1
|
Chen ST, Jheng CY, Lee YC, Huang WC, Lee SY, Chen YH. Intermittent hypoxia-reoxygenation-induced miRNAs inhibit expression of IRF and interferon genes but activate NF-κB and expression of pulmonary fibrosis markers in human small airway epithelial cells. Life Sci 2025; 370:123569. [PMID: 40120975 DOI: 10.1016/j.lfs.2025.123569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
AIM Intermittent hypoxia-reoxygenation (H/R) has been demonstrated to be associated with aviation and various respiratory diseases, and hence it is of interest to unravel the regulatory mechanisms underlying the H/R-induced innate immune and inflammatory responses in both healthy and COPD-diseased human small airway epithelial cells (SAECs). MAIN METHODS The normal healthy and COPD-diseased SAECs (i.e., N-SAECs and D-SAECs) were purchased from PromoCell biotechnology company and respectively cultured under normoxia (21 % O2) or 12/12-h cycles of H/R (i.e., 1 % O2 and 21 % O2 alternately) for 6 days in total for 2D cultures and 21 days in total for the air-liquid interface 3D cultures, followed by qPCR analyses, miRNA fluorescence in situ hybridization, luciferase reporter assays, and immunofluorescence staining. KEY FINDINGS Human SAECs cultured under 12/12-h cycles of H/R showed dramatically increased expression of HIF1A and the H/R-inducible miRNAs miR-129-1-3p, miR-1290 and miR-193b-5p, with miR-129-1-3p and miR-193b-5p targeting and inhibiting IRF5 and IRF7 mRNAs, hence downregulating both the type I and II interferon genes in SAECs cultured under H/R. In addition, miR-129-1-3p, miR-1290 and miR-193b-5p all targeted and inhibited SOCS3 mRNA, hence upregulating transactivation of NF-κB and in turn inducing expression of the inflammatory chemokine genes and pulmonary fibrosis-associated marker genes. SIGNIFICANCE We show for the first time that intermittent H/R upregulates the NF-κB-induced proinflammatory and fibrosis marker genes whereas downregulates the IRF5/7-induced type I/II interferon expression in human SAECs through distinct HIF1A-inducible miRNAs miR-129-1-3p, miR-193b-5p and miR-1290, which may serve as promising therapeutic targets for airway inflammation and pulmonary fibrosis.
Collapse
Affiliation(s)
- Shiuan-Ting Chen
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei City, Taiwan
| | - Cheng-Yu Jheng
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Yu-Chun Lee
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei City, Taiwan
| | - Wei-Chen Huang
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei City, Taiwan
| | - Shih-Yu Lee
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei City, Taiwan
| | - Yi-Hui Chen
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei City, Taiwan.
| |
Collapse
|
2
|
Langer F, Starniske I, Weingard B, Aliyev P, Mustafi M, Bals R, Wilkens H. Single lung transplantation for pulmonary fibrosis: Does side matter? JHLT OPEN 2025; 8:100229. [PMID: 40144718 PMCID: PMC11935513 DOI: 10.1016/j.jhlto.2025.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background The implementation of the Lung Allocation Score in the Eurotransplant international collaborative framework decreased waiting list mortality, but organ shortage remains a significant problem. Single lung transplantation (sLTx)-whenever possible-may decrease waiting list mortality. We have consistently employed sLTx for recipients with pulmonary fibrosis. In the current investigation, we sought to analyze if this strategy can lead to an acceptable long-term outcome and if the side of sLTx has an impact on the outcome. Methods Between 1995 and 2024, we performed 138 sLTx for patients with pulmonary fibrosis (54 ± 9 years, 88 male). Data and outcomes were analyzed retrospectively comparing recipients receiving left sLTX (n = 98) and right sLTx (n = 40). Results Survival was 83%, 59%, and 29% at 1, 5, and 10 years for the total patient cohort. Survival was similar for left and right sLTx (83 vs 81%, 58 vs 64%, and 29 vs 28% at 1, 5, and 10 years, p = 0.54). Left and right transplantations lead to similar best post-transplant forced expiratory volume per second (74% ± 20% vs 74% ± 21%, p = 0.86). While the total lung capacity (TLC) ratio TLCdonor/predicted TLCrecipient was similar between groups (104% vs 100%), the ratio TLCdonor/actual TLCrecipient was higher in left sLTx (185% vs 158%, p = 0.04). On multivariate regression analysis, postoperative pneumonia (p = 0.003, hazard ratio 3.404) and sepsis (p = 0.002, hazard ratio 10.700) were identified as predictors for early mortality. Conclusions Performing sLTx for pulmonary fibrosis patients can be an effective strategy to optimize donor utilization and improve outcomes-irrespective of graft side.
Collapse
Affiliation(s)
- Frank Langer
- Department of Thoracic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Ina Starniske
- Department of Thoracic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Bettina Weingard
- Department of Internal Medicine V, Pneumology and Intensive Care Medicine, Saarland University Medical Center, Homburg, Germany
| | - Parviz Aliyev
- Department of Thoracic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Migdat Mustafi
- Department of Thoracic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Robert Bals
- Department of Internal Medicine V, Pneumology and Intensive Care Medicine, Saarland University Medical Center, Homburg, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University Campus, Saarbrücken, Germany
| | - Heinrike Wilkens
- Department of Internal Medicine V, Pneumology and Intensive Care Medicine, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
3
|
Zhao Y, Liang L, McCaig A, Aujla T, Keshavjee S, Liu M. Activation of PANoptosis and ferroptosis during ex vivo lung perfusion in human lungs. J Heart Lung Transplant 2025:S1053-2498(25)01910-2. [PMID: 40288429 DOI: 10.1016/j.healun.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/10/2024] [Accepted: 11/26/2024] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND A recent study demonstrated upregulation of PANoptosis-related genes during reperfusion in human lung transplants. However, the impact of ex vivo lung perfusion (EVLP) on different cell death pathways and their relationship with inflammatory genes and clinical characteristics remains unknown. METHODS We conducted transcriptomic analyses on pre- and post-EVLP biopsies from 49 donation after brain death (DBD) and 39 donation after circulatory death (DCD) lungs. Gene set enrichment analysis (GSEA) and single-sample GSEA were used to assess the enrichment of cell death and inflammatory pathways. We further explored the relationships between these pathways, donor characteristics, and clinical outcomes. RESULTS DBD lungs showed significant enrichment of apoptosis and ferroptosis gene sets compared to DCD lungs. During EVLP, pyroptosis, apoptosis, necroptosis, and ferroptosis gene sets were significantly upregulated and strongly correlated with inflammatory pathways in both DBD and DCD donor lungs. Donor age, sex, and smoking history were associated with specific cell death pathways. In DCD lungs, the expression of ferroptosis-related genes was associated with recipient early outcomes. CONCLUSION The expression of cell death gene sets is donor-type specific. The identification of multiple cell death and inflammatory pathways during EVLP provides potential therapeutic targets to improve donor lung quality and enhance clinical outcomes.
Collapse
Affiliation(s)
- Yajin Zhao
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Lubiao Liang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Abby McCaig
- Latner Thoracic Surgery 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
| | - Tanroop Aujla
- 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; 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 Surgery 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.
| |
Collapse
|
4
|
Ribeiro RVP, Altarabsheh SE, Segamanasinghe DL, Lopez-Muralles IW, Garcia Moreno AS, Knop G, Al-Azzam F, Mallea J, Brownfield DG, Olson AC, Gibbard DF, Jing Y, Cheng K, Saddoughi SA. Preconditioning donor lungs with lung-derived exosomes mitigates ischemia-reperfusion injury in a warm ischemia porcine DCD model. J Heart Lung Transplant 2025:S1053-2498(25)01856-X. [PMID: 40127773 DOI: 10.1016/j.healun.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Donation after circulatory death (DCD) donors remain an underutilized source in the United States due to concerns of ischemia-reperfusion injury (IRI) after prolonged ischemic times. Lung-derived exosomes have shown potential in mitigating pulmonary fibrosis by promoting lung repair. Here, we sought to investigate the potential of lung-derived exosomes to prevent and repair lung IRI. METHODS We used a porcine DCD model to induce lung injury. Following the determination of optimal warm ischemic time (WIT), donor pigs were allocated into 3 study groups (n = 5, each): control, pre-DCD exosome treatment, and post-DCD exosomes treatment. Lungs were assessed using ex-vivo lung perfusion (EVLP) for functional parameters, histologic evaluation, and molecular analysis of inflammatory markers and oxidative stress. RESULTS A 1-hour WIT induced consistent lung injury, which was ameliorated with pre-DCD exosome treatment exhibiting significantly improved lung function during EVLP compared to controls. This group presented higher pO2, better lung compliance, lower airway pressures, and reduced pulmonary vascular resistance. Histologic analysis indicated reduced edema, vascular congestion, and leukocyte infiltration. Key inflammatory cytokines such as IL-6, IL-1β, and TNF-α were significantly downregulated, and reactive oxygen species levels were lower than controls. Despite inferior response compared to pre-DCD treatment, post-DCD exosome treatment also improved lung function and reduced edema formation, with significant decrease in inflammation. CONCLUSIONS Lung-derived exosome therapy significantly mitigates IRI in a porcine DCD model, improving lung function and reducing inflammation and oxidative stress. These findings support the potential of exosome therapy to increase donor lung utilization, warranting further mechanistic and clinical studies.
Collapse
Affiliation(s)
- Rafaela V P Ribeiro
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Ana S Garcia Moreno
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Gustavo Knop
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
| | - Fuad Al-Azzam
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
| | - Jorge Mallea
- Department of Critical Care Medicine, Mayo Clinic, Jacksonville, Florida
| | - Douglas G Brownfield
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Aaron C Olson
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Daniel F Gibbard
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yujia Jing
- Xsome Biotech Inc., Raleigh, North Carolina
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York City, New York
| | - Sahar A Saddoughi
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota; Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
5
|
Deitz RL, Chan EG, Ryan JP, Coster JN, Furukawa M, Hage CA, Sanchez PG. Adoption of a semi-elective lung transplantation practice by safely extending cold ischemic times. J Thorac Cardiovasc Surg 2025; 169:813-821. [PMID: 39393628 DOI: 10.1016/j.jtcvs.2024.09.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/05/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVE Lung transplantation is a complex surgical procedure performed by specialized teams. Practice changes to eliminate overnight lung transplants were implemented at our center and patient outcomes were evaluated. METHODS Patient and donor organ selection were performed in the standard fashion. All donors with a crossclamp after 6 pm matched to any of our listed recipients-independent of their surgical complexity or risk-were kept in a temperature-controlled iceless cooler from procurement to recipient implant. All recipients had a 7 am in-room start. Data were prospectively collected and compared with a cohort of recipients from the previous fifteen months. RESULTS In total, 82 transplants were performed at a single academic institution between July 1, 2022, and January 7, 2024, 22% of which included allografts with extended ischemic times using the iceless cooler (n = 18) with a median average temperature of 6.81 °C. Median ischemic times were 13.9 (12.5-15.6) hours, more than twice the length of ischemic times in the standard group (n = 64, 6.8 [6.1-7.4] hours). Postoperative outcomes were similar between groups, including postoperative intensive care unit duration (12 vs 9 days in the standard group), length of stay (24 vs 20 days), primary graft dysfunction grade (17% vs 20%), postoperative extracorporeal membrane oxygenation (22% vs 20%), and 6-month survival (94% vs 91%). CONCLUSIONS Donor lungs preserved in an iceless cooler were successfully transplanted after extended cold ischemic times. Adoption of a semielective transplant strategy can be successfully implemented without compromising patient outcomes. Additional advantages may be gleaned through daytime transplantation with standard transplant surgical teams rather than overnight, on-call, teams.
Collapse
Affiliation(s)
- Rachel L Deitz
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa.
| | - Ernest G Chan
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - John P Ryan
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Jenalee N Coster
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Masashi Furukawa
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Chadi A Hage
- Department of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Pablo G Sanchez
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| |
Collapse
|
6
|
Costamagna A, Balzani E, Marro M, Simonato E, Burello A, Rinaldi M, Brazzi L, Boffini M, Fanelli V. Association of Inflammatory Profile During Ex Vivo Lung Perfusion With High-Grade Primary Graft Dysfunction: A Systematic Review and Meta-Analysis. Transpl Int 2025; 38:13794. [PMID: 39944219 PMCID: PMC11815944 DOI: 10.3389/ti.2025.13794] [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: 09/12/2024] [Accepted: 01/02/2025] [Indexed: 05/09/2025]
Abstract
PGD3 is the manifestation of ischemia-reperfusion injury which results from inflammation and cell death and is associated with poor outcome. This systematic-review and meta-analysis of non-randomized controlled trials on patients undergoing Ltx with reconditioned lungs via EVLP, aims to assess the association between the levels of proinflammatory biomarkers during EVLP and PGD3 development within the firsts 72 h post-Ltx. Biomarkers were categorized by timing (1-hour, T0 and 4-hours, Tend from EVLPstart) and by their biological function (adhesion molecules, chemokines, cytokines, damage-associated-molecular-patterns, growth-factors, metabolites). We employed a four-level mixed-effects model with categorical predictors for biomarker groups to identify differences between patients with PGD3 and others. The single study and individual measurements were considered random intercepts. We included 8 studies (610 measurements at T0 and 884 at Tend). The pooled effect was 0.74 (p = 0.021) at T0, and 0.90 (p = 0.0015) at Tend. The four-level model indicated a large pooled correlation between developing PGD3 at 72 h post-Ltx and inflammatory biomarkers values, r = 0.62 (p = 0.009). Chemokine group showed the strongest association with the outcome (z-value = 1.26, p = 0.042). Pooled panels of inflammation markers, particularly chemokines, measured at T0 or at Tend, are associated with the development of PGD3 within the first 72 h after Ltx. Systematic Review Registration https://osf.io/gkxzh/.
Collapse
Affiliation(s)
| | - Eleonora Balzani
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Matteo Marro
- Cardiovascular and Thoracic Department, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Erika Simonato
- Cardiovascular and Thoracic Department, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - Mauro Rinaldi
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Luca Brazzi
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Massimo Boffini
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Vito Fanelli
- Department of Surgical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
7
|
Chu KA, Lai CY, Chen YH, Kuo FH, Chen IY, Jiang YC, Liu YL, Ko TL, Fu YS. An animal model of severe acute respiratory distress syndrome for translational research. Lab Anim Res 2025; 41:4. [PMID: 39856771 PMCID: PMC11758736 DOI: 10.1186/s42826-025-00235-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Despite the fact that an increasing number of studies have focused on developing therapies for acute lung injury, managing acute respiratory distress syndrome (ARDS) remains a challenge in intensive care medicine. Whether the pathology of animal models with acute lung injury in prior studies differed from clinical symptoms of ARDS, resulting in questionable management for human ARDS. To evaluate precisely the therapeutic effect of transplanted stem cells or medications on acute lung injury, we developed an animal model of severe ARDS with lower lung function, capable of keeping the experimental animals survive with consistent reproducibility. Establishing this animal model could help develop the treatment of ARDS with higher efficiency. RESULTS In this approach, we intratracheally delivered bleomycin (BLM, 5 mg/rat) into rats' left trachea via a needle connected with polyethylene tube, and simultaneously rotated the rats to the left side by 60 degrees. Within seven days after the injury, we found that arterial blood oxygen saturation (SpO2) significantly decreased to 83.7%, partial pressure of arterial oxygen (PaO2) markedly reduced to 65.3 mmHg, partial pressure of arterial carbon dioxide (PaCO2) amplified to 49.2 mmHg, and the respiratory rate increased over time. Morphologically, the surface of the left lung appeared uneven on Day 1, the alveoli of the left lung disappeared on Day 2, and the left lung shrank on Day 7. A histological examination revealed that considerable cell infiltration began on Day 1 and lasted until Day 7, with a larger area of cell infiltration. Serum levels of IL-5, IL-6, IFN-γ, MCP-1, MIP-2, G-CSF, and TNF-α substantially rose on Day 7. CONCLUSIONS This modified approach for BLM-induced lung injury provided a severe, stable, and one-sided (left-lobe) ARDS animal model with consistent reproducibility. The physiological symptoms observed in this severe ARDS animal model are entirely consistent with the characteristics of clinical ARDS. The establishment of this ARDS animal model could help develop treatment for ARDS.
Collapse
Affiliation(s)
- Kuo-An Chu
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, National Sun Yat-Sen University, No. 70, Lienhai Rd., Kaohsiung, Taiwan, ROC
- School of Nursing, Fooyin University, Kaohsiung, Taiwan, ROC
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan, ROC
| | - Chia-Yu Lai
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Hui Chen
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Fu-Hsien Kuo
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - I-Yuan Chen
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - You-Cheng Jiang
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Ya-Ling Liu
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Tsui-Ling Ko
- School of Medicine, College of Medicine, National Sun Yat-Sen University, No. 70, Lienhai Rd., Kaohsiung, Taiwan, ROC.
| | - Yu-Show Fu
- Department of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nung Street, Taipei, Taiwan, ROC.
| |
Collapse
|
8
|
Zaaqoq AM, Mazzeffi MA, Vogelsong MA, Roeser M, Cho SM. Lung injury in myocardial infarction-associated cardiogenic shock supported by venoarterial extracorporeal membrane oxygenation: a scoping review. BMC Cardiovasc Disord 2025; 25:40. [PMID: 39849351 PMCID: PMC11756158 DOI: 10.1186/s12872-025-04472-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Acute lung injury and acute respiratory failure are frequent complications of cardiogenic shock and are associated with increased morbidity and mortality. Even with increased use of temporary mechanical circulatory support, such as venoarterial extracorporeal membrane oxygenation (VA-ECMO), acute lung injury related to cardiogenic shock continues to have a determinantal effect on patient outcomes. OBJECTIVES To summarize potential mechanisms of acute lung injury described in patients with cardiogenic shock supported by VA-ECMO and determine current knowledge gaps. METHODS We searched literature from January 1st, 2010, to December 31st, 2023, using MEDLINE, EMBASE, and Web of Science databases on February 27th, 2024. The search strategy was split into two main domains: (a) cardiogenic shock and ECMO and (b) Acute respiratory failure and ECMO. RESULTS The search yielded 2246 citations. After 743 duplicates were removed, 1465 citations remained. Of these studies, 1456 were excluded based on the exclusion criteria, leaving the final eight studies we included in our scoping review. We identified disruption of the pulmonary blood flow in patients with cardiogenic shock, with cardiac arrest being an extreme form of cardiogenic shock. Placing the patient on VA-ECMO could intensify this process of lung injury. CONCLUSION Acute lung injury in patients with cardiogenic shock, especially when supported by VA ECMO, is a significant complication that is associated with increased morbidity and mortality. There is a limited understanding of the underlying mechanisms that could represent opportunities for future research to mitigate its development and provide the best approach to protecting and monitoring lung function.
Collapse
Affiliation(s)
- Akram M Zaaqoq
- Department of Anesthesiology, Division of Critical Care, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA.
| | - Michael A Mazzeffi
- Department of Anesthesiology, Division of Critical Care, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA
| | - Melissa A Vogelsong
- Department of Anesthesiology, Division of Cardiac Anesthesia, Stanford University, Stanford, CA, USA
| | - Mark Roeser
- Department of Surgery, Division of Cardiac Surgery, University of Virginia, Charlottesville, VA, USA
| | - Sung-Min Cho
- Department of Surgery, Division of Cardiac Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Departments of Neurology, Anesthesiology, Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
da Silva Battochio A, Tavares ER, Correia AT, de Almeida FM, Carvalho PO, Guido MC, Pêgo-Fernandes PM, Maranhão RC, Pazetti R. Effects of paclitaxel and methotrexate associated with cholesterol-rich nanoemulsions on ischemia-reperfusion injury after unilateral lung transplantation in rats. Sci Rep 2024; 14:31695. [PMID: 39738212 PMCID: PMC11686372 DOI: 10.1038/s41598-024-81337-7] [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: 07/24/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Currently, the barrier to successful lung transplantation is ischemia and reperfusion injury, which can lead to the development of bronchiolitis obliterans. Paclitaxel and methotrexate are drugs known to inhibit cell proliferation and have anti-inflammatory effects, and the association of these drugs with cholesterol-rich nanoparticles has been shown to be beneficial in the treatment of other transplanted organs. Thirty-three male Sprague Dawley rats were divided into 3 groups: Basal group, no intervention; Control group, received only nanoparticles; Drug group, paclitaxel and methotrexate treatment. Donors and recipients were treated with nanoparticle-paclitaxel and nanoparticle-methotrexate, respectively, 24 h before surgery. The donor lungs from the Drug group were perfused with a preservation solution supplemented with nanoparticles-paclitaxel. After 12 h, the left lung was implanted and reperfused for 1 h. Recipients had an increase in erythrocytes, neutrophils and hemoglobin and a decrease in lymphocytes, and an increase in oxygenation and lactate and a decrease in carbon dioxide. These animals showed an increase in urea and creatinine. The grafts showed perivascular edema and hemorrhage, as well as elevated values of airway resistance, tissue resistance and tissue elastance under mechanical ventilation. The tested drugs were not effective in attenuating the effects of ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Angela da Silva Battochio
- Programa de Pos-Graduacao em Cirurgia Toracica e Cardiovascular, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- Laboratorio de Pesquisa em Cirurgia Toracica, Departamento de Cardiopneumologia, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Elaine Rufo Tavares
- Laboratorio de Metabolismo e Lipides, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Aristides Tadeu Correia
- Laboratorio de Pesquisa em Cirurgia Toracica, Departamento de Cardiopneumologia, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Francine Maria de Almeida
- Programa de Pos-Graduacao em Cirurgia Toracica e Cardiovascular, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- Laboratorio de Pesquisa em Cirurgia Toracica, Departamento de Cardiopneumologia, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Priscila Oliveira Carvalho
- Laboratorio de Metabolismo e Lipides, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria Carolina Guido
- Laboratorio de Metabolismo e Lipides, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Paulo Manuel Pêgo-Fernandes
- Laboratorio de Pesquisa em Cirurgia Toracica, Departamento de Cardiopneumologia, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Raul Cavalcante Maranhão
- Laboratorio de Metabolismo e Lipides, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Rogerio Pazetti
- Laboratorio de Pesquisa em Cirurgia Toracica, Departamento de Cardiopneumologia, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
- Laboratorio de Pesquisa em Cirurgia Toracica-LIM61, Faculdade de Medicina da Universidade de Sao Paulo - FMUSP, Av. Dr. Arnaldo 455 - 1. Andar, sala 1220, Sao Paulo, SP, CEP 01246-903, Brazil.
| |
Collapse
|
10
|
Ye X, Pei F, Li W, Xue J, Huang X, Huang J, Zhang L. Fibroblast growth factor 21 attenuates pulmonary ischemia/reperfusion injury via inhibiting endoplasmic reticulum stress-induced ferroptosis though FGFR1/PPARδ signaling pathway. Int Immunopharmacol 2024; 143:113307. [PMID: 39366074 DOI: 10.1016/j.intimp.2024.113307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Acute lung injury is a critical life-threatening complication of pulmonary and cardiac surgery with a high rate of morbidity and mortality. Fibroblast growth factor 21 (FGF21) has been reported to play an important role in protecting vital organs from damage. This study aims to investigate the potential protective role and mechanism of FGF21 in pulmonary ischemia/reperfusion (I/R)-induced acute lung injury. METHODS A pulmonary epithelial cell line was treated with hypoxia/regeneration (H/R) in vitro and a mouse model of acute lung injury was induced with pulmonary I/R in vivo. Lung injury after pulmonary I/R was compared between FGF21-konckout (KO) mice and wild-type (WT) mice. Recombinant FGF21 was administrated in vivo and in vitro to determine its therapeutic effect. RESULTS Circulating levels of FGF21 in mice with pulmonary I/R injury were significantly higher than in those without pulmonary I/R injury. Lung injury was aggravated in FGF21-KO mice compared with WT mice and the administration of FGF21 alleviated lung injury in mouse treated with I/R and pulmonary epithelial cell injury treated with H/R. FGF21 treatment decreased endoplasmic reticulum (ER) stress, Fe2+ and lipid reactive oxygen species (ROS) contents and GPX4 expression and increased PTGS2 levels. Mechanistically, FGF21 upregulated the expression of FGFR1 and PPARδ, ameliorated ER stress and ER stress induced-ferroptosis. Furthermore, FGF21 increased the expression level of PPARδ in pulmonary epithelial cell exposed to H/R, which was inhibited by FGFR1 inhibitor (PD173074). The protective effects of FGF21 were abolished by co-treatment with PPARδ inhibitor (GSK0660), indicating FGF21 attenuated ER stress-induced ferroptosis by dependent on FGFR1/PPARδ signaling pathway. CONCLUSION Our study reveals that FGF21 protects against pulmonary I/R injury via inhibiting ER stress-induced ferroptosis though FGFR1/PPARδ signaling pathway. Boosting endogenous FGF21 or the administration of recombinant FGF21 could be promising therapeutic strategies for pulmonary IRI.
Collapse
Affiliation(s)
- Xinqiao Ye
- Department of Thoracic Surgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou 341009, Jiangxi, China
| | - Fang Pei
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou 341009, Jiangxi, China.
| | - Wei Li
- People's Hospital of Ningdu County, Ganzhou 342899, Jiangxi, China
| | - Jinping Xue
- Xinfeng Hospital of Traditional Chinese Midicine, Ganzhou 341699, Jiangxi, China
| | - Xiuyun Huang
- Dingnan Second Hospital, Ganzhou 341999, Jiangxi, China
| | - Jianming Huang
- Department of Thoracic Surgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou 341009, Jiangxi, China
| | - Liyan Zhang
- Department of Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 519041, Guangdong, China
| |
Collapse
|
11
|
Wu W, Meng F, Zhang H, Tian H, Zhang X. Neutrophil PPIF exacerbates lung ischemia-reperfusion injury after lung transplantation by promoting calcium overload-induced neutrophil extracellular traps formation. Int Immunopharmacol 2024; 142:113051. [PMID: 39236457 DOI: 10.1016/j.intimp.2024.113051] [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: 01/04/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Lung ischemia-reperfusion (I/R) injury is the main risk factor for primary graft dysfunction and patient death after lung transplantation (LTx). It is widely accepted that the main pathological mechanism of lung I/R injury are calcium overload, oxygen free radical explosion and neutrophil-mediated damage, which leading to the lack of effective treatment options. The aim of this study was to further explore the mechanisms of lung I/R injury after LTx and to provide potential therapeutic strategies. Our bioinformatics analysis revealed that the neutrophil extracellular traps (NETs) formation was closely involved in lung I/R injury after LTx, which was accompanied by up-regulation of peptidylprolyl isomerase F (PPIF) and peptidyl arginine deiminase 4 (PADI4). We further established an orthotopic LTx mouse model to simulate lung I/R injury in vivo, and found that PPIF and PADI4 inhibitors effectively reduced neutrophil infiltration, NETs formation, inflammatory response, and lung I/R injury. In the neutrophil model induced by HL-60 cell line in vitro, we found that PPIF inhibitor cyclosporin A (Cys A) better alleviated calcium overload induced inflammatory response, reactive oxygen species content and NETs formation. Further study demonstrated that interfering with neutrophil PPIF protected mitochondrial function by alleviating store-operated calcium entry (SOCE) during calcium overload and played the above positive role. On this basis, we found that the reduction of calcium content in neutrophils was accompanied by the inhibition of calcineurin (CN) and nuclear factor of activated T cells (NFAT). In conclusion, our findings suggested that neutrophil PPIF could serve as a novel biomarker and potential therapeutic target of lung I/R injury after LTx, which provided new clues for its treatment by inhibiting calcium overload-induced NETs formation.
Collapse
Affiliation(s)
- Wensi Wu
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fanqing Meng
- Department of Anesthesiology, Jinan Maternity and Child Care Hospital, Jinan, China
| | - Huiying Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Xiaojun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
12
|
Hauser BR, Estafanos M, Ayyat KS, Yun JJ, Elgharably H. Current status of routine use of veno-arterial extracorporeal membrane oxygenation during lung transplantation. Expert Rev Med Devices 2024; 21:1153-1163. [PMID: 39670791 DOI: 10.1080/17434440.2024.2442485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/27/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Recently, there has been growing experience with utilizing a veno-arterial extracorporeal membrane oxygenator (VA ECMO) routinely during lung transplantation procedures. Yet, there is a lack of consensus on the protocols, benefits, and outcomes of routine VA ECMO use in lung transplantation. AREAS COVERED This article presents an overview of the current status of routine use of VA ECMO during lung transplantation, including rationale, protocols, applications, and outcomes. EXPERT OPINION Utilization of VA ECMO during lung transplantation has emerged as an alternative mechanical circulatory support modality to cardiopulmonary bypass, with growing evidence showing lower rates of peri-operative complications. Some groups took that further into routine application of VA ECMO during lung transplantation. The current available evidence suggests that routine utilization of VA ECMO during lung transplantation is associated with lower rates of primary graft dysfunction and improved early outcomes. Use of VA ECMO allows controlled reperfusion of the allograft and avoids an unplanned "crash" on pump in case of hemodynamic instability, which carries worse outcomes after lung transplantation. As a relatively new approach, further follow-up of growing experience, as well as prospective clinical trials, is necessary to develop a consensus about routine utilization of VA ECMO during lung transplantation.
Collapse
Affiliation(s)
- Benjamin R Hauser
- School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Mina Estafanos
- Department of Surgery, Division of Cardiac Surgery, University of Rochester, Rochester, NY, USA
| | - Kamal S Ayyat
- Department of Thoracic & Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - James J Yun
- Department of Thoracic & Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Haytham Elgharably
- Department of Thoracic & Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
13
|
Wang W, Tai S, Tao J, Yang L, Cheng X, Zhou J. Innovative hydrogel-based therapies for ischemia-reperfusion injury: bridging the gap between pathophysiology and treatment. Mater Today Bio 2024; 29:101295. [PMID: 39493810 PMCID: PMC11528235 DOI: 10.1016/j.mtbio.2024.101295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/21/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Ischemia-reperfusion injury (IRI) commonly occurs in clinical settings, particularly in medical practices such as organ transplantation, cardiopulmonary resuscitation, and recovery from acute trauma, posing substantial challenges in clinical therapies. Current systemic therapies for IRI are limited by poor drug targeting, short efficacy, and significant side effects. Owing to their exceptional biocompatibility, biodegradability, excellent mechanical properties, targeting capabilities, controlled release potential, and properties mimicking the extracellular matrix (ECM), hydrogels not only serve as superior platforms for therapeutic substance delivery and retention, but also facilitate bioenvironment cultivation and cell recruitment, demonstrating significant potential in IRI treatment. This review explores the pathological processes of IRI and discusses the roles and therapeutic outcomes of various hydrogel systems. By categorizing hydrogel systems into depots delivering therapeutic agents, scaffolds encapsulating mesenchymal stem cells (MSCs), and ECM-mimicking hydrogels, this article emphasizes the selection of polymers and therapeutic substances, and details special crosslinking mechanisms and physicochemical properties, as well as summarizes the application of hydrogel systems for IRI treatment. Furthermore, it evaluates the limitations of current hydrogel treatments and suggests directions for future clinical applications.
Collapse
Affiliation(s)
- Weibo Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Supeng Tai
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Lexing Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Xi Cheng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
14
|
Radicetti-Silva J, Oliveira M, Baldavira CM, Braga CL, Santos RT, Felix NS, Silva AL, Capelozzi VL, Cruz FF, Rocco PRM, Silva PL. Distinct effects of intravenous bone marrow-derived mesenchymal stem cell therapy on ischemic and non-ischemic lungs after ischemia-reperfusion injury. Cytotherapy 2024; 26:1505-1513. [PMID: 39115513 DOI: 10.1016/j.jcyt.2024.07.009] [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: 05/29/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The preclinical efficacy of mesenchymal stem cell (MSC) therapy after intravenous infusion has been promising, but clinical studies have yielded only modest results. Although most preclinical studies have focused solely on the ischemic lung, it is crucial to evaluate both lungs after ischemia-reperfusion injury, considering the various mechanisms involved. This study aimed to bridge this gap by assessing the acute effects of bone marrow MSC(BM) infusion before ischemic insult and evaluating both ischemic and non-ischemic lungs after reperfusion. METHODS Eighteen male Wistar rats (403 ± 23 g) were anesthetized and mechanically ventilated using a protective strategy. After baseline data collection, the animals were randomized to 3 groups (n = 6/group): (1) SHAM; (2) ischemia-reperfusion (IR), and (3) intravenous MSC(BM) infusion followed by IR. Ischemia was induced by complete clamping of the left hilum, followed by 1 h of reperfusion after clamp removal. At the end of the experiment, the right and left lungs (non-ischemic and ischemic, respectively) were collected for immunohistochemistry and molecular biology analysis. RESULTS MSC(BM)s reduced endothelial cell damage and apoptosis markers and improved markers associated with endothelial cell integrity in both lungs. In addition, gene expression of catalase and nuclear factor erythroid 2-related factor 2 increased after MSC(BM) therapy. In the ischemic lung, MSC(BM) therapy mitigated endothelial cell damage and apoptosis and increased gene expression associated with endothelial cell integrity. Conversely, in the non-ischemic lung, apoptosis gene expression increased in the IR group but not after MSC(BM) therapy. CONCLUSION This study demonstrates distinct effects of MSC(BM) therapy on ischemic and non-ischemic lungs after ischemia-reperfusion injury. The findings underscore the importance of evaluating both lung types in ischemia-reperfusion studies, offering insights into the therapeutic potential of MSC(BM) therapy in the context of lung injury.
Collapse
Affiliation(s)
- Julia Radicetti-Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Milena Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Cassia Lisboa Braga
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata Trabach Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nathane Santanna Felix
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriana Lopes Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Yu Y, Huang J, Fang Q, Li J, Lou Z, Liang F, Wu M. Exploring the research progression and evolutionary trends of lung ischemia-reperfusion injury: A bibliometric analysis from 1979 to 2023. Life Sci 2024; 355:123000. [PMID: 39168238 DOI: 10.1016/j.lfs.2024.123000] [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: 05/14/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Lung ischemia-reperfusion injury (LIRI) poses a significant challenge in various clinical scenarios. Despite extensive research on the pathogenesis and potential treatments of LIRI, there is a notable absence of bibliometric analysis. MATERIALS AND METHODS We summarized the results of LIRI research through two searches on the Web of Science, covering data from 1979 to 2023 with topic words "lung" and "reperfusion injury". The collected data were analyzed and visualized based on country, author(s), and keywords by bibliometric software. The keyword "programmed cell death" was further added to explore the hotspot of the LIRI research field. RESULTS The initial analysis of 1648 research articles showed a total of 40 countries and 7031 researchers were involved in the publications, with America being the most productive country in the research field of LIRI. Keyword analysis revealed that the evolving focus of LIRI research has progressively transitioned from, lung transplantation, primary graft dysfunction, inflammation, oxidative stress, and ex vivo lung perfusion to cell death. Subsequently, 212 publications specifically addressing programmed cell death (PCD) in LIRI were identified, which clarified the recent hotspot of the LIRI field. CONCLUSION With closer international cooperation and increasing research scale, the LIRI research focused mainly on the pathogenesis and potential therapeutic interventions for LIRI. PCD in LIRI is becoming a trending topic and will continue to be a hotspot in this field. Our study may offer valuable guidance for future research endeavors concerning LIRI.
Collapse
Affiliation(s)
- Yifan Yu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China.; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China
| | - Jinghao Huang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China.; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China
| | - Qiuyu Fang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China.; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China
| | - Jinsheng Li
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China.; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China
| | - Zhiling Lou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China.; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China
| | - Fuxiang Liang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China.; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China..
| | - Ming Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China.; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China..
| |
Collapse
|
16
|
Oliveira-Melo P, Nepomuceno NA, Ruiz LM, Correia AT, Vilela VS, de Oliveira Braga KA, Manzuti GM, Feitosa DDM, Kennedy-Feitosa E, Wang A, Cypel M, Fernandes PMP. Angiotensin-converting enzyme 2 activation attenuates inflammation and oxidative stress in brain death donor followed by rat lung transplantation. Sci Rep 2024; 14:23567. [PMID: 39384890 PMCID: PMC11464679 DOI: 10.1038/s41598-024-75043-7] [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: 05/02/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
Brain death (BD) provides most of the donor organs destined for lung transplantation (LTx). However, the organs may be affected by inflammatory and oxidative processes. Based on this, we hypothesize that the angiotensin-converting enzyme 2 (ACE2) activation can reduce the lung injury associated with LTx. 3 h after BD induction, rats were injected with saline (BD group) or an ACE2 activator (ACE2a group; 15 mg/kg-1) and kept on mechanical ventilation for additional 3 h. A third group included a control ventilation (Control group) prior to transplant. After BD protocol, left LTx were performed, followed by 2 h-reperfusion. ACE2 activation was associated with better oxygenation after BD management (p = 0.01), attenuating edema (p = 0.05) followed by the reduction in tissue resistance (p = 0.01) and increase of respiratory compliance (p = 0.02). Nrf2 expression was also upregulated in the ACE2a group (p = 0.03). After transplantation, ACE2a group showed lower levels of TNF-α (p = 0.02), IL-6 (p = 0.001), IL-1β (p = 0.01), ROS (p = 0.004) and MDA (p = 0.002), in addition to higher CAT activity (p = 0.04). In conclusion, our study suggests that ACE2 activation improves anti-inflammatory and antioxidant activity in a model of LTx.
Collapse
Affiliation(s)
- Paolo Oliveira-Melo
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| | - Natalia Aparecida Nepomuceno
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Liliane Moreira Ruiz
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Aristides Tadeu Correia
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Vanessa Sana Vilela
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Karina Andrighetti de Oliveira Braga
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Giovana Maria Manzuti
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Emanuel Kennedy-Feitosa
- Departamento de Ciências da Saúde, Laboratório de Morfofisiofarmacologia, Universidade Federal Rural do Semi-Árido, Mossoró, RN, Brazil
| | - Aizhou Wang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Marcelo Cypel
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Paulo Manuel Pêgo Fernandes
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
17
|
Nakagiri T, Köhler NR, Janciauskiene S, Neubert L, Knöfel AK, Pradhan P, Ruhparwar A, Ius F, Immenschuh S. Hemopexin alleviates sterile inflammation in ischemia-reperfusion-induced lung injury. Front Immunol 2024; 15:1451577. [PMID: 39430764 PMCID: PMC11487521 DOI: 10.3389/fimmu.2024.1451577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Introduction Pulmonary ischemia-reperfusion (IR) injury (IRI) plays a significant role in various lung disorders and is a key factor in the development of primary graft dysfunction following lung transplantation. Hemopexin (Hx) is the major serum scavenger protein for heme, which is a prooxidant and pro-inflammatory compound. In the current study, we hypothesized that Hx could confer beneficial effects in sterile inflammation induced by IR-mediated lung injury. Methods To examine this hypothesis, we administered Hx in an experimental mouse model of unilateral lung IRI. Results Our results demonstrate that treatment with Hx alleviated histopathological signs of inflammation in ischemic lungs, as evidenced by a reduction in the number of infiltrating neutrophils and decreased levels of perivascular edema. In addition, thrombotic vaso-occlusion in pulmonary blood vessels of IRI lungs was reduced by Hx. Immunohistochemical analysis revealed that Hx inhibited the up-regulation of heme oxygenase-1, an enzyme highly induced by heme, in ischemic lungs. Finally, Hx administration caused a decrease in the levels of circulating B- and CD8+ T-lymphocytes in the peripheral blood of mice with pulmonary IRI. Conclusion These findings suggest that the serum heme scavenger protein Hx holds therapeutic promise in alleviating lung IRI-mediated sterile inflammation. Thus, Hx may represent a preemptive therapeutic approach in IR-related lung disorders such as primary graft dysfunction in lung transplantation.
Collapse
Affiliation(s)
- Tomoyuki Nakagiri
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Nadine R. Köhler
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- Department of Genetics and Clinical Immunology, The Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| | - Lavinia Neubert
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Ann-Kathrin Knöfel
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Pooja Pradhan
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Arjang Ruhparwar
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Fabio Ius
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Stephan Immenschuh
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| |
Collapse
|
18
|
KOCAK S, GUNER I, YAMAN MO, YILMAZ TEKIZ, MEYDANLI EEGUZEL, YELMEN N, SAHIN G. Alpha B-crystallin Ameliorates Imbalance of Redox Homeostasis, Inflammation and Apoptosis in an Acute Lung Injury Model with Rats. Medeni Med J 2024; 39:211-220. [PMID: 39350576 PMCID: PMC11572206 DOI: 10.4274/mmj.galenos.2024.82274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/09/2024] [Indexed: 11/20/2024] Open
Abstract
Objective Ischemia-reperfusion (IR) of the aorta is a significant contributor to the development of postoperative acute lung damage after abdominal aortic surgery. The aim of the present study was to examine the effect of alpha B-crystallin, a small heat shock protein (known as HspB5), on lung injury induced by abdominal aortic IR in rats. Methods Male Sprague-Dawley rats were divided into three groups: control, ischemia-reperfusion (IR, 90 min ischemia and 180 min reperfusion), and alpha B-crystallin +IR. Alpha B-crystallin (50 μg/100 g) was intraperitoneally administered 1 h before IR. Lung tissue samples were obtained for histological and biochemical analyses of oxidative stress and cytokine and apoptosis parameters in plasma, lung tissues, and bronchoalveolar lavage (BAL) fluid. Results The levels of malondialdehyde, reactive oxygen species, total oxidant status, tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), nuclear factor kappa B (NFKβ), caspase-9 (CASP-9), 8-hydroxy-2'-deoxyguanosine, total antioxidant status, superoxide dismutase, and interleukin-10 levels in lung tissues, plasma, and BAL fluid (p<0.05 versus control) increased in Aortic IR. However, alpha B-crystallin significantly reduced the lung tissue levels of oxidative, inflamatuvar, and apoptotic parameters in the plasma, lung tissues, and BAL fluid (p<0.05 versus aortic IR). Histopathological results showed that alpha B-crystallin ameliorated the morphological changes related to lung injury (p<0.001). Conclusion Alpha B-crystallin substantially restored disrupted the redox balance, inflammation, and apoptotic parameters in rats exposed to IR. The cytoprotective effect of alpha B-crystallin on redox balance might be attributed to improved lung injury.
Collapse
Affiliation(s)
- Seda KOCAK
- Kirsehir Ahi Evran University Faculty of Medicine, Department of Physiology, Kirsehir, Türkiye
| | - Ibrahim GUNER
- Tekirdag Namik Kemal University Faculty of Medicine, Department of Physiology, Tekirdag, Türkiye
| | - Muhittin Onur YAMAN
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Tugba EKIZ YILMAZ
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Histology and Embriology, Istanbul, Türkiye
| | - Emine Elif GUZEL MEYDANLI
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Histology and Embriology, Istanbul, Türkiye
| | - Nermin YELMEN
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Gulderen SAHIN
- Istanbul Aydin University Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| |
Collapse
|
19
|
Endo Y, Aoki T, Jafari D, Rolston DM, Hagiwara J, Ito-Hagiwara K, Nakamura E, Kuschner CE, Becker LB, Hayashida K. Acute lung injury and post-cardiac arrest syndrome: a narrative review. J Intensive Care 2024; 12:32. [PMID: 39227997 PMCID: PMC11370287 DOI: 10.1186/s40560-024-00745-z] [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: 04/22/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Post-cardiac arrest syndrome (PCAS) presents a multifaceted challenge in clinical practice, characterized by severe neurological injury and high mortality rates despite advancements in management strategies. One of the important critical aspects of PCAS is post-arrest lung injury (PALI), which significantly contributes to poor outcomes. PALI arises from a complex interplay of pathophysiological mechanisms, including trauma from chest compressions, pulmonary ischemia-reperfusion (IR) injury, aspiration, and systemic inflammation. Despite its clinical significance, the pathophysiology of PALI remains incompletely understood, necessitating further investigation to optimize therapeutic approaches. METHODS This review comprehensively examines the existing literature to elucidate the epidemiology, pathophysiology, and therapeutic strategies for PALI. A comprehensive literature search was conducted to identify preclinical and clinical studies investigating PALI. Data from these studies were synthesized to provide a comprehensive overview of PALI and its management. RESULTS Epidemiological studies have highlighted the substantial prevalence of PALI in post-cardiac arrest patients, with up to 50% of survivors experiencing acute lung injury. Diagnostic imaging modalities, including chest X-rays, computed tomography, and lung ultrasound, play a crucial role in identifying PALI and assessing its severity. Pathophysiologically, PALI encompasses a spectrum of factors, including chest compression-related trauma, pulmonary IR injury, aspiration, and systemic inflammation, which collectively contribute to lung dysfunction and poor outcomes. Therapeutically, lung-protective ventilation strategies, such as low tidal volume ventilation and optimization of positive end-expiratory pressure, have emerged as cornerstone approaches in the management of PALI. Additionally, therapeutic hypothermia and emerging therapies targeting mitochondrial dysfunction hold promise in mitigating PALI-related morbidity and mortality. CONCLUSION PALI represents a significant clinical challenge in post-cardiac arrest care, necessitating prompt diagnosis and targeted interventions to improve outcomes. Mitochondrial-related therapies are among the novel therapeutic strategies for PALI. Further clinical research is warranted to optimize PALI management and enhance post-cardiac arrest care paradigms.
Collapse
Affiliation(s)
- Yusuke Endo
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Tomoaki Aoki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Daniel Jafari
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Daniel M Rolston
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Jun Hagiwara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Kanako Ito-Hagiwara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Eriko Nakamura
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Cyrus E Kuschner
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA.
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
20
|
Deng J, Li N, Hao L, Li S, Aiyu N, Zhang J, Hu X. Transcription factor NF-E2-related factor 2 plays a critical role in acute lung injury/acute respiratory distress syndrome (ALI/ARDS) by regulating ferroptosis. PeerJ 2024; 12:e17692. [PMID: 39670103 PMCID: PMC11637007 DOI: 10.7717/peerj.17692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/14/2024] [Indexed: 12/14/2024] Open
Abstract
NRF2 is an important transcription factor that regulates redox homeostasis in vivo and exerts its anti-oxidative stress and anti-inflammatory response by binding to the ARE to activate and regulate the transcription of downstream protective protein genes, reducing the release of reactive oxygen species. Ferroptosis is a novel iron-dependent, lipid peroxidation-driven cell death mode, and recent studies have shown that ferroptosis is closely associated with acute lung injury/acute respiratory distress syndrome (ALI/ARDS). NRF2 is able to regulate ferroptosis through the regulation of the transcription of its target genes to ameliorate ALI/ARDS. Therefore, This article focuses on how NRF2 plays a role in ALI/ARDS by regulating ferroptosis. We further reviewed the literature and deeply analyzed the signaling pathways related to ferroptosis which were regulated by NRF2. Additionally, we sorted out the chemical molecules targeting NRF2 that are effective for ALI/ARDS. This review provides a relevant theoretical basis for further research on this theory and the prevention and treatment of ALI/ARDS. The intended audience is clinicians and researchers in the field of respiratory disease.
Collapse
Affiliation(s)
- JiaLi Deng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Na Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Liyuan Hao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shenghao Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Nie Aiyu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Junli Zhang
- Department of Infectious Disease, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - XiaoYu Hu
- Department of Infectious Disease, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Buttar SN, Møller-Sørensen H, Perch M, Kissow H, Lilleør TNB, Petersen RH, Møller CH. Porcine lungs perfused with three different flows using the 8-h open-atrium cellular ex vivo lung perfusion technique. Front Bioeng Biotechnol 2024; 12:1357182. [PMID: 38983601 PMCID: PMC11231398 DOI: 10.3389/fbioe.2024.1357182] [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: 12/17/2023] [Accepted: 05/27/2024] [Indexed: 07/11/2024] Open
Abstract
The number of lung transplantations is limited due to the shortage of donor lungs fulfilling the standard criteria. The ex vivo lung perfusion (EVLP) technique provides the ability of re-evaluating and potentially improving and treating marginal donor lungs. Accordingly, the technique has emerged as an essential tool to increase the much-needed donor lung pool. One of the major EVLP protocols, the Lund protocol, characterized by high pulmonary artery flow (100% of cardiac output [CO]), an open atrium, and a cellular perfusate, has demonstrated encouraging short-EVLP duration results. However, the potential of the longer EVLP duration of the protocol is yet to be investigated, a duration which is considered necessary to rescue more marginal donor lungs in future. This study aimed to achieve stable 8-h EVLP using an open-atrium cellular model with three different pulmonary artery flows in addition to determining the most optimal flow in terms of best lung performance, including lung electrolytes and least lung edema formation, perfusate and tissue inflammation, and histopathological changes, using the porcine model. EVLP was performed using a flow of either 40% (n = 6), 80% (n = 6), or 100% (n = 6) of CO. No flow rate demonstrated stable 8-h EVLP. Stable 2-h EVLP was observed in all three groups. Insignificant deterioration was observed in dynamic compliance, peak airway pressure, and oxygenation between the groups. Pulmonary vascular resistance increased significantly in the 40% group (p < .05). Electrolytes demonstrated an insignificant worsening trend with longer EVLP. Interleukin-8 (IL-8) in perfusate and tissue, wet-to-dry weight ratio, and histopathological changes after EVLP were insignificantly time dependent between the groups. This study demonstrated that stable 8-h EVLP was not feasible in an open-atrium cellular model regardless of the flow of 40%, 80%, or 100% of CO. No flow was superior in terms of lung performance, lung electrolytes changes, least lung edema formation, minimal IL-8 expression in perfusate and tissue, and histopathological changes.
Collapse
Affiliation(s)
- Sana N. Buttar
- Department of Cardiothoracic Surgery, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hasse Møller-Sørensen
- Department of Cardiothoracic Anaesthesiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Michael Perch
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas N. B. Lilleør
- Department of Cardiothoracic Surgery, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Rene H. Petersen
- Department of Cardiothoracic Surgery, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christian H. Møller
- Department of Cardiothoracic Surgery, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Sandiumenge A, Bello I, Coll-Torres E, Gomez-Brey A, Franco-Jarava C, Miñambres E, Pérez-Redondo M, Mosteiro F, Sánchez-Moreno L, Crowley S, Fieira E, Suberviola B, Mazo CA, Agustí A, Pont T. Systemic Inflammation Differences in Brain-vs. Circulatory-Dead Donors: Impact on Lung Transplant Recipients. Transpl Int 2024; 37:12512. [PMID: 38887494 PMCID: PMC11182341 DOI: 10.3389/ti.2024.12512] [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: 12/04/2023] [Accepted: 05/17/2024] [Indexed: 06/20/2024]
Abstract
Brain death triggers a systemic inflammatory response. Whether systemic inflammation is different in lung donors after brain- (DBD) or circulatory-death (DCD) is unknown, but this may potentially increase the incidence of primary graft dysfunction (PGD) after lung transplantation. We compared the plasma levels of interleukin (IL)-6, IL-8, IL-10 and TNF-α in BDB and DCD and their respective recipients, as well as their relationship with PGD and mortality after LT. A prospective, observational, multicenter, comparative, cohort-nested study that included 40 DBD and 40 DCD lung donors matched and their respective recipients. Relevant clinical information and blood samples were collected before/during lung retrieval in donors and before/during/after (24, 48 and 72 h) LT in recipients. Incidence of PGD and short-term mortality after LT was recorded. Plasma levels of all determined cytokines were numerically higher in DBD than in DCD donors and reached statistical significance for IL-6, IL-10 and IL-8. In recipients with PGD the donor's plasma levels of TNF-α were higher. The post-operative mortality rate was very low and similar in both groups. DBD is associated with higher systemic inflammation than DCD donors, and higher TNF-α plasma levels in donors are associated with a higher incidence of PGD.
Collapse
Affiliation(s)
- Alberto Sandiumenge
- Donation and Trasplantation Program Coordination Unit, Vall d’Hebron, University Hospital, Cell, Tissue and Organ Donation and Trasplantation Resarch Group, Vall d’Hebron Research Institute (VHIR), Barcelona, Spain
| | - Irene Bello
- Department of Thoracic Surgery, Respiratory Institute Hospital Clínic, Barcelona, Spain
- Institut d’Investigacions BIomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Aroa Gomez-Brey
- Donation and Trasplantation Program Coordination Unit, Vall d’Hebron, University Hospital, Cell, Tissue and Organ Donation and Trasplantation Resarch Group, Vall d’Hebron Research Institute (VHIR), Barcelona, Spain
| | | | - Eduardo Miñambres
- Transplant Coordination Unit and Department of Intensive Care, University Hospital Marqués de Valdecilla-IDIVAL, School of Medicine, University of Cantabria, Santander, Spain
| | - Marina Pérez-Redondo
- Departament of Critical Care, Department of Donor and Transplant Coordinator, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Fernando Mosteiro
- Departament of Critical Care, Department of Donor and Transplant Coordinator, A Coruña University Hospital, A Coruña, Spain
| | - Laura Sánchez-Moreno
- Department of Thoracic Surgery, University Hospital Marqués de Valdecilla-IDIVAL, School of Medicine, University of Cantabria, Santander, Spain
| | - Silvana Crowley
- Department of Thoracic Surgery and Lung Trasplantation, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Eva Fieira
- Department of Thoracic Surgery and Lung Trasplantation, A Coruña University Hospital, A Coruña, Spain
| | - Borja Suberviola
- Transplant Coordination Unit and Department of Intensive Care, University Hospital Marqués de Valdecilla-IDIVAL, School of Medicine, University of Cantabria, Santander, Spain
| | - Cristopher Alan Mazo
- Donation and Trasplantation Program Coordination Unit, Vall d’Hebron, University Hospital, Cell, Tissue and Organ Donation and Trasplantation Resarch Group, Vall d’Hebron Research Institute (VHIR), Barcelona, Spain
| | - Alvar Agustí
- Institut d’Investigacions BIomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Pneumology, Respiratory Institute, Barcelona, Spain
- Cátedra de Salut Respiratoria, Universidad de Barcelona, Barcelona, Spain
- CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Teresa Pont
- Donation and Trasplantation Program Coordination Unit, Vall d’Hebron, University Hospital, Cell, Tissue and Organ Donation and Trasplantation Resarch Group, Vall d’Hebron Research Institute (VHIR), Barcelona, Spain
| |
Collapse
|
23
|
Chen P, Van Hassel J, Pinezich MR, Diane M, Hudock MR, Kaslow SR, Gavaudan OP, Fung K, Kain ML, Lopez H, Saqi A, Guenthart BA, Hozain AE, Romanov A, Bacchetta M, Vunjak-Novakovic G. Recovery of extracorporeal lungs using cross-circulation with injured recipient swine. J Thorac Cardiovasc Surg 2024; 167:e106-e130. [PMID: 37741314 PMCID: PMC10954590 DOI: 10.1016/j.jtcvs.2023.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023]
Abstract
OBJECTIVE Lung transplantation remains limited by the shortage of healthy organs. Cross-circulation with a healthy swine recipient provides a durable physiologic environment to recover injured donor lungs. In a clinical application, a recipient awaiting lung transplantation could be placed on cross-circulation to recover damaged donor lungs, enabling eventual transplantation. Our objective was to assess the ability of recipient swine with respiratory compromise to tolerate cross-circulation and support recovery of donor lungs subjected to extended cold ischemia. METHODS Swine donor lungs (n = 6) were stored at 4 °C for 24 hours while recipient swine (n = 6) underwent gastric aspiration injury before cross-circulation. Longitudinal multiscale analyses (blood gas, bronchoscopy, radiography, histopathology, cytokine quantification) were performed to evaluate recipient swine and extracorporeal lungs on cross-circulation. RESULTS Recipient swine lung injury resulted in sustained, impaired oxygenation (arterial oxygen tension/inspired oxygen fraction ratio 205 ± 39 mm Hg vs 454 ± 111 mm Hg at baseline). Radiographic, bronchoscopic, and histologic assessments demonstrated bilateral infiltrates, airway cytokine elevation, and significantly worsened lung injury scores. Recipient swine provided sufficient metabolic support for extracorporeal lungs to demonstrate robust functional improvement (0 hours, arterial oxygen tension/inspired oxygen fraction ratio 138 ± 28.2 mm Hg; 24 hours, 539 ± 156 mm Hg). Multiscale analyses demonstrated improved gross appearance, aeration, and cellular regeneration in extracorporeal lungs by 24 hours. CONCLUSIONS We demonstrate that acutely injured recipient swine tolerate cross-circulation and enable recovery of donor lungs subjected to extended cold storage. This proof-of-concept study supports feasibility of cross-circulation for recipients with isolated lung disease who are candidates for this clinical application.
Collapse
Affiliation(s)
- Panpan Chen
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Surgery, Columbia University Medical Center, New York, NY
| | - Julie Van Hassel
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Surgery, Columbia University Medical Center, New York, NY
| | - Meghan R Pinezich
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Mohamed Diane
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Maria R Hudock
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Sarah R Kaslow
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Surgery, Columbia University Medical Center, New York, NY
| | | | - Kenmond Fung
- Clinical Perfusion, Columbia University Medical Center, New York, NY
| | - Mandy L Kain
- Institute of Comparative Medicine, Columbia University, New York, NY
| | - Hermogenes Lopez
- Clinical Perfusion, Columbia University Medical Center, New York, NY
| | - Anjali Saqi
- Pathology and Cell Biology, Columbia University Medical Center, New York, NY
| | - Brandon A Guenthart
- Department of Cardiothoracic Surgery, Stanford University Medical Center, Stanford, Calif
| | - Ahmed E Hozain
- Department of Surgery, Columbia University Medical Center, New York, NY
| | - Alexander Romanov
- Institute of Comparative Medicine, Columbia University, New York, NY
| | - Matthew Bacchetta
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tenn; Department of Biomedical Engineering, Vanderbilt University, Nashville, Tenn
| | | |
Collapse
|
24
|
Ta HQ, Kuppusamy M, Sonkusare SK, Roeser ME, Laubach VE. The endothelium: gatekeeper to lung ischemia-reperfusion injury. Respir Res 2024; 25:172. [PMID: 38637760 PMCID: PMC11027545 DOI: 10.1186/s12931-024-02776-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
The success of lung transplantation is limited by the high rate of primary graft dysfunction due to ischemia-reperfusion injury (IRI). Lung IRI is characterized by a robust inflammatory response, lung dysfunction, endothelial barrier disruption, oxidative stress, vascular permeability, edema, and neutrophil infiltration. These events are dependent on the health of the endothelium, which is a primary target of IRI that results in pulmonary endothelial barrier dysfunction. Over the past 10 years, research has focused more on the endothelium, which is beginning to unravel the multi-factorial pathogenesis and immunologic mechanisms underlying IRI. Many important proteins, receptors, and signaling pathways that are involved in the pathogenesis of endothelial dysfunction after IR are starting to be identified and targeted as prospective therapies for lung IRI. In this review, we highlight the more significant mediators of IRI-induced endothelial dysfunction discovered over the past decade including the extracellular glycocalyx, endothelial ion channels, purinergic receptors, kinases, and integrins. While there are no definitive clinical therapies currently available to prevent lung IRI, we will discuss potential clinical strategies for targeting the endothelium for the treatment or prevention of IRI. The accruing evidence on the essential role the endothelium plays in lung IRI suggests that promising endothelial-directed treatments may be approaching the clinic soon. The application of therapies targeting the pulmonary endothelium may help to halt this rapid and potentially fatal injury.
Collapse
Affiliation(s)
- Huy Q Ta
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mark E Roeser
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA
| | - Victor E Laubach
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA.
| |
Collapse
|
25
|
Langer F, Lepper PM, Weingard B, Aliyev P, Bals R, Wilkens H. Two single lung transplantations from one donor: lung twinning in the LAS era. Respir Res 2024; 25:131. [PMID: 38500110 PMCID: PMC10949597 DOI: 10.1186/s12931-024-02754-w] [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: 01/14/2024] [Accepted: 03/02/2024] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVES The implementation of the Lung Allocation Score (LAS) in the Eurotransplant international collaborative framework decreased waiting list mortality, but organ shortage remains a significant problem. Transplantation of two single lungs from one donor into two recipients (lung twinning) may decrease waiting list mortality. We sought to analyze if this strategy can lead to an acceptable intermediate-term outcome. METHODS Since the LAS-implementation we performed 32 paired single-lung transplantations from 16 postmortal donors. Data and outcome were analyzed retrospectively comparing recipients receiving the first lung (first twins) with recipients receiving the second lung (second twins), left versus right transplantation and restrictive versus obstructive disease. RESULTS Survival at one year was 81% and 54% at five years. Veno-venous ECMO had been successfully used as bridge-to-transplant in three patients with ECMO-explantation immediately after surgery. Bronchial anastomotic complications were not observed in any patient. First twins and second twins exhibited similar survival (p = 0.82) despite higher LAS in first twins (median 45 versus 34, p < 0.001) and longer cold ischemic time in second twins (280 ± 83 vs. 478 ± 125 min, p < 0.001). Survival of left and right transplantation was similar (p = 0.45) with similar best post-transplant FEV1 (68 ± 15% versus 62 ± 14%, p = 0.26). Survival was similar in restrictive and obstructive disease (p = 0.28) with better post-transplant FEV1 (70 ± 15% versus 57 ± 11%, p = 0.02) in restrictive disease. CONCLUSIONS Performing two single-lung transplantations from one donor can be performed safely with encouraging intermediate-term outcome and good functional capacity. Lung twinning maximizes the donor pool and may help to overcome severe organ shortage. CLINICAL TRIALS This research is not a clinical trial. Thus no registration details will be provided.
Collapse
Affiliation(s)
- Frank Langer
- Dept. of Thoracic Surgery, Saarland University Medical Center, 66424, Homburg / Saar, Germany.
| | - Philipp M Lepper
- Department of Internal Medicine V, Pneumology and Intensive Care Medicine, Saarland University Medical Center, Homburg / Saar, Germany
| | - Bettina Weingard
- Department of Internal Medicine V, Pneumology and Intensive Care Medicine, Saarland University Medical Center, Homburg / Saar, Germany
| | - Parviz Aliyev
- Dept. of Thoracic Surgery, Saarland University Medical Center, 66424, Homburg / Saar, Germany
| | - Robert Bals
- Department of Internal Medicine V, Pneumology and Intensive Care Medicine, Saarland University Medical Center, Homburg / Saar, Germany
| | - Heinrike Wilkens
- Department of Internal Medicine V, Pneumology and Intensive Care Medicine, Saarland University Medical Center, Homburg / Saar, Germany
| |
Collapse
|
26
|
Ponholzer F, Dumfarth J, Krapf C, Pircher A, Hautz T, Wolf D, Augustin F, Schneeberger S. The impact and relevance of techniques and fluids on lung injury in machine perfusion of lungs. Front Immunol 2024; 15:1358153. [PMID: 38510260 PMCID: PMC10950925 DOI: 10.3389/fimmu.2024.1358153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Primary graft dysfunction (PGD) is a common complication after lung transplantation. A plethora of contributing factors are known and assessment of donor lung function prior to organ retrieval is mandatory for determination of lung quality. Specialized centers increasingly perform ex vivo lung perfusion (EVLP) to further assess lung functionality and improve and extend lung preservation with the aim to increase lung utilization. EVLP can be performed following different protocols. The impact of the individual EVLP parameters on PGD development, organ function and postoperative outcome remains to be fully investigated. The variables relate to the engineering and function of the respective perfusion devices, such as the type of pump used, functional, like ventilation modes or physiological (e.g. perfusion solutions). This review reflects on the individual technical and fluid components relevant to EVLP and their respective impact on inflammatory response and outcome. We discuss key components of EVLP protocols and options for further improvement of EVLP in regard to PGD. This review offers an overview of available options for centers establishing an EVLP program and for researchers looking for ways to adapt existing protocols.
Collapse
Affiliation(s)
- Florian Ponholzer
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Julia Dumfarth
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Krapf
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Pircher
- Department of Haematology and Oncology, Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Theresa Hautz
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Department of Haematology and Oncology, Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Augustin
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Bojic D, Aujla T, Sugihara J, Wong A, Keshavjee S, Liu M. Thyroid hormone protects human lung epithelial cells from cold preservation and warm reperfusion-induced injury. J Transl Med 2024; 22:221. [PMID: 38429788 PMCID: PMC10908176 DOI: 10.1186/s12967-024-05024-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Cellular stress associated with static-cold storage (SCS) and warm reperfusion of donor lungs can contribute to ischemia-reperfusion (IR) injury during transplantation. Adding cytoprotective agents to the preservation solution may be conducive to reducing graft deterioration and improving post-transplant outcomes. METHODS SCS and warm reperfusion were simulated in human lung epithelial cells (BEAS-2B) by exposing cells to low potassium dextran glucose solution at 4 °C for different periods and then switching back to serum-containing culture medium at 37 °C. Transcriptomic analysis was used to explore potential cytoprotective agents. Based on its results, cell viability, caspase activity, cell morphology, mitochondrial function, and inflammatory gene expression were examined under simulated IR conditions with or without thyroid hormones (THs). RESULTS After 18 h SCS followed by 2 h warm reperfusion, genes related to inflammation and cell death were upregulated, and genes related to protein synthesis and metabolism were downregulated in BEAS-2B cells, which closely mirrored gene profiles found in thyroid glands of mice with congenital hypothyroidism. The addition of THs (T3 or T4) to the preservation solution increases cell viability, inhibits activation of caspase 3, 8 and 9, preserves cell morphology, enhances mitochondrial membrane potential, reduces mitochondrial superoxide production, and suppresses inflammatory gene expression. CONCLUSION Adding THs to lung preservation solutions may protect lung cells during SCS by promoting mitochondrial function, reducing apoptosis, and inhibiting pro-inflammatory pathways. Further in vivo testing is warranted to determine the potential clinical application of adding THs as therapeutics in lung preservation solutions.
Collapse
Affiliation(s)
- Dejan Bojic
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tanroop Aujla
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Junichi Sugihara
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Aaron Wong
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
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
|
29
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
30
|
Jenkins JA, Verdiner R, Omar A, Farina JM, Wilson R, D’Cunha J, Reck Dos Santos PA. Donor and recipient risk factors for the development of primary graft dysfunction following lung transplantation. Front Immunol 2024; 15:1341675. [PMID: 38380332 PMCID: PMC10876853 DOI: 10.3389/fimmu.2024.1341675] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/22/2024] [Indexed: 02/22/2024] Open
Abstract
Primary Graft Dysfunction (PGD) is a major cause of both short-term and long-term morbidity and mortality following lung transplantation. Various donor, recipient, and technical risk factors have been previously identified as being associated with the development of PGD. Here, we present a comprehensive review of the current literature as it pertains to PGD following lung transplantation, as well as discussing current strategies to mitigate PGD and future directions. We will pay special attention to recent advances in lung transplantation such as ex-vivo lung perfusion, thoracoabdominal normothermic regional perfusion, and up-to-date literature published in the interim since the 2016 ISHLT consensus statement on PGD and the COVID-19 pandemic.
Collapse
Affiliation(s)
- J. Asher Jenkins
- Department of Cardiothoracic Surgery, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Ricardo Verdiner
- Department of Anesthesia and Perioperative Medicine, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Ashraf Omar
- Division of Pulmonology and Critical Care Medicine, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Juan Maria Farina
- Department of Cardiothoracic Surgery, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Renita Wilson
- Department of Cardiothoracic Surgery, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Jonathan D’Cunha
- Department of Cardiothoracic Surgery, Mayo Clinic Arizona, Phoenix, AZ, United States
| | | |
Collapse
|
31
|
Di Y, Fu R, Xiang Z, Sun H, Dai M, Zhang Q, Si Y. Comparison of Prognosis for Lung Transplantation between Older and Younger Donors: A Systematic Review and Meta-Analysis Based on Cohort Studies. Ann Thorac Cardiovasc Surg 2024; 30:24-00092. [PMID: 39111865 PMCID: PMC11324350 DOI: 10.5761/atcs.ra.24-00092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/19/2024] [Indexed: 08/16/2024] Open
Abstract
PURPOSE This meta-analysis aimed to compare the prognosis of lung transplantation recipients based on donor age. METHODS A detailed search was performed in PubMed, Embase, Web of Science, and the Cochrane Library for cohort studies on lung transplantation. The prognosis of lung transplant recipients was investigated based on the donor age, with the primary outcomes being 1-year overall survival (OS), 3-year OS, 5-year OS, and 5-year chronic lung allograft dysfunction (CLAD)-free survival. RESULTS This meta-analysis included 10 cohort studies. Among the short-term outcomes, the older donor group demonstrated no significant difference from the young donor group in primary graft dysfunction within 72 hours, use of extracorporeal membrane oxygenation, length of ventilator use, and intensive care unit hours. However, a longer hospital stay was associated with the older donor group. In terms of long-term outcomes, no difference was found between the two groups in 1-year OS, 3-year OS, and 5-year OS. Notably, patients with older donors exhibited a superior 5-year CLAD-free survival. CONCLUSIONS The results of this meta-analysis indicate that older donors are not inferior to younger donors in terms of long-term and short-term recipient outcomes. Lung transplantation using older donors is a potential therapeutic option after rigorous evaluation.
Collapse
Affiliation(s)
- Yongkang Di
- Intensive Care Unit, Hospital of Zhejiang People's Armed Police, Hangzhou, Zhejiang, China
| | - Rongrong Fu
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhiyi Xiang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huiwen Sun
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Min Dai
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qiufeng Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yuexiu Si
- Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
Bejeshk MA, Najafipour H, Khaksari M, Nematollahi MH, Rajizadeh MA, Dabiri S, Beik A, Samareh-Fekri M, Sepehri G. Preparation and Evaluation of Preventive Effects of Inhalational and Intraperitoneal Injection of Myrtenol Loaded Nano-Niosomes on Lung Ischemia-Reperfusion Injury in Rats. J Pharm Sci 2024; 113:85-94. [PMID: 37931787 DOI: 10.1016/j.xphs.2023.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
INTRODUCTION Ischemia-reperfusion injury (IRI) is directly related to forming reactive oxygen species, endothelial cell injury, increased vascular permeability, and the activation of neutrophils and cytokines. Niosomes are nanocarriers and an essential part of drug delivery systems. We aimed to investigate the effects of myrtenol's inhaled and intraperitoneal niosomal form, compared to its simple form, on lung ischemia reperfusion injury (LIRI). MATERIAL AND METHOD Wistar rats were divided into ten groups. Simple and niosomal forms of myrtenol were inhaled or intraperitoneally injected daily for one week prior to LIRI. We evaluated oxidative stress, apoptotic, and inflammatory indices, nitric oxide, inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS) and histopathological indices. RESULTS Pretreatment with simple and niosomal forms of myrtenol significantly inhibited the indices of pulmonary edema, pro-inflammatory cytokines and proteins, oxidant agents, nitric oxide, iNOS, apoptotic proteins, congestion of capillaries, neutrophil infiltration, and bleeding in the alveoli. Furthermore, myrtenol increased anti-inflammatory cytokines, anti-oxidants agents, eNOS, anti-apoptotic proteins and the survival time of animals. The niosomal form of myrtenol showed a more ameliorative effect than its simple form. CONCLUSION The results showed the superior protective effect of the inhalation of myrtenol niosomal form against LIRI compared to its simple form and systemic use.
Collapse
Affiliation(s)
- Mohammad Abbas Bejeshk
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Najafipour
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Mohammad Amin Rajizadeh
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Pathology and Stem Cells Research Center, Department of Pathology, School of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Ahmad Beik
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mitra Samareh-Fekri
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Sepehri
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
33
|
Zhang Z, Li X, Guo J, He B, Wu L, Yang R, Li X, Fang D, Yang X, Yang D, Wang F, Tang M, Han Y, Jose PA, Wang H, Zeng C. β-aminoisobutyrics acid, a metabolite of BCAA, activates the AMPK/Nrf-2 pathway to prevent ferroptosis and ameliorates lung ischemia-reperfusion injury. Mol Med 2023; 29:164. [PMID: 38049750 PMCID: PMC10696792 DOI: 10.1186/s10020-023-00729-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 09/17/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Lung ischemia-reperfusion (I/R) injury is a serious clinical problem without effective treatment. Enhancing branched-chain amino acids (BCAA) metabolism can protect against cardiac I/R injury, which may be related to bioactive molecules generated by BCAA metabolites. L-β-aminoisobutyric acid (L-BAIBA), a metabolite of BCAA, has multi-organ protective effects, but whether it protects against lung I/R injury is unclear. METHODS To assess the protective effect of L-BAIBA against lung I/R injury, an animal model was generated by clamping the hilum of the left lung, followed by releasing the clamp in C57BL/6 mice. Mice with lung I/R injury were pre-treated or post-treated with L-BAIBA (150 mg/kg/day), given by gavage or intraperitoneal injection. Lung injury was assessed by measuring lung edema and analyzing blood gases. Inflammation was assessed by measuring proinflammatory cytokines in bronchoalveolar lavage fluid (BALF), and neutrophil infiltration of the lung was measured by myeloperoxidase activity. Molecular biological methods, including western blot and immunofluorescence, were used to detect potential signaling mechanisms in A549 and BEAS-2B cells. RESULTS We found that L-BAIBA can protect the lung from I/R injury by inhibiting ferroptosis, which depends on the up-regulation of the expressions of GPX4 and SLC7A11 in C57BL/6 mice. Additionally, we demonstrated that the Nrf-2 signaling pathway is key to the inhibitory effect of L-BAIBA on ferroptosis in A549 and BEAS-2B cells. L-BAIBA can induce the nuclear translocation of Nrf-2. Interfering with the expression of Nrf-2 eliminated the protective effect of L-BAIBA on ferroptosis. A screening of potential signaling pathways revealed that L-BAIBA can increase the phosphorylation of AMPK, and compound C can block the Nrf-2 nuclear translocation induced by L-BAIBA. The presence of compound C also blocked the protective effects of L-BAIBA on lung I/R injury in C57BL/6 mice. CONCLUSIONS Our study showed that L-BAIBA protects against lung I/R injury via the AMPK/Nrf-2 signaling pathway, which could be a therapeutic target.
Collapse
Affiliation(s)
- Ziyue Zhang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
- Outpatient Department, Hospital of PLA, Hanzhong, Shanxi, 96608, P. R. China
| | - Xingbing Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
- Department of Cardiology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P. R. China
| | - Jingwen Guo
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Bo He
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Lianpan Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Rongpei Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Xingyue Li
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Dandong Fang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - XiaoLi Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Donghai Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Fengxian Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Ming Tang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Yu Han
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine, Department of Physiology/Pharmacology, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Hongyong Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China.
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China.
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China.
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, P. R. China.
| |
Collapse
|
34
|
Mody S, Nadkarni S, Vats S, Kumar A, Nandavaram S, Keshavamurthy S. Lung Donor Selection and Management: An Updated Review. OBM TRANSPLANTATION 2023; 07:1-54. [DOI: 10.21926/obm.transplant.2304203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The shortage of donor lungs for transplantation is a major challenge, resulting in longer waitlist times for patients with a higher risk of waitlist mortality. It is crucial to continue promoting awareness about organ donation through legislation, public campaigns, and health care provider education. Only a small number of cadaveric donors meet the ideal criteria for lung donation, leaving many lungs unused. Donor lung utilization can be improved by carefully considering the extended-criteria donors, actively participating in donor management, and by utilizing the modalities to assess and manage the marginal lungs after retrieval from the donor. The purpose of this article is to provide an up-to-date review of donor selection, assessment of donor lungs, and donor lung management to enhance organ recovery rates for lung transplantation.
Collapse
|
35
|
Santos J, Wang P, Shemesh A, Liu F, Tsao T, Aguilar OA, Cleary SJ, Singer JP, Gao Y, Hays SR, Golden JA, Leard L, Kleinhenz ME, Kolaitis NA, Shah R, Venado A, Kukreja J, Weigt SS, Belperio JA, Lanier LL, Looney MR, Greenland JR, Calabrese DR. CCR5 drives NK cell-associated airway damage in pulmonary ischemia-reperfusion injury. JCI Insight 2023; 8:e173716. [PMID: 37788115 PMCID: PMC10721259 DOI: 10.1172/jci.insight.173716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
Primary graft dysfunction (PGD) limits clinical benefit after lung transplantation, a life-prolonging therapy for patients with end-stage disease. PGD is the clinical syndrome resulting from pulmonary ischemia-reperfusion injury (IRI), driven by innate immune inflammation. We recently demonstrated a key role for NK cells in the airways of mouse models and human tissue samples of IRI. Here, we used 2 mouse models paired with human lung transplant samples to investigate the mechanisms whereby NK cells migrate to the airways to mediate lung injury. We demonstrate that chemokine receptor ligand transcripts and proteins are increased in mouse and human disease. CCR5 ligand transcripts were correlated with NK cell gene signatures independently of NK cell CCR5 ligand secretion. NK cells expressing CCR5 were increased in the lung and airways during IRI and had increased markers of tissue residency and maturation. Allosteric CCR5 drug blockade reduced the migration of NK cells to the site of injury. CCR5 blockade also blunted quantitative measures of experimental IRI. Additionally, in human lung transplant bronchoalveolar lavage samples, we found that CCR5 ligand was associated with increased patient morbidity and that the CCR5 receptor was increased in expression on human NK cells following PGD. These data support a potential mechanism for NK cell migration during lung injury and identify a plausible preventative treatment for PGD.
Collapse
Affiliation(s)
- Jesse Santos
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Surgery, UCSF - East Bay, Oakland, California, USA
| | - Ping Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Avishai Shemesh
- Department of Medicine, UCSF, San Francisco, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Fengchun Liu
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Tasha Tsao
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Simon J. Cleary
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Ying Gao
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Steven R. Hays
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Lorriana Leard
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | | | - Rupal Shah
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Aida Venado
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - S. Sam Weigt
- Department of Medicine, UCLA, Los Angeles, California, USA
| | | | - Lewis L. Lanier
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Department of Microbiology and Immunology, and
| | - Mark R. Looney
- Department of Medicine, UCSF, San Francisco, California, USA
| | - John R. Greenland
- Department of Medicine, UCSF, San Francisco, California, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| | - Daniel R. Calabrese
- Department of Medicine, UCSF, San Francisco, California, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| |
Collapse
|
36
|
Dugbartey GJ. Therapeutic benefits of nitric oxide in lung transplantation. Biomed Pharmacother 2023; 167:115549. [PMID: 37734260 DOI: 10.1016/j.biopha.2023.115549] [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: 08/06/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023] Open
Abstract
Lung transplantation is an evolutionary procedure from its experimental origin in the twentieth century and is now recognized as an established and routine life-saving intervention for a variety of end-stage pulmonary diseases refractory to medical management. Despite the success and continuous refinement in lung transplantation techniques, the widespread application of this important life-saving intervention is severely hampered by poor allograft quality offered from donors-after-brain-death. This has necessitated the use of lung allografts from donors-after-cardiac-death (DCD) as an additional source to expand the pool of donor lungs. Remarkably, the lung exhibits unique properties that may make it ideally suitable for DCD lung transplantation. However, primary graft dysfunction (PGD), allograft rejection and other post-transplant complications arising from unavoidable ischemia-reperfusion injury (IRI) of transplanted lungs, increase morbidity and mortality of lung transplant recipients annually. In the light of this, nitric oxide (NO), a selective pulmonary vasodilator, has been identified as a suitable agent that attenuates lung IRI and prevents PGD when administered directly to lung donors prior to donor lung procurement, or to recipients during and after transplantation, or administered indirectly by supplementing lung preservation solutions. This review presents a historical account of clinical lung transplantation and discusses the lung as an ideal organ for DCD. Next, the author highlights IRI and its clinical effects in lung transplantation. Finally, the author discusses preservation solutions suitable for lung transplantation, and the protective effects and mechanisms of NO in experimental and clinical lung transplantation.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana; Accra College of Medicine, Magnolia St, JVX5+FX9, East Legon, Accra, Ghana.
| |
Collapse
|
37
|
Orlitová M, Verbelen T, Frick AE, Vanstapel A, Van Beersel D, Ordies S, Van Slambrouck J, Kaes J, Jin X, Coudyzer W, Verleden SE, Verleden GM, Vanaudenaerde BM, Van Raemdonck DE, Vos R, Ceulemans LJ, Claus P, Neyrinck AP. The hemodynamic interplay between pulmonary ischemia-reperfusion injury and right ventricular function in lung transplantation: a translational porcine model. Am J Physiol Lung Cell Mol Physiol 2023; 325:L675-L688. [PMID: 37724349 PMCID: PMC11550898 DOI: 10.1152/ajplung.00281.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 09/20/2023] Open
Abstract
Lung transplantation (LTx) is a challenging procedure. Following the process of ischemia-reperfusion injury, the transplanted pulmonary graft might become severely damaged, resulting in primary graft dysfunction. In addition, during the intraoperative window, the right ventricle (RV) is at risk of acute failure. The interaction of right ventricular function with lung injury is, however, poorly understood. We aimed to address this interaction in a translational porcine model of pulmonary ischemia-reperfusion injury. Advanced pulmonary and hemodynamic assessment was used, including right ventricular pressure-volume loop analysis. The acute model was based on clamping and unclamping of the left lung hilus, respecting the different hemodynamic phases of a clinical lung transplantation. We found that forcing entire right ventricular cardiac output through a lung suffering from ischemia-reperfusion injury increased afterload (pulmonary vascular resistance from baseline to end experiment P < 0.0001) and induced right ventricular failure (RVF) in 5/9 animals. Notably, we identified different compensation patterns in failing versus nonfailing ventricles (arterial elastance P = 0.0008; stroke volume P < 0.0001). Furthermore, increased vascular pressure and flow produced by the right ventricle resulted in higher pulmonary injury, as measured by ex vivo CT density (correlation: pressure r = 0.8; flow r = 0.85). Finally, RV ischemia as measured by troponin-T was negatively correlated with pulmonary injury (r = -0.76); however, troponin-T values did not determine RVF in all animals. In conclusion, we demonstrate a delicate balance between development of pulmonary ischemia-reperfusion injury and right ventricular function during lung transplantation. Furthermore, we provide a physiological basis for potential benefit of extracorporeal life support technology.NEW & NOTEWORTHY In contrast to the abundant literature of mechanical pulmonary artery clamping to increase right ventricular afterload, we developed a model adding a biological factor of pulmonary ischemia-reperfusion injury. We did not only focus on the right ventricular behavior, but also on the interaction with the injured lung. We are the first to describe this interaction while addressing the hemodynamic intraoperative phases of clinical lung transplantation.
Collapse
Affiliation(s)
- Michaela Orlitová
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Tom Verbelen
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Cardiac Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Anna E Frick
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Arno Vanstapel
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Dieter Van Beersel
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Sofie Ordies
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Walter Coudyzer
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Stijn E Verleden
- Antwerp Surgical Training, Anatomy and Research Center, University of Antwerp, Antwerp, Belgium
| | - Geert M Verleden
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, 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
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Piet Claus
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Housman B, Laskey D, Dawodu G, Scheinin S. Single Lung Transplant for Secondary Pulmonary Hypertension: The Right Option for the Right Patient. J Clin Med 2023; 12:6789. [PMID: 37959256 PMCID: PMC10649201 DOI: 10.3390/jcm12216789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Introduction: The optimal treatment for Secondary Pulmonary Hypertension from End-Stage Lung Disease remains controversial. Double Lung Transplantation is widely regarded as the treatment of choice as it eliminates all diseased parenchyma and introduces a large volume of physiologically normal allograft. By comparison, the role of single lung transplantation for pulmonary hypertension (PAH) is less clear. The remaining diseased lung will limit clinical improvements and permit downstream sequelae; including residual cough, recurrent infection, and continued pulmonary hypertension. But not every patient can undergo DLT. Advanced age, frailty, co-morbid conditions, and limited availability of organs will all affect surgical candidacy and can offset the benefits of double lung procedures. Studies that compare SLT and DLT do not commonly explore the utility of single lung procedures even though multiple theoretical advantages exist; including reduced waiting times, less waitlist mortality, fewer surgical complications, and lower operative mortality. Worse, multiple forms of publication and selection bias may favor DLT in registry-based studies. In this review, we present the prevailing literature on single and double lung transplants in patients with secondary pulmonary hypertension and clarify the potential utility of these procedures. Materials and Methods: A PubMed search for English-language articles exploring single and double lung transplants in the setting of secondary pulmonary hypertension was conducted from 1990 to 2023. Key words included "single lung transplant", "double lung transplant", "pulmonary hypertension", "rejection", "complications", "extracorporeal membranous oxygenation", "death", and all appropriate Boolean operators. We prioritized research from retrospective studies that evaluated clinical outcomes from single centers. Conclusions: The question is not whether DLT is better at resolving lung disease; instead, we must ask if SLT is an acceptable form of therapy in a select group of high-risk patients. Further research should focus on how best to identify recipients that may benefit from each type of procedure, and the clinical utility of perioperative VA ECMO.
Collapse
Affiliation(s)
- Brian Housman
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, NY 10029, USA; (D.L.); (G.D.); (S.S.)
| | | | | | | |
Collapse
|
39
|
Zhao J, Li J, Wei D, Gao F, Yang X, Yue B, Xiong D, Liu M, Xu H, Hu C, Chen J. Liproxstatin-1 Alleviates Lung Transplantation-induced Cold Ischemia-Reperfusion Injury by Inhibiting Ferroptosis. Transplantation 2023; 107:2190-2202. [PMID: 37202851 DOI: 10.1097/tp.0000000000004638] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
BACKGROUND Primary graft dysfunction, which is directly related to cold ischemia-reperfusion (CI/R) injury, is a major obstacle in lung transplantation (LTx). Ferroptosis, a novel mode of cell death elicited by iron-dependent lipid peroxidation, has been implicated in ischemic events. This study aimed to investigate the role of ferroptosis in LTx-CI/R injury and the effectiveness of liproxstatin-1 (Lip-1), a ferroptosis inhibitor, in alleviating LTx-CI/R injury. METHODS LTx-CI/R-induced signal pathway alterations, tissue injury, cell death, inflammatory responses, and ferroptotic features were examined in human lung biopsies, the human bronchial epithelial (BEAS-2B) cells, and the mouse LTx-CI/R model (24-h CI/4-h R). The therapeutic efficacy of Lip-1 was explored and validated both in vitro and in vivo. RESULTS In human lung tissues, LTx-CI/R activated ferroptosis-related signaling pathway, increased the tissue iron content and lipid peroxidation accumulation, and altered key protein (GPX4, COX2, Nrf2, and SLC7A11) expression and mitochondrial morphology. In BEAS-2B cells, the hallmarks of ferroptosis were significantly evidenced at the setting of both CI and CI/R compared with the control, and the effect of adding Lip-1 only during CI was much better than that of only during reperfusion by Cell Counting Kit-8. Furthermore, Lip-1 administration during CI markedly relieved LTx-CI/R injury in mice, as indicated by significant improvement in lung pathological changes, pulmonary function, inflammation, and ferroptosis. CONCLUSIONS This study revealed the existence of ferroptosis in the pathophysiology of LTx-CI/R injury. Using Lip-1 to inhibit ferroptosis during CI could ameliorate LTx-CI/R injury, suggesting that Lip-1 administration might be proposed as a new strategy for organ preservation.
Collapse
Affiliation(s)
- Jin Zhao
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jiawei Li
- Department of Intensive Care Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Dong Wei
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Fei Gao
- Department of Emergency, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Xiucheng Yang
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Bingqing Yue
- Department of Lung Transplantation, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Dian Xiong
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Mingzhao Liu
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Hongyang Xu
- Department of Intensive Care Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Chunxiao Hu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jingyu Chen
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
- Department of Lung Transplantation, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
40
|
Boffini M, Marro M, Simonato E, Scalini F, Costamagna A, Fanelli V, Barbero C, Solidoro P, Brazzi L, Rinaldi M. Cytokines Removal During Ex-Vivo Lung Perfusion: Initial Clinical Experience. Transpl Int 2023; 36:10777. [PMID: 37645241 PMCID: PMC10460908 DOI: 10.3389/ti.2023.10777] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/31/2023] [Indexed: 08/31/2023]
Abstract
Ex Vivo Lung Perfusion (EVLP) can be potentially used to manipulate organs and to achieve a proper reconditioning process. During EVLP pro-inflammatory cytokines have been shown to accumulate in perfusate over time and their production is correlated with poor outcomes of the graft. Aim of the present study is to investigate the feasibility and safety of cytokine adsorption during EVLP. From July 2011 to March 2020, 54 EVLP procedures have been carried out, 21 grafts treated with an adsorption system and 33 without. Comparing the grafts perfused during EVLP with or without cytokine adsorption, the use of a filter significantly decreased the levels of IL10 and GCSFat the end of the procedure. Among the 38 transplanted patients, the adsorption group experienced a significant decreased IL6, IL10, MCP1 and GCSF concentrations and deltas compared to the no-adsorption group, with a lower in-hospital mortality (p = 0.03) and 1-year death rate (p = 0.01). This interventional study is the first human experience suggesting the safety and efficacy of a porous polymer beads adsorption device in reducing the level of inflammatory mediators during EVLP. Clinical impact of cytokines reduction during EVLP must be evaluated in further studies.
Collapse
Affiliation(s)
- Massimo Boffini
- Cardiac Surgery Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Matteo Marro
- Cardiac Surgery Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Erika Simonato
- Cardiac Surgery Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Fabrizio Scalini
- Cardiac Surgery Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Andrea Costamagna
- Anesthesiology and Intensive Care Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Vito Fanelli
- Anesthesiology and Intensive Care Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Cristina Barbero
- Cardiac Surgery Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Paolo Solidoro
- Pulmonology Division, Medical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Luca Brazzi
- Anesthesiology and Intensive Care Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Mauro Rinaldi
- Cardiac Surgery Division, Surgical Sciences Department, Città della Salute e della Scienza, University of Turin, Turin, Italy
| |
Collapse
|
41
|
Kim JL, Gouchoe DA, Reader BF, Dumond C, Lee YG, Black SM, Whitson BA. Biometric Profiling to Quantify Lung Injury Through Ex Vivo Lung Perfusion Following Warm Ischemia. ASAIO J 2023; 69:e368-e375. [PMID: 37192317 DOI: 10.1097/mat.0000000000001988] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Standard physiologic assessment parameters of donor lung grafts may not accurately reflect lung injury or quality. A biometric profile of ischemic injury could be identified as a means to assess the quality of the donor allograft. We sought to identify a biometric profile of lung ischemic injury assessed during ex vivo lung perfusion (EVLP). A rat model of lung donation after circulatory death (DCD) warm ischemic injury with subsequent EVLP evaluation was utilized. We did not observe a significant correlation between the classical physiological assessment parameters and the duration of the ischemic. In the perfusate, solubilized lactate dehydrogenase (LDH) as well as hyaluronic acid (HA) significantly correlated with duration of ischemic injury and length of perfusion ( p < 0.05). Similarly, in perfusates, the endothelin-1 (ET-1) and Big ET-1 correlated ischemic injury ( p < 0.05) and demonstrated a measure of endothelial cell injury. In tissue protein expression, heme oxygenase-1 (HO-1), angiopoietin 1 (Ang-1), and angiopoietin 2 (Ang-2) levels were correlated with the duration of ischemic injury ( p < 0.05). Cleaved caspase-3 levels were significantly elevated at 90 and 120 minutes ( p < 0.05) demonstrating increased apoptosis. A biometric profile of solubilized and tissue protein markers correlated with cell injury is a critical tool to aid in the evaluation of lung transplantation, as accurate evaluation of lung quality is imperative and improved quality leads to better results. http://links.lww.com/ASAIO/B49.
Collapse
Affiliation(s)
- Jung-Lye Kim
- From the Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Collaboration for Organ Perfusion, Protection, Engineering and Regeneration (COPPER) Laboratory, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Doug A Gouchoe
- From the Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Collaboration for Organ Perfusion, Protection, Engineering and Regeneration (COPPER) Laboratory, The Ohio State University Wexner Medical Center, Columbus, Ohio
- 88th Surgical Operations Squadron, Wright-Patterson Medical Center, Wright-Patterson AFB, Ohio
| | - Brenda F Reader
- From the Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Curtis Dumond
- From the Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Yong Gyu Lee
- From the Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Collaboration for Organ Perfusion, Protection, Engineering and Regeneration (COPPER) Laboratory, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Sylvester M Black
- From the Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Collaboration for Organ Perfusion, Protection, Engineering and Regeneration (COPPER) Laboratory, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Bryan A Whitson
- From the Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Collaboration for Organ Perfusion, Protection, Engineering and Regeneration (COPPER) Laboratory, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Davis Heart and Lung Research Institute at The Ohio State University Wexner Medical, College of Medicine, Columbus, Ohio
| |
Collapse
|
42
|
Zhang XL, Cheng Y, Xing CL, Ying JY, Yang X, Cai XD, Lu GP. Establishment of a Rat Model of Capillary Leakage Syndrome Induced by Cardiopulmonary Resuscitation After Cardiac Arrest. Curr Med Sci 2023; 43:708-715. [PMID: 37405608 DOI: 10.1007/s11596-023-2695-8] [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: 11/10/2021] [Accepted: 11/08/2022] [Indexed: 07/06/2023]
Abstract
OBJECTIVE Cardiopulmonary resuscitation (CPR) after cardiac arrest (CA) is one of the main causes of capillary leakage syndrome (CLS). This study aimed to establish a stable CLS model following the CA and cardiopulmonary resuscitation (CA-CPR) model in Sprague-Dawley (SD) rats. METHODS We conducted a prospective, randomized, animal model study. All adult male SD rats were randomly divided into a normal group (group N), a sham operation group (group S), and a cardiopulmonary resuscitation group (group T). The SD rats of the three groups were all inserted with 24-G needles through their left femoral arteries and right femoral veins. In group S and group T, the endotracheal tube was intubated. In group T, CA induced by asphyxia (AACA) was caused by vecuronium bromide with the endotracheal tube obstructed for 8 min, and the rats were resuscitated with manual chest compression and mechanical ventilation. Preresuscitation and postresuscitation measurements, including basic vital signs (BVS), blood gas analysis (BG), routine complete blood count (CBC), wet-to-dry ratio of tissues (W/D), and the HE staining results after 6 h were evaluated. RESULTS In group T, the success rate of the CA-CPR model was 60% (18/30), and CLS occurred in 26.6% (8/30) of the rats. There were no significant differences in the baseline characteristics, including BVS, BG, and CBC, among the three groups (P>0.05). Compared with pre-asphyxia, there were significant differences in BVS, CBC, and BG, including temperature, oxygen saturation (SpO2), mean arterial pressure (MAP), central venous pressure (CVP), white blood cell count (WBC), hemoglobin, hematocrit, pH, pCO2, pO2, SO2, lactate (Lac), base excess (BE), and Na+ (P<0.05) after the return of spontaneous circulation (ROSC) in group T. At 6 h after ROSC in group T and at 6 h after surgery in groups N and S, there were significant differences in temperature, heart rate (HR), respiratory rate (RR), SpO2, MAP, CVP, WBC, pH, pCO2, Na+, and K+ among the three groups (P<0.05). Compared with the other two groups, the rats in group T showed a significantly increased W/D weight ratio (P<0.05). The HE-stained sections showed consistent severe lesions in the lung, small intestine, and brain tissues of the rats at 6 h after ROSC following AACA. CONCLUSION The CA-CPR model in SD rats induced by asphyxia could reproduce CLS with good stability and reproducibility.
Collapse
Affiliation(s)
- Xiao-Lei Zhang
- Department of Pediatric Emergency Medicine and Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Ye Cheng
- Department of Pediatric Emergency Medicine and Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Chun-Lin Xing
- Department of Pediatric Critical Care Medicine, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jia-Yun Ying
- Department of Pediatric Emergency Medicine and Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Xue Yang
- Department of Pediatric Emergency Medicine and Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Xiao-di Cai
- Department of Pediatric Emergency Medicine and Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Guo-Ping Lu
- Department of Pediatric Emergency Medicine and Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
43
|
He F, Lu Y, Mao Q, Zhou L, Chen Y, Xie Y. Effects of penehyclidine hydrochloride combined with dexmedetomidine on pulmonary function in patients undergoing heart valve surgery: a double-blind, randomized trial. BMC Anesthesiol 2023; 23:237. [PMID: 37442959 PMCID: PMC10339561 DOI: 10.1186/s12871-023-02176-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
AIM To investigate the effects of penehyclidine hydrochloride combined with dexmedetomidine on pulmonary function in patients undergoing heart valve surgery with cardiopulmonary bypass (CPB). METHODS A total of 180 patients undergoing elective heart valve surgery with CPB were randomly divided into four groups: 45 in group P (intravenous penehyclidine hydrochloride 0.02 mg/kg 10 min before anesthesia induction and at the beginning of CPB, total 0.04 mg/kg); 43 in group D (dexmedetomidine 0.5 μg/kg/h after induction of anesthesia until the end of anesthesia); 44 in group PD ( penehyclidine hydrochloride 0.04 mg/kg combined with dexmedetomidine 0.5 μg/kg/h intravenously during anesthesia); and 43 in group C (same amount of normal saline 10 min before and after anesthesia induction, to the end of anesthesia, and at the beginning of CPB). The main outcomes were the incidence and severity of postoperative pulmonary complications (PPCs). The secondary outcomes were: (1) extubation time, length of stay in intensive care, and postoperative hospital stay, and adverse events; and (2) pulmonary function evaluation indices (oxygenation index and respiratory index) and plasma inflammatory factor concentrations (tumor necrosis factor-α, interleukin-6, C-reactive protein and procalcitonin) during the perioperative period. RESULTS The incidence of PPCs in groups P, D and PD after CPB was lower than that in group C (P < 0.05), and the incidence in group PD was significantly lower than that in groups P and D (P < 0.05). The scores for PPCs in groups P, D and PD were lower than those in group C (P < 0.05). CONCLUSION Combined use of penehyclidine hydrochloride and dexmedetomidine during anesthesia reduced the occurrence of postoperative pulmonary dysfunction, and improved the prognosis of patients undergoing heart valve surgery with CPB. TRIAL REGISTRATION The trial was registered in the Chinese Clinical Trial Registry on 3/11/2020 (Registration No.: ChiCTR2000039610).
Collapse
Affiliation(s)
- Fang He
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yizhi Lu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qi Mao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lifang Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanhua Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
44
|
Zheng JL, Wang X, Song Z, Zhou P, Zhang GJ, Diao JJ, Han CE, Jia GY, Zhou X, Zhang BQ. Network pharmacology and molecular docking to explore Polygoni Cuspidati Rhizoma et Radix treatment for acute lung injury. World J Clin Cases 2023; 11:4579-4600. [PMID: 37469744 PMCID: PMC10353494 DOI: 10.12998/wjcc.v11.i19.4579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Polygoni Cuspidati Rhizoma et Radix (PCRR), a well-known traditional Chinese medicine (TCM), inhibits inflammation associated with various human diseases. However, the anti-inflammatory effects of PCRR in acute lung injury (ALI) and the underlying mechanisms of action remain unclear.
AIM To determine the ingredients related to PCRR for treatment of ALI using multiple databases to obtain potential targets for fishing.
METHODS Recognized and candidate active compounds for PCRR were obtained from Traditional Chinese Medicine Systems Pharmacology, STITCH, and PubMed databases. Target ALI databases were built using the Therapeutic Target, DrugBank, DisGeNET, Online Mendelian Inheritance in Man, and Genetic Association databases. Network pharmacology includes network construction, target prediction, topological feature analysis, and enrichment analysis. Bioinformatics resources from the Database for Annotation, Visualization and Integrated Discovery were utilized for gene ontology biological process and Kyoto Encyclopedia of Genes and Genomes network pathway enrichment analysis, and molecular docking techniques were adopted to verify the combination of major active ingredients and core targets.
RESULTS Thirteen bioactive compounds corresponding to the 433 PCRR targets were identified. In addition, 128 genes were closely associated with ALI, 60 of which overlapped with PCRR targets and were considered therapeutically relevant. Functional enrichment analysis suggested that PCRR exerted its pharmacological effects in ALI by modulating multiple pathways, including the cell cycle, cell apoptosis, drug metabolism, inflammation, and immune modulation. Molecular docking results revealed a strong associative relationship between the active ingredient and core target.
CONCLUSION PCRR alleviates ALI symptoms via molecular mechanisms predicted by network pharmacology. This study proposes a strategy to elucidate the mechanisms of TCM at the network pharmacology level.
Collapse
Affiliation(s)
- Jia-Lin Zheng
- Department of Respiratory, The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong Province, China
| | - Xiao Wang
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Zhe Song
- Department of Respiratory, The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong Province, China
| | - Peng Zhou
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Gui-Ju Zhang
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Juan-Juan Diao
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Cheng-En Han
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Guang-Yuan Jia
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Xu Zhou
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Bao-Qing Zhang
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| |
Collapse
|
45
|
Razi O, Teixeira AM, Tartibian B, Zamani N, Knechtle B. Respiratory issues in patients with multiple sclerosis as a risk factor during SARS-CoV-2 infection: a potential role for exercise. Mol Cell Biochem 2023; 478:1533-1559. [PMID: 36411399 PMCID: PMC9684932 DOI: 10.1007/s11010-022-04610-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022]
Abstract
Coronavirus disease-2019 (COVID-19) is associated with cytokine storm and is characterized by acute respiratory distress syndrome (ARDS) and pneumonia problems. The respiratory system is a place of inappropriate activation of the immune system in people with multiple sclerosis (MS), and this may cause damage to the lung and worsen both MS and infections.The concerns for patients with multiple sclerosis are because of an enhance risk of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The MS patients pose challenges in this pandemic situation, because of the regulatory defect of autoreactivity of the immune system and neurological and respiratory tract symptoms. In this review, we first indicate respiratory issues associated with both diseases. Then, the main mechanisms inducing lung damages and also impairing the respiratory muscles in individuals with both diseases is discussed. At the end, the leading role of physical exercise on mitigating respiratory issues inducing mechanisms is meticulously evaluated.
Collapse
Affiliation(s)
- Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Razi University, Kermanshah, Iran
| | - Ana Maria Teixeira
- Research Center for Sport and Physical Activity, Faculty of Sport Sciences and Physical Education, University of Coimbra, Coimbra, Portugal
| | - Bakhtyar Tartibian
- Department of Exercise Physiology, Faculty of Physical Education and Sports Sciences, Allameh Tabataba’i University, Tehran, Iran
| | - Nastaran Zamani
- Department of Biology, Faculty of Science, Payame-Noor University, Tehran, Iran
| | - Beat Knechtle
- Institute of Primary Care, University of Zurich, Zurich, Switzerland
- Medbase St. Gallen Am Vadianplatz, Vadianstrasse 26, 9001 St. Gallen, Switzerland
| |
Collapse
|
46
|
Wong W, Johnson B, Cheng PC, Josephson MB, Maeda K, Berg RA, Kawut SM, Harhay MO, Goldfarb SB, Yehya N, Himebauch AS. Primary graft dysfunction grade 3 following pediatric lung transplantation is associated with chronic lung allograft dysfunction. J Heart Lung Transplant 2023; 42:669-678. [PMID: 36639317 PMCID: PMC10811698 DOI: 10.1016/j.healun.2022.12.014] [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: 05/26/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Severe primary graft dysfunction (PGD) is associated with the development of bronchiolitis obliterans syndrome (BOS), the most common form of chronic lung allograft dysfunction (CLAD), in adults. However, PGD associations with long-term outcomes following pediatric lung transplantation are unknown. We hypothesized that PGD grade 3 (PGD 3) at 48- or 72-hours would be associated with shorter CLAD-free survival following pediatric lung transplantation. METHODS This was a single center retrospective cohort study of patients ≤ 21 years of age who underwent bilateral lung transplantation between 2005 and 2019 with ≥ 1 year of follow-up. PGD and CLAD were defined by published criteria. We evaluated the association of PGD 3 at 48- or 72-hours with CLAD-free survival by using time-to-event analyses. RESULTS Fifty-one patients were included (median age 12.7 years; 51% female). The most common transplant indications were cystic fibrosis (29%) and pulmonary hypertension (20%). Seventeen patients (33%) had PGD 3 at either 48- or 72-hours. In unadjusted analysis, PGD 3 was associated with an increased risk of CLAD or mortality (HR 2.10, 95% CI 1.01-4.37, p=0.047). This association remained when adjusting individually for multiple potential confounders. There was evidence of effect modification by sex (interaction p = 0.055) with the association of PGD 3 and shorter CLAD-free survival driven predominantly by males (HR 4.73, 95% CI 1.44-15.6) rather than females (HR 1.23, 95% CI 0.47-3.20). CONCLUSIONS PGD 3 at 48- or 72-hours following pediatric lung transplantation was associated with shorter CLAD-free survival. Sex may be a modifier of this association.
Collapse
Affiliation(s)
- Wai Wong
- Department of Pediatrics, Division of Pulmonary Medicine and Respiratory Diseases, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Brandy Johnson
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Pi Chun Cheng
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Division of Pediatric Pulmonology, Allergy, and Sleep Medicine, Indiana University School of Medicine, Riley Hospital for Children, Indianapolis, Indiana
| | - Maureen B Josephson
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Katsuhide Maeda
- Department of Surgery, Division of Cardiothoracic Surgery, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Robert A Berg
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael O Harhay
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Samuel B Goldfarb
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Minnesota, Masonic Children's Hospital, Minneapolis, Minnesota
| | - Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Adam S Himebauch
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
47
|
Gokbulut P, Kuskonmaz SM, Koc G, Onder CE, Yumusak N, Erel O, Nural AS, Culha C. Evaluation of the effects of empagliflozin on acute lung injury in rat intestinal ischemia-reperfusion model. J Endocrinol Invest 2023; 46:1017-1026. [PMID: 36495440 DOI: 10.1007/s40618-022-01978-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Empagliflozin is a selective sodium-glucose co-transporter (SGLT2) inhibitor that is approved for the treatment of type 2 diabetes. The beneficial effects of empagliflozin on other organ systems including the heart and kidneys have been proven. The aim of this study is to evaluate the role of empagliflozin on acute lung injury induced by intestinal ischemia-reperfusion (I/R). MATERIALS AND METHODS A total of 27 male Wistar albino rats were divided into three groups: sham, I/R, and I/R + empagliflozin; each group containing nine animals. Sham group rats underwent laparotomy without I/R injury. Rats in the I/R group underwent laparotomy, 1 h of after ischemia-reperfusion injury (superior mesenteric artery ligation was followed by 2 h of reperfusion). Rats in I/R were given empagliflozin (30 mg/kg) by gastric gavage for 7 days before the ischemia-reperfusion injury. All animals were killed at the end of reperfusion and lung tissue samples were obtained for immunohistochemical staining and histopathological investigation in all groups. RESULTS Serum glucose, AST, ALT, creatinine, native thiol, total thiol, and disulfide levels and disulfide-native thiol, disulfide-total thiol, and native thiol-total thiol ratios as well as the IMA levels were analyzed and compared among the groups. While intestinal I/R significantly increases serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and creatinine levels; did not cause any change in homeostasis parameters and IMA level. Empagliflozin treatment had no significant effect on biochemical parameters. Empagliflozin treatment induced a significant decrease in positive immunostaining for IL-1, IL-6, TNF-alpha, caspase 3, caspase 8, and caspase 9 compared to the I/R group in lung tissue samples. Intestinal I/R caused severe histopathological injury including edema, hemorrhage, increased thickness of the alveolar wall, and infiltration of inflammatory cells into alveolar spaces. Empagliflozin treatment significantly attenuated the severity of intestinal I/R injury. CONCLUSIONS It was concluded that empagliflozin treatment may have beneficial effects in acute lung injury, and, therefore, has the potential for clinical use.
Collapse
Affiliation(s)
- P Gokbulut
- Department of Endocrinology, Ankara Training and Research Hospital, Hacettepe Neighborhood Ulucanlar Caddesi No: 89 Altindag, 06230, Ankara, Turkey.
| | - S M Kuskonmaz
- Department of Endocrinology, Ankara Training and Research Hospital, Hacettepe Neighborhood Ulucanlar Caddesi No: 89 Altindag, 06230, Ankara, Turkey
| | - G Koc
- Department of Endocrinology, Ankara Training and Research Hospital, Hacettepe Neighborhood Ulucanlar Caddesi No: 89 Altindag, 06230, Ankara, Turkey
| | - C E Onder
- Ömer Halisdemir University Training and Research Hospital, Nigde, Turkey
| | - N Yumusak
- Faculty of Veterinary Medicine, Harran University, Sanlıurfa, Turkey
| | - O Erel
- Department of Biochemistry, Ankara City Hospital, Ankara, Turkey
| | - A S Nural
- Department of Biochemistry, Ankara City Hospital, Ankara, Turkey
| | - C Culha
- Department of Endocrinology, Ankara Training and Research Hospital, Hacettepe Neighborhood Ulucanlar Caddesi No: 89 Altindag, 06230, Ankara, Turkey
| |
Collapse
|
48
|
Wagner MJ, Hatami S, Freed DH. Thoracic organ machine perfusion: A review of concepts with a focus on reconditioning therapies. FRONTIERS IN TRANSPLANTATION 2023; 2:1060992. [PMID: 38993918 PMCID: PMC11235380 DOI: 10.3389/frtra.2023.1060992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/06/2023] [Indexed: 07/13/2024]
Abstract
Thoracic organ transplantation, including lung, heart, and heart-lung transplants are highly regarded as gold standard treatments for patients suffering from heart failure or chronic end stage lung conditions. The relatively high prevalence of conditions necessitating thoracic organ transplants combined with the lack of available organs has resulted in many either dying or becoming too ill to receive a transplant while on the waiting list. There is a dire need to increase both the number of organs available and the utilization of such organs. Improved preservation techniques beyond static storage have shown great potential to lengthen the current period of viability of thoracic organs while outside the body, promising better utilization rates, increased donation distance, and improved matching of donors to recipients. Ex-situ organ perfusion (ESOP) can also make some novel therapeutic strategies viable, and the combination of the ESOP platform with such reconditioning therapies endeavors to better improve functional preservation of organs in addition to making more organs viable for transplantation. Given the abundance of clinical and pre-clinical studies surrounding reconditioning of thoracic organs in combination with ESOP, we summarize in this review important concepts and research regarding thoracic organ machine perfusion in combination with reconditioning therapies.
Collapse
Affiliation(s)
| | - Sanaz Hatami
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Darren H Freed
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
- Alberta Transplant Institute, Edmonton, AB, Canada
| |
Collapse
|
49
|
Avtaar Singh SS, Das De S, Al-Adhami A, Singh R, Hopkins PMA, Curry PA. Primary graft dysfunction following lung transplantation: From pathogenesis to future frontiers. World J Transplant 2023; 13:58-85. [PMID: 36968136 PMCID: PMC10037231 DOI: 10.5500/wjt.v13.i3.58] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/11/2022] [Accepted: 02/17/2023] [Indexed: 03/16/2023] Open
Abstract
Lung transplantation is the treatment of choice for patients with end-stage lung disease. Currently, just under 5000 lung transplants are performed worldwide annually. However, a major scourge leading to 90-d and 1-year mortality remains primary graft dysfunction. It is a spectrum of lung injury ranging from mild to severe depending on the level of hypoxaemia and lung injury post-transplant. This review aims to provide an in-depth analysis of the epidemiology, patho physiology, risk factors, outcomes, and future frontiers involved in mitigating primary graft dysfunction. The current diagnostic criteria are examined alongside changes from the previous definition. We also highlight the issues surrounding chronic lung allograft dysfunction and identify the novel therapies available for ex-vivo lung perfusion. Although primary graft dysfunction remains a significant contributor to 90-d and 1-year mortality, ongoing research and development abreast with current technological advancements have shed some light on the issue in pursuit of future diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Sanjeet Singh Avtaar Singh
- Department of Cardiothoracic Surgery, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, United Kingdom
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Sudeep Das De
- Heart and Lung Transplant Unit, Wythenshawe Hospital, Manchester M23 9NJ, United Kingdom
| | - Ahmed Al-Adhami
- Department of Cardiothoracic Surgery, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, United Kingdom
- Department of Heart and Lung Transplant, Royal Papworth Hospital, Cambridge CB2 0AY, United Kingdom
| | - Ramesh Singh
- Mechanical Circulatory Support, Inova Health System, Falls Church, VA 22042, United States
| | - Peter MA Hopkins
- Queensland Lung Transplant Service, Prince Charles Hospital, Brisbane, QLD 4032, Australia
| | - Philip Alan Curry
- Department of Cardiothoracic Surgery, Golden Jubilee National Hospital, Glasgow G81 4DY, United Kingdom
| |
Collapse
|
50
|
Vajter J, Vachtenheim J, Prikrylova Z, Berousek J, Vymazal T, Lischke R, Martin AK, Durila M. Effect of targeted coagulopathy management and 5% albumin as volume replacement therapy during lung transplantation on allograft function: a secondary analysis of a randomized clinical trial. BMC Pulm Med 2023; 23:80. [PMID: 36894877 PMCID: PMC9996868 DOI: 10.1186/s12890-023-02372-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Primary graft dysfunction (PGD) after lung transplantation (LuTx) contributes substantially to early postoperative morbidity. Both intraoperative transfusion of a large amount of blood products during the surgery and ischemia-reperfusion injury after allograft implantation play an important role in subsequent PGD development. METHODS We have previously reported a randomized clinical trial of 67 patients where point of care (POC) targeted coagulopathy management and intraoperative administration of 5% albumin led to significant reduction of blood loss and blood product consumption during the lung transplantation surgery. A secondary analysis of the randomized clinical trial evaluating the effect of targeted coagulopathy management and intraoperative administration of 5% albumin on early lung allograft function after LuTx and 1-year survival was performed. RESULTS Compared to the patients in the control (non-POC) group, those in study (POC) group showed significantly superior graft function, represented by the Horowitz index (at 72 h after transplantation 402.87 vs 308.03 with p < 0.001, difference between means: 94.84, 95% CI: 60.18-129.51). Furthermore, the maximum doses of norepinephrine administered during first 24 h were significantly lower in the POC group (0.193 vs 0.379 with p < 0.001, difference between the means: 0.186, 95% CI: 0.105-0.267). After dichotomization of PGD (0-1 vs 2-3), significant difference between the non-POC and POC group occurred only at time point 72, when PGD grade 2-3 developed in 25% (n = 9) and 3.2% (n = 1), respectively (p = 0.003). The difference in 1-year survival was not statistically significant (10 patients died in non-POC group vs. 4 patients died in POC group; p = 0.17). CONCLUSIONS Utilization of a POC targeted coagulopathy management combined with Albumin 5% as primary resuscitative fluid may improve early lung allograft function, provide better circulatory stability during the early post-operative period, and have potential to decrease the incidence of PGD without negative effect on 1-year survival. TRIAL REGISTRATION This clinical trial was registered at ClinicalTrials.gov (NCT03598907).
Collapse
Affiliation(s)
- Jaromir Vajter
- Department of Anesthesiology and Intensive Care Medicine, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jiri Vachtenheim
- Prague Lung Transplant Program, 3rd Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic.
| | - Zuzana Prikrylova
- Department of Anesthesiology and Intensive Care Medicine, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan Berousek
- Department of Anesthesiology and Intensive Care Medicine, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Tomas Vymazal
- Department of Anesthesiology and Intensive Care Medicine, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Robert Lischke
- Prague Lung Transplant Program, 3rd Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Archer Kilbourne Martin
- Division of Cardiovascular and Thoracic Anesthesiology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Miroslav Durila
- Department of Anesthesiology and Intensive Care Medicine, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|