1
|
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
|
2
|
Edström D, Niroomand A, Stenlo M, Broberg E, Hirdman G, Ghaidan H, Hyllén S, Pierre L, Olm F, Lindstedt S. Amniotic fluid-derived mesenchymal stem cells reduce inflammation and improve lung function following transplantation in a porcine model. J Heart Lung Transplant 2024:S1053-2498(24)01800-X. [PMID: 39182800 DOI: 10.1016/j.healun.2024.08.014] [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: 10/14/2023] [Revised: 08/03/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Lung transplantation is hindered by low donor lung utilization rates. Infectious complications are reasons to decline donor grafts due to fear of post-transplant primary graft dysfunction. Mesenchymal stem cells are a promising therapy currently investigated in treating lung injury. Full-term amniotic fluid-derived lung-specific mesenchymal stem cell treatment may regenerate damaged lungs. These cells have previously demonstrated inflammatory mediation in other respiratory diseases, and we hypothesized that treatment would improve donor lung quality and postoperative outcomes. METHODS In a transplantation model, donor pigs were stratified to either the treated or the nontreated group. Acute respiratory distress syndrome was induced in donor pigs and harvested lungs were placed on ex vivo lung perfusion (EVLP) before transplantation. Treatment consisted of 3 doses of 2 × 106 cells/kg: one during EVLP and 2 after transplantation. Donors and recipients were assessed on clinically relevant parameters and recipients were followed for 3 days before evaluation for primary graft dysfunction (PGD). RESULTS Repeated injection of the cell treatment showed reductions in inflammation seen through lowered immune cell counts, reduced histology signs of inflammation, and decreased cytokines in the plasma and bronchoalveolar lavage fluid. Treated recipients showed improved pulmonary function, including increased PaO2/FiO2 ratios and reduced incidence of PGD. CONCLUSIONS Repeated injection of lung-specific cell treatment during EVLP and post transplant was associated with improved function of previously damaged lungs. Cell treatment may be considered as a potential therapy to increase the number of lungs available for transplantation and the improvement of postoperative outcomes.
Collapse
Affiliation(s)
- Dag Edström
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Anna Niroomand
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, New York
| | - Martin Stenlo
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Ellen Broberg
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Gabriel Hirdman
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Haider Ghaidan
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Snejana Hyllén
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Leif Pierre
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Franziska Olm
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
3
|
Zhang N, Zhang Q, Zhang Z, Yu J, Fu Y, Gao J, Jiang X, Jiang P, Wen Z. IRF1 and IL1A associated with PANoptosis serve as potential immune signatures for lung ischemia reperfusion injury following lung transplantation. Int Immunopharmacol 2024; 139:112739. [PMID: 39074415 DOI: 10.1016/j.intimp.2024.112739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Lung ischemia reperfusion injury (IRI) is the principal cause of primary graft dysfunction (PGD) after lung transplantation, affecting short-term and long-term mortality post-transplantation. PANoptosis, a newly identified form of regulated cell death involving apoptosis, necroptosis, and pyroptosis, is now considered a possible cause of organ damage and IRI. However, the specific role of PANoptosis to the development of lung IRI following lung transplantation is still not fully understood. METHODS In this study, we identified differentially expressed genes (DEGs) by analyzing the gene expression data from the GEO database related to lung IRI following lung transplantation. PANoptosis-IRI DEGs were determined based on the intersection of PANoptosis-related genes and screened DEGs. Hub genes associated with lung IRI were further screened using Lasso regression and the SVM-RFE algorithm. Additionally, the Cibersort algorithm was employed to assess immune cell infiltration and investigate the interaction between immune cells and hub genes. The upstream miRNAs that may regulate hub genes and compounds that may interact with hub genes were also analyzed. Moreover, an external dataset was utilized to validate the differential expression analysis of hub genes. Finally, the expressions of hub genes were ultimately confirmed using quantitative real-time PCR, western blotting, and immunohistochemistry in both animal models of lung IRI and lung transplant patients. RESULTS PANoptosis-related genes, specifically interferon regulatory factor 1 (IRF1) and interleukin 1 alpha (IL1A), have been identified as potential biomarkers for lung IRI following lung transplantation. In mouse models of lung IRI, both the mRNA and protein expression levels of IRF1 and IL1A were significantly elevated in lung tissues of the IRI group compared to the control group. Moreover, lung transplant recipients exhibited significantly higher protein levels of IRF1 and IL1A in PBMCs when compared to healthy controls. Patients who experienced PGD showed elevated levels of IRF1 and IL1A proteins in their blood samples. Furthermore, in patients undergoing lung transplantation, the protein levels of IRF1 and IL1A were notably increased in peripheral blood mononuclear cells (PBMCs) compared to healthy controls. In addition, patients who developed primary graft dysfunction (PGD) exhibited even higher protein levels of IRF1 and IL1A than those without PGD. Furthermore, PANoptosis was observed in the lung tissues of mouse models of lung IRI and in the PBMCs of patients who underwent lung transplantation. CONCLUSIONS Our research identified IRF1 and IL1A as biomarkers associated with PANoptosis in lung IRI, suggesting their potential utility as targets for diagnosing and therapeutically intervening in lung IRI and PGD following lung transplantation.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ping Jiang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
4
|
Chao BT, Sage AT, Yeung JC, Bai X, Ma J, Martinu T, Liu M, Cypel M, Van Raemdonck D, Ceulemans LJ, Neyrinck A, Verleden S, Keshavjee S. Identification of regional variation in gene expression and inflammatory proteins in donor lung tissue and ex vivo lung perfusate. J Thorac Cardiovasc Surg 2023; 166:1520-1528.e3. [PMID: 37482240 DOI: 10.1016/j.jtcvs.2023.07.013] [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: 02/03/2023] [Revised: 06/08/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
OBJECTIVE Diagnosing lung injury is a challenge in lung transplantation. It has been unclear if a single biopsy specimen is truly representative of the entire organ. Our objective was to investigate lung inflammatory biomarkers using human lung tissue biopsies and ex vivo lung perfusion perfusate. METHODS Eight human donor lungs declined for transplantation were air inflated, flash frozen, and partitioned from apex to base. Biopsies were then sampled throughout the lung. Perfusate was sampled from 4 lung lobes in 8 additional donor lungs subjected to ex vivo lung perfusion. The levels of interleukin-6, interleukin-8, interleukin-10, and interleukin-1β were measured using quantitative reverse transcription polymerase chain reaction from lung biopsies and enzyme-linked immunosorbent assay from ex vivo lung perfusion perfusate. RESULTS The median intra-biopsy equal-variance P value was .50 for messenger RNA biomarkers in tissue biopsies. The median intra-biopsy coefficient of variance was 18%. In donors with no apparent focal injuries, the biopsies in each donor showed no difference in various lung slices, with a coefficient of variance of 20%. The exception was biopsies from the lingula and injured focal areas that demonstrated larger differences. Cytokines in ex vivo lung perfusion perfusate showed minimal variation among different lobes (coefficient of variance = 4.9%). CONCLUSIONS Cytokine gene expression in lung biopsies was consistent, and the biopsy analysis reflects the whole lung, except when specimens were collected from the lingula or an area of focal injury. Ex vivo lung perfusion perfusate also provides a representative measurement of lung inflammation from the draining lobe. These results will reassure clinicians that a lung biopsy or an ex vivo lung perfusion perfusate sample can be used to inform donor lung selection.
Collapse
Affiliation(s)
- Bonnie T Chao
- Toronto Lung Transplant Program and Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Andrew T Sage
- Toronto Lung Transplant Program and Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ajmera Transplant Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan C Yeung
- Toronto Lung Transplant Program and Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ajmera Transplant Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xiaohui Bai
- Toronto Lung Transplant Program and Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jin Ma
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Tereza Martinu
- Toronto Lung Transplant Program and Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ajmera Transplant Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Toronto Lung Transplant Program and Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ajmera Transplant Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Toronto Lung Transplant Program and Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ajmera Transplant Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dirk Van Raemdonck
- BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Arne Neyrinck
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Stijn Verleden
- BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of ASTARC, University of Antwerp, Antwerp, Belgium
| | - Shaf Keshavjee
- Toronto Lung Transplant Program and Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Ajmera Transplant Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
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: 3] [Impact Index Per Article: 3.0] [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
|
6
|
Subramaniam K, Loor G, Chan EG, Bottiger BA, Ius F, Hartwig MG, Daoud D, Zhang Q, Wei Q, Villavicencio-Theoduloz MA, Osho AA, Chandrashekaran S, Noguchi Machuca T, Van Raemdonck D, Neyrinck A, Toyoda Y, Kashem MA, Huddleston S, Ryssel NR, Sanchez PG. Intraoperative Red Blood Cell Transfusion and Primary Graft Dysfunction After Lung Transplantation. Transplantation 2023; 107:1573-1579. [PMID: 36959119 DOI: 10.1097/tp.0000000000004545] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
BACKGROUND In this international, multicenter study of patients undergoing lung transplantation (LT), we explored the association between the amount of intraoperative packed red blood cell (PRBC) transfusion and occurrence of primary graft dysfunction (PGD) and associated outcomes. METHODS The Extracorporeal Life Support in LT Registry includes data on LT recipients from 9 high-volume (>40 transplants/y) transplant centers (2 from Europe, 7 from the United States). Adult patients who underwent bilateral orthotopic lung transplant from January 2016 to January 2020 were included. The primary outcome of interest was the occurrence of grade 3 PGD in the first 72 h after LT. RESULTS We included 729 patients who underwent bilateral orthotopic lung transplant between January 2016 and November 2020. LT recipient population tertiles based on the amount of intraoperative PRBC transfusion (0, 1-4, and >4 units) were significantly different in terms of diagnosis, age, gender, body mass index, mean pulmonary artery pressure, lung allocation score, hemoglobin, prior chest surgery, preoperative hospitalization, and extracorporeal membrane oxygenation requirement. Inverse probability treatment weighting logistic regression showed that intraoperative PRBC transfusion of >4 units was significantly ( P < 0.001) associated with grade 3 PGD within 72 h (odds ratio [95% confidence interval], 2.2 [1.6-3.1]). Inverse probability treatment weighting analysis excluding patients with extracorporeal membrane oxygenation support produced similar findings (odds ratio [95% confidence interval], 2.4 [1.7-3.4], P < 0.001). CONCLUSIONS In this multicenter, international registry study of LT patients, intraoperative transfusion of >4 units of PRBCs was associated with an increased risk of grade 3 PGD within 72 h. Efforts to improve post-LT outcomes should include perioperative blood conservation measures.
Collapse
Affiliation(s)
- Kathirvel Subramaniam
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Gabriel Loor
- Division of Cardiothoracic Transplantation and Mechanical Circulatory Support, Baylor College of Medicine, Houston, TX
| | - Ernest G Chan
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Brandi A Bottiger
- Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Fabio Ius
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Matthew G Hartwig
- Division of Cardiovascular and Thoracic Surgery, Duke University Medical Center, Durham, NC
| | - Daoud Daoud
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Qianzi Zhang
- Surgical Research Core, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Qi Wei
- Department of Statistics, Phastar Inc, Durham, NC
| | | | - Asishana A Osho
- Department of Cardiac Surgery, Massachusetts General Hospital, Boston, MA
| | - Satish Chandrashekaran
- Department of Pulmonary and Critical Care, McKelvey Lung Transplant Center, Emory University Hospital, Atlanta, GA
| | | | - Dirk Van Raemdonck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Arne Neyrinck
- Division of Anesthesiology and Algology, University Hospitals Leuven, Leuven, Belgium
| | - Yoshiya Toyoda
- Division of Cardiovascular Surgery, Temple University, Philadelphia, PA
| | - Mohammed A Kashem
- Division of Cardiovascular Surgery, Temple University, Philadelphia, PA
| | - Stephen Huddleston
- Division of Cardiothoracic Surgery, University of Minnesota Medical School, Minneapolis, MI
| | - Naomi R Ryssel
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Pablo G Sanchez
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
7
|
Diagnostic and Therapeutic Implications of Ex Vivo Lung Perfusion in Lung Transplantation: Potential Benefits and Inherent Limitations. Transplantation 2023; 107:105-116. [PMID: 36508647 DOI: 10.1097/tp.0000000000004414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ex vivo lung perfusion (EVLP), a technique in which isolated lungs are continually ventilated and perfused at normothermic temperature, is emerging as a promising platform to optimize donor lung quality and increase the lung graft pool. Over the past few decades, the EVLP technique has become recognized as a significant achievement and gained much attention in the field of lung transplantation. EVLP has been demonstrated to be an effective platform for various targeted therapies to optimize donor lung function before transplantation. Additionally, some physical parameters during EVLP and biological markers in the EVLP perfusate can be used to evaluate graft function before transplantation and predict posttransplant outcomes. However, despite its advantages, the clinical practice of EVLP continuously encounters multiple challenges associated with both intrinsic and extrinsic limitations. It is of utmost importance to address the advantages and disadvantages of EVLP for its broader clinical usage. Here, the pros and cons of EVLP are comprehensively discussed, with a focus on its benefits and potential approaches for overcoming the remaining limitations. Directions for future research to fully explore the clinical potential of EVLP in lung transplantation are also discussed.
Collapse
|
8
|
Assadiasl S, Nicknam MH. Cytokines in Lung Transplantation. Lung 2022; 200:793-806. [PMID: 36348053 DOI: 10.1007/s00408-022-00588-1] [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: 09/13/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022]
Abstract
Lung transplantation has developed significantly in recent years, but post-transplant care and patients' survival still need to be improved. Moreover, organ shortage urges novel modalities to improve the quality of unsuitable lungs. Cytokines, the chemical mediators of the immune system, might be used for diagnostic and therapeutic purposes in lung transplantation. Cytokine monitoring pre- and post-transplant could be applied to the prevention and early diagnosis of injurious inflammatory events including primary graft dysfunction, acute cellular rejection, bronchiolitis obliterans syndrome, restrictive allograft syndrome, and infections. In addition, preoperative cytokine removal, specific inhibition of proinflammatory cytokines, and enhancement of anti-inflammatory cytokines gene expression could be considered therapeutic options to improve lung allograft survival. Therefore, it is essential to describe the cytokines alteration during inflammatory events to gain a better insight into their role in developing the abovementioned complications. Herein, cytokine fluctuations in lung tissue, bronchoalveolar fluid, peripheral blood, and exhaled breath condensate in different phases of lung transplantation have been reviewed; besides, cytokine gene polymorphisms with clinical significance have been summarized.
Collapse
Affiliation(s)
- Sara Assadiasl
- Molecular Immunology Research Center, Tehran University of Medical Sciences, No. 142, Nosrat St., Tehran, 1419733151, Iran.
| | - Mohammad Hossein Nicknam
- Molecular Immunology Research Center, Tehran University of Medical Sciences, No. 142, Nosrat St., Tehran, 1419733151, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Bonneau S, Landry C, Bégin S, Adam D, Villeneuve L, Clavet-Lanthier MÉ, Dasilva A, Charles E, Dumont BL, Neagoe PE, Brochiero E, Menaouar A, Nasir B, Stevens LM, Ferraro P, Noiseux N, Sirois MG. Correlation between Neutrophil Extracellular Traps (NETs) Expression and Primary Graft Dysfunction Following Human Lung Transplantation. Cells 2022; 11:3420. [PMID: 36359815 PMCID: PMC9656095 DOI: 10.3390/cells11213420] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 09/08/2023] Open
Abstract
Primary graft dysfunction (PGD) is characterized by alveolar epithelial and vascular endothelial damage and inflammation, lung edema and hypoxemia. Up to one-third of recipients develop the most severe form of PGD (Grade 3; PGD3). Animal studies suggest that neutrophils contribute to the inflammatory process through neutrophil extracellular traps (NETs) release (NETosis). NETs are composed of DNA filaments decorated with granular proteins contributing to vascular occlusion associated with PGD. The main objective was to correlate NETosis in PGD3 (n = 9) versus non-PGD3 (n = 27) recipients in an exploratory study. Clinical data and blood samples were collected from donors and recipients pre-, intra- and postoperatively (up to 72 h). Inflammatory inducers of NETs' release (IL-8, IL-6 and C-reactive protein [CRP]) and components (myeloperoxidase [MPO], MPO-DNA complexes and cell-free DNA [cfDNA]) were quantified by ELISA. When available, histology, immunohistochemistry and immunofluorescence techniques were performed on lung biopsies from donor grafts collected during the surgery to evaluate the presence of activated neutrophils and NETs. Lung biopsies from donor grafts collected during transplantation presented various degrees of vascular occlusion including neutrophils undergoing NETosis. Additionally, in recipients intra- and postoperatively, circulating inflammatory (IL-6, IL-8) and NETosis biomarkers (MPO-DNA, MPO, cfDNA) were up to 4-fold higher in PGD3 recipients compared to non-PGD3 (p = 0.041 to 0.001). In summary, perioperative elevation of NETosis biomarkers is associated with PGD3 following human lung transplantation and these biomarkers might serve to identify recipients at risk of PGD3 and initiate preventive therapies.
Collapse
Affiliation(s)
- Steven Bonneau
- Research Center—Montreal Heart Institute, 5000 Belanger St., Montreal, QC H1T 1C8, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
| | - Caroline Landry
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Stéphanie Bégin
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
| | - Damien Adam
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Louis Villeneuve
- Research Center—Montreal Heart Institute, 5000 Belanger St., Montreal, QC H1T 1C8, Canada
| | | | - Ariane Dasilva
- Research Center—Montreal Heart Institute, 5000 Belanger St., Montreal, QC H1T 1C8, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
| | - Elcha Charles
- Research Center—Montreal Heart Institute, 5000 Belanger St., Montreal, QC H1T 1C8, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Benjamin L. Dumont
- Research Center—Montreal Heart Institute, 5000 Belanger St., Montreal, QC H1T 1C8, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Paul-Eduard Neagoe
- Research Center—Montreal Heart Institute, 5000 Belanger St., Montreal, QC H1T 1C8, Canada
| | - Emmanuelle Brochiero
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Ahmed Menaouar
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
| | - Basil Nasir
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Louis-Mathieu Stevens
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Pasquale Ferraro
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Nicolas Noiseux
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis St, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Martin G. Sirois
- Research Center—Montreal Heart Institute, 5000 Belanger St., Montreal, QC H1T 1C8, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, 2900 Blvd Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
10
|
Haam S. Ex Vivo Lung Perfusion in Lung Transplantation. J Chest Surg 2022; 55:288-292. [PMID: 35924535 PMCID: PMC9358162 DOI: 10.5090/jcs.22.056] [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: 07/11/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Ex vivo lung perfusion (EVLP) is a technique that enables active metabolism of the lung by creating an environment similar to that inside the body, even though the explanted lungs are outside the body. The EVLP system enables the use of lung grafts that do not satisfy the acceptance criteria for lung transplantation (LTx) by making it possible to evaluate the function of the lung grafts and repair lungs in poor condition, thereby reducing the waiting time of patients requiring LTx and consequently mortality.
Collapse
Affiliation(s)
- Seokjin Haam
- Department of Thoracic and Cardiovascular Surgery, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
11
|
Gori F, Fumagalli J, Lonati C, Carlin A, Leonardi P, Biancolilli O, Rossetti A, Righi I, Tosi D, Palleschi A, Rosso L, Morlacchi LC, Blasi F, Vivona L, Florio G, Scaravilli V, Valenza F, Zanella A, Grasselli G. Lung Biomolecular Profile and Function of Grafts from Donors after Cardiocirculatory Death with Prolonged Donor Warm Ischemia Time. J Clin Med 2022; 11:jcm11113066. [PMID: 35683455 PMCID: PMC9181171 DOI: 10.3390/jcm11113066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/30/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
The acceptable duration of donor warm ischemia time (DWIT) after cardiocirculatory death (DCD) is still debated. We analyzed the biomolecular profile and function during ex vivo lung perfusion (EVLP) of DCD lungs and their correlation with lung transplantation (LuTx) outcomes. Donor data, procurement times, recipient outcomes, and graft function up to 1 year after LuTx were collected. During EVLP, the parameters of graft function and metabolism, perfusate samples to quantify inflammation, glycocalyx breakdown products, coagulation, and endothelial activation markers were obtained. Data were compared to a cohort of extended-criteria donors after brain death (EC-DBD). Eight DBD and seven DCD grafts transplanted after EVLP were analyzed. DCD’s DWIT was 201 [188;247] minutes. Donors differed only regarding the duration of mechanical ventilation that was longer in the EC-DBD group. No difference was observed in lung graft function during EVLP. At reperfusion, “wash-out” of inflammatory cells and microthrombi was predominant in DCD grafts. Perfusate biomolecular profile demonstrated marked endothelial activation, characterized by the presence of inflammatory mediators and glycocalyx breakdown products both in DCD and EC-DBD grafts. Early graft function after LuTx was similar between DCD and EC-DBD. DCD lungs exposed to prolonged DWIT represent a potential resource for donation if properly preserved and evaluated.
Collapse
Affiliation(s)
- Francesca Gori
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.G.); (J.F.); (O.B.); (V.S.); (G.G.)
| | - Jacopo Fumagalli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.G.); (J.F.); (O.B.); (V.S.); (G.G.)
| | - Caterina Lonati
- Center of Preclinical Research, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Andrea Carlin
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
| | - Patrizia Leonardi
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
| | - Osvaldo Biancolilli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.G.); (J.F.); (O.B.); (V.S.); (G.G.)
| | - Antonello Rossetti
- Hospital Medical Direction, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Ilaria Righi
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (I.R.); (D.T.)
| | - Davide Tosi
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (I.R.); (D.T.)
| | - Alessandro Palleschi
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (I.R.); (D.T.)
| | - Lorenzo Rosso
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (I.R.); (D.T.)
| | - Letizia Corinna Morlacchi
- Respiratory Unit & Cystic Fibrosis Adult Center, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
- Respiratory Unit & Cystic Fibrosis Adult Center, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Luigi Vivona
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
| | - Gaetano Florio
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
| | - Vittorio Scaravilli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.G.); (J.F.); (O.B.); (V.S.); (G.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
| | - Franco Valenza
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
- Department of Anesthesia and Critical Care, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Alberto Zanella
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.G.); (J.F.); (O.B.); (V.S.); (G.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
- Correspondence: ; Tel.: +39-02-55033674; Fax: +39-02-55033230
| | - Giacomo Grasselli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.G.); (J.F.); (O.B.); (V.S.); (G.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.C.); (P.L.); (A.P.); (L.R.); (F.B.); (L.V.); (G.F.); (F.V.)
| |
Collapse
|
12
|
Peyneau M, de Chaisemartin L, Faille D, Messika J, Mal H, Castier Y, Mordant P, Carrasco JL, Tanaka S, Lortat Jacob B, Ferrari P, Arrault X, Ajzenberg N, Chollet-Martin S, Montravers P, Tran-Dinh A. First Experience With Extracorporeal Cytokine Adsorption Therapy After Lung Transplantation. Transpl Int 2022; 35:10319. [PMID: 35387399 PMCID: PMC8977406 DOI: 10.3389/ti.2022.10319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/14/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Marine Peyneau
- Autoimmunity and Hypersensitivity Laboratory, AP-HP, Hôpital Bichat-Claude-Bernard, Paris, France
- Faculty of Pharmacy, INSERM UMR 996, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| | - Luc de Chaisemartin
- Autoimmunity and Hypersensitivity Laboratory, AP-HP, Hôpital Bichat-Claude-Bernard, Paris, France
- Faculty of Pharmacy, INSERM UMR 996, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| | - Dorothée Faille
- Hematology Laboratory, AP-HP, Hôpital Bichat Claude Bernard, Paris, France
- Laboratory for Vascular Translational Science, INSERM UMR 1148, Université de Paris, Paris, France
| | - Jonathan Messika
- INSERM UMR 1152 PHERE, Université de Paris, Paris, France
- Pneumologie B et Transplantation Pulmonaire, AP-HP, Hôpital Bichat-Claude-Bernard, Paris, France
- Paris Transplant Group, Paris, France
| | - Hervé Mal
- INSERM UMR 1152 PHERE, Université de Paris, Paris, France
- Pneumologie B et Transplantation Pulmonaire, AP-HP, Hôpital Bichat-Claude-Bernard, Paris, France
| | - Yves Castier
- INSERM UMR 1152 PHERE, Université de Paris, Paris, France
- Service de Chirurgie Vasculaire, Thoracique et Transplantation Pulmonaire, AP-HP, Hôpital Bichat Claude Bernard, Paris, France
| | - Pierre Mordant
- INSERM UMR 1152 PHERE, Université de Paris, Paris, France
- Service de Chirurgie Vasculaire, Thoracique et Transplantation Pulmonaire, AP-HP, Hôpital Bichat Claude Bernard, Paris, France
| | - José Luis Carrasco
- Département d’Anesthésie-Réanimation, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Sébastien Tanaka
- Département d’Anesthésie-Réanimation, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
- INSERM UMR 1188 DéTROI, Université de la Réunion, Saint-Denis de la Réunion, France
| | - Brice Lortat Jacob
- Département d’Anesthésie-Réanimation, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Paola Ferrari
- Département d’Anesthésie-Réanimation, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Xavier Arrault
- Service de Pharmacie, AP-HP, Hôpital Bichat Claude Bernard, Paris, France
| | - Nadine Ajzenberg
- Hematology Laboratory, AP-HP, Hôpital Bichat Claude Bernard, Paris, France
- Laboratory for Vascular Translational Science, INSERM UMR 1148, Université de Paris, Paris, France
| | - Sylvie Chollet-Martin
- Autoimmunity and Hypersensitivity Laboratory, AP-HP, Hôpital Bichat-Claude-Bernard, Paris, France
- Faculty of Pharmacy, INSERM UMR 996, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| | - Philippe Montravers
- INSERM UMR 1152 PHERE, Université de Paris, Paris, France
- Département d’Anesthésie-Réanimation, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Alexy Tran-Dinh
- Laboratory for Vascular Translational Science, INSERM UMR 1148, Université de Paris, Paris, France
- Département d’Anesthésie-Réanimation, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
- *Correspondence: Alexy Tran-Dinh,
| |
Collapse
|
13
|
Ma X, Yan W, He N. Lidocaine attenuates hypoxia/reoxygenation‑induced inflammation, apoptosis and ferroptosis in lung epithelial cells by regulating the p38 MAPK pathway. Mol Med Rep 2022; 25:150. [PMID: 35244190 PMCID: PMC8941375 DOI: 10.3892/mmr.2022.12666] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/22/2021] [Indexed: 11/12/2022] Open
Abstract
Lung ischemia-reperfusion (I/R) injury poses a serious threat to human health, worldwide. The current study aimed to determine the role of lidocaine in A549 cells, in addition to the involvement of the p38 MAPK pathway. Oxygen deprivation/reoxygenation-induced A549 cells were utilized to simulate I/R injury in vitro. Cell viability and apoptosis were detected using MTT and TUNEL assays, respectively. The levels of IL-6, IL-8, TNF-α, malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase, iron and reactive oxygen species (ROS) were measured using corresponding commercial kits. The corresponding protein expression levels were also measured using western blotting. Moreover, a monolayer cell paracellular permeability assay was performed to determine the permeability of A549 cells. The results demonstrated that, whilst lidocaine had no influence on untreated A549 cells, it significantly increased the viability of hypoxia/reoxygenation (H/R)-induced A549 cells. A549 cell apoptosis and the release of inflammatory cytokines in the H/R group were decreased after the addition of lidocaine. When compared with the H/R group, increased MDA level and decreased SOD level were observed in H/R-induced A549 cells following lidocaine treatment. In addition, the permeability of H/R-induced A549 cells was markedly decreased following lidocaine treatment. Compared with the H/R group, the expression levels of tight junction and ferroptosis-related proteins were significantly upregulated by lidocaine, whereas the expression of transferrin was downregulated. However, p79350, an agonist of p38, reversed the effects of lidocaine on H/R-induced A549 cells. In conclusion, lidocaine exerted a protective role in HR-induced lung epithelial cell injury, which may serve as a potential agent for the treatment of patients with lung I/R injury.
Collapse
Affiliation(s)
- Xiaojun Ma
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Weihua Yan
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Na He
- Department of Anesthesiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010000, P.R. China
| |
Collapse
|
14
|
Hernández-Jiménez C, Olmos-Zúñiga JR, Baltazares-Lipp M, Jasso-Victoria R, Polo-Jerez A, Pérez-López MT, Vázquez-Justiniano LF, Díaz-Martínez NE, Gaxiola-Gaxiola M, Romero-Romero L, Guzmán-Cedillo AE, Baltazares-Lipp ME, Vázquez-Minero JC, Gutiérrez-González LH, Alonso-Gómez M, Silva-Martínez M. Endothelin-Converting Enzyme 1 and Vascular Endothelial Growth Factor as Potential Biomarkers during Ex Vivo Lung Perfusion with Prolonged Hypothermic Lung-Sparing. DISEASE MARKERS 2022; 2022:6412238. [PMID: 35178130 PMCID: PMC8844163 DOI: 10.1155/2022/6412238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 12/18/2022]
Abstract
Lung transplantation requires optimization of donor's organ use through ex vivo lung perfusion (EVLP) to avoid primary graft dysfunction. Biomarkers can aid in organ selection by providing early evidence of suboptimal lungs during EVLP and thus avoid high-risk transplantations. However, predictive biomarkers of pulmonary graft function such as endothelin-converting enzyme (ECE-1) and vascular endothelial growth factor (VEGF) have not been described under EVLP with standard prolonged hypothermic preservation, which are relevant in situations where lung procurement is difficult or far from the transplantation site. Therefore, this study is aimed at quantifying ECE-1 and VEGF, as well as determining their association with hemodynamic, gasometric, and mechanical ventilatory parameters in a swine model of EVLP with standard prolonged hypothermic preservation. Using a protocol with either immediate (I-) or delayed (D-) initiation of EVLP, ECE-1 levels over time were found to remain constant in both study groups (p > 0.05 RM-ANOVA), while the VEGF protein was higher after prolonged preservation, but it decreased throughout EVLP (p > 0.05 RM-ANOVA). Likewise, hemodynamic, gasometric, mechanical ventilatory, and histological parameters had a tendency to better results after 12 hours of hypothermic preservation in the delayed infusion group.
Collapse
Affiliation(s)
- Claudia Hernández-Jiménez
- Department of Surgical Research, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - J. Raúl Olmos-Zúñiga
- Experimental Lung Transplant Unit, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Matilde Baltazares-Lipp
- Department of Surgical Research, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Rogelio Jasso-Victoria
- Department of Surgical Research, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Adrián Polo-Jerez
- Department of Surgical Research, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - María Teresa Pérez-López
- Nursing Research Coordination, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | | | - Néstor Emmanuel Díaz-Martínez
- Laboratory of Cellular Reprogramming and Tissue Engineering, Department of Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C, Mexico City, Mexico
| | - Miguel Gaxiola-Gaxiola
- Laboratory of Morphology, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Laura Romero-Romero
- Department of Pathology, School of Veterinary Medicine and Zootechnics, UNAM, Mexico City, Mexico
| | - Axel Edmundo Guzmán-Cedillo
- Department of Surgical Research, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Mario Enrique Baltazares-Lipp
- Hemodynamics and Echocardiography Service, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Juan Carlos Vázquez-Minero
- Cardiothoracic Surgery Service, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | | | - Marcelino Alonso-Gómez
- Department of Surgical Research, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Mariana Silva-Martínez
- Experimental Lung Transplant Unit, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| |
Collapse
|
15
|
Weissenbacher A, Stone JP, Lo Faro ML, Hunter JP, Ploeg RJ, Coussios CC, Fildes JE, Friend PJ. Hemodynamics and Metabolic Parameters in Normothermic Kidney Preservation Are Linked With Donor Factors, Perfusate Cells, and Cytokines. Front Med (Lausanne) 2022; 8:801098. [PMID: 35083252 PMCID: PMC8784871 DOI: 10.3389/fmed.2021.801098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/08/2021] [Indexed: 12/29/2022] Open
Abstract
Kidney transplantation is the best renal-replacement option for most patients with end-stage renal disease. Normothermic machine preservation (NMP) of the kidney has been studied extensively during the last two decades and implemented in clinical trials. Biomarker research led to success in identifying molecules with diagnostic, predictive and therapeutic properties in chronic kidney disease. However, perfusate biomarkers and potential predictive mechanisms in NMP have not been identified yet. Twelve discarded human kidneys (n = 7 DBD, n = 5 DCD) underwent NMP for up to 24 h. Eight were perfused applying urine recirculation (URC), four with replacement of urine (UR) using Ringer's lactate. The aim of our study was to investigate biomarkers (NGAL, KIM-1, and L-FABP), cells and cytokines in the perfusate in context with donor characteristics, perfusate hemodynamics and metabolic parameters. Cold ischemia time did not correlate with any of the markers. Perfusates of DBD kidneys had a significantly lower number of leukocytes after 6 h of NMP compared to DCD. Arterial flow, pH, NGAL and L-FABP correlated with donor creatinine and eGFR. Arterial flow was higher in kidneys with lower perfusate lactate. Perfusate TNF-α was higher in kidneys with lower arterial flow. The cytokines IL-1β and GM-CSF decreased during 6 h of NMP. Kidneys with more urine output had lower perfusate KIM-1 levels. Median and 6-h values of lactate, arterial flow, pH, NGAL, KIM-1, and L-FABP correlated with each other indicating a 6-h period being applicable for kidney viability assessment. The study results demonstrate a comparable cytokine and cell profile in perfusates with URC and UR. In conclusion, clinically available perfusate and hemodynamic parameters correlate well with donor characteristics and measured biomarkers in a discarded human NMP model.
Collapse
Affiliation(s)
- Annemarie Weissenbacher
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom.,Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - John P Stone
- The ex-vivo Lab, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.,The ex-vivo Research Centre Community Interest Company (CIC), Macclesfield, United Kingdom
| | - Maria Letizia Lo Faro
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - James P Hunter
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Rutger J Ploeg
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | | | - James E Fildes
- The ex-vivo Lab, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.,The ex-vivo Research Centre Community Interest Company (CIC), Macclesfield, United Kingdom
| | - Peter J Friend
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
16
|
Current Status and Future Perspectives on Machine Perfusion: A Treatment Platform to Restore and Regenerate Injured Lungs Using Cell and Cytokine Adsorption Therapy. Cells 2021; 11:cells11010091. [PMID: 35011653 PMCID: PMC8750486 DOI: 10.3390/cells11010091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 02/06/2023] Open
Abstract
Since its advent in the 1990′s, ex vivo lung perfusion (EVLP) has been studied and implemented as a tool to evaluate the quality of a donor organ prior to transplantation. It provides an invaluable window of opportunity for therapeutic intervention to render marginal lungs viable for transplantation. This ultimately aligns with the need of the lung transplant field to increase the number of available donor organs given critical shortages. As transplantation is the only option for patients with end-stage lung disease, advancements in technology are needed to decrease wait-list time and mortality. This review summarizes the results from the application of EVLP as a therapeutic intervention and focuses on the use of the platform with regard to cell therapies, cell product therapies, and cytokine filtration among other technologies. This review will summarize both the clinical and translational science being conducted in these aspects and will highlight the opportunities for EVLP to be developed as a powerful tool to increase the donor lung supply.
Collapse
|
17
|
Clausen E, Cantu E. Primary graft dysfunction: what we know. J Thorac Dis 2021; 13:6618-6627. [PMID: 34992840 PMCID: PMC8662499 DOI: 10.21037/jtd-2021-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022]
Abstract
Many advances in lung transplant have occurred over the last few decades in the understanding of primary graft dysfunction (PGD) though effective prevention and treatment remain elusive. This review will cover prior understanding of PGD, recent findings, and directions for future research. A consensus statement updating the definition of PGD in 2016 highlights the growing complexity of lung transplant perioperative care taking into account the increasing use of high flow oxygen delivery and pulmonary vasodilators in the current era. PGD, particularly more severe grades, is associated with worse short- and long-term outcomes after transplant such as chronic lung allograft dysfunction. Growing experience have helped identify recipient, donor, and intraoperative risk factors for PGD. Understanding the pathophysiology of PGD has advanced with increasing knowledge of the role of innate immune response, humoral cell immunity, and epithelial cell injury. Supportive care post-transplant with technological advances in extracorporeal membranous oxygenation (ECMO) remain the mainstay of treatment for severe PGD. Future directions include the evolving utility of ex vivo lung perfusion (EVLP) both in PGD research and potential pre-transplant treatment applications. PGD remains an important outcome in lung transplant and the future holds a lot of potential for improvement in understanding its pathophysiology as well as development of preventative therapies and treatment.
Collapse
Affiliation(s)
- Emily Clausen
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Edward Cantu
- Division of Cardiovascular Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
18
|
Patel PM, Connolly MR, Coe TM, Calhoun A, Pollok F, Markmann JF, Burdorf L, Azimzadeh A, Madsen JC, Pierson RN. Minimizing Ischemia Reperfusion Injury in Xenotransplantation. Front Immunol 2021; 12:681504. [PMID: 34566955 PMCID: PMC8458821 DOI: 10.3389/fimmu.2021.681504] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022] Open
Abstract
The recent dramatic advances in preventing "initial xenograft dysfunction" in pig-to-non-human primate heart transplantation achieved by minimizing ischemia suggests that ischemia reperfusion injury (IRI) plays an important role in cardiac xenotransplantation. Here we review the molecular, cellular, and immune mechanisms that characterize IRI and associated "primary graft dysfunction" in allotransplantation and consider how they correspond with "xeno-associated" injury mechanisms. Based on this analysis, we describe potential genetic modifications as well as novel technical strategies that may minimize IRI for heart and other organ xenografts and which could facilitate safe and effective clinical xenotransplantation.
Collapse
Affiliation(s)
- Parth M. Patel
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Margaret R. Connolly
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Taylor M. Coe
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Anthony Calhoun
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Franziska Pollok
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - James F. Markmann
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Transplantation, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lars Burdorf
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Agnes Azimzadeh
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Joren C. Madsen
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Richard N. Pierson
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
19
|
Frick AE, Verleden SE, Ordies S, Sacreas A, Vos R, Verleden GM, Vanaudenaerde BM, Claes S, Schols D, Van Raemdonck DE, Neyrinck AP. Early protein expression profile in bronchoalveolar lavage fluid and clinical outcomes in primary graft dysfunction after lung transplantation. Eur J Cardiothorac Surg 2021; 58:379-388. [PMID: 32267918 DOI: 10.1093/ejcts/ezaa043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 01/08/2020] [Accepted: 01/16/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Primary graft dysfunction (PGD) remains a major post-transplant complication and is associated with increased morbidity and mortality. Mechanisms evoking PGD are not completely clear, but inflammation plays a central role. We investigated the association between PGD and inflammatory proteins present in immediate postoperative bronchoalveolar lavage. METHODS All double-lung recipients transplanted at our institution from 2002 to 2018 were included in our study. We retrospectively selected 80 consecutive lung transplant recipients with different PGD grades (n = 20 for each PGD grades 0-1 to 2-3). In bronchoalveolar lavage performed within the first 24 h after donor aortic cross-clamping following lung transplantation, concentrations of 30 cytokines, chemokines and growth factors were assessed by enzyme-linked immunosorbent assay (ELISA) and correlated with donor and recipient demographics and outcomes. For analysis, 2 groups were defined: 'mild' PGD (grade 0-1) and 'severe' PGD (grades 2-3). RESULTS Significant differences between mild and severe PGD were found in 8 biomarkers [interleukin (IL)-6, IL-10, IL-13, eotaxin, granulocyte colony-stimulating factor, interferon γ, macrophage inflammatory protein 1α, surfactant protein D (SP-D); P < 0.05]. Increased IL-10 and IL-13, but none of the other proteins, were associated with short-term outcome (longer time to extubation; P = 0.005 and P < 0.0001; increased intensive care unit stay; P = 0.012 and P < 0.0001; and hospital stay; P = 0.041 and P = 0.002). There were no significant differences in donor and recipient characteristics between the groups. CONCLUSIONS Expression profiles of key inflammatory mediators in bronchoalveolar lavage fluid differed significantly between lung transplant recipients with severe versus mild PGD and correlated with clinical outcome variables. Further research should focus on the early mechanisms leading to PGD.
Collapse
Affiliation(s)
- Anna E Frick
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.,Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Belgium
| | - Stijn E Verleden
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Belgium
| | - Sofie Ordies
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.,Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Belgium
| | - Annelore Sacreas
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Belgium
| | - Robin Vos
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Belgium.,Department of Pneumology, University Hospitals Leuven, Leuven, Belgium
| | - Geert M Verleden
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Belgium.,Department of Pneumology, University Hospitals Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Belgium
| | - Sandra Claes
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Belgium.,Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.,Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Lung Transplantation, Pulmonary Endothelial Inflammation, and Ex-Situ Lung Perfusion: A Review. Cells 2021; 10:cells10061417. [PMID: 34200413 PMCID: PMC8229792 DOI: 10.3390/cells10061417] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/31/2022] Open
Abstract
Lung transplantation (LTx) is the gold standard treatment for end-stage lung disease; however, waitlist mortality remains high due to a shortage of suitable donor lungs. Organ quality can be compromised by lung ischemic reperfusion injury (LIRI). LIRI causes pulmonary endothelial inflammation and may lead to primary graft dysfunction (PGD). PGD is a significant cause of morbidity and mortality post-LTx. Research into preservation strategies that decrease the risk of LIRI and PGD is needed, and ex-situ lung perfusion (ESLP) is the foremost technological advancement in this field. This review addresses three major topics in the field of LTx: first, we review the clinical manifestation of LIRI post-LTx; second, we discuss the pathophysiology of LIRI that leads to pulmonary endothelial inflammation and PGD; and third, we present the role of ESLP as a therapeutic vehicle to mitigate this physiologic insult, increase the rates of donor organ utilization, and improve patient outcomes.
Collapse
|
21
|
Natalini JG, Diamond JM. Primary Graft Dysfunction. Semin Respir Crit Care Med 2021; 42:368-379. [PMID: 34030200 DOI: 10.1055/s-0041-1728794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
Abstract
Primary graft dysfunction (PGD) is a form of acute lung injury after transplantation characterized by hypoxemia and the development of alveolar infiltrates on chest radiograph that occurs within 72 hours of reperfusion. PGD is among the most common early complications following lung transplantation and significantly contributes to increased short-term morbidity and mortality. In addition, severe PGD has been associated with higher 90-day and 1-year mortality rates compared with absent or less severe PGD and is a significant risk factor for the subsequent development of chronic lung allograft dysfunction. The International Society for Heart and Lung Transplantation released updated consensus guidelines in 2017, defining grade 3 PGD, the most severe form, by the presence of alveolar infiltrates and a ratio of PaO2:FiO2 less than 200. Multiple donor-related, recipient-related, and perioperative risk factors for PGD have been identified, many of which are potentially modifiable. Consistently identified risk factors include donor tobacco and alcohol use; increased recipient body mass index; recipient history of pulmonary hypertension, sarcoidosis, or pulmonary fibrosis; single lung transplantation; and use of cardiopulmonary bypass, among others. Several cellular pathways have been implicated in the pathogenesis of PGD, thus presenting several possible therapeutic targets for preventing and treating PGD. Notably, use of ex vivo lung perfusion (EVLP) has become more widespread and offers a potential platform to safely investigate novel PGD treatments while expanding the lung donor pool. Even in the presence of significantly prolonged ischemic times, EVLP has not been associated with an increased risk for PGD.
Collapse
Affiliation(s)
- Jake G Natalini
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua M Diamond
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
22
|
Pushing the Envelope for Donor Lungs. Semin Respir Crit Care Med 2021; 42:357-367. [PMID: 34030199 DOI: 10.1055/s-0041-1729859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The shortage of organ donors remains the major limiting factor in lung transplant, with the number of patients on the waiting list largely exceeding the number of available organ donors. Another issue is the low utilization rate seen in some types of donors. Therefore, novel strategies are continuously being explored to increase the donor pool. Advanced age, smoking history, positive serologies, and size mismatch are common criteria that decrease the rate of use when it comes to organ utilization. Questioning these limitations is one of the purposes of this review. Challenging these limitations by adapting novel donor management strategies could help to increase the rate of suitable lungs for transplantation while still maintaining good outcomes. A second goal is to present the latest advances in organ donation after controlled and uncontrolled cardiac death, and also on how to improve these lungs on ex vivo platforms for assessment and future specific therapies. Finally, pushing the limit of the donor envelope also means reviewing some of the recent improvements made in lung preservation itself, as well as upcoming experimental research fields. In summary, donor lung optimization refers to a global care strategy to increase the total numbers of available allografts, and preserve or improve organ quality without paying the price of early-, mid-, or long-term negative outcomes after transplantation.
Collapse
|
23
|
Carbonnel M, Cornet N, Revaux A, Favre-Inhofer A, Galio L, Raliou M, Couturier-Tarrade A, Giraud-Delville C, Charpigny G, Gelin V, Dubois O, Hersant B, Bosc R, Coscas R, Vialard F, Chavatte-Palmer P, Richard C, Sandra O, Ayoubi JM. Analysis of blood parameters and molecular endometrial markers during early reperfusion in two ovine models of uterus transplantation. PLoS One 2021; 16:e0251474. [PMID: 34003831 PMCID: PMC8130915 DOI: 10.1371/journal.pone.0251474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/27/2021] [Indexed: 11/30/2022] Open
Abstract
The dissection of the veins is the trickiest step of Uterine transplantation (UTx). Performing the anastomosis of a single uterine vein could bring a therapeutic benefit and simplification of surgery and serve for managing unilateral venous thromboses. The objectives of this project were to evaluate the expression of early markers of ischemia-reperfusion and to compare findings following one or two vein anastomoses. Orthotopic uterine auto-transplantations were performed on an ovine model with anastomosis of either two (group 1) or one utero-ovarian veins (group 2). Blood gases, histology and ischemia- reperfusion markers transcripts (PTGS2, IL6, IL8, SOD2, C3, BAX/BCL2 and TLR4) were analyzed as well as PTGS2 protein expression using Western Blot and fluorescence immunolocalization on endometrial biopsies after 3h of reperfusion. Ten ewes were included in the experimentation, 4 were in group1, 3 in group 2, the others being sham operated controls. No significant differences were observed between the two phenotypes. Based on these results, the anastomosis of one single uterine vein appears to be an approach consistent with short-term graft survival. Further experiments will be needed to confirm the reliability of this approach, especially the long-term follow-up of the uterine graft including its ability to support gestation to term.
Collapse
Affiliation(s)
- Marie Carbonnel
- Department of Gynaecology and Obstetrics, Foch Hospital, Suresnes, France
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
- * E-mail:
| | - Nathalie Cornet
- Department of Gynaecology and Obstetrics, Foch Hospital, Suresnes, France
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Aurélie Revaux
- Department of Gynaecology and Obstetrics, Foch Hospital, Suresnes, France
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Angéline Favre-Inhofer
- Department of Gynaecology and Obstetrics, Foch Hospital, Suresnes, France
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Laurent Galio
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Mariam Raliou
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Corinne Giraud-Delville
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Gilles Charpigny
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Valérie Gelin
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Olivier Dubois
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Barbara Hersant
- Department of Plastic, Reconstructive, Aesthetic and Maxillofacial Surgery, Henri Mondor Hospital, Créteil, France
| | - Romain Bosc
- Department of Plastic, Reconstructive, Aesthetic and Maxillofacial Surgery, Henri Mondor Hospital, Créteil, France
| | - Raphael Coscas
- Department of Vascular Surgery, Ambroise Paré University Hospital, Boulogne-Billancourt, France
- UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Boulogne-Billancourt, France
| | - François Vialard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Christophe Richard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Olivier Sandra
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Jean-Marc Ayoubi
- Department of Gynaecology and Obstetrics, Foch Hospital, Suresnes, France
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| |
Collapse
|
24
|
van Zanden JE, Leuvenink HGD, Verschuuren EAM, Veldhuis ZJ, Ottens PJ, Erasmus ME, Hottenrott MC. Ex Vivo Perfusion With Methylprednisolone Attenuates Brain Death-induced Lung Injury in Rats. Transplant Direct 2021; 7:e682. [PMID: 33748411 PMCID: PMC7969243 DOI: 10.1097/txd.0000000000001141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/24/2021] [Accepted: 02/02/2021] [Indexed: 11/25/2022] Open
Abstract
The onset of brain death (BD) leads to the deterioration of potential donor lungs. Methylprednisolone is considered to increase lung oxygenation capacity and enhance the procurement yield of donor lungs, when applied in situ, during donor management. However, whether BD-induced lung damage is ameliorated upon treatment with methylprednisolone during acellular ex vivo lung perfusion (EVLP), remains unknown. We aimed to investigate whether the quality of lungs from brain-dead donors improves upon methylprednisolone treatment during EVLP.
Collapse
Affiliation(s)
- Judith E van Zanden
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Erik A M Verschuuren
- Department of Pulmonary Diseases, University Medical Center Groningen, Groningen, The Netherlands
| | - Zwanida J Veldhuis
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Petra J Ottens
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel E Erasmus
- Department of Cardiothoracic Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Maximilia C Hottenrott
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands.,Department of Surgery, University of Regensburg, Regensburg, Germany
| |
Collapse
|
25
|
Prediction of donor related lung injury in clinical lung transplantation using a validated ex vivo lung perfusion inflammation score. J Heart Lung Transplant 2021; 40:687-695. [PMID: 33781664 DOI: 10.1016/j.healun.2021.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Ex vivo lung perfusion (EVLP) is an isolated organ assessment technique that has revolutionized the field of lung transplantation and enabled a safe increase in the number of organs transplanted. The objective of this study was to develop a protein-based assay that would provide a precision medicine approach to lung injury assessment during EVLP. METHODS Perfusate samples collected from clinical EVLP cases performed from 2009 to 2019 were separated into development (n = 281) and validation (n = 57) sets to derive and validate an inflammation score based on IL-6 and IL-8 protein levels in perfusate. The ability of an inflammation score to predict lungs suitable for transplantation and likely to produce excellent recipient outcomes (time on ventilator ≤ 3 days) was assessed. Inflammation scores were compared to conventional clinical EVLP assessment parameters and associated with outcomes, including primary graft dysfunction and patient care in the ICU. RESULTS An inflammation score accurately predicted the decision to transplant (AUROC 68% [95% CI 62-74]) at the end of EVLP and those transplants associated with short ventilator times (AUROC 73% [95% CI 66-80]). The score identified lungs more likely to develop primary graft dysfunction at 72-hours post-transplant (OR 4.0, p = 0.03). A model comprised of the inflammation score and ∆PO2 was able to determine EVLP transplants that were likely to have excellent recipient outcomes, with an accuracy of 87% [95% CI 83-92]. CONCLUSIONS The adoption of an inflammation score will improve accuracy of EVLP decision-making and increase confidence of surgical teams to determine lungs that are suitable for transplantation, thereby improving organ utilization rates and patient outcomes.
Collapse
|
26
|
Guo Y, Krupnick AS. Commentary: 'Tis the season to filter your perfusate. J Thorac Cardiovasc Surg 2021; 161:e127-e128. [PMID: 31959447 DOI: 10.1016/j.jtcvs.2019.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 01/31/2023]
Affiliation(s)
- Yizhan Guo
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | | |
Collapse
|
27
|
Natalini JG, Diamond JM, Porteous MK, Lederer DJ, Wille KM, Weinacker AB, Orens JB, Shah PD, Lama VN, McDyer JF, Snyder LD, Hage CA, Singer JP, Ware LB, Cantu E, Oyster M, Kalman L, Christie JD, Kawut SM, Bernstein EJ. Risk of primary graft dysfunction following lung transplantation in selected adults with connective tissue disease-associated interstitial lung disease. J Heart Lung Transplant 2021; 40:351-358. [PMID: 33637413 DOI: 10.1016/j.healun.2021.01.1391] [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: 09/22/2020] [Revised: 12/23/2020] [Accepted: 01/19/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Previous studies have reported similarities in long-term outcomes following lung transplantation for connective tissue disease-associated interstitial lung disease (CTD-ILD) and idiopathic pulmonary fibrosis (IPF). However, it is unknown whether CTD-ILD patients are at increased risk of primary graft dysfunction (PGD), delays in extubation, or longer index hospitalizations following transplant compared to IPF patients. METHODS We performed a multicenter retrospective cohort study of CTD-ILD and IPF patients enrolled in the Lung Transplant Outcomes Group registry who underwent lung transplantation between 2012 and 2018. We utilized mixed effects logistic regression and stratified Cox proportional hazards regression to determine whether CTD-ILD was independently associated with increased risk for grade 3 PGD or delays in post-transplant extubation and hospital discharge compared to IPF. RESULTS A total of 32.7% (33/101) of patients with CTD-ILD and 28.9% (145/501) of patients with IPF developed grade 3 PGD 48-72 hours after transplant. There were no significant differences in odds of grade 3 PGD among patients with CTD-ILD compared to those with IPF (adjusted OR 1.12, 95% CI 0.64-1.97, p = 0.69), nor was CTD-ILD independently associated with a longer post-transplant time to extubation (adjusted HR for first extubation 0.87, 95% CI 0.66-1.13, p = 0.30). However, CTD-ILD was independently associated with a longer post-transplant hospital length of stay (median 23 days [IQR 14-35 days] vs17 days [IQR 12-28 days], adjusted HR for hospital discharge 0.68, 95% CI 0.51-0.90, p = 0.008). CONCLUSION Patients with CTD-ILD experienced significantly longer postoperative hospitalizations compared to IPF patients without an increased risk of grade 3 PGD.
Collapse
Affiliation(s)
- Jake G Natalini
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua M Diamond
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mary K Porteous
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Keith M Wille
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Ann B Weinacker
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Jonathan B Orens
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pali D Shah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vibha N Lama
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - John F McDyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Laurie D Snyder
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Chadi A Hage
- Division of Pulmonary Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jonathan P Singer
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco School of Medicine, San Francisco, California
| | - Lorraine B Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Edward Cantu
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michelle Oyster
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laurel Kalman
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason D Christie
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven M Kawut
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elana J Bernstein
- Division of Rheumatology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York.
| |
Collapse
|
28
|
Oshima Y, Otsuki A, Endo R, Nakasone M, Harada T, Takahashi S, Inagaki Y. The Effects of Volatile Anesthetics on Lung Ischemia-Reperfusion Injury: Basic to Clinical Studies. J Surg Res 2020; 260:325-344. [PMID: 33373852 DOI: 10.1016/j.jss.2020.11.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023]
Abstract
Case reports from as early as the 1970s have shown that intravenous injection of even a small dose of volatile anesthetics result in fatal lung injury. Direct contact between volatile anesthetics and pulmonary vasculature triggers chemical damage in the vessel walls. A wide variety of factors are involved in lung ischemia-reperfusion injury (LIRI), such as pulmonary endothelial cells, alveolar epithelial cells, alveolar macrophages, neutrophils, mast cells, platelets, proinflammatory cytokines, and surfactant. With a constellation of factors involved, the assessment of the protective effect of volatile anesthetics in LIRI is difficult. Multiple animal studies have reported that with regards to LIRI, sevoflurane demonstrates an anti-inflammatory effect in immunocompetent cells and an anti-apoptotic effect on lung tissue. Scattered studies have dismissed a protective effect of desflurane against LIRI. While a single-center randomized controlled trial (RCT) found that volatile anesthetics including desflurane demonstrated a lung-protective effect in thoracic surgery, a multicenter RCT did not demonstrate a lung-protective effect of desflurane. LIRI is common in lung transplantation. One study, although limited due to its small sample size, found that the use of volatile anesthetics in organ procurement surgery involving "death by neurologic criteria" donors did not improve lung graft survival. Future studies on the protective effect of volatile anesthetics against LIRI must examine not only the mechanism of the protective effect but also differences in the effects of different types of volatile anesthetics, their optimal dosage, and the appropriateness of their use in the event of marked alveolar capillary barrier damage.
Collapse
Affiliation(s)
- Yoshiaki Oshima
- Department of Anesthesiology, Yonago Medical Center, Yonago, Tottori, Japan.
| | - Akihiro Otsuki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Ryo Endo
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Masato Nakasone
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tomomi Harada
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Shunsaku Takahashi
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yoshimi Inagaki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
29
|
Ball AL, Edge RJ, Amin K, Critchley WR, Howell GJ, Yonan N, Stone JP, Fildes JE. A post-preservation vascular flush removes significant populations of donor leukocytes prior to lung transplantation. Transpl Immunol 2020; 64:101356. [PMID: 33264679 DOI: 10.1016/j.trim.2020.101356] [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: 06/04/2020] [Revised: 10/30/2020] [Accepted: 11/25/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Donor leukocytes are intrinsically involved in acute lung allograft rejection, via self-presentation of donor antigens to recipient leukocytes. Therapeutic modalities to remove donor leukocytes are currently unavailable. We evaluated if a vascular flush immediately following preservation can be used for this purpose. METHODS A post-preservation flush was performed with STEEN solution in n = 6 porcine lungs following static cold storage. The first 500 ml effluent from the left atrium was collected and an inflammatory profile performed. RESULTS A total of 1.17 billion (±2.8 × 108) viable leukocytes were identified within the effluent. T cells were the dominant cell population, representing 82% of the total mobilised leukocytes, of which <0.01% were regulatory T cells. IL-18 was the most abundant cytokine, with a mean concentration of 84,216 pg (±153,552 pg). In addition, there was a mean concentration of 8819 ng (±4415) cell-free mitochondrial DNA. CONCLUSION There is an immediate transfer of donor leukocytes, cytokines and damage-associated molecular patterns following reperfusion. Such a pro-inflammatory donor load may enhance alloantigen presentation and drive recipient alloimmune responses. A post-preservation flush may therefore be an effective method for reducing the immune burden of the donor lung prior to transplantation.
Collapse
Affiliation(s)
- Alexandra L Ball
- The Transplant Centre, University of Manchester NHS Foundation Trust, Manchester M23 9LT, United Kingdom; The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, United Kingdom
| | - Rebecca J Edge
- The Transplant Centre, University of Manchester NHS Foundation Trust, Manchester M23 9LT, United Kingdom; The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, United Kingdom
| | - Kavit Amin
- The Transplant Centre, University of Manchester NHS Foundation Trust, Manchester M23 9LT, United Kingdom; The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, United Kingdom
| | - William R Critchley
- The Transplant Centre, University of Manchester NHS Foundation Trust, Manchester M23 9LT, United Kingdom; The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, United Kingdom
| | - Gareth J Howell
- Flow Cytometry Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, United Kingdom
| | - Nizar Yonan
- The Transplant Centre, University of Manchester NHS Foundation Trust, Manchester M23 9LT, United Kingdom
| | - John P Stone
- The Transplant Centre, University of Manchester NHS Foundation Trust, Manchester M23 9LT, United Kingdom; The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, United Kingdom
| | - James E Fildes
- The Transplant Centre, University of Manchester NHS Foundation Trust, Manchester M23 9LT, United Kingdom; The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, United Kingdom.
| |
Collapse
|
30
|
Leligdowicz A, Ross JT, Nesseler N, Matthay MA. The endogenous capacity to produce proinflammatory mediators by the ex vivo human perfused lung. Intensive Care Med Exp 2020; 8:56. [PMID: 32955627 PMCID: PMC7505905 DOI: 10.1186/s40635-020-00343-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/04/2020] [Indexed: 12/19/2022] Open
Abstract
Background The ex vivo human perfused lung model has enabled optimizing donor lungs for transplantation and delineating mechanisms of lung injury. Perfusate and airspace biomarkers are a proxy of the lung response to experimental conditions. However, there is a lack of studies evaluating biomarker kinetics during perfusion and after exposure to stimuli. In this study, we analyzed the ex vivo-perfused lung response to three key perturbations: exposure to the perfusion circuit, exogenous fresh whole blood, and bacteria. Results Ninety-nine lungs rejected for transplantation underwent ex vivo perfusion. One hour after reaching experimental conditions, fresh whole blood was added to the perfusate (n = 55). Two hours after reaching target temperature, Streptococcus pneumoniae was added to the perfusate (n = 42) or to the airspaces (n = 17). Perfusate and airspace samples were collected at baseline (once lungs were equilibrated for 1 h, but before blood or bacteria were added) and 4 h later. Interleukin (IL)-6, IL-8, angiopoietin (Ang)-2, and soluble tumor necrosis factor receptor (sTNFR)-1 were quantified. Baseline perfusate and airspace biomarker levels varied significantly, and this was not related to pre-procurement PaO2:FiO2 ratio, cold ischemia time, and baseline alveolar fluid clearance (AFC). After 4 h of ex vivo perfusion, the lung demonstrated a sustained production of proinflammatory mediators. The change in biomarker levels was not influenced by baseline donor lung characteristics (cold ischemia time, baseline AFC) nor was it associated with measures of experimental epithelial (final AFC) or endothelial (percent weight gain) injury. In the presence of exogenous blood, the rise in biomarkers was attenuated. Lungs exposed to intravenous (IV) bacteria relative to control lungs demonstrated a significantly higher rise in perfusate IL-6. Conclusions The ex vivo-perfused lung has a marked endogenous capacity to produce inflammatory mediators over the course of short-term perfusion that is not significantly influenced by donor lung characteristics or the presence of exogenous blood, and only minimally affected by the introduction of systemic bacteremia. The lack of association between biomarker change and donor lung cold ischemia time, final alveolar fluid clearance, and experimental percent weight gain suggests that the maintained ability of the human lung to produce biomarkers is not merely a marker of lung epithelial or endothelial injury, but may support the function of the lung as an immune cell reservoir.
Collapse
Affiliation(s)
- Aleksandra Leligdowicz
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA. .,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - James T Ross
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Nicolas Nesseler
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.,Department of Anesthesia and Critical Care, Pontchaillou, University Hospital of Rennes, Rennes, France.,Univ Rennes, CHU de Rennes, Inra, Inserm, Institut NUMECAN-UMR_A 1341, UMR_S 1241, 35000, Rennes, France.,Univ Rennes, CHU Rennes, Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), 35000, Rennes, France
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.,Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA, USA.,Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
31
|
Jin Z, Hana Z, Alam A, Rajalingam S, Abayalingam M, Wang Z, Ma D. Review 1: Lung transplant-from donor selection to graft preparation. J Anesth 2020; 34:561-574. [PMID: 32476043 PMCID: PMC7261511 DOI: 10.1007/s00540-020-02800-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 05/17/2020] [Indexed: 12/16/2022]
Abstract
For various end-stage lung diseases, lung transplantation remains one of the only viable treatment options. While the demand for lung transplantation has steadily risen over the last few decades, the availability of donor grafts is limited, which have resulted in progressively longer waiting lists. In the early years of lung transplantation, only the 'ideal' donor grafts are considered for transplantation. Due to the donor shortages, there is ongoing discussion about the safe use of 'suboptimal' grafts to expand the donor pool. In this review, we will discuss the considerations around donor selection, donor-recipient matching, graft preparation and graft optimisation.
Collapse
Affiliation(s)
- Zhaosheng Jin
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Zac Hana
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Azeem Alam
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Shamala Rajalingam
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Mayavan Abayalingam
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Zhiping Wang
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK.
| |
Collapse
|
32
|
Xenogeneic cross-circulation for extracorporeal recovery of injured human lungs. Nat Med 2020; 26:1102-1113. [PMID: 32661401 PMCID: PMC9990469 DOI: 10.1038/s41591-020-0971-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/09/2020] [Indexed: 12/15/2022]
Abstract
Patients awaiting lung transplantation face high wait-list mortality, as injury precludes the use of most donor lungs. Although ex vivo lung perfusion (EVLP) is able to recover marginal quality donor lungs, extension of normothermic support beyond 6 h has been challenging. Here we demonstrate that acutely injured human lungs declined for transplantation, including a lung that failed to recover on EVLP, can be recovered by cross-circulation of whole blood between explanted human lungs and a Yorkshire swine. This xenogeneic platform provided explanted human lungs a supportive, physiologic milieu and systemic regulation that resulted in functional and histological recovery after 24 h of normothermic support. Our findings suggest that cross-circulation can serve as a complementary approach to clinical EVLP to recover injured donor lungs that could not otherwise be utilized for transplantation, as well as a translational research platform for immunomodulation and advanced organ bioengineering.
Collapse
|
33
|
Dorrello NV, Vunjak-Novakovic G. Bioengineering of Pulmonary Epithelium With Preservation of the Vascular Niche. Front Bioeng Biotechnol 2020; 8:269. [PMID: 32351946 PMCID: PMC7174601 DOI: 10.3389/fbioe.2020.00269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/16/2020] [Indexed: 12/20/2022] Open
Abstract
The shortage of transplantable donor organs directly affects patients with end-stage lung disease, for which transplantation remains the only definitive treatment. With the current acceptance rate of donor lungs of only 20%, rescuing even one half of the rejected donor lungs would increase the number of transplantable lungs threefold, to 60%. We review recent advances in lung bioengineering that have potential to repair the epithelial and vascular compartments of the lung. Our focus is on the long-term support and recovery of the lung ex vivo, and the replacement of defective epithelium with healthy therapeutic cells. To this end, we first review the roles of the lung epithelium and vasculature, with focus on the alveolar-capillary membrane, and then discuss the available and emerging technologies for ex vivo bioengineering of the lung by decellularization and recellularization. While there have been many meritorious advances in these technologies for recovering marginal quality lungs to the levels needed to meet the standards for transplantation – many challenges remain, motivating further studies of the extended ex vivo support and interventions in the lung. We propose that the repair of injured epithelium with preservation of quiescent vasculature will be critical for the immediate blood supply to the lung and the lung survival and function following transplantation.
Collapse
Affiliation(s)
- N Valerio Dorrello
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, United States.,Department of Medicine, Columbia University, New York, NY, United States
| |
Collapse
|
34
|
Gu C, Pan X, Shi J. Progress of Clinical Application for Ex Vivo Lung Perfusion (EVLP) in Lung Transplantation. Methods Mol Biol 2020; 2204:217-224. [PMID: 32710328 DOI: 10.1007/978-1-0716-0904-0_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In recent years, medical advances make lung transplantation become a standard treatment for terminal lung diseases (such as emphysema, pulmonary fibrosis, pulmonary cystic fibrosis, and pulmonary arterial hypertension) that cannot be cured by drugs or surgery (Lund et al., J Heart Lung Transplant 34:1244, 2015). However, the current number of donor lungs that meet the transplant criteria is no longer sufficient for transplanting, causing some patients to die while waiting for a suitable lung. Current methods for improving the situation of shortage of lung transplant donors include the use of donation after cardiac death (DCD) donors, smoker donors, and Ex Vivo Lung Perfusion (EVLP). Among them, EVLP is a technique for extending lung preservation time and repairing lung injury in the field of lung transplantation. By continuously assessing and improving the function of marginal donor lungs, EVLP increases the number of lungs that meet the transplant criteria and, to some extent, alleviates the current situation of shortage of donor lungs. This chapter reviews the clinical application and research progress of EVLP in the field of lung transplantation.
Collapse
Affiliation(s)
- Chang Gu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xufeng Pan
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianxin Shi
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
35
|
Nakajima D, Watanabe Y, Ohsumi A, Pipkin M, Chen M, Mordant P, Kanou T, Saito T, Lam R, Coutinho R, Caldarone L, Juvet S, Martinu T, Iyer RK, Davies JE, Hwang DM, Waddell TK, Cypel M, Liu M, Keshavjee S. Mesenchymal stromal cell therapy during ex vivo lung perfusion ameliorates ischemia-reperfusion injury in lung transplantation. J Heart Lung Transplant 2019; 38:1214-1223. [DOI: 10.1016/j.healun.2019.07.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/15/2019] [Accepted: 07/20/2019] [Indexed: 12/21/2022] Open
|
36
|
Scozzi D, Ibrahim M, Liao F, Lin X, Hsiao HM, Hachem R, Tague LK, Ricci A, Kulkarni HS, Huang HJ, Sugimoto S, Krupnick AS, Kreisel D, Gelman AE. Mitochondrial damage-associated molecular patterns released by lung transplants are associated with primary graft dysfunction. Am J Transplant 2019; 19:1464-1477. [PMID: 30582269 PMCID: PMC6482093 DOI: 10.1111/ajt.15232] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/12/2018] [Accepted: 12/08/2018] [Indexed: 02/07/2023]
Abstract
Primary graft dysfunction (PGD) is a major limitation in short- and long-term lung transplant survival. Recent work has shown that mitochondrial damage-associated molecular patterns (mtDAMPs) can promote solid organ injury, but whether they contribute to PGD severity remains unclear. We quantitated circulating plasma mitochondrial DNA (mtDNA) in 62 patients, before lung transplantation and shortly after arrival to the intensive care unit. Although all recipients released mtDNA, high levels were associated with severe PGD development. In a mouse orthotopic lung transplant model of PGD, we detected airway cell-free damaged mitochondria and mtDNA in the peripheral circulation. Pharmacologic inhibition or genetic deletion of formylated peptide receptor 1 (FPR1), a chemotaxis sensor for N-formylated peptides released by damaged mitochondria, inhibited graft injury. An analysis of intragraft neutrophil-trafficking patterns reveals that FPR1 enhances neutrophil transepithelial migration and retention within airways but does not control extravasation. Using donor lungs that express a mitochondria-targeted reporter protein, we also show that FPR1-mediated neutrophil trafficking is coupled with the engulfment of damaged mitochondria, which in turn triggers reactive oxygen species (ROS)-induced pulmonary edema. Therefore, our data demonstrate an association between mtDAMP release and PGD development and suggest that neutrophil trafficking and effector responses to damaged mitochondria are drivers of graft damage.
Collapse
Affiliation(s)
- Davide Scozzi
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department of Clinical & Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mohsen Ibrahim
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department Medical-Surgical Science & Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Fuyi Liao
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Xue Lin
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Hsi-Min Hsiao
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Ramsey Hachem
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Laneshia K Tague
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Alberto Ricci
- Department of Clinical & Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Hrishikesh S Kulkarni
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Howard J Huang
- Houston Methodist J. C. Walter Jr. Transplant Center, Houston, Texas
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery, Okayama University Hospital, Okayama, Japan
| | - Alexander S Krupnick
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Daniel Kreisel
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew E Gelman
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department Medical-Surgical Science & Translational Medicine, Sapienza University of Rome, Rome, Italy
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
37
|
Renne J, Gutberlet M, Voskrebenzev A, Kern A, Kaireit T, Hinrichs JB, Braubach P, Falk CS, Höffler K, Warnecke G, Zardo P, Haverich A, Wacker F, Vogel-Claussen J, Zinne N. Functional Pulmonary Magnetic Resonance Imaging for Detection of Ischemic Injury in a Porcine Ex-Vivo Lung Perfusion System Prior to Transplantation. Acad Radiol 2019; 26:170-178. [PMID: 29929935 DOI: 10.1016/j.acra.2018.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 10/28/2022]
Abstract
RATIONALE AND OBJECTIVES To evaluate the feasibility of multiparametric magnetic resonance imaging (MRI) of the lungs to detect impaired organ function in a porcine model of ischemic injury within an ex-vivo lung perfusion system (EVLP) prior to transplantation. MATERIALS AND METHODS Twelve pigs were anesthetized, and left lungs were clamped to induce warm ischemia for 3 hours. Right lungs remained perfused as controls. Lungs were removed and installed in an EVLP for 12 hours. Lungs in the EVLP were imaged repeatedly using computed tomography, proton MRI (1H-MRI) and fluorine MRI (19F-MRI). Dynamic contrast-enhanced derived parenchymal blood volume, oxygen washout times, and 19F washout times were calculated. PaO2 was measured for ischemic and normal lungs, wet/dry ratio was determined, histologic samples were assessed, and cytokines in the lung tissue were analyzed. Statistical analysis was performed using nonparametric testing. RESULTS Eleven pigs were included in the final analysis. Ischemic lungs showed significantly higher wet/dry ratios (p = 0.024), as well as IL-8 tissue levels (p = 0.0098). Histologic assessment as well as morphologic scoring of computed tomography and 1H-MRI did not reveal significant differences between ischemic and control lungs. 19F washout (p = 0.966) and parenchymal blood flow (p = 0.32) were not significantly different. Oxygen washout was significantly prolonged in ischemic lungs compared to normal control lungs at the beginning (p = 0.018) and further prolonged at the end of the EVLP run (p = 0.005). CONCLUSION Multiparametric pulmonary MRI is feasible in lung allografts within an EVLP system. Oxygen-enhanced imaging seems to be a promising marker for ischemic injury, enabling detection of affected lung segments prior to transplantation.
Collapse
|
38
|
Ali A, Keshavjee S, Cypel M. Rising to the Challenge of Unmet Need: Expanding the Lung Donor Pool. CURRENT PULMONOLOGY REPORTS 2018. [DOI: 10.1007/s13665-018-0205-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
39
|
Verleden SE, Martens A, Ordies S, Neyrinck AP, Van Raemdonck DE, Verleden GM, Vanaudenaerde BM, Vos R. Immediate post-operative broncho-alveolar lavage IL-6 and IL-8 are associated with early outcomes after lung transplantation. Clin Transplant 2018; 32:e13219. [PMID: 29405435 DOI: 10.1111/ctr.13219] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2018] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Previous studies demonstrated that increased cytokine and chemokine levels, either shortly before or after lung transplantation, were associated with post-transplant outcome. However, small patient cohorts were mostly used, focusing on 1 molecule and 1 outcome. In a large single-center cohort, we investigated the predictive value of immediate post-operative broncho-alveolar lavage (BAL) expression of IL-6 and IL-8 on multiple key outcomes, including PGD, CLAD, graft survival, as well as several secondary outcomes. MATERIAL AND METHODS All patients undergoing a first lung transplant in whom routine bronchoscopy with BAL was performed during the first 48 hours post-transplantation were included. IL-6 and IL-8 protein levels were measured in BAL via ELISA. RESULTS A total of 336 patients were included. High IL-6 levels measured within 24 hours of transplantation were associated with longer time on ICU and time to hospital discharge; and increased prevalence of PGD grade 3. Increased IL-8 levels, measured within 24 hours, were associated with PGD3, more ECMO use, higher donor paO2 , younger donor age, but not with other short-or long-term outcome. IL-6 and IL-8 measured between 24 and 48 hours of transplantation were not associated with any outcome parameters. CONCLUSION Recipient BAL IL-6 and IL-8 within 24 hours post-transplant were associated with an increased incidence of PGD3.
Collapse
Affiliation(s)
- Stijn E Verleden
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| | - An Martens
- Department of cardiovascular sciences, KU Leuven, Leuven, Belgium
| | - Sofie Ordies
- Department of cardiovascular sciences, KU Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Department of cardiovascular sciences, KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| |
Collapse
|
40
|
Fisher A, Andreasson A, Chrysos A, Lally J, Mamasoula C, Exley C, Wilkinson J, Qian J, Watson G, Lewington O, Chadwick T, McColl E, Pearce M, Mann K, McMeekin N, Vale L, Tsui S, Yonan N, Simon A, Marczin N, Mascaro J, Dark J. An observational study of Donor Ex Vivo Lung Perfusion in UK lung transplantation: DEVELOP-UK. Health Technol Assess 2018; 20:1-276. [PMID: 27897967 DOI: 10.3310/hta20850] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Many patients awaiting lung transplantation die before a donor organ becomes available. Ex vivo lung perfusion (EVLP) allows initially unusable donor lungs to be assessed and reconditioned for clinical use. OBJECTIVE The objective of the Donor Ex Vivo Lung Perfusion in UK lung transplantation study was to evaluate the clinical effectiveness and cost-effectiveness of EVLP in increasing UK lung transplant activity. DESIGN A multicentre, unblinded, non-randomised, non-inferiority observational study to compare transplant outcomes between EVLP-assessed and standard donor lungs. SETTING Multicentre study involving all five UK officially designated NHS adult lung transplant centres. PARTICIPANTS Patients aged ≥ 18 years with advanced lung disease accepted onto the lung transplant waiting list. INTERVENTION The study intervention was EVLP assessment of donor lungs before determining suitability for transplantation. MAIN OUTCOME MEASURES The primary outcome measure was survival during the first 12 months following lung transplantation. Secondary outcome measures were patient-centred outcomes that are influenced by the effectiveness of lung transplantation and that contribute to the health-care costs. RESULTS Lungs from 53 donors unsuitable for standard transplant were assessed with EVLP, of which 18 (34%) were subsequently transplanted. A total of 184 participants received standard donor lungs. Owing to the early closure of the study, a non-inferiority analysis was not conducted. The Kaplan-Meier estimate of survival at 12 months was 0.67 [95% confidence interval (CI) 0.40 to 0.83] for the EVLP arm and 0.80 (95% CI 0.74 to 0.85) for the standard arm. The hazard ratio for overall 12-month survival in the EVLP arm relative to the standard arm was 1.96 (95% CI 0.83 to 4.67). Patients in the EVLP arm required ventilation for a longer period and stayed longer in an intensive therapy unit (ITU) than patients in the standard arm, but duration of overall hospital stay was similar in both groups. There was a higher rate of very early grade 3 primary graft dysfunction (PGD) in the EVLP arm, but rates of PGD did not differ between groups after 72 hours. The requirement for extracorporeal membrane oxygenation (ECMO) support was higher in the EVLP arm (7/18, 38.8%) than in the standard arm (6/184, 3.2%). There were no major differences in rates of chest radiograph abnormalities, infection, lung function or rejection by 12 months. The cost of EVLP transplants is approximately £35,000 higher than the cost of standard transplants, as a result of the cost of the EVLP procedure, and the increased ECMO use and ITU stay. Predictors of cost were quality of life on joining the waiting list, type of transplant and number of lungs transplanted. An exploratory model comparing a NHS lung transplant service that includes EVLP and standard lung transplants with one including only standard lung transplants resulted in an incremental cost-effectiveness ratio of £73,000. Interviews showed that patients had a good understanding of the need for, and the processes of, EVLP. If EVLP can increase the number of usable donor lungs and reduce waiting, it is likely to be acceptable to those waiting for lung transplantation. Study limitations include small numbers in the EVLP arm, limiting analysis to descriptive statistics and the EVLP protocol change during the study. CONCLUSIONS Overall, one-third of donor lungs subjected to EVLP were deemed suitable for transplant. Estimated survival over 12 months was lower than in the standard group, but the data were also consistent with no difference in survival between groups. Patients receiving these additional transplants experience a higher rate of early graft injury and need for unplanned ECMO support, at increased cost. The small number of participants in the EVLP arm because of early study termination limits the robustness of these conclusions. The reason for the increased PGD rates, high ECMO requirement and possible differences in lung injury between EVLP protocols needs evaluation. TRIAL REGISTRATION Current Controlled Trials ISRCTN44922411. FUNDING This project was funded by the NIHR Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 20, No. 85. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Andrew Fisher
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Anders Andreasson
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Alexandros Chrysos
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Joanne Lally
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | | | - Catherine Exley
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | | | - Jessica Qian
- Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, UK
| | - Gillian Watson
- Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, UK
| | | | - Thomas Chadwick
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Elaine McColl
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK.,Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Pearce
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Kay Mann
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Nicola McMeekin
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Luke Vale
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Steven Tsui
- Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Nizar Yonan
- University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Andre Simon
- Royal Brompton and Harefield Hospital NHS Foundation Trust, London, UK
| | - Nandor Marczin
- Royal Brompton and Harefield Hospital NHS Foundation Trust, London, UK
| | - Jorge Mascaro
- Queen Elizabeth Hospital NHS Foundation Trust, Birmingham, UK
| | - John Dark
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
41
|
Hamilton BCS, Dincheva GR, Zhuo H, Golden JA, Brzezinski M, Singer JP, Matthay MA, Kukreja J. Elevated donor plasminogen activator inhibitor-1 levels and the risk of primary graft dysfunction. Clin Transplant 2018; 32:e13210. [PMID: 29377268 DOI: 10.1111/ctr.13210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2018] [Indexed: 01/11/2023]
Abstract
Primary graft dysfunction (PGD) following lung transplantation is associated with elevated recipient plasma levels of plasminogen activator inhibitor-1 (PAI-1) and the receptor for advanced glycation end products (RAGE). However, the significance of these biomarkers in the donor plasma is uncertain. We hypothesized that elevated donor plasma levels of PAI-1 and RAGE would be associated with recipient PGD. We carried out a prospective unmatched case-control study of double-lung transplant recipients between May 2014 and September 2015. We compared donor plasma levels of PAI-1 and RAGE using rank-sum tests and t tests, in 12 recipients who developed PGD grade 2 or 3 within 72 hours postoperatively with 13 recipients who did not. Recipients who developed PGD had higher donor plasma levels of PAI-1 than recipients who did not (median 2.7 ng/mL vs 1.4; P = .03). Recipients with PGD also had numerically higher donor plasma levels of RAGE than recipients without PGD, although this difference did not achieve statistical significance (median 1061 pg/mL vs 679; P = .12). Systemic inflammatory responses in the donor, as reflected by elevated plasma levels of PAI-1, may contribute to the risk of developing PGD. Rapid biomarker assessment of easily available plasma samples may assist in donor lung selection and risk stratification.
Collapse
Affiliation(s)
| | | | - Hanjing Zhuo
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Jeffrey A Golden
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Marek Brzezinski
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Jonathan P Singer
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Michael A Matthay
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|
42
|
Affiliation(s)
- Robert Jeen-Chen Chen
- 1 Cardiothoracic Surgery Taipei Tzuchi Hospital Buddhist Tzuchi Medical Foundation New Taipei City, Taiwan.,2 Tzuchi University College of Medicine Buddhist Tzuchi Medical Foundation Hualian, Taiwan and
| | - Wei-Hsuan Yu
- 3 Biochemistry & Molecular Biology National Taiwan University College of Medicine Taipei, Taiwan
| |
Collapse
|
43
|
Abstract
Primary graft dysfunction is a form of acute injury after lung transplantation that is associated with significant short- and long-term morbidity and mortality. Multiple mechanisms contribute to the pathogenesis of primary graft dysfunction, including ischemia reperfusion injury, epithelial cell death, endothelial cell dysfunction, innate immune activation, oxidative stress, and release of inflammatory cytokines and chemokines. This article reviews the epidemiology, pathogenesis, risk factors, prevention, and treatment of primary graft dysfunction.
Collapse
Affiliation(s)
- Mary K Porteous
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Drive, Philadelphia, PA 19104, USA.
| | - James C Lee
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
44
|
Mazo C, Borgatta B, Pont T, Sandiumenge A, Moyano S, Roman A, Rello J. Procalcitonin accurately predicts lung transplant adults with low risk of pulmonary graft dysfunction and intensive care mortality. J Crit Care 2017; 44:142-147. [PMID: 29112904 DOI: 10.1016/j.jcrc.2017.10.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/27/2017] [Accepted: 10/28/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE We evaluated the association of procalcitonin (PCT), IL-6-8-10 plasma levels during the first 72h after lung transplantation (LT) with ICU-mortality, oxygenation, primary graft dysfunction (PGD), and one-year graft function after LT. MATERIAL AND METHODS Prospective, observational study. PCT and IL-6-8-10 plasma levels were measured at 24h, 48h and 72h after LT from 100 lung transplant recipients (LTr). Patients were followed until one year after LT. End-points were ICU survival, grade 3 PGD at 72h and one-year graft function. RESULTS Higher PCT at 24h was associated with lower PaO2/FIO2 ratio and Grade 3 PGD over the first 72h after LT (p<0.05). PCT at 24h was higher in the 9 patients who died (2.90 vs 1.47ng/mL, p<0.05), with AUC=0.74 for predicting ICU-mortality. All patients with PCT<2ng/mL at 24h following LT, survived in the ICU (p<0.05). PCT and IL-10 at 48h were correlated with FEV1 (rho=-0.35) and FVC (rho=-0.29) one year after LT. (p<0.05). CONCLUSIONS A breakpoint of PCT<2ng/mL within 24h has a high predictive value to exclude grade 3 PGD at 72h and for ICU survival. Moreover, both PCT and IL-10 within 48h were associated with significantly better graft function one year after surgery.
Collapse
Affiliation(s)
- Cristopher Mazo
- Critical Care Department, Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; CIBERES, Instituto de Salud Carlos III, Madrid, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain; Transplant Coordination Department, Vall d'Hebron University Hospital, Barcelona, Spain.
| | - Bárbara Borgatta
- Critical Care Department, Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; CIBERES, Instituto de Salud Carlos III, Madrid, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Teresa Pont
- Critical Care Department, Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Transplant Coordination Department, Vall d'Hebron University Hospital, Barcelona, Spain.
| | - Alberto Sandiumenge
- Critical Care Department, Vall d'Hebron University Hospital, Barcelona, Spain; CIBERES, Instituto de Salud Carlos III, Madrid, Spain; Transplant Coordination Department, Vall d'Hebron University Hospital, Barcelona, Spain.
| | - Silvia Moyano
- Vall d'Hebron Institut de Recerca, Barcelona, Spain.
| | - Antonio Roman
- Vall d'Hebron Institut de Recerca, Barcelona, Spain; CIBERES, Instituto de Salud Carlos III, Madrid, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain; Pulmonary Department, Vall d'Hebron University Hospital, Barcelona, Spain.
| | - Jordi Rello
- Critical Care Department, Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; CIBERES, Instituto de Salud Carlos III, Madrid, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
45
|
Andreasson ASI, Karamanou DM, Gillespie CS, Özalp F, Butt T, Hill P, Jiwa K, Walden HR, Green NJ, Borthwick LA, Clark SC, Pauli H, Gould KF, Corris PA, Ali S, Dark JH, Fisher AJ. Profiling inflammation and tissue injury markers in perfusate and bronchoalveolar lavage fluid during human ex vivo lung perfusion. Eur J Cardiothorac Surg 2017; 51:577-586. [PMID: 28082471 PMCID: PMC5400024 DOI: 10.1093/ejcts/ezw358] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/12/2016] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES: Availability of donor lungs suitable for transplant falls short of current demand and contributes to waiting list mortality. Ex vivo lung perfusion (EVLP) offers the opportunity to objectively assess and recondition organs unsuitable for immediate transplant. Identifying robust biomarkers that can stratify donor lungs during EVLP to use or non-use or for specific interventions could further improve its clinical impact. METHODS: In this pilot study, 16 consecutive donor lungs unsuitable for immediate transplant were assessed by EVLP. Key inflammatory mediators and tissue injury markers were measured in serial perfusate samples collected hourly and in bronchoalveolar lavage fluid (BALF) collected before and after EVLP. Levels were compared between donor lungs that met criteria for transplant and those that did not. RESULTS: Seven of the 16 donor lungs (44%) improved during EVLP and were transplanted with uniformly good outcomes. Tissue and vascular injury markers lactate dehydrogenase, HMGB-1 and Syndecan-1 were significantly lower in perfusate from transplanted lungs. A model combining IL-1β and IL-8 concentrations in perfusate could predict final EVLP outcome after 2 h assessment. In addition, perfusate IL-1β concentrations showed an inverse correlation to recipient oxygenation 24 h post-transplant. CONCLUSIONS: This study confirms the feasibility of using inflammation and tissue injury markers in perfusate and BALF to identify donor lungs most likely to improve for successful transplant during clinical EVLP. These results support examining this issue in a larger study.
Collapse
Affiliation(s)
- Anders S I Andreasson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Danai M Karamanou
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Colin S Gillespie
- School of Mathematics & Statistics, Newcastle University, Newcastle upon Tyne, UK
| | - Faruk Özalp
- Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Tanveer Butt
- Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Paul Hill
- Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Kasim Jiwa
- Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Hannah R Walden
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Nicola J Green
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Lee A Borthwick
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Stephen C Clark
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Henning Pauli
- Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Kate F Gould
- Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Paul A Corris
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Simi Ali
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - John H Dark
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Andrew J Fisher
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Cardiopulmonary Transplantation, Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
46
|
Morrison MI, Pither TL, Fisher AJ. Pathophysiology and classification of primary graft dysfunction after lung transplantation. J Thorac Dis 2017; 9:4084-4097. [PMID: 29268419 DOI: 10.21037/jtd.2017.09.09] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The term primary graft dysfunction (PGD) incorporates a continuum of disease severity from moderate to severe acute lung injury (ALI) within 72 h of lung transplantation. It represents the most significant obstacle to achieving good early post-transplant outcomes, but is also associated with increased incidence of bronchiolitis obliterans syndrome (BOS) subsequently. PGD is characterised histologically by diffuse alveolar damage, but is graded on clinical grounds with a combination of PaO2/FiO2 (P/F) and the presence of radiographic infiltrates, with 0 being absence of disease and 3 being severe PGD. The aetiology is multifactorial but commonly results from severe ischaemia-reperfusion injury (IRI), with tissue-resident macrophages largely responsible for stimulating a secondary 'wave' of neutrophils and lymphocytes that produce severe and widespread tissue damage. Donor history, recipient health and operative factors may all potentially contribute to the likelihood of PGD development. Work that aims to minimise the incidence of PGD in ongoing, with techniques such as ex vivo perfusion of donor lungs showing promise both in research and in clinical studies. This review will summarise the current clinical status of PGD before going on to discuss its pathophysiology, current therapies available and future directions for clinical management of PGD.
Collapse
Affiliation(s)
- Morvern Isabel Morrison
- Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne, UK.,Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Thomas Leonard Pither
- Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne, UK.,Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Andrew John Fisher
- Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne, UK.,Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
47
|
Bowser JL, Lee JW, Yuan X, Eltzschig HK. The hypoxia-adenosine link during inflammation. J Appl Physiol (1985) 2017; 123:1303-1320. [PMID: 28798196 DOI: 10.1152/japplphysiol.00101.2017] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 07/18/2017] [Accepted: 08/06/2017] [Indexed: 12/23/2022] Open
Abstract
Hypoxic tissue conditions occur during a number of inflammatory diseases and are associated with the breakdown of barriers and induction of proinflammatory responses. At the same time, hypoxia is also known to induce several adaptive and tissue-protective pathways that dampen inflammation and protect tissue integrity. Hypoxia-inducible factors (HIFs) that are stabilized during inflammatory or hypoxic conditions are at the center of mediating these responses. In the past decade, several genes regulating extracellular adenosine metabolism and signaling have been identified as being direct targets of HIFs. Here, we discuss the relationship between inflammation, hypoxia, and adenosine and that HIF-driven adenosine metabolism and signaling is essential in providing tissue protection during inflammatory conditions, including myocardial injury, inflammatory bowel disease, and acute lung injury. We also discuss how the hypoxia-adenosine link can be targeted therapeutically in patients as a future treatment approach for inflammatory diseases.
Collapse
Affiliation(s)
- Jessica L Bowser
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas
| | - Jae W Lee
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas
| | - Xiaoyi Yuan
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas
| | - Holger K Eltzschig
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas
| |
Collapse
|
48
|
Choi CYU, Reimers K, Allmeling C, Kall S, Choi YH, Vogt PM. Inhibition of Apoptosis by Expression of Antiapoptotic Proteins in Recombinant Human Keratinocytes. Cell Transplant 2017; 16:663-674. [DOI: 10.3727/000000007783465037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The Fas ligand/Fas interaction plays an important role in the regulation of immune responses. Allografted cells undergo Fas-mediated apoptosis induced by CD8+ T cells. Our objective was to prevent human keratinocytes from immunologically induced apoptosis. We focused on three proteins with inhibitory function on Fas-mediated apoptosis. Human keratinocytes were transfected with either Flip, Faim, or Lifeguard (LFG). The treatment proved to be practicable and efficient. The recombinant keratinocytes with expression of our target proteins were cocultured with CD8+ T cells and the apoptotic activity was then evaluated. Activation of caspase-8 was detectable in control but not in the recombinant cells. Quantitative analysis revealed significant induction of T-cell-induced apoptosis in nontransfected keratinocytes (p = 0.04, n = 12) but not in Flip (p = 0.66), Faim (p = 0.42), or LFG (p = 0.44) expressing cells. Our results suggest that heterotopic expression of antiapoptotic proteins can induce the resistance of keratinocytes to a major mechanism of rejection.
Collapse
Affiliation(s)
- Claudia Y. U. Choi
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover, D-30625 Hannover, Germany
| | - Kerstin Reimers
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover, D-30625 Hannover, Germany
| | - Christina Allmeling
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover, D-30625 Hannover, Germany
| | - Susanne Kall
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover, D-30625 Hannover, Germany
| | - Yeong-Hoon Choi
- Department of Cardiac Surgery, Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peter M. Vogt
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover, D-30625 Hannover, Germany
| |
Collapse
|
49
|
Eskandani M, Vandghanooni S, Barar J, Nazemiyeh H, Omidi Y. Cell physiology regulation by hypoxia inducible factor-1: Targeting oxygen-related nanomachineries of hypoxic cells. Int J Biol Macromol 2017; 99:46-62. [DOI: 10.1016/j.ijbiomac.2016.10.113] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/26/2016] [Indexed: 12/27/2022]
|
50
|
Andreasson ASI, Borthwick LA, Gillespie C, Jiwa K, Scott J, Henderson P, Mayes J, Romano R, Roman M, Ali S, Fildes JE, Marczin N, Dark JH, Fisher AJ. The role of interleukin-1β as a predictive biomarker and potential therapeutic target during clinical ex vivo lung perfusion. J Heart Lung Transplant 2017; 36:985-995. [PMID: 28551353 PMCID: PMC5578478 DOI: 10.1016/j.healun.2017.05.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/09/2017] [Accepted: 05/09/2017] [Indexed: 11/23/2022] Open
Abstract
Background Extended criteria donor lungs deemed unsuitable for immediate transplantation can be reconditioned using ex vivo lung perfusion (EVLP). Objective identification of which donor lungs can be successfully reconditioned and will function well post-operatively has not been established. This study assessed the predictive value of markers of inflammation and tissue injury in donor lungs undergoing EVLP as part of the DEVELOP-UK study. Methods Longitudinal samples of perfusate, bronchoalveolar lavage, and tissue from 42 human donor lungs undergoing clinical EVLP assessments were analyzed for markers of inflammation and tissue injury. Levels were compared according to EVLP success and post-transplant outcomes. Neutrophil adhesion to human pulmonary microvascular endothelial cells (HPMECs) conditioned with perfusates from EVLP assessments was investigated on a microfluidic platform. Results The most effective markers to differentiate between in-hospital survival and non-survival post-transplant were perfusate interleukin (IL)-1β (area under the curve = 1.00, p = 0.002) and tumor necrosis factor-α (area under the curve = 0.95, p = 0.006) after 30 minutes of EVLP. IL-1β levels in perfusate correlated with upregulation of intracellular adhesion molecule-1 in donor lung vasculature (R2 = 0.68, p < 0.001) and to a lesser degree upregulation of intracellular adhesion molecule-1 (R2 = 0.30, p = 0.001) and E-selectin (R2 = 0.29, p = 0.001) in conditioned HPMECs and neutrophil adhesion to conditioned HPMECs (R2 = 0.33, p < 0.001). Neutralization of IL-1β in perfusate effectively inhibited neutrophil adhesion to conditioned HPMECs (91% reduction, p = 0.002). Conclusions Donor lungs develop a detectable and discriminatory pro-inflammatory signature in perfusate during EVLP. Blocking the IL-1β pathway during EVLP may reduce endothelial activation and subsequent neutrophil adhesion on reperfusion; this requires further investigation in vivo.
Collapse
Affiliation(s)
- Anders S I Andreasson
- Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, United Kingdom; Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lee A Borthwick
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Colin Gillespie
- School of Mathematics & Statistics, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kasim Jiwa
- Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Jonathan Scott
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Paul Henderson
- Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Jonny Mayes
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | - Simi Ali
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - James E Fildes
- University Hospital of South Manchester, Manchester, United Kingdom; Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom
| | | | - John H Dark
- Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, United Kingdom; Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew J Fisher
- Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, United Kingdom; Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | | |
Collapse
|