1
|
Kang M, Park HK, Kim KS, Choi D. Animal models for transplant immunology: bridging bench to bedside. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:354-376. [PMID: 39233453 PMCID: PMC11732767 DOI: 10.4285/ctr.24.0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 09/06/2024]
Abstract
The progress of transplantation has been propelled forward by animal experiments. Animal models have not only provided opportunities to understand complex immune mechanisms in transplantation but also served as a platform to assess therapeutic interventions. While small animals have been instrumental in uncovering new therapeutic concepts related to immunosuppression and immune tolerance, the progression to human trials has largely been driven by studies in large animals. Recent research has begun to explore the potential of porcine organs to address the shortage of available organs. The consistent progress in transplant immunology research can be attributed to a thorough understanding of animal models. This review provides a comprehensive overview of the available animal models, detailing their modifications, strengths, and weaknesses, as well as their historical applications, to aid researchers in selecting the most suitable model for their specific research needs.
Collapse
Affiliation(s)
- Minseok Kang
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Hwon Kyum Park
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Kyeong Sik Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Korea
| |
Collapse
|
2
|
Sun R, Wang N, Zheng S, Wang H, Xie H. Nanotechnology-based Strategies for Molecular Imaging, Diagnosis, and Therapy of Organ Transplantation. Transplantation 2024; 108:1730-1748. [PMID: 39042368 DOI: 10.1097/tp.0000000000004913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Organ transplantation is the preferred paradigm for patients with end-stage organ failures. Despite unprecedented successes, complications such as immune rejection, ischemia-reperfusion injury, and graft dysfunction remain significant barriers to long-term recipient survival after transplantation. Conventional immunosuppressive drugs have limited efficacy because of significant drug toxicities, high systemic immune burden, and emergence of transplant infectious disease, leading to poor quality of life for patients. Nanoparticle-based drug delivery has emerged as a promising medical technology and offers several advantages by enhancing the delivery of drug payloads to their target sites, reducing systemic toxicity, and facilitating patient compliance over free drug administration. In addition, nanotechnology-based imaging approaches provide exciting diagnostic methods for monitoring molecular and cellular changes in transplanted organs, visualizing immune responses, and assessing the severity of rejection. These noninvasive technologies are expected to help enhance the posttransplantation patient survival through real time and early diagnosis of disease progression. Here, we present a comprehensive review of nanotechnology-assisted strategies in various aspects of organ transplantation, including organ protection before transplantation, mitigation of ischemia-reperfusion injury, counteraction of immune rejection, early detection of organ dysfunction posttransplantation, and molecular imaging and diagnosis of immune rejection.
Collapse
Affiliation(s)
- Ruiqi Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Province, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Zhejiang Province, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang Province, Hangzhou, China
| | - Ning Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Province, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Zhejiang Province, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang Province, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Province, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Zhejiang Province, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang Province, Hangzhou, China
| | - Hangxiang Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Province, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Zhejiang Province, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang Province, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Province, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Zhejiang Province, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang Province, Hangzhou, China
| |
Collapse
|
3
|
Belousova N, Cheng A, Matelski J, Vasileva A, Wu JKY, Ghany R, Martinu T, Ryan CM, Chow CW. Effects of donor smoking history on early post-transplant lung function measured by oscillometry. Front Med (Lausanne) 2024; 11:1328395. [PMID: 38654829 PMCID: PMC11037252 DOI: 10.3389/fmed.2024.1328395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Prior studies assessing outcomes of lung transplants from cigarette-smoking donors found mixed results. Oscillometry, a non-invasive test of respiratory impedance, detects changes in lung function of smokers prior to diagnosis of COPD, and identifies spirometrically silent episodes of rejection post-transplant. We hypothesise that oscillometry could identify abnormalities in recipients of smoking donor lungs and discriminate from non-smoking donors. Methods This prospective single-center cohort study analysed 233 double-lung recipients. Oscillometry was performed alongside routine conventional pulmonary function tests (PFT) post-transplant. Multivariable regression models were constructed to compare oscillometry and conventional PFT parameters between recipients of lungs from smoking vs non-smoking donors. Results The analysis included 109 patients who received lungs from non-smokers and 124 from smokers. Multivariable analysis identified significant differences between recipients of smoking and non-smoking lungs in the oscillometric measurements R5-19, X5, AX, R5z and X5z, but no differences in %predicted FEV1, FEV1/FVC, %predicted TLC or %predicted DLCO. An analysis of the smoking group also demonstrated associations between increasing smoke exposure, quantified in pack years, and all the oscillometry parameters, but not the conventional PFT parameters. Conclusion An interaction was identified between donor-recipient sex match and the effect of smoking. The association between donor smoking and oscillometry outcomes was significant predominantly in the female donor/female recipient group.
Collapse
Affiliation(s)
- Natalia Belousova
- Toronto Lung Transplant Program, Ajmera Multi-Organ Transplant Program and Division of Respirology, University Health Network, Tonronto, ON, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Pneumology, Aduch Cystic Fibrosis and Lung Transplantation Department, Foch Hospital, Suresnes, France
| | - Albert Cheng
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - John Matelski
- Pneumology, Aduch Cystic Fibrosis and Lung Transplantation Department, Foch Hospital, Suresnes, France
| | - Anastasiia Vasileva
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Joyce K. Y. Wu
- Toronto General Pulmonary Function Laboratory, University Health Network, Toronto, ON, Canada
| | - Rasheed Ghany
- Toronto Lung Transplant Program, Ajmera Multi-Organ Transplant Program and Division of Respirology, University Health Network, Tonronto, ON, Canada
| | - Tereza Martinu
- Toronto Lung Transplant Program, Ajmera Multi-Organ Transplant Program and Division of Respirology, University Health Network, Tonronto, ON, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Clodagh M. Ryan
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Pulmonary Function Laboratory, University Health Network, Toronto, ON, Canada
| | - Chung-Wai Chow
- Toronto Lung Transplant Program, Ajmera Multi-Organ Transplant Program and Division of Respirology, University Health Network, Tonronto, ON, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Kerckhof P, Ambrocio GPL, Beeckmans H, Kaes J, Geudens V, Bos S, Willems L, Vermaut A, Vermant M, Goos T, De Fays C, Aversa L, Mohamady Y, Vanstapel A, Orlitová M, Van Slambrouck J, Jin X, Varghese V, Josipovic I, Boone MN, Dupont LJ, Weynand B, Dubbeldam A, Van Raemdonck DE, Ceulemans LJ, Gayan-Ramirez G, De Sadeleer LJ, McDonough JE, Vanaudenaerde BM, Vos R. Ventilatory capacity in CLAD is driven by dysfunctional airway structure. EBioMedicine 2024; 101:105030. [PMID: 38394744 PMCID: PMC10897920 DOI: 10.1016/j.ebiom.2024.105030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) encompasses three main phenotypes: bronchiolitis obliterans syndrome (BOS), restrictive allograft syndrome (RAS) and a Mixed phenotype combining both pathologies. How the airway structure in its entirety is affected in these phenotypes is still poorly understood. METHODS A detailed analysis of airway morphometry was applied to gain insights on the effects of airway remodelling on the distribution of alveolar ventilation in end-stage CLAD. Ex vivo whole lung μCT and tissue-core μCT scanning of six control, six BOS, three RAS and three Mixed explant lung grafts (9 male, 9 female, 2014-2021, Leuven, Belgium) were used for digital airway reconstruction and calculation of airway dimensions in relation to luminal obstructions. FINDINGS BOS and Mixed explants demonstrated airway obstructions of proximal bronchioles (starting at generation five), while RAS explants particularly had airway obstructions in the most distal bronchioles (generation >12). In BOS and Mixed explants 76% and 84% of bronchioles were obstructed, respectively, while this was 22% in RAS. Bronchiolar obstructions were mainly caused by lymphocytic inflammation of the airway wall or fibrotic remodelling, i.e. constrictive bronchiolitis. Proximal bronchiolectasis and imbalance in distal lung ventilation were present in all CLAD phenotypes and explain poor lung function and deterioration of specific lung function parameters. INTERPRETATION Alterations in the structure of conducting bronchioles revealed CLAD to affect alveolar ventilatory distribution in a regional fashion. The significance of various obstructions, particularly those associated with mucus, is highlighted. FUNDING This research was funded with the National research fund Flanders (G060322N), received by R.V.
Collapse
Affiliation(s)
- Pieterjan Kerckhof
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Gene P L Ambrocio
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Division of Pulmonary Medicine, Department of Internal Medicine, University of the Philippines - Philippine General Hospital, Manilla, The Philippines
| | - Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Vincent Geudens
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Saskia Bos
- Newcastle University, Translational and Clinical Research Institute, Newcastle upon Tyne, United Kingdom
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Astrid Vermaut
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Tinne Goos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Charlotte De Fays
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Lucia Aversa
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Yousry Mohamady
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | | | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Vimi Varghese
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Heart and Lung Transplant, Yashoda Hospitals, Hyderabad, India
| | - Iván Josipovic
- Department of Physics and Astronomy, UGCT, Radiation Physics, Ghent University, Gent, Belgium
| | - Matthieu N Boone
- Department of Physics and Astronomy, UGCT, Radiation Physics, Ghent University, Gent, Belgium
| | - Lieven J Dupont
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Adriana Dubbeldam
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | | | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Laurens J De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Cell Circuits in Systems Medicine of Lung Disease (Schiller Lab), Institute of Lung Health and Immunity (LHI) / Comprehensive Pneumology Centre (CPC), German Centre for Lung Research, Helmholtz Zentrum München, München, Germany
| | - John E McDonough
- Department of Medicine, McMaster University, Firestone Institute of Respiratory Health, Hamilton, Canada
| | - Bart M Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
5
|
Khatri A, Todd JL, Kelly FL, Nagler A, Ji Z, Jain V, Gregory SG, Weinhold KJ, Palmer SM. JAK-STAT activation contributes to cytotoxic T cell-mediated basal cell death in human chronic lung allograft dysfunction. JCI Insight 2023; 8:167082. [PMID: 36946463 PMCID: PMC10070100 DOI: 10.1172/jci.insight.167082] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/01/2023] [Indexed: 03/23/2023] Open
Abstract
Chronic lung allograft dysfunction (CLAD) is the leading cause of death in lung transplant recipients. CLAD is characterized clinically by a persistent decline in pulmonary function and histologically by the development of airway-centered fibrosis known as bronchiolitis obliterans. There are no approved therapies to treat CLAD, and the mechanisms underlying its development remain poorly understood. We performed single-cell RNA-Seq and spatial transcriptomic analysis of explanted tissues from human lung recipients with CLAD, and we performed independent validation studies to identify an important role of Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling in airway epithelial cells that contributes to airway-specific alloimmune injury. Specifically, we established that activation of JAK-STAT signaling leads to upregulation of major histocompatibility complex 1 (MHC-I) in airway basal cells, an important airway epithelial progenitor population, which leads to cytotoxic T cell-mediated basal cell death. This study provides mechanistic insight into the cell-to-cell interactions driving airway-centric alloimmune injury in CLAD, suggesting a potentially novel therapeutic strategy for CLAD prevention or treatment.
Collapse
Affiliation(s)
- Aaditya Khatri
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Jamie L Todd
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Fran L Kelly
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Andrew Nagler
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Zhicheng Ji
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| | - Kent J Weinhold
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Scott M Palmer
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
6
|
Aoki Y, Walker NM, Misumi K, Mimura T, Vittal R, McLinden AP, Fitzgerald L, Combs MP, Lyu D, Osterholzer JJ, Pinsky DJ, Lama VN. The mitigating effect of exogenous carbon monoxide on chronic allograft rejection and fibrosis post-lung transplantation. J Heart Lung Transplant 2023; 42:317-326. [PMID: 36522238 DOI: 10.1016/j.healun.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 10/22/2022] [Accepted: 11/15/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Small airway inflammation and fibrosis or bronchiolitis obliterans (BO) is the predominant presentation of chronic lung allograft dysfunction (CLAD) post-lung transplantation. Carbon monoxide (CO) is a critical endogenous signaling transducer with known anti-inflammatory and anti-fibrotic effects but its therapeutic potential in CLAD remains to be fully elucidated. METHODS Here we investigate the effect of inhaled CO in modulating chronic lung allograft rejection pathology in a murine orthotopic lung transplant model of BO (B6D2F1/J→DBA/2J). Additionally, the effects of CO on the activated phenotype of mesenchymal cells isolated from human lung transplant recipients with CLAD were studied. RESULTS Murine lung allografts treated with CO (250 ppm × 30 minutes twice daily from days 7 to 40 post-transplantation) demonstrated decreased immune cell infiltration, fibrosis, and airway obliteration by flow cytometry, trichrome staining, and morphometric analysis, respectively. Decreased total collagen, with levels comparable to isografts, was noted in CO-treated allografts by quantitative hydroxyproline assay. In vitro, CO (250 ppm × 16h) was effective in reversing the fibrotic phenotype of human CLAD mesenchymal cells with decreased collagen I and β-catenin expression as well as an inhibitory effect on ERK1/2 MAPK, and mTORC1/2 signaling. Sildenafil, a phosphodiesterase 5 inhibitor, partially mimicked the effects of CO on CLAD mesenchymal cells and was partially effective in decreasing collagen deposition in murine allografts, suggesting the contribution of cGMP-dependent and -independent mechanisms in mediating the effect of CO. CONCLUSION These results suggest a potential role for CO in alleviating allograft fibrosis and mitigating chronic rejection pathology post-lung transplant.
Collapse
Affiliation(s)
- Yoshiro Aoki
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Natalie M Walker
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Keizo Misumi
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Takeshi Mimura
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Ragini Vittal
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Aidan P McLinden
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Linda Fitzgerald
- Department of Pharmacy Services, University of Michigan Health System, Ann Arbor, Michigan
| | - Michael P Combs
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Dennis Lyu
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - John J Osterholzer
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan; Pulmonary Section, VA Ann Arbor Health System, Ann Arbor, Michigan
| | - David J Pinsky
- Cardiology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Vibha N Lama
- Divisions of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan.
| |
Collapse
|
7
|
Fraebel J, Engelhardt BG, Kim TK. Noninfectious Pulmonary Complications after Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2023; 29:82-93. [PMID: 36427785 DOI: 10.1016/j.jtct.2022.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/31/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
Pulmonary complications after hematopoietic stem cell transplantation (HSCT) are important sources of morbidity and mortality. Improvements in infection-related complications have made noninfectious pulmonary complications an increasingly significant driver of transplantation-related mortality. Broadly, these complications can be characterized as either early or late complications, with idiopathic pneumonia syndrome and bronchiolitis obliterans syndrome the most prevalent early and late complications, respectively. Outcomes with historical treatment consisting mainly of corticosteroids are often poor, highlighting the need for a deeper understanding of these complications' underlying disease biology to guide the adoption of novel therapies that are being increasingly used in the modern era.
Collapse
Affiliation(s)
- Johnathan Fraebel
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brian G Engelhardt
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt-Ingram Cancer Center, Nashville, Tennessee; Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Immunobiology, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt-Ingram Cancer Center, Nashville, Tennessee; Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee.
| |
Collapse
|
8
|
Lynch TJ, Ahlers BA, Swatek AM, Ievlev V, Pai AC, Brooks L, Tang Y, Evans IA, Meyerholz DK, Engelhardt JF, Parekh KR. Ferret Lung Transplantation Models Differential Lymphoid Aggregate Morphology Between Restrictive and Obstructive Forms of Chronic Lung Allograft Dysfunction. Transplantation 2022; 106:1974-1989. [PMID: 35442232 PMCID: PMC9529760 DOI: 10.1097/tp.0000000000004148] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Long-term survival after lung transplantation remains limited by chronic lung allograft dysfunction (CLAD). CLAD has 2 histologic phenotypes, namely obliterative bronchiolitis (OB) and restrictive alveolar fibroelastosis (AFE), which have distinct clinical presentations, pathologies, and outcomes. Understanding of OB versus AFE pathogenesis would improve with better animal models. METHODS We utilized a ferret orthotopic single-lung transplantation model to characterize allograft fibrosis as a histologic measure of CLAD. Native lobes and "No CLAD" allografts lacking aberrant histology were used as controls. We used morphometric analysis to evaluate the size and abundance of B-cell aggregates and tertiary lymphoid organs (TLOs) and their cell composition. Quantitative RNA expression of 47 target genes was performed simultaneously using a custom QuantiGene Plex Assay. RESULTS Ferret lung allografts develop the full spectrum of human CLAD histology including OB and AFE subtypes. While both OB and AFE allografts developed TLOs, TLO size and number were greater with AFE histology. More activated germinal center cells marked by B-cell lymphoma 6 Transcription Repressor, (B-cell lymphoma 6) expression and fewer cells expressing forkhead box P3 correlated with AFE, congruent with greater diffuse immunoglobulin, plasma cell abundance, and complement 4d staining. Furthermore, forkhead box P3 RNA induction was significant in OB allografts specifically. RNA expression changes were seen in native lobes of animals with AFE but not OB when compared with No CLAD native lobes. CONCLUSIONS The orthotopic ferret single-lung transplant model provides unique opportunities to better understand factors that dispose allografts to OB versus AFE. This will help develop potential immunomodulatory therapies and antifibrotic approaches for lung transplant patients.
Collapse
Affiliation(s)
- Thomas J. Lynch
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Bethany A. Ahlers
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Anthony M. Swatek
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Vitaly Ievlev
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Albert C. Pai
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Leonard Brooks
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Yinghua Tang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Idil A. Evans
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - David K. Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Kalpaj R. Parekh
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
9
|
Bos S, Milross L, Filby AJ, Vos R, Fisher AJ. Immune processes in the pathogenesis of chronic lung allograft dysfunction: identifying the missing pieces of the puzzle. Eur Respir Rev 2022; 31:31/165/220060. [PMID: 35896274 DOI: 10.1183/16000617.0060-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/19/2022] [Indexed: 11/05/2022] Open
Abstract
Lung transplantation is the optimal treatment for selected patients with end-stage chronic lung diseases. However, chronic lung allograft dysfunction remains the leading obstacle to improved long-term outcomes. Traditionally, lung allograft rejection has been considered primarily as a manifestation of cellular immune responses. However, in reality, an array of complex, interacting and multifactorial mechanisms contribute to its emergence. Alloimmune-dependent mechanisms, including T-cell-mediated rejection and antibody-mediated rejection, as well as non-alloimmune injuries, have been implicated. Moreover, a role has emerged for autoimmune responses to lung self-antigens in the development of chronic graft injury. The aim of this review is to summarise the immune processes involved in the pathogenesis of chronic lung allograft dysfunction, with advanced insights into the role of innate immune pathways and crosstalk between innate and adaptive immunity, and to identify gaps in current knowledge.
Collapse
Affiliation(s)
- Saskia Bos
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK.,Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Luke Milross
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Andrew J Filby
- Flow Cytometry Core and Innovation, Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Robin Vos
- Dept of CHROMETA, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.,University Hospitals Leuven, Dept of Respiratory Diseases, Leuven, Belgium
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK .,Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| |
Collapse
|
10
|
Mauer J, Kuckhahn A, Ramsperger-Gleixner M, Ensminger SM, Distler JH, Weyand M, Heim C. Nintedanib reduces alloimmune-induced chronic airway changes in murine tracheal allografts. Transpl Immunol 2022; 73:101608. [DOI: 10.1016/j.trim.2022.101608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/12/2022] [Accepted: 04/23/2022] [Indexed: 01/01/2023]
|
11
|
Varma R, Marin‐Araujo AE, Rostami S, Waddell TK, Karoubi G, Haykal S. Short-Term Preclinical Application of Functional Human Induced Pluripotent Stem Cell-Derived Airway Epithelial Patches. Adv Healthc Mater 2021; 10:e2100957. [PMID: 34569180 DOI: 10.1002/adhm.202100957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/15/2021] [Indexed: 12/17/2022]
Abstract
Airway pathologies including cancer, trauma, and stenosis lack effective treatments, meanwhile airway transplantation and available tissue engineering approaches fail due to epithelial dysfunction. Autologous progenitors do not meet the clinical need for regeneration due to their insufficient expansion and differentiation, for which human induced pluripotent stem cells (hiPSCs) are promising alternatives. Airway epithelial patches are engineered by differentiating hiPSC-derived airway progenitors into physiological proportions of ciliated (73.9 ± 5.5%) and goblet (2.1 ± 1.4%) cells on a silk fibroin-collagen vitrigel membrane (SF-CVM) composite biomaterial for transplantation in porcine tracheal defects ex vivo and in vivo. Evaluation of ex vivo tracheal repair using hiPSC-derived SF-CVM patches demonstrate native-like tracheal epithelial metabolism and maintenance of mucociliary epithelium to day 3. In vivo studies demonstrate SF-CVM integration and maintenance of airway patency, showing 80.8 ± 3.6% graft coverage with an hiPSC-derived pseudostratified epithelium and 70.7 ± 2.3% coverage with viable cells, 3 days postoperatively. The utility of bioengineered, hiPSC-derived epithelial patches for airway repair is demonstrated in a short-term preclinical survival model, providing a significant leap for airway reconstruction approaches.
Collapse
Affiliation(s)
- Ratna Varma
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
- Institute of Biomedical Engineering (BME) University of Toronto 164 College St Toronto ON M5S 3G9 Canada
| | - Alba E. Marin‐Araujo
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
| | - Sara Rostami
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
| | - Thomas K. Waddell
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
- Institute of Biomedical Engineering (BME) University of Toronto 164 College St Toronto ON M5S 3G9 Canada
- Institute of Medical Sciences University of Toronto 27 King's College Cir Toronto ON M5S 1A8 Canada
| | - Golnaz Karoubi
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
- Department of Mechanical and Industrial Engineering University of Toronto 5 King's College Circle Toronto ON M5S 3G8 Canada
- Department of Laboratory Medicine and Pathobiology University of Toronto 1 King's College Circle Toronto ON M5S 1A8 Canada
| | - Siba Haykal
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
- Institute of Medical Sciences University of Toronto 27 King's College Cir Toronto ON M5S 1A8 Canada
- Division of Plastic and Reconstructive Surgery Department of Surgery University of Toronto 200 Elizabeth Street 8N‐869 Toronto ON M5G2P7 Canada
| |
Collapse
|
12
|
Nemska S, Daubeuf F, Obrecht A, Israel-Biet D, Stern M, Kessler R, Roux A, Tavakoli R, Villa P, Tissot A, Danger R, Reber L, Durand E, Foureau A, Brouard S, Magnan A, Frossard N. Overexpression of the MSK1 Kinase in Patients With Chronic Lung Allograft Dysfunction and Its Confirmed Role in a Murine Model. Transplantation 2021; 105:1212-1224. [PMID: 33560725 DOI: 10.1097/tp.0000000000003606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) and its obstructive form, the obliterative bronchiolitis (OB), are the main long-term complications related to high mortality rate postlung transplantation. CLAD treatment lacks a significant success in survival. Here, we investigated a new strategy through inhibition of the proinflammatory mitogen- and stress-activated kinase 1 (MSK1) kinase. METHODS MSK1 expression was assessed in a mouse OB model after heterotopic tracheal allotransplantation. Pharmacological inhibition of MSK1 (H89, fasudil, PHA767491) was evaluated in the murine model and in a translational model using human lung primary fibroblasts in proinflammatory conditions. MSK1 expression was graded over time in biopsies from a cohort of CLAD patients. RESULTS MSK1 mRNA progressively increased during OB (6.4-fold at D21 posttransplantation). Inhibition of MSK1 allowed to counteract the damage to the epithelium (56% restoration for H89), and abolished the recruitment of MHCII+ (94%) and T cells (100%) at the early inflammatory phase of OB. In addition, it markedly decreased the late fibroproliferative obstruction in allografts (48%). MSK1 inhibitors decreased production of IL-6 (whose transcription is under the control of MSK1) released from human lung fibroblasts (96%). Finally, we confirmed occurrence of a 2.9-fold increased MSK1 mRNA expression in lung biopsies in patients at 6 months before CLAD diagnosis as compared to recipients with stable lung function. CONCLUSIONS These findings suggest the overall interest of the MSK1 kinase either as a marker or as a potential therapeutic target in lung dysfunction posttransplantation.
Collapse
Affiliation(s)
- Simona Nemska
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - François Daubeuf
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
- Plateforme de Chimie Biologie Intégrative de Strasbourg (PCBIS) UMS 3286 CNRS, Université de Strasbourg, Labex Medalis, 300 Bld Brant, Illkirch, France
| | - Adeline Obrecht
- Plateforme de Chimie Biologie Intégrative de Strasbourg (PCBIS) UMS 3286 CNRS, Université de Strasbourg, Labex Medalis, 300 Bld Brant, Illkirch, France
| | | | - Marc Stern
- Hôpital Foch, Suresnes, INRAe UMR 0892, Université de Versailles Saint-Quentin Paris-Saclay, Paris, France
| | - Romain Kessler
- Service de Pneumologie, CHU Strasbourg, Strasbourg, France
| | - Antoine Roux
- Hôpital Foch, Suresnes, INRAe UMR 0892, Université de Versailles Saint-Quentin Paris-Saclay, Paris, France
| | - Reza Tavakoli
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Pascal Villa
- Plateforme de Chimie Biologie Intégrative de Strasbourg (PCBIS) UMS 3286 CNRS, Université de Strasbourg, Labex Medalis, 300 Bld Brant, Illkirch, France
| | - Adrien Tissot
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
- Service de Pneumologie, L'institut du thorax, CHU Nantes, Nantes, France
| | - Richard Danger
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
- Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), Labex IGO, Nantes, France
| | - Laurent Reber
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Eugénie Durand
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
| | - Aurore Foureau
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
- Service de Pneumologie, L'institut du thorax, CHU Nantes, Nantes, France
| | - Sophie Brouard
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
- Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), Labex IGO, Nantes, France
| | - Antoine Magnan
- Service de Pneumologie, L'institut du thorax, CHU Nantes, Nantes, France
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| |
Collapse
|
13
|
IL-17A Contributes to Lung Fibrosis in a Model of Chronic Pulmonary Graft-versus-host Disease. Transplantation 2020; 103:2264-2274. [PMID: 31658231 DOI: 10.1097/tp.0000000000002837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chronic pulmonary graft-versus-host disease (cpGVHD) after hematopoietic cell transplant (HCT) manifests as progressive airway and parenchymal lung fibrosis. On the basis of our prior data, mice that undergo allogeneic HCT with Tbet-knockout donors (AlloTbet) have increased lung Th17 cells and IL-17A and develop fibrosis resembling human cpGVHD. The role of IL-17A in posttransplant pulmonary fibrosis remains incompletely understood. We hypothesized that IL-17A is necessary for development of murine cpGVHD in this model. METHODS AlloTbet mice received weekly intraperitoneal anti-IL-17A or IgG (200 μg/mouse) starting 2 weeks post-HCT and were sacrificed after week 5. Histologic airway and parenchymal fibrosis were semiquantitatively graded in a blinded fashion. Lung cells and proteins were measured by flow cytometry, ELISA, and multicytokine assays. RESULTS Anti-IL-17A modestly decreased airway and parenchymal lung fibrosis, along with a striking reduction in pulmonary neutrophilia, IL-6, MIP-1α, MIP-1β, CXCL1, and CXCL5 in AlloTbet mice. Additionally, anti-IL-17A decreased CCL2, inflammatory monocytes and macrophages, and Th17 cells. CONCLUSIONS In the setting of murine AlloHCT with Tbet donors, IL-17A blockade decreases fibrotic features of cpGVHD. This may be mediated by the observed reduction in neutrophils or specific lung monocyte and macrophage populations or alternatively via a direct effect on fibroblasts. Collectively, our results further suggest that anti-IL-17A strategies could prove useful in preventing alloimmune-driven fibrotic lung diseases.
Collapse
|
14
|
Gubareva EA, Kuevda EV, Basov AA, Sotnichenko AS, Bolotin SN, Dzhimak SS. Biophysical methods for quality evaluation of decellularized and recellularized tissue-engineered constructs of organs and tissues. J Biosci 2019; 44:140. [PMID: 31894121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Tissue engineering is rapidly growing now and can become a promising alternative to transplantation of organs and tissues, as it is devoid of major shortcomings of transplantology, such as acute shortage, complexity of selection, delivery and storage of donor material, lifelong immunosuppressive therapy. One of the most widely known methods of obtaining biological scaffolds for the subsequent creation of tissue-engineered constructs of organs and tissues is decellularization. The evaluation of the quality of the obtained scaffolds, based on the study of the viability of cell structures in decellularized and recellularized matrices, is one of the priorities of modern regenerative medicine worldwide. In this investigation, the biophysical criteria of decellularization and recellularization of tissue-engineered constructs based on the evaluation of the generation of free radicals in native, decellularized and recellularized tissues by EPR spectroscopy and chemoluminescence in a complex assessment of the quality of biological matrixes obtained are considered using intrathoracic organs and tissues of rats. It has been established that the intensity indices of free radical generation in native and recellularized tissues of animal organs, as well as in decellularized matrices, can serve as one of the express criteria for quantitative assessment of cell structures viability.
Collapse
|
15
|
Gubareva EA, Kuevda EV, Basov AA, Sotnichenko AS, Bolotin SN, Dzhimak SS. Biophysical methods for quality evaluation of decellularized and recellularized tissue-engineered constructs of organs and tissues. J Biosci 2019. [DOI: 10.1007/s12038-019-9953-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|