ArticlePublisher preview available

Cellular recovery after prolonged warm ischaemia of the whole body

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

After cessation of blood flow or similar ischaemic exposures, deleterious molecular cascades commence in mammalian cells, eventually leading to their death1,2. Yet with targeted interventions, these processes can be mitigated or reversed, even minutes or hours post mortem, as also reported in the isolated porcine brain using BrainEx technology3. To date, translating single-organ interventions to intact, whole-body applications remains hampered by circulatory and multisystem physiological challenges. Here we describe OrganEx, an adaptation of the BrainEx extracorporeal pulsatile-perfusion system and cytoprotective perfusate for porcine whole-body settings. After 1 h of warm ischaemia, OrganEx application preserved tissue integrity, decreased cell death and restored selected molecular and cellular processes across multiple vital organs. Commensurately, single-nucleus transcriptomic analysis revealed organ- and cell-type-specific gene expression patterns that are reflective of specific molecular and cellular repair processes. Our analysis comprises a comprehensive resource of cell-type-specific changes during defined ischaemic intervals and perfusion interventions spanning multiple organs, and it reveals an underappreciated potential for cellular recovery after prolonged whole-body warm ischaemia in a large mammal. OrganEx—an extracorporeal pulsatile-perfusion system with cytoprotective perfusate for porcine whole-body settings—preserved tissue integrity, decreased cell death and restored selected molecular and cellular processes across multiple vital organs after 1 h of warm ischaemia in pigs.
Functional characterization and metabolic activity of selected organs a–c, Measurement of 2-NBDG uptake in the brain (a), heart (b) and kidney (c). n = 3. d, Observed QRS complexes in ECMO- and OrganEx-treated animals 3 h into the perfusion period (left). Statistical analysis was performed using χ² tests. Right, representative EKG trances in the OrganEx and ECMO groups 3 h into the perfusion period. e, Measurement of cardiomyocyte contraction velocity in acute heart slices in the 0 h WIT, OrganEx and ECMO groups (left), and cardiomyocyte contraction duration (right). n = 5. f, Representative confocal images of immunolabelling for albumin in the liver. Scale bars, 50 μm. g, Measurement of normalized immunolabelling signal intensity of albumin in the liver demonstrating similar expression between the OrganEx and 0 h WIT groups. n = 3. h, Representative images of organotypic hippocampal slices after 1 and 14 days in culture. Scale bar, 500 μm. i, Quantification of hippocampal slice integrity. n = 4–5. j, Representative confocal images of newly synthesized proteins (AHA, Click-iT Chemistry) with DAPI counterstaining in the long-term organotypic hippocampal slice culture. Scale bar, 100 μm. k, Quantification of AHA relative intensity in the CA1. n = 3–5. For a–c, g, i and k, data are mean ± s.e.m. Statistical analysis was performed using one-way ANOVA with post hoc Dunnett adjustment; *P < 0.05, **P < 0.01, ***P < 0.001. AU, arbitrary units. HIP, hippocampus.
… 
This content is subject to copyright. Terms and conditions apply.
Nature | Vol 608 | 11 August 2022 | 405
Article
Cellular recovery after prolonged warm
ischaemia of the whole body
David Andrijevic1,18, Zvonimir Vrselja1,18, Taras Lysyy1,2,18, Shupei Zhang1,3,18, Mario Skarica1,
Ana Spajic1, David Dellal1,4, Stephanie L. Thorn5, Robert B. Duckrow6, Shaojie Ma1,
Phan Q. Duy1,7,8 , Atagun U. Isiktas1, Dan Liang1, Mingfeng Li1, Suel-Kee Kim1,
Stefano G. Daniele1,8, Khadija Banu9, Sudhir Perincheri10, Madhav C. Menon9, Anita Huttner10,
Kevin N. Sheth6,7, Kevin T. Gobeske6, Gregory T. Tietjen2,4, Hitten P. Zaveri6,
Stephen R. Latham11, Albert J. Sinusas3,4,12,13 & Nenad Sestan1,3,14,15,1 6,17 ✉
After cessation of blood ow or similar ischaemic exposures, deleterious molecular
cascades commence in mammalian cells, eventually leading to their death1,2. Yet with
targeted interventions, these processes can be mitigated or reversed, even minutes or
hours post mortem, as also reported in the isolated porcine brain using BrainEx
technology3. To date, translating single-organ interventions to intact, whole-body
applications remains hampered by circulatory and multisystem physiological
challenges. Here we describe OrganEx, an adaptation of the BrainEx extracorporeal
pulsatile-perfusion system and cytoprotective perfusate for porcine whole-body
settings. After 1 h of warm ischaemia, OrganEx application preserved tissue integrity,
decreased cell death and restored selected molecular and cellular processes across
multiple vital organs. Commensurately, single-nucleus transcriptomic analysis
revealed organ- and cell-type-specic gene expression patterns that are reective of
specic molecular and cellular repair processes. Our analysis comprises a
comprehensive resource of cell-type-specic changes during dened ischaemic
intervals and perfusion interventions spanning multiple organs, and it reveals an
underappreciated potential for cellular recovery after prolonged whole-body warm
ischaemia in a large mammal.
Mammalian cells require oxygen to maintain cellular and tissue viability4.
In just minutes after ischaemia, intracellular acidosis and oedema
develop and trigger secondary injury to membranes and organelles,
often causing cell death1. At the whole-body scale, there is a systemic
release of hormones and cytokines, followed by activation of autonomic
nervous, immune and coagulation systems, leading to end-organ injury
culminating in systemic metabolic acidosis and hyperkalaemia2,5,6.
However, recent studies question the inevitability of cell death even
after hours of circulatory interruption. Viable cells can be collected
from multiple organs and maintained using invitro culture after
prolonged ischaemia
7
. Similarly, cellular recovery can be promoted
using ex vivo perfusion of isolated whole organs, including heart, liver,
kidneys and lungs
8–11
. Our perfusion-based BrainEx technology also
demonstrated restored circulation and cellular activity hours post
mortem in isolated porcine brains—the organ that is most vulnerable
to ischaemia2,6.
Nevertheless, translating solutions from isolated-organ models
for molecular and cellular recovery to whole-body applications after
extended ischaemia still presents substantial challenges. Reperfu-
sion of the whole body with autologous blood has several hindrances,
including coagulation, microvascular dysfunction, inflammation and
blood-intrinsic cellular dysfunction
2,5
. This has restricted whole-body
reperfusion and recovery times to 20 min in large mammals12, although
catastrophic/fatal consequences are widespread after several minutes
in humans
13
. A new approach may reinstate systemic circulation while
adding targeted molecular and cellular recovery strategies for specific
organs in the whole-body setting. From this, achieving recovery after
1 h of warm ischaemia may facilitate the development of opportunities
across various clinical disciplines.
Towards these goals, we translated principles from BrainEx technol-
ogy3 to develop OrganEx, a perfusion system and synthetic, acellular,
cytoprotective perfusate, for whole-body use in large mammals. We
https://doi.org/10.1038/s41586-022-05016-1
Received: 9 September 2021
Accepted: 23 June 2022
Published online: 3 August 2022
Check for updates
1Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA. 2Department of Surgery, Yale School of Medicine New Haven, New Haven, CT, USA. 3Department of Genetics, Yale
School of Medicine, New Haven, CT, USA. 4Department of Biomedical Engineering, Yale University, New Haven, CT, USA. 5Yale Translational Research Imaging Center, Department of Medicine,
Yale School of Medicine, New Haven, CT, USA. 6Department of Neurology, Yale University School of Medicine, New Haven, CT, USA. 7Department of Neurosurgery, Yale School of Medicine, New
Haven, CT, USA. 8Medical Scientist Training Program (MD-PhD), Yale School of Medicine, New Haven, CT, USA. 9Department of Nephrology, Yale School of Medicine, New Haven, CT, USA.
10Department of Pathology, Yale School of Medicine, New Haven, CT, USA. 11Interdisciplinary Center for Bioethics, Yale University, New Haven, CT, USA. 12Vascular Biology and Therapeutics
Program, Yale School of Medicine, New Haven, CT, USA. 13Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA. 14Department of Psychiatry, Yale
School of Medicine, New Haven, CT, USA. 15Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA. 16Program in Cellular Neuroscience, Neurodegeneration and
Repair, Yale School of Medicine, New Haven, CT, USA. 17Yale Child Study Center, New Haven, CT, USA. 18These authors contributed equally: David Andrijevic, Zvonimir Vrselja, Taras Lysyy,
Shupei Zhang. e-mail: nenad.sestan@yale.edu
Content courtesy of Springer Nature, terms of use apply. Rights reserved
... Shortly after death, intracellular acidosis and oedema elicit secondary injury to membranes and organelles, causing an irreversible cascade of apoptosis, necrosis, and axonal damage. This is followed by activation of innate immune responses, leading to endorgan injury and widespread metabolic acidosis [24][25][26][27] , which could alter ADAR and A-to-I editing. Moreover, DNA is relatively stable over extended postmortem periods, RNA is much more chemically labile and sensitive 28 . ...
Article
Full-text available
Adenosine-to-inosine (A-to-I) editing is a prevalent post-transcriptional RNA modification within the brain. Yet, most research has relied on postmortem samples, assuming it is an accurate representation of RNA biology in the living brain. We challenge this assumption by comparing A-to-I editing between postmortem and living prefrontal cortical tissues. Major differences were found, with over 70,000 A-to-I sites showing higher editing levels in postmortem tissues. Increased A-to-I editing in postmortem tissues is linked to higher ADAR and ADARB1 expression, is more pronounced in non-neuronal cells, and indicative of postmortem activation of inflammation and hypoxia. Higher A-to-I editing in living tissues marks sites that are evolutionarily preserved, synaptic, developmentally timed, and disrupted in neurological conditions. Common genetic variants were also found to differentially affect A-to-I editing levels in living versus postmortem tissues. Collectively, these discoveries offer more nuanced and accurate insights into the regulatory mechanisms of RNA editing in the human brain.
... Advances in biotechnology, particularly in transcriptomics, have enabled a deeper understanding of the molecular mechanisms underlying ischemic brain injury (17,18,19). Single-cell RNA-seq sequencing elucidates gene expression heterogeneity at the cellular level, offering a powerful tool for identifying distinct immune cell subgroups (20,21,22). ...
Preprint
Full-text available
This study extensively investigated immune-inflammatory responses following ischemic brain injury using cutting-edge technologies. By analyzing the changes in T cells, neutrophils, and monocytes in patients' blood and identifying key immune cells in necrotic brain zones, the study uncovered crucial insights into the dynamics of immune cells during brain injury. Moreover, targeting the Hspa8 gene effectively reduced neutrophil infiltration and ROS production, suggesting a promising therapeutic approach for alleviating ischemic brain injury. Overall, the study not only provided new theoretical perspectives but also proposed a potential strategy for managing functional decline in ischemic brain injury.
... 13 Single-cell transcriptomic technology enables a comprehensive analysis of cellular physiology of organ transplantation and the recipient's immune response. [14][15][16][17] In this study, we conducted comprehensive single-cell RNA sequencing (scRNAseq) analyses on the first porcine-to-human kidney xenografts, as well as on longitudinal peripheral blood mononuclear cell (PBMC) samples, to characterize the intricate cellular and molecular dynamics of xenograft-recipient interactions. Such comprehensive analyses enabled the detection of major transcriptomic changes in both human and porcine cells upon xenotransplantation. ...
... In sum, while NMP allows for prolonged perfusion after extraction, which can buy extra time for liver graft assessment and repair, 49 its medical benefits may not yet quite equal those of NRP, although they may increase as a result of further research on perfusates and biomarkers. 50 Proponents of in situ perfusion, particularly those focused on obtaining hearts, argue that, since TA-NRP is less expensive and resource intensive than NMP, it can be adopted faster and more equitably than NMP. The estimated costs of NMP appear to be higher than those of NRP because NMP requires the purchase and maintenance of a purpose-built machine, whereas NRP uses already purchased ECMO machines and NMP relies more on disposable supplies. ...
Article
Full-text available
In transplant medicine, the use of normothermic regional perfusion (NRP) in donation after circulatory determination of death raises ethical difficulties. NRP is objectionable because it restores the donor's circulation, thus invalidating a death declaration based on the permanent cessation of circulation. NRP's defenders respond with arguments that are tortuous and factually inaccurate and depend on introducing extraneous concepts into the law. However, results comparable to NRP's—more and higher‐quality organs and more efficient allocation—can be achieved by removing organs from deceased donors and using normothermic machine perfusion (NMP) to support the organs outside the body, without jeopardizing confidence in transplantation's legal and ethical foundations. Given the controversy that NRP generates and the convoluted justifications made for it, we recommend a prudential approach we call “ethical parsimony,” which holds that, in the choice between competing means of achieving a result, the ethically simpler one is to be preferred. This approach makes clear that policy‐makers should favor NMP over NRP .
... It has been demonstrated that at subnormothermia (20−32°C), the oxygen demand of the graft can be met using a cell-free perfusate, thereby negating the need for RBCs [18][19][20][21] . Moreover, it was recently demonstrated that at subnormothermia (28°C) there is sufficient metabolic activity to support molecular and cellular repair processes 22 . ...
Article
Full-text available
The growing disparity between the demand for transplants and the available donor supply, coupled with an aging donor population and increasing prevalence of chronic diseases, highlights the urgent need for the development of platforms enabling reconditioning, repair, and regeneration of deceased donor organs. This necessitates the ability to preserve metabolically active kidneys ex vivo for days. However, current kidney normothermic machine perfusion (NMP) approaches allow metabolic preservation only for hours. Here we show that human kidneys discarded for transplantation can be preserved in a metabolically active state up to 4 days when perfused with a cell-free perfusate supplemented with TCA cycle intermediates at subnormothermia (25 °C). Using spatially resolved isotope tracing we demonstrate preserved metabolic fluxes in the kidney microenvironment up to Day 4 of perfusion. Beyond Day 4, significant changes were observed in renal cell populations through spatial lipidomics, and increases in injury markers such as LDH, NGAL and oxidized lipids. Finally, we demonstrate that perfused kidneys maintain functional parameters up to Day 4. Collectively, these findings provide evidence that this approach enables metabolic and functional preservation of human kidneys over multiple days, establishing a solid foundation for future clinical investigations.
... Shortly after death, intracellular acidosis and oedema elicit secondary injury to membranes and organelles, causing an irreversible cascade of apoptosis, necrosis and axonal damage. This is followed by activation of innate immune responses, leading to end-organ injury and wide-spread metabolic acidosis [24][25][26][27] , which could alter ADAR and A-to-I editing. Moreover, DNA is relatively stable over extended postmortem periods, RNA is much more chemically labile and sensitive 28 . ...
Preprint
Full-text available
Adenosine-to-inosine (A-to-I) editing is a prevalent post-transcriptional RNA modification within the brain. Yet, most research has relied on postmortem samples, assuming it is an accurate representation of RNA biology in the living brain. We challenge this assumption by comparing A-to-I editing between postmortem and living prefrontal cortical tissues. Major differences were found, with over 70,000 A-to-I sites showing higher editing levels in postmortem tissues. Increased A-to-I editing in postmortem tissues is linked to higher ADAR1 and ADARB1 expression, is more pronounced in non-neuronal cells, and indicative of postmortem activation of inflammation and hypoxia. Higher A-to-I editing in living tissues marks sites that are evolutionarily preserved, synaptic, developmentally timed, and disrupted in neurological conditions. Common genetic variants were also found to differentially affect A-to-I editing levels in living versus postmortem tissues. Collectively, these discoveries illuminate the nuanced functions and intricate regulatory mechanisms of RNA editing within the human brain.
Article
Full-text available
Since the invention of cardiopulmonary bypass, cardioprotective strategies have been investigated to mitigate ischemic injury to the heart during aortic cross-clamping and reperfusion injury with cross-clamp release. With advances in cardiac surgical and percutaneous techniques and post-operative management strategies including mechanical circulatory support, cardiac surgeons are able to operate on more complex patients. Therefore, there is a growing need for improved cardioprotective strategies to optimize outcomes in these patients. This review provides an overview of the basic principles of cardioprotection in the setting of cardiac surgery, including mechanisms of cardiac injury in the context of cardiopulmonary bypass, followed by a discussion of the specific approaches to optimizing cardioprotection in cardiac surgery, including refinements in cardiopulmonary bypass and cardioplegia, ischemic conditioning, use of specific anesthetic and pharmaceutical agents, and novel mechanical circulatory support technologies. Finally, translational strategies that investigate cardioprotection in the setting of cardiac surgery will be reviewed, with a focus on promising research in the areas of cell-based and gene therapy. Advances in this area will help cardiologists and cardiac surgeons mitigate myocardial ischemic injury, improve functional post-operative recovery, and optimize clinical outcomes in patients undergoing cardiac surgery.
Article
Full-text available
In a previous study, heart xenografts from 10-gene-edited pigs transplanted into two human decedents did not show evidence of acute-onset cellular- or antibody-mediated rejection. Here, to better understand the detailed molecular landscape following xenotransplantation, we carried out bulk and single-cell transcriptomics, lipidomics, proteomics and metabolomics on blood samples obtained from the transplanted decedents every 6 h, as well as histological and transcriptomic tissue profiling. We observed substantial early immune responses in peripheral blood mononuclear cells and xenograft tissue obtained from decedent 1 (male), associated with downstream T cell and natural killer cell activity. Longitudinal analyses indicated the presence of ischemia reperfusion injury, exacerbated by inadequate immunosuppression of T cells, consistent with previous findings of perioperative cardiac xenograft dysfunction in pig-to-nonhuman primate studies. Moreover, at 42 h after transplantation, substantial alterations in cellular metabolism and liver-damage pathways occurred, correlating with profound organ-wide physiological dysfunction. By contrast, relatively minor changes in RNA, protein, lipid and metabolism profiles were observed in decedent 2 (female) as compared to decedent 1. Overall, these multi-omics analyses delineate distinct responses to cardiac xenotransplantation in the two human decedents and reveal new insights into early molecular and immune responses after xenotransplantation. These findings may aid in the development of targeted therapeutic approaches to limit ischemia reperfusion injury-related phenotypes and improve outcomes.
Article
Full-text available
Heart transplantation with donation after circulatory death (DCD) provides excellent patient outcomes and increases donor heart availability. However, unlike conventional grafts obtained through donation after brain death, DCD cardiac grafts are not only exposed to warm, unprotected ischemia, but also to a potentially damaging pre-ischemic phase after withdrawal of life-sustaining therapy (WLST). In this review, we aim to bring together knowledge about changes in cardiac energy metabolism and its regulation that occur in DCD donors during WLST, circulatory arrest, and following the onset of warm ischemia. Acute metabolic, hemodynamic, and biochemical changes in the DCD donor expose hearts to high circulating catecholamines, hypoxia, and warm ischemia, all of which can negatively impact the heart. Further metabolic changes and cellular damage occur with reperfusion. The altered energy substrate availability prior to organ procurement likely plays an important role in graft quality and post-ischemic cardiac recovery. These aspects should, therefore, be considered in clinical protocols, as well as in pre-clinical DCD models. Notably, interventions prior to graft procurement are limited for ethical reasons in DCD donors; thus, it is important to understand these mechanisms to optimize conditions during initial reperfusion in concert with graft evaluation and re-evaluation for the purpose of tailoring and adjusting therapies and ensuring optimal graft quality for transplantation.
Article
Full-text available
Importance Ischemic cold storage (ICS) of livers for transplant is associated with serious posttransplant complications and underuse of liver allografts. Objective To determine whether portable normothermic machine perfusion preservation of livers obtained from deceased donors using the Organ Care System (OCS) Liver ameliorates early allograft dysfunction (EAD) and ischemic biliary complications (IBCs). Design, Setting, and Participants This multicenter randomized clinical trial (International Randomized Trial to Evaluate the Effectiveness of the Portable Organ Care System Liver for Preserving and Assessing Donor Livers for Transplantation) was conducted between November 2016 and October 2019 at 20 US liver transplant programs. The trial compared outcomes for 300 recipients of livers preserved using either OCS (n = 153) or ICS (n = 147). Participants were actively listed for liver transplant on the United Network of Organ Sharing national waiting list. Interventions Transplants were performed for recipients randomly assigned to receive donor livers preserved by either conventional ICS or the OCS Liver initiated at the donor hospital. Main Outcomes and Measures The primary effectiveness end point was incidence of EAD. Secondary end points included OCS Liver ex vivo assessment capability of donor allografts, extent of reperfusion syndrome, incidence of IBC at 6 and 12 months, and overall recipient survival after transplant. The primary safety end point was the number of liver graft–related severe adverse events within 30 days after transplant. Results Of 293 patients in the per-protocol population, the primary analysis population for effectiveness, 151 were in the OCS Liver group (mean [SD] age, 57.1 [10.3] years; 102 [67%] men), and 142 were in the ICS group (mean SD age, 58.6 [10.0] years; 100 [68%] men). The primary effectiveness end point was met by a significant decrease in EAD (27 of 150 [18%] vs 44 of 141 [31%]; P = .01). The OCS Liver preserved livers had significant reduction in histopathologic evidence of ischemia-reperfusion injury after reperfusion (eg, less moderate to severe lobular inflammation: 9 of 150 [6%] for OCS Liver vs 18 of 141 [13%] for ICS; P = .004). The OCS Liver resulted in significantly higher use of livers from donors after cardiac death (28 of 55 [51%] for the OCS Liver vs 13 of 51 [26%] for ICS; P = .007). The OCS Liver was also associated with significant reduction in incidence of IBC 6 months (1.3% vs 8.5%; P = .02) and 12 months (2.6% vs 9.9%; P = .02) after transplant. Conclusions and Relevance This multicenter randomized clinical trial provides the first indication, to our knowledge, that normothermic machine perfusion preservation of deceased donor livers reduces both posttransplant EAD and IBC. Use of the OCS Liver also resulted in increased use of livers from donors after cardiac death. Together these findings indicate that OCS Liver preservation is associated with superior posttransplant outcomes and increased donor liver use. Trial Registration ClinicalTrials.gov Identifier: NCT02522871
Article
Full-text available
The hippocampal-entorhinal system supports cognitive functions, has lifelong neurogenic capabilities in many species, and is selectively vulnerable to Alzheimer’s disease. To investigate neurogenic potential and cellular diversity, we profiled single-nucleus transcriptomes in five hippocampal-entorhinal subregions in humans, macaques, and pigs. Integrated cross-species analysis revealed robust transcriptomic and histologic signatures of neurogenesis in the adult mouse, pig, and macaque but not humans. Doublecortin (DCX), a widely accepted marker of newly generated granule cells, was detected in diverse human neurons, but it did not define immature neuron populations. To explore species differences in cellular diversity and implications for disease, we characterized subregion-specific, transcriptomically defined cell types and transitional changes from the three-layered archicortex to the six-layered neocortex. Notably, METTL7B defined subregion-specific excitatory neurons and astrocytes in primates, associated with endoplasmic reticulum and lipid droplet proteins, including Alzheimer’s disease-related proteins. This resource reveals cell-type- and species-specific properties shaping hippocampal-entorhinal neurogenesis and function.
Article
Full-text available
In donation after circulatory death (DCD), (thoraco)abdominal regional perfusion (RP) restores circulation to a region of the body following death declaration. We systematically reviewed outcomes of solid organ transplantation after RP by searching PubMed, Embase, and Cochrane libraries. Eighty-eight articles reporting on outcomes of liver, kidney, pancreas, heart, and lung transplants or donor/organ utilisation were identified. Meta-analyses were conducted when possible. Methodological quality was assessed using National Institutes of Health (NIH)-scoring tools. Case reports (13/88), case series (44/88), retrospective cohort studies (35/88), retrospective matched cohort studies (5/88), and case-control studies (2/88) were identified, with overall fair quality. As blood viscosity and rheology change below 20°C, studies were grouped as hypothermic (HRP, ≤20°C) or normothermic (NRP, >20°C) regional perfusion. Data demonstrate that RP is a safe alternative to in situ cold preservation (ISP) in uncontrolled and controlled DCDs. The scarce HRP data are from before 2005. NRP appears to reduce post-transplant complications, especially biliary complications in controlled DCD livers, compared to ISP. Comparisons for kidney and pancreas with ISP are needed but there is no evidence NRP is detrimental. Additional data on NRP in thoracic organs are needed. Whether RP increases donor or organ utilisation needs further research.
Article
Full-text available
Ischemia reperfusion injury (IRI) is associated with a broad array of life-threatening medical conditions including myocardial infarct, cerebral stroke, and organ transplant. Although the pathobiology and clinical manifestations of IRI are well reviewed by previous publications, IRI-related transcriptomic alterations are less studied. This study aimed to reveal a transcriptomic hallmark for IRI by using the RNA-sequencing data provided by several studies on non-human preclinical experimental models. In this regard, we focused on the transcriptional responses of IRI in an acute time-point up to 48 h. We compiled a list of highly reported genes in the current literature that are affected in the context of IRI. We conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses and found many of the up-regulated genes to be involved in cell survival, cell surface signaling, response to oxidative stress, and inflammatory response, while down-regulated genes were predominantly involved in ion transport. Furthermore, by GO analysis, we found that multiple inflammatory and stress response processes were affected after IRI. Tumor necrosis factor alpha (TNF) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways were also highlighted in the Kyoto Encyclopedia of Genes and Genomes enrichment analysis. In the last section, we discuss the treatment approaches and their efficacy for IRI by comparing RNA sequencing data from therapeutic interventions with the results of our cross-comparison of differentially expressed genes and pathways across IRI.
Article
Full-text available
The simultaneous measurement of multiple modalities represents an exciting frontier for single-cell genomics and necessitates computational methods that can define cellular states based on multimodal data. Here, we introduce “weighted-nearest neighbor” analysis, an unsupervised framework to learn the relative utility of each data type in each cell, enabling an integrative analysis of multiple modalities. We apply our procedure to a CITE-seq dataset of 211,000 human peripheral blood mononuclear cells (PBMCs) with panels extending to 228 antibodies to construct a multimodal reference atlas of the circulating immune system. Multimodal analysis substantially improves our ability to resolve cell states, allowing us to identify and validate previously unreported lymphoid subpopulations. Moreover, we demonstrate how to leverage this reference to rapidly map new datasets and to interpret immune responses to vaccination and coronavirus disease 2019 (COVID-19). Our approach represents a broadly applicable strategy to analyze single-cell multimodal datasets and to look beyond the transcriptome toward a unified and multimodal definition of cellular identity.
Article
Full-text available
Currently, pyroptosis has received more and more attention because of its association with innate immunity and disease. The research scope of pyroptosis has expanded with the discovery of the gasdermin family. A great deal of evidence shows that pyroptosis can affect the development of tumors. The relationship between pyroptosis and tumors is diverse in different tissues and genetic backgrounds. In this review, we provide basic knowledge of pyroptosis, explain the relationship between pyroptosis and tumors, and focus on the significance of pyroptosis in tumor treatment. In addition, we further summarize the possibility of pyroptosis as a potential tumor treatment strategy and describe the side effects of radiotherapy and chemotherapy caused by pyroptosis. In brief, pyroptosis is a double-edged sword for tumors. The rational use of this dual effect will help us further explore the formation and development of tumors, and provide ideas for patients to develop new drugs based on pyroptosis.
Article
Full-text available
Understanding global communications among cells requires accurate representation of cell-cell signaling links and effective systems-level analyses of those links. We construct a database of interactions among ligands, receptors and their cofactors that accurately represent known heteromeric molecular complexes. We then develop CellChat, a tool that is able to quantitatively infer and analyze intercellular communication networks from single-cell RNA-sequencing (scRNA-seq) data. CellChat predicts major signaling inputs and outputs for cells and how those cells and signals coordinate for functions using network analysis and pattern recognition approaches. Through manifold learning and quantitative contrasts, CellChat classifies signaling pathways and delineates conserved and context-specific pathways across different datasets. Applying CellChat to mouse and human skin datasets shows its ability to extract complex signaling patterns. Our versatile and easy-to-use toolkit CellChat and a web-based Explorer (http://www.cellchat.org/) will help discover novel intercellular communications and build cell-cell communication atlases in diverse tissues.
Article
Mortality on the liver waitlist remains unacceptably high. Donation after circulatory determination of death (DCD) donors are considered marginal but are a potentially underutilized resource. Thoraco‐abdominal normothermic perfusion (TA‐NRP) in DCD donors might result in higher quality livers and offset waitlist mortality. We retrospectively reviewed outcomes of the first 13 livers transplanted from TA‐NRP donors in the US. Nine centers transplanted livers from 8 organ procurement organizations. Median donor age was 25 years; median agonal phase was 13 minutes. Median recipient age was 60 years; median lab MELD score was 21. Three patients (23%) met early allograft dysfunction (EAD) criteria. Three received simultaneous liver‐kidney transplants; neither had EAD nor delayed renal allograft function. One recipient died 186 days post‐transplant from sepsis but had normal pre‐sepsis liver function. One patient developed a biliary anastomotic stricture, managed endoscopically; no recipient developed clinical evidence of ischemic cholangiopathy (IC). Twelve of 13 (92%) patients are alive with good liver function at 439 days median follow‐up; 1 patient has extrahepatic recurrent HCC. TA‐NRP DCD livers in these recipients all functioned well, particularly with respect to IC, and provide a valuable option to decrease deaths on the waiting list. This article is protected by copyright. All rights reserved
Article
Objective This pilot study sought to evaluate the feasibility of our donation after circulatory death (DCD) heart transplant protocol using cardiopulmonary bypass (CPB) for normothermic regional reperfusion (NRP). Methods Suitable local DCD candidates were transferred to our institution. Life support was withdrawn in the operating room (OR). Upon declaration of circulatory death, sternotomy was performed and the aortic arch vessels were ligated. CPB was initiated with left ventricular venting. The heart was reperfused with correction of any metabolic abnormalities. CPB was weaned and cardiac function assessed at 30-minute intervals. If accepted, the heart was procured with cold preservation and transplanted into recipients in a nearby OR. Results From January 2020 to January 2021, a total of 8 DCD heart transplants were performed: six isolated hearts, one heart-lung, and one combined heart-kidney. All donor hearts were successfully resuscitated and weaned from CPB without inotropic support. Average lactate and potassium levels decreased from 9.39 ± 1.47 mmol/L to 7.20 ± 0.13 mmol/L and 7.49 ± 1.32 mmol/L to 4.36 ± 0.67 mmol/L, respectively. Post-transplant, the heart-lung recipient required venoarterial extracorporeal membrane oxygenation for primary lung graft dysfunction, but was decannulated on postoperative day 3 and recovered uneventfully. All other recipients required minimal inotropic support without mechanical circulatory support. Survival was 100% with median follow-up of 304 days (interquartile range, 105–371 days). Conclusions DCD heart transplant outcomes have been excellent. Our DCD protocol is adoptable for more widespread use, and will increase donor availability in the United States.
Article
The susceptibility of the brain to ischaemic injury dramatically limits its viability following interruptions in blood flow. However, data from studies of dissociated cells, tissue specimens, isolated organs and whole bodies have brought into question the temporal limits within which the brain is capable of tolerating prolonged circulatory arrest. This Review assesses cell type-specific mechanisms of global cerebral ischaemia, and examines the circumstances in which the brain exhibits heightened resilience to injury. We suggest strategies for expanding such discoveries to fuel translational research into novel cytoprotective therapies, and describe emerging technologies and experimental concepts. By doing so, we propose a new multimodal framework to investigate brain resuscitation following extended periods of circulatory arrest.