1
|
Gedaly R, Orozco G, Lewis LJ, Valvi D, Chapelin F, Khurana A, Hidalgo GE, Shmookler A, Tripathi A, Zhang C, Zwischenberger JB, Marti F. Effect of mitochondrial oxidative stress on Regulatory T Cell manufacturing for clinical application in transplantation: results from a pilot study. Am J Transplant 2024:S1600-6135(24)00684-1. [PMID: 39515758 DOI: 10.1016/j.ajt.2024.10.024] [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: 01/25/2024] [Revised: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
The manufacturing process of Regulatory T (Treg) cells for clinical application begins with the positive selection of CD25+ cells using superparamagnetic iron-oxide nanoparticle (SPION)-conjugated anti-CD25 antibodies (spCD25) and immunomagnetic cell separation technology. Our findings revealed that the interaction of spCD25 with its cell target induced the internalization of the complex spCD25-Interleukin-2 Receptor. Accumulation of intracellular spCD25 triggered oxidative stress, causing delayed Treg expansion and temporary reduction in suppressor activity. This activation delay hindered the efficient generation of clinically competent cells. During this early phase, Treg cells exhibited elevated mitochondrial superoxide and lipid peroxidation levels, with concomitant decrease on mitochondrial respiration rates. The results uncovered the increased mitochondrial unfolded protein response (mitoUPR). This protective, redox-sensitive activity is inherent of Tregs when contrasted with homologous, spCD25-treated, conventional T cells. While the temporary effects of spCD25 on clinically competent cells did not impede their use in a safety/feasibility pilot study with kidney transplant recipients*, it is reasonable to anticipate a potential reduction in their therapeutic efficacy. The mechanistic understanding of the adverse effects triggered by spCD25 is crucial for improving the manufacturing process of clinically competent Treg cells, a pivotal step in the successful implementation of immune cell therapy in transplantation. *Clinical trial registration number NCT03284242 at ClinicalTrials.gov.
Collapse
Affiliation(s)
- Roberto Gedaly
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA; Lucillle Parker Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA; Division of Transplantation, Quality and Biostatistics Section, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA; Alliance Research Initiative (TILT Alliance), University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA
| | - Gabriel Orozco
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Lillie J Lewis
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Deepa Valvi
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Fanny Chapelin
- Lucillle Parker Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA; Alliance Research Initiative (TILT Alliance), University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA; Department of Biomedical Engineering, University of Kentucky, College of Medicine, Lexington, Kentucky 40506, USA
| | - Aman Khurana
- Lucillle Parker Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA; Alliance Research Initiative (TILT Alliance), University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA; Department of Radiology, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA
| | - Giovanna E Hidalgo
- Pathology and Laboratory Medicine, University of Kentucky, College of Medicine, Lexington, Kentucky 40506, USA
| | - Aaron Shmookler
- Pathology and Laboratory Medicine, University of Kentucky, College of Medicine, Lexington, Kentucky 40506, USA
| | - Aashutosh Tripathi
- Department of Microbiology, Immunology, and Molecular Genetics. University of Kentucky, College of Medicine, Lexington, Kentucky 40506, USA
| | - Cuiping Zhang
- Flow Cytometry & Immune Monitoring Core Facility, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA
| | - Joseph B Zwischenberger
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA; Lucillle Parker Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA
| | - Francesc Marti
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA; Lucillle Parker Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA; Division of Transplantation, Quality and Biostatistics Section, University of Kentucky, College of Medicine, Lexington, Kentucky 40536, USA; Alliance Research Initiative (TILT Alliance), University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA.
| |
Collapse
|
2
|
Herr F, Dekeyser M, Le Pavec J, Desterke C, Chiron AS, Bargiel K, Mercier O, Vernochet A, Fadel E, Durrbach A. mTOR Inhibition Impairs the Activation and Function of Belatacept-Resistant CD4 +CD57 + T Cells In Vivo and In Vitro. Pharmaceutics 2023; 15:pharmaceutics15041299. [PMID: 37111784 PMCID: PMC10142381 DOI: 10.3390/pharmaceutics15041299] [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: 02/20/2023] [Revised: 03/28/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Calcineurin inhibitors have improved graft survival in solid-organ transplantation but their use is limited by toxicity, requiring a switch to another immunosuppressor in some cases. Belatacept is one option that has been shown to improve graft and patient survival despite being associated with a higher risk of acute cellular rejection. This risk of acute cellular rejection is correlated with the presence of belatacept-resistant T cells. We performed a transcriptomic analysis of in vitro-activated cells to identify pathways affected by belatacept in belatacept-sensitive cells (CD4+CD57-) but not in belatacept-resistant CD4+CD57+ T cells. mTOR was significantly downregulated in belatacept-sensitive but not belatacept-resistant T cells. The inhibition of mTOR strongly decreases the activation and cytotoxicity of CD4+CD57+ cells. In humans, the use of a combination of mTOR inhibitor and belatacept prevents graft rejection and decreases the expression of activation markers on CD4 and CD8 T cells. mTOR inhibition decreases the functioning of belatacept-resistant CD4+CD57+ T cells in vitro and in vivo. It could potentially be used in association with belatacept to prevent acute cellular rejection in cases of calcineurin intolerance.
Collapse
Affiliation(s)
- Florence Herr
- Institut Gustave Roussy, Inserm, Immunologie Intégrative des Tumeurs et Immunothérapie des Cancers, Université Paris-Saclay, 94805 Villejuif, France
| | - Manon Dekeyser
- Institut Gustave Roussy, Inserm, Immunologie Intégrative des Tumeurs et Immunothérapie des Cancers, Université Paris-Saclay, 94805 Villejuif, France
- Hôpital Henri Mondor, Service de Néphrologie, Assistance Publique-Hôpitaux de Paris, 94010 Creteil, France
| | - Jerome Le Pavec
- Inserm, Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Université Paris-Saclay, 92350 Le Plessis Robinson, France
- Centre Hospitalier Marie Lannelongue, 92350 Le Plessis Robinson, France
| | - Christophe Desterke
- Inserm, Modèles de Cellules Souches Malignes et Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France
| | - Andrada-Silvana Chiron
- Unité des Technologies Chimiques et Biologiques pour la Santé, CNRS, INSERM, UTCBS, Université de Paris, 75006 Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, 94270 Le Kremlin-Bicetre, France
| | - Karen Bargiel
- Institut Gustave Roussy, Inserm, Immunologie Intégrative des Tumeurs et Immunothérapie des Cancers, Université Paris-Saclay, 94805 Villejuif, France
| | - Olaf Mercier
- Inserm, Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Université Paris-Saclay, 92350 Le Plessis Robinson, France
- Centre Hospitalier Marie Lannelongue, 92350 Le Plessis Robinson, France
| | - Amelia Vernochet
- Institut Gustave Roussy, Inserm, Immunologie Intégrative des Tumeurs et Immunothérapie des Cancers, Université Paris-Saclay, 94805 Villejuif, France
| | - Elie Fadel
- Inserm, Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Université Paris-Saclay, 92350 Le Plessis Robinson, France
- Centre Hospitalier Marie Lannelongue, 92350 Le Plessis Robinson, France
| | - Antoine Durrbach
- Institut Gustave Roussy, Inserm, Immunologie Intégrative des Tumeurs et Immunothérapie des Cancers, Université Paris-Saclay, 94805 Villejuif, France
- Hôpital Henri Mondor, Service de Néphrologie, Assistance Publique-Hôpitaux de Paris, 94010 Creteil, France
| |
Collapse
|
3
|
Gedaly R, Cornea V, Turcios L, Edmisson JS, Harris DD, Watt DS, Chapelin F, Khurana A, Mei X, Liu C, Taylor I, Gonzalez-Valdivieso J, Mitchel H, Ruffing A, Chishti A, Orozco G, Zwischenberger J, Evers BM, Marti F. Anti-neoplastic sulfonamides alter the metabolic homeostasis and disrupt the suppressor activity of regulatory T cells. Sci Rep 2022; 12:19112. [PMID: 36352020 PMCID: PMC9646802 DOI: 10.1038/s41598-022-23601-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
Regulatory T cells (Tregs) are essential to maintain self-tolerance and immune homeostasis but, as components of the tumor microenvironment (TME), are also a major barrier to effective cancer immunosurveillance and immunotherapy. FH535 and its derivative Y3 are two N-aryl-benzene-sulfonamides (NABs) that inhibit HCC cell proliferation and tumor progression. However, the impact of NABs on the immune cells in the TME is not yet known. Analyses of explanted livers from patients with hepatocellular carcinoma (HCC) showed that high levels of tumor-infiltrating Tregs were associated with poor tumor differentiation. These results lead us to investigate the immunomodulatory effects of NABs in regulatory and effector T cells. Exposure of primary human Tregs to NABs induced a rapid but temporary increase of cell expansion, a gradual disruption of suppressor activity, and concomitant bioenergetics and autophagic flux dysregulations. In contrast to Tregs, no gross effects were observed in effector T cells. Addition of Rapamycin prevented the functional decay of Tregs and restored their metabolic profile, suggesting that NAB effects require the integrity of the mTOR pathway. This study revealed the immunomodulatory properties of NABs with a preferential impact on Treg activity and provided novel insights into the anti-tumor potential of sulfonamides.
Collapse
Affiliation(s)
- Roberto Gedaly
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
- Lucillle Parker Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
- Division of Transplantation, Section for Quality and Biostatistics, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
- Alliance Research Initiative (TILT Alliance), College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
- Department of Surgery, Transplant Center, 740 South Limestone, K 301, Rm 312, Lexington, KY, 40536-0284, USA.
| | - Virgilius Cornea
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Lucillle Parker Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Lilia Turcios
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Jacob S Edmisson
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Dwight D Harris
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - David S Watt
- Lucillle Parker Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - Fanny Chapelin
- Lucillle Parker Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Alliance Research Initiative (TILT Alliance), College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Department of Biomedical Engineering, College of Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Aman Khurana
- Lucillle Parker Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Alliance Research Initiative (TILT Alliance), College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Department of Radiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Xiaonan Mei
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Division of Transplantation, Section for Quality and Biostatistics, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Chunming Liu
- Lucillle Parker Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Isaac Taylor
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Juan Gonzalez-Valdivieso
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Hunter Mitchel
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Department of Science & Health, School of Science, Health & Mathematics, Asbury University, Wilmore, KY, 40390, USA
| | - Alexis Ruffing
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
- Department of Science & Health, School of Science, Health & Mathematics, Asbury University, Wilmore, KY, 40390, USA
| | - Asir Chishti
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Gabriel Orozco
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Joseph Zwischenberger
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - B Mark Evers
- Lucillle Parker Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Francesc Marti
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
- Lucillle Parker Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
- Alliance Research Initiative (TILT Alliance), College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
- Department of Surgery, Transplant Center, Peter P. Bosomworth Health Sciences Research Building (HSRB), Office: Room# 363 / Lab: Room# 361, 1095 Veterans Drive, Lexington, KY, 40536-0305, USA.
| |
Collapse
|
4
|
Zhou K, Chen X, Zhang L, Yang Z, Zhu H, Guo D, Su R, Chen H, Li H, Song P, Xu X, Wang H, Zheng S, Xie H. Targeting peripheral immune organs with self-assembling prodrug nanoparticles ameliorates allogeneic heart transplant rejection. Am J Transplant 2021; 21:3871-3882. [PMID: 34212503 DOI: 10.1111/ajt.16748] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 01/25/2023]
Abstract
Organ transplantation has become a mainstay of therapy for patients with end-stage organ diseases. However, long-term administration of immunosuppressive agents, a scheme for improving the survival of transplant recipients, has been compromised by severe side effects and posttransplant complications. Therapeutic delivery targeting immune organs has the potential to address these unmet medical issues. Here, through screening of a small panel of mammalian target of rapamycin complex kinase inhibitor (TORKinib) compounds, a TORKinib PP242 is identified to be able to inhibit T cell function. Further chemical derivatization of PP242 using polyunsaturated fatty acids (i.e., docosahexaenoic acid) transforms this water-insoluble hydrophobic agent into a self-assembling nanoparticle (DHA-PP242 nanoparticle [DPNP]). Surface PEGylation of DPNP with amphiphilic copolymers renders the nanoparticles aqueously soluble for preclinical studies. Systemically administered DPNP shows tropism for macrophages within peripheral immune organs. Furthermore, DPNP regulates differentiation of adoptively transferred T cells in a macrophage-dependent manner in Rag1-/- mouse model. In an experimental model of heart transplantation, DPNP significantly extends the survival of grafts through inducing immune suppression, thus reducing the inflammatory response of the recipients. These findings suggest that targeted delivery of TORKinibs exploiting prodrug-assembled nanoparticle scaffolds may provide a therapeutic option against organ rejection.
Collapse
Affiliation(s)
- Ke Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Xiaona Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Liang Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Zhentao Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Hai Zhu
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Danjing Guo
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Rong Su
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Hui Chen
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Hui Li
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Xiao Xu
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Hangxiang Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, China
| |
Collapse
|
5
|
Johansen KH, Golec DP, Thomsen JH, Schwartzberg PL, Okkenhaug K. PI3K in T Cell Adhesion and Trafficking. Front Immunol 2021; 12:708908. [PMID: 34421914 PMCID: PMC8377255 DOI: 10.3389/fimmu.2021.708908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
PI3K signalling is required for activation, differentiation, and trafficking of T cells. PI3Kδ, the dominant PI3K isoform in T cells, has been extensively characterised using PI3Kδ mutant mouse models and PI3K inhibitors. Furthermore, characterisation of patients with Activated PI3K Delta Syndrome (APDS) and mouse models with hyperactive PI3Kδ have shed light on how increased PI3Kδ activity affects T cell functions. An important function of PI3Kδ is that it acts downstream of TCR stimulation to activate the major T cell integrin, LFA-1, which controls transendothelial migration of T cells as well as their interaction with antigen-presenting cells. PI3Kδ also suppresses the cell surface expression of CD62L and CCR7 which controls the migration of T cells across high endothelial venules in the lymph nodes and S1PR1 which controls lymph node egress. Therefore, PI3Kδ can control both entry and exit of T cells from lymph nodes as well as the recruitment to and retention of T cells within inflamed tissues. This review will focus on the regulation of adhesion receptors by PI3Kδ and how this contributes to T cell trafficking and localisation. These findings are relevant for our understanding of how PI3Kδ inhibitors may affect T cell redistribution and function.
Collapse
Affiliation(s)
- Kristoffer H Johansen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.,Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, United States
| | - Dominic P Golec
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, United States
| | - Julie H Thomsen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
mTOR Inhibitor Everolimus in Regulatory T Cell Expansion for Clinical Application in Transplantation. Transplantation 2019; 103:705-715. [PMID: 30451741 DOI: 10.1097/tp.0000000000002495] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Experimental and preclinical evidence suggest that adoptive transfer of regulatory T (Treg) cells could be an appropriate therapeutic strategy to induce tolerance and improve graft survival in transplanted patients. The University of Kentucky Transplant Service Line is developing a novel phase I/II clinical trial with ex vivo expanded autologous Treg cells as an adoptive cellular therapy in renal transplant recipients who are using everolimus (EVR)-based immunosuppressive regimen. METHODS The aim of this study was to determine the mechanisms of action and efficacy of EVR for the development of functionally competent Treg cell-based adoptive immunotherapy in transplantation to integrate a common EVR-based regimen in vivo (in the patient) and ex vivo (in the expansion of autologous Treg cells). CD25 Treg cells were selected from leukapheresis product with a GMP-compliant cell separation system and placed in 5-day (short) or 21-day (long) culture with EVR or rapamycin (RAPA). Multi-parametric flow cytometry analyses were used to monitor the expansion rates, phenotype, autophagic flux, and suppressor function of the cells. phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathway profiles of treated cells were analyzed by Western blot and cell bioenergetic parameters by extracellular flux analysis. RESULTS EVR-treated cells showed temporary slower growth, lower metabolic rates, and reduced phosphorylation of protein kinase B compared with RAPA-treated cells. In spite of these differences, the expansion rates, phenotype, and suppressor function of long-term Treg cells in culture with EVR were similar to those with RAPA. CONCLUSIONS Our results support the feasibility of EVR to expand functionally competent Treg cells for their clinical use.
Collapse
|
7
|
Jiang L, Liu T, Xie L, Ouyang C, Ji J, Huang T. AICAR prolongs corneal allograft survival via the AMPK-mTOR signaling pathway in mice. Biomed Pharmacother 2019; 113:108558. [PMID: 30856534 DOI: 10.1016/j.biopha.2019.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/06/2019] [Accepted: 01/06/2019] [Indexed: 12/11/2022] Open
Abstract
Immune rejection is a critical complication that results in the graft failure after corneal transplantation. Thus, there remains a need for new therapies for allograft rejection. AICAR (aminoimidazole-4-carboxamide ribonucleoside) is an, as adenosine monophosphate-activated protein kinase (AMPK) activator and a purine nucleoside with a wide range of metabolic effects, including activation of AMPK. More recently, it was reported that it is possible to inhibiting organs rejection and prolong the graft survival time in various models of organ transplantation. In this study, we systematically evaluated the efficacy of AICAR as a treatment modality for inhibiting allograft rejection in a mouse model of corneal transplantation. We found that AICAR significantly suppressed the opacity, edema, and vascularization of the graft, resulting in prolonged corneal allograft survival. AICAR treatment also significantly decreased central corneal thickness. Moreover, the AICAR-treated group showed decreased expression of IB4 and VEGF as compared to the control group. In addition, the mRNA expression of T helper 1 cytokines (IL-2, INF-γ, and TNF-α) was suppressed, and the expression of T helper 2 cytokines (IL-4, IL-5, and IL-13) was elevated by AICAR. Furthermore, the western blotting results revealed that AICAR stimulated AMPK activation and inhibited angiogenesis and inflammation possibly by subsequently suppressing mTOR phosphorylation. By contrast, the AMPK inhibitor Compound C (also called dorsomorphin) had the opposite effect. Our results showed that Compound C blocked AMPK-mTOR signaling and promoted the angiogenesis and inflammation, thus compromising the graft survival. These results suggest that AICAR may be a potential option for inhibiting the corneal graft rejection and for prolonging the graft survival.
Collapse
Affiliation(s)
- Li Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, PR China
| | - Tingting Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, PR China
| | - Lijie Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, PR China
| | - Chen Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, PR China
| | - Jianping Ji
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, PR China
| | - Ting Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, PR China.
| |
Collapse
|
8
|
Peptide Tk-PQ induces immunosuppression in skin allogeneic transplantation via increasing Foxp3 + Treg and impeding nuclear translocation of NF-κB. Mol Immunol 2018; 101:597-607. [PMID: 30001873 DOI: 10.1016/j.molimm.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/09/2018] [Accepted: 05/09/2018] [Indexed: 12/14/2022]
Abstract
Solid organ transplantation is used as the last resort for patients with end-stage disease, but allograft rejection is an unsolved problem. Here, we showed that Tk-PQ, a peptide derived from trichosanthin, had an immune-suppressive effect without obvious cytotoxicity in vitro and in a mouse skin allo-transplantation model. In vitro, treatment of Tk-PQ administrated type 2 T helper cell (Th2)/regulatory T-cell (Treg) cytokines, and increased the ratio of CD4+CD25+Foxp3+ Treg by repressing the PI3K/mTOR pathway. In addition, Tk-PQ decreased NF-κB activation to downregulate pro-inflammatory cytokines. Tk-PQ treatment in the mouse skin transplantation model also caused the similar molecular and cellular phenotypes. Furthermore, Tk-PQ enhanced the suppressive function of Treg by increasing Foxp3 expression, and substantially improved allograft survival. These finding demonstrate that Tk-PQ has the potential to be used in clinical allogeneic transplantation.
Collapse
|