1
|
Ríos I, Herrero C, Torres-Torresano M, López-Navarro B, Schiaffino MT, Díaz Crespo F, Nieto-Valle A, Samaniego R, Sierra-Palomares Y, Oliver E, Revuelta-Salgado F, García-Luján R, Sánchez-Mateos P, Delgado R, Puig-Kröger A, Corbí AL. Reprogramming of GM-CSF-dependent alveolar macrophages through GSK3 activity modulation. eLife 2025; 14:RP102659. [PMID: 40365863 PMCID: PMC12077879 DOI: 10.7554/elife.102659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Monocyte-derived macrophages recruited into inflamed tissues can acquire an array of functional states depending on the extracellular environment. Since the anti-inflammatory/pro-fibrotic macrophage profile is determined by MAFB, whose activity/protein levels are regulated by GSK3, we addressed the macrophage reprogramming potential of GSK3 modulation. GM-CSF-dependent (GM-MØ) and M-CSF-dependent monocyte-derived macrophages (M-MØ) exhibited distinct levels of inactive GSK3, and inhibiting GSK3 in GM-MØ led to the acquisition of transcriptional, phenotypic, and functional properties characteristic of M-MØ (enhanced expression of IL-10 and monocyte-recruiting factors, and higher efferocytosis). These reprogramming effects were also observed upon GSK3α/β knockdown and through GSK3 inhibition in ex vivo isolated human alveolar macrophages (AMØ). Notably, GSK3 downmodulation potentiated the transcriptional signature of interstitial macrophages (IMØ) while suppressing the AMØ-specific gene profile. Indeed, heightened levels of inactive GSK3 and MAFB-dependent proteins were observed in severe COVID-19 patients' lung macrophages, highlighting the GSK3-MAFB axis as a therapeutic target for macrophage reprogramming.
Collapse
Affiliation(s)
- Israel Ríos
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSICMadridSpain
| | - Cristina Herrero
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSICMadridSpain
| | - Mónica Torres-Torresano
- Laboratorio de Inmuno-Metabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio MarañónMadridSpain
| | - Baltasar López-Navarro
- Laboratorio de Inmuno-Metabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio MarañónMadridSpain
| | - María Teresa Schiaffino
- Laboratorio de Inmuno-Metabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio MarañónMadridSpain
| | - Francisco Díaz Crespo
- Servicio de Anatomía Patológica, Hospital General Universitario Gregorio MarañónMadridSpain
| | - Alicia Nieto-Valle
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio MarañónMadridSpain
| | - Rafael Samaniego
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio MarañónMadridSpain
| | - Yolanda Sierra-Palomares
- Experimental Pharmacology and New Tragets in Cardiopulmonary Disorders, Centro de Investigaciones Biológicas, CSICMadridSpain
| | - Eduardo Oliver
- Experimental Pharmacology and New Tragets in Cardiopulmonary Disorders, Centro de Investigaciones Biológicas, CSICMadridSpain
| | | | | | - Paloma Sánchez-Mateos
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio MarañónMadridSpain
- Department of Immunology, Ophthalmology and ENT, Universidad Complutense School of MedicineMadridSpain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario de Octubre (imas12)MadridSpain
- Universidad Complutense School of MedicineMadridSpain
| | - Amaya Puig-Kröger
- Laboratorio de Inmuno-Metabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio MarañónMadridSpain
| | - Angel L Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSICMadridSpain
| |
Collapse
|
2
|
Creyns B, MacKenzie B, Jannini Sa YAP, Coelho AL, Christensen D, Parimon T, Windsor B, Hogaboam CM. Caveolin Scaffolding Domain (CSD) Peptide LTI-2355 Modulates the Phagocytic and Synthetic Activity of Lung-Derived Myeloid Cells in Idiopathic Pulmonary Fibrosis (IPF) and Post-Acute Sequelae of COVID Fibrosis (PASC-F). Biomedicines 2025; 13:796. [PMID: 40299362 PMCID: PMC12024842 DOI: 10.3390/biomedicines13040796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
Rationale: The role of the innate immune system in idiopathic pulmonary fibrosis (IPF) remains poorly understood. However, a functional myeloid compartment is required to remove dying cells and cellular debris, as well as to mediate innate immune responses against pathogens. Aberrant macrophage activity has been described in patients with post-acute sequelae of COVID fibrosis (PASC-F), and caveolin scaffolding domain (CSD) peptides have been found to attenuate inflammation and fibrosis in mouse lung injury models. Therefore, we examined, for the first time, the effects of CSD peptide LTI-2355 on the functional and synthetic properties of human myeloid cells isolated from lung explant tissue of donor lungs as well as IPF and PASC-F lung explant tissue. Methods and Results: CD45+ myeloid cells isolated from lung explant tissue from IPF and PASC-F patients exhibited an impaired capacity to clear autologous dead cells and cellular debris. The uptake of pathogen-coated bioparticles was impaired in myeloid cells from both fibrotic patient groups independent of the type of pathogen, highlighting an intrinsic functional cell impairment. LTI-2355 improved the phagocytic activity of both IPF and PASC-F myeloid cells, and this improvement was paired with decreased proinflammatory and pro-fibrotic synthetic activity. LTI-2355 was also shown to primarily target CD206-expressing IPF and PASC-F myeloid cells. Conclusions: Primary myeloid cells from IPF and PASC-F patients exhibit dysfunctional phagocytic and synthetic properties that are modulated by LTI-2355. LTI-2355 treatment of IPF myeloid cells resulted in significantly reduced sCD163, IFN-α2, IFN-γ, IL-2, IL-10, IL-12p40, and MMP-1 in the cell supernatant. This study highlights an additional mechanism of action of the CSD peptide in the treatment of IPF and progressive fibrotic lung disease.
Collapse
Affiliation(s)
- Brecht Creyns
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - BreAnne MacKenzie
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| | - Yago Amigo Pinho Jannini Sa
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Ana Lucia Coelho
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Dale Christensen
- Division of Hematology, Department of Medicine, Duke University, Durham, NC 27708, USA;
| | - Tanyalak Parimon
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Brian Windsor
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| | - Cory M. Hogaboam
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| |
Collapse
|
3
|
Woods PS, Mutlu GM. Differences in glycolytic metabolism between tissue-resident alveolar macrophages and recruited lung macrophages. Front Immunol 2025; 16:1535796. [PMID: 40092977 PMCID: PMC11906440 DOI: 10.3389/fimmu.2025.1535796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Immunometabolism has emerged as a key area of focus in immunology and has the potential to lead to new treatments for immune-related diseases. It is well-established that glycolytic metabolism is essential for adaptation to hypoxia and for macrophage inflammatory function. Macrophages have been shown to upregulate their glycolytic metabolism in response to pathogens and pathogen-associated molecular patterns such as LPS. As a direct link to the external environment, the lungs' distinctive nutrient composition and multiple macrophage subtypes provide a unique opportunity to study macrophage metabolism. This review aims to highlight how the steady-state airway and severely inflamed airway offer divergent environments for macrophage glycolytic metabolism. We describe the differences in glycolytic metabolism between tissue-resident alveolar macrophages, and other lung macrophages at steady-state and during inflammation/injury. We also provide an overview of experimental guidelines on how to assess metabolism at the cellular level using Seahorse-based bioenergetic analysis including a review of pharmacologic agents used to inhibit or activate glycolysis.
Collapse
Affiliation(s)
| | - Gökhan M. Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University
of Chicago, Chicago, IL, United States
| |
Collapse
|
4
|
Pöpperl P, Stoff M, Beineke A. Alveolar Macrophages in Viral Respiratory Infections: Sentinels and Saboteurs of Lung Defense. Int J Mol Sci 2025; 26:407. [PMID: 39796262 PMCID: PMC11721917 DOI: 10.3390/ijms26010407] [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: 12/09/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Respiratory viral infections continue to cause pandemic and epidemic outbreaks in humans and animals. Under steady-state conditions, alveolar macrophages (AlvMϕ) fulfill a multitude of tasks in order to maintain tissue homeostasis. Due to their anatomic localization within the deep lung, AlvMϕ are prone to detect and react to inhaled viruses and thus play a role in the early pathogenesis of several respiratory viral infections. Here, detection of viral pathogens causes diverse antiviral and proinflammatory reactions. This fact not only makes them promising research targets, but also suggests them as potential targets for therapeutic and prophylactic approaches. This review aims to give a comprehensive overview of the current knowledge about the role of AlvMϕ in respiratory viral infections of humans and animals.
Collapse
Affiliation(s)
- Pauline Pöpperl
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Melanie Stoff
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| |
Collapse
|
5
|
Panahipoor Javaherdehi A, Ghanbari S, Mahdavi P, Zafarani A, Razizadeh MH. The role of alveolar macrophages in viral respiratory infections and their therapeutic implications. Biochem Biophys Rep 2024; 40:101826. [PMID: 39324036 PMCID: PMC11422589 DOI: 10.1016/j.bbrep.2024.101826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
Alveolar macrophages are pivotal components of the lung's innate immune defense against respiratory virus infections. Their multifaceted role spans from viral clearance to modulation of immune responses, making them essential players in shaping disease outcomes. In this comprehensive review collection, we look into the intricate interplay between Alveolar macrophages and various respiratory viruses, shedding light on their dynamic contributions to immune resilience. From influenza to respiratory syncytial virus, Alveolar macrophages emerge as sentinels of the airways, actively participating in viral detection and initiating rapid antiviral responses. Their ability to recognize viral pathogens triggers a cascade of events, including cytokine and chemokine production that guides the recruitment and activation of immune effectors. Furthermore, Alveolar macrophages impact the fate of adaptive immune responses by modulating the activation of T lymphocytes and the secretion of key cytokines. These reviews encompass a range of insights, including the regulation of inflammasome activation, the influence of Alveolar macrophages on cytokine dysregulation, and their role in preventing secondary bacterial pneumonia post-infection. Collectively, they highlight the significance of Alveolar macrophages in preserving pulmonary integrity and immune homeostasis during viral challenges.
Collapse
Affiliation(s)
| | | | - Pooya Mahdavi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Zafarani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Razizadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Tazawa R, Ohashi R, Kitamura N, Tanaka T, Nakagaki K, Yuki S, Fujiwara A, Nakata K. Repeated inhalation of GM-CSF by nonhuman primates induces bronchus-associated lymphoid tissue along the lower respiratory tract. Respir Res 2024; 25:402. [PMID: 39523334 PMCID: PMC11550524 DOI: 10.1186/s12931-024-03003-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Repeated inhalation of granulocyte-macrophage colony-stimulating factor (GM-CSF) was recently approved in Japan as a treatment for autoimmune pulmonary alveolar proteinosis. However, the detailed physiological and pathological effects of repeated inhalation in the long term, especially at increasing doses, remain unclear. METHODS In this chronic safety study, we administered 24 cynomolgus monkeys (Macaca fascicularis) aged 2-3 years with aerosolized sargramostim (a yeast-derived recombinant human GM-CSF [rhGM-CSF]) biweekly for 26 weeks across four dosing groups (0, 5, 100, and 500 µg/kg/day). We measured the serum GM-CSF antibody (GM-Ab) concentration by an ELISA and assessed the neutralizing capacity of GM-Ab using the GM-CSF-dependent cell line TF-1. We subjected lung tissue samples taken from all monkeys at 27 weeks to histopathological assessment using a sargramostim-specific monoclonal antibody to detect localization of residual sargramostim. RESULTS All the animals maintained good body condition and showed steady weight gain throughout the study. The pathological analyses of the lung revealed the formation of induced bronchus-associated lymphoid tissue (iBALT) in the lower respiratory tract, even at the clinical dose of 5 µg/kg/day. There was a relationship between the number or size of BALT and sargramostim dose or the serum GM-Ab levels. Immunohistochemical analyses revealed GM-Ab-producing cells in the follicular region of iBALT, with residual sargramostim in the follicles. Leucocyte counts were inversely correlated with GM-Ab levels in the high-dose groups. Additionally, serum GM-Ab from the treated animals significantly suppressed the alveolar macrophage proliferation activity of both Cynomolgus recombinant and rhGM-CSF in vitro. CONCLUSION Long-term repeated inhalation of sargramostim led to iBALT formation in the lower respiratory tract, even at the clinical dose of 5 µg/kg/day, with the extent of iBALT formation increasing in a dose-dependent manner. Inhaled sargramostim was localized to the follicular region of iBALT nodules, which may induce the production of GM-Ab.
Collapse
Affiliation(s)
- Ryushi Tazawa
- Health Administration Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Riuko Ohashi
- Division of Molecular and Diagnostic Pathology, Histopathology Core Facility, Center for Research Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Nobutaka Kitamura
- Division of Pioneering Advanced Therapeutics, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Niigata, 951-8520, Japan
| | - Takahiro Tanaka
- The Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Kazuhide Nakagaki
- Division of Pioneering Advanced Therapeutics, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Niigata, 951-8520, Japan
| | - Sachiko Yuki
- Division of Pioneering Advanced Therapeutics, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Niigata, 951-8520, Japan
| | | | - Koh Nakata
- Division of Pioneering Advanced Therapeutics, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Niigata, 951-8520, Japan.
| |
Collapse
|
7
|
Debeuf N, Deckers J, Lameire S, Bosteels C, Hammad H, Lambrecht BN. Inhaled GM-CSF administered during ongoing pneumovirus infection alters myeloid and CD8 T cell immunity without affecting disease outcome. Front Immunol 2024; 15:1439789. [PMID: 39439800 PMCID: PMC11493702 DOI: 10.3389/fimmu.2024.1439789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Granulocyte-macrophage colony stimulating factor (GM-CSF) is a pleiotropic cytokine, able to promote both myelopoiesis and activation of immune cells. Particularly in the lung, GM-CSF plays an important homeostatic role in the development and maintenance of alveolar macrophages, and is therefore considered to play a role in respiratory virus infections such as influenza and SARS-CoV-2, although the benefits of GM-CSF treatment in clinical studies remain inconclusive. To address this, we tested inhaled GM-CSF treatment in the Pneumonia Virus of Mice (PVM) mouse model. Our findings show that local GM-CSF therapy during PVM disease increased local neutrophilia and monocyte-derived cell influx, but diminished CD8+ T cells responses. Despite this, the observed effects on T cells and myeloid cells did not result in an altered clinical outcome during PVM infection. We conclude that inhaled GM-CSF therapy cannot be considered as a universal protective therapy in respiratory virus infections.
Collapse
Affiliation(s)
- Nincy Debeuf
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Julie Deckers
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sahine Lameire
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Cedric Bosteels
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N. Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
8
|
Da Silva Filho J, Herder V, Gibbins MP, Dos Reis MF, Melo GC, Haley MJ, Judice CC, Val FFA, Borba M, Tavella TA, de Sousa Sampaio V, Attipa C, McMonagle F, Wright D, de Lacerda MVG, Costa FTM, Couper KN, Marcelo Monteiro W, de Lima Ferreira LC, Moxon CA, Palmarini M, Marti M. A spatially resolved single-cell lung atlas integrated with clinical and blood signatures distinguishes COVID-19 disease trajectories. Sci Transl Med 2024; 16:eadk9149. [PMID: 39259811 DOI: 10.1126/scitranslmed.adk9149] [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: 09/26/2023] [Revised: 02/15/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
COVID-19 is characterized by a broad range of symptoms and disease trajectories. Understanding the correlation between clinical biomarkers and lung pathology during acute COVID-19 is necessary to understand its diverse pathogenesis and inform more effective treatments. Here, we present an integrated analysis of longitudinal clinical parameters, peripheral blood markers, and lung pathology in 142 Brazilian patients hospitalized with COVID-19. We identified core clinical and peripheral blood signatures differentiating disease progression between patients who recovered from severe disease compared with those who succumbed to the disease. Signatures were heterogeneous among fatal cases yet clustered into two patient groups: "early death" (<15 days until death) and "late death" (>15 days). Progression to early death was characterized systemically and in lung histopathological samples by rapid endothelial and myeloid activation and the presence of thrombi associated with SARS-CoV-2+ macrophages. In contrast, progression to late death was associated with fibrosis, apoptosis, and SARS-CoV-2+ epithelial cells in postmortem lung tissue. In late death cases, cytotoxicity, interferon, and T helper 17 (TH17) signatures were only detectable in the peripheral blood after 2 weeks of hospitalization. Progression to recovery was associated with higher lymphocyte counts, TH2 responses, and anti-inflammatory-mediated responses. By integrating antemortem longitudinal blood signatures and spatial single-cell lung signatures from postmortem lung samples, we defined clinical parameters that could be used to help predict COVID-19 outcomes.
Collapse
Affiliation(s)
- João Da Silva Filho
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Vanessa Herder
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Matthew P Gibbins
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Monique Freire Dos Reis
- Department of Education and Research, Oncology Control Centre of Amazonas State (FCECON), Manaus, Brazil
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Federal University of Amazonas, Manaus, Brazil
- Amazonas Oncology Control Center Foundation, Manaus, Brazil
| | | | - Michael J Haley
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Carla Cristina Judice
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Fernando Fonseca Almeida Val
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Mayla Borba
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Delphina Rinaldi Abdel Aziz Emergency Hospital (HPSDRA), Manaus, Brazil
| | - Tatyana Almeida Tavella
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
- INSERM U1016, CNRS UMR8104, University of Paris Cité, Institut Cochin, Paris, France
| | | | - Charalampos Attipa
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Fiona McMonagle
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Glasgow Imaging Facility/School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Derek Wright
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marcus Vinicius Guimaraes de Lacerda
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
- Instituto Leônidas e Maria Deane, Fiocruz, Manaus, Brazil
- University of Texas Medical Branch, Galveston, TX, USA
| | | | - Kevin N Couper
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Wuelton Marcelo Monteiro
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Luiz Carlos de Lima Ferreira
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Christopher Alan Moxon
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- (C.A.M.)
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- (M.P.)
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
- (M.M.)
| |
Collapse
|
9
|
Ruscitti C, Abinet J, Maréchal P, Meunier M, de meeûs C, Vanneste D, Janssen P, Dourcy M, Thiry M, Bureau F, Schneider C, Machiels B, Hidalgo A, Ginhoux F, Dewals B, Guiot J, Schleich F, Garigliany MM, Bellahcène A, Radermecker C, Marichal T. Recruited atypical Ly6G + macrophages license alveolar regeneration after lung injury. Sci Immunol 2024; 9:eado1227. [PMID: 39093958 PMCID: PMC7616420 DOI: 10.1126/sciimmunol.ado1227] [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: 01/17/2024] [Accepted: 05/31/2024] [Indexed: 08/04/2024]
Abstract
The lung is constantly exposed to airborne pathogens and particles that can cause alveolar damage. Hence, appropriate repair responses are essential for gas exchange and life. Here, we deciphered the spatiotemporal trajectory and function of an atypical population of macrophages after lung injury. Post-influenza A virus (IAV) infection, short-lived monocyte-derived Ly6G-expressing macrophages (Ly6G+ Macs) were recruited to the alveoli of lung perilesional areas. Ly6G+ Macs engulfed immune cells, exhibited a high metabolic potential, and clustered with alveolar type 2 epithelial cells (AT2s) in zones of active epithelial regeneration. Ly6G+ Macs were partially dependent on granulocyte-macrophage colony-stimulating factor and interleukin-4 receptor signaling and were essential for AT2-dependent alveolar regeneration. Similar macrophages were recruited in other models of injury and in the airspaces of lungs from patients with suspected pneumonia. This study identifies perilesional alveolar Ly6G+ Macs as a spatially restricted, short-lived macrophage subset promoting epithelial regeneration postinjury, thus representing an attractive therapeutic target for treating lung damage.
Collapse
Affiliation(s)
- C. Ruscitti
- Laboratory of Immunophysiology, GIGA Institute, University of Liège; Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
| | - J. Abinet
- Laboratory of Immunophysiology, GIGA Institute, University of Liège; Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
| | - P. Maréchal
- Laboratory of Immunophysiology, GIGA Institute, University of Liège; Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
| | - M. Meunier
- Laboratory of Immunophysiology, GIGA Institute, University of Liège; Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
| | - C. de meeûs
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
- Department of Pathology, FARAH Institute, University of Liège; Liège, Belgium
| | - D. Vanneste
- Laboratory of Immunophysiology, GIGA Institute, University of Liège; Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
| | - P. Janssen
- Laboratory of Immunophysiology, GIGA Institute, University of Liège; Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
| | - M. Dourcy
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
- Laboratory of Immunology-Vaccinology, FARAH Institute, University of Liège; Liège, Belgium
| | - M. Thiry
- Laboratory of Cellular and Tissular Biology, GIGA Institute, University of Liège; Liège, Belgium
| | - F. Bureau
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, University of Liège; Liège, Belgium
| | - C. Schneider
- Institute of Physiology, University of Zurich; Zurich, Switzerland
| | - B. Machiels
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
- Laboratory of Immunology-Vaccinology, FARAH Institute, University of Liège; Liège, Belgium
| | - A. Hidalgo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III; Madrid, Spain
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine; New Haven, CT, USA
| | - F. Ginhoux
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine; Shanghai, China
- Inserm U1015, Gustave Roussy, Bâtiment de Médecine Moléculaire ; Villejuif, France
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR); Singapore, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre; Singapore, Singapore
| | - B.G. Dewals
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
- Laboratory of Immunology-Vaccinology, FARAH Institute, University of Liège; Liège, Belgium
| | - J. Guiot
- Laboratory of Pneumology, GIGA Institute, University of Liège; Liège, Belgium
- Department of Respiratory Medicine, CHU University Hospital; Liège, Belgium
| | - F. Schleich
- Laboratory of Pneumology, GIGA Institute, University of Liège; Liège, Belgium
- Department of Respiratory Medicine, CHU University Hospital; Liège, Belgium
| | - M-M. Garigliany
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
- Department of Pathology, FARAH Institute, University of Liège; Liège, Belgium
| | - A. Bellahcène
- Metastasis Research Laboratory, GIGA Institute, University of Liège; Liège, Belgium
| | - C. Radermecker
- Laboratory of Immunophysiology, GIGA Institute, University of Liège; Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
| | - T. Marichal
- Laboratory of Immunophysiology, GIGA Institute, University of Liège; Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège; Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute; Wavre, Belgium
| |
Collapse
|
10
|
Bosteels V, Van Duyse J, Ruyssinck E, Van der Borght K, Nguyen L, Gavel J, Janssens S, Van Isterdael G. Automated antibody dispensing to improve high-parameter flow cytometry throughput and analysis. Cytometry A 2024; 105:464-473. [PMID: 38456613 DOI: 10.1002/cyto.a.24835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 03/09/2024]
Abstract
Over the past decade, the flow cytometry field has witnessed significant advancements in the number of fluorochromes that can be detected. This enables researchers to analyze more than 40 markers simultaneously on thousands of cells per second. However, with this increased complexity and multiplicity of markers, the manual dispensing of antibodies for flow cytometry experiments has become laborious, time-consuming, and prone to errors. An automated antibody dispensing system could provide a potential solution by enhancing the efficiency, and by improving data quality by faithfully dispensing the fluorochrome-conjugated antibodies and by enabling the easy addition of extra controls. In this study, a comprehensive comparison of different liquid handlers for dispensing fluorochrome-labeled antibodies was conducted for the preparation of flow cytometry stainings. The evaluation focused on key criteria including dispensing time, dead volume, and reliability of dispensing. After benchmarking, the I.DOT, a non-contact liquid handler, was selected and optimized in more detail. In the end, the I.DOT was able to prepare a 25-marker panel in 20 min, including the full stain, all FMOs and all single stain controls for cells and beads. Having all these controls improved the validation of the panel, visualization, and analysis of the data. Thus, automated antibody dispensing by dispensers such as the I.DOT reduces time and errors, enhances data quality, and can be easily integrated in an automated workflow to prepare samples for flow cytometry.
Collapse
Affiliation(s)
- Victor Bosteels
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
| | - Julie Van Duyse
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB Flow Core, VIB Center for Inflammation Research, Ghent, Belgium
| | - Elien Ruyssinck
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB Flow Core, VIB Center for Inflammation Research, Ghent, Belgium
| | - Katrien Van der Borght
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB Flow Core, VIB Center for Inflammation Research, Ghent, Belgium
| | - Long Nguyen
- VIB Screening Core, VIB, Ghent, Belgium
- Center for Bioassay Development and Screening, Ghent University, Ghent, Belgium
| | - Jannes Gavel
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Immunoregulation and Mucosal Immunology Lab, VIB Center for Inflammation Research, Ghent, Belgium
| | - Sophie Janssens
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
| | - Gert Van Isterdael
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB Flow Core, VIB Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
11
|
Lafon T, Chapuis N, Guerin E, Daix T, Otranto M, Boumediene A, Jeannet R, Fontenay M, Henri Hani K, Vignon P, Monneret G, François B, Jean-Philippe J, Feuillard J. Along with PaO2/FiO2 ratio and lymphopenia, low HLA-DR monocytes are the only additional parameter that independently predicts the clinical course of undifferentiated SARS-CoV-2 patients in emergency departments. J Leukoc Biol 2024; 115:1131-1142. [PMID: 38366559 DOI: 10.1093/jleuko/qiae022] [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: 09/30/2023] [Revised: 12/28/2023] [Accepted: 01/01/2024] [Indexed: 02/18/2024] Open
Abstract
Because one-third of patients deteriorate after their admission to the emergency department, assessing the prognosis of COVID-19 patients is of great importance. However, to date, only lymphopenia and the partial pressure of oxygen/fraction of inspired oxygen (PaO2/FiO2) ratio have been reported as partly predictive of COVID-19-related further deterioration, and their association has not been evaluated. We asked whether other key biomarkers of SARS-CoV-2 immunologic defects-increase in circulating immature granulocytes, loss of monocyte HLA-DR (mHLA-DR) expression, and monocyte differentiation blockade-could also predict further COVID-19 deterioration. A series of 284 consecutive COVID-19 patients, with the sole inclusion criterion of being an adult, were prospectively enrolled at emergency department admission (day 0) of 2 different hospitals: 1 for the exploratory cohort (180 patients) and 1 for the confirmatory cohort (104 patients). Deterioration was assessed over the next 7 days. Neither increased immature granulocyte levels nor monocyte differentiation blockade predicted patient worsening. Among more than 30 clinical, biological, and radiological parameters, the value of decreased P/F ratio and lymphopenia for prediction of further COVID-19 deterioration was strongly confirmed, and the loss of mHLA-DR was the only additional independent marker. Combined together in a simple OxyLymphoMono score, the 3 variables perfectly predicted patients who did not worsen and correctly predicted worsening in 59% of cases. By highlighting lymphocyte and monocyte defects as preceding COVID-19 deterioration, these results point on early immunosuppression in COVID-19 deterioration. Combining P/F ratio, lymphopenia, and loss of mHLA-DR together in a simple and robust score could offer a pragmatic method for COVID-19 patient stratification.
Collapse
Affiliation(s)
- Thomas Lafon
- Emergency Department, University Hospital Center of Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Nicolas Chapuis
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut Cochin, Université Paris Cité, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
- Service d'Hématologie Biologique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Centre-Université Paris Cité, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Estelle Guerin
- Laboratoire d'Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87042 Limoges, France
| | - Thomas Daix
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
- Réanimation Polyvalente, Centre Hospitalier Universitaire de Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1092, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Marcela Otranto
- Emergency Department, University Hospital Center of Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Ahmed Boumediene
- Laboratoire Immunologie, Centre Hospitalier Universitaire de Limoges, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87042 Limoges, France
| | - Robin Jeannet
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
- Unité Mixte de Recherche Centre National de Recherche Scientifique 7276/Institut National de la Santé et de la Recherche Médicale U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87025 Limoges, France
| | - Michaela Fontenay
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut Cochin, Université Paris Cité, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
- Service d'Hématologie Biologique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Centre-Université Paris Cité, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Karam Henri Hani
- Emergency Department, University Hospital Center of Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Philippe Vignon
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
- Réanimation Polyvalente, Centre Hospitalier Universitaire de Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1092, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Guillaume Monneret
- Laboratoire d'Immunologie, Hôpital E. Herriot, Hospices Civils de Lyon, 5 Pl. d'Arsonval, 69003 Lyon, France
| | - Bruno François
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
- Réanimation Polyvalente, Centre Hospitalier Universitaire de Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1092, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Jais Jean-Philippe
- Imagine Institute, Université Paris Cité, 24 Bd du Montparnasse, 75015 Paris, France
- Biostatistic Unit, Necker University Hospital, Assistance Publique-Hôpitaux de Paris, 149 rue de Sèvre, 75015 Paris, France
- Human Genetics of Infectious Diseases: Complex Predisposition, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1163, 24 Bd du Montparnasse, 75015 Paris, France
| | - Jean Feuillard
- Laboratoire d'Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87042 Limoges, France
- Unité Mixte de Recherche Centre National de Recherche Scientifique 7276/Institut National de la Santé et de la Recherche Médicale U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87025 Limoges, France
| |
Collapse
|
12
|
Pervizaj-Oruqaj L, Ferrero MR, Matt U, Herold S. The guardians of pulmonary harmony: alveolar macrophages orchestrating the symphony of lung inflammation and tissue homeostasis. Eur Respir Rev 2024; 33:230263. [PMID: 38811033 PMCID: PMC11134199 DOI: 10.1183/16000617.0263-2023] [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: 12/20/2023] [Accepted: 03/20/2024] [Indexed: 05/31/2024] Open
Abstract
Recent breakthroughs in single-cell sequencing, advancements in cellular and tissue imaging techniques, innovations in cell lineage tracing, and insights into the epigenome collectively illuminate the enigmatic landscape of alveolar macrophages in the lung under homeostasis and disease conditions. Our current knowledge reveals the cellular and functional diversity of alveolar macrophages within the respiratory system, emphasising their remarkable adaptability. By synthesising insights from classical cell and developmental biology studies, we provide a comprehensive perspective on alveolar macrophage functional plasticity. This includes an examination of their ontology-related features, their role in maintaining tissue homeostasis under steady-state conditions and the distinct contribution of bone marrow-derived macrophages (BMDMs) in promoting tissue regeneration and restoring respiratory system homeostasis in response to injuries. Elucidating the signalling pathways within inflammatory conditions, the impact of various triggers on tissue-resident alveolar macrophages (TR-AMs), as well as the recruitment and polarisation of macrophages originating from the bone marrow, presents an opportunity to propose innovative therapeutic approaches aimed at modulating the equilibrium between phenotypes to induce programmes associated with a pro-regenerative or homeostasis phenotype of BMDMs or TR-AMs. This, in turn, can lead to the amelioration of disease outcomes and the attenuation of detrimental inflammation. This review comprehensively addresses the pivotal role of macrophages in the orchestration of inflammation and resolution phases after lung injury, as well as ageing-related shifts and the influence of clonal haematopoiesis of indeterminate potential mutations on alveolar macrophages, exploring altered signalling pathways and transcriptional profiles, with implications for respiratory homeostasis.
Collapse
Affiliation(s)
- Learta Pervizaj-Oruqaj
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Maximiliano Ruben Ferrero
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Buenos Aires, Argentina
| | - Ulrich Matt
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
13
|
Tzetzo SL, Kramer ED, Mohammadpour H, Kim M, Rosario SR, Yu H, Dolan MR, Oturkar CC, Morreale BG, Bogner PN, Stablewski AB, Benavides FJ, Brackett CM, Ebos JM, Das GM, Opyrchal M, Nemeth MJ, Evans SS, Abrams SI. Downregulation of IRF8 in alveolar macrophages by G-CSF promotes metastatic tumor progression. iScience 2024; 27:109187. [PMID: 38420590 PMCID: PMC10901102 DOI: 10.1016/j.isci.2024.109187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 01/16/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Tissue-resident macrophages (TRMs) are abundant immune cells within pre-metastatic sites, yet their functional contributions to metastasis remain incompletely understood. Here, we show that alveolar macrophages (AMs), the main TRMs of the lung, are susceptible to downregulation of the immune stimulatory transcription factor IRF8, impairing anti-metastatic activity in models of metastatic breast cancer. G-CSF is a key tumor-associated factor (TAF) that acts upon AMs to reduce IRF8 levels and facilitate metastasis. Translational relevance of IRF8 downregulation was observed among macrophage precursors in breast cancer and a CD68hiIRF8loG-CSFhi gene signature suggests poorer prognosis in triple-negative breast cancer (TNBC), a G-CSF-expressing subtype. Our data highlight the underappreciated, pro-metastatic roles of AMs in response to G-CSF and identify the contribution of IRF8-deficient AMs to metastatic burden. AMs are an attractive target of local neoadjuvant G-CSF blockade to recover anti-metastatic activity.
Collapse
Affiliation(s)
- Stephanie L. Tzetzo
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Elliot D. Kramer
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hemn Mohammadpour
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Minhyung Kim
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Spencer R. Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Han Yu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Melissa R. Dolan
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Chetan C. Oturkar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Brian G. Morreale
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Paul N. Bogner
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Aimee B. Stablewski
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Fernando J. Benavides
- Department of Epigenetics and Molecular Carcinogenesis, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Craig M. Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - John M.L. Ebos
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Gokul M. Das
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Mateusz Opyrchal
- Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Michael J. Nemeth
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Sharon S. Evans
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Scott I. Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
14
|
Nguyen TTT, Kim YT, Jeong G, Jin M. Immunopathology of and potential therapeutics for secondary hemophagocytic lymphohistiocytosis/macrophage activation syndrome: a translational perspective. Exp Mol Med 2024; 56:559-569. [PMID: 38448692 PMCID: PMC10984945 DOI: 10.1038/s12276-024-01182-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/21/2023] [Accepted: 12/19/2023] [Indexed: 03/08/2024] Open
Abstract
Secondary hemophagocytic lymphohistiocytosis/macrophage activation syndrome (sHLH/MAS) is a life-threatening immune disorder triggered by rheumatic disease, infections, malignancies, or medications. Characterized by the presence of hemophagocytic macrophages and a fulminant cytokine storm, sHLH/MAS leads to hyperferritinemia and multiorgan failure and rapidly progresses to death. The high mortality rate and the lack of specific treatments necessitate the development of a new drug. However, the complex and largely unknown immunopathologic mechanisms of sHLH/MAS, which involve dysfunction of various immune cells, diverse etiologies, and different clinical contexts make this effort challenging. This review introduces the terminology, diagnosis, and clinical features of sHLH/MAS. From a translational perspective, this review focuses on the immunopathological mechanisms linked to various etiologies, emphasizing potential drug targets, including key molecules and signaling pathways. We also discuss immunomodulatory biologics, existing drugs under clinical evaluation, and novel therapies in clinical trials. This systematic review aims to provide insights and highlight opportunities for the development of novel sHLH/MAS therapeutics.
Collapse
Affiliation(s)
- Tram T T Nguyen
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Yoon Tae Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Geunyeol Jeong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Mirim Jin
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.
- Department of Microbiology, College of Medicine, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
15
|
Creyns B, MacKenzie B, Sa Y, Coelho AL, Christensen D, Parimon T, Windsor B, Hogaboam CM. Caveolin scaffolding domain (CSD) peptide LTI-2355 modulates the phagocytic and synthetic activity of lung derived myeloid cells in Idiopathic Pulmonary Fibrosis (IPF) and Post-acute sequelae of COVID-fibrosis (PASC-F). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.01.569608. [PMID: 38654821 PMCID: PMC11037873 DOI: 10.1101/2023.12.01.569608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Rationale The role of the innate immune system in Idiopathic Pulmonary Fibrosis (IPF) remains poorly understood. However, a functional myeloid compartment is required to remove dying cells and cellular debris, and to mediate innate immune responses against pathogens. Aberrant macrophage activity has been described in patients with Post-acute sequelae of COVID fibrosis (PASC-F). Therefore, we examined the functional and synthetic properties of myeloid cells isolated from normal donor lung and lung explant tissue from both IPF and PASC-F patients and explored the effect of LTI-2355, a Caveolin Scaffolding Domain (CSD) peptide, on these cells. Methods & Results CD45 + myeloid cells isolated from lung explant tissue from IPF and PASC-F patients exhibited an impaired capacity to clear autologous dead cells and cellular debris. Uptake of pathogen-coated bioparticles was impaired in myeloid cells from both fibrotic patient groups independent of type of pathogen highlighting a cell intrinsic functional impairment. LTI-2355 improved the phagocytic activity of both IPF and PASC-F myeloid cells, and this improvement was paired with decreased pro-inflammatory and pro-fibrotic synthetic activity. LTI-2355 was also shown to primarily target CD206-expressing IPF and PASC-F myeloid cells. Conclusions Primary myeloid cells from IPF and PASC-F patients exhibit dysfunctional phagocytic and synthetic properties that are reversed by LTI-2355. Thus, these studies highlight an additional mechanism of action of a CSD peptide in the treatment of IPF and progressive fibrotic lung disease.
Collapse
|
16
|
Shimasaki S, Baba T, Ogura T, Akasaka K, Matsushima H, Izumi S, Takasaki J, Tsushima K, Kinouchi T, Kichikawa Y, Awashima M, Izumo T, Awano N, Nishimura N, Tazawa R, Mikami A, Kitamura N, Ishii H, Kurihara Y, Taniguchi M, Aikawa S, Okada M, Morita Y, Ishikawa Y, Ohinata A, Nakata K. Short-term inhalation of sargramostim with concomitant high-dose steroids does not hasten recovery in moderate COVID-19 pneumonia: a double-blind, randomised, placebo-controlled trial. Infect Dis (Lond) 2023; 55:857-873. [PMID: 37729076 DOI: 10.1080/23744235.2023.2254380] [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: 05/10/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Granulocyte-macrophage colony stimulating factor (GM-CSF) inhalation may alleviate pulmonary inflammation caused by viral pneumonia. To investigate this, we evaluated its efficacy on COVID-19 pneumonia. METHODS This double-blind, randomised, placebo-controlled study (ClinicalTrials.gov: NCT04642950) evaluated patients in the first half of 2021 at seven Japanese hospitals. Hospitalised patients with COVID-19 pneumonia with moderate hypoxaemia inhaled sargramostim or placebo for 5 days. The primary endpoint was days to achieve a ≥ 2-category improvement from baseline on a modified 7-category ordinal scale. Secondary endpoints included degree of oxygenation, defined by amount of oxygen supply, and serum CCL17 level. RESULTS Seventy-five patients were randomly assigned in a 2:1 ratio to receive sargramostim or placebo, of which 47 and 23 were analysed, respectively. No difference was observed between groups regarding the primary endpoint (8.0 and 7.0 days for sargramostim and placebo, respectively) or in the secondary endpoints, except for CCL17. A post hoc sub-analysis indicated that endpoint assessments were influenced by concomitant corticosteroid therapy. When the cumulative corticosteroid dose was ≤500 mg during Days 1-5, recovery and oxygenation were faster in the sargramostim group than for placebo. Bolus dose corticosteroids were associated with temporarily impaired oxygenation and delayed clinical recovery. The increase in serum CCL17, a candidate prognostic factor, reflected improvement with sargramostim inhalation. The number of adverse events was similar between groups. Two serious adverse events were observed in the sargramostim group without causal relation. CONCLUSIONS Inhaled sargramostim was likely to be effective for COVID-19 pneumonia unless the concomitant corticosteroid dose was high.
Collapse
Affiliation(s)
| | - Tomohisa Baba
- Department of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | - Takashi Ogura
- Department of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | - Keiichi Akasaka
- Department of Respiratory Medicine, Saitama Red Cross Hospital, Saitama, Japan
| | - Hidekazu Matsushima
- Department of Respiratory Medicine, Saitama Red Cross Hospital, Saitama, Japan
| | - Shinyu Izumi
- Department of Respiratory Medicine, National Center for Global Health and Medicine, Shinjuku-ku, Japan
| | - Jin Takasaki
- Department of Respiratory Medicine, National Center for Global Health and Medicine, Shinjuku-ku, Japan
| | - Kenji Tsushima
- Department of Pulmonary Medicine, International University of Health and Welfare (IUHW), Chiba, Japan
| | - Toru Kinouchi
- Department of Pulmonary Medicine, International University of Health and Welfare (IUHW), Chiba, Japan
| | - Yoshiko Kichikawa
- Department of Respiratory Medicine, Federation of National Public Service Personnel Mutual Aid Associations, Mishuku Hospital, Meguro-ku, Japan
| | - Maiko Awashima
- Department of Respiratory Medicine, Federation of National Public Service Personnel Mutual Aid Associations, Mishuku Hospital, Meguro-ku, Japan
| | - Takehiro Izumo
- Department of Respiratory Medicine, Japanese Red Cross Medical Center, Shibuya-ku, Japan
| | - Nobuyasu Awano
- Department of Respiratory Medicine, Japanese Red Cross Medical Center, Shibuya-ku, Japan
| | - Naoki Nishimura
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, Chuo-ku, Japan
| | - Ryushi Tazawa
- Health Administration Center, Student Support and Health Administration Organization, Tokyo Medical and Dental University, Bunkyo-ku, Japan
| | - Ayako Mikami
- National Center for Global Health and Medicine, Center for Clinical Sciences, Shinjuku-ku, Japan
| | - Nobutaka Kitamura
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Haruyuki Ishii
- Department of Respiratory Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | - Yasuyuki Kurihara
- Department of Radiology, St. Luke's International Hospital, Chuo-ku, Japan
| | | | | | | | | | | | | | - Koh Nakata
- Center for Medical Innovation, Division of Pioneering Advanced Therapeutics, Niigata University Medical Dental Hospital, Niigata, Japan
| |
Collapse
|
17
|
da Silva Antunes R, Weiskopf D, Sidney J, Rubiro P, Peters B, Arlehamn CSL, Grifoni A, Sette A. The MegaPool Approach to Characterize Adaptive CD4+ and CD8+ T Cell Responses. Curr Protoc 2023; 3:e934. [PMID: 37966108 PMCID: PMC10662678 DOI: 10.1002/cpz1.934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Epitopes recognized by T cells are a collection of short peptide fragments derived from specific antigens or proteins. Immunological research to study T cell responses is hindered by the extreme degree of heterogeneity of epitope targets, which are usually derived from multiple antigens; within a given antigen, hundreds of different T cell epitopes can be recognized, differing from one individual to the next because T cell epitope recognition is restricted by the epitopes' ability to bind to MHC molecules, which are extremely polymorphic in different individuals. Testing large pools encompassing hundreds of peptides is technically challenging because of logistical considerations regarding solvent-induced toxicity. To address this issue, we developed the MegaPool (MP) approach based on sequential lyophilization of large numbers of peptides that can be used in a variety of assays to measure T cell responses, including ELISPOT, intracellular cytokine staining, and activation-induced marker assays, and that has been validated in the study of infectious diseases, allergies, and autoimmunity. Here, we describe the procedures for generating and testing MPs, starting with peptide synthesis and lyophilization, as well as a step-by-step guide and recommendations for their handling and experimental usage. Overall, the MP approach is a powerful strategy for studying T cell responses and understanding the immune system's role in health and disease. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Generation of peptide pools ("MegaPools") Basic Protocol 2: MegaPool testing and quantitation of antigen-specific T cell responses.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Paul Rubiro
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | | | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| |
Collapse
|
18
|
Malainou C, Abdin SM, Lachmann N, Matt U, Herold S. Alveolar macrophages in tissue homeostasis, inflammation, and infection: evolving concepts of therapeutic targeting. J Clin Invest 2023; 133:e170501. [PMID: 37781922 PMCID: PMC10541196 DOI: 10.1172/jci170501] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
Alveolar macrophages (AMs) are the sentinel cells of the alveolar space, maintaining homeostasis, fending off pathogens, and controlling lung inflammation. During acute lung injury, AMs orchestrate the initiation and resolution of inflammation in order to ultimately restore homeostasis. This central role in acute lung inflammation makes AMs attractive targets for therapeutic interventions. Single-cell RNA-Seq and spatial omics approaches, together with methodological advances such as the generation of human macrophages from pluripotent stem cells, have increased understanding of the ontogeny, function, and plasticity of AMs during infectious and sterile lung inflammation, which could move the field closer to clinical application. However, proresolution phenotypes might conflict with proinflammatory and antibacterial responses. Therefore, therapeutic targeting of AMs at vulnerable time points over the course of infectious lung injury might harbor the risk of serious side effects, such as loss of antibacterial host defense capacity. Thus, the identification of key signaling hubs that determine functional fate decisions in AMs is of the utmost importance to harness their therapeutic potential.
Collapse
Affiliation(s)
- Christina Malainou
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Shifaa M. Abdin
- German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology and
- REBIRTH Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology and
- REBIRTH Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- RESIST (Resolving Infection Susceptibility), Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Ulrich Matt
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Susanne Herold
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
19
|
Sueblinvong V, Fan X, Hart C, Molina S, Koval M, Guidot DM. Ethanol-exposed lung fibroblasts cause airway epithelial barrier dysfunction. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1839-1849. [PMID: 37864530 DOI: 10.1111/acer.15174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Chronic alcohol ingestion predisposes to lung injury and disrepair during sepsis. Our previous studies outlined roles for transforming growth factor-beta 1 (TGFβ1) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in epithelial barrier homeostasis and how alcohol perturbs their expression and signaling. Here we hypothesize that ethanol-exposed lung fibroblasts (LF) are a source of dysregulated TGFβ1 and GM-CSF and thereby alter airway epithelial barrier function. METHODS Human or rat LF were cultured ± ethanol for 2 weeks and then co-cultured with human or rat airway epithelial cells (AEC) seeded on Transwell permeable supports. In selected groups, a TGFβ1 receptor type 1 (TGFβR1) inhibitor (SB431542) or a TGFβ1 neutralizing antibody was applied. Transepithelial electrical resistance (TER) was measured prior to co-culture and on day 5 of co-culture. AEC were then analyzed for the expression of selected tight junction and mesenchymal proteins, and transwell membranes were analyzed by immunofluorescence microscopy for ZO-1 expression and localization. TGFβ1 and GM-CSF levels in conditioned media from the co-cultures were quantified by ELISA. RESULTS AEC co-cultured with ethanol-exposed LF (ELF) showed a significant reduction in TER and corresponding decreases in ZO-1 expression, whereas collagen type 1A1 and α-smooth muscle actin protein expression were increased. In parallel, in conditioned media from the ELF + AEC co-cultures, activated TGFβ1 levels increased and GM-CSF levels decreased. Notably, all the effects of ELF on the AEC were prevented by blocking TGFβ1 activity. CONCLUSIONS Prior ethanol exposure to LF induces barrier dysfunction in naive AEC in a paracrine fashion through activation of TGFβ1 signaling and suppression of GM-CSF. These experimental findings provide a potential mechanism by which chronic alcohol ingestion impairs airway epithelial integrity and renders individuals susceptible to lung injury.
Collapse
Affiliation(s)
- Viranuj Sueblinvong
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Xian Fan
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Craishun Hart
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Samuel Molina
- FUJIFILM Irvine Scientific, Warminster, Pennsylvania, USA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David M Guidot
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
20
|
Imbiakha B, Ezzatpour S, Buchholz DW, Sahler J, Ye C, Olarte-Castillo XA, Zou A, Kwas C, O’Hare K, Choi A, Adeleke RA, Khomandiak S, Goodman L, Jager MC, Whittaker GR, Martinez-Sobrido L, August A, Aguilar HC. Age-dependent acquisition of pathogenicity by SARS-CoV-2 Omicron BA.5. SCIENCE ADVANCES 2023; 9:eadj1736. [PMID: 37738347 PMCID: PMC10516498 DOI: 10.1126/sciadv.adj1736] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/23/2023] [Indexed: 09/24/2023]
Abstract
Pathology studies of SARS-CoV-2 Omicron variants of concern (VOC) are challenged by the lack of pathogenic animal models. While Omicron BA.1 and BA.2 replicate in K18-hACE2 transgenic mice, they cause minimal to negligible morbidity and mortality, and less is known about more recent Omicron VOC. Here, we show that in contrast to Omicron BA.1, BA.5-infected mice exhibited high levels of morbidity and mortality, correlating with higher early viral loads. Neither Omicron BA.1 nor BA.5 replicated in brains, unlike most prior VOC. Only Omicron BA.5-infected mice exhibited substantial weight loss, high pathology scores in lungs, and high levels of inflammatory cells and cytokines in bronchoalveolar lavage fluid, and 5- to 8-month-old mice exhibited 100% fatality. These results identify a rodent model for pathogenesis or antiviral countermeasure studies for circulating SARS-CoV-2 Omicron BA.5. Further, differences in morbidity and mortality between Omicron BA.1 and BA.5 provide a model for understanding viral determinants of pathogenicity.
Collapse
Affiliation(s)
- Brian Imbiakha
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, 14853, USA
| | - David W. Buchholz
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Julie Sahler
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Ximena A. Olarte-Castillo
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
- James A. Baker Institute for Animal Health, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Anna Zou
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Cole Kwas
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Katelyn O’Hare
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Richard Ayomide Adeleke
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Solomiia Khomandiak
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Laura Goodman
- James A. Baker Institute for Animal Health, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
- Department of Public & Ecosystem Health, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Mason C. Jager
- Department of Population Medicine and Diagnostic Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
- Department of Public & Ecosystem Health, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | | | - Avery August
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
- Department of Microbiology, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, 14853, USA
| |
Collapse
|
21
|
Ray A, Kale SL, Ramonell RP. Bridging the Gap between Innate and Adaptive Immunity in the Lung: Summary of the Aspen Lung Conference 2022. Am J Respir Cell Mol Biol 2023; 69:266-280. [PMID: 37043828 PMCID: PMC10503303 DOI: 10.1165/rcmb.2023-0057ws] [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: 02/16/2023] [Accepted: 04/12/2023] [Indexed: 04/14/2023] Open
Abstract
Although significant strides have been made in the understanding of pulmonary immunology, much work remains to be done to comprehensively explain coordinated immune responses in the lung. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic only served to highlight the inadequacy of current models of host-pathogen interactions and reinforced the need for current and future generations of immunologists to unravel complex biological questions. As part of that effort, the 64th Annual Thomas L. Petty Aspen Lung Conference was themed "Bridging the Gap between Innate and Adaptive Immunity in the Lung" and featured exciting work from renowned immunologists. This report summarizes the proceedings of the 2022 Aspen Lung Conference, which was convened to discuss the roles played by innate and adaptive immunity in disease pathogenesis, evaluate the interface between the innate and adaptive immune responses, assess the role of adaptive immunity in the development of autoimmunity and autoimmune lung disease, discuss lessons learned from immunologic cancer treatments and approaches, and define new paradigms to harness the immune system to prevent and treat lung diseases.
Collapse
Affiliation(s)
- Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sagar L. Kale
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Richard P. Ramonell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| |
Collapse
|
22
|
Van Damme KFA, Hoste L, Declercq J, De Leeuw E, Maes B, Martens L, Colman R, Browaeys R, Bosteels C, Verwaerde S, Vermeulen N, Lameire S, Debeuf N, Deckers J, Stordeur P, Depuydt P, Van Braeckel E, Vandekerckhove L, Guilliams M, Schetters STT, Haerynck F, Tavernier SJ, Lambrecht BN. A complement atlas identifies interleukin-6-dependent alternative pathway dysregulation as a key druggable feature of COVID-19. Sci Transl Med 2023; 15:eadi0252. [PMID: 37611083 DOI: 10.1126/scitranslmed.adi0252] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023]
Abstract
Improvements in COVID-19 treatments, especially for the critically ill, require deeper understanding of the mechanisms driving disease pathology. The complement system is not only a crucial component of innate host defense but can also contribute to tissue injury. Although all complement pathways have been implicated in COVID-19 pathogenesis, the upstream drivers and downstream effects on tissue injury remain poorly defined. We demonstrate that complement activation is primarily mediated by the alternative pathway, and we provide a comprehensive atlas of the complement alterations around the time of respiratory deterioration. Proteomic and single-cell sequencing mapping across cell types and tissues reveals a division of labor between lung epithelial, stromal, and myeloid cells in complement production, in addition to liver-derived factors. We identify IL-6 and STAT1/3 signaling as an upstream driver of complement responses, linking complement dysregulation to approved COVID-19 therapies. Furthermore, an exploratory proteomic study indicates that inhibition of complement C5 decreases epithelial damage and markers of disease severity. Collectively, these results support complement dysregulation as a key druggable feature of COVID-19.
Collapse
Affiliation(s)
- Karel F A Van Damme
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Levi Hoste
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
| | - Jozefien Declercq
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Elisabeth De Leeuw
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bastiaan Maes
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Liesbet Martens
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
| | - Roos Colman
- Biostatistics Unit, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Robin Browaeys
- Bioinformatics Expertise Unit, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Cédric Bosteels
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Respiratory Infection and Defense Lab, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Stijn Verwaerde
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Nicky Vermeulen
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
| | - Sahine Lameire
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Nincy Debeuf
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Julie Deckers
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Patrick Stordeur
- Belgian National Reference Center for the Complement System, Laboratory of Immunology, LHUB-ULB, Université Libre de Bruxelles, Brussels, Belgium
| | - Pieter Depuydt
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Intensive Care Unit, Ghent University Hospital, Ghent, Belgium
| | - Eva Van Braeckel
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Respiratory Infection and Defense Lab, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Linos Vandekerckhove
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University and Ghent University Hospital, 9000 Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
| | - Sjoerd T T Schetters
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
| | - Simon J Tavernier
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
23
|
Dib L, Koneva LA, Edsfeldt A, Zurke YX, Sun J, Nitulescu M, Attar M, Lutgens E, Schmidt S, Lindholm MW, Choudhury RP, Cassimjee I, Lee R, Handa A, Goncalves I, Sansom SN, Monaco C. Lipid-associated macrophages transition to an inflammatory state in human atherosclerosis increasing the risk of cerebrovascular complications. NATURE CARDIOVASCULAR RESEARCH 2023; 2:656-672. [PMID: 38362263 PMCID: PMC7615632 DOI: 10.1038/s44161-023-00295-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 05/31/2023] [Indexed: 02/17/2024]
Abstract
The immune system is integral to cardiovascular health and disease. Targeting inflammation ameliorates adverse cardiovascular outcomes. Atherosclerosis, a major underlying cause of cardiovascular disease (CVD), is conceptualised as a lipid-driven inflammation where macrophages play a non-redundant role. However, evidence emerging so far from single cell atlases suggests a dichotomy between lipid associated and inflammatory macrophage states. Here, we present an inclusive reference atlas of human intraplaque immune cell communities. Combining scRNASeq of human surgical carotid endarterectomies in a discovery cohort with bulk RNASeq and immunohistochemistry in a validation cohort (the Carotid Plaque Imaging Project-CPIP), we reveal the existence of PLIN2hi/TREM1hi macrophages as a toll-like receptor-dependent inflammatory lipid-associated macrophage state linked to cerebrovascular events. Our study shifts the current paradigm of lipid-driven inflammation by providing biological evidence for a pathogenic macrophage transition to an inflammatory lipid-associated phenotype and for its targeting as a new treatment strategy for CVD.
Collapse
Grants
- FS/18/63/34184 British Heart Foundation
- Novo Nordisk Fonden (Novo Nordisk Foundation)
- British Heart Foundation (BHF)
- Fondation Leducq
- European Commission (EC)
- Kennedy Trust for Rheumatology Research (KENN161701, KENN202101, KENN192004), Oxford NIHR Biomedical Research Centre.
- Vetenskapsrådet (Swedish Research Council)
- The Swedish Society for Medical Research, Crafoord foundation; The Swedish Society of Medicine, the Swedish Heart and Lung Foundation, Diabetes foundation, SUS foundation, Lund University Diabetes Center, The Knut and Alice Wallenberg foundation, the Medical Faculty at Lund University and Region Skåne.
- Kennedy Trust for Rheumatology Research (KENN161701, KENN202101, KENN192004)
- Netcare-Physicians-Partnership trust
- Stiftelsen för Strategisk Forskning (Swedish Foundation for Strategic Research)
Collapse
Affiliation(s)
- Lea Dib
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Lada A. Koneva
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Clinical Research Center, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Yasemin-Xiomara Zurke
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Jiangming Sun
- Department of Clinical Sciences Malmö, Clinical Research Center, Lund University, Malmö, Sweden
| | - Mihaela Nitulescu
- Department of Clinical Sciences Malmö, Clinical Research Center, Lund University, Malmö, Sweden
| | - Moustafa Attar
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Esther Lutgens
- Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN USA
| | - Steffen Schmidt
- Roche Pharma Research and Early Development, RNA Therapeutics Research, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Marie W. Lindholm
- Roche Pharma Research and Early Development, RNA Therapeutics Research, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | | | - Ismail Cassimjee
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Ashok Handa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Clinical Research Center, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Stephen N. Sansom
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Rodriguez-Rodriguez L, Gillet L, Machiels B. Shaping of the alveolar landscape by respiratory infections and long-term consequences for lung immunity. Front Immunol 2023; 14:1149015. [PMID: 37081878 PMCID: PMC10112541 DOI: 10.3389/fimmu.2023.1149015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/15/2023] [Indexed: 04/07/2023] Open
Abstract
Respiratory infections and especially viral infections, along with other extrinsic environmental factors, have been shown to profoundly affect macrophage populations in the lung. In particular, alveolar macrophages (AMs) are important sentinels during respiratory infections and their disappearance opens a niche for recruited monocytes (MOs) to differentiate into resident macrophages. Although this topic is still the focus of intense debate, the phenotype and function of AMs that recolonize the niche after an inflammatory insult, such as an infection, appear to be dictated in part by their origin, but also by local and/or systemic changes that may be imprinted at the epigenetic level. Phenotypic alterations following respiratory infections have the potential to shape lung immunity for the long-term, leading to beneficial responses such as protection against allergic airway inflammation or against other infections, but also to detrimental responses when associated with the development of immunopathologies. This review reports the persistence of virus-induced functional alterations in lung macrophages, and discusses the importance of this imprinting in explaining inter-individual and lifetime immune variation.
Collapse
|
25
|
Loos P, Baiwir J, Maquet C, Javaux J, Sandor R, Lallemand F, Marichal T, Machiels B, Gillet L. Dampening type 2 properties of group 2 innate lymphoid cells by a gammaherpesvirus infection reprograms alveolar macrophages. Sci Immunol 2023; 8:eabl9041. [PMID: 36827420 DOI: 10.1126/sciimmunol.abl9041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Immunological dysregulation in asthma is associated with changes in exposure to microorganisms early in life. Gammaherpesviruses (γHVs), such as Epstein-Barr virus, are widespread human viruses that establish lifelong infection and profoundly shape host immunity. Using murid herpesvirus 4 (MuHV-4), a mouse γHV, we show that after infection, lung-resident and recruited group 2 innate lymphoid cells (ILC2s) exhibit a reduced ability to expand and produce type 2 cytokines in response to house dust mites, thereby contributing to protection against asthma. In contrast, MuHV-4 infection triggers GM-CSF production by those lung ILC2s, which orders the differentiation of monocytes (Mos) into alveolar macrophages (AMs) without promoting their type 2 functions. In the context of γHV infection, ILC2s are therefore essential cells within the pulmonary niche that imprint the tissue-specific identity of Mo-derived AMs and shape their function well beyond the initial acute infection.
Collapse
Affiliation(s)
- Pauline Loos
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Jérôme Baiwir
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Céline Maquet
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Justine Javaux
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Rémy Sandor
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - François Lallemand
- Centre Hospitalier Universitaire de Liège, Département de Physique Médicale, Service médical de radiothérapie, Liège 4000, Belgium
| | - Thomas Marichal
- Laboratory of Immunophysiology, GIGA-Research and Faculty of Veterinary Medicine, ULiège, Liège 4000, Belgium
| | - Bénédicte Machiels
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Laurent Gillet
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| |
Collapse
|
26
|
Lazarus HM, Pitts K, Wang T, Lee E, Buchbinder E, Dougan M, Armstrong DG, Paine R, Ragsdale CE, Boyd T, Rock EP, Gale RP. Recombinant GM-CSF for diseases of GM-CSF insufficiency: Correcting dysfunctional mononuclear phagocyte disorders. Front Immunol 2023; 13:1069444. [PMID: 36685591 PMCID: PMC9850113 DOI: 10.3389/fimmu.2022.1069444] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Endogenous granulocyte-macrophage colony-stimulating factor (GM-CSF), identified by its ability to support differentiation of hematopoietic cells into several types of myeloid cells, is now known to support maturation and maintain the metabolic capacity of mononuclear phagocytes including monocytes, macrophages, and dendritic cells. These cells sense and attack potential pathogens, present antigens to adaptive immune cells, and recruit other immune cells. Recombinant human (rhu) GM-CSF (e.g., sargramostim [glycosylated, yeast-derived rhu GM-CSF]) has immune modulating properties and can restore the normal function of mononuclear phagocytes rendered dysfunctional by deficient or insufficient endogenous GM-CSF. Methods We reviewed the emerging biologic and cellular effects of GM-CSF. Experts in clinical disease areas caused by deficient or insufficient endogenous GM-CSF examined the role of GM-CSF in mononuclear phagocyte disorders including autoimmune pulmonary alveolar proteinosis (aPAP), diverse infections (including COVID-19), wound healing, and anti-cancer immune checkpoint inhibitor therapy. Results We discuss emerging data for GM-CSF biology including the positive effects on mitochondrial function and cell metabolism, augmentation of phagocytosis and efferocytosis, and immune cell modulation. We further address how giving exogenous rhu GM-CSF may control or treat mononuclear phagocyte dysfunction disorders caused or exacerbated by GM-CSF deficiency or insufficiency. We discuss how rhu GM-CSF may augment the anti-cancer effects of immune checkpoint inhibitor immunotherapy as well as ameliorate immune-related adverse events. Discussion We identify research gaps, opportunities, and the concept that rhu GM-CSF, by supporting and restoring the metabolic capacity and function of mononuclear phagocytes, can have significant therapeutic effects. rhu GM-CSF (e.g., sargramostim) might ameliorate multiple diseases of GM-CSF deficiency or insufficiency and address a high unmet medical need.
Collapse
Affiliation(s)
- Hillard M. Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH, United States
| | - Katherine Pitts
- Medical Affairs, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Tisha Wang
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elinor Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elizabeth Buchbinder
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Michael Dougan
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - David G. Armstrong
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Robert Paine
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, United States
| | | | - Timothy Boyd
- Clinical Development, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Edwin P. Rock
- Clinical Development, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Robert Peter Gale
- Hematology Centre, Department of Immunology and Inflammation, Imperial College, London, United Kingdom
| |
Collapse
|