1
|
Aktay-Cetin Ö, Pullamsetti SS, Herold S, Savai R. Lung tumor immunity: redirecting macrophages through infection-induced inflammation. Trends Immunol 2025:S1471-4906(25)00096-1. [PMID: 40382244 DOI: 10.1016/j.it.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
Macrophages play a central role in maintaining tissue homeostasis and in surveillance against pathogens and disease. In the lung, they can adopt either proinflammatory or anti-inflammatory states depending on the nature of the stimulus. As the predominant immune cells in both the lung tumor microenvironment and in sites of lung infection, the functional plasticity of macrophages makes them key players in determining disease outcome. Accurately defining their inflammatory profiles offers an opportunity to reprogram infection-associated macrophages towards enhanced tumor-killing phenotypes. This review explores how acute inflammation can drive macrophage-mediated antitumor immunity and highlights key molecules and signaling pathways that may be leveraged to therapeutically modulate macrophage function.
Collapse
Affiliation(s)
- Öznur Aktay-Cetin
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Soni Savai Pullamsetti
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany; Department of Internal Medicine II, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Susanne Herold
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Department of Internal Medicine V, German Center for Lung Research (DZL), German Center for Infection Research (DZIF), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany; Department of Internal Medicine II, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.
| |
Collapse
|
2
|
von Ehr A, Steenbuck ID, Häfele C, Remmersmann F, Vico TA, Ehlert C, Lindner D, Wolf D, Tholen S, Schilling O, Czerny M, Westermann D, Hilgendorf I. Experimental evidence on colchicine's mode of action in human carotid artery plaques. Atherosclerosis 2025; 406:119239. [PMID: 40381496 DOI: 10.1016/j.atherosclerosis.2025.119239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/27/2025] [Accepted: 05/03/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND AND AIMS Atherosclerosis, driven by inflammation, is a leading cause of cardiovascular events. Recent clinical trials have highlighted the therapeutic potential of anti-inflammatory treatments. Consequently, colchicine is being recommended for secondary prevention in current guidelines, although the drug's mechanistic actions are not fully understood. METHODS To this end, we conducted a multiomic investigation of colchicine's effect on human carotid plaques. Sections from endarterectomy specimens were exposed to colchicine at concentrations of 2 ng/ml and 10 ng/ml ex vivo for 24 h and compared to untreated segments of the same plaque. Gene expression changes were analyzed by bulk RNA sequencing, and plaque secretomes underwent mass spectrometry for proteomic analysis. In situ cell proliferation was assessed by histology. RESULTS Our data indicate, that colchicine suppresses neutrophil and platelet degranulation and activation, collagen degradation and atheromatous plaque macrophage proliferation in a dose-dependent manner in human plaques, while stimulating myofibroblast activation. Unexpectedly, interleukine (IL)-1beta release from colchicine treated plaques was not reduced. These results indicate that the inflammasome may not be the predominant target of low-dose colchicine in human carotid artery plaques. CONCLUSION Our study identifies multifactorial pathways through which colchicine, the first cardiovascular guideline-recommended anti-inflammatory drug, predominantly acts on human atherosclerotic lesions beyond the inflammasome. Targeting neutrophil and platelet degranulation, collagen degradation and macrophage proliferation, selectively, may provide substantial therapeutic benefit in atherosclerotic cardiovascular disease without colchicine's undesired side effects.
Collapse
Affiliation(s)
- Alexander von Ehr
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Ines Derya Steenbuck
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Häfele
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix Remmersmann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tamara A Vico
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carolin Ehlert
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Tholen
- Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
3
|
Liu Y, Li B, Zhang J, Zhao B, Chen L, Chen B. Temporal Proteome Profiling of Anterior Cruciate Ligament Tear Remnants: Secretory Proteins in the Acute Phase Potentially Promote Tissue Repair. J Proteome Res 2025; 24:2302-2313. [PMID: 40192091 DOI: 10.1021/acs.jproteome.4c00792] [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: 05/03/2025]
Abstract
Previous studies reported that preserving the anterior cruciate ligament (ACL) remnants following ACL rupture during reconstruction surgery could promote graft healing. However, the temporal proteomic expression of ACL remnants remains unclear. Based on previous reports, we have redefined the initial 6 weeks following ACL rupture as the acute phase and the subsequent 6 weeks to 6 months as the subacute phase. High-throughput proteomic sequencing on ACL remnants from the two groups was utilized. Our study unveiled a total of 381 differential expression proteins (DEPs), with 136 upregulated and 245 downregulated proteins in the acute phase. By intersecting these findings with secretory protein databases, we identified 26 upregulated secretory proteins and 19 downregulated in the acute phase. The upregulation of MMP9 and VTN and the downregulation of COL1A1 and POSTN in the acute phase were further confirmed by immunohistochemistry. These findings suggest that the elevated expression of secretory proteins in the acute phase may play crucial roles in promoting cell proliferation, angiogenesis, and tissue repair of the graft. This study not only enhances our understanding of repair mechanisms in ACL remnant preservation but also provides a theoretical foundation for guiding rational clinical surgical timing.
Collapse
Affiliation(s)
- Yiming Liu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, China
| | - Bin Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, China
| | - Jun Zhang
- Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China
| | - Boming Zhao
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shannxi 710000, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, China
| | - Biao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, China
| |
Collapse
|
4
|
Boyd DF, Jordan SV, Balachandran S. ZBP1-driven cell death in severe influenza. Trends Microbiol 2025; 33:521-532. [PMID: 39809680 DOI: 10.1016/j.tim.2024.12.008] [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: 10/16/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Influenza A virus (IAV) infections can cause life-threatening illness in humans. The severity of disease is directly linked to virus replication in the alveoli of the lower respiratory tract. In particular, the lytic death of infected alveolar epithelial cells (AECs) is a major driver of influenza severity. Recent studies have begun to define the molecular mechanisms by which IAV triggers lytic cell death. Z-form nucleic-acid-binding protein 1 (ZBP1) senses replicating IAV and drives programmed cell death (PCD) in infected cells, including apoptosis and necroptosis in AECs and pyroptosis in myeloid cells. Necroptosis and pyroptosis, both lytic forms of death, contribute to pathogenesis during severe infections. Pharmacological blockade of necroptosis shows strong therapeutic potential in mouse models of lethal influenza. We suggest that targeting ZBP1-initiated necroinflammatory cell lysis, either alone or in combination antiviral drugs, will provide clinical benefit in severe influenza.
Collapse
Affiliation(s)
- David F Boyd
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA.
| | - Summer Vaughn Jordan
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA
| | | |
Collapse
|
5
|
Iliakis CS, Crotta S, Wack A. The Interplay Between Innate Immunity and Nonimmune Cells in Lung Damage, Inflammation, and Repair. Annu Rev Immunol 2025; 43:395-422. [PMID: 40036704 DOI: 10.1146/annurev-immunol-082323-031852] [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: 03/06/2025]
Abstract
As the site of gas exchange, the lung is critical for organismal survival. It is also subject to continual environmental insults inflicted by pathogens, particles, and toxins. Sometimes, these insults result in structural damage and the initiation of an innate immune response. Operating in parallel, the immune response aims to eliminate the threat, while the repair process ensures continual physiological function of the lung. The inflammatory response and repair processes are thus inextricably linked in time and space but are often studied in isolation. Here, we review the interplay of innate immune cells and nonimmune cells during lung insult and repair. We highlight how cellular cross talk can fine-tune the circuitry of the immune response, how innate immune cells can facilitate or antagonize proper organ repair, and the prolonged changes to lung immunity and physiology that can result from acute immune responses and repair processes.
Collapse
Affiliation(s)
- Chrysante S Iliakis
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Stefania Crotta
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
6
|
Wang G, Yi Q, Hu B, Peng M, Fu T, Huang E. The regulatory role of BMP9 on lipopolysaccharide-induced matrix metalloproteinases in human stem cells from the apical papilla. Arch Oral Biol 2025; 171:106154. [PMID: 39689436 DOI: 10.1016/j.archoralbio.2024.106154] [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: 08/28/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVE The aim of this study was to investigate changes in the expression of members of the matrix metalloproteinases (MMPs) family in response to lipopolysaccharide (LPS) stimulation and to investigate the regulatory effects of BMP9 on MMPs. DESIGN The extracted human stem cells from the apical papilla (hSCAPs) were identified by flow cytometry, Alizarin Red staining, Oil Red O staining, and alkaline phosphatase staining. The appropriate LPS concentration for inducing inflammation in hSCAPs was determined using real-time quantitative PCR (RT-qPCR) and Cell Counting Kit-8 (CCK-8) assays. MMP expression in LPS-stimulated hSCAPs was evaluated by RT-qPCR. BMP9 was overexpressed in hSCAPs via recombinant adenovirus, and its effects on MMP regulation were assessed using RT-qPCR, Western blotting, and ELISA. All experiments were conducted in vitro. Data were analyzed by one-way ANOVA followed by Tukey's post-hoc comparison, with p < 0.05 considered significant. RESULTS The results showed that on the 3rd and 5th day after LPS stimulation, the expression of MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, MMP-10, MMP-12, and MMP-13 in hSCAPs was significantly upregulated. On the 7th day after LPS induction, the expression of MMP-3, MMP-8, MMP-9 and MMP-13 in hSCAPs was significantly increased. When BMP9 was overexpressed in hSCAPs, the elevated MMPs were inhibited to varying degrees. CONCLUSIONS In the LPS-induced inflammatory environment, certain MMPs are elevated in hSCAP, with MMP-13 being the most significant. Overexpression of BMP9 can significantly inhibit elevated MMPs, suggesting that BMP9 may provide new insights and targets for the treatment of periapical periodontitis.
Collapse
Affiliation(s)
- Gang Wang
- Chongqing Key Laboratory of Oral Diseases, Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Qin Yi
- Chongqing Key Laboratory of Oral Diseases, Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Butu Hu
- Chongqing Key Laboratory of Oral Diseases, Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Mengtian Peng
- Chongqing Key Laboratory of Oral Diseases, Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Tiwei Fu
- Chongqing Key Laboratory of Oral Diseases, Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Enyi Huang
- Chongqing Key Laboratory of Oral Diseases, Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
7
|
Crowley LE, Stockley RA, Thickett DR, Dosanjh D, Scott A, Parekh D. Neutrophil dynamics in pulmonary fibrosis: pathophysiological and therapeutic perspectives. Eur Respir Rev 2024; 33:240139. [PMID: 39603661 PMCID: PMC11600124 DOI: 10.1183/16000617.0139-2024] [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: 06/18/2024] [Accepted: 08/23/2024] [Indexed: 11/29/2024] Open
Abstract
The shared pathobiological mechanisms driving progressive fibrosis in interstitial lung diseases (ILDs) remain unclear. Neutrophils, the most common immune cells in the human body, contain an extensive array of proteinases that are important for cell function, including tissue repair and remodelling. Increasing observational studies have reported elevated neutrophil counts in the respiratory tract and circulation of patients with ILD and suggest a role as a biomarker of disease severity. Neutrophils and their contents (including the formation of neutrophil extracellular traps (NETs)) are present in fibrotic lung tissue. Proteinases and NETs may drive fibrogenesis in animal and in vitro models and may impact transforming growth factor-β1 activation. However, the effect of neutrophil action, whether reparative or pathologically destructive to the delicate lung architecture, has yet to be determined. This review aims to summarise the current literature surrounding the potential role of the neutrophil as a biomarker and contributor to the pathogenesis of ILD. There is currently a paucity of treatment options in ILD driven by the knowledge gap underlying the overall disease mechanisms. This review concludes that neutrophils warrant further evaluation as manipulation of recruitment and function could provide a novel and much needed therapeutic strategy.
Collapse
Affiliation(s)
- Louise Elizabeth Crowley
- Birmingham Acute Care Research Group, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, UK
- Respiratory Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Robert Andrew Stockley
- Birmingham Acute Care Research Group, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, UK
| | - David Richard Thickett
- Birmingham Acute Care Research Group, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, UK
- Respiratory Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Davinder Dosanjh
- Birmingham Acute Care Research Group, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, UK
| | - Aaron Scott
- Birmingham Acute Care Research Group, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, UK
- Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Joint senior authors
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, UK
- Respiratory Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Joint senior authors
| |
Collapse
|
8
|
Noda K, Atale N, Al‐Zahrani A, Furukawa M, Snyder ME, Ren X, Sanchez PG. Heparanase-induced endothelial glycocalyx degradation exacerbates lung ischemia/reperfusion injury in male mice. Physiol Rep 2024; 12:e70113. [PMID: 39448392 PMCID: PMC11502304 DOI: 10.14814/phy2.70113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
The endothelial glycocalyx (eGC) is a carbohydrate-rich layer on the vascular endothelium, and its damage can lead to endothelial and organ dysfunction. Heparanase (HPSE) degrades the eGC in response to cellular stress, but its role in organ dysfunction remains unclear. This study investigates HPSE's role in lung ischemia-reperfusion (I/R) injury. A left lung hilar occlusion model was used in B6 wildtype (WT) and HPSE genetic knockout (-/-) mice to induce I/R injury in vivo. The left lungs were ischemic for 1 h followed by reperfusion for 4 h prior to investigations of lung function and eGC status. Data were compared between uninjured lungs and I/R-injured lungs in WT and HPSE-/- mice. WT lungs showed significant functional impairment after I/R injury, whereas HPSE-/- lungs did not. Inhibition or knockout of HPSE prevented eGC damage, inflammation, and cellular migration after I/R injury by reducing matrix metalloproteinase activities. HPSE-/- mice exhibited compensatory regulation of related gene expressions. HPSE facilitates eGC degradation leading to inflammation and impaired lung function after I/R injury. HPSE may be a therapeutic target to attenuate graft damage in lung transplantation.
Collapse
Affiliation(s)
- Kentaro Noda
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Neha Atale
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Amer Al‐Zahrani
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Masashi Furukawa
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Mark E. Snyder
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Xi Ren
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Pablo G. Sanchez
- Section of Thoracic Surgery, Department of SurgeryUniversity of ChicagoChicagoIllinoisUSA
| |
Collapse
|
9
|
Schaaf KR, Landstreet SR, Putz ND, Gonski SK, Lin J, Buggs CJ, Gibson D, Langouët-Astrié CJ, Jetter CS, Negretti NM, Sucre JMS, Schmidt EP, Ware LB, Bastarache JA, Shaver CM. Matrix metalloproteinases mediate influenza A-associated shedding of the alveolar epithelial glycocalyx. PLoS One 2024; 19:e0308648. [PMID: 39312544 PMCID: PMC11419339 DOI: 10.1371/journal.pone.0308648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/28/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND The alveolar epithelium is protected by a heparan sulfate-rich, glycosaminoglycan layer called the epithelial glycocalyx. It is cleaved in patients with acute respiratory distress syndrome (ARDS) and in murine models of influenza A (IAV) infection, shedding fragments into the airspace from the cell surface. Glycocalyx shedding results in increased permeability of the alveolar-capillary barrier, amplifying acute lung injury. The mechanisms underlying alveolar epithelial glycocalyx shedding in IAV infection are unknown. We hypothesized that induction of host sheddases such as matrix metalloproteinases (MMPs) during IAV infection results in glycocalyx shedding and increased lung injury. MATERIALS AND METHODS We measured glycocalyx shedding and lung injury during IAV infection with and without treatment with the pan-MMP inhibitor Ilomastat (ILO) and in an MMP-7 knock out (MMP-7KO) mouse. C57BL/6 or MMP-7KO male and female mice were given IAV A/PR/8/34 (H1N1) at 30,000 PFU/mouse or PBS intratracheally. For some experiments, C56BL/6 mice were infected in the presence of ILO (100mg/kg) or vehicle given daily by IP injection. Bronchoalveolar lavage (BAL) and lung tissue were collected on day 1, 3, and 7 for analysis of glycocalyx shedding (BAL Syndecan-1) and lung injury (histology, BAL protein, BAL cytokines, BAL immune cell infiltrates, BAL RAGE). Expression and localization of the sheddase MMP-7 and its inhibitor TIMP-1 was examined by RNAScope. For in vitro experiments, MLE-12 mouse lung epithelial cells were cultured and treated with active or heat-inactivated heparinase (2.5 U/mL) prior to infection with IAV (MOI 1) and viral load and MMP-7 and TIMP-1 expression analyzed. RESULTS IAV infection caused shedding of the epithelial glycocalyx into the BAL. Inhibition of MMPs with ILO reduced glycocalyx shedding by 36% (p = 0.0051) and reduced lung epithelial injury by 40% (p = 0.0404). ILO also reduced viral load by 68% (p = 0.027), despite having no significant effect on lung cytokine production. Both MMP-7 and its inhibitor TIMP-1 were upregulated in IAV infected mice: MMP-7 colocalized with IAV, while TIMP-1 was limited to cells adjacent to infection. However, MMP-7KO mice had similar glycocalyx shedding, epithelial injury, and viral load compared to WT littermates, suggesting redundancy in MMP sheddase function in the lung. In vitro, heparinase treatment before infection led to a 52% increase in viral load (p = 0.0038) without altering MMP-7 or TIMP-1 protein levels. CONCLUSIONS Glycocalyx shedding and MMPs play key roles in IAV-induced epithelial injury, with significant impact on IAV viral load. Further studies are needed to understand which specific MMPs regulate lung epithelial glycocalyx shedding.
Collapse
Affiliation(s)
- Kaitlyn R. Schaaf
- Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Stuart R. Landstreet
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Nathan D. Putz
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Samantha K. Gonski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jason Lin
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Charity J. Buggs
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Dustin Gibson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Christophe J. Langouët-Astrié
- Division of Pulmonary and Critical Care, Department of Medicine, University of Colorado Anschutz, Denver, Colorado, United States of America
| | - Christopher S. Jetter
- Department of Neonatology, Monroe Caroll Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
| | - Nicolas M. Negretti
- Department of Neonatology, Monroe Caroll Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
| | - Jennifer M. S. Sucre
- Department of Neonatology, Monroe Caroll Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
| | - Eric P. Schmidt
- Division of Pulmonary and Critical Care, Department of Medicine, University of Colorado Anschutz, Denver, Colorado, United States of America
- Division of Pulmonary and Critical Care, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Lorraine B. Ware
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Julie A. Bastarache
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Development Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ciara M. Shaver
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
10
|
Varshney A, Ram VS, Kumar P. Beyond troponins: Emerging diagnostic significance of novel markers in NSTEMI. Glob Cardiol Sci Pract 2024; 2024:e202433. [PMID: 39351477 PMCID: PMC11439417 DOI: 10.21542/gcsp.2024.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/23/2024] [Indexed: 10/04/2024] Open
Abstract
Objective: This study aims to comprehensively analyze a multiple-marker panel consisting of 55 morphofunctional and biochemical markers in 123 patients diagnosed with non-ST-segment elevation myocardial infarction (NSTEMI). The goal of this study was to identify novel pathogenetic landmarks and diagnostic predictors associated with NSTEMI. Methods: The study includes 123 patients diagnosed with NSTEMI based on ESC Guidelines criteria. Clinical characteristics, morphofunctional markers, and serum levels of 53 biochemical markers related to inflammation, oxidative stress, endothelial dysfunction, cellular injury, hemostasis, and myocardial remodeling were assessed. A control group of 47 healthy individuals was included for comparison. Results: NSTEMI patients exhibited an activated inflammatory status, oxidative stress, and endothelial dysfunction. Notable increases in inflammation markers, alterations in adipokines, and changes in oxidative stress markers were observed. Endothelial dysfunction markers indicated vascular remodeling and dysfunction. Cellular injury markers, including cMyBP-C, suggested myocardial necrotic injury. Hemostasis markers showed impaired anticoagulant systems, and ECM remodeling markers indicated increased matrix metalloproteinases. Conclusion: The multiple-marker panel provides insights into novel pathogenetic entities associated with NSTEMI. Markers such as MPO, MMP-8, E-selectin, PhA2, Ang 2, FE, MF, and cMyBP-C demonstrate potential diagnostic and prognostic value. This comprehensive analysis enhances our understanding of NSTEMI pathogenesis and offers potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Amit Varshney
- Department of General Medicine, Noida International Institute of Medical Sciences, Gautam Budh Nagar, Uttar Pradesh, India
| | - Vidya S. Ram
- Department of Medicine, Uttar Pradesh University of Medical Sciences, Saifai, Etawah, Uttar Pradesh, India
| | - Pankaj Kumar
- Department of Medicine, Uttar Pradesh University of Medical Sciences, Saifai, Etawah, Uttar Pradesh, India
| |
Collapse
|
11
|
Cheetham CJ, McKelvey MC, McAuley DF, Taggart CC. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. Int J Mol Sci 2024; 25:5492. [PMID: 38791530 PMCID: PMC11122108 DOI: 10.3390/ijms25105492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.
Collapse
Affiliation(s)
- Coby J. Cheetham
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK;
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| |
Collapse
|
12
|
Nguyen TK, Paone S, Baxter AA, Mayfosh AJ, Phan TK, Chan E, Peter K, Poon IKH, Thomas SR, Hulett MD. Heparanase promotes the onset and progression of atherosclerosis in apolipoprotein E gene knockout mice. Atherosclerosis 2024; 392:117519. [PMID: 38581737 DOI: 10.1016/j.atherosclerosis.2024.117519] [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: 09/27/2023] [Revised: 01/31/2024] [Accepted: 03/12/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis is the primary underlying cause of myocardial infarction and stroke, which are the major causes of death globally. Heparanase (Hpse) is a pro-inflammatory extracellular matrix degrading enzyme that has been implicated in atherogenesis. However, to date the precise roles of Hpse in atherosclerosis and its mechanisms of action are not well defined. This study aims to provide new insights into the contribution of Hpse in different stages of atherosclerosis in vivo. METHODS We generated Hpse gene-deficient mice on the atherosclerosis-prone apolipoprotein E gene knockout (ApoE-/-) background to investigate the impact of Hpse gene deficiency on the initiation and progression of atherosclerosis after 6 and 14 weeks high-fat diet feeding, respectively. Atherosclerotic lesion development, blood serum profiles, lesion composition and aortic immune cell populations were evaluated. RESULTS Hpse-deficient mice exhibited significantly reduced atherosclerotic lesion burden in the aortic sinus and aorta at both time-points, independent of changes in plasma cholesterol levels. A significant reduction in the necrotic core size and an increase in smooth muscle cell content were also observed in advanced atherosclerotic plaques of Hpse-deficient mice. Additionally, Hpse deficiency reduced circulating and aortic levels of VCAM-1 at the initiation and progression stages of disease and circulating MCP-1 levels in the initiation but not progression stage. Moreover, the aortic levels of total leukocytes and dendritic cells in Hpse-deficient ApoE-/- mice were significantly decreased compared to control ApoE-/-mice at both disease stages. CONCLUSIONS This study identifies Hpse as a key pro-inflammatory enzyme driving the initiation and progression of atherosclerosis and highlighting the potential of Hpse inhibitors as novel anti-inflammatory treatments for cardiovascular disease.
Collapse
Affiliation(s)
- Tien K Nguyen
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Stephanie Paone
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Amy A Baxter
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Alyce J Mayfosh
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Thanh Kha Phan
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Enoch Chan
- Department of Pathology, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Ivan K H Poon
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Shane R Thomas
- Department of Pathology, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Mark D Hulett
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
| |
Collapse
|
13
|
Rizo-Téllez SA, Filep JG. Beyond host defense and tissue injury: the emerging role of neutrophils in tissue repair. Am J Physiol Cell Physiol 2024; 326:C661-C683. [PMID: 38189129 PMCID: PMC11193466 DOI: 10.1152/ajpcell.00652.2023] [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: 11/29/2023] [Revised: 12/31/2023] [Accepted: 12/31/2023] [Indexed: 01/09/2024]
Abstract
Neutrophils, the most abundant immune cells in human blood, play a fundamental role in host defense against invading pathogens and tissue injury. Neutrophils carry potentially lethal weaponry to the affected site. Inadvertent and perpetual neutrophil activation could lead to nonresolving inflammation and tissue damage, a unifying mechanism of many common diseases. The prevailing view emphasizes the dichotomy of their function, host defense versus tissue damage. However, tissue injury may also persist during neutropenia, which is associated with disease severity and poor outcome. Numerous studies highlight neutrophil phenotypic heterogeneity and functional versatility, indicating that neutrophils play more complex roles than previously thought. Emerging evidence indicates that neutrophils actively orchestrate resolution of inflammation and tissue repair and facilitate return to homeostasis. Thus, neutrophils mobilize multiple mechanisms to limit the inflammatory reaction, assure debris removal, matrix remodeling, cytokine scavenging, macrophage reprogramming, and angiogenesis. In this review, we will summarize the homeostatic and tissue-reparative functions and mechanisms of neutrophils across organs. We will also discuss how the healing power of neutrophils might be harnessed to develop novel resolution and repair-promoting therapies while maintaining their defense functions.
Collapse
Affiliation(s)
- Salma A Rizo-Téllez
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Martin KR, Gamell C, Tai TY, Bonelli R, Hansen J, Tatoulis J, Alhamdoosh M, Wilson N, Wicks I. Whole blood transcriptomics reveals granulocyte colony-stimulating factor as a mediator of cardiopulmonary bypass-induced systemic inflammatory response syndrome. Clin Transl Immunology 2024; 13:e1490. [PMID: 38375330 PMCID: PMC10875393 DOI: 10.1002/cti2.1490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 12/20/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Objectives Systemic inflammatory response syndrome (SIRS) is a frequent complication of cardiopulmonary bypass (CPB). SIRS is associated with significant morbidity and mortality, but its pathogenesis remains incompletely understood, and as a result, biomarkers are lacking and treatment remains expectant and supportive. This study aimed to understand the pathophysiological mechanisms driving SIRS induced by CPB and identify novel therapeutic targets that might reduce systemic inflammation and improve patient outcomes. Methods Twenty-one patients undergoing cardiac surgery and CPB were recruited, and blood was sampled before, during and after surgery. SIRS was defined using the American College of Chest Physicians/Society of Critical Care Medicine criteria. We performed immune cell profiling and whole blood transcriptomics and measured individual mediators in plasma/serum to characterise SIRS induced by CPB. Results Nineteen patients fulfilled criteria for SIRS, with a mean duration of 2.7 days. Neutrophil numbers rose rapidly with CPB and remained elevated for at least 48 h afterwards. Transcriptional signatures associated with neutrophil activation and degranulation were enriched during CPB. We identified a network of cytokines governing these transcriptional changes, including granulocyte colony-stimulating factor (G-CSF), a regulator of neutrophil production and function. Conclusions We identified neutrophils and G-CSF as major regulators of CPB-induced systemic inflammation. Short-term targeting of G-CSF could provide a novel therapeutic strategy to limit neutrophil-mediated inflammation and tissue damage in SIRS induced by CPB.
Collapse
Affiliation(s)
- Katherine R Martin
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
| | | | - Tsin Yee Tai
- WEHIParkvilleVICAustralia
- CSL Innovation, Bio21 InstituteParkvilleVICAustralia
| | - Roberto Bonelli
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- CSL Innovation, Bio21 InstituteParkvilleVICAustralia
| | | | - James Tatoulis
- Cardiothoracic SurgeryRoyal Melbourne HospitalParkvilleVICAustralia
- Department of SurgeryUniversity of MelbourneParkvilleVICAustralia
| | | | | | - Ian Wicks
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- Department of RheumatologyRoyal Melbourne HospitalParkvilleVICAustralia
| |
Collapse
|
15
|
Nudelman A, Shenoy A, Allouche-Arnon H, Fisler M, Rosenhek-Goldian I, Dayan L, Abou Karam P, Porat Z, Solomonov I, Regev-Rudzki N, Bar-Shir A, Sagi I. Proteolytic Vesicles Derived from Salmonella enterica Serovar Typhimurium-Infected Macrophages: Enhancing MMP-9-Mediated Invasion and EV Accumulation. Biomedicines 2024; 12:434. [PMID: 38398037 PMCID: PMC10886541 DOI: 10.3390/biomedicines12020434] [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: 01/04/2024] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Proteolysis of the extracellular matrix (ECM) by matrix metalloproteinases (MMPs) plays a crucial role in the immune response to bacterial infections. Here we report the secretion of MMPs associated with proteolytic extracellular vesicles (EVs) released by macrophages in response to Salmonella enterica serovar Typhimurium infection. Specifically, we used global proteomics, in vitro, and in vivo approaches to investigate the composition and function of these proteolytic EVs. Using a model of S. Typhimurium infection in murine macrophages, we isolated and characterized a population of small EVs. Bulk proteomics analysis revealed significant changes in protein cargo of naïve and S. Typhimurium-infected macrophage-derived EVs, including the upregulation of MMP-9. The increased levels of MMP-9 observed in immune cells exposed to S. Typhimurium were found to be regulated by the toll-like receptor 4 (TLR-4)-mediated response to bacterial lipopolysaccharide. Macrophage-derived EV-associated MMP-9 enhanced the macrophage invasion through Matrigel as selective inhibition of MMP-9 reduced macrophage invasion. Systemic administration of fluorescently labeled EVs into immunocompromised mice demonstrated that EV-associated MMP activity facilitated increased accumulation of EVs in spleen and liver tissues. This study suggests that macrophages secrete proteolytic EVs to enhance invasion and ECM remodeling during bacterial infections, shedding light on an essential aspect of the immune response.
Collapse
Affiliation(s)
- Alon Nudelman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (A.N.); (A.S.); (L.D.); (I.S.)
| | - Anjana Shenoy
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (A.N.); (A.S.); (L.D.); (I.S.)
| | - Hyla Allouche-Arnon
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel; (H.A.-A.); (M.F.); (A.B.-S.)
| | - Michal Fisler
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel; (H.A.-A.); (M.F.); (A.B.-S.)
| | - Irit Rosenhek-Goldian
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel;
| | - Lior Dayan
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (A.N.); (A.S.); (L.D.); (I.S.)
| | - Paula Abou Karam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel; (P.A.K.); (N.R.-R.)
| | - Ziv Porat
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel;
| | - Inna Solomonov
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (A.N.); (A.S.); (L.D.); (I.S.)
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel; (P.A.K.); (N.R.-R.)
| | - Amnon Bar-Shir
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel; (H.A.-A.); (M.F.); (A.B.-S.)
| | - Irit Sagi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (A.N.); (A.S.); (L.D.); (I.S.)
| |
Collapse
|
16
|
Popescu MC, Haddock NL, Burgener EB, Rojas-Hernandez LS, Kaber G, Hargil A, Bollyky PL, Milla CE. The Inovirus Pf4 Triggers Antiviral Responses and Disrupts the Proliferation of Airway Basal Epithelial Cells. Viruses 2024; 16:165. [PMID: 38275975 PMCID: PMC10818373 DOI: 10.3390/v16010165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The inovirus Pf4 is a lysogenic bacteriophage of Pseudomonas aeruginosa (Pa). People with Cystic Fibrosis (pwCF) experience chronic airway infection with Pa and a significant proportion have high numbers of Pf4 in their airway secretions. Given the known severe damage in the airways of Pa-infected pwCF, we hypothesized a high Pf4 burden can affect airway healing and inflammatory responses. In the airway, basal epithelial cells (BCs) are a multipotent stem cell population critical to epithelium homeostasis and repair. We sought to investigate the transcriptional responses of BCs under conditions that emulate infection with Pa and exposure to high Pf4 burden. METHODS Primary BCs isolated from pwCF and wild-type (WT) donors were cultured in vitro and exposed to Pf4 or bacterial Lipopolysaccharide (LPS) followed by transcriptomic and functional assays. RESULTS We found that BCs internalized Pf4 and this elicits a strong antiviral response as well as neutrophil chemokine production. Further, we found that BCs that take up Pf4 demonstrate defective migration and proliferation. CONCLUSIONS Our findings are highly suggestive of Pf4 playing a role in the pathogenicity of Pa in the airways. These findings provide additional evidence for the ability of inoviruses to interact with mammalian cells and disrupt cell function.
Collapse
Affiliation(s)
- Medeea C. Popescu
- Department of Infectious Diseases, Stanford University, Stanford, CA 94305, USA (P.L.B.)
- Immunology Program, Stanford University, Stanford, CA 94305, USA
| | - Naomi L. Haddock
- Department of Infectious Diseases, Stanford University, Stanford, CA 94305, USA (P.L.B.)
- Immunology Program, Stanford University, Stanford, CA 94305, USA
| | - Elizabeth B. Burgener
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura S. Rojas-Hernandez
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gernot Kaber
- Department of Infectious Diseases, Stanford University, Stanford, CA 94305, USA (P.L.B.)
| | - Aviv Hargil
- Department of Infectious Diseases, Stanford University, Stanford, CA 94305, USA (P.L.B.)
| | - Paul L. Bollyky
- Department of Infectious Diseases, Stanford University, Stanford, CA 94305, USA (P.L.B.)
| | - Carlos E. Milla
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
17
|
Casalino-Matsuda SM, Chen F, Gonzalez-Gonzalez FJ, Matsuda H, Nair A, Abdala-Valencia H, Budinger GS, Dong JT, Beitel GJ, Sporn PH. Myeloid Zfhx3 deficiency protects against hypercapnia-induced suppression of host defense against influenza A virus. JCI Insight 2024; 9:e170316. [PMID: 38227369 PMCID: PMC11143927 DOI: 10.1172/jci.insight.170316] [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: 03/10/2023] [Accepted: 01/10/2024] [Indexed: 01/17/2024] Open
Abstract
Hypercapnia, elevation of the partial pressure of CO2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that hypercapnia inhibits multiple macrophage and neutrophil antimicrobial functions and that elevated CO2 increases the mortality of bacterial and viral pneumonia in mice. Here, we show that normoxic hypercapnia downregulates innate immune and antiviral gene programs in alveolar macrophages (AMØs). We also show that zinc finger homeobox 3 (Zfhx3) - a mammalian ortholog of zfh2, which mediates hypercapnic immune suppression in Drosophila - is expressed in mouse and human macrophages. Deletion of Zfhx3 in the myeloid lineage blocked the suppressive effect of hypercapnia on immune gene expression in AMØs and decreased viral replication, inflammatory lung injury, and mortality in hypercapnic mice infected with influenza A virus. To our knowledge, our results establish Zfhx3 as the first known mammalian mediator of CO2 effects on immune gene expression and lay the basis for future studies to identify therapeutic targets to interrupt hypercapnic immunosuppression in patients with advanced lung disease.
Collapse
Affiliation(s)
- S. Marina Casalino-Matsuda
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Fei Chen
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Francisco J. Gonzalez-Gonzalez
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hiroaki Matsuda
- Department of Physical Sciences and Engineering, Wilbur Wright College, Chicago, Illinois, USA
| | - Aisha Nair
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - G.R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Greg J. Beitel
- Department of Molecular Biosciences, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois, USA
| | - Peter H.S. Sporn
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
18
|
Sinton MC, Chandrasegaran PRG, Capewell P, Cooper A, Girard A, Ogunsola J, Perona-Wright G, M Ngoyi D, Kuispond N, Bucheton B, Camara M, Kajimura S, Bénézech C, Mabbott NA, MacLeod A, Quintana JF. IL-17 signalling is critical for controlling subcutaneous adipose tissue dynamics and parasite burden during chronic murine Trypanosoma brucei infection. Nat Commun 2023; 14:7070. [PMID: 37923768 PMCID: PMC10624677 DOI: 10.1038/s41467-023-42918-8] [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: 05/26/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023] Open
Abstract
In the skin, Trypanosoma brucei colonises the subcutaneous white adipose tissue, and is proposed to be competent for forward transmission. The interaction between parasites, adipose tissue, and the local immune system is likely to drive the adipose tissue wasting and weight loss observed in cattle and humans infected with T. brucei. However, mechanistically, events leading to subcutaneous white adipose tissue wasting are not fully understood. Here, using several complementary approaches, including mass cytometry by time of flight, bulk and single cell transcriptomics, and in vivo genetic models, we show that T. brucei infection drives local expansion of several IL-17A-producing cells in the murine WAT, including TH17 and Vγ6+ cells. We also show that global IL-17 deficiency, or deletion of the adipocyte IL-17 receptor protect from infection-induced WAT wasting and weight loss. Unexpectedly, we find that abrogation of adipocyte IL-17 signalling results in a significant accumulation of Dpp4+ Pi16+ interstitial preadipocytes and increased extravascular parasites in the WAT, highlighting a critical role for IL-17 signalling in controlling preadipocyte fate, subcutaneous WAT dynamics, and local parasite burden. Taken together, our study highlights the central role of adipocyte IL-17 signalling in controlling WAT responses to infection, suggesting that adipocytes are critical coordinators of tissue dynamics and immune responses to T. brucei infection.
Collapse
Affiliation(s)
- Matthew C Sinton
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK.
- Division of Cardiovascular Science, University of Manchester, Manchester, UK.
| | - Praveena R G Chandrasegaran
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Paul Capewell
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Anneli Cooper
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Alex Girard
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - John Ogunsola
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Georgia Perona-Wright
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Dieudonné M Ngoyi
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of Congo
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Nono Kuispond
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of Congo
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Bruno Bucheton
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
- Institut de Recherche pour le Développement, Unité Mixte de Recherche IRD-CIRAD 177, Campus International de Baillarguet, Montpellier, France
| | - Mamadou Camara
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine, Ministère de la Santé, Conakry, Guinea
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Cécile Bénézech
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Juan F Quintana
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK.
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
- Division of Immunology, Immunity to Infection and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
19
|
Meteva D, Vinci R, Seppelt C, Abdelwahed YS, Pedicino D, Nelles G, Skurk C, Haghikia A, Rauch-Kröhnert U, Gerhardt T, Straessler E, Zhao Y, Golla F, Joner M, Rai H, Kratzer A, Arnal HG, Liuzzo G, Klotsche J, Crea F, Landmesser U, Leistner DM, Kränkel N. Toll-like receptor 2, hyaluronan, and neutrophils play a key role in plaque erosion: the OPTICO-ACS study. Eur Heart J 2023; 44:3892-3907. [PMID: 37381760 DOI: 10.1093/eurheartj/ehad379] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND AND AIMS In one-third of patients with acute coronary syndrome (ACS), thrombosis occurs despite an intact fibrous cap (IFC) (IFC-ACS, 'plaque erosion'). Recent studies emphasize neutrophils as the immediate inflammatory response in this pathology, but their exact molecular activation patterns are still poorly understood and may represent future therapeutic targets. METHODS AND RESULTS Thirty-two patients with IFC-ACS and matched patients with ACS with ruptured fibrous cap (RFC) (RFC-ACS) from the OPTICO-ACS study were included, and blood samples were collected from the local site of the culprit lesion and the systemic circulation. Neutrophil surface marker expression was quantified by flow cytometry. Neutrophil cytotoxicity towards endothelial cells was examined in an ex vivo co-culture assay. Secretion of active matrix metalloproteinase 9 (MMP9) by neutrophils was evaluated using zymography in supernatants and in plasma samples. Optical coherence tomography (OCT)-embedded thrombi were used for immunofluorescence analysis. Toll-like receptor 2 (TLR2) expression was higher on neutrophils from IFC-ACS than RFC-ACS patients. TLR2 stimulation increased the release of active MMP9 from local IFC-ACS-derived neutrophils, which also aggravated endothelial cell death independently of TLR2. Thrombi of IFC-ACS patients exhibited more hyaluronidase 2 with concomitant increase in local plasma levels of the TLR2 ligand: hyaluronic acid. CONCLUSION The current study provides first in-human evidence for distinct TLR2-mediated neutrophil activation in IFC-ACS, presumably triggered by elevated soluble hyaluronic acid. Together with disturbed flow conditions, neutrophil-released MMP9 might be promoting endothelial cell loss-triggered thrombosis and therefore providing a potential future target for a phenotype-specific secondary therapeutic approach in IFC-ACS.
Collapse
Affiliation(s)
- Denitsa Meteva
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Ramona Vinci
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Department of Cardiovascular Sciences, IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo Francesco Vito 1, Rome 00168, Italy
| | - Claudio Seppelt
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Department of Cardiology and Angiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60598, Germany
| | - Youssef S Abdelwahed
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Daniela Pedicino
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Department of Cardiovascular Sciences, IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo Francesco Vito 1, Rome 00168, Italy
| | - Gregor Nelles
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Carsten Skurk
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Arash Haghikia
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
| | - Ursula Rauch-Kröhnert
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Teresa Gerhardt
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
| | - Elisabeth Straessler
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Yingjie Zhao
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Felix Golla
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Michael Joner
- Department of Cardiology and ISAR Research Centre, German Heart Centre Munich, Lazarettstrasse 36, Munich 80636, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Munich, Munich 80636, Germany
| | - Himanshu Rai
- Cardiovascular Research Institute Dublin, Mater Private Network, 73 Eccles Street, Dublin D07 YH66, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, 123 St. Stephan's Green, Dublin D02 YN77, Ireland
| | - Adelheid Kratzer
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Hector Giral Arnal
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Giovanna Liuzzo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Department of Cardiovascular Sciences, IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo Francesco Vito 1, Rome 00168, Italy
| | - Jens Klotsche
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- German Rheumatism Research Centre (DRFZ) and Institute for Social Medicine, Epidemiology and Health Economy, Charitė University Medicine Berlin, Campus Charite Mitte, Charitėplatz 1, Berlin 10117, Germany
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Department of Cardiovascular Sciences, IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo Francesco Vito 1, Rome 00168, Italy
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
| | - David M Leistner
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
- Department of Cardiology and Angiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60598, Germany
| | - Nicolle Kränkel
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| |
Collapse
|
20
|
Huang K, Liu X, Lv Z, Zhang D, Zhou Y, Lin Z, Guo J. MMP9-Responsive Graphene Oxide Quantum Dot-Based Nano-in-Micro Drug Delivery System for Combinatorial Therapy of Choroidal Neovascularization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207335. [PMID: 36871144 DOI: 10.1002/smll.202207335] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Indexed: 06/18/2023]
Abstract
Age-related macular degeneration (AMD), especially wet AMD with choroidal neovascularization (CNV), commonly causes blindness in older patients and disruption of the choroid followed by second-wave injuries, including chronic inflammation, oxidative stress, and excessive matrix metalloproteinase 9 (MMP9) expression. Increased macrophage infiltrate in parallel with microglial activation and MMP9 overexpression on CNV lesions is shown to contribute to the inflammatory process and then enhance pathological ocular angiogenesis. Graphene oxide quantum dots (GOQDs), as natural antioxidants, exert anti-inflammatory effects and minocycline is a specific macrophage/microglial inhibitor that can suppress both macrophage/microglial activation and MMP9 activity. Herein, an MMP9-responsive GOQD-based minocycline-loaded nano-in-micro drug delivery system (C18PGM) is developed by chemically bonding GOQDs to an octadecyl-modified peptide sequence (C18-GVFHQTVS, C18P) that can be specifically cleaved by MMP9. Using a laser-induced CNV mouse model, the prepared C18PGM shows significant MMP9 inhibitory activity and anti-inflammatory action followed by antiangiogenic effects. Moreover, C18PGM combined with antivascular endothelial growth factor antibody bevacizumab markedly increases the antiangiogenesis effect by interfering with the "inflammation-MMP9-angiogenesis" cascade. The prepared C18PGM shows a good safety profile and no obvious ophthalmic or systemic side effects. The results taken together suggest that C18PGM is an effective and novel strategy for combinatorial therapy of CNV.
Collapse
Affiliation(s)
- Keke Huang
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Xin Liu
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Ziru Lv
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Di Zhang
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Yuling Zhou
- Department of ophthalmology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, 435000, P. R. China
| | - Zhiqing Lin
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Juan Guo
- Department of Ophthalmology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| |
Collapse
|
21
|
Morales-Vázquez MM, Meza-Serrano E, Lara-Pereyra I, Acuña-González RJ, Alonso-Morales R, Hayen-Valles S, Boeta AM, Zarco L, Lozano-Cuenca J, López-Canales JS, Flores-Herrera H. Equine Placentitis in Mares Induces the Secretion of Pro-Inflammatory Cytokine eIL-1β and the Active Extracellular Matrix Metalloproteinase (MMP)-9. Vet Sci 2023; 10:532. [PMID: 37756054 PMCID: PMC10536981 DOI: 10.3390/vetsci10090532] [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: 06/19/2023] [Revised: 08/05/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
Equine placentitis is characterized by infection and inflammation of the placenta. Different biomarkers associated with this inflammatory response have been evaluated in experimentally induced equine placentitis, but not in pregnant mares with spontaneous placentitis. The aim of the current study was to determine the concentration of eIL-1β and the activity of proMMP-2 and proMMP-9 in the serum of healthy mares and mares with placentitis on days 240 and 320 of gestation to explore whether these biomarkers are associated with equine maternal placentitis and/or with the birth of an infected or inviable foals. Serum samples were collected from sixteen pregnant English Thoroughbred mares, retrospectively classified as follows: (1) healthy mares with full-term gestation; and (2) mares with ultrasonographic signs of placentitis. The health of each foal was examined at birth, and it was decided to classify the cases into four groups: (1) healthy mares delivering a healthy foals (HM-HF, n = 6); (2) mares with USP delivering a healthy foal (USP-HF, n = 3); (3) mares with USP delivering a live septic foal (USP-LSeF, n = 4); and (4) mares with USP delivering a dead foal (USP-DF, n = 3). eIL-1β was quantified by ELISA, and proMMP-2 and proMMP-9 activity by gelatin zymography electrophoresis. In healthy mares, the serum concentrations of eIL-1β underwent a significant 16.5-fold increase from day 240 to day 320 of gestation. Although similar results were found in the mares with ultrasonographic signs of placentitis that delivered a healthy foal, those delivering a live septic or nonviable foal exhibited much higher concentrations of eIL-1β. proMMP-2 and proMMP-9 activity was not associated with maternal placentitis, foal infection, or death. Hence, the presence of placentitis severe enough to affect the health of the foal can be confirmed or discarded by determining the eIL-1β concentration in mares that have shown ultrasonographic signs of placentitis.
Collapse
Affiliation(s)
- María Margarita Morales-Vázquez
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Europa Meza-Serrano
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Irlando Lara-Pereyra
- Departamento de Ginecología, Hospital General de Zona 252, Instituto Mexicano del Seguro Social, Atlacomulco 28984, Mexico
| | - Ricardo Josué Acuña-González
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
| | - Rogelio Alonso-Morales
- Genética, Laboratorio de Biotecnologías, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico;
| | - Sergio Hayen-Valles
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Ana Myriam Boeta
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Luis Zarco
- Centro de Enseñanza, Investigación y Extensión en Producción Ovina, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Tres Marías, Ciudad de México 62515, Mexico;
| | - Jair Lozano-Cuenca
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, Mexico; (J.L.-C.); (J.S.L.-C.)
| | - Jorge Skiold López-Canales
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, Mexico; (J.L.-C.); (J.S.L.-C.)
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Héctor Flores-Herrera
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
| |
Collapse
|
22
|
Wei KH, Lin IT, Chowdhury K, Lim KL, Liu KT, Ko TM, Chang YM, Yang KC, Lai SL(B. Comparative single-cell profiling reveals distinct cardiac resident macrophages essential for zebrafish heart regeneration. eLife 2023; 12:e84679. [PMID: 37498060 PMCID: PMC10411971 DOI: 10.7554/elife.84679] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 07/26/2023] [Indexed: 07/28/2023] Open
Abstract
Zebrafish exhibit a robust ability to regenerate their hearts following injury, and the immune system plays a key role in this process. We previously showed that delaying macrophage recruitment by clodronate liposome (-1d_CL, macrophage-delayed model) impairs neutrophil resolution and heart regeneration, even when the infiltrating macrophage number was restored within the first week post injury (Lai et al., 2017). It is thus intriguing to learn the regenerative macrophage property by comparing these late macrophages vs. control macrophages during cardiac repair. Here, we further investigate the mechanistic insights of heart regeneration by comparing the non-regenerative macrophage-delayed model with regenerative controls. Temporal RNAseq analyses revealed that -1d_CL treatment led to disrupted inflammatory resolution, reactive oxygen species homeostasis, and energy metabolism during cardiac repair. Comparative single-cell RNAseq profiling of inflammatory cells from regenerative vs. non-regenerative hearts further identified heterogeneous macrophages and neutrophils, showing alternative activation and cellular crosstalk leading to neutrophil retention and chronic inflammation. Among macrophages, two residential subpopulations (hbaa+ Mac and timp4.3+ Mac 3) were enriched only in regenerative hearts and barely recovered after +1d_CL treatment. To deplete the resident macrophage without delaying the circulating macrophage recruitment, we established the resident macrophage-deficient model by administrating CL earlier at 8 d (-8d_CL) before cryoinjury. Strikingly, resident macrophage-deficient zebrafish still exhibited defects in revascularization, cardiomyocyte survival, debris clearance, and extracellular matrix remodeling/scar resolution without functional compensation from the circulating/monocyte-derived macrophages. Our results characterized the diverse function and interaction between inflammatory cells and identified unique resident macrophages prerequisite for zebrafish heart regeneration.
Collapse
Affiliation(s)
- Ke-Hsuan Wei
- Graduate Institute of Life Sciences, National Defense Medical CenterTaipeiTaiwan
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - I-Ting Lin
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - Kaushik Chowdhury
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Khai Lone Lim
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Kuan-Ting Liu
- Department of Biological Science & Technology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Tai-Ming Ko
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Department of Biological Science & Technology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - Kai-Chien Yang
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Department and Graduate Institute of Pharmacology, National Taiwan University College of MedicineTaipeiTaiwan
| | - Shih-Lei (Ben) Lai
- Graduate Institute of Life Sciences, National Defense Medical CenterTaipeiTaiwan
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
23
|
Arora A, Singh A. Exploring the role of neutrophils in infectious and noninfectious pulmonary disorders. Int Rev Immunol 2023; 43:41-61. [PMID: 37353973 DOI: 10.1080/08830185.2023.2222769] [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: 03/25/2022] [Accepted: 05/31/2023] [Indexed: 06/25/2023]
Abstract
With the change in global environment, respiratory disorders are becoming more threatening to the health of people all over the world. These diseases are closely linked to performance of immune system. Within the innate arm of immune system, Neutrophils are an important moiety to serve as an immune defense barrier. They are one of the first cells recruited to the site of infection and plays a critical role in pathogenesis of various pulmonary diseases. It is established that the migration and activation of neutrophils can lead to inflammation either directly or indirectly and this inflammation caused is very crucial for the clearance of pathogens and resolution of infection. However, the immunopathological mechanisms involved to carry out the same is very complex and not well understood. Despite there being studies concentrating on the role of neutrophils in multiple respiratory diseases, there is still a long way to go in order to completely understand the complexity of the participation of neutrophils and mechanisms involved in the development of these respiratory diseases. In the present article, we have reviewed the literature to comprehensively provide an insight in the current development and advancements about the role of neutrophils in infectious respiratory disorders including viral respiratory disorders such as Coronavirus disease (COVID-19) and bacterial pulmonary disorders with a focused review on pulmonary tuberculosis as well as in noninfectious disorders like Chronic obstructive pulmonary disease (COPD) and asthma. Also, future directions into research and therapeutic targets have been discussed for further exploration.
Collapse
Affiliation(s)
- Alisha Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
24
|
Herrera-Uribe J, Lim KS, Byrne KA, Daharsh L, Liu H, Corbett RJ, Marco G, Schroyen M, Koltes JE, Loving CL, Tuggle CK. Integrative profiling of gene expression and chromatin accessibility elucidates specific transcriptional networks in porcine neutrophils. Front Genet 2023; 14:1107462. [PMID: 37287538 PMCID: PMC10242145 DOI: 10.3389/fgene.2023.1107462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 04/27/2023] [Indexed: 06/09/2023] Open
Abstract
Neutrophils are vital components of the immune system for limiting the invasion and proliferation of pathogens in the body. Surprisingly, the functional annotation of porcine neutrophils is still limited. The transcriptomic and epigenetic assessment of porcine neutrophils from healthy pigs was performed by bulk RNA sequencing and transposase accessible chromatin sequencing (ATAC-seq). First, we sequenced and compared the transcriptome of porcine neutrophils with eight other immune cell transcriptomes to identify a neutrophil-enriched gene list within a detected neutrophil co-expression module. Second, we used ATAC-seq analysis to report for the first time the genome-wide chromatin accessible regions of porcine neutrophils. A combined analysis using both transcriptomic and chromatin accessibility data further defined the neutrophil co-expression network controlled by transcription factors likely important for neutrophil lineage commitment and function. We identified chromatin accessible regions around promoters of neutrophil-specific genes that were predicted to be bound by neutrophil-specific transcription factors. Additionally, published DNA methylation data from porcine immune cells including neutrophils were used to link low DNA methylation patterns to accessible chromatin regions and genes with highly enriched expression in porcine neutrophils. In summary, our data provides the first integrative analysis of the accessible chromatin regions and transcriptional status of porcine neutrophils, contributing to the Functional Annotation of Animal Genomes (FAANG) project, and demonstrates the utility of chromatin accessible regions to identify and enrich our understanding of transcriptional networks in a cell type such as neutrophils.
Collapse
Affiliation(s)
- Juber Herrera-Uribe
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Kyu-Sang Lim
- Department of Animal Science, Iowa State University, Ames, IA, United States
- Department of Animal Resource Science, Kongju National University, Yesan, Republic of Korea
| | - Kristen A. Byrne
- USDA-Agriculture Research Service, National Animal Disease Center, Food Safety and Enteric Pathogens Research Unit, Ames, IA, United States
| | - Lance Daharsh
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Haibo Liu
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Ryan J. Corbett
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Gianna Marco
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Martine Schroyen
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - James E. Koltes
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Crystal L. Loving
- USDA-Agriculture Research Service, National Animal Disease Center, Food Safety and Enteric Pathogens Research Unit, Ames, IA, United States
| | | |
Collapse
|
25
|
Bandyopadhyay SS, Halder AK, Saha S, Chatterjee P, Nasipuri M, Basu S. Assessment of GO-Based Protein Interaction Affinities in the Large-Scale Human-Coronavirus Family Interactome. Vaccines (Basel) 2023; 11:549. [PMID: 36992133 PMCID: PMC10059867 DOI: 10.3390/vaccines11030549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
SARS-CoV-2 is a novel coronavirus that replicates itself via interacting with the host proteins. As a result, identifying virus and host protein-protein interactions could help researchers better understand the virus disease transmission behavior and identify possible COVID-19 drugs. The International Committee on Virus Taxonomy has determined that nCoV is genetically 89% compared to the SARS-CoV epidemic in 2003. This paper focuses on assessing the host-pathogen protein interaction affinity of the coronavirus family, having 44 different variants. In light of these considerations, a GO-semantic scoring function is provided based on Gene Ontology (GO) graphs for determining the binding affinity of any two proteins at the organism level. Based on the availability of the GO annotation of the proteins, 11 viral variants, viz., SARS-CoV-2, SARS, MERS, Bat coronavirus HKU3, Bat coronavirus Rp3/2004, Bat coronavirus HKU5, Murine coronavirus, Bovine coronavirus, Rat coronavirus, Bat coronavirus HKU4, Bat coronavirus 133/2005, are considered from 44 viral variants. The fuzzy scoring function of the entire host-pathogen network has been processed with ~180 million potential interactions generated from 19,281 host proteins and around 242 viral proteins. ~4.5 million potential level one host-pathogen interactions are computed based on the estimated interaction affinity threshold. The resulting host-pathogen interactome is also validated with state-of-the-art experimental networks. The study has also been extended further toward the drug-repurposing study by analyzing the FDA-listed COVID drugs.
Collapse
Affiliation(s)
- Soumyendu Sekhar Bandyopadhyay
- Department of Computer Science and Engineering, Jadavpur University, Kolkata 700032, India
- Department of Computer Science and Engineering, School of Engineering and Technology, Adamas University, Kolkata 700126, India
| | - Anup Kumar Halder
- Faculty of Mathematics and Information Sciences, Warsaw University of Technology, 00-662 Warsaw, Poland
| | - Sovan Saha
- Department of Computer Science and Engineering (Artificial Intelligence and Machine Learning), Techno Main Salt Lake, Sector V, Kolkata 700091, India
| | - Piyali Chatterjee
- Department of Computer Science and Engineering, Netaji Subhash Engineering College, Kolkata 700152, India
| | - Mita Nasipuri
- Department of Computer Science and Engineering, Jadavpur University, Kolkata 700032, India
| | - Subhadip Basu
- Department of Computer Science and Engineering, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
26
|
Sutherland TE, Dyer DP, Allen JE. The extracellular matrix and the immune system: A mutually dependent relationship. Science 2023; 379:eabp8964. [PMID: 36795835 DOI: 10.1126/science.abp8964] [Citation(s) in RCA: 178] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/22/2022] [Indexed: 02/18/2023]
Abstract
For decades, immunologists have studied the role of circulating immune cells in host protection, with a more recent appreciation of immune cells resident within the tissue microenvironment and the intercommunication between nonhematopoietic cells and immune cells. However, the extracellular matrix (ECM), which comprises at least a third of tissue structures, remains relatively underexplored in immunology. Similarly, matrix biologists often overlook regulation of complex structural matrices by the immune system. We are only beginning to understand the scale at which ECM structures determine immune cell localization and function. Additionally, we need to better understand how immune cells dictate ECM complexity. This review aims to highlight the potential for biological discovery at the interface of immunology and matrix biology.
Collapse
Affiliation(s)
- Tara E Sutherland
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
- School of Medicine, Medical Sciences and Dentistry, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Douglas P Dyer
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Salford M6 8HD, UK
| | - Judith E Allen
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
27
|
Kasumba DM, Huot S, Caron E, Fortin A, Laflamme C, Zamorano Cuervo N, Lamontagne F, Pouliot M, Grandvaux N. DUOX2 regulates secreted factors in virus-infected respiratory epithelial cells that contribute to neutrophil attraction and activation. FASEB J 2023; 37:e22765. [PMID: 36607642 PMCID: PMC10107641 DOI: 10.1096/fj.202201205r] [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: 07/29/2022] [Revised: 12/10/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
The first line of defense against respiratory viruses relies on the antiviral and proinflammatory cytokine response initiated in infected respiratory epithelial cells. The cytokine response not only restricts virus replication and spreading, but also orchestrates the subsequent immune response. The epithelial Dual Oxidase 2 (DUOX2) has recently emerged as a regulator of the interferon antiviral response. Here, we investigated the role of DUOX2 in the inflammatory cytokine response using a model of A549 cells deficient in DUOX2 generated using Crispr-Cas9 and infected by Sendai virus. We found that the absence of DUOX2 selectively reduced the induction of a restricted panel of 14 cytokines and chemokines secreted in response to Sendai virus by 20 to 89%. The secreted factors produced by epithelial cells upon virus infection promoted the migration, adhesion, and degranulation of primary human neutrophils, in part through the DUOX2-dependent secretion of TNF and chemokines. In contrast, DUOX2 expression did not impact neutrophil viability or NETosis, thereby highlighting a selective impact of DUOX2 in neutrophil functions. Overall, this study unveils previously unrecognized roles of epithelial DUOX2 in the epithelial-immune cells crosstalk during respiratory virus infection.
Collapse
Affiliation(s)
- Dacquin M Kasumba
- Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Sandrine Huot
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Québec, Canada.,Axe maladies infectieuses et immunitaires, Centre de Recherche du CHU de Québec - Université Laval, Québec City, Québec, Canada
| | - Elise Caron
- Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Audray Fortin
- Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Cynthia Laflamme
- Axe maladies infectieuses et immunitaires, Centre de Recherche du CHU de Québec - Université Laval, Québec City, Québec, Canada
| | - Natalia Zamorano Cuervo
- Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Felix Lamontagne
- Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Marc Pouliot
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Québec, Canada.,Axe maladies infectieuses et immunitaires, Centre de Recherche du CHU de Québec - Université Laval, Québec City, Québec, Canada
| | - Nathalie Grandvaux
- Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
28
|
Differential Effects of Cytokine Versus Hypoxic Preconditioning of Human Mesenchymal Stromal Cells in Pulmonary Sepsis Induced by Antimicrobial-Resistant Klebsiella pneumoniae. Pharmaceuticals (Basel) 2023. [DOI: 10.3390/ph16020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background: Pulmonary sepsis is a leading cause of hospital mortality, and sepses arising from antimicrobial-resistant (AMR) bacterial strains are particularly difficult to treat. Here we investigated the potential of mesenchymal stromal cells (MSCs) to combat established Klebsiella pneumoniae pneumosepsis and further evaluated MSC preconditioning and pre-activation methods. Methods: The potential for naïve and preconditioned MSCs to enhance wound healing, reduce inflammation, preserve metabolic activity, and enhance bacterial killing was assessed in vitro. Rats were subjected to intratracheal K. pneumoniae followed by the intravenous administration of MSCs. Physiological indices, blood, bronchoalveolar lavage (BAL), and tissues were obtained 72 h later. Results: In vitro assays confirmed that preconditioning enhances MSC function, accelerating pulmonary epithelial wound closure, reducing inflammation, attenuating cell death, and increasing bacterial killing. Cytomix-pre-activated MSCs are superior to naïve and hypoxia-exposed MSCs in attenuating Klebsiella pneumosepsis, improving lung compliance and oxygenation, reducing bacteria, and attenuating histologic injuries in lungs. BAL inflammatory cytokines were reduced, correlating with decreases in polymorphonuclear (PMN) cells. MSCs increased PMN apoptosis and the CD4:CD8 ratio in BAL. Systemically, granulocytes, classical monocytes, and the CD4:CD8 ratio were reduced, and nonclassical monocytes were increased. Conclusions: Preconditioning with cytokines, but not hypoxia, enhances the therapeutic potential of MSCs in clinically relevant models of K. pneumoniae-induced pneumosepsis.
Collapse
|
29
|
Biasella F, Plössl K, Baird PN, Weber BHF. The extracellular microenvironment in immune dysregulation and inflammation in retinal disorders. Front Immunol 2023; 14:1147037. [PMID: 36936905 PMCID: PMC10014728 DOI: 10.3389/fimmu.2023.1147037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) as well as genetically complex retinal phenotypes represent a heterogenous group of ocular diseases, both on account of their phenotypic and genotypic characteristics. Therefore, overlaps in clinical features often complicate or even impede their correct clinical diagnosis. Deciphering the molecular basis of retinal diseases has not only aided in their disease classification but also helped in our understanding of how different molecular pathologies may share common pathomechanisms. In particular, these relate to dysregulation of two key processes that contribute to cellular integrity, namely extracellular matrix (ECM) homeostasis and inflammation. Pathological changes in the ECM of Bruch's membrane have been described in both monogenic IRDs, such as Sorsby fundus dystrophy (SFD) and Doyne honeycomb retinal dystrophy (DHRD), as well as in the genetically complex age-related macular degeneration (AMD) or diabetic retinopathy (DR). Additionally, complement system dysfunction and distorted immune regulation may also represent a common connection between some IRDs and complex retinal degenerations. Through highlighting such overlaps in molecular pathology, this review aims to illuminate how inflammatory processes and ECM homeostasis are linked in the healthy retina and how their interplay may be disturbed in aging as well as in disease.
Collapse
Affiliation(s)
- Fabiola Biasella
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Karolina Plössl
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Paul N. Baird
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
- Department of Surgery, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Paul N. Baird, ; Bernhard H. F. Weber,
| | - Bernhard H. F. Weber
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
- Institute of Clinical Human Genetics, University Hospital Regensburg, Regensburg, Germany
- *Correspondence: Paul N. Baird, ; Bernhard H. F. Weber,
| |
Collapse
|
30
|
Thomas PG, Shubina M, Balachandran S. ZBP1/DAI-Dependent Cell Death Pathways in Influenza A Virus Immunity and Pathogenesis. Curr Top Microbiol Immunol 2023; 442:41-63. [PMID: 31970498 DOI: 10.1007/82_2019_190] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Influenza A viruses (IAV) are members of the Orthomyxoviridae family of negative-sense RNA viruses. The greatest diversity of IAV strains is found in aquatic birds, but a subset of strains infects other avian as well as mammalian species, including humans. In aquatic birds, infection is largely restricted to the gastrointestinal tract and spread is through feces, while in humans and other mammals, respiratory epithelial cells are the primary sites supporting productive replication and transmission. IAV triggers the death of most cell types in which it replicates, both in culture and in vivo. When well controlled, such cell death is considered an effective host defense mechanism that eliminates infected cells and limits virus spread. Unchecked or inopportune cell death also results in immunopathology. In this chapter, we discuss the impact of cell death in restricting virus spread, supporting the adaptive immune response and driving pathogenesis in the mammalian respiratory tract. Recent studies have begun to shed light on the signaling pathways underlying IAV-activated cell death. These pathways, initiated by the pathogen sensor protein ZBP1 (also called DAI and DLM1), cause infected cells to undergo apoptosis, necroptosis, and pyroptosis. We outline mechanisms of ZBP1-mediated cell death signaling following IAV infection.
Collapse
Affiliation(s)
- Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, MS 351, 262 Danny Thomas Place, 38105, Memphis, TN, USA.
| | - Maria Shubina
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Sun H, Morihara R, Feng T, Bian Z, Yu H, Hu X, Hu X, Bian Y, Sasaki R, Fukui Y, Takemoto M, Yunoki T, Nakano Y, Abe K, Yamashita T. Human Cord Blood-Endothelial Progenitor Cells Alleviate Intimal Hyperplasia of Arterial Damage in a Rat Stroke Model. Cell Transplant 2023; 32:9636897231193069. [PMID: 37615293 PMCID: PMC10467372 DOI: 10.1177/09636897231193069] [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: 03/26/2023] [Revised: 06/28/2023] [Accepted: 07/18/2023] [Indexed: 08/25/2023] Open
Abstract
Human cord blood-endothelial progenitor cells (hCB-EPCs) isolated from the human umbilical cord can be used to repair damaged arteries. In this study, we used an animal model with pathological changes that mimics artery wall damage caused by stent retrievers in humans. We injected hCB-EPCs to investigate their effect on endothelial hyperplasia and dysfunction during intimal repair. Four groups were established based on the length of reperfusion (3 and 28 days), as well as the presence or absence of hCB-EPC therapy. Damage to the internal carotid artery was evaluated by hematoxylin-eosin and immunohistochemical staining. Stroke volume was not significantly different between non-EPC and EPC groups although EPC treatment alleviated intimal hyperplasia 28 days after intimal damage. Vascular endothelial growth factor (VEGF) and eNOS expression were significantly higher in the EPC-treated group than in the non-EPC group 3 days after intimal damage. In addition, MMP9 and 4HNE expression in the EPC-treated group was significantly lower than in the non-EPC group. Ultimately, this study found that venous transplantation of hCB-EPCs could inhibit neointimal hyperplasia, alleviate endothelial dysfunction, suppress intimal inflammation, and reduce oxidative stress during healing of intimal damage.
Collapse
Affiliation(s)
- Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tian Feng
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xiao Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryo Sasaki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Taijun Yunoki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koji Abe
- National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
32
|
Resveratrol Downmodulates Neutrophil Extracellular Trap (NET) Generation by Neutrophils in Patients with Severe COVID-19. Antioxidants (Basel) 2022; 11:antiox11091690. [PMID: 36139764 PMCID: PMC9495554 DOI: 10.3390/antiox11091690] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022] Open
Abstract
The formation of microthrombi in lung autopsies indicates the involvement of NETs in the immunopathogenesis of severe COVID-19. Therefore, supplements inhibiting NET formation, in association with drugs with fewer adverse effects, should be a relevant strategy to attenuate the disease. Resveratrol (RESV) is a natural polyphenol with an important antiviral and antioxidant role. To modulate neutrophils from patients infected with SARS-CoV-2, we evaluated the in vitro effect of RESV on NET formation. Herein, we investigated 190 patients hospitalized with moderate, severe, and critical symptoms at Hospital das Clínicas, Brazil. We observed that neutrophilia in patients with severe COVID-19 infection is composed of neutrophils with activated profile able to release NET spontaneously. Notably, RESV decreased the neutrophil-activated status and the release of free DNA, inhibiting NET formation even under the specific PMA stimulus. At present, there is no evidence of the role of RESV in neutrophils from patients with COVID-19 infection. These findings suggest that adjunctive therapies with RESV may help decrease the inflammation of viral or bacterial infection, improving patient outcomes.
Collapse
|
33
|
Taylor RS, Ruiz Daniels R, Dobie R, Naseer S, Clark TC, Henderson NC, Boudinot P, Martin SA, Macqueen DJ. Single cell transcriptomics of Atlantic salmon ( Salmo salar L.) liver reveals cellular heterogeneity and immunological responses to challenge by Aeromonas salmonicida. Front Immunol 2022; 13:984799. [PMID: 36091005 PMCID: PMC9450062 DOI: 10.3389/fimmu.2022.984799] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
The liver is a multitasking organ with essential functions for vertebrate health spanning metabolism and immunity. In contrast to mammals, our understanding of liver cellular heterogeneity and its role in regulating immunological status remains poorly defined in fishes. Addressing this knowledge gap, we generated a transcriptomic atlas of 47,432 nuclei isolated from the liver of Atlantic salmon (Salmo salar L.) contrasting control fish with those challenged with a pathogenic strain of Aeromonas salmonicida, a problematic bacterial pathogen in global aquaculture. We identified the major liver cell types and their sub-populations, revealing poor conservation of many hepatic cell marker genes utilized in mammals, while identifying novel heterogeneity within the hepatocyte, lymphoid, and myeloid lineages. This included polyploid hepatocytes, multiple T cell populations including γδ T cells, and candidate populations of monocytes/macrophages and dendritic cells. A dominant hepatocyte population radically remodeled its transcriptome following infection to activate the acute phase response and other defense functions, while repressing routine functions such as metabolism. These defense-specialized hepatocytes showed strong activation of genes controlling protein synthesis and secretion, presumably to support the release of acute phase proteins into circulation. The infection response further involved up-regulation of numerous genes in an immune-cell specific manner, reflecting functions in pathogen recognition and killing, antigen presentation, phagocytosis, regulation of inflammation, B cell differentiation and T cell activation. Overall, this study greatly enhances our understanding of the multifaceted role played by liver immune and non-immune cells in host defense and metabolic remodeling following infection and provides many novel cell-specific marker genes to empower future studies of this organ in fishes.
Collapse
Affiliation(s)
- Richard S. Taylor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Rose Ruiz Daniels
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross Dobie
- Centre for Inflammation Research, The Queen’s Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, United Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Shahmir Naseer
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Thomas C. Clark
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Neil C. Henderson
- Centre for Inflammation Research, The Queen’s Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, United Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Pierre Boudinot
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Samuel A.M. Martin
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Daniel J. Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
34
|
Falcinelli E, Petito E, Gresele P. The role of platelets, neutrophils and endothelium in COVID-19 infection. Expert Rev Hematol 2022; 15:727-745. [PMID: 35930267 DOI: 10.1080/17474086.2022.2110061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION COVID-19 is associated to an increased risk of thrombosis, as a result of a complex process that involves the activation of vascular and circulating cells, the release of soluble inflammatory and thrombotic mediators and blood clotting activation. AREAS COVERED This article reviews the pathophysiological role of platelets, neutrophils and the endothelium, and of their interactions, in the thrombotic complications of COVID-19 patients, and the current and future therapeutic approaches targeting these cell types. EXPERT OPINION Virus-induced platelet, neutrophil and endothelial cell changes are crucial triggers of the thrombotic complications and of the adverse evolution of COVID-19. Both the direct interaction with the virus and the associated cytokine storm concur to trigger cell activation in a classical thromboinflammatory vicious circle. Although heparin has proven to be an effective prophylactic and therapeutic weapon for the prevention and treatment of COVID-19-associated thrombosis, it acts downstream of the cascade of events triggered by SARS-CoV-2. The identification of specific molecular targets interrupting the thromboinflammatory cascade upstream, and more specifically acting either on the interaction of SARS-CoV-2 with blood and vascular cells or on the specific signalling mechanisms associated with their COVID-19-associated activation, might theoretically offer greater protection with potentially lesser side effects.
Collapse
Affiliation(s)
- E Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - E Petito
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - P Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
35
|
Tang F, Brune JE, Chang MY, Reeves SR, Altemeier WA, Frevert CW. Defining the versican interactome in lung health and disease. Am J Physiol Cell Physiol 2022; 323:C249-C276. [PMID: 35649251 PMCID: PMC9291419 DOI: 10.1152/ajpcell.00162.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022]
Abstract
The extracellular matrix (ECM) imparts critical mechanical and biochemical information to cells in the lungs. Proteoglycans are essential constituents of the ECM and play a crucial role in controlling numerous biological processes, including regulating cellular phenotype and function. Versican, a chondroitin sulfate proteoglycan required for embryonic development, is almost absent from mature, healthy lungs and is reexpressed and accumulates in acute and chronic lung disease. Studies using genetically engineered mice show that the versican-enriched matrix can be pro- or anti-inflammatory depending on the cellular source or disease process studied. The mechanisms whereby versican develops a contextual ECM remain largely unknown. The primary goal of this review is to provide an overview of the interaction of versican with its many binding partners, the "versican interactome," and how through these interactions, versican is an integrator of complex extracellular information. Hopefully, the information provided in this review will be used to develop future studies to determine how versican and its binding partners can develop contextual ECMs that control select biological processes. Although this review focuses on versican and the lungs, what is described can be extended to other proteoglycans, tissues, and organs.
Collapse
Affiliation(s)
- Fengying Tang
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Jourdan E Brune
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Mary Y Chang
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Stephen R Reeves
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington
| | - William A Altemeier
- Center for Lung Biology, The University of Washington at South Lake Union, 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, The University of Washington at South Lake Union, 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
|
36
|
Moniot A, Braux J, Siboni R, Guillaume C, Audonnet S, Allart-Simon I, Sapi J, Tirouvanziam R, Gérard S, Gangloff SC, Velard F. Inhibition of Recruitment and Activation of Neutrophils by Pyridazinone-Scaffold-Based Compounds. Int J Mol Sci 2022; 23:ijms23137226. [PMID: 35806233 PMCID: PMC9266889 DOI: 10.3390/ijms23137226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/07/2022] Open
Abstract
In inflammatory diseases, polymorphonuclear neutrophils (PMNs) are known to produce elevated levels of pro-inflammatory cytokines and proteases. To limit ensuing exacerbated cell responses and tissue damage, novel therapeutic agents are sought. 4aa and 4ba, two pyridazinone-scaffold-based phosphodiesterase-IV inhibitors are compared in vitro to zardaverine for their ability to: (1) modulate production of pro-inflammatory mediators, reactive oxygen species (ROS), and phagocytosis; (2) modulate degranulation by PMNs after transepithelial lung migration. Compound 4ba and zardaverine were tested in vivo for their ability to limit tissue recruitment of PMNs in a murine air pouch model. In vitro treatment of lipopolysaccharide-stimulated PMNs with compounds 4aa and 4ba inhibited the release of interleukin-8, tumor necrosis factor-α, and matrix metalloproteinase-9. PMNs phagocytic ability, but not ROS production, was reduced following treatment. Using a lung inflammation model, we proved that PMNs transmigration led to reduced expression of the CD16 phagocytic receptor, which was significantly blunted after treatment with compound 4ba or zardaverine. Using the murine air pouch model, LPS-induced PMNs recruitment was significantly decreased upon addition of compound 4ba or zardaverine. Our data suggest that new pyridazinone derivatives have therapeutic potential in inflammatory diseases by limiting tissue recruitment and activation of PMNs.
Collapse
Affiliation(s)
- Aurélie Moniot
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Julien Braux
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Renaud Siboni
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Christine Guillaume
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Sandra Audonnet
- Université de Reims Champagne-Ardenne, URCACyt, 51 Rue Cognacq Jay, 51100 Reims, France;
| | - Ingrid Allart-Simon
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Janos Sapi
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Center for CF and Airways Disease Research, Children’s Healthcare of Atlanta, 2015 Uppergate Road, Atlanta, GA 30322, USA
| | - Stéphane Gérard
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Sophie C. Gangloff
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Frédéric Velard
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
- Correspondence: ; Tel.: +33-3-26-91-80-10
| |
Collapse
|
37
|
Oishi K, Horiuchi S, Frere J, Schwartz RE, tenOever BR. A diminished immune response underlies age-related SARS-CoV-2 pathologies. Cell Rep 2022; 39:111002. [PMID: 35714615 PMCID: PMC9181267 DOI: 10.1016/j.celrep.2022.111002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/27/2022] [Accepted: 06/03/2022] [Indexed: 12/02/2022] Open
Abstract
Morbidity and mortality in response to SARS-CoV-2 infection are significantly elevated in people of advanced age. To understand the underlying biology of this phenotype, we utilize the golden hamster model to compare how the innate and adaptive immune responses to SARS-CoV-2 infection differed between younger and older animals. We find that while both hamster cohorts showed similar virus kinetics in the lungs, the host response in older animals was dampened, with diminished tissue repair in the respiratory tract post-infection. Characterization of the adaptive immune response also revealed age-related differences, including fewer germinal center B cells in older hamsters, resulting in reduced potency of neutralizing antibodies. Moreover, older animals demonstrate elevated suppressor T cells and neutrophils in the respiratory tract, correlating with an increase in TGF-β and IL-17 induction. Together, these data support that diminished immunity is one of the underlying causes of age-related morbidity.
Collapse
Affiliation(s)
- Kohei Oishi
- Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Shu Horiuchi
- Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Justin Frere
- Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Robert E Schwartz
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
38
|
Zhang Y, Wang Q, Mackay CR, Ng LG, Kwok I. Neutrophil subsets and their differential roles in viral respiratory diseases. J Leukoc Biol 2022; 111:1159-1173. [PMID: 35040189 PMCID: PMC9015493 DOI: 10.1002/jlb.1mr1221-345r] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 12/19/2022] Open
Abstract
Neutrophils play significant roles in immune homeostasis and as neutralizers of microbial infections. Recent evidence further suggests heterogeneity of neutrophil developmental and activation states that exert specialized effector functions during inflammatory disease conditions. Neutrophils can play multiple roles during viral infections, secreting inflammatory mediators and cytokines that contribute significantly to host defense and pathogenicity. However, their roles in viral immunity are not well understood. In this review, we present an overview of neutrophil heterogeneity and its impact on the course and severity of viral respiratory infectious diseases. We focus on the evidence demonstrating the crucial roles neutrophils play in the immune response toward respiratory infections, using influenza as a model. We further extend the understanding of neutrophil function with the studies pertaining to COVID-19 disease and its neutrophil-associated pathologies. Finally, we discuss the relevance of these results for future therapeutic options through targeting and regulating neutrophil-specific responses.
Collapse
Affiliation(s)
- Yuning Zhang
- Department of ResearchNational Skin CentreSingaporeSingapore
| | - Quanbo Wang
- School of Pharmaceutical Sciences, Shandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Charles R Mackay
- School of Pharmaceutical Sciences, Shandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
- Department of Microbiology, Infection and Immunity ProgramBiomedicine Discovery Institute, Monash UniversityMelbourneAustralia
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN)A*STAR (Agency for Science, Technology and Research)BiopolisSingapore
- State Key Laboratory of Experimental HematologyInstitute of Hematology, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Department of Microbiology and ImmunologyImmunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of SingaporeSingaporeSingapore
- National Cancer Centre SingaporeSingaporeSingapore
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN)A*STAR (Agency for Science, Technology and Research)BiopolisSingapore
| |
Collapse
|
39
|
Network pharmacology combined with GEO database identifying the mechanisms and molecular targets of Polygoni Cuspidati Rhizoma on Peri-implants. Sci Rep 2022; 12:8227. [PMID: 35581339 PMCID: PMC9114011 DOI: 10.1038/s41598-022-12366-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/10/2022] [Indexed: 11/08/2022] Open
Abstract
Peri-implants is a chronic disease leads to the bone resorption and loss of implants. Polygoni Cuspidati Rhizoma (PCRER), a traditional Chinese herbal has been used to treat diseases of bone metabolism. However, its mechanism of anti-bone absorption still remains unknown. We aimed to identify its molecular target and the mechanism involved in PCRER potential treatment theory to Peri-implants by network pharmacology. The active ingredients of PCRER and potential disease-related targets were retrieved from TCMSP, Swiss Target Prediction, SEA databases and then combined with the Peri-implants disease differential genes obtained in the GEO microarray database. The crossed genes were used to protein–protein interaction (PPI) construction and Gene Ontology (GO) and KEGG enrichment analysis. Using STRING database and Cytoscape plug-in to build protein interaction network and screen the hub genes and verified through molecular docking by AutoDock vina software. A total of 13 active compounds and 90 cross targets of PCRER were selected for analysis. The GO and KEGG enrichment analysis indicated that the anti-Peri-implants targets of PCRER mainly play a role in the response in IL-17 signaling, Calcium signaling pathway, Toll-like receptor signaling pathway, TNF signaling pathway among others. And CytoHubba screened ten hub genes (MMP9, IL6, MPO, IL1B, SELL, IFNG, CXCL8, CXCL2, PTPRC, PECAM1). Finally, the molecular docking results indicated the good binding ability with active compounds and hub genes. PCRER’s core components are expected to be effective drugs to treat Peri-implants by anti-inflammation, promotes bone metabolism. Our study provides new thoughts into the development of natural medicine for the prevention and treatment of Peri-implants.
Collapse
|
40
|
Lee AJ, Feng E, Chew MV, Balint E, Poznanski SM, Giles E, Zhang A, Marzok A, Revill SD, Vahedi F, Dubey A, Ayaub E, Jimenez-Saiz R, McGrath JJC, Ritchie TM, Jordana M, Jonigk DD, Ackermann M, Ask K, Miller M, Richards CD, Ashkar AA. Type I interferon regulates proteolysis by macrophages to prevent immunopathology following viral infection. PLoS Pathog 2022; 18:e1010471. [PMID: 35512020 PMCID: PMC9113601 DOI: 10.1371/journal.ppat.1010471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 05/17/2022] [Accepted: 03/24/2022] [Indexed: 11/18/2022] Open
Abstract
The ability to treat severe viral infections is limited by our understanding of the mechanisms behind virus-induced immunopathology. While the role of type I interferons (IFNs) in early control of viral replication is clear, less is known about how IFNs can regulate the development of immunopathology and affect disease outcomes. Here, we report that absence of type I IFN receptor (IFNAR) is associated with extensive immunopathology following mucosal viral infection. This pathology occurred independent of viral load or type II immunity but required the presence of macrophages and IL-6. The depletion of macrophages and inhibition of IL-6 signaling significantly abrogated immunopathology. Tissue destruction was mediated by macrophage-derived matrix metalloproteinases (MMPs), as MMP inhibition by doxycycline and Ro 28–2653 reduced the severity of tissue pathology. Analysis of post-mortem COVID-19 patient lungs also displayed significant upregulation of the expression of MMPs and accumulation of macrophages. Overall, we demonstrate that IFNs inhibit macrophage-mediated MMP production to prevent virus-induced immunopathology and uncover MMPs as a therapeutic target towards viral infections.
Collapse
Affiliation(s)
- Amanda J. Lee
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Emily Feng
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Marianne V. Chew
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Elizabeth Balint
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Sophie M. Poznanski
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Elizabeth Giles
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Ali Zhang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Art Marzok
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Spencer D. Revill
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Firestone Institute of Respiratory Health, McMaster University, Hamilton, Ontario, Canada
| | - Fatemeh Vahedi
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Anisha Dubey
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Firestone Institute of Respiratory Health, McMaster University, Hamilton, Ontario, Canada
| | - Ehab Ayaub
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Firestone Institute of Respiratory Health, McMaster University, Hamilton, Ontario, Canada
| | - Rodrigo Jimenez-Saiz
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Joshua J. C. McGrath
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Tyrah M. Ritchie
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Manel Jordana
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Danny D. Jonigk
- Institute of Pathology, Hannover Medical School, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Maximilian Ackermann
- Institute of Pathology and Molecular Pathology, Helios University Clinic Wuppertal, University of Witten/Herdecke, Wuppertal, Germany
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kjetil Ask
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Firestone Institute of Respiratory Health, McMaster University, Hamilton, Ontario, Canada
| | - Matthew Miller
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Carl D. Richards
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Ali A. Ashkar
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
41
|
Chronic Exposure to the Food Additive tBHQ Modulates Expression of Genes Related to SARS-CoV-2 and Influenza Viruses. Life (Basel) 2022; 12:life12050642. [PMID: 35629310 PMCID: PMC9147452 DOI: 10.3390/life12050642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 11/16/2022] Open
Abstract
Background. tert-butylhydroquinone (tBHQ) is an antioxidant commonly used as a food additive. Studies suggest that tBHQ could modulate immune responses to influenza and SARS-CoV-2 infection. In our transcriptomic analysis we explored the molecular mechanisms behind tBHQ’s modulatory properties and the relationships to respiratory viral infections. Methods. tBHQ was administered per os to BALB/c mice (1.5% [w/w]) for 20 days. Splenic T cells were isolated with magnetic separation and subjected to transcriptomic analysis. Gene-set enrichment analysis and g:Profiler was conducted to provide a functional interpretation of significantly changed genes. Further analysis for AHR/NRF2 binding sites was performed with GeneHancer. Results. In CD4+ cells, we found significantly altered expression of 269 genes by tBHQ. Of them, many had relevance in influenza infection such as genes responsible for virus entry (Anxa1/2, Cd14), interferon signaling (Dusp10, Tnfsf13), or prostaglandin synthesis (Ptgs1/2). In SARS-CoV-2 infections, interferon signaling (Ifitm1), proteolytic enzymes (CtsB), and also cell-surface proteins (Cd14, Cd151) were among the prominent alterations after tBHQ exposure. Of these genes, many had one or more binding sites for AHR and NRF2, two major xenosensors triggered by tBHQ. Conclusions. Our results strongly suggest that a common food additive, tBHQ, can modulate virus-dependent processes in both influenza and SARS-CoV-2 infections.
Collapse
|
42
|
Zhang N, Aiyasiding X, Li WJ, Liao HH, Tang QZ. Neutrophil degranulation and myocardial infarction. Cell Commun Signal 2022; 20:50. [PMID: 35410418 PMCID: PMC8996539 DOI: 10.1186/s12964-022-00824-4] [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: 07/12/2021] [Accepted: 12/26/2021] [Indexed: 11/21/2022] Open
Abstract
Myocardial infarction (MI) is one of the most common cardiac emergencies with high morbidity and is a leading cause of death worldwide. Since MI could develop into a life-threatening emergency and could also seriously affect the life quality of patients, continuous efforts have been made to create an effective strategy to prevent the occurrence of MI and reduce MI-related mortality. Numerous studies have confirmed that neutrophils play important roles in inflammation and innate immunity, which provide the first line of defense against microorganisms by producing inflammatory cytokines and chemokines, releasing reactive oxygen species, and degranulating components of neutrophil cytoplasmic granules to kill pathogens. Recently, researchers reported that neutrophils are closely related to the severity and prognosis of patients with MI, and neutrophil to lymphocyte ratio in post-MI patients had predictive value for major adverse cardiac events. Neutrophils have been increasingly recognized to exert important functions in MI. Especially, granule proteins released by neutrophil degranulation after neutrophil activation have been suggested to involve in the process of MI. This article reviewed the current research progress of neutrophil granules in MI and discusses neutrophil degranulation associated diagnosis and treatment strategies. Video abstract
Collapse
Affiliation(s)
- Nan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Xiahenazi Aiyasiding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Wen-Jing Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Hai-Han Liao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
43
|
Parker J, Guslund NC, Jentoft S, Roth O. Characterization of Pipefish Immune Cell Populations Through Single-Cell Transcriptomics. Front Immunol 2022; 13:820152. [PMID: 35154138 PMCID: PMC8828949 DOI: 10.3389/fimmu.2022.820152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/10/2022] [Indexed: 01/16/2023] Open
Abstract
Teleost adaptive immune systems have evolved with more flexibility than previously assumed. A particularly enigmatic system to address immune system modifications in the evolutionary past is represented by the Syngnathids, the family of pipefishes, seahorses and seadragons. These small fishes with their unique male pregnancy have lost the spleen as an important immune organ as well as a functional major histocompatibility class II (MHC II) pathway. How these evolutionary changes have impacted immune cell population dynamics have up to this point remained unexplored. Here, we present the first immune cell repertoire characterization of a syngnathid fish (Syngnathus typhle) using single-cell transcriptomics. Gene expression profiles of individual cells extracted from blood and head-kidney clustered in twelve putative cell populations with eight belonging to those with immune function. Upregulated cell marker genes identified in humans and teleosts were used to define cell clusters. While the suggested loss of CD4+ T-cells accompanied the loss of the MHC II pathway was supported, the upregulation of specific subtype markers within the T-cell cluster indicates subpopulations of regulatory T-cells (il2rb) and cytotoxic T-cells (gzma). Utilizing single-cell RNA sequencing this report is the first to characterize immune cell populations in syngnathids and provides a valuable foundation for future cellular classification and experimental work within the lineage.
Collapse
Affiliation(s)
- Jamie Parker
- Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany.,Marine Evolutionary Biology, Christian-Albrechts-University, Kiel, Germany
| | - Naomi Croft Guslund
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Oslo, Norway.,Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sissel Jentoft
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Olivia Roth
- Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany.,Marine Evolutionary Biology, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
44
|
Schimke LF, Marques AHC, Baiocchi GC, de Souza Prado CA, Fonseca DLM, Freire PP, Rodrigues Plaça D, Salerno Filgueiras I, Coelho Salgado R, Jansen-Marques G, Rocha Oliveira AE, Peron JPS, Cabral-Miranda G, Barbuto JAM, Camara NOS, Calich VLG, Ochs HD, Condino-Neto A, Overmyer KA, Coon JJ, Balnis J, Jaitovich A, Schulte-Schrepping J, Ulas T, Schultze JL, Nakaya HI, Jurisica I, Cabral-Marques O. Severe COVID-19 Shares a Common Neutrophil Activation Signature with Other Acute Inflammatory States. Cells 2022; 11:cells11050847. [PMID: 35269470 PMCID: PMC8909161 DOI: 10.3390/cells11050847] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Severe COVID-19 patients present a clinical and laboratory overlap with other hyperinflammatory conditions such as hemophagocytic lymphohistiocytosis (HLH). However, the underlying mechanisms of these conditions remain to be explored. Here, we investigated the transcriptome of 1596 individuals, including patients with COVID-19 in comparison to healthy controls, other acute inflammatory states (HLH, multisystem inflammatory syndrome in children [MIS-C], Kawasaki disease [KD]), and different respiratory infections (seasonal coronavirus, influenza, bacterial pneumonia). We observed that COVID-19 and HLH share immunological pathways (cytokine/chemokine signaling and neutrophil-mediated immune responses), including gene signatures that stratify COVID-19 patients admitted to the intensive care unit (ICU) and COVID-19_nonICU patients. Of note, among the common differentially expressed genes (DEG), there is a cluster of neutrophil-associated genes that reflects a generalized hyperinflammatory state since it is also dysregulated in patients with KD and bacterial pneumonia. These genes are dysregulated at the protein level across several COVID-19 studies and form an interconnected network with differentially expressed plasma proteins that point to neutrophil hyperactivation in COVID-19 patients admitted to the intensive care unit. scRNAseq analysis indicated that these genes are specifically upregulated across different leukocyte populations, including lymphocyte subsets and immature neutrophils. Artificial intelligence modeling confirmed the strong association of these genes with COVID-19 severity. Thus, our work indicates putative therapeutic pathways for intervention.
Collapse
Affiliation(s)
- Lena F. Schimke
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
- Correspondence: (L.F.S.); (O.C.-M.); Tel.: +55-11-943661555 (L.F.S.); +55-11-974642022 (O.C.-M.)
| | - Alexandre H. C. Marques
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Gabriela Crispim Baiocchi
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Caroline Aliane de Souza Prado
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.A.d.S.P.); (D.L.M.F.); (D.R.P.); (A.E.R.O.); (H.I.N.)
| | - Dennyson Leandro M. Fonseca
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.A.d.S.P.); (D.L.M.F.); (D.R.P.); (A.E.R.O.); (H.I.N.)
| | - Paula Paccielli Freire
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Desirée Rodrigues Plaça
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.A.d.S.P.); (D.L.M.F.); (D.R.P.); (A.E.R.O.); (H.I.N.)
| | - Igor Salerno Filgueiras
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Ranieri Coelho Salgado
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Gabriel Jansen-Marques
- Information Systems, School of Arts, Sciences and Humanities, University of Sao Paulo, São Paulo 03828-000, Brazil;
| | - Antonio Edson Rocha Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.A.d.S.P.); (D.L.M.F.); (D.R.P.); (A.E.R.O.); (H.I.N.)
| | - Jean Pierre Schatzmann Peron
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Gustavo Cabral-Miranda
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - José Alexandre Marzagão Barbuto
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
- Laboratory of Medical Investigation in Pathogenesis, Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Niels Olsen Saraiva Camara
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Vera Lúcia Garcia Calich
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Hans D. Ochs
- Department of Pediatrics, Seattle Children’s Research Institute, University of Washington School of Medicine, Seattle, WA 98101, USA;
| | - Antonio Condino-Neto
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
| | - Katherine A. Overmyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; (K.A.O.); (J.J.C.)
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Joshua J. Coon
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; (K.A.O.); (J.J.C.)
- Morgridge Institute for Research, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53506, USA
| | - Joseph Balnis
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; (J.B.); (A.J.)
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; (J.B.); (A.J.)
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Jonas Schulte-Schrepping
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany; (J.S.-S.); (J.L.S.)
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), University of Bonn, 53127 Bonn, Germany;
| | - Thomas Ulas
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), University of Bonn, 53127 Bonn, Germany;
- German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, University of Bonn, 53127 Bonn, Germany
| | - Joachim L. Schultze
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany; (J.S.-S.); (J.L.S.)
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), University of Bonn, 53127 Bonn, Germany;
- German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, University of Bonn, 53127 Bonn, Germany
| | - Helder I. Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.A.d.S.P.); (D.L.M.F.); (D.R.P.); (A.E.R.O.); (H.I.N.)
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil
- Scientific Platform Pasteur, University of São Paulo, São Paulo 05508-020, Brazil
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada;
- Departments of Medical Biophysics and Computer Science, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1L7, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Otávio Cabral-Marques
- Department of Imunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (A.H.C.M.); (G.C.B.); (P.P.F.); (I.S.F.); (R.C.S.); (J.P.S.P.); (G.C.-M.); (J.A.M.B.); (N.O.S.C.); (V.L.G.C.); (A.C.-N.)
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.A.d.S.P.); (D.L.M.F.); (D.R.P.); (A.E.R.O.); (H.I.N.)
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo 05508-000, Brazil
- Correspondence: (L.F.S.); (O.C.-M.); Tel.: +55-11-943661555 (L.F.S.); +55-11-974642022 (O.C.-M.)
| |
Collapse
|
45
|
Salazar F, Bignell E, Brown GD, Cook PC, Warris A. Pathogenesis of Respiratory Viral and Fungal Coinfections. Clin Microbiol Rev 2022; 35:e0009421. [PMID: 34788127 PMCID: PMC8597983 DOI: 10.1128/cmr.00094-21] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Individuals suffering from severe viral respiratory tract infections have recently emerged as "at risk" groups for developing invasive fungal infections. Influenza virus is one of the most common causes of acute lower respiratory tract infections worldwide. Fungal infections complicating influenza pneumonia are associated with increased disease severity and mortality, with invasive pulmonary aspergillosis being the most common manifestation. Strikingly, similar observations have been made during the current coronavirus disease 2019 (COVID-19) pandemic. The copathogenesis of respiratory viral and fungal coinfections is complex and involves a dynamic interplay between the host immune defenses and the virulence of the microbes involved that often results in failure to return to homeostasis. In this review, we discuss the main mechanisms underlying susceptibility to invasive fungal disease following respiratory viral infections. A comprehensive understanding of these interactions will aid the development of therapeutic modalities against newly identified targets to prevent and treat these emerging coinfections.
Collapse
Affiliation(s)
- Fabián Salazar
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Elaine Bignell
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Gordon D. Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Peter C. Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
46
|
Lai Y, Han T, Lao Z, Li G, Xiao J, Liu X. Phillyrin for COVID-19 and Influenza Co-infection: A Potential Therapeutic Strategy Targeting Host Based on Bioinformatics Analysis. Front Pharmacol 2021; 12:754241. [PMID: 34803696 PMCID: PMC8599367 DOI: 10.3389/fphar.2021.754241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/13/2021] [Indexed: 12/20/2022] Open
Abstract
Background: The risk of co-epidemic between COVID-19 and influenza is very high, so it is urgent to find a treatment strategy for the co-infection. Previous studies have shown that phillyrin can not only inhibit the replication of the two viruses, but also has a good anti-inflammatory effect, which is expected to become a candidate compound against COVID-19 and influenza. Objective: To explore the possibility of phillyrin as a candidate compound for the treatment of COVID-19 and influenza co-infection and to speculate its potential regulatory mechanism. Methods: We used a series of bioinformatics network pharmacology methods to understand and characterize the pharmacological targets, biological functions, and therapeutic mechanisms of phillyrin in COVID-19 and influenza co-infection and discover its therapeutic potential. Results: We revealed potential targets, biological processes, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, and upstream pathway activity of phillyrin against COVID-19 and influenza co-infection. We constructed protein-protein interaction (PPI) network and identified 50 hub genes, such as MMP9, IL-2, VEGFA, AKT, and HIF-1A. Furthermore, our findings indicated that the treatment of phillyrin for COVID-19 and influenza co-infection was associated with immune balance and regulation of hypoxia-cytokine storm, including HIF-1 signaling pathway, PI3K-Akt signaling pathway, Ras signaling pathway, and T cell receptor signaling pathway. Conclusion: For the first time, we uncovered the potential targets and biological pathways of phillyrin for COVID-19 and influenza co-infection. These findings should solve the urgent problem of co-infection of COVID-19 and influenza that the world will face in the future, but clinical drug trials are needed for verification in the future.
Collapse
Affiliation(s)
- Yanni Lai
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tiantian Han
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zizhao Lao
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Geng Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianyong Xiao
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
47
|
Chan L, Karimi N, Morovati S, Alizadeh K, Kakish JE, Vanderkamp S, Fazel F, Napoleoni C, Alizadeh K, Mehrani Y, Minott JA, Bridle BW, Karimi K. The Roles of Neutrophils in Cytokine Storms. Viruses 2021; 13:v13112318. [PMID: 34835125 PMCID: PMC8624379 DOI: 10.3390/v13112318] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 12/16/2022] Open
Abstract
A cytokine storm is an abnormal discharge of soluble mediators following an inappropriate inflammatory response that leads to immunopathological events. Cytokine storms can occur after severe infections as well as in non-infectious situations where inflammatory cytokine responses are initiated, then exaggerated, but fail to return to homeostasis. Neutrophils, macrophages, mast cells, and natural killer cells are among the innate leukocytes that contribute to the pathogenesis of cytokine storms. Neutrophils participate as mediators of inflammation and have roles in promoting homeostatic conditions following pathological inflammation. This review highlights the advances in understanding the mechanisms governing neutrophilic inflammation against viral and bacterial pathogens, in cancers, and in autoimmune diseases, and how neutrophils could influence the development of cytokine storm syndromes. Evidence for the destructive potential of neutrophils in their capacity to contribute to the onset of cytokine storm syndromes is presented across a multitude of clinical scenarios. Further, a variety of potential therapeutic strategies that target neutrophils are discussed in the context of suppressing multiple inflammatory conditions.
Collapse
Affiliation(s)
- Lily Chan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Negar Karimi
- Department of Clinical Sciences, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad 91779-4897, Iran;
| | - Solmaz Morovati
- Division of Biotechnology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz 71557-13876, Iran;
| | - Kasra Alizadeh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Julia E. Kakish
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Sierra Vanderkamp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Fatemeh Fazel
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Christina Napoleoni
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Kimia Alizadeh
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA;
| | - Yeganeh Mehrani
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
- Department of Clinical Sciences, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad 91779-4897, Iran;
| | - Jessica A. Minott
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Byram W. Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
- Correspondence: (B.W.B.); (K.K.); Tel.: +1-519-824-4120 (ext. 54657) (B.W.B.); +1-519-824-4120 (ext. 54668) (K.K.)
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
- Correspondence: (B.W.B.); (K.K.); Tel.: +1-519-824-4120 (ext. 54657) (B.W.B.); +1-519-824-4120 (ext. 54668) (K.K.)
| |
Collapse
|
48
|
Rapid orderly migration of neutrophils after traumatic brain injury depends on MMP9/13. Biochem Biophys Res Commun 2021; 579:161-167. [PMID: 34601201 DOI: 10.1016/j.bbrc.2021.09.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
Macrophages and granulocytes play an important role in various injuries and post-traumatic repair. Due to the limited number of neutrophils in the brain, their role in traumatic brain injury has rarely been mentioned. Here, neutrophils were found to take over the role of macrophages after brain injury in the absence of macrophages. Neutrophils have the characteristics of long residence time and number advantage to actively remove the apoptotic debris. The number of neutrophils recruited was effectively reduced by inhibiting IL-1β. Interestingly, neutrophils migrated regularly and rapidly to the wound during the early stages of brain injury through three paths. They first infiltrated the wound mainly through blood circulation around the eyes, then became unscrupulous and began to move directly across the brain. In addition, MMP9 and MMP13 were found to be related to the migration of neutrophils, and inhibition of MMP could significantly inhibit the number and speed of neutrophils' migration. Our study showed that neutrophils rely on MMP9 and MMP13 for a rapid and orderly response to brain injury to maintain central nervous system stability in the absence or decrease of macrophages.
Collapse
|
49
|
Leite-Avalca MCG, Zampronio A, Lehmann C. Cannabinoid Receptor 1 and 2 Signaling Pathways Involved in Sepsis. Shock 2021; 56:673-681. [PMID: 33625115 DOI: 10.1097/shk.0000000000001763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Sepsis is defined as a life-threatening organ dysfunction, caused by a dysregulated host response to an infection and can progress to septic shock, which represents a major challenge in critical care with a high mortality rate. Currently, there is no definitive treatment available for the dysregulated immune response in sepsis. Therefore, a better understanding of the pathophysiological mechanisms may be useful for elucidating the molecular basis of sepsis and may contribute to the development of new therapeutic strategies. The endocannabinoid system is an emerging research topic for the modulation of the host immune response under various pathological conditions. Cannabinoid receptors include the cannabinoid type 1 receptor (CB1) and the cannabinoid type 2 receptor (CB2). This review addresses the main functionality of CB1 and CB2 in sepsis, which can contribute to a better understanding about the pathophysiology of sepsis. Specifically, we discuss the role of CB1 in the cardiovascular system which is one of the biological systems that are strongly affected by sepsis and septic shock. We are also reviewing the role of CB2 in sepsis, specially CB2 activation, which exerts anti-inflammatory activities with potential benefit in sepsis.
Collapse
Affiliation(s)
| | - Aleksander Zampronio
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Christian Lehmann
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
50
|
McMahon M, Ye S, Pedrina J, Dlugolenski D, Stambas J. Extracellular Matrix Enzymes and Immune Cell Biology. Front Mol Biosci 2021; 8:703868. [PMID: 34527702 PMCID: PMC8436118 DOI: 10.3389/fmolb.2021.703868] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Remodelling of the extracellular matrix (ECM) by ECM metalloproteinases is increasingly being associated with regulation of immune cell function. ECM metalloproteinases, including Matrix Metalloproteinases (MMPs), A Disintegrin and Metalloproteinases (ADAMs) and ADAMs with Thombospondin-1 motifs (ADAMTS) play a vital role in pathogen defence and have been shown to influence migration of immune cells. This review provides a current summary of the role of ECM enzymes in immune cell migration and function and discusses opportunities and limitations for development of diagnostic and therapeutic strategies targeting metalloproteinase expression and activity in the context of infectious disease.
Collapse
Affiliation(s)
- Meagan McMahon
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Siying Ye
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Jess Pedrina
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Daniel Dlugolenski
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - John Stambas
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|