1
|
Neiens V, Hansbauer EM, Jaquin TJ, Peper-Gabriel JK, Mahavadi P, Snyder ME, Grill MJ, Wurzenberger C, Konitsiotis A, Estrada-Bernal A, Heinig K, Fysikopoulos A, Schwenck N, Grüner S, Bartoschek D, Mosebach T, Kerstan S, Wrennall J, Richter M, Noda K, Hoetzenecker K, Burgess JK, Tarran R, Wurzenberger C, Wichmann KR, Biehler J, Müller KW, Guenther A, Eickelberg O, Fitzgerald MF, Olwill SA, Matschiner G, Pavlidou M. Preclinical concept studies showing advantage of an inhaled anti-CTGF/CCN2 protein for pulmonary fibrosis treatment. Nat Commun 2025; 16:3251. [PMID: 40185752 PMCID: PMC11971276 DOI: 10.1038/s41467-025-58568-x] [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: 01/25/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Inhaled therapeutics have high potential for the treatment of chronic respiratory diseases of high unmet medical need, such as idiopathic pulmonary fibrosis (IPF). Preclinical and early clinical evidence show that cellular communication network factor 2 (CCN2), previously called connective tissue growth factor (CTGF), is a promising target for the treatment of IPF. In recent phase 3 clinical trials, however, systemic CCN2 inhibition failed to demonstrate a clinically meaningful benefit. Here, we present the preclinical profile of the inhaled anti-CCN2 Anticalin® protein PRS-220. Our study demonstrates that efficient pulmonary delivery directly translates into superior efficacy in relevant models of pulmonary fibrosis when compared to systemic CCN2 inhibition. Moreover, we present a holistic approach for the preclinical characterization of inhaled PRS-220 from state-of-the art in vitro and in vivo models to novel human ex vivo and in silico models, highlighting the advantage of inhaled drug delivery for treatment of respiratory disease.
Collapse
Affiliation(s)
- Vanessa Neiens
- Pieris Pharmaceuticals GmbH, 85399, Hallbergmoos, Germany
| | | | | | | | - Poornima Mahavadi
- Department of Internal Medicine, Justus Liebig University (JLU) Giessen, 35392, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, 35392, Giessen, Germany
| | - Mark E Snyder
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Starzl Transplantation Institute, Pittsburgh, PA, 15213, USA
| | | | | | | | - Adriana Estrada-Bernal
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | | | | | | | - Stefan Grüner
- Pieris Pharmaceuticals GmbH, 85399, Hallbergmoos, Germany
| | | | | | - Sandra Kerstan
- Pieris Pharmaceuticals GmbH, 85399, Hallbergmoos, Germany
| | - Joe Wrennall
- University of North Carolina Department of Cell Biology & Physiology, Chapel Hill, NC, 27599, USA
| | | | - Kentaro Noda
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery Vienna, Medical University of Vienna, 1090, Vienna, Austria
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, 9713 GZ, Groningen, the Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ, Groningen, the Netherlands
| | - Robert Tarran
- University of North Carolina Department of Cell Biology & Physiology, Chapel Hill, NC, 27599, USA
| | | | | | | | | | - Andreas Guenther
- Department of Internal Medicine, Justus Liebig University (JLU) Giessen, 35392, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, 35392, Giessen, Germany
- Agaplesion Evangelisches Krankenhaus Mittelhessen, 35398, Giessen, Germany
- European IPF Network and European IPF Registry, 35392, Giessen, Germany
- Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | | | - Shane A Olwill
- Pieris Pharmaceuticals GmbH, 85399, Hallbergmoos, Germany
| | | | | |
Collapse
|
2
|
Noda K, Atale N, Austin T, Geller DA, Gerlach J, Sanchez PG. Hepatic conditioning results in better lung endothelial cell preservation under hypoxic environment in vitro. Int J Artif Organs 2025; 48:84-91. [PMID: 39885637 DOI: 10.1177/03913988251315092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
BACKGROUND as we look to extend ex vivo lung perfusion times (EVLP) to improve preservation, the metabolic activity of the lungs will require support from other organ functions. Active functional liver support, including detoxification, synthesis, and regulation, can improve lung preservation during EVLP. This study aimed to demonstrate the effects of hepatic conditioning of the EVLP perfusate on lung endothelium, via the receptor of advanced glycation end-products (RAGE)-nuclear-factor-κB (NF-κB) signaling in vitro. METHODS we performed in vitro experiments using human lung microvascular endothelial cells (HLMVECs), human hepatocytes, and perfusate (Steen solution). Four experimental groups: 1) fresh Steen (negative controls, NC), 2) EVLP'ed Steen control, this solution collected after 12 h of EVLP of human lungs, 3) hepatocyte conditioned EVLP'ed Steen (Hep-cond.), and 4) a RAGE inhibitor added in EVLP'ed Steen (RAGE inhibitor). HLMVECs were incubated in each testing condition and exposed to hypoxia (1% O2/8% CO2) for 24 h. Media were collected to investigate NF-κB signaling and endothelial glycocalyx damage. RESULTS HLMVECs incubated under hypoxia in EVLP'ed Steen showed significantly upregulated NF-κB signal and endothelial damage denoted by increased glycosaminoglycans and matrix metalloproteinase-2 activity among the groups. The Hep-cond. solution significantly attenuated those findings, while the RAGE inhibitor attenuated the NF-κB signal but not endothelial glycocalyx damage. CONCLUSION Our study demonstrates that hepatic function incorporated into EVLP can ameliorate pulmonary endothelial cells injury under hypoxic normothermic perfusion exposure. Our data supports the concept of incorporating other organ functions into an organ perfusion platform, to enhance lung graft preservation.
Collapse
Affiliation(s)
- Kentaro Noda
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center Health System, Pittsburgh, PA, USA
| | - Neha Atale
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center Health System, Pittsburgh, PA, USA
| | - Taylor Austin
- Department of Surgery, University of Pittsburgh Medical Center Health System, Pittsburgh, PA, USA
| | - David A Geller
- Department of Surgery, University of Pittsburgh Medical Center Health System, Pittsburgh, PA, USA
| | - Jorg Gerlach
- Department of Surgery, University of Pittsburgh Medical Center Health System, Pittsburgh, PA, USA
- Departments of Surgery and Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pablo G Sanchez
- Departments of Surgery and Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Section of Thoracic Surgery, Department of Surgery, University of Chicago Medicine & Biological Sciences, Chicago, IL, USA
| |
Collapse
|
3
|
Xie D, Lu G, Mai G, Guo Q, Xu G. Tissue-resident memory T cells in diseases and therapeutic strategies. MedComm (Beijing) 2025; 6:e70053. [PMID: 39802636 PMCID: PMC11725047 DOI: 10.1002/mco2.70053] [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: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are crucial components of the immune system that provide rapid, localized responses to recurrent pathogens at mucosal and epithelial barriers. Unlike circulating memory T cells, TRM cells are located within peripheral tissues, and they play vital roles in antiviral, antibacterial, and antitumor immunity. Their unique retention and activation mechanisms, including interactions with local epithelial cells and the expression of adhesion molecules, enable their persistence and immediate functionality in diverse tissues. Recent advances have revealed their important roles in chronic inflammation, autoimmunity, and cancer, illuminating both their protective and their pathogenic potential. This review synthesizes current knowledge on TRM cells' molecular signatures, maintenance pathways, and functional dynamics across different tissues. We also explore the interactions of TRM cells with other immune cells, such as B cells, macrophages, and dendritic cells, highlighting the complex network that underpins the efficacy of TRM cells in immune surveillance and response. Understanding the nuanced regulation of TRM cells is essential for developing targeted therapeutic strategies, including vaccines and immunotherapies, to enhance their protective roles while mitigating adverse effects. Insights into TRM cells' biology hold promise for innovative treatments for infectious diseases, cancer, and autoimmune conditions.
Collapse
Affiliation(s)
- Daoyuan Xie
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Guanting Lu
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Gang Mai
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaAcademy of Chinese Medical SciencesBeijingChina
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research UnitThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
4
|
Calabrese DR, Ekstrand CA, Yellamilli S, Singer JP, Hays SR, Leard LE, Shah RJ, Venado A, Kolaitis NA, Perez A, Combes A, Greenland JR. Macrophage and CD8 T cell discordance are associated with acute lung allograft dysfunction progression. J Heart Lung Transplant 2024; 43:1074-1086. [PMID: 38367738 PMCID: PMC11230518 DOI: 10.1016/j.healun.2024.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Acute lung allograft dysfunction (ALAD) is an imprecise syndrome denoting concern for the onset of chronic lung allograft dysfunction (CLAD). Mechanistic biomarkers are needed that stratify risk of ALAD progression to CLAD. We hypothesized that single cell investigation of bronchoalveolar lavage (BAL) cells at the time of ALAD would identify immune cells linked to progressive graft dysfunction. METHODS We prospectively collected BAL from consenting lung transplant recipients for single cell RNA sequencing. ALAD was defined by a ≥10% decrease in FEV1 not caused by infection or acute rejection and samples were matched to BAL from recipients with stable lung function. We examined cell compositional and transcriptional differences across control, ALAD with decline, and ALAD with recovery groups. We also assessed cell-cell communication. RESULTS BAL was assessed for 17 ALAD cases with subsequent decline (ALAD declined), 13 ALAD cases that resolved (ALAD recovered), and 15 cases with stable lung function. We observed broad differences in frequencies of the 26 unique cell populations across groups (p = 0.02). A CD8 T cell (p = 0.04) and a macrophage cluster (p = 0.01) best identified ALAD declined from the ALAD recovered and stable groups. This macrophage cluster was distinguished by an anti-inflammatory signature and the CD8 T cell cluster resembled a Tissue Resident Memory subset. Anti-inflammatory macrophages signaled to activated CD8 T cells via class I HLA, fibronectin, and galectin pathways (p < 0.05 for each). Recipients with discordance between these cells had a nearly 5-fold increased risk of severe graft dysfunction or death (HR 4.6, 95% CI 1.1-19.2, adjusted p = 0.03). We validated these key findings in 2 public lung transplant genomic datasets. CONCLUSIONS BAL anti-inflammatory macrophages may protect against CLAD by suppressing CD8 T cells. These populations merit functional and longitudinal assessment in additional cohorts.
Collapse
Affiliation(s)
- Daniel R Calabrese
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California.
| | | | - Shivaram Yellamilli
- Department of Pathology, University of California, San Francisco, California
| | - Jonathan P Singer
- Department of Medicine, University of California, San Francisco, California
| | - Steven R Hays
- Department of Medicine, University of California, San Francisco, California
| | - Lorriana E Leard
- Department of Medicine, University of California, San Francisco, California
| | - Rupal J Shah
- Department of Medicine, University of California, San Francisco, California
| | - Aida Venado
- Department of Medicine, University of California, San Francisco, California
| | | | - Alyssa Perez
- Department of Medicine, University of California, San Francisco, California
| | - Alexis Combes
- Department of Pathology, University of California, San Francisco, California
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California
| |
Collapse
|
5
|
Bell PT, Gelman AE. Alveolar macrophage-CD8 T cell interactions after acute lung allograft dysfunction: Insights from single-cell RNA sequencing. J Heart Lung Transplant 2024; 43:1087-1089. [PMID: 38490571 DOI: 10.1016/j.healun.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Affiliation(s)
- Peter T Bell
- Frazer Institute, at the Translational Research Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, St Louis, Missouri.
| |
Collapse
|
6
|
Schworer SA, Moran TP. Pulmonary Interstitial Macrophages stIMulate Regulatory T-Cell Responses. Am J Respir Cell Mol Biol 2024; 70:429-430. [PMID: 38445963 PMCID: PMC11160415 DOI: 10.1165/rcmb.2024-0049ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/05/2024] [Indexed: 03/07/2024] Open
Affiliation(s)
- Stephen A Schworer
- Department of Medicine University of North Carolina School of Medicine Chapel Hill, North Carolina
| | - Timothy P Moran
- Department of Pediatrics University of North Carolina School of Medicine Chapel Hill, North Carolina
| |
Collapse
|
7
|
Saito T, Fujino N, Kyogoku Y, Yamada M, Okutomo K, Ono Y, Konno S, Endo T, Itakura K, Matsumoto S, Sano H, Aizawa H, Numakura T, Onodera K, Okada Y, Hussell T, Ichinose M, Sugiura H. Identification of Siglec-1-negative alveolar macrophages with proinflammatory phenotypes in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2024; 326:L672-L686. [PMID: 38530936 DOI: 10.1152/ajplung.00303.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 03/28/2024] Open
Abstract
Alveolar macrophages (AMs) in patients with chronic obstructive pulmonary disease (COPD) orchestrate persistent inflammation in the airway. However, subpopulations of AMs participating in chronic inflammation have been poorly characterized. We previously reported that Siglec-1 expression on AMs, which is important for bacteria engulfment, was decreased in COPD. Here, we show that Siglec-1-negative AMs isolated from COPD lung tissues exhibit a proinflammatory phenotype and are associated with poor clinical outcomes in patients with COPD. Using flow cytometry, we segregated three subsets of AMs based on the expression of Siglec-1 and their side scattergram (SSC) and forward scattergram (FSC) properties: Siglec-1+SSChiFSChi, Siglec-1-SSChiFSChi, and Siglec-1-SSCloFSClo subsets. The Siglec-1-SSCloFSClo subset number was increased in COPD. RNA sequencing revealed upregulation of multiple proinflammatory signaling pathways and emphysema-associated matrix metalloproteases in the Siglec-1-SSCloFSClo subset. Gene set enrichment analysis indicated that the Siglec-1-SSCloFSClo subset adopted intermediate phenotypes between monocytes and mature alveolar macrophages. Functionally, these cells produced TNF-α, IL-6, and IL-8 at baseline, and these cytokines were significantly increased in response to viral RNA. The increase in Siglec-1-negative AMs in induced sputum is associated with future exacerbation risk and lung function decline in patients with COPD. Collectively, the novel Siglec-1-SSCloFSClo subset of AMs displays proinflammatory properties, and their emergence in COPD airways may be associated with poor clinical outcomes.NEW & NOTEWORTHY Alveolar macrophages (AMs) in patients with chronic obstructive pulmonary disease (COPD) orchestrate persistent inflammation in the airway. We find that Siglec-1-negative alveolar macrophages have a wide range of proinflammatory landscapes and a protease-expressing phenotype. Moreover, this subset is associated with the pathogenesis of COPD and responds to viral stimuli.
Collapse
Affiliation(s)
- Takuya Saito
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoya Fujino
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yorihiko Kyogoku
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koji Okutomo
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshinao Ono
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuichi Konno
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuto Endo
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koji Itakura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuichiro Matsumoto
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirohito Sano
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Aizawa
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tadahisa Numakura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Katsuhiro Onodera
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Tracy Hussell
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| | | | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
8
|
Camp B, Jorde I, Sittel F, Pausder A, Jeron A, Bruder D, Schreiber J, Stegemann-Koniszewski S. Comprehensive analysis of lung macrophages and dendritic cells in two murine models of allergic airway inflammation reveals model- and subset-specific accumulation and phenotypic alterations. Front Immunol 2024; 15:1374670. [PMID: 38529288 PMCID: PMC10961404 DOI: 10.3389/fimmu.2024.1374670] [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: 01/22/2024] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Allergic asthma has been mainly attributed to T helper type 2 (Th2) and proinflammatory responses but many cellular processes remain elusive. There is increasing evidence for distinct roles for macrophage and dendritic cell (DC) subsets in allergic airway inflammation (AAI). At the same time, there are various mouse models for allergic asthma that have been of utmost importance in identifying key inflammatory pathways in AAI but that differ in the allergen and/or route of sensitization. It is unclear whether and how the accumulation and activation of specialized macrophage and DC subsets depend on the experimental model chosen for analyses. Methods In our study, we employed high-parameter spectral flow cytometry to comprehensively assess the accumulation and phenotypic alterations of different macrophage- and DC-subsets in the lung in an OVA- and an HDM-mediated mouse model of AAI. Results We observed subset-specific as well as model-specific characteristics with respect to cell numbers and functional marker expression. Generally, alveolar as opposed to interstitial macrophages showed increased MHCII surface expression in AAI. Between the models, we observed significantly increased numbers of alveolar macrophages, CD103+ DC and CD11b+ DC in HDM-mediated AAI, concurrent with significantly increased airway interleukin-4 but decreased total serum IgE levels. Further, increased expression of CD80 and CD86 on DC was exclusively detected in HDM-mediated AAI. Discussion Our study demonstrates a model-specific involvement of macrophage and DC subsets in AAI. It further highlights spectral flow cytometry as a valuable tool for their comprehensive analysis under inflammatory conditions in the lung.
Collapse
Affiliation(s)
- Belinda Camp
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Ilka Jorde
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Franka Sittel
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Alexander Pausder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andreas Jeron
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jens Schreiber
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Sabine Stegemann-Koniszewski
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
9
|
Bondonese A, Craig A, Fan L, Valenzi E, Bain W, Lafyatis R, Sembrat J, Chen K, Snyder ME. Impact of enzymatic digestion on single cell suspension yield from peripheral human lung tissue. Cytometry A 2023; 103:777-785. [PMID: 37449375 PMCID: PMC10592386 DOI: 10.1002/cyto.a.24777] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/26/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
An increasing number of translational investigations of lung biology rely on analyzing single cell suspensions obtained from human lungs. To obtain these single cell suspensions, human lungs from biopsies or research-consented organ donors must be subjected to mechanical and enzymatic digestion prior to analysis with either flow cytometry or single cell RNA sequencing. A variety of enzymes have been used to perform tissue digestion, each with potential limitations. To better understand the limitations of each enzymatic digestion protocol and to establish a framework for comparing studies across protocols, we performed five commonly published protocols in parallel from identical samples obtained from 6 human lungs. Following mechanical (gentleMACS™) and enzymatic digestion, we quantified cell count and viability using a Nexcelom Cellometer and determined cell phenotype using multiparameter spectral flow cytometry (Cytek™ Aurora). We found that all protocols were superior in cellular yield and viability when compared to mechanical digestion alone. Protocols high in dispase cleaved immune markers CD4, CD8, CD69, and CD103 and contributed to an increased monocyte to macrophage yield. Similarly, dispase led to a differential epithelial cell yield, with increased TSPN8+ and ITGA6+ epithelial cells and reduced CD66e+ cells. When compared to collagenase D, collagenase P protocols yielded increased AT1 and AT2 cells and decreased endothelial cells. These results provide a framework for selecting an enzymatic digestion protocol best suited to the scientific question and allow for comparison of studies using different protocols.
Collapse
Affiliation(s)
| | - Andrew Craig
- Department of Medicine, University of Pittsburgh
| | - Li Fan
- Department of Medicine, University of Pittsburgh
| | | | - William Bain
- Department of Medicine, University of Pittsburgh
| | | | - John Sembrat
- Department of Medicine, University of Pittsburgh
| | - Kong Chen
- Department of Medicine, University of Pittsburgh
| | - Mark E. Snyder
- Department of Medicine, University of Pittsburgh
- Department of Immunology, University of Pittsburgh
- Starzl Transplantation Institute
| |
Collapse
|
10
|
Wang Y, Xiang X, Chen H, Zhou L, Chen S, Zhang G, Liu X, Ren X, Liu J, Kuang M, Jiang J, She J, Zhang Z, Xue R, Jiang H, Wang J, Peng S. Intratumoral erythroblastic islands restrain anti-tumor immunity in hepatoblastoma. Cell Rep Med 2023; 4:101044. [PMID: 37196629 DOI: 10.1016/j.xcrm.2023.101044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/28/2022] [Accepted: 04/19/2023] [Indexed: 05/19/2023]
Abstract
Erythroblastic islands (EBIs) are the specialized structures for erythropoiesis, but they have never been found functional in tumors. As the most common pediatric liver malignancy, hepatoblastoma (HB) requires more effective and safer therapies to prevent progression and the lifelong impact of complications on young children. However, developing such therapies is impeded by a lack of comprehensive understanding of the tumor microenvironment. By single-cell RNA sequencing of 13 treatment-naive HB patients, we discover an immune landscape characterized by aberrant accumulation of EBIs, formed by VCAM1+ macrophages and erythroid cells, which is inversely correlated with survival of HB. Erythroid cells inhibit the function of dendritic cells (DCs) via the LGALS9/TIM3 axis, leading to impaired anti-tumor T cell immune responses. Encouragingly, TIM3 blockades relieve the inhibitory effect of erythroid cells on DCs. Our study provides an immune evasion mechanism mediated by intratumoral EBIs and proposes TIM3 as a promising therapeutic target for HB.
Collapse
Affiliation(s)
- Yuanqi Wang
- Department of Liver Surgery, Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao Xiang
- Department of Liver Surgery, Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huadong Chen
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Luyao Zhou
- Division of Interventional Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuling Chen
- Division of Interventional Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guopei Zhang
- Department of Liver Surgery, Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofei Liu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuxin Ren
- Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Juncheng Liu
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Kuang
- Department of Liver Surgery, Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Interventional Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Juan Jiang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinbiao She
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhichong Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruidong Xue
- Translational Cancer Research, Peking University First Hospital, Beijing, China
| | - Hong Jiang
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Ji Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Sui Peng
- Department of Liver Surgery, Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
11
|
Wang C, Hyams B, Allen NC, Cautivo K, Monahan K, Zhou M, Dahlgren MW, Lizama CO, Matthay M, Wolters P, Molofsky AB, Peng T. Dysregulated lung stroma drives emphysema exacerbation by potentiating resident lymphocytes to suppress an epithelial stem cell reservoir. Immunity 2023; 56:576-591.e10. [PMID: 36822205 PMCID: PMC10578134 DOI: 10.1016/j.immuni.2023.01.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/29/2022] [Accepted: 01/30/2023] [Indexed: 02/24/2023]
Abstract
Aberrant tissue-immune interactions are the hallmark of diverse chronic lung diseases. Here, we sought to define these interactions in emphysema, a progressive disease characterized by infectious exacerbations and loss of alveolar epithelium. Single-cell analysis of human emphysema lungs revealed the expansion of tissue-resident lymphocytes (TRLs). Murine studies identified a stromal niche for TRLs that expresses Hhip, a disease-variant gene downregulated in emphysema. Stromal-specific deletion of Hhip induced the topographic expansion of TRLs in the lung that was mediated by a hyperactive hedgehog-IL-7 axis. 3D immune-stem cell organoids and animal models of viral exacerbations demonstrated that expanded TRLs suppressed alveolar stem cell growth through interferon gamma (IFNγ). Finally, we uncovered an IFNγ-sensitive subset of human alveolar stem cells that was preferentially lost in emphysema. Thus, we delineate a stromal-lymphocyte-epithelial stem cell axis in the lung that is modified by a disease-variant gene and confers host susceptibility to emphysema.
Collapse
Affiliation(s)
- Chaoqun Wang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Ben Hyams
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nancy C Allen
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelly Cautivo
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kiara Monahan
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Minqi Zhou
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Madelene W Dahlgren
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carlos O Lizama
- Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael Matthay
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Paul Wolters
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ari B Molofsky
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Tien Peng
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
12
|
Noda K, Chan EG, Furukawa M, Ryan JP, Clifford S, Luketich JD, Sanchez PG. Single-center experience of ex vivo lung perfusion and subsequent lung transplantation. Clin Transplant 2023; 37:e14901. [PMID: 36588340 DOI: 10.1111/ctr.14901] [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: 09/30/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND The safety of lung transplantation using ex vivo lung perfusion (EVLP) has been confirmed in multiple clinical studies; however, limited evidence is currently available regarding the potential effects of EVLP on posttransplant graft complications and survival with mid- to long-term follow-up. In this study, we reviewed our institutional data to better understand the impact of EVLP. METHODS Lungs placed on EVLP from 2014 through 2020 and transplant outcomes were retrospectively analyzed. Data were compared between lungs transplanted and declined after EVLP, between patients with severe primary graft dysfunction (PGD3) and no PGD3 after EVLP, and between matched patients with lungs transplanted with and without EVLP. RESULTS In total, 98 EVLP cases were performed. Changes in metabolic indicators during EVLP were correlated with graft quality and transplantability, but not changes in physiological parameters. Among 58 transplanted lungs after EVLP, PGD3 at 72 h occurred in 36.9% and was associated with preservation time, mechanical support prior to transplant, and intraoperative transfusion volume. Compared with patients without EVLP, patients who received lungs screened with EVLP had a higher incidence of PGD3 and longer ICU and hospital stays. Lung grafts placed on EVLP exhibited a significantly higher chance of developing airway anastomotic ischemic injury by 30 days posttransplant. Acute and chronic graft rejection, pulmonary function, and posttransplant survival were not different between patients with lungs screened on EVLP versus lungs with no EVLP. CONCLUSION EVLP use is associated with an increase of early posttransplant adverse events, but graft functional outcomes and patient survival are preserved.
Collapse
Affiliation(s)
- Kentaro Noda
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ernest G Chan
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Masashi Furukawa
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John P Ryan
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sarah Clifford
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - James D Luketich
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pablo G Sanchez
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
13
|
Jiang J, Huang Y, Zeng Z, Zhao C. Harnessing Engineered Immune Cells and Bacteria as Drug Carriers for Cancer Immunotherapy. ACS NANO 2023; 17:843-884. [PMID: 36598956 DOI: 10.1021/acsnano.2c07607] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Immunotherapy continues to be in the spotlight of oncology therapy research in the past few years and has been proven to be a promising option to modulate one's innate and adaptive immune systems for cancer treatment. However, the poor delivery efficiency of immune agents, potential off-target toxicity, and nonimmunogenic tumors significantly limit its effectiveness and extensive application. Recently, emerging biomaterial-based drug carriers, including but not limited to immune cells and bacteria, are expected to be potential candidates to break the dilemma of immunotherapy, with their excellent natures of intrinsic tumor tropism and immunomodulatory activity. More than that, the tiny vesicles and physiological components derived from them have similar functions with their source cells due to the inheritance of various surface signal molecules and proteins. Herein, we presented representative examples about the latest advances of biomaterial-based delivery systems employed in cancer immunotherapy, including immune cells, bacteria, and their derivatives. Simultaneously, opportunities and challenges of immune cells and bacteria-based carriers are discussed to provide reference for their future application in cancer immunotherapy.
Collapse
Affiliation(s)
- Jingwen Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Zishan Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| |
Collapse
|
14
|
Barber PR, Mustapha R, Flores-Borja F, Alfano G, Ng K, Weitsman G, Dolcetti L, Suwaidan AA, Wong F, Vicencio JM, Galazi M, Opzoomer JW, Arnold JN, Thavaraj S, Kordasti S, Doyle J, Greenberg J, Dillon MT, Harrington KJ, Forster M, Coolen ACC, Ng T. Predicting progression-free survival after systemic therapy in advanced head and neck cancer: Bayesian regression and model development. eLife 2022; 11:e73288. [PMID: 36562609 PMCID: PMC9815805 DOI: 10.7554/elife.73288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/22/2022] [Indexed: 12/24/2022] Open
Abstract
Background Advanced head and neck squamous cell carcinoma (HNSCC) is associated with a poor prognosis, and biomarkers that predict response to treatment are highly desirable. The primary aim was to predict progression-free survival (PFS) with a multivariate risk prediction model. Methods Experimental covariates were derived from blood samples of 56 HNSCC patients which were prospectively obtained within a Phase 2 clinical trial (NCT02633800) at baseline and after the first treatment cycle of combined platinum-based chemotherapy with cetuximab treatment. Clinical and experimental covariates were selected by Bayesian multivariate regression to form risk scores to predict PFS. Results A 'baseline' and a 'combined' risk prediction model were generated, each of which featuring clinical and experimental covariates. The baseline risk signature has three covariates and was strongly driven by baseline percentage of CD33+CD14+HLADRhigh monocytes. The combined signature has six covariates, also featuring baseline CD33+CD14+HLADRhigh monocytes but is strongly driven by on-treatment relative change of CD8+ central memory T cells percentages. The combined model has a higher predictive power than the baseline model and was successfully validated to predict therapeutic response in an independent cohort of nine patients from an additional Phase 2 trial (NCT03494322) assessing the addition of avelumab to cetuximab treatment in HNSCC. We identified tissue counterparts for the immune cells driving the models, using imaging mass cytometry, that specifically colocalized at the tissue level and correlated with outcome. Conclusions This immune-based combined multimodality signature, obtained through longitudinal peripheral blood monitoring and validated in an independent cohort, presents a novel means of predicting response early on during the treatment course. Funding Daiichi Sankyo Inc, Cancer Research UK, EU IMI2 IMMUCAN, UK Medical Research Council, European Research Council (335326), Merck Serono. Cancer Research Institute, National Institute for Health Research, Guy's and St Thomas' NHS Foundation Trust and The Institute of Cancer Research. Clinical trial number NCT02633800.
Collapse
Affiliation(s)
- Paul R Barber
- UCL Cancer Institute, Paul O'Gorman Building, University College LondonLondonUnited Kingdom
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, King’s College LondonLondonUnited Kingdom
| | - Rami Mustapha
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUnited Kingdom
| | - Fabian Flores-Borja
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College LondonLondonUnited Kingdom
| | - Giovanna Alfano
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUnited Kingdom
| | - Kenrick Ng
- UCL Cancer Institute, Paul O'Gorman Building, University College LondonLondonUnited Kingdom
| | - Gregory Weitsman
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUnited Kingdom
| | - Luigi Dolcetti
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUnited Kingdom
| | - Ali Abdulnabi Suwaidan
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUnited Kingdom
| | - Felix Wong
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUnited Kingdom
| | - Jose M Vicencio
- UCL Cancer Institute, Paul O'Gorman Building, University College LondonLondonUnited Kingdom
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUnited Kingdom
| | - Myria Galazi
- UCL Cancer Institute, Paul O'Gorman Building, University College LondonLondonUnited Kingdom
| | - James W Opzoomer
- Tumor Immunology Group, School of Cancer & Pharmaceutical Sciences, King’s College LondonLondonUnited Kingdom
| | - James N Arnold
- Tumor Immunology Group, School of Cancer & Pharmaceutical Sciences, King’s College LondonLondonUnited Kingdom
| | - Selvam Thavaraj
- Centre for Clinical, Oral & Translational Science, King’s College LondonLondonUnited Kingdom
| | - Shahram Kordasti
- Systems Cancer Immunology, School of Cancer & Pharmaceutical Sciences, King’s College LondonLondonUnited Kingdom
| | - Jana Doyle
- Daiichi Sankyo IncorporatedNewarkUnited States
| | | | | | | | - Martin Forster
- UCL Cancer Institute, Paul O'Gorman Building, University College LondonLondonUnited Kingdom
| | - Anthony CC Coolen
- Institute for Mathematical and Molecular Biomedicine, King’s College LondonLondonUnited Kingdom
- Saddle Point Science LtdLondonUnited Kingdom
| | - Tony Ng
- UCL Cancer Institute, Paul O'Gorman Building, University College LondonLondonUnited Kingdom
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUnited Kingdom
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College LondonLondonUnited Kingdom
| |
Collapse
|
15
|
Characteristic Genes and Immune Infiltration Analysis for Acute Rejection after Kidney Transplantation. DISEASE MARKERS 2022; 2022:6575052. [PMID: 36393969 PMCID: PMC9646319 DOI: 10.1155/2022/6575052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Background Renal transplantation can significantly improve the survival rate and quality of life of patients with end-stage renal disease, but the probability of acute rejection (AR) in adult renal transplant recipients is still approximately 12.2%. Machine learning (ML) is superior to traditional statistical methods in various clinical scenarios. However, the current AR model is constructed only through simple difference analysis or a single queue, which cannot guarantee the accuracy of prediction. Therefore, this study identified and validated new gene sets that contribute to the early prediction of AR and the prognosis prediction of patients after renal transplantation by constructing a more accurate AR gene signature through ML technology. Methods Based on the Gene Expression Omnibus (GEO) database and multiple bioinformatic analyses, we identified differentially expressed genes (DEGs) and built a gene signature via LASSO regression and SVM analysis. Immune cell infiltration and immunocyte association analyses were also conducted. Furthermore, we investigated the relationship between AR genes and graft survival status. Results Twenty-four DEGs were identified. A 5 gene signature (CPA6, EFNA1, HBM, THEM5, and ZNF683) were obtained by LASSO analysis and SVM analysis, which had a satisfied ability to differentiate AR and NAR in the training cohort, internal validation cohort and external validation cohort. Additionally, ZNF683 was associated with graft survival. Conclusion A 5 gene signature, particularly ZNF683, provided insight into a precise therapeutic schedule and clinical applications for AR patients.
Collapse
|
16
|
Tian Y, Wang H, Guan W, Zhang X, Sun Y, Qian C, Song X, Peng B, Cui X. GBP2 serves as a novel prognostic biomarker and potential immune microenvironment indicator in renal cell carcinoma. Mol Carcinog 2022; 61:1082-1098. [PMID: 36222186 DOI: 10.1002/mc.23447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/18/2022] [Accepted: 06/29/2022] [Indexed: 11/11/2022]
Abstract
Since the application of immune checkpoint therapy (ICT) has gradually become a new strategy for clear cell renal cell carcinoma (ccRCC) treatment, biomarkers that predict the individual response to ICT is needed. This study aimed to identify a new clinical indicator for postoperative surveillance of ccRCC and prediction of ICT response. We investigated the GBP2 expression and its relation with immune cell infiltration in tumor microenvironment using public databases, clinical specimens and ccRCC cell lines. Bioinformatic analysis using public database revealed that GBP2 expression is higher in cancer tissues than in adherent normal tissues among different cancer types including ccRCC, and the same results were acquired from clinical tissue samples tested by Western Blot and PCR. In ccRCC cell lines, CCk-8 proliferation assay and apoptosis assessment suggested GBP2 facilitates the malignancy of ccRCC. 286 ccRCC patients were randomly divided into a training or validation cohort, and immunohistochemistry (IHC) and Kaplan-Meier analysis revealed that higher GBP2 expression is related to worse prognosis. C-index analysis implied that integrating GBP2 expression with TNM stage improved the accuracy in predicting prognosis of ccRCC patients compared to the solitary use of either. Bioinformatic analysis implied a relation between GBP2 and immunity, and GBP2 expression is positively related with suppressive immune markers in ccRCC microenvironment. Taken together, our study demonstrated the potential of GBP2 to sever as a prognostic predictor of ccRCC, and an association between GBP2 and tumor-infiltrating lymphocytes in ccRCC was observed, making it a promising indicator of ICT response.
Collapse
Affiliation(s)
- Yijun Tian
- Department of Urinary Surgery, The Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Hongru Wang
- Department of Urinary Surgery, The Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Wenbin Guan
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiangmin Zhang
- Department of Urinary Surgery, Luodian Hospital, Shanghai, China
| | - Ye Sun
- Department of Urinary Surgery, Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Cheng Qian
- Department of Urinary Surgery, Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xu Song
- Department of Urology, Shanghai Seventh People's Hospital, Shanghai, China
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xingang Cui
- Department of Urinary Surgery, The Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China.,Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
17
|
Liu Z, Liao F, Zhu J, Zhou D, Heo GS, Leuhmann HP, Scozzi D, Parks A, Hachem R, Byers DE, Tague LK, Kulkarni HS, Cano M, Wong BW, Li W, Huang HJ, Krupnick AS, Kreisel D, Liu Y, Gelman AE. Reprogramming alveolar macrophage responses to TGF-β reveals CCR2+ monocyte activity that promotes bronchiolitis obliterans syndrome. J Clin Invest 2022; 132:159229. [PMID: 36189800 PMCID: PMC9525120 DOI: 10.1172/jci159229] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is a major impediment to lung transplant survival and is generally resistant to medical therapy. Extracorporeal photophoresis (ECP) is an immunomodulatory therapy that shows promise in stabilizing BOS patients, but its mechanisms of action are unclear. In a mouse lung transplant model, we show that ECP blunts alloimmune responses and inhibits BOS through lowering airway TGF-β bioavailability without altering its expression. Surprisingly, ECP-treated leukocytes were primarily engulfed by alveolar macrophages (AMs), which were reprogrammed to become less responsive to TGF-β and reduce TGF-β bioavailability through secretion of the TGF-β antagonist decorin. In untreated recipients, high airway TGF-β activity stimulated AMs to express CCL2, leading to CCR2+ monocyte-driven BOS development. Moreover, we found TGF-β receptor 2-dependent differentiation of CCR2+ monocytes was required for the generation of monocyte-derived AMs, which in turn promoted BOS by expanding tissue-resident memory CD8+ T cells that inflicted airway injury through Blimp-1-mediated granzyme B expression. Thus, through studying the effects of ECP, we have identified an AM functional plasticity that controls a TGF-β-dependent network that couples CCR2+ monocyte recruitment and differentiation to alloimmunity and BOS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ramsey Hachem
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Derek E. Byers
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laneshia K. Tague
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hrishikesh S. Kulkarni
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marlene Cano
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Howard J. Huang
- Houston Methodist J.C. Walter Jr. Transplant Center, Houston, Texas, USA
| | - Alexander S. Krupnick
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Daniel Kreisel
- Department of Surgery
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yongjian Liu
- Houston Methodist J.C. Walter Jr. Transplant Center, Houston, Texas, USA
| | - Andrew E. Gelman
- Department of Surgery
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
18
|
Aegerter H, Lambrecht BN, Jakubzick CV. Biology of lung macrophages in health and disease. Immunity 2022; 55:1564-1580. [PMID: 36103853 DOI: 10.1016/j.immuni.2022.08.010] [Citation(s) in RCA: 282] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 12/14/2022]
Abstract
Tissue-resident alveolar and interstitial macrophages and recruited macrophages are critical players in innate immunity and maintenance of lung homeostasis. Until recently, assessing the differential functional contributions of tissue-resident versus recruited macrophages has been challenging because they share overlapping cell surface markers, making it difficult to separate them using conventional methods. This review describes how scRNA-seq and spatial transcriptomics can separate these subpopulations and help unravel the complexity of macrophage biology in homeostasis and disease. First, we provide a guide to identifying and distinguishing lung macrophages from other mononuclear phagocytes in humans and mice. Second, we outline emerging concepts related to the development and function of the various lung macrophages in the alveolar, perivascular, and interstitial niches. Finally, we describe how different tissue states profoundly alter their functions, including acute and chronic lung disease, cancer, and aging.
Collapse
Affiliation(s)
- Helena Aegerter
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent 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-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, ErasmusMC, Rotterdam, the Netherlands
| | - Claudia V Jakubzick
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA.
| |
Collapse
|
19
|
Zhang M, Li N, He Y, Shi T, Jie Z. Pulmonary resident memory T cells in respiratory virus infection and their inspiration on therapeutic strategies. Front Immunol 2022; 13:943331. [PMID: 36032142 PMCID: PMC9412965 DOI: 10.3389/fimmu.2022.943331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
The immune system generates memory cells on infection with a virus for the first time. These memory cells play an essential role in protection against reinfection. Tissue-resident memory T (TRM) cells can be generated in situ once attacked by pathogens. TRM cells dominate the defense mechanism during early stages of reinfection and have gradually become one of the most popular focuses in recent years. Here, we mainly reviewed the development and regulation of various TRM cell signaling pathways in the respiratory tract. Moreover, we explored the protective roles of TRM cells in immune response against various respiratory viruses, such as Respiratory Syncytial Virus (RSV) and influenza. The complex roles of TRM cells against SARS-CoV-2 infection are also discussed. Current evidence supports the therapeutic strategies targeting TRM cells, providing more possibilities for treatment. Rational utilization of TRM cells for therapeutics is vital for defense against respiratory viruses.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yanchao He
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
- *Correspondence: Zhijun Jie,
| |
Collapse
|
20
|
Rahimi RA, Cho JL, Jakubzick CV, Khader SA, Lambrecht BN, Lloyd CM, Molofsky AB, Talbot S, Bonham CA, Drake WP, Sperling AI, Singer BD. Advancing Lung Immunology Research: An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2022; 67:e1-18. [PMID: 35776495 PMCID: PMC9273224 DOI: 10.1165/rcmb.2022-0167st] [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: 11/24/2022] Open
Abstract
The mammalian airways and lungs are exposed to a myriad of inhaled particulate matter, allergens, and pathogens. The immune system plays an essential role in protecting the host from respiratory pathogens, but a dysregulated immune response during respiratory infection can impair pathogen clearance and lead to immunopathology. Furthermore, inappropriate immunity to inhaled antigens can lead to pulmonary diseases. A complex network of epithelial, neural, stromal, and immune cells has evolved to sense and respond to inhaled antigens, including the decision to promote tolerance versus a rapid, robust, and targeted immune response. Although there has been great progress in understanding the mechanisms governing immunity to respiratory pathogens and aeroantigens, we are only beginning to develop an integrated understanding of the cellular networks governing tissue immunity within the lungs and how it changes after inflammation and over the human life course. An integrated model of airway and lung immunity will be necessary to improve mucosal vaccine design as well as prevent and treat acute and chronic inflammatory pulmonary diseases. Given the importance of immunology in pulmonary research, the American Thoracic Society convened a working group to highlight central areas of investigation to advance the science of lung immunology and improve human health.
Collapse
|
21
|
Abstract
Tissue-resident immune cells span both myeloid and lymphoid cell lineages, have been found in multiple human tissues, and play integral roles at all stages of the immune response, from maintaining homeostasis to responding to infectious challenges to resolution of inflammation to tissue repair. In humans, studying immune cells and responses in tissues is challenging, although recent advances in sampling and high-dimensional profiling have provided new insights into the ontogeny, maintenance, and functional role of tissue-resident immune cells. Each tissue contains a specific complement of resident immune cells. Moreover, resident immune cells for each lineage share core properties, along with tissue-specific adaptations. Here we propose a five-point checklist for defining resident immune cell types in humans and describe the currently known features of resident immune cells, their mechanisms of development, and their putative functional roles within various human organs. We also consider these aspects of resident immune cells in the context of future studies and therapeutics.
Collapse
Affiliation(s)
- Joshua I Gray
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, USA;
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, USA;
- Department of Surgery, Columbia University Irving Medical Center, New York, USA
| |
Collapse
|
22
|
Oja AE, van Lier RAW, Hombrink P. Two sides of the same coin: Protective versus pathogenic CD4 + resident memory T cells. Sci Immunol 2022; 7:eabf9393. [PMID: 35394815 DOI: 10.1126/sciimmunol.abf9393] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The ability of the adaptive immune system to form memory is key to providing protection against secondary infections. Resident memory T cells (TRM) are specialized T cell populations that reside within tissue sites where they await reencounter with their cognate antigen. TRM are distinct from circulating memory cells, including central and effector memory T cells, both functionally and transcriptionally. Since the discovery of TRM, most research has focused on CD8+ TRM, despite that CD4+ TRM are also abundant in most tissues. In the past few years, more evidence has emerged that CD4+ TRM can contribute both protective and pathogenic roles in disease. A complexity inherent to the CD4+ TRM field is the ability of CD4+ T cells to polarize into a multitude of distinct subsets and recognize not only viruses and intracellular bacteria but also extracellular bacteria, fungi, and parasites. In this review, we outline the key features of CD4+ TRM in health and disease, including their contributions to protection against SARS-CoV-2 and potential contributions to immunopathology associated with COVID-19.
Collapse
Affiliation(s)
- Anna E Oja
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - René A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Pleun Hombrink
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
23
|
Snyder ME, Moghbeli K, Bondonese A, Craig A, Popescu I, Fan L, Tabib T, Lafyatis R, Chen K, Trejo Bittar HE, Lendermon E, Pilewski J, Johnson B, Kilaru S, Zhang Y, Sanchez PG, Alder JK, Sims PA, McDyer JF. Modulation of tissue resident memory T cells by glucocorticoids after acute cellular rejection in lung transplantation. J Exp Med 2022; 219:e20212059. [PMID: 35285873 PMCID: PMC8924935 DOI: 10.1084/jem.20212059] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/13/2021] [Accepted: 02/04/2022] [Indexed: 01/01/2023] Open
Abstract
Acute cellular rejection is common after lung transplantation and is associated with an increased risk of early chronic rejection. We present combined single-cell RNA and TCR sequencing on recipient-derived T cells obtained from the bronchoalveolar lavage of three lung transplant recipients with rejection and compare them with T cells obtained from the same patients after treatment of rejection with high-dose systemic glucocorticoids. At the time of rejection, we found an oligoclonal expansion of cytotoxic CD8+ T cells that all persisted as tissue resident memory T cells after successful treatment. Persisting CD8+ allograft-resident T cells have reduced gene expression for cytotoxic mediators after therapy with glucocorticoids but accumulate around airways. This clonal expansion is discordant with circulating T cell clonal expansion at the time of rejection, suggesting in situ expansion. We thus highlight the accumulation of cytotoxic, recipient-derived tissue resident memory T cells within the lung allograft that persist despite the administration of high-dose systemic glucocorticoids. The long-term clinical consequences of this persistence have yet to be characterized.
Collapse
Affiliation(s)
- Mark E. Snyder
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| | - Kaveh Moghbeli
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Anna Bondonese
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Andrew Craig
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Iulia Popescu
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Li Fan
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Tracy Tabib
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Robert Lafyatis
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Kong Chen
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Joseph Pilewski
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Bruce Johnson
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Silpa Kilaru
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY
| | - John F. McDyer
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
24
|
Zheng MZM, Wakim LM. Tissue resident memory T cells in the respiratory tract. Mucosal Immunol 2022; 15:379-388. [PMID: 34671115 PMCID: PMC8526531 DOI: 10.1038/s41385-021-00461-z] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/04/2023]
Abstract
Owing to their capacity to rapidly spread across the population, airborne pathogens represent a significant risk to global health. Indeed, several of the past major global pandemics have been instigated by respiratory pathogens. A greater understanding of the immune cells tasked with protecting the airways from infection will allow for the development of strategies that curb the spread and impact of these airborne diseases. A specific subset of memory T-cell resident in both the upper and lower respiratory tract, termed tissue-resident memory (Trm), have been shown to play an instrumental role in local immune responses against a wide breadth of both viral and bacterial infections. In this review, we discuss factors that influence respiratory tract Trm development, longevity, and immune surveillance and explore vaccination regimes that harness these cells, such approaches represent exciting new strategies that may be utilized to tackle the next global pandemic.
Collapse
Affiliation(s)
- Ming Z. M. Zheng
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Linda M. Wakim
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| |
Collapse
|
25
|
Yang Y, Wang Y. Autocrine, Paracrine, and Endocrine Signals That Can Alter Alveolar Macrophages Function. Rev Physiol Biochem Pharmacol 2022; 186:177-198. [PMID: 36472676 DOI: 10.1007/112_2022_76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alveolar macrophages (AMs) are extremely versatile cells with complex functions involved in health or diseases such as pneumonia, asthma, and pulmonary alveolar proteinosis. In recent years, it has been widely identified that the different functions and states of macrophages are the results from the complex interplay between microenvironmental signals and macrophage lineage. Diverse and complicated signals to which AMs respond are mentioned when they are described individually or in a particular state of AMs. In this review, the microenvironmental signals are divided into autocrine, paracrine, and endocrine signals based on their secreting characteristics. This new perspective on classification provides a more comprehensive and systematic introduction to the complex signals around AMs and is helpful for understanding the roles of AMs affected by physiological environment. The existing possible treatments of AMs are also mentioned in it. The thorough understanding of AMs signals modulation may be contributed to the development of more effective therapies for AMs-related lung diseases.
Collapse
Affiliation(s)
- Yue Yang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
26
|
Martin FP, Jacqueline C, Poschmann J, Roquilly A. Alveolar Macrophages: Adaptation to Their Anatomic Niche during and after Inflammation. Cells 2021; 10:cells10102720. [PMID: 34685700 PMCID: PMC8534884 DOI: 10.3390/cells10102720] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
At the early stages of life development, alveoli are colonized by embryonic macrophages, which become resident alveolar macrophages (ResAM) and self-sustain by local division. Genetic and epigenetic signatures and, to some extent, the functions of ResAM are dictated by the lung microenvironment, which uses cytokines, ligand-receptor interactions, and stroma cells to orchestrate lung homeostasis. In resting conditions, the lung microenvironment induces in ResAM a tolerogenic programming that prevents unnecessary and potentially harmful inflammation responses to the foreign bodies, which continuously challenge the airways. Throughout life, any episode of acute inflammation, pneumonia being likely the most frequent cause, depletes the pool of ResAM, leaving space for the recruitment of inflammatory monocytes that locally develop in monocyte-derived alveolar macrophages (InfAM). During lung infection, the local microenvironment induces a temporary inflammatory signature to the recruited InfAM to handle the tissue injury and eliminate the pathogens. After a few days, the recruited InfAM, which locally self-sustain and develop as new ResAM, gain profibrotic functions required for tissue healing. After the complete resolution of the infectious episode, the functional programming of both embryonic and monocyte-derived ResAM remains altered for months and possibly for the entire life. Adult lungs thus contain a wide diversity of ResAM since every infection brings new waves of InfAM which fill the room left open by the inflammatory process. The memory of these innate cells called trained immunity constitutes an immunologic scar left by inflammation, notably pneumonia. This memory of ResAM has advantages and drawbacks. In some cases, lung-trained immunity offers better defense capacities against autoimmune disorders and the long-term risk of infection. At the opposite, it can perpetuate a harmful process and lead to a pathological state, as is the case among critically ill patients who have immune paralysis and are highly susceptible to hospital-acquired pneumonia and acute respiratory distress syndrome. The progress in understanding the kinetics of response of alveolar macrophages (AM) to lung inflammation is paving the way to new treatments of pneumonia and lung inflammatory process.
Collapse
Affiliation(s)
- Florian Pierre Martin
- EA3826 Host Pathogen Interactions, Inflammation and Mucosal Immunity, Department of Anesthesiology and Intensive Medicine, Hôtel Dieu, CHU Nantes, University of Nantes, F-44000 Nantes, France; (F.P.M.); (C.J.)
| | - Cédric Jacqueline
- EA3826 Host Pathogen Interactions, Inflammation and Mucosal Immunity, Department of Anesthesiology and Intensive Medicine, Hôtel Dieu, CHU Nantes, University of Nantes, F-44000 Nantes, France; (F.P.M.); (C.J.)
| | - Jeremie Poschmann
- Centre de Recherche en Transplantation et Immunologie, University of Nantes, UMR 1064, ITUN, Inserm, F-44000 Nantes, France;
| | - Antoine Roquilly
- EA3826 Host Pathogen Interactions, Inflammation and Mucosal Immunity, Department of Anesthesiology and Intensive Medicine, Hôtel Dieu, CHU Nantes, University of Nantes, F-44000 Nantes, France; (F.P.M.); (C.J.)
- Correspondence: ; Tel.: +33-253482230
| |
Collapse
|
27
|
Humphries DC, O’Connor RA, Larocque D, Chabaud-Riou M, Dhaliwal K, Pavot V. Pulmonary-Resident Memory Lymphocytes: Pivotal Orchestrators of Local Immunity Against Respiratory Infections. Front Immunol 2021; 12:738955. [PMID: 34603321 PMCID: PMC8485048 DOI: 10.3389/fimmu.2021.738955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
There is increasing evidence that lung-resident memory T and B cells play a critical role in protecting against respiratory reinfection. With a unique transcriptional and phenotypic profile, resident memory lymphocytes are maintained in a quiescent state, constantly surveying the lung for microbial intruders. Upon reactivation with cognate antigen, these cells provide rapid effector function to enhance immunity and prevent infection. Immunization strategies designed to induce their formation, alongside novel techniques enabling their detection, have the potential to accelerate and transform vaccine development. Despite most data originating from murine studies, this review will discuss recent insights into the generation, maintenance and characterisation of pulmonary resident memory lymphocytes in the context of respiratory infection and vaccination using recent findings from human and non-human primate studies.
Collapse
Affiliation(s)
- Duncan C. Humphries
- Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh BioQuarter, The University of Edinburgh, Edinburgh, United Kingdom
- Sanofi Pasteur, R&D, Marcy l’Etoile, Lyon, France
| | - Richard A. O’Connor
- Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh BioQuarter, The University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Kevin Dhaliwal
- Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh BioQuarter, The University of Edinburgh, Edinburgh, United Kingdom
| | | |
Collapse
|
28
|
Soma S, Harriff MJ. Ex Vivo Lung Perfusion Provides New Insights into Human Lung-Resident Immune Cell Localization and Functional Interactions. Am J Respir Crit Care Med 2021; 203:1207-1208. [PMID: 33357022 PMCID: PMC8456468 DOI: 10.1164/rccm.202012-4358ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Shogo Soma
- Division of Pulmonary and Critical Care Medicine Oregon Health & Science University Portland, Oregon
| | - Melanie J Harriff
- Division of Pulmonary and Critical Care Medicine and.,Department of Molecular Microbiology and Immunology Oregon Health & Science University Portland, Oregon, and.,VA Portland Health Care System Portland, Oregon
| |
Collapse
|