1
|
Komaru Y, Ning L, Lama C, Suresh A, Kefaloyianni E, Miller MJ, Kawana S, Shepherd HM, Li W, Kreisel D, Herrlich A. Acute kidney injury triggers hypoxemia by lung intravascular neutrophil retention that reduces capillary blood flow. J Clin Invest 2025; 135:e186705. [PMID: 40048367 PMCID: PMC12077900 DOI: 10.1172/jci186705] [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: 09/04/2024] [Accepted: 02/27/2025] [Indexed: 03/12/2025] Open
Abstract
Sterile acute kidney injury (AKI) is common in the clinic and frequently associated with unexplained hypoxemia that does not improve with dialysis. AKI induces remote lung inflammation with neutrophil recruitment in mice and humans, but which cellular cues establish neutrophilic inflammation and how it contributes to hypoxemia is not known. Here we report that AKI induced rapid intravascular neutrophil retention in lung alveolar capillaries without extravasation into tissue or alveoli, causing hypoxemia by reducing lung capillary blood flow in the absence of substantial lung interstitial or alveolar edema. In contrast to direct ischemic lung injury, lung neutrophil recruitment during remote lung inflammation did not require cues from intravascular nonclassical monocytes or tissue-resident alveolar macrophages. Instead, lung neutrophil retention depended on the neutrophil chemoattractant CXCL2 released by activated classical monocytes. Comparative single-cell RNA-Seq analysis of direct and remote lung inflammation revealed that alveolar macrophages were highly activated and produced CXCL2 only in direct lung inflammation. Establishing a CXCL2 gradient into the alveolus by intratracheal CXCL2 administration during AKI-induced remote lung inflammation enabled neutrophils to extravasate. We thus discovered important differences in lung neutrophil recruitment in direct versus remote lung inflammation and identified lung capillary neutrophil retention that negatively affected oxygenation by causing a ventilation-perfusion mismatch as a driver of AKI-induced hypoxemia.
Collapse
Affiliation(s)
| | - Liang Ning
- Division of Nephrology, Department of Medicine
| | - Carine Lama
- Division of Nephrology, Department of Medicine
| | | | | | | | | | | | | | - Daniel Kreisel
- Department of Surgery, and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andreas Herrlich
- Division of Nephrology, Department of Medicine
- VA Saint Louis Health Care System, John Cochran Division, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Arias E, Haynes ME, Nadkarni NA, Lipfert ZK, Muller WA, Batra A, Sullivan DP. EdU tracking of leukocyte recruitment in mouse models of ischemic stroke and sterile lung inflammation. J Cell Sci 2025; 138:jcs263835. [PMID: 40260638 DOI: 10.1242/jcs.263835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
The discovery of copper(I)-catalyzed azide-alkyne cycloaddition (click chemistry) has significantly advanced the detection of proliferating cells by utilizing 5-ethynyl-2'-deoxyuridine (EdU). EdU, a thymidine analogue, is incorporated into DNA during replication and detected by the direct reaction with an azide-conjugated fluorophore. Traditionally, dividing cells are labeled using 5-bromodeoxyuridine (BrdU), another nucleotide analogue. However, BrdU detection is a harsh method that requires substantial sample processing, unlike EdU detection. EdU is classically used to identify proliferating cells; however, we report a streamlined methodology that uses EdU to label and track leukocyte recruitment that is compatible with flow cytometry and microscopy and preserves transgenic fluorophores. EdU labeling was performed in two different models of sterile inflammation: ischemic stroke and hydrochloric acid aspiration. EdU injection was timed to differentially label circulating monocytes, neutrophils and T cells. Tissue analysis showed EdU-positive monocytes and T cells were enriched in both inflammatory models. This suggests that recently divided monocytes and T cells are preferentially recruited to these vascular beds during inflammation and highlights the utility of this labeling approach to track leukocyte subtypes longitudinally during inflammation.
Collapse
Affiliation(s)
- Erika Arias
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
| | - Maureen E Haynes
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
| | - Neil A Nadkarni
- Department of Neurology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Zoie K Lipfert
- Department of Neurology, Northwestern University, Chicago, IL 60643, USA
| | - William A Muller
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
| | - Ayush Batra
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
- Department of Neurology, Northwestern University, Chicago, IL 60643, USA
| | - David P Sullivan
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
| |
Collapse
|
3
|
Nelson NC, Wong KK, Mahoney IJ, Malik T, Rudym D, Lesko MB, Qayum S, Lewis TC, Chang SH, Chan JCY, Geraci TC, Li Y, Pamar P, Schnier J, Singh R, Collazo D, Chang M, Kyeremateng Y, McCormick C, Borghi S, Patel S, Darawshy F, Barnett CR, Sulaiman I, Kugler MC, Brosnahan SB, Singh S, Tsay JCJ, Wu BG, Pass HI, Angel LF, Segal LN, Natalini JG. Lung allograft dysbiosis associates with immune response and primary graft dysfunction. J Heart Lung Transplant 2025; 44:422-434. [PMID: 39561864 PMCID: PMC11956144 DOI: 10.1016/j.healun.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Lower airway enrichment with oral commensals has been previously associated with severe primary graft dysfunction (PGD) after lung transplantation (LT). We aimed to determine whether this dysbiotic signature is present across all PGD severity grades and whether it is associated with a distinct host inflammatory endotype. METHODS Lower airway samples from 96 LT recipients were used to evaluate the lung allograft microbiota via 16S rRNA gene sequencing. Bronchoalveolar lavage (BAL) cytokine concentrations and cell differential percentages were compared across PGD grades. In a subset of samples, we evaluated the lower airway host transcriptome using RNA sequencing methods. RESULTS Differential analyses demonstrated lower airway enrichment with supraglottic-predominant taxa (SPT) in moderate and severe PGD. Dirichlet multinomial mixtures modeling identified 2 distinct microbial clusters. A greater percentage of subjects with moderate-severe PGD than no PGD were identified within the dysbiotic cluster (C-SPT, 48% and 29%, respectively) though this did not reach statistical significance (p = 0.06). PGD severity associated with increased BAL neutrophil concentration (p = 0.03) and correlated with BAL concentrations of MCP-1/CCL2, IP-10/CXCL10, IL-10, and TNF-α (p < 0.05). Furthermore, signatures of dysbiosis correlated with neutrophils, MCP-1/CCL-2, IL-10, and TNF-α (p < 0.05). C-SPT exhibited differential expression of TNF, SERPINE1, MPO, and MMP1 genes and upregulation of MAPK pathways, host signling associated with neutrophilic inflammation. CONCLUSIONS Lower airway dysbiosis within the lung allograft is associated with a neutrophilic inflammatory endotype, an immune profile commonly recognized as the hallmark for PGD. These data highlight a putative role of lower airway microbial dysbiosis in the pathogenesis of this syndrome.
Collapse
Affiliation(s)
- Nathaniel C Nelson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Kendrew K Wong
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Ian J Mahoney
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Tahir Malik
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Darya Rudym
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Melissa B Lesko
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Seema Qayum
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Tyler C Lewis
- NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Stephanie H Chang
- NYU Langone Transplant Institute, NYU Langone Health, New York, New York; Department of Cardiothoracic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Justin C Y Chan
- NYU Langone Transplant Institute, NYU Langone Health, New York, New York; Department of Cardiothoracic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Travis C Geraci
- NYU Langone Transplant Institute, NYU Langone Health, New York, New York; Department of Cardiothoracic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Yonghua Li
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Prerna Pamar
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Joseph Schnier
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Rajbir Singh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Destiny Collazo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Miao Chang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Yaa Kyeremateng
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Colin McCormick
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Sara Borghi
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
| | - Shrey Patel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Fares Darawshy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; The Institute of Pulmonology, Hadassah Medical Center, Jerusalem, Israel; Department of Medicine, The Faculty of Medicine at the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Clea R Barnett
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Imran Sulaiman
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; Department of Respiratory Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Respiratory Medicine, Beaumont Hospital, Dublin, Ireland
| | - Matthias C Kugler
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Shari B Brosnahan
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Shivani Singh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Jun-Chieh J Tsay
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, VA New York Harbor Healthcare System, New York, New York
| | - Benjamin G Wu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, VA New York Harbor Healthcare System, New York, New York
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Luis F Angel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Leopoldo N Segal
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Jake G Natalini
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York.
| |
Collapse
|
4
|
Bery AI, Belousova N, Hachem RR, Roux A, Kreisel D. Chronic Lung Allograft Dysfunction: Clinical Manifestations and Immunologic Mechanisms. Transplantation 2025; 109:454-466. [PMID: 39104003 PMCID: PMC11799353 DOI: 10.1097/tp.0000000000005162] [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] [Indexed: 08/07/2024]
Abstract
The term "chronic lung allograft dysfunction" has emerged to describe the clinical syndrome of progressive, largely irreversible dysfunction of pulmonary allografts. This umbrella term comprises 2 major clinical phenotypes: bronchiolitis obliterans syndrome and restrictive allograft syndrome. Here, we discuss the clinical manifestations, diagnostic challenges, and potential therapeutic avenues to address this major barrier to improved long-term outcomes. In addition, we review the immunologic mechanisms thought to propagate each phenotype of chronic lung allograft dysfunction, discuss the various models used to study this process, describe potential therapeutic targets, and identify key unknowns that must be evaluated by future research strategies.
Collapse
Affiliation(s)
- Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Natalia Belousova
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
| | - Ramsey R Hachem
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Antoine Roux
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
- Paris Transplant Group, INSERM U 970s, Paris, France
| | - Daniel Kreisel
- Departments of Surgery, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
5
|
Miceli V, Ferrigno P, Centi C, Carcione C, Iannolo G, Agnese V, Lo Iacono G, Liotta R, Conaldi PG, Pinzani M, De Monte L, Bertani A. Differentially expressed microRNAs in pre-transplant lung biopsies target immune checkpoint proteins and can predict primary graft dysfunction in lung transplantation. Heliyon 2025; 11:e42515. [PMID: 40028527 PMCID: PMC11869042 DOI: 10.1016/j.heliyon.2025.e42515] [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: 05/10/2024] [Revised: 01/18/2025] [Accepted: 02/06/2025] [Indexed: 03/02/2025] Open
Abstract
Lung transplantation (LTx) significantly improves outcomes for patients with end-stage respiratory failure. However, primary graft dysfunction (PGD) remains one of the most relevant hurdles. Although PGD is attributed to ischemia-reperfusion injury (IRI), immune responses, primarily T cell-mediated, may play a pivotal role in its pathogenesis. Additionally, innate immune activation following IRI links PGD to adaptive alloimmunity, highlighting the impact of early events on LTx outcomes. Immune checkpoints (ICPs) such as PD-1/PD-L1, CD40/CD40LG, and OX40/OX40L, regulate post-LTx T cell responses, and dysregulation of microRNAs (miRNAs) has been implicated in altering ICP expression, influencing the amplification of immune responses. In this preliminary study, we used the taqMan low-density array (TLDA) cards to investigate miRNA dysregulation's prognostic potential as a PGD marker in pre-transplant back-table lung biopsies. Our analysis revealed differential miRNA expression in donor lung tissues, potentially associated with PGD onset, targeting immune regulatory pathways. Specifically, deregulated miRNAs targeted key ICP proteins, including PD-L1, CD40LG, and OX40L. Moreover, the differential expression of these miRNAs was observed in grafts with future PGD compared to grafts without PGD, suggesting a potential prognostic benefit and a possible role for lung tissue miRNAs in the onset of early graft dysfunction. These findings provide a basis for future investigations into their mechanistic roles and therapeutic potential for PGD. Although based on a limited number of cases, our results imply that miRNAs might be involved in early graft dysfunction. While requiring validation in larger cohorts, our data raise the possibility that the evaluation of the aforementioned markers during the pre-transplant phase, might offer a prognostic benefit in monitoring the onset of PGD. Additionally, the use of compounds that can modulate the function of these molecules could be evaluated for the management of LTx patients.
Collapse
Affiliation(s)
- Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Pia Ferrigno
- Division of Thoracic Surgery and Lung Transplantation, Chest Center, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
- UPMCI (University of Pittsburgh Medical Center Italy), Palermo, Italy
| | - Claudio Centi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | | | - Gioacchin Iannolo
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Valentina Agnese
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Giovanna Lo Iacono
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Rosa Liotta
- Pathology Unit, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
- UPMCI (University of Pittsburgh Medical Center Italy), Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Massimo Pinzani
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Lavinia De Monte
- Division of Thoracic Surgery and Lung Transplantation, Chest Center, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | - Alessandro Bertani
- Division of Thoracic Surgery and Lung Transplantation, Chest Center, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
- UPMCI (University of Pittsburgh Medical Center Italy), Palermo, Italy
| |
Collapse
|
6
|
Zhang F, Lu S, Wang G, Xu H, Huang D, Li X. Predictive value of the monocyte count for determining the risk of postoperative moderate-to-severe ARDS in patients undergoing one-lung ventilation during radical treatment of esophageal cancer. Front Med (Lausanne) 2025; 12:1510788. [PMID: 40007585 PMCID: PMC11850344 DOI: 10.3389/fmed.2025.1510788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Background This study aimed to screen for risk factors and to assess the predictive value of the monocyte count for the development of moderate-to-severe acute respiratory distress syndrome (ARDS) in patients undergoing one-lung ventilation (OLV) during radical surgery for esophageal cancer. Methods In this retrospective study, patients with esophageal cancer admitted to the Department of Thoracic Surgery of Wuxi People's Hospital between January 2017 and January 2021 were selected. Demographic, preoperative, intraoperative, and postoperative (within 2 h) data were collected. Patients were categorized into moderate-to-severe ARDS and non-moderate-to-severe ARDS groups. Multifactorial logistic regression, receiver operating characteristic (ROC), curve-fitting, and Spearman correlation analysis were used to analyze the data. Results After screening, 255 patients were enrolled, with 18% in moderate-to-severe ARDS group. Regression analysis revealed that postoperative monocyte count was an independent predictor for severe ARDS after surgery (OR = 2.916, 95% CI: 1.082-7.863, p < 0.05). The optimal cut-off value of postoperative monocyte count in predicting moderate-to-severe ARDS was 0.56 × 109/L (AUC = 0.708) with a sensitivity of 67.4% and a specificity of 66.5%. The difference of predictive value between postoperative monocyte count and prediction model (AUC = 0.760) was not statistically significant (p = 0.142). Additionally, a nonlinear connection between postoperative monocyte count and severe ARDS was found using curve fitting. Conclusion The postoperative monocyte count is an ideal predictor of postoperative moderate-to-severe ARDS in this patient population and can be used for the early diagnosis of patients with severe postoperative ARDS.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Emergency Medicine, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
- Department of Intensive Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Shunmei Lu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Guilong Wang
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Hongyang Xu
- Department of Intensive Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Dongxiao Huang
- Department of Anesthesiology, Jiangnan University Medical Center, Wuxi, Jiangsu, China
| | - Xiaomin Li
- Department of Emergency Medicine, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| |
Collapse
|
7
|
Vanluyten C, Vos R, Ceulemans LJ. Nr4a1: A multilevel target to overcome PGD in lung transplantation. J Heart Lung Transplant 2025; 44:261-262. [PMID: 39577508 DOI: 10.1016/j.healun.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024] Open
Affiliation(s)
- Cedric Vanluyten
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.
| | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Kawana S, Okazaki M, Sakaue T, Hashimoto K, Nakata K, Choshi H, Tanaka S, Miyoshi K, Ohtani S, Ohara T, Sugimoto S, Matsukawa A, Toyooka S. Loss of Nr4a1 ameliorates endothelial cell injury and vascular leakage in lung transplantation from circulatory-death donor. J Heart Lung Transplant 2025; 44:249-260. [PMID: 39369968 DOI: 10.1016/j.healun.2024.09.028] [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: 02/22/2024] [Revised: 09/20/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) stands as a major trigger for primary graft dysfunction (PGD) in lung transplantation (LTx). Especially in LTx from donation after cardiac death (DCD), effective control of IRI following warm ischemia (WIRI) is crucial to prevent PGD. This study aimed to identify the key factors affecting WIRI in LTx from DCD. METHODS Previously reported RNA-sequencing dataset of lung WIRI was reanalyzed to identify nuclear receptor subfamily 4 group A member 1 (NR4A1) as the immediate early gene for WIRI. Dynamics of NR4A1 expression were verified using a mouse hilar clamp model. To investigate the role of NR4A1 in WIRI, a mouse model of LTx from DCD was established using Nr4a1 knockout (Nr4a1-/-) mice. RESULTS NR4A1 was located around vascular cells, and its protein levels in the lungs increased rapidly and transiently during WIRI. LTx from Nr4a1-/- donors significantly improved pulmonary graft function compared to wild-type donors. Histological analysis showed decreased microvascular endothelial cell death, neutrophil infiltration, and albumin leakage. Evans blue permeability assay demonstrated maintained pulmonary microvascular barrier integrity in grafts from Nr4a1-/- donors, correlating with diminished pulmonary edema. However, NR4A1 did not significantly affect the inflammatory response during WIRI, and IRI was not suppressed when a wild-type donor lung was transplanted into the Nr4a1-/- recipient. CONCLUSIONS Donor NR4A1 plays a specialized role in the positive regulation of endothelial cell injury and microvascular hyperpermeability. These findings demonstrate the potential of targeting NR4A1 interventions to alleviate PGD and improve outcomes in LTx from DCD.
Collapse
Affiliation(s)
- Shinichi Kawana
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mikio Okazaki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Tomohisa Sakaue
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan; Department of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Kohei Hashimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaro Nakata
- Department of Surgery, Division of Cardiovascular and Thoracic Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Haruki Choshi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shin Tanaka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaroh Miyoshi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinji Ohtani
- Department of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
9
|
Jeong JC, Gelman AE, Chong AS. Update on the immunological mechanisms of primary graft dysfunction and chronic lung allograft dysfunction. Curr Opin Organ Transplant 2024; 29:412-419. [PMID: 39422603 PMCID: PMC11537820 DOI: 10.1097/mot.0000000000001175] [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] [Indexed: 10/19/2024]
Abstract
PURPOSE OF REVIEW Primary graft dysfunction (PGD) and chronic lung allograft dysfunction (CLAD) are the leading causes of graft loss in lung transplant recipients. The development of mouse lung transplant models has allowed for the genetic dissection of cellular and molecular pathways that prevent graft survival. This review provides an overview into recent mechanistic insights into PGD and CLAD. RECENT FINDINGS Mouse orthotopic lung transplant models and investigations of human lung transplant recipeints have revealed new molecular and cellular targets that promote PGD and CLAD. Donor and recipient-derived innate immune cells promote PGD and CLAD. PGD is driven by communication between classical monocytes and tissue-resident nonclassical monocytes activating alveolar macrophages to release chemokines that recruit neutrophils. Products of cell damage trigger neutrophil NET release, which together with NK cells, antibodies and complement, that further promote PGD. The development of CLAD involves circuits that activate B cells, CD8 + T cells, classical monocytes, and eosinophils. SUMMARY Effective targeted management of PGD and CLAD in lung transplant recipient to improve their long-term outcome remains a critical unmet need. Current mechanistic studies and therapeutic studies in mouse models and humans identify new possibilities for prevention and treatment.
Collapse
Affiliation(s)
- Jong Cheol Jeong
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, Illinois, USA
| | - Andrew E. Gelman
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Anita S Chong
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
10
|
Wong A, Duong A, Wilson G, Yeung J, MacParland S, Han H, Cypel M, Keshavjee S, Liu M. Ischemia-reperfusion responses in human lung transplants at the single-cell resolution. Am J Transplant 2024; 24:2199-2211. [PMID: 39197591 DOI: 10.1016/j.ajt.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/01/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Ischemia-reperfusion is an unavoidable step of organ transplantation. Development of therapeutics for lung injury during transplantation has proved challenging; understanding lung injury from human data at the single-cell resolution is required to accelerate the development of therapeutics. Donor lung biopsies from 6 human lung transplant cases were collected at the end of cold preservation and 2-hour reperfusion and underwent single-cell RNA sequencing. Donor and recipient origin of cells from the reperfusion timepoint were deconvolved. Gene expression profiles were: (1) compared between each donor cell type between timepoints and (2) compared between donor and recipient cells. Inflammatory responses from donor lung macrophages were found after reperfusion with upregulation of multiple cytokines and chemokines, especially IL-1β and IL-1α. Significant inflammatory responses were found in alveolar epithelial cells (featured by CXCL8) and lung endothelial cells (featured by IL-6 upregulation). Different inflammatory responses were noted between donor and recipient monocytes and CD8+ T cells. The inflammatory signals and differences between donor and recipient cells observed provide insight into the cellular and molecular mechanisms of ischemia-reperfusion induced lung injury. Further investigations may lead to the development of novel targeted therapeutics.
Collapse
Affiliation(s)
- Aaron Wong
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Allen Duong
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gavin Wilson
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Yeung
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sonya MacParland
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hong Han
- Centre for Discovery in Cancer Research and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Liu X, Ren Z, Tan C, Núñez-Santana FL, Kelly ME, Yan Y, Sun H, Abdala-Valencia H, Yang W, Wu Q, Toyoda T, Milisav M, Casalino-Matsuda SM, Lecuona E, Cerier EJ, Heung LJ, Abazeed ME, Perlman H, Gao R, Chandel NS, Budinger GS, Bharat A. Inducible CCR2+ nonclassical monocytes mediate the regression of cancer metastasis. J Clin Invest 2024; 134:e179527. [PMID: 39545417 PMCID: PMC11563681 DOI: 10.1172/jci179527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024] Open
Abstract
A major limitation of immunotherapy is the development of resistance resulting from cancer-mediated inhibition of host lymphocytes. Cancer cells release CCL2 to recruit classical monocytes expressing its receptor CCR2 for the promotion of metastasis and resistance to immunosurveillance. In the circulation, some CCR2-expressing classical monocytes lose CCR2 and differentiate into intravascular nonclassical monocytes that have anticancer properties but are unable to access extravascular tumor sites. We found that in mice and humans, an ontogenetically distinct subset of naturally underrepresented CCR2-expressing nonclassical monocytes was expanded during inflammatory states such as organ transplant and COVID-19 infection. These cells could be induced during health by treatment of classical monocytes with small-molecule activators of NOD2. The presence of CCR2 enabled these inducible nonclassical monocytes to infiltrate both intra- and extravascular metastatic sites of melanoma, lung, breast, and colon cancer in murine models, and they reversed the increased susceptibility of Nod2-/- mutant mice to cancer metastasis. Within the tumor colonies, CCR2+ nonclassical monocytes secreted CCL6 to recruit NK cells that mediated tumor regression, independent of T and B lymphocytes. Hence, pharmacological induction of CCR2+ nonclassical monocytes might be useful for immunotherapy-resistant cancers.
Collapse
Affiliation(s)
- Xianpeng Liu
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | | | - Can Tan
- Division of Cardiology, Department of Medicine, and
| | - Félix L. Núñez-Santana
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Megan E. Kelly
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Yuanqing Yan
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Haiying Sun
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Hiam Abdala-Valencia
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Wenbin Yang
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Qiang Wu
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Takahide Toyoda
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Marija Milisav
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - S. Marina Casalino-Matsuda
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Emilia Lecuona
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Emily Jeong Cerier
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Lena J. Heung
- Division of Infectious Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | | | - Ruli Gao
- Department of Biochemistry, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Navdeep S. Chandel
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - G.R. Scott Budinger
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ankit Bharat
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
12
|
Gouchoe DA, Zhang Z, Kim JL, Lee YG, Whitson BA, Zhu H. Improving lung allograft function in the early post-operative period through the inhibition of pyroptosis. MEDICAL REVIEW (2021) 2024; 4:384-394. [PMID: 39444796 PMCID: PMC11495470 DOI: 10.1515/mr-2023-0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/04/2024] [Indexed: 10/25/2024]
Abstract
Lung transplantation is the only definitive therapy for end-stage pulmonary disease. Less than 20 % of offered lungs are successfully transplanted due to a limited ischemic time window and poor donor lung quality manifested by pulmonary edema, hypoxia, or trauma. Therefore, poor donor organ recovery and utilization are significant barriers to wider implementation of the life-saving therapy of transplantation. While ischemia reperfusion injury (IRI) is often identified as the underlying molecular insult leading to immediate poor lung function in the post-operative period, this injury encompasses several pathways of cellular injury in addition to the recruitment of the innate immune system to the site of injury to propagate this inflammatory cascade. Pyroptosis is a central molecular inflammatory pathway that is the most significant contributor to injury in this early post-operative phase. Pyroptosis is another form of programmed cell death and is often associated with IRI. The mitigation of pyroptosis in the early post-operative period following lung transplantation is a potential novel way to prevent poor allograft function and improve outcomes for all recipients. Here we detail the pyroptotic pathway, its importance in lung transplantation, and several therapeutic modalities that can mitigate this harmful inflammatory pathway.
Collapse
Affiliation(s)
- Doug A. Gouchoe
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- COPPER Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Zhentao Zhang
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jung-Lye Kim
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- COPPER Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yong Gyu Lee
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- COPPER Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bryan A. Whitson
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- COPPER Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hua Zhu
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
13
|
Li C, Gao P, Zhuang F, Wang T, Wang Z, Wu G, Zhou Z, Xie H, Xie D, Zhao D, Wu J, Chen C. Inhibition of ALOX12-12-HETE Alleviates Lung Ischemia-Reperfusion Injury by Reducing Endothelial Ferroptosis-Mediated Neutrophil Extracellular Trap Formation. RESEARCH (WASHINGTON, D.C.) 2024; 7:0473. [PMID: 39268501 PMCID: PMC11391482 DOI: 10.34133/research.0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024]
Abstract
Lung ischemia-reperfusion injury (IRI) stands as the primary culprit behind primary graft dysfunction (PGD) after lung transplantation, yet viable therapeutic options are lacking. In the present study, we used a murine hilar clamp (1 h) and reperfusion (3 h) model to study IRI. The left lung tissues were harvested for metabolomics, transcriptomics, and single-cell RNA sequencing. Metabolomics of plasma from human lung transplantation recipients was also performed. Lung histology, pulmonary function, pulmonary edema, and survival analysis were measured in mice. Integrative analysis of metabolomics and transcriptomics revealed a marked up-regulation of arachidonate 12-lipoxygenase (ALOX12) and its metabolite 12-hydroxyeicosatetraenoic acid (12-HETE), which played a pivotal role in promoting ferroptosis and neutrophil extracellular trap (NET) formation during lung IRI. Additionally, single-cell RNA sequencing revealed that ferroptosis predominantly occurred in pulmonary endothelial cells. Importantly, Alox12-knockout (KO) mice exhibited a notable decrease in ferroptosis, NET formation, and tissue injury. To investigate the interplay between endothelial ferroptosis and NET formation, a hypoxia/reoxygenation (HR) cell model using 2 human endothelial cell lines was established. By incubating conditioned medium from HR cell model with neutrophils, we found that the liberation of high mobility group box 1 (HMGB1) from endothelial cells undergoing ferroptosis facilitated the formation of NETs by activating the TLR4/MYD88 pathway. Last, the administration of ML355, a targeted inhibitor of Alox12, mitigated lung IRI in both murine hilar clamp/reperfusion and rat left lung transplant models. Collectively, our study indicates ALOX12 as a promising therapeutic strategy for lung IRI.
Collapse
Affiliation(s)
- Chongwu Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peigen Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Fenghui Zhuang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Tao Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Zeyu Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Guodong Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Ziheng Zhou
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| |
Collapse
|
14
|
Jeon JE, Rajapaksa Y, Keshavjee S, Liu M. Applications of transcriptomics in ischemia reperfusion research in lung transplantation. J Heart Lung Transplant 2024; 43:1501-1513. [PMID: 38513917 DOI: 10.1016/j.healun.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
Ischemia-reperfusion (IR) injury contributes to primary graft dysfunction, a major cause of early mortality after lung transplantation. Transcriptomics uses high-throughput techniques to profile the RNA transcripts within a sample and provides a unique view of the mechanisms underlying various biological processes. This review aims to highlight the applications of transcriptomics in lung IR injury studies, which have thus far revealed inflammatory responses to be the major event activated by IR, identified potential biomarkers and therapeutic targets, and investigated the mechanisms of therapeutic interventions. Ex vivo lung perfusion, together with advanced bioinformatic and transcriptomic techniques, including single-cell RNA-sequencing, microRNA profiling, and multi-omics, continue to expand the capabilities of transcriptomics. In the future, the construction of biospecimen banks and the promotion of international collaborations among clinicians and researchers have the potential to advance our understanding of IR injury and improve the management of lung transplant recipients.
Collapse
Affiliation(s)
- Jamie E Jeon
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yasal Rajapaksa
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
15
|
Yu J, Fu Y, Gao J, Zhang Q, Zhang N, Zhang Z, Jiang X, Chen C, Wen Z. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol 2024; 74:103231. [PMID: 38861835 PMCID: PMC11209641 DOI: 10.1016/j.redox.2024.103231] [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: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024] Open
Abstract
Primary graft dysfunction (PGD) is a severe form of acute lung injury resulting from lung ischemia/reperfusion injury (I/R) in lung transplantation (LTx), associated with elevated post-transplant morbidity and mortality rates. Neutrophils infiltrating during reperfusion are identified as pivotal contributors to lung I/R injury by releasing excessive neutrophil extracellular traps (NETs) via NETosis. While alveolar macrophages (AMs) are involved in regulating neutrophil chemotaxis and infiltration, their role in NETosis during lung I/R remains inadequately elucidated. Extracellular histones constitute the main structure of NETs and can activate AMs. In this study, we confirmed the significant involvement of extracellular histone-induced M1 phenotype of AMs (M1-AMs) in driving NETosis during lung I/R. Using secretome analysis, public protein databases, and transwell co-culture models of AMs and neutrophils, we identified Cathepsin C (CTSC) derived from AMs as a major mediator in NETosis. Further elucidating the molecular mechanisms, we found that CTSC induced NETosis through a pathway dependent on NADPH oxidase-mediated production of reactive oxygen species (ROS). CTSC could significantly activate p38 MAPK, resulting in the phosphorylation of the NADPH oxidase subunit p47phox, thereby facilitating the trafficking of cytoplasmic subunits to the cell membrane and activating NADPH oxidase. Moreover, CTSC up-regulated and activated its substrate membrane proteinase 3 (mPR3), resulting in an increased release of NETosis-related inflammatory factors. Inhibiting CTSC revealed great potential in mitigating NETosis-related injury during lung I/R. These findings suggests that CTSC from AMs may be a crucial factor in mediating NETosis during lung I/R, and targeting CTSC inhition may represent a novel intervention for PGD in LTx.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
16
|
Zhang N, Zhang Q, Zhang Z, Yu J, Fu Y, Gao J, Jiang X, Jiang P, Wen Z. IRF1 and IL1A associated with PANoptosis serve as potential immune signatures for lung ischemia reperfusion injury following lung transplantation. Int Immunopharmacol 2024; 139:112739. [PMID: 39074415 DOI: 10.1016/j.intimp.2024.112739] [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: 05/13/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Lung ischemia reperfusion injury (IRI) is the principal cause of primary graft dysfunction (PGD) after lung transplantation, affecting short-term and long-term mortality post-transplantation. PANoptosis, a newly identified form of regulated cell death involving apoptosis, necroptosis, and pyroptosis, is now considered a possible cause of organ damage and IRI. However, the specific role of PANoptosis to the development of lung IRI following lung transplantation is still not fully understood. METHODS In this study, we identified differentially expressed genes (DEGs) by analyzing the gene expression data from the GEO database related to lung IRI following lung transplantation. PANoptosis-IRI DEGs were determined based on the intersection of PANoptosis-related genes and screened DEGs. Hub genes associated with lung IRI were further screened using Lasso regression and the SVM-RFE algorithm. Additionally, the Cibersort algorithm was employed to assess immune cell infiltration and investigate the interaction between immune cells and hub genes. The upstream miRNAs that may regulate hub genes and compounds that may interact with hub genes were also analyzed. Moreover, an external dataset was utilized to validate the differential expression analysis of hub genes. Finally, the expressions of hub genes were ultimately confirmed using quantitative real-time PCR, western blotting, and immunohistochemistry in both animal models of lung IRI and lung transplant patients. RESULTS PANoptosis-related genes, specifically interferon regulatory factor 1 (IRF1) and interleukin 1 alpha (IL1A), have been identified as potential biomarkers for lung IRI following lung transplantation. In mouse models of lung IRI, both the mRNA and protein expression levels of IRF1 and IL1A were significantly elevated in lung tissues of the IRI group compared to the control group. Moreover, lung transplant recipients exhibited significantly higher protein levels of IRF1 and IL1A in PBMCs when compared to healthy controls. Patients who experienced PGD showed elevated levels of IRF1 and IL1A proteins in their blood samples. Furthermore, in patients undergoing lung transplantation, the protein levels of IRF1 and IL1A were notably increased in peripheral blood mononuclear cells (PBMCs) compared to healthy controls. In addition, patients who developed primary graft dysfunction (PGD) exhibited even higher protein levels of IRF1 and IL1A than those without PGD. Furthermore, PANoptosis was observed in the lung tissues of mouse models of lung IRI and in the PBMCs of patients who underwent lung transplantation. CONCLUSIONS Our research identified IRF1 and IL1A as biomarkers associated with PANoptosis in lung IRI, suggesting their potential utility as targets for diagnosing and therapeutically intervening in lung IRI and PGD following lung transplantation.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ping Jiang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
17
|
Cerier E, Kurihara C, Kaiho T, Toyoda T, Manerikar A, Kandula V, Thomae B, Yagi Y, Yeldandi A, Kim S, Avella-Patino D, Pandolfino J, Perlman H, Singer B, Scott Budinger GR, Lung K, Alexiev B, Bharat A. Temporal correlation between postreperfusion complement deposition and severe primary graft dysfunction in lung allografts. Am J Transplant 2024; 24:577-590. [PMID: 37977230 PMCID: PMC10982049 DOI: 10.1016/j.ajt.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/07/2023] [Accepted: 11/12/2023] [Indexed: 11/19/2023]
Abstract
Growing evidence implicates complement in the pathogenesis of primary graft dysfunction (PGD). We hypothesized that early complement activation postreperfusion could predispose to severe PGD grade 3 (PGD-3) at 72 hours, which is associated with worst posttransplant outcomes. Consecutive lung transplant patients (n = 253) from January 2018 through June 2023 underwent timed open allograft biopsies at the end of cold ischemia (internal control) and 30 minutes postreperfusion. PGD-3 at 72 hours occurred in 14% (35/253) of patients; 17% (44/253) revealed positive C4d staining on postreperfusion allograft biopsy, and no biopsy-related complications were encountered. Significantly more patients with PGD-3 at 72 hours had positive C4d staining at 30 minutes postreperfusion compared with those without (51% vs 12%, P < .001). Conversely, patients with positive C4d staining were significantly more likely to develop PGD-3 at 72 hours (41% vs 8%, P < .001) and experienced worse long-term outcomes. In multivariate logistic regression, positive C4d staining remained highly predictive of PGD-3 (odds ratio 7.92, 95% confidence interval 2.97-21.1, P < .001). Hence, early complement deposition in allografts is highly predictive of PGD-3 at 72 hours. Our data support future studies to evaluate the role of complement inhibition in patients with early postreperfusion complement activation to mitigate PGD and improve transplant outcomes.
Collapse
Affiliation(s)
- Emily Cerier
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Chitaru Kurihara
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Taisuke Kaiho
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Takahide Toyoda
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adwaiy Manerikar
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Viswajit Kandula
- Department of Cardiothoracic Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Benjamin Thomae
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yuriko Yagi
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Anjana Yeldandi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Samuel Kim
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Diego Avella-Patino
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - John Pandolfino
- Department of Gastroenterology and Hepatology Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Harris Perlman
- Department of Rheumatology Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Benjamin Singer
- Department of Pulmonary and Critical Care Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | - G R Scott Budinger
- Department of Pulmonary and Critical Care Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | - Kalvin Lung
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Borislav Alexiev
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ankit Bharat
- Department of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
18
|
Trindade AJ. A Hospitable Home? Cell-Free DNA and the Inflammatory Milieu in Lung Transplant Candidates. Am J Respir Crit Care Med 2024; 209:627-628. [PMID: 38226872 PMCID: PMC10945066 DOI: 10.1164/rccm.202312-2242ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024] Open
Affiliation(s)
- Anil J Trindade
- Division of Allergy, Pulmonary, and Critical Care Medicine and Vanderbilt Transplant Center Vanderbilt University Medical Center Nashville, Tennessee
| |
Collapse
|
19
|
Li Q, Nie H. Advances in lung ischemia/reperfusion injury: unraveling the role of innate immunity. Inflamm Res 2024; 73:393-405. [PMID: 38265687 DOI: 10.1007/s00011-023-01844-7] [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: 10/18/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Lung ischemia/reperfusion injury (LIRI) is a common occurrence in clinical practice and represents a significant complication following pulmonary transplantation and various diseases. At the core of pulmonary ischemia/reperfusion injury lies sterile inflammation, where the innate immune response plays a pivotal role. This review aims to investigate recent advancements in comprehending the role of innate immunity in LIRI. METHODS A computer-based online search was performed using the PubMed database and Web of Science database for published articles concerning lung ischemia/reperfusion injury, cell death, damage-associated molecular pattern molecules (DAMPs), innate immune cells, innate immunity, inflammation. RESULTS During the process of lung ischemia/reperfusion, cellular injury even death can occur. When cells are injured or undergo cell death, endogenous ligands known as DAMPs are released. These molecules can be recognized and bound by pattern recognition receptors (PRRs), leading to the recruitment and activation of innate immune cells. Subsequently, a cascade of inflammatory responses is triggered, ultimately exacerbating pulmonary injury. These steps are complex and interrelated rather than being in a linear relationship. In recent years, significant progress has been made in understanding the immunological mechanisms of LIRI, involving novel types of cell death, the ability of receptors other than PRRs to recognize DAMPs, and a more detailed mechanism of action of innate immune cells in ischemia/reperfusion injury (IRI), laying the groundwork for the development of novel diagnostic and therapeutic approaches. CONCLUSIONS Various immune components of the innate immune system play critical roles in lung injury after ischemia/reperfusion. Preventing cell death and the release of DAMPs, interrupting DAMPs receptor interactions, disrupting intracellular inflammatory signaling pathways, and minimizing immune cell recruitment are essential for lung protection in LIRI.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| |
Collapse
|
20
|
Komaru Y, Bai YZ, Kreisel D, Herrlich A. Interorgan communication networks in the kidney-lung axis. Nat Rev Nephrol 2024; 20:120-136. [PMID: 37667081 DOI: 10.1038/s41581-023-00760-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 09/06/2023]
Abstract
The homeostasis and health of an organism depend on the coordinated interaction of specialized organs, which is regulated by interorgan communication networks of circulating soluble molecules and neuronal connections. Many diseases that seemingly affect one primary organ are really multiorgan diseases, with substantial secondary remote organ complications that underlie a large part of their morbidity and mortality. Acute kidney injury (AKI) frequently occurs in critically ill patients with multiorgan failure and is associated with high mortality, particularly when it occurs together with respiratory failure. Inflammatory lung lesions in patients with kidney failure that could be distinguished from pulmonary oedema due to volume overload were first reported in the 1930s, but have been largely overlooked in clinical settings. A series of studies over the past two decades have elucidated acute and chronic kidney-lung and lung-kidney interorgan communication networks involving various circulating inflammatory cytokines and chemokines, metabolites, uraemic toxins, immune cells and neuro-immune pathways. Further investigations are warranted to understand these clinical entities of high morbidity and mortality, and to develop effective treatments.
Collapse
Affiliation(s)
- Yohei Komaru
- Department of Medicine, Division of Nephrology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Yun Zhu Bai
- Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Andreas Herrlich
- Department of Medicine, Division of Nephrology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
- VA Saint Louis Health Care System, John Cochran Division, St. Louis, MO, USA.
| |
Collapse
|
21
|
Farahnak K, Bai YZ, Yokoyama Y, Morkan DB, Liu Z, Amrute JM, De Filippis Falcon A, Terada Y, Liao F, Li W, Shepherd HM, Hachem RR, Puri V, Lavine KJ, Gelman AE, Bharat A, Kreisel D, Nava RG. B cells mediate lung ischemia/reperfusion injury by recruiting classical monocytes via synergistic B cell receptor/TLR4 signaling. J Clin Invest 2024; 134:e170118. [PMID: 38488011 PMCID: PMC10940088 DOI: 10.1172/jci170118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 01/17/2024] [Indexed: 03/18/2024] Open
Abstract
Ischemia/reperfusion injury-mediated (IRI-mediated) primary graft dysfunction (PGD) adversely affects both short- and long-term outcomes after lung transplantation, a procedure that remains the only treatment option for patients suffering from end-stage respiratory failure. While B cells are known to regulate adaptive immune responses, their role in lung IRI is not well understood. Here, we demonstrated by intravital imaging that B cells are rapidly recruited to injured lungs, where they extravasate into the parenchyma. Using hilar clamping and transplant models, we observed that lung-infiltrating B cells produce the monocyte chemokine CCL7 in a TLR4-TRIF-dependent fashion, a critical step contributing to classical monocyte (CM) recruitment and subsequent neutrophil extravasation, resulting in worse lung function. We found that synergistic BCR-TLR4 activation on B cells is required for the recruitment of CMs to the injured lung. Finally, we corroborated our findings in reperfused human lungs, in which we observed a correlation between B cell infiltration and CM recruitment after transplantation. This study describes a role for B cells as critical orchestrators of lung IRI. As B cells can be depleted with currently available agents, our study provides a rationale for clinical trials investigating B cell-targeting therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew E. Gelman
- Department of Surgery
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ankit Bharat
- Department of Surgery, Northwestern University, Chicago, Illinois, USA
| | - Daniel Kreisel
- Department of Surgery
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | |
Collapse
|
22
|
Qian J, Xu Z, Yin M, Qin Z, Pinhu L. Bioinformatics analyses of immune-related genes and immune infiltration associated with lung ischemia-reperfusion injury. Transpl Immunol 2023; 81:101926. [PMID: 37652362 DOI: 10.1016/j.trim.2023.101926] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/21/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is a significant complication that can occur following lung transplantation and is known to contribute to poor prognosis. Our research aimed to investigate the potential molecular targets and mechanisms involved in lung IRI (LIRI), in order to improve our understanding of this condition. METHOD We downloaded gene expression datasets (GSE127003 and GSE18995) linked to LIRI from the GEO database. Using WGCNA, we identified LIRI-related modules. Functional enrichment analyses were performed on the modules showing significant correlation with LIRI. Core immune-related genes (IRGs) were identified and validated using the GSE18995 dataset. A rat LIRI model was established to validate the expression changes of core IRGs. The LIRI groups were subjected to 60 min of warm ischemia followed by 120 min of reperfusion. Additionally, the xCell algorithm was used to characterize the immune landscape and analyze the relationships between hub IRGs and infiltrating immune cells. RESULTS A total of 483 genes from the turquoise module were identified through WGCNA, with a predominant enrichment in immune- and inflammation-related pathways. Three IRGs (PTGS2, CCL2, and RELB) were found to be up-regulated after reperfusion in both GSE127003 and GSE18995 datasets, and this was further confirmed using the rat LIRI model. The xCell analysis revealed that immune score, CD8+ naive T cells, eosinophils, neutrophils, NK cells, and Tregs were upregulated after reperfusion. PTGS2, CCL2, and RELB showed positive correlations with CD8+ naive T cells, monocytes, neutrophils, and Tregs. CONCLUSION PTGS2, CCL2, and RELB were found to be potential biomarkers for LIRI. Immune and microenvironment scores were higher after reperfusion compared to before reperfusion. PTGS2, CCL2, and RELB appear to play a crucial role in the development of LIRI and may contribute to it by increasing the number of immune cells. Our findings offer new perspectives on potential treatment targets and the pathogenesis of LIRI.
Collapse
Affiliation(s)
- Jing Qian
- Department of Cardiothoracic Intensive Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhanyu Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Mingjing Yin
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhidan Qin
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Liao Pinhu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
23
|
Yang W, Lecuona E, Wu Q, Liu X, Sun H, Alam H, Nadig SN, Bharat A. The role of lung-restricted autoantibodies in the development of primary and chronic graft dysfunction. FRONTIERS IN TRANSPLANTATION 2023; 2:1237671. [PMID: 38993924 PMCID: PMC11235341 DOI: 10.3389/frtra.2023.1237671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/18/2023] [Indexed: 07/13/2024]
Abstract
Lung transplantation is a life-saving treatment for both chronic end-stage lung diseases and acute respiratory distress syndrome, including those caused by infectious agents like COVID-19. Despite its increasing utilization, outcomes post-lung transplantation are worse than other solid organ transplants. Primary graft dysfunction (PGD)-a condition affecting more than half of the recipients post-transplantation-is the chief risk factor for post-operative mortality, transplant-associated multi-organ dysfunction, and long-term graft loss due to chronic rejection. While donor-specific antibodies targeting allogenic human leukocyte antigens have been linked to transplant rejection, the role of recipient's pre-existing immunoglobulin G autoantibodies against lung-restricted self-antigens (LRA), like collagen type V and k-alpha1 tubulin, is less understood in the context of lung transplantation. Recent studies have found an increased risk of PGD development in lung transplant recipients with LRA. This review will synthesize past and ongoing research-utilizing both mouse models and human subjects-aimed at unraveling the mechanisms by which LRA heightens the risk of PGD. Furthermore, it will explore prospective approaches designed to mitigate the impact of LRA on lung transplant patients.
Collapse
Affiliation(s)
- Wenbin Yang
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Emilia Lecuona
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Qiang Wu
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Xianpeng Liu
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Haiying Sun
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hasan Alam
- Division of Trauma & Acute Care Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Satish N. Nadig
- Division of Abdominal Transplant, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ankit Bharat
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
24
|
Li J, Han Z, Zhu Z, Wei L. LncRNA H19 aggravates primary graft dysfunction after lung transplantation via KLF5-mediated activation of CCL28. Am J Transplant 2023; 23:1536-1550. [PMID: 37394140 DOI: 10.1016/j.ajt.2023.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/19/2023] [Accepted: 06/25/2023] [Indexed: 07/04/2023]
Abstract
The present study aims to elucidate the possible involvement of H19 in primary graft dysfunction (PGD) following lung transplantation (LT) and the underlying mechanism. The transcriptome data were obtained through high-throughput sequencing analysis, and the differential long noncoding RNAs and messenger RNAs were screened for coexpression analysis. The interaction among H19, KLF5 and CCL28 was analyzed. A hypoxia-induced human pulmonary microvascular endothelial cell injury model was established, in which H19 was knocked down to elucidate its effect on the lung function, inflammatory response, and cell apoptosis. An orthotopic left LT model was constructed for in vivo mechanistic validation. High-throughput transcriptome sequencing analysis revealed the involvement of the H19/KLF5/CCL28 signaling axis in PGD. Silencing of H19 reduced inflammatory response and thus improved PGD. CCL28 secreted by human pulmonary microvascular endothelial cells after LT recruited neutrophils and macrophages. Mechanistic investigations indicated that H19 augmented the expression of CCL28 by binding to the transcription factor KLF5. Abundant expression of CCL28 reversed the alleviating effect of H19 silencing on PGD. In conclusion, the results point out that H19 exerts a promoting effect on PGD through increasing KLF5 expression and the subsequent CCL28 expression. Our study provides a novel insight into the mechanism of action of H19.
Collapse
Affiliation(s)
- Jiwei Li
- Department of Thoracic Surgery, Zhengzhou Key Laboratory for Surgical Treatment for End-stage Lung Disease, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.
| | - Zhijun Han
- Department of Thoracic Surgery, Zhengzhou Key Laboratory for Surgical Treatment for End-stage Lung Disease, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zibo Zhu
- Department of Thoracic Surgery, Zhengzhou Key Laboratory for Surgical Treatment for End-stage Lung Disease, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Li Wei
- Department of Thoracic Surgery, Zhengzhou Key Laboratory for Surgical Treatment for End-stage Lung Disease, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
25
|
Li W, Shepherd HM, Terada Y, Shay AE, Bery AI, Gelman AE, Lavine KJ, Serhan CN, Kreisel D. Resolvin D1 prevents injurious neutrophil swarming in transplanted lungs. Proc Natl Acad Sci U S A 2023; 120:e2302938120. [PMID: 37487095 PMCID: PMC10400944 DOI: 10.1073/pnas.2302938120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/27/2023] [Indexed: 07/26/2023] Open
Abstract
Neutrophils are the primary cell type involved in lung ischemia-reperfusion injury (IRI), which remains a frequent and morbid complication after organ transplantation. Endogenous lipid mediators that become activated during acute inflammation-resolution have gained increasing recognition for their protective role(s) in promoting the restoration of homeostasis, but their influence on early immune responses following transplantation remains to be uncovered. Resolvin D1, 7S,8R,17S-trihydroxy-4Z,9E,11E,13Z,15E,19Z-docosahexaenoic acid (RvD1), is a potent stereoselective mediator that exhibits proresolving and anti-inflammatory actions in the setting of tissue injury. Here, using metabololipidomics, we demonstrate that endogenous proresolving mediators including RvD1 are increased in human and murine lung grafts immediately following transplantation. In mouse grafts, we observe lipid mediator class switching early after reperfusion. We use intravital two-photon microscopy to reveal that RvD1 treatment significantly limits early neutrophil infiltration and swarming, thereby ameliorating early graft dysfunction in transplanted syngeneic lungs subjected to severe IRI. Through integrated analysis of single-cell RNA sequencing data of donor and recipient immune cells from lung grafts, we identify transcriptomic changes induced by RvD1. These results support a role for RvD1 as a potent modality for preventing early neutrophil-mediated tissue damage after lung IRI that may be therapeutic in the clinics.
Collapse
Affiliation(s)
- Wenjun Li
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110
| | - Hailey M Shepherd
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110
| | - Yuriko Terada
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110
| | - Ashley E Shay
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Amit I Bery
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110
| | - Andrew E Gelman
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Kory J Lavine
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110
| | - Charles N Serhan
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
26
|
Chacon-Alberty L, Fernandez R, Jindra P, King M, Rosas I, Hochman-Mendez C, Loor G. Primary Graft Dysfunction in Lung Transplantation: A Review of Mechanisms and Future Applications. Transplantation 2023; 107:1687-1697. [PMID: 36650643 DOI: 10.1097/tp.0000000000004503] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lung allograft recipients have worse survival than all other solid organ transplant recipients, largely because of primary graft dysfunction (PGD), a major form of acute lung injury affecting a third of lung recipients within the first 72 h after transplant. PGD is the clinical manifestation of ischemia-reperfusion injury and represents the predominate cause of early morbidity and mortality. Despite PGD's impact on lung transplant outcomes, no targeted therapies are currently available; hence, care remains supportive and largely ineffective. This review focuses on molecular and innate immune mechanisms of ischemia-reperfusion injury leading to PGD. We also discuss novel research aimed at discovering biomarkers that could better predict PGD and potential targeted interventions that may improve outcomes in lung transplantation.
Collapse
Affiliation(s)
| | - Ramiro Fernandez
- Division of Cardiothoracic Transplantation and Mechanical Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Peter Jindra
- Division of Cardiothoracic Transplantation and Mechanical Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Madelyn King
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, TX
| | - Ivan Rosas
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | | | - Gabriel Loor
- Division of Cardiothoracic Transplantation and Mechanical Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
- Cardiothoracic Surgery Professional Staff, The Texas Heart Institute, Houston, TX
| |
Collapse
|
27
|
Yamanashi K, Ohsumi A, Oda H, Tanaka S, Yamada Y, Nakajima D, Date H. Reduction of donor mononuclear phagocytes with clodronate-liposome during ex vivo lung perfusion attenuates ischemia-reperfusion injury. J Thorac Cardiovasc Surg 2023; 165:e181-e203. [PMID: 36404143 DOI: 10.1016/j.jtcvs.2022.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Clodronate-liposome is used for depleting mononuclear phagocytes associated with ischemia-reperfusion injury. We hypothesized that administration of clodronate-liposome into the perfusate during ex vivo lung perfusion could reduce mononuclear phagocytes and attenuate ischemia-reperfusion injury. METHODS First, the number of mononuclear phagocytes in flushed grafts (minimum cold ischemic time, 6-hour cold ischemic time, 15-hour cold ischemic time, and 18-hour cold ischemic time; n = 6 each) was determined using flow cytometry. Second, grafts (15-hour cold ischemic time) were allocated to control or clodronate (n = 5 each). In the clodronate group, clodronate-liposome is administered into the perfusate. After 4 hours of ex vivo lung perfusion, the number of mononuclear phagocytes in the perfusate and lung tissues was measured. Third, grafts (15-hour cold ischemic time) were allocated to control or clodronate (n = 6 each). After 4 hours of ex vivo lung perfusion, the left lungs were transplanted and reperfused for 2 hours. Lung function was evaluated, and samples were analyzed. RESULTS First, mononuclear phagocytes remain in flushed grafts after prolonged cold ischemia. Second, the number of mononuclear phagocytes in lung tissues after ex vivo lung perfusion was significantly reduced in the clodronate group (P = .008). Third, lung compliance and vascular resistance during ex vivo lung perfusion were significantly improved in the clodronate group (P < .001 for both). Blood oxygenation and pulmonary edema were significantly improved in the clodronate group after 2 hours of reperfusion (P = .015 and P = .026, respectively). Histological findings showed reduced lung injury in the clodronate group (P = .013). CONCLUSIONS Administration of clodronate-liposome into the perfusate during ex vivo lung perfusion resulted in a significant reduction of mononuclear phagocytes in donor lungs, leading to attenuation of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Keiji Yamanashi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hiromi Oda
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satona Tanaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshito Yamada
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
28
|
Shao L, Wu B, Liu C, Chong W. VALPROIC ACID INHIBITS CLASSICAL MONOCYTE-DERIVED TISSUE FACTOR AND ALLEVIATES HEMORRHAGIC SHOCK-INDUCED ACUTE LUNG INJURY IN RATS. Shock 2023; 59:449-459. [PMID: 36443067 PMCID: PMC9997640 DOI: 10.1097/shk.0000000000002064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/27/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
ABSTRACT Background: Monocytes and monocyte-derived tissue factor (TF) promote the development of sepsis-induced acute lung injury (ALI). Classical monocytes (C-Mcs) can be induced to express TF. Valproic acid (VPA) alleviates hemorrhagic shock (HS)-induced ALI (HS/ALI) and inhibits TF expression in monocytes. We hypothesized that C-Mcs and C-Mc-derived TF promoted HS/ALI and that VPA could inhibit C-Mc-derived TF expression and attenuate HS/ALI. Methods: Wistar rats and THP-1 cells were used to evaluate our hypothesis. Monocyte subtypes were analyzed by flow cytometry; mRNA expression was measured by fluorescence quantitative polymerase chain reaction; protein expression was measured by Western blotting, immunofluorescence, or immunohistology; inflammatory cytokines levels were measured by enzyme-linked immunosorbent assay; and ALI scores were used to determine the degree of ALI. Results: The blood %C-Mcs and C-Mcs/non-C-Mcs ratios, monocyte TF levels, serum and/or lung inflammatory cytokine levels, and ALI scores of HS rats were significantly increased ( P < 0.05). After monocyte depletion and thrombin inhibition, the inflammatory cytokine levels and ALI scores were significantly decreased ( P < 0.05). VPA reduced the %C-Mcs and C-Mc/non-C-Mc ratios, TF expression, inflammatory cytokine levels, and ALI scores during HS ( P < 0.05) and inhibited HS-induced monocyte Egr-1 and p-ERK1/2 expression ( P < 0.05). VPA inhibited hypoxia-induced TF expression in THP-1 cells by regulating the p-ERK1/2-Egr-1 axis. Conclusion: C-Mcs and C-Mc-derived TF accelerate the development of HS/ALI by increasing thrombin production. VPA inhibits HS-induced C-Mc production of TF by regulating the p-ERK1/2-Egr-1 axis and alleviates HS/ALI.
Collapse
Affiliation(s)
- Lina Shao
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
- Intensive Care Unit, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
- Intensive Care Unit, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province, China
- Intensive Care Unit, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Bing Wu
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Chang Liu
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Chong
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
29
|
McGinniss JE, Whiteside SA, Deek RA, Simon-Soro A, Graham-Wooten J, Oyster M, Brown MD, Cantu E, Diamond JM, Li H, Christie JD, Bushman FD, Collman RG. The Lung Allograft Microbiome Associates with Pepsin, Inflammation, and Primary Graft Dysfunction. Am J Respir Crit Care Med 2022; 206:1508-1521. [PMID: 36103583 PMCID: PMC9757091 DOI: 10.1164/rccm.202112-2786oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 09/14/2022] [Indexed: 12/24/2022] Open
Abstract
Rationale: Primary graft dysfunction (PGD) is the principal cause of early morbidity and mortality after lung transplantation. The lung microbiome has been implicated in later transplantation outcomes but has not been investigated in PGD. Objectives: To define the peritransplant bacterial lung microbiome and relationship to host response and PGD. Methods: This was a single-center prospective cohort study. Airway lavage samples from donor lungs before organ procurement and recipient allografts immediately after implantation underwent bacterial 16S ribosomal ribonucleic acid gene sequencing. Recipient allograft samples were analyzed for cytokines by multiplex array and pepsin by ELISA. Measurements and Main Results: We enrolled 139 transplant subjects and obtained donor lung (n = 109) and recipient allograft (n = 136) samples. Severe PGD (persistent grade 3) developed in 15 subjects over the first 72 hours, and 40 remained without PGD (persistent grade 0). The microbiome of donor lungs differed from healthy lungs, and recipient allograft microbiomes differed from donor lungs. Development of severe PGD was associated with enrichment in the immediate postimplantation lung of oropharyngeal anaerobic taxa, particularly Prevotella. Elevated pepsin, a gastric biomarker, and a hyperinflammatory cytokine profile were present in recipient allografts in severe PGD and strongly correlated with microbiome composition. Together, immediate postimplantation allograft Prevotella/Streptococcus ratio, pepsin, and indicator cytokines were associated with development of severe PGD during the 72-hour post-transplantation period (area under the curve = 0.81). Conclusions: Lung allografts that develop PGD have a microbiome enriched in anaerobic oropharyngeal taxa, elevated gastric pepsin, and hyperinflammatory phenotype. These findings suggest a possible role for peritransplant aspiration in PGD, a potentially actionable mechanism that warrants further investigation.
Collapse
Affiliation(s)
- John E. McGinniss
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | | | | | - Aurea Simon-Soro
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | | | - Michelle Oyster
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Melanie D. Brown
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | | | - Joshua M. Diamond
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Hongzhe Li
- Department of Epidemiology, Biostatistics, and Informatics
| | - Jason D. Christie
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Frederic D. Bushman
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ronald G. Collman
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Shepherd HM, Gauthier JM, Terada Y, Li W, Krupnick AS, Gelman AE, Kreisel D. Updated Views on Neutrophil Responses in Ischemia-Reperfusion Injury. Transplantation 2022; 106:2314-2324. [PMID: 35749228 PMCID: PMC9712152 DOI: 10.1097/tp.0000000000004221] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ischemia-reperfusion injury is an inevitable event during organ transplantation and represents a primary risk factor for the development of early graft dysfunction in lung, heart, liver, and kidney transplant recipients. Recent studies have implicated recipient neutrophils as key mediators of this process and also have found that early innate immune responses after transplantation can ultimately augment adaptive alloimmunity and affect late graft outcomes. Here, we discuss signaling pathways involved in neutrophil recruitment and activation after ischemia-mediated graft injury in solid organ transplantation with an emphasis on lung allografts, which have been the focus of recent studies. These findings suggest novel therapeutic interventions that target ischemia-reperfusion injury-mediated graft dysfunction in transplant recipients.
Collapse
Affiliation(s)
- Hailey M. Shepherd
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Jason M. Gauthier
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Yuriko Terada
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Wenjun Li
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | | | - Andrew E. Gelman
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
31
|
Research Highlights. Transplantation 2022. [DOI: 10.1097/tp.0000000000004407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
32
|
Yang W, Cerier EJ, Núñez-Santana FL, Wu Q, Yan Y, Kurihara C, Liu X, Yeldandi A, Khurram N, Avella-Patino D, Sun H, Budinger GS, Kreisel D, Mohanakumar T, Lecuona E, Bharat A. IL-1β-dependent extravasation of preexisting lung-restricted autoantibodies during lung transplantation activates complement and mediates primary graft dysfunction. J Clin Invest 2022; 132:157975. [PMID: 36250462 PMCID: PMC9566897 DOI: 10.1172/jci157975] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Preexisting lung-restricted autoantibodies (LRAs) are associated with a higher incidence of primary graft dysfunction (PGD), although it remains unclear whether LRAs can drive its pathogenesis. In syngeneic murine left lung transplant recipients, preexisting LRAs worsened graft dysfunction, which was evident by impaired gas exchange, increased pulmonary edema, and activation of damage-associated pathways in lung epithelial cells. LRA-mediated injury was distinct from ischemia-reperfusion injury since deletion of donor nonclassical monocytes and host neutrophils could not prevent graft dysfunction in LRA-pretreated recipients. Whole LRA IgG molecules were necessary for lung injury, which was mediated by the classical and alternative complement pathways and reversed by complement inhibition. However, deletion of Fc receptors in donor macrophages or mannose-binding lectin in recipient mice failed to rescue lung function. LRA-mediated injury was localized to the transplanted lung and dependent on IL-1β-mediated permeabilization of pulmonary vascular endothelium, which allowed extravasation of antibodies. Genetic deletion or pharmacological inhibition of IL-1R in the donor lungs prevented LRA-induced graft injury. In humans, preexisting LRAs were an independent risk factor for severe PGD and could be treated with plasmapheresis and complement blockade. We conclude that preexisting LRAs can compound ischemia-reperfusion injury to worsen PGD for which complement inhibition may be effective.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - G.R. Scott Budinger
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniel Kreisel
- Departments of Surgery, Pathology & Immunology, Washington University, St. Louis, Missouri, USA
| | | | | | - Ankit Bharat
- Division of Thoracic Surgery
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
33
|
Miller CL, O JM, Allan JS, Madsen JC. Novel approaches for long-term lung transplant survival. Front Immunol 2022; 13:931251. [PMID: 35967365 PMCID: PMC9363671 DOI: 10.3389/fimmu.2022.931251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Allograft failure remains a major barrier in the field of lung transplantation and results primarily from acute and chronic rejection. To date, standard-of-care immunosuppressive regimens have proven unsuccessful in achieving acceptable long-term graft and patient survival. Recent insights into the unique immunologic properties of lung allografts provide an opportunity to develop more effective immunosuppressive strategies. Here we describe advances in our understanding of the mechanisms driving lung allograft rejection and highlight recent progress in the development of novel, lung-specific strategies aimed at promoting long-term allograft survival, including tolerance.
Collapse
Affiliation(s)
- Cynthia L. Miller
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA, United States
| | - Jane M. O
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA, United States
| | - James S. Allan
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA, United States
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| | - Joren C. Madsen
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA, United States
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
34
|
Querrey M, Chiu S, Lecuona E, Wu Q, Sun H, Anderson M, Kelly M, Ravi S, Misharin AV, Kreisel D, Bharat A, Budinger GS. CD11b suppresses TLR activation of nonclassical monocytes to reduce primary graft dysfunction after lung transplantation. J Clin Invest 2022; 132:157262. [PMID: 35838047 PMCID: PMC9282933 DOI: 10.1172/jci157262] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/25/2022] [Indexed: 02/03/2023] Open
Abstract
Primary graft dysfunction (PGD) is the leading cause of postoperative mortality in lung transplant recipients and the most important risk factor for development of chronic lung allograft dysfunction. The mechanistic basis for the variability in the incidence and severity of PGD between lung transplant recipients is not known. Using a murine orthotopic vascularized lung transplant model, we found that redundant activation of Toll-like receptors 2 and 4 (TLR2 and -4) on nonclassical monocytes activates MyD88, inducing the release of the neutrophil attractant chemokine CXCL2. Deletion of Itgam (encodes CD11b) in nonclassical monocytes enhanced their production of CXCL2 and worsened PGD, while a CD11b agonist, leukadherin-1, administered only to the donor lung prior to lung transplantation, abrogated CXCL2 production and PGD. The damage-associated molecular pattern molecule HMGB1 was increased in peripheral blood samples from patients undergoing lung transplantation after reperfusion and induced CXCL2 production in nonclassical monocytes via TLR4/MyD88. An inhibitor of HMGB1 administered to the donor and recipient prior to lung transplantation attenuated PGD. Our findings suggest that CD11b acts as a molecular brake to prevent neutrophil recruitment by nonclassical monocytes following lung transplantation, revealing an attractive therapeutic target in the donor lung to prevent PGD in lung transplant recipients.
Collapse
Affiliation(s)
- Melissa Querrey
- Division of Pulmonary and Critical Care Medicine and,Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Stephen Chiu
- Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Emilia Lecuona
- Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Qiang Wu
- Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Haiying Sun
- Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Megan Anderson
- Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Megan Kelly
- Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sowmya Ravi
- Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Ankit Bharat
- Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - G.R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine and,Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
35
|
Peng X, Wu H. Inflammatory Links Between Hypertriglyceridemia and Atherogenesis. Curr Atheroscler Rep 2022; 24:297-306. [PMID: 35274230 PMCID: PMC9575332 DOI: 10.1007/s11883-022-01006-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Recent studies indicate an association between hypertriglyceridemia (HTG) and atherosclerotic cardiovascular disease (ASCVD). The purpose of this review is to discuss the potential mechanism connecting HTG and ASCVD risk and the potential efficacy of HTG-targeting therapies in ASCVD prevention. RECENT FINDINGS HTG, with elevations in triglyceride-rich lipoproteins (TGRL) and their remnants, are causal ASCVD risk factors. The mechanisms whereby HTG increases ASCVD risk are not well understood but may include multiple factors. Inflammation plays a crucial role in atherosclerosis. TGRL compared to low-density lipoproteins (LDL) correlate better with inflammation. TGRL remnants can penetrate endothelium and interact with macrophages leading to foam cell formation and inflammation in arterial walls, thereby contributing to atherogenesis. In addition, circulating monocytes can take up TGRL and become lipid-laden foamy monocytes, which infiltrate the arterial wall and may also contribute to atherogenesis. Novel therapies targeting HTG or inflammation are in development and have potential of reducing residual ASCVD risk associated with HTG. Clinical and preclinical studies show a causal role of HTG in promoting ASCVD, in which inflammation plays a vital role. Novel therapies targeting HTG or inflammation have potential of reducing residual ASCVD risk.
Collapse
Affiliation(s)
- Xueying Peng
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China.
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
36
|
Lee ACH, Edobor A, Wigakumar T, Lysandrou M, Johnston LK, McMullen P, Mirle V, Diaz A, Piech R, Rose R, Jendrisak M, di Sabato D, Shanmugarajah K, Fung J, Donington J, Madariaga ML. Donor leukocyte trafficking during human ex vivo lung perfusion. Clin Transplant 2022; 36:e14670. [PMID: 35396887 PMCID: PMC9540615 DOI: 10.1111/ctr.14670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/09/2022] [Accepted: 04/04/2022] [Indexed: 11/30/2022]
Abstract
Background Ex vivo lung perfusion (EVLP) is used to assess and preserve lungs prior to transplantation. However, its inherent immunomodulatory effects are not completely understood. We examine perfusate and tissue compartments to determine the change in immune cell composition in human lungs maintained on EVLP. Methods Six human lungs unsuitable for transplantation underwent EVLP. Tissue and perfusate samples were obtained during cold storage and at 1‐, 3‐ and 6‐h during perfusion. Flow cytometry, immunohistochemistry, and bead‐based immunoassays were used to measure leukocyte composition and cytokines. Mean values between baseline and time points were compared by Student's t test. Results During the 1st hour of perfusion, perfusate neutrophils increased (+22.2 ± 13.5%, p < 0.05), monocytes decreased (−77.5 ± 8.6%, p < 0.01) and NK cells decreased (−61.5 ± 22.6%, p < 0.01) compared to cold storage. In contrast, tissue neutrophils decreased (−22.1 ± 12.2%, p < 0.05) with no change in monocytes and NK cells. By 6 h, perfusate neutrophils, NK cells, and tissue neutrophils were similar to baseline. Perfusate monocytes remained decreased, while tissue monocytes remained unchanged. There was no significant change in B cells or T cell subsets. Pro‐inflammatory cytokines (IL‐1b, G‐CSF, IFN‐gamma, CXCL2, CXCL1 granzyme A, and granzyme B) and lymphocyte activating cytokines (IL‐2, IL‐4, IL‐6, IL‐8) increased during perfusion. Conclusions Early mobilization of innate immune cells occurs in both perfusate and tissue compartments during EVLP, with neutrophils and NK cells returning to baseline and monocytes remaining depleted after 6 h. The immunomodulatory effect of EVLP may provide a therapeutic window to decrease the immunogenicity of lungs prior to transplantation.
Collapse
Affiliation(s)
| | - Arianna Edobor
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | | | - Maria Lysandrou
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Laura K Johnston
- Office of Shared Research Facilities, University of Chicago, Chicago, Illinois, USA
| | - Phillip McMullen
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Vikranth Mirle
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Ashley Diaz
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Ryan Piech
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Rebecca Rose
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | | | - Diego di Sabato
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | | | - John Fung
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Jessica Donington
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
37
|
Necroptosis triggers spatially restricted neutrophil-mediated vascular damage during lung ischemia reperfusion injury. Proc Natl Acad Sci U S A 2022; 119:e2111537119. [PMID: 35238643 PMCID: PMC8917381 DOI: 10.1073/pnas.2111537119] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Intravital imaging, oxidative lipidomics, and a transplant model were used to define mechanisms that regulate neutrophil recruitment into lungs during ischemia reperfusion injury, a clinically relevant form of sterile inflammation. We found that early neutrophil-mediated damage is largely confined to the subpleural vasculature, a process that is orchestrated by a spatially restricted distribution of nonclassical monocytes that produce chemokines following necroptosis of pulmonary cells. Neutrophils disrupt the integrity of subpleural capillaries, which is associated with impaired lung function. Neutrophil-mediated vascular leakage is dependent on TLR4 expression on vascular endothelium, NOX4 signaling, and formation of neutrophil extracellular traps. Our research provides insights into mechanisms that regulate neutrophil recruitment during sterile lung inflammation and lays the foundation for developing new therapies. Ischemia reperfusion injury represents a common pathological condition that is triggered by the release of endogenous ligands. While neutrophils are known to play a critical role in its pathogenesis, the tissue-specific spatiotemporal regulation of ischemia-reperfusion injury is not understood. Here, using oxidative lipidomics and intravital imaging of transplanted mouse lungs that are subjected to severe ischemia reperfusion injury, we discovered that necroptosis, a nonapoptotic form of cell death, triggers the recruitment of neutrophils. During the initial stages of inflammation, neutrophils traffic predominantly to subpleural vessels, where their aggregation is directed by chemoattractants produced by nonclassical monocytes that are spatially restricted in this vascular compartment. Subsequent neutrophilic disruption of capillaries resulting in vascular leakage is associated with impaired graft function. We found that TLR4 signaling in vascular endothelial cells and downstream NADPH oxidase 4 expression mediate the arrest of neutrophils, a step upstream of their extravasation. Neutrophil extracellular traps formed in injured lungs and their disruption with DNase prevented vascular leakage and ameliorated primary graft dysfunction. Thus, we have uncovered mechanisms that regulate the initial recruitment of neutrophils to injured lungs, which result in selective damage to subpleural pulmonary vessels and primary graft dysfunction. Our findings could lead to the development of new therapeutics that protect lungs from ischemia reperfusion injury.
Collapse
|
38
|
Van Slambrouck J, Van Raemdonck D, Vos R, Vanluyten C, Vanstapel A, Prisciandaro E, Willems L, Orlitová M, Kaes J, Jin X, Jansen Y, Verleden GM, Neyrinck AP, Vanaudenaerde BM, Ceulemans LJ. A Focused Review on Primary Graft Dysfunction after Clinical Lung Transplantation: A Multilevel Syndrome. Cells 2022; 11:cells11040745. [PMID: 35203392 PMCID: PMC8870290 DOI: 10.3390/cells11040745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 02/01/2023] Open
Abstract
Primary graft dysfunction (PGD) is the clinical syndrome of acute lung injury after lung transplantation (LTx). However, PGD is an umbrella term that encompasses the ongoing pathophysiological and -biological mechanisms occurring in the lung grafts. Therefore, we aim to provide a focused review on the clinical, physiological, radiological, histological and cellular level of PGD. PGD is graded based on hypoxemia and chest X-ray (CXR) infiltrates. High-grade PGD is associated with inferior outcome after LTx. Lung edema is the main characteristic of PGD and alters pulmonary compliance, gas exchange and circulation. A conventional CXR provides a rough estimate of lung edema, while a chest computed tomography (CT) results in a more in-depth analysis. Macroscopically, interstitial and alveolar edema can be distinguished below the visceral lung surface. On the histological level, PGD correlates to a pattern of diffuse alveolar damage (DAD). At the cellular level, ischemia-reperfusion injury (IRI) is the main trigger for the disruption of the endothelial-epithelial alveolar barrier and inflammatory cascade. The multilevel approach integrating all PGD-related aspects results in a better understanding of acute lung failure after LTx, providing novel insights for future therapies.
Collapse
Affiliation(s)
- Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Dirk Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Cedric Vanluyten
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Elena Prisciandaro
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Pulmonary Circulation Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium;
| | - Michaela Orlitová
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.O.); (A.P.N.)
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yanina Jansen
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Geert M. Verleden
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Arne P. Neyrinck
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.O.); (A.P.N.)
- Department of Anesthesiology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
| | - Laurens J. Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Correspondence:
| |
Collapse
|
39
|
Batra A, Bui TM, Rehring JF, Yalom LK, Muller WA, Sullivan DP, Sumagin R. Experimental Colitis Enhances Temporal Variations in CX3CR1 Cell Colonization of the Gut and Brain Following Irradiation. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:295-307. [PMID: 34767810 PMCID: PMC8908021 DOI: 10.1016/j.ajpath.2021.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 02/03/2023]
Abstract
Peripheral monocyte-derived CX3C chemokine receptor 1 positive (CX3CR1+) cells play important roles in tissue homeostasis and gut repopulation. Increasing evidence also supports their role in immune repopulation of the brain parenchyma in response to systemic inflammation. Adoptive bone marrow transfer from CX3CR1 fluorescence reporter mice and high-resolution confocal microscopy was used to assess the time course of CX3CR1+ cell repopulation of steady-state and dextran sodium sulfate (DSS)-inflamed small intestine/colon and the brain over 4 weeks after irradiation. CX3CR1+ cell colonization and morphologic polarization into fully ramified cells occurred more rapidly in the small intestine than in the colon. For both organs, the crypt/mucosa was more densely populated than the serosa/muscularis layer, indicating preferential temporal and spatial occupancy. Repopulation of the brain was delayed compared with that of gut tissue, consistent with the immune privilege of this organ. However, DSS-induced colon injury accelerated the repopulation. Expression analyses confirmed increased chemokine levels and macrophage colonization within the small intestine/colon and the brain by DSS-induced injury. Early increases of transmembrane protein 119 and ionized calcium binding adaptor molecule 1 expression within the brain after colon injury suggest immune-priming effect of brain resident microglia in response to systemic inflammation. These findings identify temporal differences in immune repopulation of the gut and brain in response to inflammation and show that gut inflammation can impact immune responses within the brain.
Collapse
Affiliation(s)
- Ayush Batra
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Triet M Bui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jacob F Rehring
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Lenore K Yalom
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - William A Muller
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David P Sullivan
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
40
|
Snyder ME, Bondonese A, Craig A, Popescu I, Morrell MR, Myerburg MM, Iasella CJ, Lendermon E, Pilweski J, Johnson B, Kilaru S, Zhang Y, Trejo Bittar HE, Wang X, Sanchez PG, Lakkis F, McDyer J. Rate of recipient-derived alveolar macrophage development and major histocompatibility complex cross-decoration after lung transplantation in humans. Am J Transplant 2022; 22:574-587. [PMID: 34431221 PMCID: PMC9161707 DOI: 10.1111/ajt.16812] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/02/2021] [Accepted: 08/14/2021] [Indexed: 02/03/2023]
Abstract
Alveolar macrophages (AM) play critical roles in lung tissue homeostasis, host defense, and modulating lung injury. The rate of AM turnover (donor AM replacement by circulating monocytes) after transplantation has been incompletely characterized. Furthermore, the anatomic pattern of recipient-derived lung macrophages repopulation has not been reported, nor has their ability to accumulate and present donor major histocompatibility complex (a process we refer to as MHC cross-decoration). We longitudinally characterized the myeloid content of bronchoalveolar lavage (BAL) and biopsy specimens of lung transplant recipients and found a biphasic rate in AM turnover in the allograft, with a rapid turnover perioperatively, accelerated by both the type of induction immunosuppression and the presence of primary graft dysfunction. We found that recipient myeloid cells with cell surface AM phenotype repopulated the lung in a disorganized pattern, comprised mainly of large clusters of cells. Finally, we show that recipient AM take up and present donor peptide-MHC complexes yet are not able to independently induce an in vitro alloreactive response by circulating recipient T cells.
Collapse
Affiliation(s)
- Mark E. Snyder
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania,Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anna Bondonese
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew Craig
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Iulia Popescu
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew R. Morrell
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Carlo J. Iasella
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania,Department of Pharmacology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Joseph Pilweski
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bruce Johnson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Silpa Kilaru
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Xingan Wang
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania,Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pablo G. Sanchez
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fadi Lakkis
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania,Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John McDyer
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania,Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
41
|
Ex-vivo lung perfusion therapies. Curr Opin Organ Transplant 2022; 27:204-210. [DOI: 10.1097/mot.0000000000000961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
42
|
Role of NR4A family members in myeloid cells and leukemia. CURRENT RESEARCH IN IMMUNOLOGY 2022; 3:23-36. [PMID: 35496823 PMCID: PMC9040138 DOI: 10.1016/j.crimmu.2022.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/01/2022] [Accepted: 02/10/2022] [Indexed: 11/24/2022] Open
Abstract
The myeloid cellular compartment comprises monocytes, dendritic cells (DCs), macrophages and granulocytes. As diverse as this group of cells may be, they are all an important part of the innate immune system and are therefore linked by the necessity to be acutely sensitive to their environment and to rapidly and appropriately respond to any changes that may occur. The nuclear orphan receptors NR4A1, NR4A2 and NR4A3 are encoded by immediate early genes as their expression is rapidly induced in response to various signals. It is perhaps because of this characteristic that this family of transcription factors has many known roles in myeloid cells. In this review, we will regroup and discuss the diverse roles NR4As have in different myeloid cell subsets, including in differentiation, migration, activation, and metabolism. We will also highlight the importance these molecules have in the development of myeloid leukemia. NR4A1-3 have important roles in the different cells of the myeloid compartment. These orphan receptors homeostasis, differentiation, and activation. NR4A family is important in suppressing the development of myeloid leukemias. NR4As have been linked to several diseases and could be pharmacological targets.
Collapse
|
43
|
MiRNA-122 Promotes Ischemia-Reperfusion Injury after Lung Transplantation via the Toll-like Receptor Signaling Pathway. Curr Med Sci 2021; 41:1231-1238. [PMID: 34939145 DOI: 10.1007/s11596-021-2487-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 10/22/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE MiRNAs have been recently implicated in the pathogenesis of ischemia-reperfusion (IR) injury. This study aimed to investigate the miRNA expression profiles in the early stages after lung transplantation (LT) and to study the involvement of the Toll-like receptor (TLR) signaling pathway in lung IR injury following LT. METHODS We established the left LT model in mice and selected the miRNA-122 as a research target. The mice were injected with a miRNA-122-specific inhibitor, following which pathological changes in the lung tissue were studied using different lung injury indicators. In addition, we performed deep sequencing of transplanted lung tissues to identify differentially expressed (DE) miRNAs and their target genes. These target genes were used to further perform gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. RESULTS A total of 12 DE miRNAs were selected, and 2476 target genes were identified. The GO enrichment analysis predicted 6063 terms, and the KEGG analysis predicted 1554 biological pathways. Compared with the control group, inhibiting the expression of miRNA-122 significantly reduced the lung injury and lung wet/dry ratio (P<0.05). In addition, the activity of myeloperoxidase and the expression levels of tumor necrosis factor-alpha and TLR2/4 were decreased (P<0.05); whereas the expression of interleukin-10 was increased (P<0.05). Furthermore, the inhibition of miRNA-122 suppressed the IR injury-induced activation of the TLR signaling pathway. CONCLUSION Our findings showed the differential expression of several miRNAs in the early inflammatory response following LT. Of these, miRNA-122 promoted IR injury following LT, whereas its inhibition prevented IR injury in a TLR-dependent manner.
Collapse
|
44
|
Shepherd HM, Gauthier JM, Li W, Krupnick AS, Gelman AE, Kreisel D. Innate immunity in lung transplantation. J Heart Lung Transplant 2021; 40:562-568. [PMID: 34020867 PMCID: PMC10977655 DOI: 10.1016/j.healun.2021.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 01/11/2023] Open
Abstract
Innate immune pathways early after pulmonary transplantation have been shown to cause primary graft dysfunction (PGD) and also predispose to late graft failure. Recent studies in animal models have elucidated critical mechanisms governing such innate immune responses. Here, we discuss pathways of inflammatory cell death, triggers for sterile and infectious inflammation, and signaling cascades that mediate lung injury early after transplantation. These studies highlight potential avenues for lung-specific therapies early following lung transplantation to dampen innate immune responses and improve outcomes.
Collapse
Affiliation(s)
- Hailey M Shepherd
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Jason M Gauthier
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Wenjun Li
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | | | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri.
| |
Collapse
|
45
|
Shepherd HM, Gauthier JM, Kreisel D. Tolerance, immunosuppression, and immune modulation: impacts on lung allograft survival. Curr Opin Organ Transplant 2021; 26:328-332. [PMID: 33782247 PMCID: PMC8523032 DOI: 10.1097/mot.0000000000000871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Immune responses following lung transplantation continue to result in high rates of allograft failure and rejection, and current immunosuppression does not address the unique immunologic properties of the lung. Here, we review recent studies on lung allograft tolerance and alloimmunity and discuss implications for immunosuppression. RECENT FINDINGS Processes governing tolerance and alloimmunity in lung allografts differ from other solid organs. Recent studies have suggested that allorecognition is regulated at the level of the lung graft. Furthermore, certain cell populations essential for lung allograft tolerance may facilitate rejection in other organs. Induction of lung allograft tolerance is associated with the formation of tertiary lymphoid organs, which are enriched in regulatory T cells and play an important role in preventing rejection. SUMMARY Recent discoveries regarding alloactivation and the regulation of tolerance following lung transplantation have introduced exciting potential avenues for the development of lung-specific immunosuppression.
Collapse
Affiliation(s)
- Hailey M. Shepherd
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Jason M. Gauthier
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
46
|
Hong J, Arneson D, Umar S, Ruffenach G, Cunningham CM, Ahn IS, Diamante G, Bhetraratana M, Park JF, Said E, Huynh C, Le T, Medzikovic L, Humbert M, Soubrier F, Montani D, Girerd B, Trégouët DA, Channick R, Saggar R, Eghbali M, Yang X. Single-Cell Study of Two Rat Models of Pulmonary Arterial Hypertension Reveals Connections to Human Pathobiology and Drug Repositioning. Am J Respir Crit Care Med 2021; 203:1006-1022. [PMID: 33021809 PMCID: PMC8048757 DOI: 10.1164/rccm.202006-2169oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: The cellular and molecular landscape and translational value of commonly used models of pulmonary arterial hypertension (PAH) are poorly understood. Single-cell transcriptomics can enhance molecular understanding of preclinical models and facilitate their rational use and interpretation.Objectives: To determine and prioritize dysregulated genes, pathways, and cell types in lungs of PAH rat models to assess relevance to human PAH and identify drug repositioning candidates.Methods: Single-cell RNA sequencing was performed on the lungs of monocrotaline (MCT), Sugen-hypoxia (SuHx), and control rats to identify altered genes and cell types, followed by validation using flow-sorted cells, RNA in situ hybridization, and immunofluorescence. Relevance to human PAH was assessed by histology of lungs from patients and via integration with human PAH genetic loci and known disease genes. Candidate drugs were predicted using Connectivity Map.Measurements and Main Results: Distinct changes in genes and pathways in numerous cell types were identified in SuHx and MCT lungs. Widespread upregulation of NF-κB signaling and downregulation of IFN signaling was observed across cell types. SuHx nonclassical monocytes and MCT conventional dendritic cells showed particularly strong NF-κB pathway activation. Genes altered in SuHx nonclassical monocytes were significantly enriched for PAH-associated genes and genetic variants, and candidate drugs predicted to reverse the changes were identified. An open-access online platform was developed to share single-cell data and drug candidates (http://mergeomics.research.idre.ucla.edu/PVDSingleCell/).Conclusions: Our study revealed the distinct and shared dysregulation of genes and pathways in two commonly used PAH models for the first time at single-cell resolution and demonstrated their relevance to human PAH and utility for drug repositioning.
Collapse
Affiliation(s)
- Jason Hong
- Division of Pulmonary and Critical Care Medicine
| | | | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, and
| | | | | | - In Sook Ahn
- Department of Integrative Biology and Physiology
| | | | - May Bhetraratana
- Division of Cardiology, University of California Los Angeles, Los Angeles, California
| | - John F. Park
- Department of Anesthesiology and Perioperative Medicine, and
| | - Emma Said
- Department of Anesthesiology and Perioperative Medicine, and
| | | | - Trixie Le
- Department of Anesthesiology and Perioperative Medicine, and
| | | | - Marc Humbert
- Department of Respiratory and Intensive Care Medicine, Bicêtre Hospital, University of Paris-Saclay, National Institute of Health and Medical Research Joint Research Unit S 999, Public Assistance Hospitals of Paris, Le Kremlin-Bicêtre, France
| | - Florent Soubrier
- Institut Hospitalo–Universitaire Cardiométabolisme et Nutrition, Paris, France; and
| | - David Montani
- Department of Respiratory and Intensive Care Medicine, Bicêtre Hospital, University of Paris-Saclay, National Institute of Health and Medical Research Joint Research Unit S 999, Public Assistance Hospitals of Paris, Le Kremlin-Bicêtre, France
| | - Barbara Girerd
- Department of Respiratory and Intensive Care Medicine, Bicêtre Hospital, University of Paris-Saclay, National Institute of Health and Medical Research Joint Research Unit S 999, Public Assistance Hospitals of Paris, Le Kremlin-Bicêtre, France
| | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center, University of Bordeaux, National Institute of Health and Medical Research Joint Research Unit 1219, Bordeaux, France
| | | | - Rajan Saggar
- Division of Pulmonary and Critical Care Medicine
| | | | - Xia Yang
- Department of Integrative Biology and Physiology
| |
Collapse
|
47
|
Zhou P, Guo H, Li Y, Liu Q, Qiao X, Lu Y, Mei PY, Zheng Z, Li J. Monocytes promote pyroptosis of endothelial cells during lung ischemia-reperfusion via IL-1R/NF-κB/NLRP3 signaling. Life Sci 2021; 276:119402. [PMID: 33785335 DOI: 10.1016/j.lfs.2021.119402] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/07/2021] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
In our previous study, we observed that donor pulmonary intravascular nonclassical monocytes play a major role in early PGF, but the specific mechanism remained unclear. In this study, we investigated the mechanistic role of monocytes in inducing pyroptosis of human pulmonary microvascular endothelial cells (HPMECs) during IRI. A murine hilar ligation model of IRI was utilized whereby left lungs underwent 1 h of ischemia and 23 h of reperfusion. Monocyte depletion by intraperitoneal clodronate-liposome treatment on pulmonary edema and pyroptosis activation were determined. In vitro experiments, we performed the co-culture experiments under hypoxia-reoxygenation (H/R) conditions to mimic the IRI environment. We monitored the expression of NLRP3, caspase-1 and IL-1β in co-cultures of monocytes (U937 cells) and HPMECs under H/R conditions. NLRP3, IL-1β and IL-1R siRNA knockdown, caspase-1 and NF-κB pathway inhibitors were employed to elucidate the mechanism modulating HPMEC pyroptosis during H/R. Treatment of mice with clodronate-liposome attenuated IR-induced pulmonary edema, cytokine production and pyroptosis activation. In vitro, NLRP3 knockdown in monocytes reduced caspase-1 and IL-1β secretion in co-cultures of monocytes and HPMECs. Reduced HPMEC pyroptosis was also observed either containing HPMECs with genetically engineered IL-1R knockdown or in co-culture treated with a Triplotide inhibitor that disrupts NF-κB signaling. Monocytes play a vital role in the development of transplant-associated ischemia-reperfusion injury. A potential role is that monocytes secrete IL-1β to induce HPMEC pyroptosis via the IL-1R/NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Guo
- Department of Thyroid and Breast Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan Liu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinwei Qiao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Lu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei-Yuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhikun Zheng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jinsong Li
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
48
|
Yang W, Chiu S, Querrey M, Liu X, Wu Q, Cerier E, Islam MBAR, Schwulst SJ, Budinger GRS, Mohanakumar T, Lecuona E, Bharat A. Nonclassical Monocytes Promote Edema in Lung Allografts from Traumatic Brain Injury Donors. Am J Respir Cell Mol Biol 2021; 64:391-394. [PMID: 33646091 DOI: 10.1165/rcmb.2020-0137le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Wenbin Yang
- Northwestern University Chicago, Illinois and
| | | | | | | | - Qiang Wu
- Northwestern University Chicago, Illinois and
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kurihara C, Lecuona E, Wu Q, Yang W, Núñez-Santana FL, Akbarpour M, Liu X, Ren Z, Li W, Querrey M, Ravi S, Anderson ML, Cerier E, Sun H, Kelly ME, Abdala-Valencia H, Shilatifard A, Mohanakumar T, Budinger GRS, Kreisel D, Bharat A. Crosstalk between nonclassical monocytes and alveolar macrophages mediates transplant ischemia-reperfusion injury through classical monocyte recruitment. JCI Insight 2021; 6:147282. [PMID: 33621212 PMCID: PMC8026186 DOI: 10.1172/jci.insight.147282] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 12/24/2022] Open
Abstract
Primary graft dysfunction (PGD) is the predominant cause of early graft loss following lung transplantation. We recently demonstrated that donor pulmonary intravascular nonclassical monocytes (NCM) initiate neutrophil recruitment. Simultaneously, host-origin classical monocytes (CM) permeabilize the vascular endothelium to allow neutrophil extravasation necessary for PGD. Here, we show that a CCL2-CCR2 axis is necessary for CM recruitment. Surprisingly, although intravital imaging and multichannel flow cytometry revealed that depletion of donor NCM abrogated CM recruitment, single cell RNA sequencing identified donor alveolar macrophages (AM) as predominant CCL2 secretors. Unbiased transcriptomic analysis of murine tissues combined with murine KOs and chimeras indicated that IL-1β production by donor NCM was responsible for the early activation of AM and CCL2 release. IL-1β production by NCM was NLRP3 inflammasome dependent and inhibited by treatment with a clinically approved sulphonylurea. Production of CCL2 in the donor AM occurred through IL-1R-dependent activation of the PKC and NF-κB pathway. Accordingly, we show that IL-1β-dependent paracrine interaction between donor NCM and AM leads to recruitment of recipient CM necessary for PGD. Since depletion of donor NCM, IL-1β, or IL-1R antagonism and inflammasome inhibition abrogated recruitment of CM and PGD and are feasible using FDA-approved compounds, our findings may have potential for clinical translation.
Collapse
Affiliation(s)
| | | | - Qiang Wu
- Division of Thoracic Surgery and
| | | | | | | | | | - Ziyou Ren
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Wenjun Li
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | | | | | | | | | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniel Kreisel
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ankit Bharat
- Division of Thoracic Surgery and.,Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
50
|
Ma Y, Yan G, Guo J, Li F, Zheng H, Wang C, Chen Y, Ye Y, Dai H, Qi Z, Zhuang G. Berberine Prolongs Mouse Heart Allograft Survival by Activating T Cell Apoptosis via the Mitochondrial Pathway. Front Immunol 2021; 12:616074. [PMID: 33732240 PMCID: PMC7959711 DOI: 10.3389/fimmu.2021.616074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/18/2021] [Indexed: 11/25/2022] Open
Abstract
Berberine, which is a traditional Chinese medicine can inhibit tumorigenesis by inducing tumor cell apoptosis. However, the immunoregulatory of effects berberine on T cells remains poorly understood. Here, we first examined whether berberine can prolong allograft survival by regulating the recruitment and function of T cells. Using a major histocompatibility complex complete mismatch mouse heterotopic cardiac transplantation model, we found that the administration of moderate doses (5 mg/kg) of berberine significantly prolonged heart allograft survival to 19 days and elicited no obvious berberine-related toxicity. Compared to that with normal saline treatment, berberine treatment decreased alloreactive T cells in recipient splenocytes and lymph node cells. It also inhibited the activation, proliferation, and function of alloreactive T cells. Most importantly, berberine treatment protected myocardial cells by decreasing CD4+ and CD8+ T cell infiltration and by inhibiting T cell function in allografts. In vivo and in vitro assays revealed that berberine treatment eliminated alloreactive T lymphocytes via the mitochondrial apoptosis pathway, which was validated by transcriptome sequencing. Taken together, we demonstrated that berberine prolongs allograft survival by inducing apoptosis of alloreactive T cells. Thus, our study provides more evidence supporting the potential use of berberine in translational medicine.
Collapse
Affiliation(s)
- Yunhan Ma
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute, Xiamen University, Xiamen, China
| | - Guoliang Yan
- School of Medicine, Xiamen University, Xiamen, China
| | - Junjun Guo
- School of Medicine, Xiamen University, Xiamen, China
| | - Fujun Li
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haiping Zheng
- School of Medicine, Xiamen University, Xiamen, China
| | - Chenxi Wang
- School of Medicine, Xiamen University, Xiamen, China
| | - Yingyu Chen
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute, Xiamen University, Xiamen, China
| | - Yuhan Ye
- Department of Pathology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Helong Dai
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, China.,Clinical Immunology Center, Central South University, Changsha, China
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning, China
| | - Guohong Zhuang
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute, Xiamen University, Xiamen, China
| |
Collapse
|