate that ATM-deficient immune cells play major roles in causing the lung pathologies in A-T. Based on these results, we examined the impact of inhibiting the aberrant inflammatory responses caused by ATM-deficiency with reparixin, a CXCR1/CXCR2 chemokine receptor antagonist. We demonstrated that reparixin treatment reduces neutrophil recruitment, edema and tissue damage in ATM mutant lungs. Thus, our findings indicate that targeted inhibition of CXCR1/CXCR2 attenuates pulmonary phenotypes caused by ATM-deficiency and suggest that this treatment approach represents a viable therapeutic strategy for A-T lung disease.
Collapse
Affiliation(s)
- Rudel A Saunders
- Department of Internal Medicine, University of Michigan, 109 Zina Pitcher Place, 2063 BSRB, Box 2200, Ann Arbor, MI, 48109, USA
| | - Thomas F Michniacki
- Department of Pediatric Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Courtney Hames
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Hilary A Moale
- Department of Internal Medicine, University of Michigan, 109 Zina Pitcher Place, 2063 BSRB, Box 2200, Ann Arbor, MI, 48109, USA
| | - Carol Wilke
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Molly E Kuo
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Johnathan Nguyen
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Bethany B Moore
- Department of Internal Medicine, University of Michigan, 109 Zina Pitcher Place, 2063 BSRB, Box 2200, Ann Arbor, MI, 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - JoAnn M Sekiguchi
- Department of Internal Medicine, University of Michigan, 109 Zina Pitcher Place, 2063 BSRB, Box 2200, Ann Arbor, MI, 48109, USA.
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
35
|
Lin WC, Fessler MB. Regulatory mechanisms of neutrophil migration from the circulation to the airspace. Cell Mol Life Sci 2021; 78:4095-4124. [PMID: 33544156 PMCID: PMC7863617 DOI: 10.1007/s00018-021-03768-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/22/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
The neutrophil, a short-lived effector leukocyte of the innate immune system best known for its proteases and other degradative cargo, has unique, reciprocal physiological interactions with the lung. During health, large numbers of ‘marginated’ neutrophils reside within the pulmonary vasculature, where they patrol the endothelial surface for pathogens and complete their life cycle. Upon respiratory infection, rapid and sustained recruitment of neutrophils through the endothelial barrier, across the extravascular pulmonary interstitium, and again through the respiratory epithelium into the airspace lumen, is required for pathogen killing. Overexuberant neutrophil trafficking to the lung, however, causes bystander tissue injury and underlies several acute and chronic lung diseases. Due in part to the unique architecture of the lung’s capillary network, the neutrophil follows a microanatomic passage into the distal airspace unlike that observed in other end-organs that it infiltrates. Several of the regulatory mechanisms underlying the stepwise recruitment of circulating neutrophils to the infected lung have been defined over the past few decades; however, fundamental questions remain. In this article, we provide an updated review and perspective on emerging roles for the neutrophil in lung biology, on the molecular mechanisms that control the trafficking of neutrophils to the lung, and on past and ongoing efforts to design therapeutics to intervene upon pulmonary neutrophilia in lung disease.
Collapse
Affiliation(s)
- Wan-Chi Lin
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
36
|
Khalil BA, Elemam NM, Maghazachi AA. Chemokines and chemokine receptors during COVID-19 infection. Comput Struct Biotechnol J 2021; 19:976-988. [PMID: 33558827 PMCID: PMC7859556 DOI: 10.1016/j.csbj.2021.01.034] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Chemokines are crucial inflammatory mediators needed during an immune response to clear pathogens. However, their excessive release is the main cause of hyperinflammation. In the recent COVID-19 outbreak, chemokines may be the direct cause of acute respiratory disease syndrome, a major complication leading to death in about 40% of severe cases. Several clinical investigations revealed that chemokines are directly involved in the different stages of SARS-CoV-2 infection. Here, we review the role of chemokines and their receptors in COVID-19 pathogenesis to better understand the disease immunopathology which may aid in developing possible therapeutic targets for the infection.
Collapse
Key Words
- AECs, airway epithelial cells
- AP-1, Activator Protein 1
- ARDS
- ARDS, acute respiratory disease syndrome
- BALF, bronchial alveolar lavage fluid
- CAP, community acquired pneumonia
- COVID-19
- CRS, cytokine releasing syndrome
- Chemokine Receptors
- Chemokines
- DCs, dendritic cells
- ECM, extracellular matrix
- GAGs, glycosaminoglycans
- HIV, human immunodeficiency virus
- HRSV, human respiratory syncytial virus
- IFN, interferon
- IMM, inflammatory monocytes and macrophages
- IP-10, IFN-γ-inducible protein 10
- IRF, interferon regulatory factor
- Immunity
- MERS-CoV, Middle East respiratory syndrome coronavirus
- NETs, neutrophil extracellular traps
- NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells
- NK cells, natural killer cells
- PBMCs, peripheral blood mononuclear cells
- PRR, pattern recognition receptors
- RSV, rous sarcoma virus
- SARS-CoV, severe acute respiratory syndrome coronavirus
- SARS-CoV-2
- TLR, toll like receptor
- TRIF, TIR-domain-containing adapter-inducing interferon-β
Collapse
Affiliation(s)
- Bariaa A. Khalil
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Immuno-Oncology Group, Sharjah Institute for Medical Research (SIMR), Sharjah, United Arab Emirates
| | - Noha Mousaad Elemam
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Immuno-Oncology Group, Sharjah Institute for Medical Research (SIMR), Sharjah, United Arab Emirates
| | - Azzam A. Maghazachi
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Immuno-Oncology Group, Sharjah Institute for Medical Research (SIMR), Sharjah, United Arab Emirates
| |
Collapse
|
37
|
Maxwell AJ, Ding J, You Y, Dong Z, Chehade H, Alvero A, Mor Y, Draghici S, Mor G. Identification of key signaling pathways induced by SARS-CoV2 that underlie thrombosis and vascular injury in COVID-19 patients. J Leukoc Biol 2021; 109:35-47. [PMID: 33242368 PMCID: PMC7753679 DOI: 10.1002/jlb.4covr0920-552rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
The SARS-CoV-2 pandemic has led to hundreds of thousands of deaths and billions of dollars in economic damage. The immune response elicited from this virus is poorly understood. An alarming number of cases have arisen where COVID-19 patients develop complications on top of the symptoms already associated with SARS, such as thrombosis, injuries of vascular system, kidney, and liver, as well as Kawasaki disease. In this review, a bioinformatics approach was used to elucidate the immune response triggered by SARS-CoV-2 infection in primary human lung epithelial and transformed human lung alveolar. Additionally, examined the potential mechanism behind several complications that have been associated with COVID-19 and determined that a specific cytokine storm is leading to excessive neutrophil recruitment. These neutrophils are directly leading to thrombosis, organ damage, and complement activation via neutrophil extracellular trap release.
Collapse
Affiliation(s)
- Anthony J Maxwell
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Jiahui Ding
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Zhong Dong
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Ayesha Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Yechiel Mor
- Department of Internal Medicine Wayne State University, Detroit, Michigan, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, Michigan, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
38
|
Alam MJ, Xie L, Ang C, Fahimi F, Willingham SB, Kueh AJ, Herold MJ, Mackay CR, Robert R. Therapeutic blockade of CXCR2 rapidly clears inflammation in arthritis and atopic dermatitis models: demonstration with surrogate and humanized antibodies. MAbs 2020; 12:1856460. [PMID: 33347356 PMCID: PMC7757791 DOI: 10.1080/19420862.2020.1856460] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Neutrophils are the most abundant effector cells of the innate immune system and represent the first line of defense against infection. However, in many common pathologies, including autoimmune diseases, excessive recruitment and activation of neutrophils can drive a chronic inflammatory response leading to unwanted tissue destruction. Several strategies have been investigated to tackle pathologic neutrophil biology, and thus provide a novel therapy for chronic inflammatory diseases. The chemokine receptor CXCR2 plays a crucial role in regulating neutrophil homeostasis and is a promising pharmaceutical target. In this study, we report the discovery and validation of a humanized anti-human CXCR2 monoclonal antibody. To enable in vivo studies, we developed a surrogate anti-mouse CXCR2 antibody, as well as a human knock-in CXCR2 mouse. When administered in models of atopic dermatitis (AD) and rheumatoid arthritis (RA), the antibodies rapidly clear inflammation. Our findings support further developments of anti-CXCR2 mAb approaches not only for RA and AD, but also for other neutrophil-mediated inflammatory conditions where neutrophils are pathogenic and medical needs are unmet.
Collapse
Affiliation(s)
- Md Jahangir Alam
- Department of Microbiology, Biomedicine Discovery Institute, Monash University , Clayton, Victoria, Australia
| | - Liang Xie
- Department of Microbiology, Biomedicine Discovery Institute, Monash University , Clayton, Victoria, Australia
| | - Caroline Ang
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University , Clayton, Victoria, Australia
| | - Farnaz Fahimi
- Department of Physiology, Biomedicine Discovery Institute, Monash University , Clayton, Victoria, Australia
| | | | - Andrew J Kueh
- Walter and Eliza Hall Institute of Medical Research , Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne , Parkville, VIC, Australia
| | - Marco J Herold
- Walter and Eliza Hall Institute of Medical Research , Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne , Parkville, VIC, Australia
| | - Charles R Mackay
- Department of Microbiology, Biomedicine Discovery Institute, Monash University , Clayton, Victoria, Australia
| | - Remy Robert
- Department of Physiology, Biomedicine Discovery Institute, Monash University , Clayton, Victoria, Australia
| |
Collapse
|
39
|
Vollrath JT, Stoermann P, Becker N, Wutzler S, Hildebrand F, Marzi I, Relja B. Early local neutralization of CC16 in sepsis‑induced ALI following blunt chest trauma leads to delayed mortality without benefitting overall survival. Int J Mol Med 2020; 46:2207-2215. [PMID: 33125155 PMCID: PMC7595659 DOI: 10.3892/ijmm.2020.4767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
Blunt thoracic trauma (TxT) is a common injury pattern in polytraumatized patients. When combined with a secondary trigger, TxT often results in acute lung injury (ALI), which negatively affects outcomes. Recent findings suggest that ALI is caused by both local and systemic inflammatory reactions. Club cell protein (CC)16 is an anti‑inflammatory peptide associated with lung injury following TxT. Recently, the anti‑inflammatory properties of endogenous CC16 in a murine model of TxT with subsequent cecal‑ligation and puncture (CLP) as the secondary hit were demonstrated by our group. The present study aimed to determine whether CC16 neutralization improves survival following 'double‑hit'‑induced ALI. For this purpose, a total of 120 C57BL/6N mice were subjected to TxT, followed by CLP after 24 h. Sham‑operated animals underwent anesthesia without the induction of TxT + CLP. CC16 neutralization was performed by providing a CC16 antibody intratracheally following TxT (early) or following CLP (late). Survival was assessed in 48 animals for 6 days after CLP. Sacrifice was performed 6 or 24 h post‑CLP to evaluate the anti‑inflammatory effect of CC16. The results revealed that CC16 neutralization enhanced pro‑inflammatory CXCL1 levels, thereby confirming the anti‑inflammatory characteristics of CC16 in this model. Early CC16 neutralization immediately following TxT significantly prolonged survival within 60 h; however, the survival rate did not change until 6 days post‑trauma. Late CC16 neutralization did not provide any survival benefits. On the whole, the present study demonstrated that neutralizing CC16 confirmed its anti‑inflammatory potential in this double‑hit ALI model. Early CC16 neutralization prolonged survival within 60 h; however, no survival benefits were observed after 6 days post‑CLP in any group.
Collapse
Affiliation(s)
- Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| | - Philipp Stoermann
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| | - Nils Becker
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| | - Sebastian Wutzler
- Department of Trauma, Hand and Orthopedic Surgery, Helios Horst Schmidt Clinic, D‑65199 Wiesbaden, Germany
| | - Frank Hildebrand
- Department of Trauma Surgery, RWTH University, D‑52062 Aachen, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| |
Collapse
|
40
|
Bashant KR, Aponte AM, Randazzo D, Rezvan Sangsari P, Wood AJ, Bibby JA, West EE, Vassallo A, Manna ZG, Playford MP, Jordan N, Hasni S, Gucek M, Kemper C, Conway Morris A, Morgan NY, Toepfner N, Guck J, Mehta NN, Chilvers ER, Summers C, Kaplan MJ. Proteomic, biomechanical and functional analyses define neutrophil heterogeneity in systemic lupus erythematosus. Ann Rheum Dis 2020; 80:209-218. [PMID: 32988843 DOI: 10.1136/annrheumdis-2020-218338] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Low-density granulocytes (LDGs) are a distinct subset of proinflammatory and vasculopathic neutrophils expanded in systemic lupus erythematosus (SLE). Neutrophil trafficking and immune function are intimately linked to cellular biophysical properties. This study used proteomic, biomechanical and functional analyses to further define neutrophil heterogeneity in the context of SLE. METHODS Proteomic/phosphoproteomic analyses were performed in healthy control (HC) normal density neutrophils (NDNs), SLE NDNs and autologous SLE LDGs. The biophysical properties of these neutrophil subsets were analysed by real-time deformability cytometry and lattice light-sheet microscopy. A two-dimensional endothelial flow system and a three-dimensional microfluidic microvasculature mimetic (MMM) were used to decouple the contributions of cell surface mediators and biophysical properties to neutrophil trafficking, respectively. RESULTS Proteomic and phosphoproteomic differences were detected between HC and SLE neutrophils and between SLE NDNs and LDGs. Increased abundance of type 1 interferon-regulated proteins and differential phosphorylation of proteins associated with cytoskeletal organisation were identified in SLE LDGs relative to SLE NDNs. The cell surface of SLE LDGs was rougher than in SLE and HC NDNs, suggesting membrane perturbances. While SLE LDGs did not display increased binding to endothelial cells in the two-dimensional assay, they were increasingly retained/trapped in the narrow channels of the lung MMM. CONCLUSIONS Modulation of the neutrophil proteome and distinct changes in biophysical properties are observed alongside differences in neutrophil trafficking. SLE LDGs may be increasingly retained in microvasculature networks, which has important pathogenic implications in the context of lupus organ damage and small vessel vasculopathy.
Collapse
Affiliation(s)
- Kathleen R Bashant
- NIAMS, National Institutes of Health, Bethesda, Maryland, USA.,Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Angel M Aponte
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | - Davide Randazzo
- NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Alexander Jt Wood
- Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Jack A Bibby
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | - Erin E West
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | - Arlette Vassallo
- Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Zerai G Manna
- NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Natasha Jordan
- Rheumatology Department, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sarfaraz Hasni
- NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | - Marjan Gucek
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | - Claudia Kemper
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Nicole Y Morgan
- NIBIB, National Institutes of Health, Bethesda, Maryland, USA
| | - Nicole Toepfner
- Department of Pediatrics/Carl Gustav Carus University Hospital, Technical University Dresden, Dresden, Sachsen, Germany
| | - Jochen Guck
- Biological Optomechanics Division, Max Planck Institute for the Science of Light, Erlangen, Bayern, Germany
| | - Nehal N Mehta
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Charlotte Summers
- Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | | |
Collapse
|
41
|
Wu F, Chen X, Zhai L, Wang H, Sun M, Song C, Wang T, Qian Z. CXCR2 antagonist attenuates neutrophil transmigration into brain in a murine model of LPS induced neuroinflammation. Biochem Biophys Res Commun 2020; 529:839-845. [PMID: 32616311 DOI: 10.1016/j.bbrc.2020.05.124] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 05/17/2020] [Indexed: 10/24/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is a devastating neurological complication of sepsis with intolerable high motility. SAE is accompanied with brain vascular injury, endothelial hyperpermeability, and neutrophil infiltration into the brain tissue, key inflammatory processes leading to further brain edema and neuronal cell apoptosis. Recent studies from us and others suggest that the chemokine receptor C-X-C Motif Chemokine Receptor 2 (CXCR2) is crucial for neutrophil recruitment during SAE. Here we use CXCR2 antagonist SB225002 to characterize the role of CXCR2 in brain infiltration of neutrophil in a murine model of SAE. Systemic administration of high-dose LPS (10 mg/kg) induced evident neutrophil infiltration into the cerebral cortex in wild-type mice. However, CXCR2 antagonist SB225002 markedly attenuated neutrophil infiltration into brain. The CXCR2 expression on neutrophils in the peripheral circulation was dramatically downregulated in response to this LPS dose, and endothelial CXCR2 was significantly upregulated, suggesting endothelial but not neutrophil CXCR2 plays a more important role in neutrophil infiltration into brain. Strikingly, although these CXCR2 antagonist SB225002 treated mice displayed reduced neutrophil infiltration, no change in neutrophil rolling and adhesion was observed. Furthermore, we confirmed that CXCR2 agonist CXCL1 induced a marked increase in actin stress fiber synthesis and paracellular gap formation in cultured cerebral endothelial cells, which is attenuated by SB225002. Thus, these results demonstrate a selective role for endothelial CXCR2 to regulate cerebral vascular permeability and neutrophil transmigration in high-dose LPS induced neuroinflammation, and also suggest a therapeutic potential of CXCR2 antagonist SB225002 in SAE.
Collapse
Affiliation(s)
- Fengjiao Wu
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Xiaofen Chen
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Liqian Zhai
- Department of Histology and Embryology, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Hongtao Wang
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Meiqun Sun
- Department of Histology and Embryology, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Chuanwang Song
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Ting Wang
- Department of Internal Medicine, University of Arizona, Phoenix, AZ, 85004, USA.
| | - Zhongqing Qian
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China.
| |
Collapse
|
42
|
Shin JY, Choi JW, Kim DG, Zhou ZQ, Shin YK, Seo JH, Song HJ, Choi BM, Bae GS, Park SJ. Protective effects of Coenzyme Q10 against acute pancreatitis. Int Immunopharmacol 2020; 88:106900. [PMID: 32829089 DOI: 10.1016/j.intimp.2020.106900] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
Acute pancreatitis (AP) refers to inflammation in the pancreas, which may lead to death in severe cases. Coenzyme Q10 (Q10), generally known to generate energy, plays an important role as an anti-oxidant and anti-inflammatory effector. Here, we showed the effect of Q10 on inflammatory response in murine AP model. For this study, we induced AP by injection of cerulein intraperitoneally or pancreatic duct ligation (PDL) in mice. The level of cytokines and digestive enzymes were measured in pancreas, and blood. All pancreatic tissues were excised for investigation such as histological changes, infiltration of immune cells. Administration of Q10 attenuated the severity of AP and its associated pulmonary complication as shown by reduction of acinar cell death, parenchymal edema, inflammatory cell infiltration and alveolar thickening in both cerulein-induced AP and PDL-induced AP. Moreover, reduction of the cytokines such as interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α were observed in pancreas and pancreatic acinar cells by Q10. Furthermore, Q10 reduced the infiltration of immune cells such as monocytes and neutrophils and augmentation of chemokines such as CC chemokine-2 (CCL2) and C-X-C chemokine-2 (CXCL2) in pancreas of AP mice. In addition, Q10 deactivates the phosphorylation of extracellular signal-regulated kinase (ERK) and c-jun NH2-terminal kinase (JNK) in pancreas. In conclusion, these observations suggest that Q10 could attenuate the pancreatic damage and its associated pulmonary complications via inhibition of inflammatory cytokines and inflammatory cell infiltration and that the deactivation of ERK and JNK by Q10 might contribute to the attenuation of AP.
Collapse
Affiliation(s)
- Joon Yeon Shin
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Ji-Won Choi
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea; Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Dong-Gu Kim
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Zi Qi Zhou
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Yong Kook Shin
- Department of Bio Pharmaceutical Industry, Semyung University, Semyeong-ro 65, Jecheon, Chungcheongbuk-do 27136, Republic of Korea
| | - Jae Ho Seo
- Department of Biochemistry, School of Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Ho-Joon Song
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Byung-Min Choi
- Department of Biochemistry, School of Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea.
| | - Gi-Sang Bae
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea; Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea; Research Center of Traditional Korean Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea.
| | - Sung-Joo Park
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea; Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan-daero 460, Iksan, Jeollabuk-do 54538, Republic of Korea.
| |
Collapse
|
43
|
Tischler BY, Tosini NL, Cramer RA, Hohl TM. Platelets are critical for survival and tissue integrity during murine pulmonary Aspergillus fumigatus infection. PLoS Pathog 2020; 16:e1008544. [PMID: 32407390 PMCID: PMC7252636 DOI: 10.1371/journal.ppat.1008544] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/27/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
Beyond their canonical roles in hemostasis and thrombosis, platelets function in the innate immune response by interacting directly with pathogens and by regulating the recruitment and activation of immune effector cells. Thrombocytopenia often coincides with neutropenia in patients with hematologic malignancies and in allogeneic hematopoietic cell transplant recipients, patient groups at high risk for invasive fungal infections. While neutropenia is well established as a major clinical risk factor for invasive fungal infections, the role of platelets in host defense against human fungal pathogens remains understudied. Here, we examined the role of platelets in murine Aspergillus fumigatus infection using two complementary approaches to induce thrombocytopenia without concurrent neutropenia. Thrombocytopenic mice were highly susceptible to A. fumigatus challenge and rapidly succumbed to infection. Although platelets regulated early conidial phagocytosis by neutrophils in a spleen tyrosine kinase (Syk)-dependent manner, platelet-regulated conidial phagocytosis was dispensable for host survival. Instead, our data indicated that platelets primarily function to maintain hemostasis and lung integrity in response to exposed fungal antigens, since thrombocytopenic mice exhibited severe hemorrhage into the airways in response to fungal challenge in the absence of overt angioinvasion. Challenge with swollen, heat-killed, conidia was lethal in thrombocytopenic hosts and could be reversed by platelet transfusion, consistent with the model that fungus-induced inflammation in platelet-depleted mice was sufficient to induce lethal hemorrhage. These data provide new insights into the role of platelets in the anti-Aspergillus host response and expand their role to host defense against filamentous molds. Aspergillus fumigatus is a ubiquitous environmental mold that forms airborne spores, termed conidia. When inhaled by immune compromised individuals, A. fumigatus conidia can germinate into tissue-invasive hyphae and cause invasive aspergillosis, a major cause of infectious morbidity and mortality in patients with leukemia and in bone marrow transplant recipients. Although a low platelet count has been identified as a risk factor for clinical outcomes in patients with invasive aspergillosis, the precise role of platelets in the anti-fungal host response remains poorly understood. Here, we report an essential requirement for platelets in anti-Aspergillus host defence in a mouse model of fungal pneumonia. Although platelets play a role in activating the innate immune system after infection, they are critical for preventing lethal hemorrhage after A. fumigatus challenge. Our findings raise the question as to whether platelets can be used as a basis for therapeutic strategies in vulnerable patient populations.
Collapse
Affiliation(s)
- Benjamin Y. Tischler
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Nicholas L. Tosini
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Robert A. Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Tobias M. Hohl
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
44
|
Stackowicz J, Jönsson F, Reber LL. Mouse Models and Tools for the in vivo Study of Neutrophils. Front Immunol 2020; 10:3130. [PMID: 32038641 PMCID: PMC6985372 DOI: 10.3389/fimmu.2019.03130] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and critical actors of the immune system. Many neutrophil functions and facets of their activity in vivo were revealed by studying genetically modified mice or by tracking fluorescent neutrophils in animals using imaging approaches. Assessing the roles of neutrophils can be challenging, especially when exact molecular pathways are questioned or disease states are interrogated that alter normal neutrophil homeostasis. This review discusses the main in vivo models for the study of neutrophils, their advantages and limitations. The side-by-side comparison underlines the necessity to carefully choose the right model(s) to answer a given scientific question, and exhibit caveats that need to be taken into account when designing experimental procedures. Collectively, this review suggests that at least two models should be employed to legitimately conclude on neutrophil functions.
Collapse
Affiliation(s)
- Julien Stackowicz
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Center for Pathophysiology Toulouse-Purpan (CPTP), UMR 1043, University of Toulouse, INSERM, CNRS, Toulouse, France
| |
Collapse
|
45
|
Fonseca KL, Maceiras AR, Matos R, Simoes-Costa L, Sousa J, Cá B, Barros L, Fernandes AI, Mereiter S, Reis R, Gomes J, Tapia G, Rodríguez-Martínez P, Martín-Céspedes M, Vashakidze S, Gogishvili S, Nikolaishvili K, Appelberg R, Gärtner F, Rodrigues PNS, Vilaplana C, Reis CA, Magalhães A, Saraiva M. Deficiency in the glycosyltransferase Gcnt1 increases susceptibility to tuberculosis through a mechanism involving neutrophils. Mucosal Immunol 2020; 13:836-848. [PMID: 32203062 PMCID: PMC7434595 DOI: 10.1038/s41385-020-0277-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 02/12/2020] [Indexed: 02/04/2023]
Abstract
Modulation of immunity and disease by glycans is increasingly recognized. However, how host glycosylation shapes and is shaped by tuberculosis remains poorly understood. We show that deficiency in the glucosaminyl (N-acetyl) transferase 1 (Gcnt1), a key enzyme for core-2 O-glycans biosynthesis, drives susceptibility to Mycobacterium tuberculosis infection. The increased susceptibility of Gcnt1 deficient mice was characterized by extensive lung immune pathology, mechanistically related to neutrophils. Uninfected Gcnt1 deficient mice presented bone marrow, blood and lung neutrophilia, which further increased with infection. Blood neutrophilia required Gcnt1 deficiency in the hematopoietic compartment, relating with enhanced granulopoiesis, but normal cellular egress from the bone marrow. Interestingly, for the blood neutrophilia to translate into susceptibility to M. tuberculosis infection, Gnct1 deficiency in the stroma was also necessary. Complete Gcnt1 deficiency associated with increased lung expression of the neutrophil chemoattractant CXCL2. Lastly, we demonstrate that the transcript levels of various glycosyltransferase-encoding genes were altered in whole blood of active tuberculosis patients and that sialyl Lewis x, a glycan widely present in human neutrophils, was detected in the lung of tuberculosis patients. Our findings reveal a previously unappreciated link between Gcnt1, neutrophilia and susceptibility to M. tuberculosis infection, uncovering new players balancing the immune response in tuberculosis.
Collapse
Affiliation(s)
- Kaori L. Fonseca
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.418346.c0000 0001 2191 3202Programa de Pós-Graduação Ciência para o Desenvolvimento (PGCD), Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Ana Raquel Maceiras
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Rita Matos
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Luisa Simoes-Costa
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Jeremy Sousa
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Baltazar Cá
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Leandro Barros
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Ana Isabel Fernandes
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Stefan Mereiter
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Ricardo Reis
- CDP-Centro de Diagnóstico Pneumológico do Porto, Porto, Portugal
| | - Joana Gomes
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Gustavo Tapia
- grid.411438.b0000 0004 1767 6330UAB—Pathology Department, Universitat Autònoma de Barcelona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Paula Rodríguez-Martínez
- grid.411438.b0000 0004 1767 6330UAB—Pathology Department, Universitat Autònoma de Barcelona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Montse Martín-Céspedes
- grid.411438.b0000 0004 1767 6330UAB—Pathology Department, Universitat Autònoma de Barcelona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Sergo Vashakidze
- National Center for Tuberculosis and Lung Diseases (NCTLD), Tbilisi, Georgia
| | - Shota Gogishvili
- National Center for Tuberculosis and Lung Diseases (NCTLD), Tbilisi, Georgia
| | - Keti Nikolaishvili
- National Center for Tuberculosis and Lung Diseases (NCTLD), Tbilisi, Georgia
| | - Rui Appelberg
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Fátima Gärtner
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Pedro N. S. Rodrigues
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Cristina Vilaplana
- UAB—Experimental Tuberculosis Unit. Universitat Autònoma de Barcelona, CIBER Enfermedades Respiratorias. Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - Celso A. Reis
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226FMUP—Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Ana Magalhães
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Margarida Saraiva
- grid.5808.50000 0001 1503 7226i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
46
|
Radermecker C, Sabatel C, Vanwinge C, Ruscitti C, Maréchal P, Perin F, Schyns J, Rocks N, Toussaint M, Cataldo D, Johnston SL, Bureau F, Marichal T. Locally instructed CXCR4 hi neutrophils trigger environment-driven allergic asthma through the release of neutrophil extracellular traps. Nat Immunol 2019; 20:1444-1455. [PMID: 31591573 PMCID: PMC6859073 DOI: 10.1038/s41590-019-0496-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
Low exposure to microbial products, respiratory viral infections and air pollution is a major risk factor for allergic asthma, yet the mechanistic links between such conditions and host susceptibility to type 2 allergic disorders remain unclear. Through the use of single-cell RNA sequencing (scRNA-seq), we characterized lung neutrophils in mice exposed to a pro-allergic, low dose of lipopolysaccharides (LPSlo) or a protective, high dose of LPS (LPShi) before exposure to house dust mite (HDM). Unlike exposure to LPShi, exposure to LPSlo instructed recruited neutrophils to upregulate the expression of the chemokine receptor CXCR4 and to release neutrophil extracellular traps (NETs). The LPSlo-induced neutrophils and NETs potentiated the uptake of HDM by CD11b+Ly-6C+ dendritic cells (DCs) and type 2 allergic airway inflammation in response to HDM. NETs derived from CXCR4hi neutrophils were also needed to mediate allergic asthma triggered by infection with influenza virus or exposure to ozone. Our study indicates that apparently unrelated environmental risk factors can shape recruited lung neutrophils to promote the initiation of allergic asthma.
Collapse
Affiliation(s)
- Coraline Radermecker
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium.,Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Catherine Sabatel
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Céline Vanwinge
- Laboratory of Tumor and Development Biology, GIGA Institute, Liege University, Liege, Belgium
| | - Cecilia Ruscitti
- Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Pauline Maréchal
- Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Fabienne Perin
- Laboratory of Tumor and Development Biology, GIGA Institute, Liege University, Liege, Belgium
| | - Joey Schyns
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium.,Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Natacha Rocks
- Laboratory of Tumor and Development Biology, GIGA Institute, Liege University, Liege, Belgium
| | - Marie Toussaint
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK.,Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Didier Cataldo
- Laboratory of Tumor and Development Biology, GIGA Institute, Liege University, Liege, Belgium
| | - Sebastian L Johnston
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK.,Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.,Imperial College Healthcare National Health Service Trust, London, UK
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium. .,Faculty of Veterinary Medicine, Liege University, Liege, Belgium. .,Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium.
| | - Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium. .,Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium. .,Faculty of Veterinary Medicine, Liege University, Liege, Belgium. .,Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium.
| |
Collapse
|
47
|
Oliveira AC, Fu C, Lu Y, Williams MA, Pi L, Brantly ML, Ventetuolo CE, Raizada MK, Mehrad B, Scott EW, Bryant AJ. Chemokine signaling axis between endothelial and myeloid cells regulates development of pulmonary hypertension associated with pulmonary fibrosis and hypoxia. Am J Physiol Lung Cell Mol Physiol 2019; 317:L434-L444. [PMID: 31364370 PMCID: PMC6842914 DOI: 10.1152/ajplung.00156.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/11/2019] [Accepted: 07/24/2019] [Indexed: 12/14/2022] Open
Abstract
Pulmonary hypertension complicates the care of many patients with chronic lung diseases (defined as Group 3 pulmonary hypertension), yet the mechanisms that mediate the development of pulmonary vascular disease are not clearly defined. Despite being the most prevalent form of pulmonary hypertension, to date there is no approved treatment for patients with disease. Myeloid-derived suppressor cells (MDSCs) and endothelial cells in the lung express the chemokine receptor CXCR2, implicated in the evolution of both neoplastic and pulmonary vascular remodeling. However, precise cellular contribution to lung disease is unknown. Therefore, we used mice with tissue-specific deletion of CXCR2 to investigate the role of this receptor in Group 3 pulmonary hypertension. Deletion of CXCR2 in myeloid cells attenuated the recruitment of polymorphonuclear MDSCs to the lungs, inhibited vascular remodeling, and protected against pulmonary hypertension. Conversely, loss of CXCR2 in endothelial cells resulted in worsened vascular remodeling, associated with increased MDSC migratory capacity attributable to increased ligand availability, consistent with analyzed patient sample data. Taken together, these data suggest that CXCR2 regulates MDSC activation, informing potential therapeutic application of MDSC-targeted treatments.
Collapse
Affiliation(s)
- Aline C Oliveira
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida
| | - Chunhua Fu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Yuanqing Lu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Mason A Williams
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Liya Pi
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Mark L Brantly
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Edward W Scott
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida
| | - Andrew J Bryant
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida
| |
Collapse
|
48
|
Margraf A, Ley K, Zarbock A. Neutrophil Recruitment: From Model Systems to Tissue-Specific Patterns. Trends Immunol 2019; 40:613-634. [PMID: 31175062 PMCID: PMC6745447 DOI: 10.1016/j.it.2019.04.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/22/2019] [Accepted: 04/25/2019] [Indexed: 12/11/2022]
Abstract
Neutrophil recruitment is not only vital for host defense, but also relevant in pathological inflammatory reactions, such as sepsis. Model systems have been established to examine different steps of the leukocyte recruitment cascade in vivo and in vitro under inflammatory conditions. Recently, tissue-specific recruitment patterns have come into focus, requiring modification of formerly generalized assumptions. Here, we summarize existing models of neutrophil recruitment and highlight recent discoveries in organ-specific recruitment patterns. New techniques show that previously stated assumptions of integrin activation and tissue invasion may need revision. Similarly, neutrophil recruitment to specific organs can rely on different organ properties, adhesion molecules, and chemokines. To advance our understanding of neutrophil recruitment, organ-specific intravital microscopy methods are needed.
Collapse
Affiliation(s)
- Andreas Margraf
- Department of Anesthesiology, Intensive Care Therapy and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care Therapy and Pain Medicine, University Hospital Muenster, Muenster, Germany.
| |
Collapse
|
49
|
Chemokines in COPD: From Implication to Therapeutic Use. Int J Mol Sci 2019; 20:ijms20112785. [PMID: 31174392 PMCID: PMC6600384 DOI: 10.3390/ijms20112785] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
: Chronic Obstructive Pulmonary Disease (COPD) represents the 3rd leading cause of death in the world. The underlying pathophysiological mechanisms have been the focus of extensive research in the past. The lung has a complex architecture, where structural cells interact continuously with immune cells that infiltrate into the pulmonary tissue. Both types of cells express chemokines and chemokine receptors, making them sensitive to modifications of concentration gradients. Cigarette smoke exposure and recurrent exacerbations, directly and indirectly, impact the expression of chemokines and chemokine receptors. Here, we provide an overview of the evidence regarding chemokines involvement in COPD, and we hypothesize that a dysregulation of this tightly regulated system is critical in COPD evolution, both at a stable state and during exacerbations. Targeting chemokines and chemokine receptors could be highly attractive as a mean to control both chronic inflammation and bronchial remodeling. We present a special focus on the CXCL8-CXCR1/2, CXCL9/10/11-CXCR3, CCL2-CCR2, and CXCL12-CXCR4 axes that seem particularly involved in the disease pathophysiology.
Collapse
|
50
|
Karpova D, Rettig MP, Ritchey J, Cancilla D, Christ S, Gehrs L, Chendamarai E, Evbuomwan MO, Holt M, Zhang J, Abou-Ezzi G, Celik H, Wiercinska E, Yang W, Gao F, Eissenberg LG, Heier RF, Arnett SD, Meyers MJ, Prinsen MJ, Griggs DW, Trumpp A, Ruminski PG, Morrow DM, Bonig HB, Link DC, DiPersio JF. Targeting VLA4 integrin and CXCR2 mobilizes serially repopulating hematopoietic stem cells. J Clin Invest 2019; 129:2745-2759. [PMID: 31085833 DOI: 10.1172/jci124738] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mobilized peripheral blood has become the primary source of hematopoietic stem and progenitor cells (HSPCs) for stem cell transplantation, with a five-day course of granulocyte colony stimulating factor (G-CSF) as the most common regimen used for HSPC mobilization. The CXCR4 inhibitor, plerixafor, is a more rapid mobilizer, yet not potent enough when used as a single agent, thus emphasizing the need for faster acting agents with more predictable mobilization responses and fewer side effects. We sought to improve hematopoietic stem cell transplantation by developing a new mobilization strategy in mice through combined targeting of the chemokine receptor CXCR2 and the very late antigen 4 (VLA4) integrin. Rapid and synergistic mobilization of HSPCs along with an enhanced recruitment of true HSCs was achieved when a CXCR2 agonist was co-administered in conjunction with a VLA4 inhibitor. Mechanistic studies revealed involvement of CXCR2 expressed on BM stroma in addition to stimulation of the receptor on granulocytes in the regulation of HSPC localization and egress. Given the rapid kinetics and potency of HSPC mobilization provided by the VLA4 inhibitor and CXCR2 agonist combination in mice compared to currently approved HSPC mobilization methods, it represents an exciting potential strategy for clinical development in the future.
Collapse
Affiliation(s)
- Darja Karpova
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Michael P Rettig
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Julie Ritchey
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel Cancilla
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stephanie Christ
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Leah Gehrs
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ezhilarasi Chendamarai
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Moses O Evbuomwan
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA
| | - Matthew Holt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jingzhu Zhang
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Grazia Abou-Ezzi
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hamza Celik
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Eliza Wiercinska
- German Red Cross Blood Service and Institute for Transfusion Medicine and Immunohematology of the Goethe University, Frankfurt, Germany
| | - Wei Yang
- Genome Technology Access Center, Washington University, St. Louis, Missouri, USA
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Linda G Eissenberg
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Richard F Heier
- Center for World Health and Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Stacy D Arnett
- Center for World Health and Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Marvin J Meyers
- Center for World Health and Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Michael J Prinsen
- Center for World Health and Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - David W Griggs
- Center for World Health and Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Peter G Ruminski
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Center for World Health and Medicine, Saint Louis University, St. Louis, Missouri, USA
| | | | - Halvard B Bonig
- German Red Cross Blood Service and Institute for Transfusion Medicine and Immunohematology of the Goethe University, Frankfurt, Germany.,University of Washington, Department of Medicine/Hematology, Seattle, Washington, USA
| | - Daniel C Link
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|