1
|
Robertson FP, Cuff AO, Male V, Wright GP, Pallett LJ, Fuller BJ, Davidson BR. Inflammatory Monocytes Are Rapidly Recruited to the Post-Ischaemic Liver in Patients Undergoing Liver Transplantation and Cytokines Associated with Their Activation Correlate with Graft Outcomes. Curr Issues Mol Biol 2025; 47:49. [PMID: 39852164 PMCID: PMC11763458 DOI: 10.3390/cimb47010049] [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: 11/26/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Liver ischaemia-reperfusion (IR) injury remains a major cause of morbidity and mortality following liver transplantation and resection. CD4+ T cells have been shown to play a key role in murine models; however, there is currently a lack of data that support their role in human patients. Methods: Data on clinical outcomes and complications were documented prospectively in 28 patients undergoing first elective liver transplant surgery. Peripheral blood samples were collected at baseline (pre-op), 2 h post graft reperfusion, immediately post-op, and 24 h post-op. A post-reperfusion biopsy was analysed in all patients, and in five patients, a donor liver biopsy was available pre-implantation. Circulating cytokines were measured, and T cells were analysed for activation markers and cytokine production. Results: Circulating levels of cytokines associated with innate immune cell recruitment and activation were significantly elevated in the peri-transplant period. High circulating IL-10 levels corresponded with the development of graft-specific complications. The proportion of CD4+ T cells in the peripheral circulation fell throughout the peri-operative period, suggesting CD4+ T cell recruitment to the graft. Although TNFα was the predominant cytokine produced by CD4+ T cells in the intrahepatic environment, the production of IFNγ was significantly upregulated by circulating CD4+ T cells. Furthermore, we demonstrated clear recruitment of inflammatory monocytes in the peri-operative period. In donor-and-recipient pairs with a mismatch at the HLA-A2 or A3 allele, we demonstrated that inflammatory monocytes in the liver are recipient-derived. Discussion: This is the first study to our knowledge that tracks early immune cell responses in humans undergoing liver transplantation. The recruitment of inflammatory monocytes from the recipient and their cytokine release is associated with liver-specific complications. Inflammatory monocytes would be an attractive target to ameliorate ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Francis P. Robertson
- Division of Interventional and Surgical Science, Royal Free Campus, University College London, London NW3 2QG, UK; (B.J.F.); (B.R.D.)
- Department of Surgery, School of Medicine, Gilmorehill Campus, University of Glasgow Medical School, Glasgow G12 8QQ, UK
| | - Antonia O. Cuff
- Division of Biomedical Sciences, Warwick Medical School, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK;
| | - Victoria Male
- Department of Metabolism, Digestion and Reproduction, Chelsea and Westminster Hospital Campus, Imperial College London, London W12 0NN, UK;
| | - Graham P. Wright
- School of Applied Science, Edinburgh Napier University, Edinburgh EH11 4BN, UK;
| | - Laura J. Pallett
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK;
| | - Barry J. Fuller
- Division of Interventional and Surgical Science, Royal Free Campus, University College London, London NW3 2QG, UK; (B.J.F.); (B.R.D.)
| | - Brian R. Davidson
- Division of Interventional and Surgical Science, Royal Free Campus, University College London, London NW3 2QG, UK; (B.J.F.); (B.R.D.)
- Department of HPB and Liver Transplant Surgery, Royal Free Hospital, London NW3 2QG, UK
| |
Collapse
|
2
|
Shi X, Du J, Li T, Pan L. Platelet count as a potential predictor in refractory Takayasu arteritis. Rheumatol Int 2023; 43:2251-2260. [PMID: 37349635 DOI: 10.1007/s00296-023-05368-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023]
Abstract
Platelet parameters have been recognized as important markers for disease severity in various types of diseases. The aim of our study was to investigate whether platelet count could be used as a potential predictor of refractory Takayasu arteritis (TAK). In this retrospective study, fifty-seven patients were selected as development data group to identify the associated risk factors and potential predictors of refractory TAK. Ninety-two TAK patients were included in the validation data group to verify the predictive value of platelet count for refractory TAK. Refractory TAK patients had higher levels of platelet (PLT) than non-refractory TAK patients (305.5 vs. 272.0 × 109/L, P = 0.043). For PLT, the best cut-off value was 296.5 × 109/L to predict refractory TAK. Elevated PLT (> 296.5 × 109/L) was found to be statistically related to refractory TAK (OR [95%CI] 4.000 [1.233-12.974], p = 0.021). In the validation data group, the proportion of refractory TAK in patients with elevated PLT was significantly higher than that in patients with non-elevated PLT (55.6% vs. 32.2%, P = 0.037). The 1-, 3- and 5-year cumulative incidence of refractory TAK were 37.0%, 44.4% and 55.6% in patients with elevated PLT, respectively. Elevated PLT (p = 0.035, hazard ratio (HR) 2.106) was identified as a potential predictor of refractory TAK. Clinicians should pay close attention to platelet levels in patients with TAK. For TAK patients with PLT greater than 296.5 × 109/L, closer monitoring of the disease and comprehensive assessment of disease activity are recommended to be alert to the occurrence of refractory TAK.
Collapse
Affiliation(s)
- Xuemei Shi
- Department of Rheumatology and Immunology, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Juan Du
- Department of Rheumatology and Immunology, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Taotao Li
- Department of Rheumatology and Immunology, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Lili Pan
- Department of Rheumatology and Immunology, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
3
|
Heo MJ, Suh JH, Poulsen KL, Ju C, Kim KH. Updates on the Immune Cell Basis of Hepatic Ischemia-Reperfusion Injury. Mol Cells 2023; 46:527-534. [PMID: 37691258 PMCID: PMC10495686 DOI: 10.14348/molcells.2023.0099] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 06/19/2023] [Accepted: 07/21/2023] [Indexed: 09/12/2023] Open
Abstract
Liver ischemia-reperfusion injury (IRI) is the main cause of organ dysfunction and failure after liver surgeries including organ transplantation. The mechanism of liver IRI is complex and numerous signals are involved but cellular metabolic disturbances, oxidative stress, and inflammation are considered the major contributors to liver IRI. In addition, the activation of inflammatory signals exacerbates liver IRI by recruiting macrophages, dendritic cells, and neutrophils, and activating NK cells, NKT cells, and cytotoxic T cells. Technological advances enable us to understand the role of specific immune cells during liver IRI. Accordingly, therapeutic strategies to prevent or treat liver IRI have been proposed but no definitive and effective therapies exist yet. This review summarizes the current update on the immune cell functions and discusses therapeutic potentials in liver IRI. A better understanding of this complex and highly dynamic process may allow for the development of innovative therapeutic approaches and optimize patient outcomes.
Collapse
Affiliation(s)
- Mi Jeong Heo
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ji Ho Suh
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kyle L. Poulsen
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Cynthia Ju
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kang Ho Kim
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
4
|
Morris SM, Chauhan A. The role of platelet mediated thromboinflammation in acute liver injury. Front Immunol 2022; 13:1037645. [PMID: 36389830 PMCID: PMC9647048 DOI: 10.3389/fimmu.2022.1037645] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022] Open
Abstract
Acute liver injuries have wide and varied etiologies and they occur both in patients with and without pre-existent chronic liver disease. Whilst the pathophysiological mechanisms remain distinct, both acute and acute-on-chronic liver injury is typified by deranged serum transaminase levels and if severe or persistent can result in liver failure manifest by a combination of jaundice, coagulopathy and encephalopathy. It is well established that platelets exhibit diverse functions as immune cells and are active participants in inflammation through processes including immunothrombosis or thromboinflammation. Growing evidence suggests platelets play a dualistic role in liver inflammation, shaping the immune response through direct interactions and release of soluble mediators modulating function of liver sinusoidal endothelial cells, stromal cells as well as migrating and tissue-resident leucocytes. Elucidating the pathways involved in initiation, propagation and resolution of the immune response are of interest to identify therapeutic targets. In this review the provocative role of platelets is outlined, highlighting beneficial and detrimental effects in a spatial, temporal and disease-specific manner.
Collapse
Affiliation(s)
- Sean M. Morris
- The Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom
| | - Abhishek Chauhan
- The Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Abhishek Chauhan,
| |
Collapse
|
5
|
Gao F, Qiu X, Wang K, Shao C, Jin W, Zhang Z, Xu X. Targeting the Hepatic Microenvironment to Improve Ischemia/Reperfusion Injury: New Insights into the Immune and Metabolic Compartments. Aging Dis 2022; 13:1196-1214. [PMID: 35855339 PMCID: PMC9286916 DOI: 10.14336/ad.2022.0109] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/09/2022] [Indexed: 12/12/2022] Open
Abstract
Hepatic ischemia/reperfusion injury (IRI) is mainly characterized by high activation of immune inflammatory responses and metabolic responses. Understanding the molecular and metabolic mechanisms underlying development of hepatic IRI is critical for developing effective therapies for hepatic IRI. Recent advances in research have improved our understanding of the pathogenesis of IRI. During IRI, hepatocyte injury and inflammatory responses are mediated by crosstalk between the immune cells and metabolic components. This crosstalk can be targeted to treat or reverse hepatic IRI. Thus, a deep understanding of hepatic microenvironment, especially the immune and metabolic responses, can reveal new therapeutic opportunities for hepatic IRI. In this review, we describe important cells in the liver microenvironment (especially non-parenchymal cells) that regulate immune inflammatory responses. The role of metabolic components in the diagnosis and prevention of hepatic IRI are discussed. Furthermore, recent updated therapeutic strategies based on the hepatic microenvironment, including immune cells and metabolic components, are highlighted.
Collapse
Affiliation(s)
- Fengqiang Gao
- 1Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,6Zhejiang University School of Medicine, Hangzhou, China
| | - Xun Qiu
- 1Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,6Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Wang
- 1Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chuxiao Shao
- 7Department of Hepatobiliary and Pancreatic Surgery, Affiliated Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Wenjian Jin
- 8Department of Hepatobiliary Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhen Zhang
- 6Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Xu
- 1Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,2Zhejiang University Cancer Center, Hangzhou, China.,3Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,4NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,5Institute of Organ Transplantation, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Tao L, Ren X, Zhai W, Chen Z. Progress and Prospects of Non-Canonical NF-κB Signaling Pathway in the Regulation of Liver Diseases. Molecules 2022; 27:molecules27134275. [PMID: 35807520 PMCID: PMC9268066 DOI: 10.3390/molecules27134275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Non-canonical nuclear factor kappa B (NF-κB) signaling pathway regulates many physiological and pathological processes, including liver homeostasis and diseases. Recent studies demonstrate that non-canonical NF-κB signaling pathway plays an essential role in hyperglycemia, non-alcoholic fatty liver disease, alcoholic liver disease, liver regeneration, liver injury, autoimmune liver disease, viral hepatitis, and hepatocellular carcinoma. Small-molecule inhibitors targeting to non-canonical NF-κB signaling pathway have been developed and shown promising results in the treatment of liver injuries. Here, the recent advances and future prospects in understanding the roles of the non-canonical NF-κB signaling pathways in the regulation of liver diseases are discussed.
Collapse
Affiliation(s)
- Li Tao
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Xiaomeng Ren
- College of Pharmaceutical and Biology Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| | - Wenhui Zhai
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| |
Collapse
|
7
|
Funken D, Brüggemann A, Mende K, Lerchenberger M, Rentsch M, Khandoga A. Blocking of CX3CL1 attenuates platelet and leukocyte recruitment in murine hepatic I/R. Eur Surg Res 2022; 63:000524024. [PMID: 35279656 PMCID: PMC9808741 DOI: 10.1159/000524024] [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: 09/28/2021] [Accepted: 01/26/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND The chemokine fractalkine (CX3CL1) is critically involved in the pathophysiology of different inflammatory diseases and myocardial ischemia-reperfusion (I/R). This study aims to analyze the role of fractalkine in the activation of platelets and leukocytes during hepatic I/R. METHODS Under inhalation anesthesia, C57BL6 mice were subjected to warm hepatic I/R (90min/240min). The animals were pretreated either with a function-blocking anti-mouse CX3CL1 antibody or IgG control administered systemically before ischemia. Sham-operated animals served as controls (n=7 each group). The inflammatory response and sinusoidal perfusion failure were evaluated by intravital microscopy. Hepatic transaminases plasma levels and histopathological tissue damage were determined as markers of hepatocellular injury. RESULTS Sinusoidal perfusion failure, leukocyte recruitment to the liver, and transaminase activities were sharply increased upon I/R compared to sham-operated mice. Firm adhesion of platelets and concordantly leukocytes to endothelial cells is reduced significantly by a function-blocking anti-fractalkine antibody. We demonstrate that inhibition of fractalkine signaling attenuates leukocyte adhesion in the postischemic liver but does not significantly ameliorate overall perfusion failure and hepatocellular injury. DISCUSSION/CONCLUSION Our in vivo data demonstrate a mild attenuating effect of CX3CL1 blockade on platelet and leukocyte, but not CD4+ T cell accumulation and activation in hepatic I/R injury. We report a significant effect of blocking chemokine fractalkine on sinusoidal perfusion failure without considerably improving overall hepatocellular injury during early reperfusion.
Collapse
Affiliation(s)
- Dominik Funken
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University Hospital of Munich, Ludwig-Maximilians-University of Munich, Munich, Germany,Walter-Brendel Centre for Experimental Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany,Department of Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany,*Dominik Funken,
| | - Alexandra Brüggemann
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University Hospital of Munich, Ludwig-Maximilians-University of Munich, Munich, Germany,Walter-Brendel Centre for Experimental Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Konstantin Mende
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University Hospital of Munich, Ludwig-Maximilians-University of Munich, Munich, Germany,Walter-Brendel Centre for Experimental Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Maximilian Lerchenberger
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University Hospital of Munich, Ludwig-Maximilians-University of Munich, Munich, Germany,Walter-Brendel Centre for Experimental Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Markus Rentsch
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University Hospital of Munich, Ludwig-Maximilians-University of Munich, Munich, Germany,Walter-Brendel Centre for Experimental Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Andrej Khandoga
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University Hospital of Munich, Ludwig-Maximilians-University of Munich, Munich, Germany,Walter-Brendel Centre for Experimental Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany,Department of General, Visceral and Vascular Surgery, Main-Kinzig-Clinics, Gelnhausen, Germany,**Andrej Khandoga,
| |
Collapse
|
8
|
Ramavath NN, Gadipudi LL, Provera A, Gigliotti LC, Boggio E, Bozzola C, Albano E, Dianzani U, Sutti S. Inducible T-Cell Costimulator Mediates Lymphocyte/Macrophage Interactions During Liver Repair. Front Immunol 2021; 12:786680. [PMID: 34925367 PMCID: PMC8678521 DOI: 10.3389/fimmu.2021.786680] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
The liver capacity to recover from acute liver injury is a critical factor in the development of acute liver failure (ALF) caused by viral infections, ischemia/reperfusion or drug toxicity. Liver healing requires the switching of pro-inflammatory monocyte-derived macrophages(MoMFs) to a reparative phenotype. However, the mechanisms involved are still incompletely characterized. In this study we investigated the contribution of T-lymphocyte/macrophage interaction through the co-stimulatory molecule Inducible T-cell co-stimulator (ICOS; CD278) and its ligand (ICOSL; CD275) in modulating liver repair. The role of ICOS/ICOSL dyad was investigated during the recovery from acute liver damage induced by a single dose of carbon tetrachloride (CCl4). Flow cytometry of non-parenchymal liver cells obtained from CCl4-treated wild-type mice revealed that the recovery from acute liver injury associated with a specific up-regulation of ICOS in CD8+ T-lymphocytes and with an increase in ICOSL expression involving CD11bhigh/F4-80+ hepatic MoMFs. Although ICOS deficiency did not influence the severity of liver damage and the evolution of inflammation, CCl4-treated ICOS knockout (ICOS-/- ) mice showed delayed clearance of liver necrosis and increased mortality. These animals were also characterized by a significant reduction of hepatic reparative MoMFs due to an increased rate of cell apoptosis. An impaired liver healing and loss of reparative MoMFs was similarly evident in ICOSL-deficient mice or following CD8+ T-cells ablation in wild-type mice. The loss of reparative MoMFs was prevented by supplementing CCl4-treated ICOS-/- mice with recombinant ICOS (ICOS-Fc) which also stimulated full recovery from liver injury. These data demonstrated that CD8+ T-lymphocytes play a key role in supporting the survival of reparative MoMFs during liver healing trough ICOS/ICOSL-mediated signaling. These observations open the possibility of targeting ICOS/ICOSL dyad as a novel tool for promoting efficient healing following acute liver injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | | | | |
Collapse
|
9
|
The posttraumatic response of CD4+ regulatory T cells is modulated by direct cell-cell contact via CD40L- and P-selectin-dependent pathways. Cent Eur J Immunol 2021; 46:283-294. [PMID: 34764800 PMCID: PMC8574106 DOI: 10.5114/ceji.2021.109171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
CD4+ FoxP3+ regulatory T cells (CD4+ Tregs) are important for the posttraumatic anti-inflammatory host response. As described previously, platelets are able to modulate CD4+ Treg activity in a reciprocally activating interaction following injury. The underlying mechanisms of the posttraumatic interaction between platelets and CD4+ Tregs remain unclear. We investigated the potential influence of CD40L and P-selectin, molecules known to be involved in direct cell contact of these cell types. In a murine burn injury model, the potential interaction pathways were addressed using CD40L- and P-selectin-deficient mice. Draining lymph nodes were harvested following trauma (1 h) and following a sham procedure. Early rapid activation of CD4+ Tregs was assessed by phospho-flow cytometry (signaling molecules (p)PKC-δ and (p)ZAP-70). Platelet function was analyzed performing rotational thromboelastometry (ROTEM). We hypothesized that disruption of the direct cell-cell contact via CD40L and P-selectin would affect posttraumatic activation of CD4+ Tregs and influence the hemostatic function of platelets. Indeed, while injury induced early activation of CD4+ Tregs in wild-type mice (ZAP-70: p = 0.13, pZAP-70: p < 0.05, PKC-δ: p < 0.05, pPKC-δ: p < 0.05), disruption of CD40L-dependent interaction (ZAP-70: p = 0.57, pZAP-70: p = 0.68, PKC-δ: p = 0.68, pPKC-δ: p = 0.9) or P-selectin-dependent interaction (ZAP-70: p = 0.78, pZAP-70: p = 0.58, PKC-δ: p = 0.81, pPKC-δ: p = 0.73) resulted in reduced posttraumatic activation. Furthermore, hemostatic function was impaired towards hypocoagulability in either deficiency. Our results suggest that the posttraumatic activation of CD4+ Tregs and hemostatic function of platelets are affected by direct cell-cell-signaling via CD40L and P-selectin.
Collapse
|
10
|
Valade G, Libert N, Martinaud C, Vicaut E, Banzet S, Peltzer J. Therapeutic Potential of Mesenchymal Stromal Cell-Derived Extracellular Vesicles in the Prevention of Organ Injuries Induced by Traumatic Hemorrhagic Shock. Front Immunol 2021; 12:749659. [PMID: 34659252 PMCID: PMC8511792 DOI: 10.3389/fimmu.2021.749659] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
Severe trauma is the principal cause of death among young people worldwide. Hemorrhagic shock is the leading cause of death after severe trauma. Traumatic hemorrhagic shock (THS) is a complex phenomenon associating an absolute hypovolemia secondary to a sudden and significant extravascular blood loss, tissue injury, and, eventually, hypoxemia. These phenomena are responsible of secondary injuries such as coagulopathy, endotheliopathy, microcirculation failure, inflammation, and immune activation. Collectively, these dysfunctions lead to secondary organ failures and multi-organ failure (MOF). The development of MOF after severe trauma is one of the leading causes of morbidity and mortality, where immunological dysfunction plays a central role. Damage-associated molecular patterns induce an early and exaggerated activation of innate immunity and a suppression of adaptive immunity. Severe complications are associated with a prolonged and dysregulated immune–inflammatory state. The current challenge in the management of THS patients is preventing organ injury, which currently has no etiological treatment available. Modulating the immune response is a potential therapeutic strategy for preventing the complications of THS. Mesenchymal stromal cells (MSCs) are multipotent cells found in a large number of adult tissues and used in clinical practice as therapeutic agents for immunomodulation and tissue repair. There is growing evidence that their efficiency is mainly attributed to the secretion of a wide range of bioactive molecules and extracellular vesicles (EVs). Indeed, different experimental studies revealed that MSC-derived EVs (MSC-EVs) could modulate local and systemic deleterious immune response. Therefore, these new cell-free therapeutic products, easily stored and available immediately, represent a tremendous opportunity in the emergency context of shock. In this review, the pathophysiological environment of THS and, in particular, the crosstalk between the immune system and organ function are described. The potential therapeutic benefits of MSCs or their EVs in treating THS are discussed based on the current knowledge. Understanding the key mechanisms of immune deregulation leading to organ damage is a crucial element in order to optimize the preparation of EVs and potentiate their therapeutic effect.
Collapse
Affiliation(s)
- Guillaume Valade
- Institut de Recherche Biomédicale des Armées (IRBA), Inserm UMRS-MD-1197, Clamart, France
| | - Nicolas Libert
- Service d'Anesthésie-Réanimation, Hôpital d'instruction des armées Percy, Clamart, France
| | - Christophe Martinaud
- Unité de Médicaments de Thérapie Innovante, Centre de Transfusion Sanguine des Armées, Clamart, France
| | - Eric Vicaut
- Laboratoire d'Etude de la Microcirculation, Université de Paris, UMRS 942 INSERM, Paris, France
| | - Sébastien Banzet
- Institut de Recherche Biomédicale des Armées (IRBA), Inserm UMRS-MD-1197, Clamart, France
| | - Juliette Peltzer
- Institut de Recherche Biomédicale des Armées (IRBA), Inserm UMRS-MD-1197, Clamart, France
| |
Collapse
|
11
|
Lack of gamma delta T cells ameliorates inflammatory response after acute intestinal ischemia reperfusion in mice. Sci Rep 2021; 11:18628. [PMID: 34545104 PMCID: PMC8452610 DOI: 10.1038/s41598-021-96525-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/29/2021] [Indexed: 01/07/2023] Open
Abstract
T-cells have been demonstrated to modulate ischemia–reperfusion injury (IRI) in the kidney, lung, liver, and intestine. Whereas most T-cell subpopulations contribute primarily to the antigen-specific effector and memory phases of immunity, γδ-T-cells combine adaptive features with rapid, innate-like responses that can place them in the initiation phase of immune reactions. Therefore, we aimed to clarify the role of γδ-T-cells in intestinal IRI. Adult wild-type (WT) and γδ-T-cell-deficient mice were subjected to acute intestinal IRI. Gene expression of pro-inflammatory cytokines and influx of leukocyte subpopulations in the gut were assessed by qPCR and flow cytometry. Serum transaminases were measured as an indicator of distant organ IRI. Intestinal IRI led to increased influx of neutrophils, pro-inflammatory cytokine expression and LDH/ALT/AST elevation. Selective deficiency of γδ-T-cells significantly decreased pro-inflammatory cytokine levels and neutrophil infiltration in the gut following IRI compared to controls. Furthermore, γδ-T-cell deficiency resulted in decreased LDH and transaminases levels in sera, indicating amelioration of distant organ injury. Increasing evidence demonstrates a key role of T-cell subpopulations in IRI. We demonstrate that γδ-T-cell deficiency ameliorated pro-inflammatory cytokine production, neutrophil recruitment and distant organ injury. Thus, γδ-T-cells may be considered as mediators contributing to the inflammatory response in the acute phase of intestinal IRI.
Collapse
|
12
|
Kageyama S, Kadono K, Hirao H, Nakamura K, Ito T, Gjertson DW, Sosa RA, Reed EF, Kaldas FM, Busuttil RW, Kupiec-Weglinski JW, Zhai Y. Ischemia-reperfusion Injury in Allogeneic Liver Transplantation: A Role of CD4 T Cells in Early Allograft Injury. Transplantation 2021; 105:1989-1997. [PMID: 33065722 PMCID: PMC8046839 DOI: 10.1097/tp.0000000000003488] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND A major discrepancy between clinical and most experimental settings of liver ischemia-reperfusion injury (IRI) is the allogenicity. METHODS In the current study, we first established a murine model of allogeneic orthotopic liver transplantation with extended cold ischemia time (18 h). Roles of CD4 T cells in the pathogenesis of IRI in liver allografts were determined using a depleting anti-CD4 antibody. The clinical relevance of CD4 as a marker of liver IRI was analyzed retrospectively in 55 liver transplant patients. RESULTS CD4 depletion in both donors and recipients resulted in the most effective protection of liver allografts from IRI, as measured by serum transaminase levels and liver histology. CD4 depletion inhibited IR-induced intragraft neutrophil/macrophage infiltration and proinflammatory gene expressions. Quantitative reverse-transcriptase polymerase chain reaction analysis of human liver biopsies (2 h postreperfusion) revealed that posttransplant, rather than pretransplant, CD4 transcript levels correlated positively with proinflammatory gene expression profile. When we divided patients into subgroups according to intragraft CD4 levels, the high CD4 cohort developed a more severe hepatocellular damage than that with low CD4 levels. CONCLUSIONS CD4 T cells play a key pathogenic role in IRI of allogeneic liver transplants, and intragraft CD4 levels in the early postreperfusion phase may serve as a potential biomarker and therapeutic target to ameliorate liver IRI and improve orthotopic liver transplantation outcomes.
Collapse
Affiliation(s)
- Shoichi Kageyama
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| | - Kentaro Kadono
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| | - Hirofumi Hirao
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| | - Kojiro Nakamura
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| | - Takahiro Ito
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| | - David W. Gjertson
- Department of Biostatistics, UCLA School of Public Health University of California, Los Angeles, CA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA
| | - Rebecca A. Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA
| | - Fady M. Kaldas
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| | - Ronald W. Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, University of California, Los Angeles, CA
| |
Collapse
|
13
|
Guo L, Shen S, Rowley JW, Tolley ND, Jia W, Manne BK, McComas KN, Bolingbroke B, Kosaka Y, Krauel K, Denorme F, Jacob SP, Eustes AS, Campbell RA, Middleton EA, He X, Brown SM, Morrell CN, Weyrich AS, Rondina MT. Platelet MHC class I mediates CD8+ T-cell suppression during sepsis. Blood 2021; 138:401-416. [PMID: 33895821 PMCID: PMC8343546 DOI: 10.1182/blood.2020008958] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Circulating platelets interact with leukocytes to modulate host immune and thrombotic responses. In sepsis, platelet-leukocyte interactions are increased and have been associated with adverse clinical events, including increased platelet-T-cell interactions. Sepsis is associated with reduced CD8+ T-cell numbers and functional responses, but whether platelets regulate CD8+ T-cell responses during sepsis remains unknown. In our current study, we systemically evaluated platelet antigen internalization and presentation through major histocompatibility complex class I (MHC-I) and their effects on antigen-specific CD8+ T cells in sepsis in vivo and ex vivo. We discovered that both human and murine platelets internalize and proteolyze exogenous antigens, generating peptides that are loaded onto MHC-I. The expression of platelet MHC-I, but not platelet MHC-II, is significantly increased in human and murine platelets during sepsis and in human megakaryocytes stimulated with agonists generated systemically during sepsis (eg, interferon-γ and lipopolysaccharide). Upregulation of platelet MHC-I during sepsis increases antigen cross-presentation and interactions with CD8+ T cells in an antigen-specific manner. Using a platelet lineage-specific MHC-I-deficient mouse strain (B2Mf/f-Pf4Cre), we demonstrate that platelet MHC-I regulates antigen-specific CD8+ T-cell proliferation in vitro, as well as the number and functional responses of CD8+ T cells in vivo, during sepsis. Loss of platelet MHC-I reduces sepsis-associated mortality in mice in an antigen-specific setting. These data identify a new mechanism by which platelets, through MHC-I, process and cross-present antigens, engage antigen-specific CD8+ T cells, and regulate CD8+ T-cell numbers, functional responses, and outcomes during sepsis.
Collapse
Affiliation(s)
- Li Guo
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Sikui Shen
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- West China Hospital, Sichuan University, Chengdu, China
| | - Jesse W Rowley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Pulmonary and Critical Care Division, Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT
| | - Neal D Tolley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Wenwen Jia
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | | | - Kyra N McComas
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Ben Bolingbroke
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT
| | - Yasuhiro Kosaka
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Krystin Krauel
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| | - Frederik Denorme
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Shancy P Jacob
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Alicia S Eustes
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Internal Medicine, University of Iowa, Iowa City, IA
| | - Robert A Campbell
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Division of General Internal Medicine, Department of Medicine, School of Medicine, and
| | - Elizabeth A Middleton
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Pulmonary and Critical Care Division, Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT
| | - Xiao He
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Samuel M Brown
- Pulmonary and Critical Care Division, Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT
- Center for Humanizing Critical Care, Intermountain Healthcare, Murray, UT
- Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Murray, UT
| | - Craig N Morrell
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY; and
| | - Andrew S Weyrich
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Pulmonary and Critical Care Division, Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Division of General Internal Medicine, Department of Medicine, School of Medicine, and
- Department of Pathology, University of Utah, Salt Lake City, UT
- Department of Internal Medicine, George E. Wahlen VA Medical Center and Geriatric Research Education Clinical Center (GRECC), Salt Lake City, UT
| |
Collapse
|
14
|
Shi L, Lu BL, Qiu Y, Huang L, Huang SY, Mao R, Lin JJ, Du JF, Feng ST, Li ZP, Sun CH, Li XH. Hepatic mosaic enhancement pattern correlates with increased inflammatory activity and adverse therapeutic outcomes in patients with Crohn's disease. Abdom Radiol (NY) 2021; 46:3149-3158. [PMID: 33646351 DOI: 10.1007/s00261-021-02979-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/25/2021] [Accepted: 02/09/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE This study aimed to evaluate the role of hepatic mosaic enhancement pattern (HMEP) on computed tomography images in the disease activity and therapeutic outcome of Crohn's Disease (CD). METHODS Twenty-five CD patients with HMEP comprised the HMEP group, and 25 CD patients without HMEP, who had a similar onset age, sex, and disease course with those in the HMEP group, comprised the non-HMEP group. No underlying liver/biliary disease was observed in any of the patients. Clinical characteristics, laboratory test results, Lémann index, and CD endoscopic index of severity (CDEIS) were compared between the groups using the Student t-, Mann-Whitney U, Chi square, or Fisher's exact tests. Patients received top-down, step-up, or traditional treatment during the follow-up period. After the 1-year follow-up, therapeutic outcomes (active inflammation [CDEIS > 3.5 if the endoscopic data were available, or C-reactive protein level > 5 mg/L if the endoscopic data were unavailable] or remission) were evaluated. RESULTS The occurrence rate of fistulas/abscesses was higher in the HMEP group (84%, 21/25) than in the non-HMEP group (48%, 12/25) with no statistical significance (P = 0.056). The HMEP group showed a higher C-reactive protein level (P = 0.001), erythrocyte sedimentation rate (P = 0.013), and blood platelet count (P = 0.005). There was no significant difference in therapeutic strategies between the groups (P = 0.509). The HMEP group showed a significantly lower remission ratio after anti-inflammatory treatment than the non-HMEP group (P = 0.045). CONCLUSIONS HMEP was correlated with increased inflammatory activity and adverse therapeutic outcomes in CD. This finding provided insights regarding novel markers of CD diagnosis and treatment.
Collapse
Affiliation(s)
- Li Shi
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
- Department of Radiology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150, People's Republic of China
| | - Bao-Lan Lu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Yun Qiu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Li Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Si-Yun Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Jin-Jiang Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Jin-Fang Du
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Zi-Ping Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Can-Hui Sun
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China.
| | - Xue-Hua Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
15
|
Rossaint J, Thomas K, Mersmann S, Skupski J, Margraf A, Tekath T, Jouvene CC, Dalli J, Hidalgo A, Meuth SG, Soehnlein O, Zarbock A. Platelets orchestrate the resolution of pulmonary inflammation in mice by T reg cell repositioning and macrophage education. J Exp Med 2021; 218:212168. [PMID: 34014253 PMCID: PMC8142284 DOI: 10.1084/jem.20201353] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/29/2020] [Accepted: 04/19/2021] [Indexed: 12/14/2022] Open
Abstract
Beyond hemostasis, platelets actively participate in immune cell recruitment and host defense, yet their potential in the resolution of inflammatory processes remains unknown. Here, we demonstrate that platelets are recruited into the lung together with neutrophils during the onset of inflammation and alongside regulatory T (T reg) cells during the resolution phase. This partnering dichotomy is regulated by differential adhesion molecule expression during resolution. Mechanistically, intravascular platelets form aggregates with T reg cells, a prerequisite for their recruitment into the lung. This interaction relies on platelet activation by sCD40L and platelet P-selectin binding to PSGL-1 on T reg cells. Physical platelet–T reg cell interactions are necessary to modulate the transcriptome and instruct T reg cells to release the anti-inflammatory mediators IL-10 and TGFβ. Notably, the presence of platelet–T reg cell aggregates in the lung was also required for macrophage transcriptional reprogramming, polarization toward an anti-inflammatory phenotype, and effective resolution of pulmonary inflammation. Thus, platelets partner with successive immune cell subsets to orchestrate both the initiation and resolution of inflammation.
Collapse
Affiliation(s)
- Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Katharina Thomas
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Sina Mersmann
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Jennifer Skupski
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Tobias Tekath
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Charlotte C Jouvene
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Andres Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Sven G Meuth
- Clinic of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Institute for Cardiovascular Prevention, Ludwig-Maximillians-Universität München, Munich, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
16
|
Zhong Y, Xu M, Hu J, Huang X, Lin N, Deng M. Inhibiting Th1/2 cells influences hepatic capillarization by adjusting sinusoidal endothelial fenestrae through Rho-ROCK-myosin pathway. Aging (Albany NY) 2021; 13:5069-5086. [PMID: 33535174 PMCID: PMC7950229 DOI: 10.18632/aging.202425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 11/10/2020] [Indexed: 12/24/2022]
Abstract
CD4+ T cells are considered to be vital in chronic liver diseases, but their exact roles in hepatic capillarization, the typical characteristic of liver fibrosis, are poorly understood. This study aimed to assess the roles of typical subtype of CD4+ T cells, named T helper 1 (Th1) and Th2 cells in liver fibrosis. Taking advantage of well established fibrotic rat model, we conducted in vitro and in vivo experiments to explore the interactions between liver sinusoidal endothelial cells (LSECs) and Th1/2 cells; meanwhile we evaluated the degree of hepatic capillarization when inhibiting these interactions with inhibitory antibodies. Our results showed that prohibiting interactions between Th2 cells and LSECs caused the restoration of fenestrae, increased cytokine level of Th1 cells and reduction of hepatic capillarization; inhibiting the interaction between Th1 cells and LSECs produced the opposite effects. Moreover, increased Rho and myosin light chain phosphorylation were observed when Th1 cells were inhibited with the corresponding inhibitory antibody; Th2 cell inhibition yielded the opposite results. This study indicated that Th1/2 cells steer the capillarization process in different directions and this effect is probably mediated by the Rho-Rho kinase (ROCK)-myosin signaling pathway.
Collapse
Affiliation(s)
- Yuesi Zhong
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Mingxing Xu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Jingxiong Hu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Xi Huang
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Nan Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Meihai Deng
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| |
Collapse
|
17
|
Tang T, Cheng X, Truong B, Sun L, Yang X, Wang H. Molecular basis and therapeutic implications of CD40/CD40L immune checkpoint. Pharmacol Ther 2020; 219:107709. [PMID: 33091428 DOI: 10.1016/j.pharmthera.2020.107709] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/15/2020] [Indexed: 12/22/2022]
Abstract
The CD40 receptor and its ligand CD40L is one of the most critical molecular pairs of the stimulatory immune checkpoints. Both CD40 and CD40L have a membrane form and a soluble form generated by proteolytic cleavage or alternative splicing. CD40 and CD40L are widely expressed in various types of cells, among which B cells and myeloid cells constitutively express high levels of CD40, and T cells and platelets express high levels of CD40L upon activation. CD40L self-assembles into functional trimers which induce CD40 trimerization and downstream signaling. The canonical CD40/CD40L signaling is mediated by recruitment of TRAFs and NF-κB activation, which is supplemented by signal pathways such as PI3K/AKT, MAPKs and JAK3/STATs. CD40/CD40L immune checkpoint leads to activation of both innate and adaptive immune cells via two-way signaling. CD40/CD40L interaction also participates in regulating thrombosis, tissue inflammation, hematopoiesis and tumor cell fate. Because of its essential role in immune activation, CD40/CD40L interaction has been regarded as an attractive immunotherapy target. In recent years, significant advance has been made in CD40/CD40L-targeted therapy. Various types of agents, including agonistic/antagonistic monoclonal antibodies, cellular vaccines, adenoviral vectors and protein antagonist, have been developed and evaluated in early-stage clinical trials for treating malignancies, autoimmune diseases and allograft rejection. In general, these agents have demonstrated favorable safety and some of them show promising clinical efficacy. The mechanisms of benefits include immune cell activation and tumor cell lysis/apoptosis in malignancies, or immune cell inactivation in autoimmune diseases and allograft rejection. This review provides a comprehensive overview of the structure, processing, cellular expression pattern, signaling and effector function of CD40/CD40L checkpoint molecules. In addition, we summarize the progress, targeted diseases and outcomes of current ongoing and completed clinical trials of CD40/CD40L-targeted therapy.
Collapse
Affiliation(s)
- TingTing Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Billy Truong
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - LiZhe Sun
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Department of Cardiovascular Medicine, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - XiaoFeng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA.
| |
Collapse
|
18
|
Marcoux G, Laroche A, Espinoza Romero J, Boilard E. Role of platelets and megakaryocytes in adaptive immunity. Platelets 2020; 32:340-351. [PMID: 32597341 DOI: 10.1080/09537104.2020.1786043] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system is comprised of two principal interconnected components called innate and adaptive immunity. While the innate immune system mounts a nonspecific response that provides protection against the spread of foreign pathogens, the adaptive immune system has developed to specifically recognize a given pathogen and lead to immunological memory. Platelets are small fragments produced from megakaryocytes in bone marrow and lungs. They circulate throughout the blood to monitor the integrity of the vasculature and to prevent bleeding. Given their large repertoire of immune receptors and inflammatory molecules, platelets and megakaryocytes can contribute to both innate and adaptive immunity. In adaptive immunity, platelets and megakaryocytes can process and present antigens to lymphocytes. Moreover, platelets, via FcγRIIA, rapidly respond to pathogens in an immune host when antibodies are present. This manuscript reviews the reported contributions of platelets and megakaryocytes with emphasis on antigen presentation and antibody response in adaptive immunity.
Collapse
Affiliation(s)
- Genevieve Marcoux
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Audrée Laroche
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Jenifer Espinoza Romero
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Eric Boilard
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| |
Collapse
|
19
|
Skoric B, Fabijanovic D, Pasalic M, Planinc AR, Botonjic H, Cikes M, Planinc I, Ljubas-Macek J, Gasparovic H, Milicic D. Lower Platelet Count Following Rabbit Antithymocyte Globulin Induction Is Associated With Less Acute Cellular Rejection in Heart Transplant Recipients. Transplant Proc 2020; 53:335-340. [PMID: 32571710 DOI: 10.1016/j.transproceed.2020.02.172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/16/2020] [Accepted: 02/23/2020] [Indexed: 11/15/2022]
Abstract
BACKGROUND Unlike lymphodepletion, a decrease in platelet count following induction immunosuppressive therapy with polyclonal rabbit antithymocyte globulin (rATG) is deemed as an adverse event. However, this phenomenon may represent a particular rATG antirejection mechanism. METHODS This retrospective single-center study included 156 patients who received a heart transplant (HTx) between 2010 and 2018. All patients received rATG induction therapy for 5 days. Absolute lymphocyte count (ALC) and platelet counts were assessed on days 0, 7, and 14 following HTx. The primary outcome of the study was the first occurrence of acute cellular rejection (ACR) defined as grade ≥ 1B within 24 months after HTx. RESULTS Both ALC and platelet counts decreased rapidly after induction. During the 24-month follow-up period, 17% of patients had ACR. Patients with ACR had significantly higher platelet count on day 7 (145 vs 104, P < .001) and higher ALC on day 14 (162 vs 130, P = .035) than those without rejection. Patients in the highest platelet count quartile showed more ACR (50% in quartile 4 vs 0% in quartile 1, P = .006) as well as a higher cumulative total rejection score. Univariate analysis showed that ACR was associated with platelet count on day 7, recipient age, and pretransplant cytomegalovirus IgG serology. In multivariable regression analysis, platelet count on day 7 was the most accurate predictor of ACR. CONCLUSIONS Lower platelet count after induction with rATG is associated with less ACR. This suggests platelet involvement in antirejection mechanisms of rATG and a possible rationale for targeting platelets in future immunosuppressive strategies.
Collapse
Affiliation(s)
- Bosko Skoric
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Dora Fabijanovic
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Marijan Pasalic
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ana Reschner Planinc
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Hata Botonjic
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Maja Cikes
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ivo Planinc
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Jana Ljubas-Macek
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Hrvoje Gasparovic
- Department of Cardiac Surgery, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Davor Milicic
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| |
Collapse
|
20
|
Saif-Elnasr M, Abdel Fattah SM, Swailam HM. Treatment of hepatotoxicity induced by γ-radiation using platelet-rich plasma and/or low molecular weight chitosan in experimental rats. Int J Radiat Biol 2019; 95:1517-1528. [PMID: 31290709 DOI: 10.1080/09553002.2019.1642538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/09/2019] [Accepted: 06/27/2019] [Indexed: 10/26/2022]
Abstract
Background and aim: Platelet-rich plasma (PRP) is rich in growth factors and plays an important role in tissue healing and cytoprotection. Also, it has been proved that low molecular weight chitosan (LMC) possesses many outstanding health benefits. The aim of this study was to assess the possibility of using PRP and/or fungal LMC to treat hepatotoxicity induced by γ-radiation in albino rats.Materials and methods: Forty-eight adult male albino rats were randomly divided into eight groups. Group I (control), Group II (PRP alone), Group III (LMC alone), Group IV (PRP + LMC), Group V (γ-irradiated alone), Group VI (γ-irradiated + PRP), Group VII (γ-irradiated + LMC), and Group VIII (γ-irradiated + PRP + LMC). The irradiated rats were whole body exposed to γ-radiation (8 Gy) as fractionated doses (2 Gy) twice a week for 2 consecutive weeks. The treated groups received PRP (0.5 mL/kg body weight, s.c.) and/or LMC (10 mg/kg body weight, s.c.) 2 days a week 1 h after every dose of γ-radiation and continued for another week after the last dose of radiation. Serum alanine transaminase (ALT) and aspartate transaminase (AST) activities, as well as reduced glutathione (GSH) content, malondialdehyde (MDA), total antioxidant capacity (TAC), and nuclear factor erythroid 2-related factor 2 (Nrf2) levels in the liver tissue and relative expression of microRNA-21 (miR-21) in serum were measured, in addition to histopathological examination.Results: Exposure of rats to γ-radiation resulted in a significant increase in serum ALT and AST activities, hepatic MDA levels, and serum miR-21 relative expression, along with a significant decrease in hepatic GSH content, TAC, and Nrf2 levels. Treatment with PRP and/or fungal LMC after exposure to γ-radiation ameliorated these parameters and improved the histopathological changes induced by γ-radiation.Conclusions: The results demonstrated that PRP and/or LMC inhibited γ-radiation-induced hepatotoxicity and using both of them together seems more effective. They can be a candidate to be studied toward the development of a therapeutic strategy for liver diseases.
Collapse
Affiliation(s)
- Mostafa Saif-Elnasr
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Salma M Abdel Fattah
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Hesham M Swailam
- Radiation Microbiology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
21
|
Li S, Fujino M, Takahara T, Li XK. Protective role of heme oxygenase-1 in fatty liver ischemia-reperfusion injury. Med Mol Morphol 2019; 52:61-72. [PMID: 30171344 PMCID: PMC6542780 DOI: 10.1007/s00795-018-0205-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022]
Abstract
Ischemia-reperfusion (IR) injury is a kind of injury resulting from the restoration of the blood supply after blood vessel closure during liver transplantation and is the main cause of graft failure. The pathophysiological mechanisms of hepatic IR include a variety of oxidative stress responses. Hepatic IR is characterized by ischemia and hypoxia inducing oxidative stress, immune response and apoptosis. Fat-denatured livers are also used as donors due to the lack of liver donors. Fatty liver is less tolerant to IR than normal liver. Heme oxygenase (HO) is an enzyme that breaks down hemoglobin to bilirubin, ferrous iron and carbon monoxide (CO). Inducible HO subtype HO-1 is an important protective molecule in mammalian cells used to improve acute and chronic liver injury owing to its characteristic anti-inflammatory and anti-apoptotic qualities. HO-1 degrades heme, and its reaction product CO has been shown to reduce hepatic IR injury and increase the survival rate of grafts. As an induced form of HO, HO-1 also exerts a protective effect against liver IR injury and may be useful as a new strategy of ameliorating this kind of damage. This review summarizes the protective effects of HO-1 in liver IR injury, especially in fatty liver.
Collapse
Affiliation(s)
- Shaowei Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Terumi Takahara
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan.
| |
Collapse
|
22
|
Rossaint J, Margraf A, Zarbock A. Role of Platelets in Leukocyte Recruitment and Resolution of Inflammation. Front Immunol 2018; 9:2712. [PMID: 30515177 PMCID: PMC6255980 DOI: 10.3389/fimmu.2018.02712] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/02/2018] [Indexed: 12/30/2022] Open
Abstract
Platelets are most often recognized for their crucial role in the control of acute hemorrhage. However, current research has greatly expanded the appreciation of platelets beyond their contribution to primary hemostasis, indicating that platelets also actively participate in leukocyte recruitment and the regulation of the host defense in response to exogenous pathogens and sterile injury. Early recruitment of leukocytes, especially neutrophils, is the evolutionary stronghold of the innate immune response to successfully control exogenous infections. Platelets have been shown to physically interact with different leukocyte subsets during inflammatory processes. This interaction holds far-reaching implications for the leukocyte recruitment into peripheral tissues as well as the regulation of leukocyte cell autonomous functions, including the formation and liberation of neutrophil extracellular traps. These functions critically depend on the interaction of platelets with leukocytes. The host immune response and leukocyte recruitment must be tightly regulated to avoid excessive tissue and organ damage and to avoid chronification of inflammation. Thus, platelet-leukocyte interactions and the resulting leukocyte activation and recruitment also underlies tight regulation by several inherited feedback mechanisms to limit the extend of vascular inflammation and to protect the host from collateral damage caused by overshooting immune system activation. After the acute inflammatory phase has been overcome the host defense response must eventually be terminated to allow for resolution from inflammation and restoration of tissue and organ function. Besides their essential role for leukocyte recruitment and the initiation and propagation of vascular inflammation, platelets have lately also been implicated in the resolution process. Here, their contribution to phagocyte clearance, T cell recruitment and macrophage reprogramming is also of outmost importance. This review will focus on the role of platelets in leukocyte recruitment during the initiation of the host defense and we will also discuss the participation of platelets in the resolution process after acute inflammation.
Collapse
Affiliation(s)
- Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany.,Interdisciplinary Centre for Clinical Research, University Hospital Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
23
|
Chae MS, Lee N, Choi HJ, Chung HS, Park CS, Lee J, Choi JH, Hong SH. Comparison of Liver Graft Regeneration Between ABO-Compatible and ABO-Incompatible Living Donor Liver Transplantation: A Propensity Score Matching Analysis. Ann Transplant 2018; 23:507-519. [PMID: 30050031 PMCID: PMC6248061 DOI: 10.12659/aot.908787] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND ABO-incompatible (ABOi) living donor liver transplantation (LDLT) was accepted as a feasible therapy for end-stage liver disease after the introduction of rituximab. The present study investigated the association between ABO incompatibility and graft regeneration in patients who underwent LDLT. MATERIAL AND METHODS A total of 335 adult patients who underwent elective LDLT were divided into ABO-compatible (ABOc) and ABOi LDLT groups using propensity score (PS) matching of graft regeneration-related factors. Postoperative serial changes in graft volumes were compared between the groups. The factors associated with graft volume on postoperative day (POD) 21 were investigated in patients who underwent ABOi LDLT. RESULTS In total, 300 (89.6%) patients underwent ABOc LDLT and 35 (10.4%) patients underwent ABOi LDLT. After PS matching, the ABOc and ABOi groups each included 32 paired patients. The absolute liver graft volumes on POD 21 were significantly lower in the ABOi group than those in the ABOc group in the PS-matched patients (1098.4 [964.0-1,162.0] vs. 1202.0 [1107.8-1455.2] mL; p=0.007). Major complications, including overall patient mortality during the follow-up period, did not differ between the groups. In patients who underwent ABOi LDLT, the preoperative graft volume/standard liver volume ratio and CD4+ cell level on POD 14 were independent factors related to liver graft volume on POD 21. CONCLUSIONS These results suggest that ABO incompatibility could affect postoperative liver graft regeneration. Therefore, graft regeneration must be investigated using a volumetric assessment in patients who have undergone ABOi LDLT.
Collapse
Affiliation(s)
- Min Suk Chae
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Nuri Lee
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyun Sik Chung
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Chul Soo Park
- Anesthesiology and pain medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, , South Korea
| | - Jaemin Lee
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jong Ho Choi
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sang Hyun Hong
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
24
|
The posttraumatic activation of CD4+ T regulatory cells is modulated by TNFR2- and TLR4-dependent pathways, but not by IL-10. Cell Immunol 2018; 331:137-145. [PMID: 29954581 DOI: 10.1016/j.cellimm.2018.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/10/2018] [Accepted: 06/18/2018] [Indexed: 12/21/2022]
Abstract
Platelets modulate the immune system following injury by interacting with CD4+ T regulatory cells (CD4+ Tregs). The underlying mechanisms remain unsolved. We hypothesize paracrine interactions via Tumor necrosis factor-alpha (TNFα)-, Toll like receptor-4 (TLR4)-, and Interleukin-10 (IL-10). In the murine burn injury model, CD4+ Treg activation pathways were selectively addressed using TNFR2-, TLR4- and IL-10-deficient mice. The CD4+ Treg signalling molecule PKC-θ was analyzed using phospho-flow cytometry to detect rapid cell activation. Thromboelastometry (ROTEM®) was used to assess platelet activation. Injury induced significant early activation of CD4+ Tregs, disruption of TNFR2 and TLR4 activation pathways resulted in lower activity. The disruption of IL-10 crosstalk had no significant impact. Selective disruption of paracrine interactions is associated with changes in posttraumatic hemostasis parameters. TNFR2- and TLR4-dependent pathways modulate the activation of CD4+ Tregs following trauma. In contrast, we did not observe a role of IL-10 in the posttraumatic activation of CD4+ Tregs. ONE SENTENCE SUMMARY TLR4- and TNFR2-dependent mechanisms, but not IL-10-dependent pathways, modulate the anti-inflammatory response of CD4+ Tregs following trauma.
Collapse
|
25
|
Abstract
Platelets are key players in thrombosis and hemostasis. Alterations in platelet count and function are common in liver disease, and may contribute to bleeding or thrombotic complications in liver diseases and during liver surgery. In addition to their hemostatic function, platelets may modulate liver diseases by mechanisms that are incompletely understood. Here, we present clinical evidence for a role of platelets in the progression of chronic and acute liver diseases, including cirrhosis, acute liver failure, and hepatocellular carcinoma. We also present clinical evidence that platelets promote liver regeneration following partial liver resection. Subsequently, we summarize studies in experimental animal models that support these clinical observations, and also highlight studies that are in contrast with clinical observations. The combined results of clinical and experimental studies suggest that platelets may be a therapeutic target in the treatment of liver injury and repair, but the gaps in our understanding of mechanisms involved in platelet-mediated modulation of liver diseases call for caution in clinical application of these findings.
Collapse
Affiliation(s)
- Ton Lisman
- Section of Hepatobiliary Surgery and Liver Transplantation and Surgical Research Laboratory, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - James P. Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
26
|
Finsterbusch M, Schrottmaier WC, Kral-Pointner JB, Salzmann M, Assinger A. Measuring and interpreting platelet-leukocyte aggregates. Platelets 2018; 29:677-685. [PMID: 29461910 PMCID: PMC6178087 DOI: 10.1080/09537104.2018.1430358] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelets, besides their specialised role in haemostasis and atherothrombosis, actively modulate innate and adaptive immune responses with crucial roles in immune surveillance, inflammation and host defence during infection. An important prerequisite for platelet-mediated changes of immune functions involves direct engagement with different types of leukocytes. Indeed, increased platelet-leukocyte aggregates (PLAs) within the circulation and/or locally at the site of inflammation represent markers of many thrombo-inflammatory diseases, such as cardiovascular diseases, acute lung injury, renal and cerebral inflammation. Therefore, measurement of PLAs could provide an attractive and easily accessible prognostic and/or diagnostic tool for many diseases. To measure PLAs in different (patho-)physiological settings in human and animal models flow cytometric and microscopic approaches have been applied. These techniques represent complementary tools to study different aspects relating to the involvement of leukocyte subtypes and molecules, as well as location of PLAs within tissues, dynamics of their interactions and/or dynamic changes in leukocyte and platelet behaviour. This review summarises various approaches to measure and interpret PLAs and discusses potential experimental factors influencing platelet binding to leukocytes. Furthermore, we summarise insights gained from studies regarding the underlying mechanism of platelet-leukocyte interactions and discuss implications of these interactions in health and disease.
Collapse
Affiliation(s)
- Michaela Finsterbusch
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Waltraud C Schrottmaier
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Julia B Kral-Pointner
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Manuel Salzmann
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Alice Assinger
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| |
Collapse
|
27
|
Abstract
Hepatic ischemia/reperfusion (I/R) injury is a major complication of liver surgery, including liver resection, liver transplantation, and trauma surgery. Much has been learned about the inflammatory injury response induced by I/R, including the cascade of proinflammatory mediators and recruitment of activated leukocytes. In this review, we discuss the complex network of events that culminate in liver injury after I/R, including cellular, protein, and molecular mechanisms. In addition, we address the known endogenous regulatory mediators that function to maintain homeostasis and resolve injury. Finally, we cover more recent insights into how the liver repairs and regenerates after I/R injury, a setting in which physical mass remains unchanged, but functional liver mass is greatly reduced. In this regard, we focus on recent work highlighting a novel role of CXC chemokines as important regulators of hepatocyte proliferation and liver regeneration after I/R injury.
Collapse
Affiliation(s)
- Takanori Konishi
- Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Alex B. Lentsch
- Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
28
|
Kolachala VL, Palle S, Shen M, Feng A, Shayakhmetov D, Gupta NA. Loss of L-selectin-guided CD8 + , but not CD4 + , cells protects against ischemia reperfusion injury in a steatotic liver. Hepatology 2017; 66:1258-1274. [PMID: 28543181 PMCID: PMC5605411 DOI: 10.1002/hep.29276] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 04/30/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022]
Abstract
UNLABELLED Steatotic liver responds with increased hepatocellular injury when exposed to an ischemic-reperfusion insult. Increasing evidence supports the role of immune cells as key mediators of this injury in a normal (lean) state, but data about their role in a steatotic liver are practically nonexistent. The objective of the current study was to delineate the contribution of specific phenotypes of T cells and adhesion molecules in exacerbated cell death in steatotic liver injury. RNA sequencing was performed on isolated steatotic primary hepatocytes, and T-cell markers were assessed in hepatic lymphocytes after ischemia reperfusion injury (IRI) in high-fat diet (HFD)-fed mice. Cluster of differentiation 8 knockout (CD8-/- ) and CD4-/- mice along with CD8 and L-selectin antibody-treated mice were fed an HFD, and hepatocellular injury was assessed by histology, propidium iodide injection, and alanine aminotransferase after IRI. RNA sequencing demonstrated a strikingly differential gene profile in steatotic hepatocytes versus lean hepatocytes. After injury, the HFD liver showed increased necrosis, infiltrating CD8+ cells, alanine aminotransferase, and proinflammatory cytokines. Hepatic lymphocytes demonstrated increased CD8+ /CD62L+ (L-selectin) cells in HFD-fed mice after IRI. CD8-/- mice and CD8-depleted C57BL/6 mice demonstrated significant protection from injury, which was not seen in CD4-/- mice. L-selectin blockade also demonstrated significant hepatoprotection from IRI. L-selectin ligand MECA-79 was increased in HFD-fed mice undergoing IRI. CONCLUSION Blockade of CD8 and L-selectin, but not CD4, ameliorated hepatocellular injury, confirming that CD8+ cells are critical drivers of injury in a steatotic liver; this represents a therapeutic target in steatotic liver injury, underlining the importance of development of therapies specific to a steatotic liver. (Hepatology 2017;66:1258-1274).
Collapse
Affiliation(s)
| | - Sirish Palle
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Ming Shen
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Alayna Feng
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | | | - Nitika A. Gupta
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA,Transplant services, Children’s Healthcare of Atlanta. Atlanta, GA
| |
Collapse
|
29
|
Xu D, Chen L, Chen X, Wen Y, Yu C, Yao J, Wu H, Wang X, Xia Q, Kong X. The triterpenoid CDDO-imidazolide ameliorates mouse liver ischemia-reperfusion injury through activating the Nrf2/HO-1 pathway enhanced autophagy. Cell Death Dis 2017; 8:e2983. [PMID: 28796242 PMCID: PMC5596572 DOI: 10.1038/cddis.2017.386] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 02/07/2023]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated induction of antioxidants has been implicated to have protective roles in ischemia-reperfusion (I/R) injury in many animal models. However, the in vivo effects of CDDO-imidazole (CDDO-Im) (1-[2-cyano-3-,12-dioxooleana-1,9(11)-dien-28-oyl] imidazole), a Nrf2 activator, in hepatic I/R injury is lacking and its exact molecular mechanisms are still not very clear. The goals of this study were to determine whether CDDO-Im can prevent liver injury induced by I/R in the mouse, and to elucidate the molecular target of drug action. Mice were randomly equally divided into two groups and administered intraperitoneally with either DMSO control or CDDO-Im (2 mg/kg) 3 h before subjected to 90-min hepatic 70% ischemia followed by reperfusion. Subsequently, the Liver and blood samples of these mice were collected to evaluate liver injury. CDDO-Im pretreatment markedly improve hepatic I/R injury by attenuating hepatic necrosis and apoptosis, reducing reactive oxygen species (ROS) levels and inflammatory responses, and ameliorating mitochondrial dysfunction. Mechanistically, by using Nrf2 Knockout mice and hemeoxygenase 1 (HO-1) inhibitor, we found that these CDDO-Im protection effects are attributed to enhanced autophagy, which is mediated by activating Nrf2/HO-1 pathway. By accelerating autophagy and clearance of damaged mitochondria, CDDO-Im reduced the mtDNA release and ROS overproduction, and in turn decreased damage-associated molecular patterns induced inflammatory responses and the following secondary liver injury. These results indicate that by enhancing autophagy, CDDO-Im-mediated activation of Nrf2/HO-1 signaling could be a novel therapeutic strategy to minimize the adverse effects of hepatic I/R injury.
Collapse
Affiliation(s)
- Dongwei Xu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lili Chen
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaosong Chen
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yankai Wen
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Chang Yu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jufang Yao
- Animal Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hailong Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xin Wang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoni Kong
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
30
|
NKT cells are important mediators of hepatic ischemia-reperfusion injury. Transpl Immunol 2017; 45:15-21. [PMID: 28797737 PMCID: PMC5694034 DOI: 10.1016/j.trim.2017.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/04/2017] [Accepted: 08/05/2017] [Indexed: 12/16/2022]
Abstract
Introduction IRI results from the interruption then reinstatement of an organ's blood supply, and this poses a significant problem in liver transplantation and resectional surgery. In this paper, we explore the role T cells play in the pathogenesis of this injury. Materials & methods We used an in vivo murine model of warm partial hepatic IRI, genetically-modified mice, in vivo antibody depletion, adoptive cell transfer and flow cytometry to determine which lymphocyte subsets contribute to pathology. Injury was assessed by measuring serum alanine aminotransfersase (ALT) and by histological examination of liver tissue sections. Results The absence of T cells (CD3εKO) is associated with significant protection from injury (p = 0.010). Through a strategy of antibody depletion it appears that NKT cells (p = 0.0025), rather than conventional T (CD4 + or CD8 +) (p = 0.11) cells that are the key mediators of injury. Discussion Our results indicate that tissue-resident NKT cells, but not other lymphocyte populations are responsible for the injury in hepatic IRI. Targeting the activation of NKT cells and/or their effector apparatus would be a novel approach in protecting the liver during transplantation and resection surgery; this may allow us to expand our current criteria for surgery. Hepatic IRI worsens outcome in liver transplantation. T cells are important in hepatic IRI. These are tissue-resident rather than recruited T cells. NKT, but not conventional T or NK cells, are key mediators of hepatic IRI. Targeting NKT activation or their effector apparatus may offer therapeutic potential.
Collapse
|
31
|
Funken D, Ishikawa-Ankerhold H, Uhl B, Lerchenberger M, Rentsch M, Mayr D, Massberg S, Werner J, Khandoga A. In situ targeting of dendritic cells sets tolerogenic environment and ameliorates CD4 + T-cell response in the postischemic liver. FASEB J 2017; 31:4796-4808. [PMID: 28720647 DOI: 10.1096/fj.201601358r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 07/05/2017] [Indexed: 12/27/2022]
Abstract
CD4+ T cells recruited to the liver play a key role in the pathogenesis of ischemia/reperfusion (I/R) injury. The mechanism of their activation during alloantigen-independent I/R is not completely understood. We hypothesized that liver-resident dendritic cells (DCs) interact with CD4+ T cells in the postischemic liver and that modulation of DCs or T-cell-DC interactions attenuates liver inflammation. In mice, warm hepatic I/R (90/120-240 min) was induced. Tolerogenic DCs were generated in situ by pretreatment of animals with the vitamin D analog paricalcitol. A mAb-CD44 was used for blockade of CD4+ T-cell-DC interactions. As shown by 2-photon in vivo microscopy as well as confocal microscopy, CD4+ T cells were closely colocalized with DCs in the postischemic liver. Pretreatment with paricalcitol attenuated I/R-induced maturation of DCs (flow cytometry), CD4+ T-cell recruitment into the liver (intravital microscopy), and hepatocellular/microvascular damage (intravital microscopy, alanine aminotransferase/aspartate aminotransferase, histology). However, interruption of T-cell-DC interaction increased proinflammatory DC maturation and even enhanced tissue damage. Simultaneous treatment with an anti-CD44mAb completely abolished the beneficial effect of paricalcitol on T-cell migration and tissue injury. Our study demonstrates for the first time that hepatic DCs interact with CD4+ T cells in the postischemic liver in vivo; modulation of DCs and/or generation of tolerogenic DCs attenuates intrahepatic CD4+ T-cell recruitment and reduces I/R injury; and interruption of CD44-dependent CD4+ T-cell-DC interactions enhances tissue injury by preventing the modulatory effect of hepatic DCs on T cells, especially type 1 T helper effector cells. Thus, hepatic DCs are strongly involved in the promotion of CD4+ T-cell-dependent postischemic liver inflammation.-Funken, D., Ishikawa-Ankerhold, H., Uhl, B., Lerchenberger, M., Rentsch, M., Mayr, D., Massberg, S., Werner, J., Khandoga, A. In situ targeting of dendritic cells sets tolerogenic environment and ameliorates CD4+ T-cell response in the postischemic liver.
Collapse
Affiliation(s)
- Dominik Funken
- Department of General, Visceral, and Transplant Surgery, University of Munich, Munich, Germany
| | - Hellen Ishikawa-Ankerhold
- Walter-Brendel Centre of Experimental Medicine, University of Munich, Munich, Germany.,Department of Cardiology, University of Munich, Munich, Germany
| | - Bernd Uhl
- Walter-Brendel Centre of Experimental Medicine, University of Munich, Munich, Germany
| | | | - Markus Rentsch
- Department of General, Visceral, and Transplant Surgery, University of Munich, Munich, Germany
| | - Doris Mayr
- Department of Pathology, University of Munich, Munich, Germany
| | | | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, University of Munich, Munich, Germany
| | - Andrej Khandoga
- Department of General, Visceral, and Transplant Surgery, University of Munich, Munich, Germany;
| |
Collapse
|
32
|
Sawada T, Inoue K, Tanabe D, Kawamoto S, Tsuji T, Tashiro S. Experimental Studies on Protective Effects of FK506 Against Hepatic Ischemia-Reperfusion Injury. THE JOURNAL OF MEDICAL INVESTIGATION 2017; 63:262-9. [PMID: 27644569 DOI: 10.2152/jmi.63.262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Purposes; FK506 (strong immunosuppressive agent) was investigated experimentally whether to protect the hepatic IRI. Methods; Warm ischemic experiment using pigs and rats were performed and examined whether FK506 is effective. Results; The results obtained are as follows. 1. Warm ischemia allowed time of the pigs without FK506 was 150 minutes, but as for that of FK506 group, the extension of 30 minutes was got in 180 minutes. 2. Biliary excretion rate of BSP after reperfusion were better in the group of 180 minutes ischemia with FK506 than in without FK506 group. 3. Chemiluminescence intensity in the peripheral neutrophils and adhered and infiltrated leukocytes in the liver were suppressed markedly by FK506. 4. The vascular endothelium with the scanning electron microscope was relatively preserved in the FK506 group comparing to the placebo group on 30 minutes after reperfusion. 5. Stress gastric ulcer was controlled and myeloperoxidase activity in the gastric mucosa was suppressed by FK506. Conclusion; Based on the results of theses experiments, it was suggested that FK506 has a protective effect on IRI by suppressing: the impairment of sinusoidal endothelial cells; the activation of KCs; the disturbance of micro-circulation; oxidative stress; inflammation; and the accumulation of leukocytes. J. Med. Invest. 63: 262-269, August, 2016.
Collapse
|
33
|
Zimmerman MA, Martin A, Yee J, Schiller J, Hong JC. Natural Killer T Cells in Liver Ischemia-Reperfusion Injury. J Clin Med 2017; 6:jcm6040041. [PMID: 28368299 PMCID: PMC5406773 DOI: 10.3390/jcm6040041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/10/2017] [Accepted: 03/20/2017] [Indexed: 02/06/2023] Open
Abstract
Restoration of blood flow to an ischemic organ results in significant tissue injury. In the field of liver transplantation, ischemia–reperfusion injury (IRI) has proven to be a formidable clinical obstacle. In addition to metabolic stress and inflammation, IRI results in profound graft dysfunction and loss. The severity of IRI further limits the ability to expand the donor pool by using partial grafts and marginal organs. As such, the inflammatory response to reperfusion of the liver continues to be an area of intense investigation. Among the various leukocytes involved in IRI, new insights suggest that natural killer T (NKT) cells may be a central driver of hepatocellular injury. Herein, we examine recent experimental observations that provide a mechanistic link between NKT cell recruitment to liver and post-perfusion tissue injury.
Collapse
Affiliation(s)
- Michael A Zimmerman
- Department of Surgery, Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Alicia Martin
- Department of Surgery, Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Jennifer Yee
- Department of Surgery, Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Jennifer Schiller
- Department of Surgery, Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Histocompatibility and Immunogenetics, Blood Center of Wisconsin, Milwaukee, WI 53201, USA.
| | - Johnny C Hong
- Department of Surgery, Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
34
|
Brown DL. Immunopathology of the Hepatobiliary System. MOLECULAR AND INTEGRATIVE TOXICOLOGY 2017:329-417. [DOI: 10.1007/978-3-319-47385-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
35
|
Platelets modulate the immune response following trauma by interaction with CD4+ T regulatory cells in a mouse model. Immunol Res 2016; 64:508-17. [PMID: 26471021 DOI: 10.1007/s12026-015-8726-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
CD4+ T regulatory cells (Tregs) play a pivotal role in the anti-inflammatory immune response following trauma. The mechanisms of CD4+ Treg activation are mostly unknown. Here, we hypothesize that platelets regulate CD4+ Treg activation following trauma. In a murine burn injury model (male C57Bl/6N mice), depletion of platelets or CD4+ Tregs was conducted. Draining lymph nodes, blood and spleen were harvested 2 h and 7 days after trauma. CD4+ Treg activation was measured using phospho- and conventional flow cytometry. Platelet activation was analyzed using thromboelastometry and flow cytometry. Trauma differentially activates CD4+ T cells, early after trauma only CD4+ Tregs are activated. Following burn injury, platelets augment the activation of CD4+ Tregs. This effect could only be seen early after trauma. While CD4+ Tregs influence hemostasis early following trauma, platelet activation markers were unchanged. Beyond their role in hemostasis, platelets are able to modulate the immunologic host response to trauma-induced injury by augmenting the activation of CD4+ Tregs. CD4+ Treg activation following trauma is considered protective. In addition, CD4+ Tregs are capable of modulating the hemostatic function of platelets. For the first time, we could show reciprocal activation of platelets and CD4+ Tregs as part of the protective immune response following trauma.
Collapse
|
36
|
Abstract
Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.
Collapse
Affiliation(s)
- Theodore Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Christopher P. Baines
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
37
|
Chauhan A, Adams DH, Watson SP, Lalor PF. Platelets: No longer bystanders in liver disease. Hepatology 2016; 64:1774-1784. [PMID: 26934463 PMCID: PMC5082495 DOI: 10.1002/hep.28526] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 02/17/2016] [Accepted: 02/21/2016] [Indexed: 12/17/2022]
Abstract
UNLABELLED Growing lines of evidence recognize that platelets play a central role in liver homeostasis and pathobiology. Platelets have important roles at every stage during the continuum of liver injury and healing. These cells contribute to the initiation of liver inflammation by promoting leukocyte recruitment through sinusoidal endothelium. They can activate effector cells, thus amplifying liver damage, and by modifying the hepatic cellular and cytokine milieu drive both hepatoprotective and hepatotoxic processes. CONCLUSION In this review we summarize how platelets drive such pleiotropic actions and attempt to reconcile the paradox of platelets being both deleterious and beneficial to liver function; with increasingly novel methods of manipulating platelet function at our disposal, we highlight avenues for future therapeutic intervention in liver disease. (Hepatology 2016;64:1774-1784).
Collapse
Affiliation(s)
- Abhishek Chauhan
- Centre for Liver Research, and NIHR Birmingham Liver Biomedical Research Unit, Institute of Biomedical Research, Birmingham, UK.
| | - David H. Adams
- Centre for Liver Research, and NIHR Birmingham Liver Biomedical Research UnitInstitute of Biomedical ResearchBirminghamUK
| | - Steve P. Watson
- Institute for Cardiovascular Sciences, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Patricia F. Lalor
- Centre for Liver Research, and NIHR Birmingham Liver Biomedical Research UnitInstitute of Biomedical ResearchBirminghamUK
| |
Collapse
|
38
|
Pratschke S, Arnold H, Zollner A, Heise M, Pascher A, Schemmer P, Scherer MN, Bauer A, Jauch KW, Werner J, Guba M, Angele MK. Results of the TOP Study: Prospectively Randomized Multicenter Trial of an Ex Vivo Tacrolimus Rinse Before Transplantation in EDC Livers. Transplant Direct 2016; 2:e76. [PMID: 27500266 PMCID: PMC4946517 DOI: 10.1097/txd.0000000000000588] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Organ shortage results in the transplantation of extended donor criteria (EDC) livers which is associated with increased ischemia-reperfusion injury (IRI). Experimental studies indicate that an organ rinse with the calcineurin inhibitor tacrolimus before implantation protects against IRI. The tacrolimus organ perfusion study was initiated to examine the effects of ex vivo tacrolimus perfusion on IRI in transplantation of EDC livers. METHODS A prospective randomized multicenter trial comparing ex vivo perfusion of marginal liver grafts (≥2 EDC according to Eurotransplant manual) with tacrolimus (20 ng/mL) or histidine-tryptophane-ketoglutarate solution (control) was carried out at 5 German liver transplant centers (Munich Ludwig-Maximilians University, Berlin, Heidelberg, Mainz, Regensburg) between October 2011 and July 2013. Primary endpoint was the maximum alanine transaminase (ALT) level within 48 hours after transplantation. Secondary endpoints were aspartate transaminase (AST), prothrombine ratio, and graft-patient survival within an observation period of 1 week. After an interim analysis, the study was terminated by the scientific committee after the treatment of 24 patients (tacrolimus n = 11, Control n = 13). RESULTS Tacrolimus rinse did not reduce postoperative ALT peaks compared with control (P = 0.207; tacrolimus: median, 812; range, 362-3403 vs control: median, 652; range, 147-2034). Moreover, ALT (P = 0.100), prothrombine ratio (P = 0.553), and bilirubin (P = 0.815) did not differ between the groups. AST was higher in patients treated with tacrolimus (P = 0.011). Survival was comparable in both groups (P > 0.05). CONCLUSIONS Contrary to experimental findings, tacrolimus rinse failed to improve the primary endpoint of the study (ALT). Because 1 secondary endpoint (AST) was even higher in the intervention group, the study was terminated prematurely. Thus, tacrolimus rinse cannot be recommended in transplantation of EDC livers.
Collapse
Affiliation(s)
- Sebastian Pratschke
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Hannah Arnold
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Alfred Zollner
- Münchner Studienzentrum, University Hospital Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael Heise
- Department of General, Visceral and Transplantation Surgery, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Andreas Pascher
- Department of General, Visceral and Transplantation Surgery, Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Peter Schemmer
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Ruprecht-Karls-University, Heidelberg, Germany
| | - Marcus N Scherer
- Department of Surgery, University Hospital Regensburg, University of Regensburg, Regensburg, Germany
| | - Andreas Bauer
- Department of Anaesthesiology, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Karl-Walter Jauch
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Markus Guba
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Martin K Angele
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
39
|
Model for end-stage liver disease score in the first 3 weeks after liver transplantation as a predictor for long-term outcome. Eur J Gastroenterol Hepatol 2016; 28:153-8. [PMID: 26545081 DOI: 10.1097/meg.0000000000000505] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Early allograft dysfunction after liver transplantation (LTX) is not well defined. The aim of this study was to evaluate the value of early post-transplant model for end-stage liver disease (MELD) scores for predicting long-term outcome after transplantation. METHODS In this single-center retrospective study, 362 consecutive patients after LTX were included. MELD scores at 7, 14, and 21 postoperative days (PODs) were calculated from primary lab values. Receiver operating characteristic (ROC) analyses were carried out to determine the critical cutoff MELD scores for patient and graft survival. RESULTS One year after transplantation, the patient and graft survival rates were 85 and 69%, respectively. Although pretransplant MELD scores were similar, they were significantly different at POD7, POD14, and POD21 between patients who died and those who survived the first year after transplantation. As shown by ROC curves, for patient survival, the optimal time point is POD14 with a cutoff MELD of 17. At this time point, patients with a MELD below 17 showed a 1-year survival rate of 94.3% and patients with a MELD of 17 and higher showed a 1-year survival rate of only 75.4%. For graft survival, the optimal time point was day 7 and a cutoff MELD of 29 (92% at MELD<29; 56.4% at MELD≥29). A multivariate analysis of potential risk factors indicated a significant role of serum bilirubin and MELD score determined on POD14 for patient survival. CONCLUSION In conclusion, early postoperative MELD scores predict outcome after LTX. The postoperative MELD score at POD14 is a good predictor for patient survival and at POD7 for the graft survival after LTX.
Collapse
|
40
|
Richards JA, Bucsaiova M, Hesketh EE, Ventre C, Henderson NC, Simpson K, Bellamy COC, Howie SEM, Anderton SM, Hughes J, Wigmore SJ. Acute Liver Injury Is Independent of B Cells or Immunoglobulin M. PLoS One 2015; 10:e0138688. [PMID: 26406765 PMCID: PMC4583453 DOI: 10.1371/journal.pone.0138688] [Citation(s) in RCA: 7] [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: 06/15/2015] [Accepted: 09/02/2015] [Indexed: 01/08/2023] Open
Abstract
Background & Aims Acute liver injury is a clinically important pathology and results in the release of Danger Associated Molecular Patterns, which initiate an immune response. Withdrawal of the injurious agent and curtailing any pathogenic secondary immune response may allow spontaneous resolution of injury. The role B cells and Immunoglobulin M (IgM) play in acute liver injury is largely unknown and it was proposed that B cells and/or IgM would play a significant role in its pathogenesis. Methods Tissue from 3 models of experimental liver injury (ischemia-reperfusion injury, concanavalin A hepatitis and paracetamol-induced liver injury) and patients transplanted following paracetamol overdose were stained for evidence of IgM deposition. Mice deficient in B cells (and IgM) were used to dissect out the role B cells and/or IgM played in the development or resolution of injury. Serum transfer into mice lacking IgM was used to establish the role IgM plays in injury. Results Significant deposition of IgM was seen in the explanted livers of patients transplanted following paracetamol overdose as well as in 3 experimental models of acute liver injury (ischemia-reperfusion injury, concanavalin A hepatitis and paracetamol-induced liver injury). Serum transfer into IgM-deficient mice failed to reconstitute injury (p = 0.66), despite successful engraftment of IgM. Mice deficient in both T and B cells (RAG1-/-) mice (p<0.001), but not B cell deficient (μMT) mice (p = 0.93), were significantly protected from injury. Further interrogation with T cell deficient (CD3εKO) mice confirmed that the T cell component is a key mediator of sterile liver injury. Mice deficient in B cells and IgM mice did not have a significant delay in resolution following acute liver injury. Discussion IgM deposition appears to be common feature of both human and murine sterile liver injury. However, neither IgM nor B cells, play a significant role in the development of or resolution from acute liver injury. T cells appear to be key mediators of injury. In conclusion, the therapeutic targeting of IgM or B cells (e.g. with Rituximab) would have limited benefit in protecting patients from acute liver injury.
Collapse
Affiliation(s)
- James A. Richards
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
- Clinical Surgery, The University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| | - Martina Bucsaiova
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Emily E. Hesketh
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Chiara Ventre
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Neil C. Henderson
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
- Hepatology, Division of Health Sciences, School of Clinical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Kenneth Simpson
- Hepatology, Division of Health Sciences, School of Clinical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Christopher O. C. Bellamy
- The University of Edinburgh, Edinburgh, United Kingdom
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Sarah E. M. Howie
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen M. Anderton
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jeremy Hughes
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen J. Wigmore
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
- Clinical Surgery, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
41
|
Heymann F, Niemietz PM, Peusquens J, Ergen C, Kohlhepp M, Mossanen JC, Schneider C, Vogt M, Tolba RH, Trautwein C, Martin C, Tacke F. Long term intravital multiphoton microscopy imaging of immune cells in healthy and diseased liver using CXCR6.Gfp reporter mice. J Vis Exp 2015:52607. [PMID: 25866988 PMCID: PMC4401387 DOI: 10.3791/52607] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Liver inflammation as a response to injury is a highly dynamic process involving the infiltration of distinct subtypes of leukocytes including monocytes, neutrophils, T cell subsets, B cells, natural killer (NK) and NKT cells. Intravital microscopy of the liver for monitoring immune cell migration is particularly challenging due to the high requirements regarding sample preparation and fixation, optical resolution and long-term animal survival. Yet, the dynamics of inflammatory processes as well as cellular interaction studies could provide critical information to better understand the initiation, progression and regression of inflammatory liver disease. Therefore, a highly sensitive and reliable method was established to study migration and cell-cell-interactions of different immune cells in mouse liver over long periods (about 6 hr) by intravital two-photon laser scanning microscopy (TPLSM) in combination with intensive care monitoring. The method provided includes a gentle preparation and stable fixation of the liver with minimal perturbation of the organ; long term intravital imaging using multicolor multiphoton microscopy with virtually no photobleaching or phototoxic effects over a time period of up to 6 hr, allowing tracking of specific leukocyte subsets; and stable imaging conditions due to extensive monitoring of mouse vital parameters and stabilization of circulation, temperature and gas exchange. To investigate lymphocyte migration upon liver inflammation CXCR6.gfp knock-in mice were subjected to intravital liver imaging under baseline conditions and after acute and chronic liver damage induced by intraperitoneal injection(s) of carbon tetrachloride (CCl4). CXCR6 is a chemokine receptor expressed on lymphocytes, mainly on Natural Killer T (NKT)-, Natural Killer (NK)- and subsets of T lymphocytes such as CD4 T cells but also mucosal associated invariant (MAIT) T cells1. Following the migratory pattern and positioning of CXCR6.gfp+ immune cells allowed a detailed insight into their altered behavior upon liver injury and therefore their potential involvement in disease progression.
Collapse
Affiliation(s)
- Felix Heymann
- Department of Medicine III, RWTH University-Hospital Aachen;
| | | | | | - Can Ergen
- Department of Medicine III, RWTH University-Hospital Aachen
| | | | | | | | - Michael Vogt
- IZKF Aachen Core Facility "Two-Photon Imaging", RWTH University-Hospital Aachen
| | - Rene H Tolba
- Institute for Laboratory Animal Science & Experimental Surgery, RWTH Aachen University
| | | | | | - Frank Tacke
- Department of Medicine III, RWTH University-Hospital Aachen;
| |
Collapse
|
42
|
Modulating CD4+ T cell migration in the postischemic liver: hepatic stellate cells as new therapeutic target? Transplantation 2015; 99:41-7. [PMID: 25360872 DOI: 10.1097/tp.0000000000000461] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND CD4+ T cells play a critical role during hepatic ischemia-reperfusion (I/R) injury although the mechanisms of their migration in the postischemic liver remain unclear. We answered the questions of whether hepatic stellate cells (HSCs) interact with CD4+ T cells during I/R of the liver and whether modulation of HSC activity affects T cell-dependent I/R injury. METHODS In mice, migration of CD4+ T cells was analyzed in vivo using conventional intravital microscopy and two-photon microscopy. CD4+ T cell-HSC interactions were visualized after infusion of fluorescence-labeled CD4+ T cells into Cx3CR1 mice (mice exhibiting GFP-labeled HSCs) after I/R. Because the activation of HSC is controlled by endocannabinoid receptors, CB-1 and CB-2, the mice received treatment before I/R with the CB-2 agonist JWH-133 to reach HSC depletion or the CB-1 agonist arachidonylcyclopropylamide to activate HSCs. Sinusoidal perfusion and liver transaminases were used as markers of I/R injury. RESULTS Hepatic I/R induced CD4+ T cell recruitment in sinusoids. More than 25% of adherent CD4+ T cells were colocalized with HSCs during reperfusion, suggesting a direct cell-cell interaction. The HSC deactivation with JWH-133 significantly attenuated the CD4+ T cell recruitment in the postischemic liver and reduced I/R injury as compared to the vehicle-treated group. The HSC hyperactivation by CB-1, however, did not affect T-cell migration and even increased perfusion failure. CONCLUSION Our in vivo data suggest that CD4+ T cells interact with HSCs on their migration into the hepatic parenchyma, and a depletion or deactivation of HSCs protects the liver from T cell-dependent I/R injury.
Collapse
|
43
|
Chin JL, Hisamuddin SH, O'Sullivan A, Chan G, McCormick PA. Thrombocytopenia, Platelet Transfusion, and Outcome Following Liver Transplantation. Clin Appl Thromb Hemost 2014; 22:351-60. [PMID: 25430936 DOI: 10.1177/1076029614559771] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Thrombocytopenia affects patients undergoing liver transplantation. Intraoperative platelet transfusion has been shown to independently influence survival after liver transplantation at 1 and 5 years. We examined the impact of thrombocytopenia and intraoperative platelet transfusion on short-term graft and overall survival after orthotopic liver transplantation (OLT). A total of 399 patients undergoing first OLT were studied. Graft and overall survival in patients with different degrees of thrombocytopenia and with or without intraoperative platelet transfusion were described. The degree of thrombocytopenia prior to OLT did not affect graft or overall survival after transplant. However, graft survival in patients receiving platelets was significantly reduced at 1 year (P= .023) but not at 90 days (P= .093). Overall survival was significantly reduced at both 90 days (P= .040) and 1 year (P= .037) in patients receiving platelets. We conclude that a consistently lower graft and overall survival were observed in patients receiving intraoperative platelet transfusion.
Collapse
Affiliation(s)
- Jun Liong Chin
- Liver Unit, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | | | - Aoife O'Sullivan
- Blood bank, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Grace Chan
- Liver Unit, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - P Aiden McCormick
- Liver Unit, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| |
Collapse
|
44
|
Khandoga A, Mende K, Iskandarov E, Rosentreter D, Schelcher C, Reifart J, Jauch KW, Thasler WE. Augmenter of liver regeneration attenuates inflammatory response in the postischemic mouse liver in vivo. J Surg Res 2014; 192:187-94. [DOI: 10.1016/j.jss.2014.05.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/02/2014] [Accepted: 05/13/2014] [Indexed: 12/12/2022]
|
45
|
Yang J, Wang X, Song S, Liu F, Fu Z, Wang Q. Near-term anti-CD25 monoclonal antibody administration protects murine liver from ischemia-reperfusion injury due to reduced numbers of CD4+ T cells. PLoS One 2014; 9:e106892. [PMID: 25188007 PMCID: PMC4154778 DOI: 10.1371/journal.pone.0106892] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/03/2014] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND CD4(+) T cell is acknowledged as a key factor in the initiation phase of liver ischemia reperfusion injury. The purpose of current study is to demonstrate the effect of antecedent near-term anti-CD25 monoclonal antibody treatment on IR-induced liver injury by modulation of CD4(+) T cells. METHODS 70% liver warm IR was induced in male C57BL/6 mice after anti-CD25 mAb or non-specific IgG administration. Liver function, histological damage, in vitro Proliferation, FACS, cytokine production, and immunofluorescence were assessed to evaluate the impact of antecedent near-term PC61 treatment on IR-induced liver injury. RESULTS After 70% liver ischemia, mice preconditioned with PC61 displayed significantly preserved liver function as characterized by less histological damage and reduced serum enzymes level. Mechanistic studies revealed that the protection effect of anti-CD25 mAb was associated with ameliorated intrahepatic inflammatory milieu and reduced CD4(+) T lymphocytes as manifested by the decrease of proinflammatory cytokine production (less expression of TNF-α, IFN-γ, IL-2, and IL-6) and the lower CD4/CD8 proportion. CONCLUSIONS Our results provide first line of evidence indicating that near-term treatment with anti-CD25 monoclonal antibody might provide protection for livers against IR-induced injury by reducing CD4(+) T cells, but not influencing functional Treg population. Therefore, our results demonstrate a potential function of anti-CD25 monoclonal antibody which was neglected in the past, and may be helpful in various clinical conditions, particularly in liver and kidney transplantations.
Collapse
Affiliation(s)
- Jinghui Yang
- Department of Organ Transplantation, Shanghai ChangZheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoyu Wang
- Department of Organ Transplantation, Shanghai ChangZheng Hospital, Second Military Medical University, Shanghai, China
| | - Shaohua Song
- Department of Organ Transplantation, Shanghai ChangZheng Hospital, Second Military Medical University, Shanghai, China
| | - Fang Liu
- Department of Organ Transplantation, Shanghai ChangZheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhiren Fu
- Department of Organ Transplantation, Shanghai ChangZheng Hospital, Second Military Medical University, Shanghai, China
| | - Quanxing Wang
- National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, China
| |
Collapse
|
46
|
Gao W, Liu D, Li D, Che X, Cui G. Effects of hypercapnia on T cells in lung ischemia/reperfusion injury after lung transplantation. Exp Biol Med (Maywood) 2014; 239:1597-605. [PMID: 25013165 DOI: 10.1177/1535370214542072] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
T cells play a key role in lung ischemia/reperfusion injury (IRI). Hypercapnia has been indicated to decrease IRI and inhibit immunity. This study aimed to evaluate the effects of hypercapnia on T cells during lung IRI and to identify the underlying mechanism of these effects. In the in vivo study, rat recipients of lung transplants were randomized into a control group M and a hypercapnia group H. Peripheral blood T cells and cytokines were analyzed during reperfusion. In the in vitro study, we analyzed the T cells and cytokine levels in culture media from phytohemagglutinin-stimulated T cells from normal rats, stimulated under the normal (group C), hypercapnic (group H), or buffer hypercapnic (group BH) condition. In the in vivo study, the CD3+/CD4+ T-cell ratio and interleukin (IL)-2, IL-8, interferon (IFN)-γ, intracellular adhesion molecule (ICAM)-1, and P-selectin levels were decreased, but the IL-4 and IL-10 levels were increased, after reperfusion in group H compared to group M. In the in vitro study, groups H and BH exhibited a decreased CD2+/CD28+ ratio and IL-2 and IFN-γ levels, but elevated IL-4 and IL-10 levels, compared to group C. The CD2+/CD28+ ratio was not different between groups BH and H; however, group H evidenced a lower IL-2 level and higher IL-4 and IL-10 levels compared to group BH. Hypercapnia decreased the CD3+/CD4+ T-cell ratio and pro-inflammatory cytokine levels, but promoted anti-inflammatory factors in lung IRI. Hypercapnia inhibits CD2 and CD28 in T cells by CO2 and modulates T-cell cytokines via acidosis.
Collapse
Affiliation(s)
- Wei Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, 150000 Harbin, China
| | - Dongdong Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, 150000 Harbin, China
| | - Di Li
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, 150000 Harbin, China
| | - Xiangyu Che
- Department of Anesthesiology, The Fifth Hospital of Harbin, 150000 Harbin, China
| | - Guangxiao Cui
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, 150000 Harbin, China
| |
Collapse
|
47
|
Targeting platelet migration in the postischemic liver by blocking protease-activated receptor 4. Transplantation 2014; 97:154-60. [PMID: 24434483 DOI: 10.1097/01.tp.0000437430.89485.a0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Platelets play a critical role during hepatic ischemia/reperfusion (I/R). Antiplatelet strategies during liver transplantation are, however, limited because of bleeding complications. Thrombin is activated during reperfusion and regulates platelet and endothelial cell function via protease-activated receptor 4 (PAR-4). Interventions at the level of PAR-4, the main platelet receptor for thrombin, are assumed to attenuate the proinflammatory effects of thrombin without affecting blood coagulation. The aim of our study was to analyze the impact of PAR-4 blockade on platelet recruitment and microvascular injury during hepatic I/R. METHODS C57BL/6 mice undergoing hepatic I/R (90 min/60 min and 240 min) were treated either with a selective PAR-4 antagonist TcY-NH2 or vehicle. Sham-operated animals served as controls. Recruitment of freshly isolated and fluorescence-labeled platelets and CD4 T cells was analyzed using intravital video fluorescence microscopy. Parameters of tissue injury, regeneration, and blood coagulation were assessed in tissue/blood samples. RESULTS Results show that treatment with TcY-NH2 attenuated I/R-induced platelet and CD4 T-cell recruitment, improved sinusoidal perfusion failure, and reduced apoptotic and necrotic injury. The protective effect of PAR-4 blockade did not suppress hemostasis or liver regeneration. CONCLUSION Our in vivo data suggest PAR-4 as a potential target for future therapeutic strategies against platelet-mediated liver injury on transplantation.
Collapse
|
48
|
Murata S, Maruyama T, Nowatari T, Takahashi K, Ohkohchi N. Signal transduction of platelet-induced liver regeneration and decrease of liver fibrosis. Int J Mol Sci 2014; 15:5412-5425. [PMID: 24686514 PMCID: PMC4013572 DOI: 10.3390/ijms15045412] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 03/16/2014] [Accepted: 03/20/2014] [Indexed: 12/16/2022] Open
Abstract
Platelets contain three types of granules: alpha granules, dense granules, and lysosomal granules. Each granule contains various growth factors, cytokines, and other physiological substances. Platelets trigger many kinds of biological responses, such as hemostasis, wound healing, and tissue regeneration. This review presents experimental evidence of platelets in accelerating liver regeneration and improving liver fibrosis. The regenerative effect of liver by platelets consists of three mechanisms; i.e., the direct effect on hepatocytes, the cooperative effect with liver sinusoidal endothelial cells, and the collaborative effect with Kupffer cells. Many signal transduction pathways are involved in hepatocyte proliferation. One is activation of Akt and extracellular signal-regulated kinase (ERK)1/2, which are derived from direct stimulation from growth factors in platelets. The other is signal transducer and activator of transcription-3 (STAT3) activation by interleukin (IL)-6 derived from liver sinusoidal endothelial cells and Kupffer cells, which are stimulated by contact with platelets during liver regeneration. Platelets also improve liver fibrosis in rodent models by inactivating hepatic stellate cells to decrease collagen production. The level of intracellular cyclic adenosine monophosphate (cyclic AMP) is increased by adenosine through its receptors on hepatic stellate cells, resulting in inactivation of these cells. Adenosine is produced by the degradation of adenine nucleotides such as adenosine diphosphate (ADP) and adenosine tri-phosphate (ATP), which are stored in abundance within the dense granules of platelets.
Collapse
Affiliation(s)
- Soichiro Murata
- Department of Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Takehito Maruyama
- Department of Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Takeshi Nowatari
- Department of Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Kazuhiro Takahashi
- Department of Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Nobuhiro Ohkohchi
- Department of Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
49
|
Iwase H, Ezzelarab MB, Ekser B, Cooper DKC. The role of platelets in coagulation dysfunction in xenotransplantation, and therapeutic options. Xenotransplantation 2014; 21:201-20. [PMID: 24571124 DOI: 10.1111/xen.12085] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 01/08/2014] [Indexed: 12/11/2022]
Abstract
Xenotransplantation could resolve the increasing discrepancy between the availability of deceased human donor organs and the demand for transplantation. Most advances in this field have resulted from the introduction of genetically engineered pigs, e.g., α1,3-galactosyltransferase gene-knockout (GTKO) pigs transgenic for one or more human complement-regulatory proteins (e.g., CD55, CD46, CD59). Failure of these grafts has not been associated with the classical features of acute humoral xenograft rejection, but with the development of thrombotic microangiopathy in the graft and/or consumptive coagulopathy in the recipient. Although the precise mechanisms of coagulation dysregulation remain unclear, molecular incompatibilities between primate coagulation factors and pig natural anticoagulants exacerbate the thrombotic state within the xenograft vasculature. Platelets play a crucial role in thrombosis and contribute to the coagulation disorder in xenotransplantation. They are therefore important targets if this barrier is to be overcome. Further genetic manipulation of the organ-source pigs, such as pigs that express one or more coagulation-regulatory genes (e.g., thrombomodulin, endothelial protein C receptor, tissue factor pathway inhibitor, CD39), is anticipated to inhibit platelet activation and the generation of thrombus. In addition, adjunctive pharmacologic anti-platelet therapy may be required. The genetic manipulations that are currently being tested are reviewed, as are the potential pharmacologic agents that may prove beneficial.
Collapse
Affiliation(s)
- Hayato Iwase
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
50
|
Effect of Platelet-Rich Plasma on CCl4-Induced Chronic Liver Injury in Male Rats. Int J Hepatol 2014; 2014:932930. [PMID: 24707405 PMCID: PMC3953414 DOI: 10.1155/2014/932930] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/02/2014] [Indexed: 11/30/2022] Open
Abstract
Platelet-rich plasma (PRP) has been of great concern to the scientists and doctors who are involved in wound healing and regenerative medicine which focuses on repairing and replacing damaged cells and tissues. Growth factors of platelet-rich plasma are cost-effective, available, and is more stable than recombinant human growth factors. Given these valuable properties, we decided to assess the effect of PRP on CCl4-induced hepatotoxicity on rats. The rats received CCl4 (1 mL/kg, i.p. 1 : 1 in olive oil) twice per week for 8 weeks. Five weeks after CCl4 injection, the rats also received PRP (0.5 mL/kg, s.c.) two days a week for three weeks. Twenty-four hours after last CCl4 injection, the animals bled and their livers dissected for biochemical and histopathological studies. Blood analysis was performed to evaluate enzyme activity. The results showed that PRP itself was not toxic for liver and could protect the liver from CCl4-induced histological damages and attenuated oxidative stress by increase in glutathione content and decrease in lipid peroxidative marker of liver tissue. The results of the present study lend support to our beliefs in hepatoprotective effects of PRP.
Collapse
|