51
|
Xu Z, Tsai HI, Xiao Y, Wu Y, Su D, Yang M, Zha H, Yan F, Liu X, Cheng F, Chen H. Engineering Programmed Death Ligand-1/Cytotoxic T-Lymphocyte-Associated Antigen-4 Dual-Targeting Nanovesicles for Immunosuppressive Therapy in Transplantation. ACS NANO 2020; 14:7959-7969. [PMID: 32515579 DOI: 10.1021/acsnano.9b09065] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
T cell activation by immune allorecognition is a major contributing factor toward the triggering of organ rejection. Immunosuppressive drugs have to be taken after organ transplantation, but long-term use of these drugs increases the risks of infection and other serious disorders. Here, we showed dysregulation of programmed cell death-ligand 1/programmed cell death 1 (PD-L1/PD-1) and cytotoxic T-lymphocyte-associated protein 4/cluster of differentiation 80 (CTLA-4/CD80) in the spleen of two organ transplantation models. Using a bioengineering approach, cellular exosome-like nanovesicles (NVs) displaying PD-L1/CTLA-4 dual-targeting cargos were designed, and their specificity to bind their ligands PD-1 and CD80 on T cell and dendritic cell surfaces was confirmed. These NVs consequently enhanced PD-L1/PD-1 and CTLA-4/CD80 immune inhibitory pathways, two key immune checkpoints to co-inhibit T cell activation and maintain peripheral tolerance. It was also confirmed that PD-L1/CTLA-4 NVs led to the reduction of T cell activation and proliferation in vitro and in vivo. Finally, it was demonstrated that PD-L1/CTLA-4 NVs reduced density of CD8+ T cells and cytokine production, enriched regulatory T cells, and prolonged the survival of mouse skin and heart grafts. Taken together, these data supported the idea that PD-L1/CTLA-4 dual-targeting NVs exert immune inhibitory effects and may be used as a prospective immunosuppressant in organ transplantation.
Collapse
Affiliation(s)
- Zhanxue Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Hsiang-I Tsai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Youmei Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Yingyi Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Dandan Su
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Min Yang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Hualian Zha
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Fuxia Yan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Xiaoyan Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| |
Collapse
|
52
|
Moosavi MM, Duncan A, Stowell SR, Roback JD, Sullivan HC. Passenger Lymphocyte Syndrome; a Review of the Diagnosis, Treatment, and Proposed Detection Protocol. Transfus Med Rev 2020; 34:178-187. [PMID: 32826130 DOI: 10.1016/j.tmrv.2020.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Passenger lymphocyte syndrome (PLS) is caused by the transfer of B-lymphocytes present in the donor graft into the recipient circulation following solid organ or hematopoietic stem cell transplantation. These cells may produce antibodies against the recipient's red blood cells, thereby triggering antibody dependent cytotoxicity and erythroid clearance, with potential resulting hemolysis and jaundice. Although uncommon, the true incidence is unknown because many cases are subclinical, with only serologic findings or with non significant levels of hemolysis detectable clinically or by laboratory monitoring. Thus, these cases may not be detected in the immediate perioperative period. No standardized consensus exists on screening for PLS in patients. Through a review of the literature from 2009 to 2019, we aim to approximate the incidence of this condition in different solid organ transplant settings, as well as to streamline recognition, detection, and management of PLS early in the disease course to prevent adverse outcomes and minimize invasive therapy. The resultant literature review yielded 22 case reports and 8 case series comprising 71 solid organ transplant patients. Hematopoietic stem cell transplant cases were excluded, as PLS cases related to solid organ transplant were the primary focus of this review. Our institution has traditionally handled PLS on a case-by-case basis, although we hope to improve this process through an introduction of an algorithm based on review of the literature and formalized communication with primary caregivers.
Collapse
Affiliation(s)
- Mitchell M Moosavi
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Alexander Duncan
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Sean R Stowell
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - John D Roback
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Harold Clifford Sullivan
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA.
| |
Collapse
|
53
|
Elsanhoury A, Tschöpe C, Van Linthout S. A Toolbox of Potential Immune-Related Therapies for Inflammatory Cardiomyopathy. J Cardiovasc Transl Res 2020; 14:75-87. [PMID: 32440911 PMCID: PMC7892499 DOI: 10.1007/s12265-020-10025-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
Myocarditis is a multifactorial disorder, characterized by an inflammatory reaction in the myocardium, predominantly triggered by infectious agents, but also by antigen mimicry or autoimmunity in susceptible individuals. Unless spontaneously resolved, a chronic inflammatory course concludes with cardiac muscle dysfunction portrayed by ventricular dilatation, clinically termed inflammatory cardiomyopathy (Infl-CM). Treatment strategies aim to resolve chronic inflammation and preserve cardiac function. Beside standard heart failure treatments, which only play a supportive role in this condition, systemic immunosuppressants are used to diminish inflammatory cell function at the cost of noxious side effects. To date, the treatment protocols are expert-based without large clinical evidence. This review describes concept and contemporary strategies to alleviate myocardial inflammation and sheds light on potential inflammatory targets in an evidence-based order.
Collapse
Affiliation(s)
- Ahmed Elsanhoury
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Föhrerstrasse 15, 13353, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Föhrerstrasse 15, 13353, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.,Department of Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Föhrerstrasse 15, 13353, Berlin, Germany. .,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.
| |
Collapse
|
54
|
Clinical periodontal and dental findings in liver transplant patients: a systematic review and meta-analysis. Br Dent J 2020; 228:108-116. [PMID: 31980788 DOI: 10.1038/s41415-020-1196-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objectives This systematic review aimed to investigate the oral health of post-liver transplant recipients (postLTx), in terms of dental and periodontal clinical results.Data sources Pubmed/MEDLINE and Cochrane Library databases were searched electronically.Data selection Prospective, cross-sectional and case-control studies comparing periodontal and dental health of postLTx patients with healthy and/or pre-liver transplant patients.Data extraction This was performed independently by two authors. Quality assessment was done by using tools from the National Heart, Lung, and Blood Institute.Data synthesis Nine studies were eligible for inclusion in the systematic review. A meta-analysis was performed on gingival inflammation, expressed by scores, on four studies including healthy controls. The gingival index scores were significantly higher in postLTx patients compared with healthy controls (p <0.001). The weighted mean difference was 0.91 (95% confidence interval, 0.44 to 1.38, heterogeneity I2 = 99.5%). Increased tendency of gingival enlargement was noticed in postLTx patients, especially after immunosuppression with Cyclosporine A. Controversial results were reported for the prevalence of caries in postLTx recipients, but there seems to be an increase.Conclusion The results suggest that patients with liver transplants, due to systemic condition and immunosuppressive therapy, exhibit a higher degree of gingival inflammation compared with unexposed patients.Prospero registration number: CRD42019121893.
Collapse
|
55
|
Zhuang Q, Li H, Yu M, Peng B, Liu S, Luo M, Stefano GB, Kream RM, Ming Y. Profiles of B-cell subsets in immunologically stable renal allograft recipients and end-stage renal disease patients. Transpl Immunol 2020; 58:101249. [DOI: 10.1016/j.trim.2019.101249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/27/2022]
|
56
|
Brami I, Ini D, Sassonker N, Zaknoun M, Zuckerman T, Lewis EC. Immunosuppressive Drugs Alter α1-Antitrypsin Production in Hepatocytes: Implications for Epithelial Gap Repair. Biol Blood Marrow Transplant 2019; 26:625-633. [PMID: 31899361 DOI: 10.1016/j.bbmt.2019.12.764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/18/2019] [Accepted: 12/25/2019] [Indexed: 01/04/2023]
Abstract
Immunosuppressive drugs are an inherent component of hematopoietic stem cell transplantation (HSCT) for the prevention of acute graft-versus-host disease (GVHD). Circulating α1-antitrypsin (AAT), a serine-protease inhibitor produced predominantly by hepatocytes that rises during acute phase responses, is lost in patient's stool due to gastrointestinal GVHD, and its augmentation has been found to attenuate GVHD. Here we explored the effect of immunosuppressive drugs on hepatocyte production of AAT and intestinal epithelial gap repair. The effect of commonly used immunosuppressants on AAT production was examined in vitro using HepG2 cells and primary mouse hepatocytes, and their impact on human intestinal epithelial cell line gap repair was evaluated. Sera from 12 allogeneic HSCT recipients, obtained at 14 days post-transplantation, predating the diagnosis of GVHD (n = 6), were examined for reepithelialization, with added clinical-grade AAT. Rapamycin compromised AAT production under inflammatory conditions. Mycophenolate mofetil and cyclosporine A (CSA) inhibited reepithelialization; AAT minimized the effect of CSA. Patient sera displayed superior gap repair with exogenous AAT. Functional insufficiency in circulating AAT may be the result of drug toxicities leading to ineffective gut reepithelization and compromised gut lining. Taken together, our data strengthen the rationale for incorporating AAT augmentation therapy into immunosuppressive treatment protocols.
Collapse
Affiliation(s)
- Ido Brami
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel.
| | - Dor Ini
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel
| | - Nofit Sassonker
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel
| | - Melodie Zaknoun
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel
| | - Tsila Zuckerman
- Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel
| |
Collapse
|
57
|
Nanga TM, Doan TTP, Marquet P, Musuamba FT. Toward a robust tool for pharmacokinetic-based personalization of treatment with tacrolimus in solid organ transplantation: A model-based meta-analysis approach. Br J Clin Pharmacol 2019; 85:2793-2823. [PMID: 31471970 DOI: 10.1111/bcp.14110] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
AIMS The objective of this study is to develop a generic model for tacrolimus pharmacokinetics modelling using a meta-analysis approach, that could serve as a first step towards a prediction tool to inform pharmacokinetics-based optimal dosing of tacrolimus in different populations and indications. METHODS A systematic literature review was performed and a meta-model developed with NONMEM software using a top-down approach. Historical (previously published) data were used for model development and qualification. In-house individual rich and sparse tacrolimus blood concentration profiles from adult and paediatric kidney, liver, lung and heart transplant patients were used for model validation. Model validation was based on successful numerical convergence, adequate precision in parameter estimation, acceptable goodness of fit with respect to measured blood concentrations with no indication of bias, and acceptable performance of visual predictive checks. External validation was performed by fitting the model to independent data from 3 external cohorts and remaining previously published studies. RESULTS A total of 76 models were found relevant for meta-model building from the literature and the related parameters recorded. The meta-model developed using patient level data was structurally a 2-compartment model with first-order absorption, absorption lag time and first-time varying elimination. Population values for clearance, intercompartmental clearance, central and peripheral volume were 22.5 L/h, 24.2 L/h, 246.2 L and 109.9 L, respectively. The absorption first-order rate and the lag time were fixed to 3.37/h and 0.33 hours, respectively. Transplanted organ and time after transplantation were found to influence drug apparent clearance whereas body weight influenced both the apparent volume of distribution and the apparent clearance. The model displayed good results as regards the internal and external validation. CONCLUSION A meta-model was successfully developed for tacrolimus in solid organ transplantation that can be used as a basis for the prediction of concentrations in different groups of patients, and eventually for effective dose individualization in different subgroups of the population.
Collapse
Affiliation(s)
- Tom M Nanga
- INSERM UMR 1248, Université de Limoges, FHU support, Limoges Cédex, 87025, France
| | - Thao T P Doan
- INSERM UMR 1248, Université de Limoges, FHU support, Limoges Cédex, 87025, France
| | - Pierre Marquet
- INSERM UMR 1248, Université de Limoges, FHU support, Limoges Cédex, 87025, France
| | - Flora T Musuamba
- Federal Agency for Medicines and Health Products, Brussels, Belgium.,Faculté des sciences pharmaceutiques, Université de Lubumbashi, Lubumbashi, Democratic Republic of the Congo
| |
Collapse
|
58
|
Gastrodin Ameliorates Acute Rejection via IRE1 α/TRAF2/NF- κB in Rats Receiving Liver Allografts. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9276831. [PMID: 31828147 PMCID: PMC6886336 DOI: 10.1155/2019/9276831] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/05/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022]
Abstract
Background Liver transplantation (LT) is currently an effective treatment for end-stage liver disease, but the occurrence of acute rejection (AR) is still the main problem to be solved. The present study aimed to evaluate the effect of gastrodin (GAS) on LT. Methods Rat transplant models were established and divided into SHAM, LT, GAS-L (50 mg/kg GAS), and GAS-H (100 mg/kg GAS) groups. The liver function, inflammatory factors, liver histopathology, survival of rats, number of M2-type macrophages, liver cell apoptosis, and pathway proteins were assayed at 7 days and 14 days after the operations. Results With increasing GAS concentrations, liver function, expression of proinflammatory factors in the liver, and expression of M2-type molecules in macrophages were significantly improved, and the survival time of rats was significantly prolonged (P < 0.05). All rats treated with low or high doses of GAS were judged to have nondeterministic acute rejection. Flow cytometry showed that liver cell apoptosis was decreased significantly in the GAS-L and GAS-H groups after GAS administration compared with apoptosis and differentiation in the LT group (P < 0.05). Expression levels of Caspase-3, Bad, and Bax proteins were decreased, and the expression of the antiapoptotic protein Bcl-2 was increased in the GAS-L and GAS-H groups (P < 0.05). Mechanistically, the ERS-related IRE1α/TRAF2/NF-κB pathway was suppressed by GAS, and GAS acted mainly on intrahepatic macrophages to affect AR and reduce ROS production (P < 0.05). Conclusion GAS ameliorated AR by inhibiting the IRE1α/TRAF2/NF-κB pathway in LT.
Collapse
|
59
|
Gianardi D, Bianchini M, Palmeri M, Di Franco G, Morelli L. Is there a role of robotic surgery in abdominal organs transplantations? J Robot Surg 2019; 14:677-678. [PMID: 31531754 DOI: 10.1007/s11701-019-01002-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Desirée Gianardi
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, Pisa, 56124, Italy.
| | - Matteo Bianchini
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, Pisa, 56124, Italy
| | - Matteo Palmeri
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, Pisa, 56124, Italy
| | - Gregorio Di Franco
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, Pisa, 56124, Italy
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, Pisa, 56124, Italy
- EndoCAS (Centre for Computer Assisted Surgery), University of Pisa, Pisa, Italy
| |
Collapse
|
60
|
Recent Topics on The Mechanisms of Immunosuppressive Therapy-Related Neurotoxicities. Int J Mol Sci 2019; 20:ijms20133210. [PMID: 31261959 PMCID: PMC6651704 DOI: 10.3390/ijms20133210] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023] Open
Abstract
Although transplantation procedures have been developed for patients with end-stage hepatic insufficiency or other diseases, allograft rejection still threatens patient health and lifespan. Over the last few decades, the emergence of immunosuppressive agents such as calcineurin inhibitors (CNIs) and mammalian target of rapamycin (mTOR) inhibitors have strikingly increased graft survival. Unfortunately, immunosuppressive agent-related neurotoxicity commonly occurs in clinical practice, with the majority of neurotoxicity cases caused by CNIs. The possible mechanisms through which CNIs cause neurotoxicity include increasing the permeability or injury of the blood–brain barrier, alterations of mitochondrial function, and alterations in the electrophysiological state. Other immunosuppressants can also induce neuropsychiatric complications. For example, mTOR inhibitors induce seizures, mycophenolate mofetil induces depression and headaches, methotrexate affects the central nervous system, the mouse monoclonal immunoglobulin G2 antibody (used against the cluster of differentiation 3) also induces headaches, and patients using corticosteroids usually experience cognitive alteration. Therapeutic drug monitoring, individual therapy based on pharmacogenetics, and early recognition of symptoms help reduce neurotoxic events considerably. Once neurotoxicity occurs, a reduction in the drug dosage, switching to other immunosuppressants, combination therapy with drugs used to treat the neuropsychiatric manifestation, or blood purification therapy have proven to be effective against neurotoxicity. In this review, we summarize recent topics on the mechanisms of immunosuppressive drug-related neurotoxicity. In addition, information about the neuroprotective effects of several immunosuppressants is also discussed.
Collapse
|
61
|
Rezahosseini O, Drabe CH, Sørensen SS, Rasmussen A, Perch M, Ostrowski SR, Nielsen SD. Torque-Teno virus viral load as a potential endogenous marker of immune function in solid organ transplantation. Transplant Rev (Orlando) 2019; 33:137-144. [PMID: 30981537 DOI: 10.1016/j.trre.2019.03.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/29/2019] [Accepted: 03/31/2019] [Indexed: 12/27/2022]
Abstract
Solid organ transplantation (SOT) recipients receive immunosuppressive therapy to avoid rejection of the transplanted organ. Immunosuppressive therapy increases the risk of infections. However, no existing marker reliably reveals the status of the immune function in SOT recipients. Torque-Teno virus or Transfusion-transmitted virus (TTV) has gained attention as a possible endogenous marker of the immune function. TTV is a non-enveloped, circular single strand DNA virus, and it may be considered a part of the human virome. In a bidirectional relationship, the immune system detects TTV and TTV may also modulate the activity of immune system. These characteristics have made the virus a possible candidate indicator of immune function. In this systematic review, we describe the role and potential function of TTV viral load as an endogenous marker of the immune function and consequently the level of immune suppression in SOT recipients.
Collapse
Affiliation(s)
- Omid Rezahosseini
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Heldbjerg Drabe
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Allan Rasmussen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Michael Perch
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
62
|
Lopez J, Gourin CG, Tufaro AP. Aggressive Cutaneous Malignancies: A New and Dangerous Phenomenon in Transplant Patients. CURRENT SURGERY REPORTS 2019. [DOI: 10.1007/s40137-019-0223-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
63
|
Bösmüller C, Messner F, Margreiter C, Öllinger R, Maglione M, Oberhuber R, Scheidl S, Neuwirt H, Öfner D, Margreiter R, Schneeberger S. Good Results with Individually Adapted Long-Term Immunosuppression Following Alemtuzumab Versus ATG Induction Therapy in Combined Kidney-Pancreas Transplantation: A Single-Center Report. Ann Transplant 2019; 24:52-56. [PMID: 30679414 PMCID: PMC6363467 DOI: 10.12659/aot.911712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Claudia Bösmüller
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Franka Messner
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Margreiter
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Öllinger
- Department of General Surgery, Charité Virchow Hospital Berlin, Berlin, Germany
| | - Manuel Maglione
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Rupert Oberhuber
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Scheidl
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Hannes Neuwirt
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Öfner
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Raimund Margreiter
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Schneeberger
- Department of Transplant Surgery, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
64
|
Gentry BG, Bogner E, Drach JC. Targeting the terminase: An important step forward in the treatment and prophylaxis of human cytomegalovirus infections. Antiviral Res 2018; 161:116-124. [PMID: 30472161 DOI: 10.1016/j.antiviral.2018.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/07/2018] [Accepted: 11/13/2018] [Indexed: 10/27/2022]
Abstract
A key step in the replication of human cytomegalovirus (HCMV) in the host cell is the generation and packaging of unit-length genomes into preformed capsids. Enzymes required for this process are so-called terminases, first described for double-stranded DNA bacteriophages. The HCMV terminase consists of the two subunits, the ATPase pUL56 and the nuclease pUL89, and a potential third component pUL51. The terminase subunits are essential for virus replication and are highly conserved throughout the Herpesviridae family. Together with the portal protein pUL104 they form a powerful biological nanomotor. It has been shown for tailed dsDNA bacteriophages that DNA translocation into preformed capsid needs an extraordinary amount of energy. The HCMV terminase subunit pUL56 provides the required ATP hydrolyzing activity. The necessary nuclease activity to cleave the concatemers into unit-length genomes is mediated by the terminase subunit pUL89. Whether this cleavage is mediated by site-specific duplex nicking has not been demonstrated, however, it is required for packaging. Binding to the portal is a prerequisite for DNA translocation. To date, it is a common view that during translocation the terminase moves along some domains of the DNA by a binding and release mechanism. These critical structures have proven to be outstanding targets for drugs to treat HCMV infections because corresponding structures do not exist in mammalian cells. Herein we examine the HCMV terminase as a target for drugs and review several inhibitors discovered by both lead-directed medicinal chemistry and by target-specific design. In addition to producing clinically active compounds the research also has furthered the understanding of the role and function of the terminase itself.
Collapse
Affiliation(s)
- Brian G Gentry
- Drake University College of Pharmacy and Health Sciences, 2507 University Ave., Des Moines, 50311, IA, USA.
| | - Elke Bogner
- Institute of Virology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - John C Drach
- University of Michigan School of Dentistry, 1101 N. University Ave., Ann Arbor, 48109, MI, USA.
| |
Collapse
|
65
|
Arasaratnam RJ, Tzannou I, Gray T, Aguayo-Hiraldo PI, Kuvalekar M, Naik S, Gaikwad A, Liu H, Miloh T, Vera JF, Himes RW, Munoz FM, Leen AM. Dynamics of virus-specific T cell immunity in pediatric liver transplant recipients. Am J Transplant 2018; 18:2238-2249. [PMID: 29900673 PMCID: PMC6117219 DOI: 10.1111/ajt.14967] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/11/2018] [Accepted: 06/06/2018] [Indexed: 01/25/2023]
Abstract
Immunosuppression following solid organ transplantation (SOT) has a deleterious effect on cellular immunity leading to frequent and prolonged viral infections. To better understand the relationship between posttransplant immunosuppression and circulating virus-specific T cells, we prospectively monitored the frequency and function of T cells directed to a range of latent (CMV, EBV, HHV6, BK) and lytic (AdV) viruses in 16 children undergoing liver transplantation for up to 1 year posttransplant. Following transplant, there was an immediate decline in circulating virus-specific T cells, which recovered posttransplant, coincident with the introduction and subsequent routine tapering of immunosuppression. Furthermore, 12 of 14 infections/reactivations that occurred posttransplant were successfully controlled with immunosuppression reduction (and/or antiviral use) and in all cases we detected a temporal increase in the circulating frequency of virus-specific T cells directed against the infecting virus, which was absent in 2 cases where infections remained uncontrolled by the end of follow-up. Our study illustrates the dynamic changes in virus-specific T cells that occur in children following liver transplantation, driven both by active viral replication and modulation of immunosuppression.
Collapse
Affiliation(s)
- R J Arasaratnam
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - I Tzannou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - T Gray
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - P I Aguayo-Hiraldo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - M Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - S Naik
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - A Gaikwad
- Department of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - H Liu
- Biostatistics Core of the Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - T Miloh
- Department of Pediatrics, Division of Pediatric Gastroenterology and Nutrition, Texas Children's Hospital, Houston, TX, USA
| | - J F Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - R W Himes
- Department of Pediatrics, Division of Pediatric Gastroenterology and Nutrition, Texas Children's Hospital, Houston, TX, USA
| | - F M Munoz
- Departments of Pediatrics, Infectious Diseases Section, and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - A M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
66
|
Lim S, Kirkiles-Smith NC, Pober JS, Bothwell ALM, Choi JM. Regulation of human T cell responses by dNP2-ctCTLA-4 inhibits human skin and microvessel graft rejection. Biomaterials 2018; 183:128-138. [PMID: 30165256 DOI: 10.1016/j.biomaterials.2018.08.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 12/30/2022]
Abstract
Manipulation of human T cell functioning by delivery of macromolecules such as DNA, RNA, or protein is limited, unless the human T cells have been stimulated or electropermeabilized. To achieve successful adaptation and survival of a grafted organ, the alloreactive T cells that induce graft rejection must be regulated. Corticosteroids, calcineurin inhibitors, and mTOR inhibitors, which are systemic immunosuppressants, are currently used for transplantation, with significant side effects. In this study, we demonstrated that a cell-permeable peptide (CPP), dNP2, could efficiently deliver proteins into human CD4 and CD8 T cells. We confirmed regulatory functioning of the cytoplasmic domain of CTLA-4 conjugated with dNP2 (dNP2-ctCTLA-4) in human T cell activation, proliferation, and chemokine receptor expression. We utilized a human skin allograft system in SCID/beige mice to examine whether dNP2-ctCTLA-4 could inhibit allograft rejection by controlling T cell responses. The grafted skin tissue inflammation, allogeneic T cell infiltration, and blood cytokine level was markedly reduced by dNP2-ctCTLA-4, resulting in successful transplantation. In addition, it also inhibited T cell alloresponses against microvessels formed form Bcl-2-transduced human umbilical vein endothelial cells implanted into Balb/c Rag1-/-/IL-2Rγ-/- double knockout (DKO) mice, assessed as reduced T cell infiltration and granzyme B expression. These results collectively suggest that dNP2 peptide conjugation offers a valuable tool for delivering macromolecules like proteins into human T cells, and dNP2-ctCTLA-4 is a novel agent that shows potential in controlling human T cell responses to allow successful adaptation of grafted tissues.
Collapse
Affiliation(s)
- Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Nancy C Kirkiles-Smith
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
67
|
van Aalst M, Lötsch F, Spijker R, van der Meer JTM, Langendam MW, Goorhuis A, Grobusch MP, de Bree GJ. Incidence of invasive pneumococcal disease in immunocompromised patients: A systematic review and meta-analysis. Travel Med Infect Dis 2018; 24:89-100. [PMID: 29860151 DOI: 10.1016/j.tmaid.2018.05.016] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/10/2018] [Accepted: 05/29/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Invasive pneumococcal disease (IPD) is associated with high morbidity and mortality, with immunocompromised patients (ICPs) at particular risk. Therefore, guidelines recommend pneumococcal vaccination for these patients. However, guidelines are scarcely underpinned with references to incidence studies of IPD in this population. This, potentially results in unawareness of the importance of vaccination and low vaccination rates. The objective of this systematic review and meta-analysis was to assess the incidence of IPD in ICPs. METHODS We systematically searched PubMed and Embase to identify studies in English published before December 6th, 2017 that included terms related to 'incidence', 'rate', 'pneumococcal', 'pneumoniae', 'meningitis', 'septicemia', or 'bacteremia'. We focused on patients with HIV, transplantation and chronic inflammatory diseases. RESULTS We included 45 studies in the systematic review reporting an incidence or rate of IPD, defined as isolation of Streptococcus pneumoniae from a normally sterile site. Random effects meta-analysis of 38 studies showed a pooled IPD incidence of 331/100,000 person years in patients with HIV in the late-antiretroviral treatment era in non-African countries, and 318/100,000 in African countries; 696 and 812/100,000 in patients who underwent an autologous or allogeneic stem cell transplantation, respectively; 465/100,000 in patients with a solid organ transplantation; and 65/100,000 in patients with chronic inflammatory diseases. In healthy control cohorts, the pooled incidence was 10/100,000. DISCUSSION ICPs are at increased risk of contracting IPD, especially those with HIV, and those who underwent transplantation. Based on our findings, we recommend pneumococcal vaccination in immunocompromised patients. PROSPERO REGISTRATION ID: CRD42016048438.
Collapse
Affiliation(s)
- Mariëlle van Aalst
- Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
| | - Felix Lötsch
- Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands; Clinical Division of Infection and Tropical Medicine, Medical University of Vienna, Splitalgasse 23, 1090, Vienna, Austria
| | - René Spijker
- Medical Library, Academic Medical Center, Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands; Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan T M van der Meer
- Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
| | - Miranda W Langendam
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
| | - Abraham Goorhuis
- Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
| | - Martin P Grobusch
- Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands; Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.
| | - Godelieve J de Bree
- Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands; Amsterdam Institute for Global Health and Development, Paasheuvelweg 25, 1105BP, Amsterdam, The Netherlands.
| |
Collapse
|
68
|
Zhang Q, Dai Y, Cai Z, Mou L. HDAC Inhibitors: Novel Immunosuppressants for Allo- and Xeno- Transplantation. ChemistrySelect 2018. [DOI: 10.1002/slct.201702295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Qing Zhang
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center; Institute of Translational Medicine, Shenzhen Second People's Hospital; Sungang Road 3002, Futian District, Shenzhen Guangdong China
| | - Yifan Dai
- Department Jiangsu Key Laboratory of Xenotransplantation; Nanjing Medical University; Nanjing, Jiangsu 210029 China
| | - Zhiming Cai
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center; Institute of Translational Medicine, Shenzhen Second People's Hospital; Sungang Road 3002, Futian District, Shenzhen Guangdong China
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center; Institute of Translational Medicine, Shenzhen Second People's Hospital; Sungang Road 3002, Futian District, Shenzhen Guangdong China
| |
Collapse
|