1
|
Leung ML, Barrett‐Chan E, Levings MK, Vercauteren S, Blydt‐Hansen TD. Evaluating Activated Regulatory T Cells as a Biomarker of Chronic Allograft Inflammation in Pediatric Kidney Transplant Recipients. Pediatr Transplant 2025; 29:e70041. [PMID: 39924341 PMCID: PMC11807789 DOI: 10.1111/petr.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND There is a need for noninvasive immunological biomarkers that can identify stable kidney allograft immune quiescence to inform individualized immunosuppression. METHODS We conducted a cross-sectional, pilot cohort study evaluating the relative abundance of regulatory T cells (Tregs) to effector T-cell (Teff) populations as a surrogate marker of long-term graft tolerance. We obtained fresh peripheral blood mononuclear cell samples from stable pediatric kidney transplant recipients, most with recent surveillance biopsies to identify the presence or absence of chronic inflammation. Tregs were sub-phenotyped as naïve, memory, and activated Tregs (aTreg). Treg/Teff ratios were modeled for association with chronic inflammation and in the context of potential clinical features. RESULTS Twenty-seven patient samples were included on standard immunosuppression (tacrolimus, mycophenolate, and prednisone) with a mean age of 9.2 ± 5.0 years, at 30.2 ± 21.7 months posttransplant. The ratio of aTreg (FOXP3++CD45RA-) to Th17 cells (CD4+IL-17+) was significantly greater in patients without inflammation than in patients with graft inflammation (p < 0.01). Similarly, there was a trend toward greater aTreg/CD4+ T cells and aTreg/CD8+ Teff in patients without inflammation (p = 0.05 and 0.09, respectively). There was no significant association for inflammation with naïve or memory Treg/Teff ratios. Multiple logistic regression with all three aTreg/Teff ratios modeled allograft inflammation with high sensitivity and specificity (AUC = 0.83, 95% CI 0.67-0.98). CONCLUSIONS The proportion of peripheral blood aTregs/Teff cells in this pilot cohort of stable pediatric kidney transplant recipients was associated with immune quiescence. These data support further investigation into aTreg/Teff monitoring to inform precision immunosuppressive treatment.
Collapse
Affiliation(s)
- Macyn L. Leung
- BC Children's Hospital Research InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ella Barrett‐Chan
- BC Children's Hospital Research InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Megan K. Levings
- BC Children's Hospital Research InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of SurgeryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Suzanne Vercauteren
- BC Children's Hospital Research InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Tom D. Blydt‐Hansen
- BC Children's Hospital Research InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of PediatricsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
2
|
Avalos-de Leon CG, Thomson AW. Regulatory Immune Cell-derived Exosomes: Modes of Action and Therapeutic Potential in Transplantation. Transplantation 2025:00007890-990000000-00994. [PMID: 39865513 DOI: 10.1097/tp.0000000000005309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Reduced dependence on antirejection agents, improved long-term allograft survival, and induction of operational tolerance remain major unmet needs in organ transplantation due to the limitations of current immunosuppressive therapies. To address this challenge, investigators are exploring the therapeutic potential of adoptively transferred host- or donor-derived regulatory immune cells. Extracellular vesicles of endosomal origin (exosomes) secreted by these cells seem to be important contributors to their immunoregulatory properties. Twenty years ago, it was first reported that donor-derived exosomes could extend the survival of transplanted organs in rodents. Recent studies have revealed that regulatory immune cells, such as regulatory myeloid cells (dendritic cells, macrophages, or myeloid-derived suppressor cells), regulatory T cells, or mesenchymal stem/stromal cells can suppress graft rejection via exosomes that express a cargo of immunosuppressive molecules. These include cell surface molecules that interact with adaptive immune cell receptors, immunoregulatory enzymes, and micro- and long noncoding RNAs that can regulate inflammatory gene expression via posttranscriptional changes and promote tolerance through promotion of regulatory T cells. This overview analyzes the diverse molecules and mechanisms that enable regulatory immune cell-derived exosomes to modulate alloimmunity and promote experimental transplant tolerance. We also discuss the potential benefits and limitations of their application as therapeutic entities in organ transplantation.
Collapse
Affiliation(s)
- Cindy G Avalos-de Leon
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh PA
| | - Angus W Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh PA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA
| |
Collapse
|
3
|
Papa-Gobbi R, Stringa P, Gentilini MV, Ivanoff I, Machuca M, Arreola NM, Serradilla J, Estefanía-Fernández K, Talayero P, Velayos M, Sánchez—Zapardiel E, Gondolesi G, Andrés-Moreno A, Rumbo M, Hernández-Oliveros F. Low regulatory T-cells frequency is associated with graft rejection after small bowel transplantation: Clinical and experimental evidence. PLoS One 2025; 20:e0307534. [PMID: 39854413 PMCID: PMC11761612 DOI: 10.1371/journal.pone.0307534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 07/07/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Intestinal transplantation (ITx) represents the only curative option for patients with irreversible intestinal failure. Nevertheless, its rejection rate surpasses that of other solid organ transplants due to the heightened immunological load of the gut. Regulatory T-cells (Tregs) are key players in the induction and maintenance of peripheral tolerance, suggesting their potential involvement in modulating host vs. graft responses after ITx. Thus, we investigated the association of Tregs with allograft outcomes in pediatric patients and in an experimental model of small bowel transplantation. METHODS Treg frequency in human samples was analyzed by Flow cytometry (CD4+CD25highCD127-, blood samples) and immunohistochemistry (FoxP3, graft samples). Experimental allogenic-heterotopic small bowel transplantation was performed in rats and animals divided into 3 groups: non-immunosuppressant treatment, rapamycin (2 mg/kg), and tacrolimus (0.6 mg/kg) treatment. Acute cellular rejection (ACR) was diagnosed based on clinical and histological findings, graft gene expression of pro- and anti-inflammatory mediators assessed by RT-qPCR, serum IL-6 and IL-10 levels by Luminex, and Treg frequency analyzed by flow cytometry (CD4+CD25highFoxP3+). RESULTS Blood samples from patients undergoing ACR exhibited a significant reduction in the Treg number compared to those with normo-functional grafts. Similarly, a diminished number of FoxP3+ cells was observed in mucosa samples with ACR. In the experimental model, rapamycin-treated animals displayed clinical and histological findings resembling those not receiving immunosuppression treatment. Notably, ACR correlated with a high CD8/CD4 ratio, loss of T-cell chimerism, mRNA upregulation of pro-inflammatory genes and diminished graft Treg frequency. In contrast, tacrolimus treatment prevented ACR and facilitate blood and graft Treg expansion. Remarkably, recipients who achieved Treg expansion within the graft remained free of ACR even after discontinuation of the immunosuppressant treatment and this phenomenon was associated with increased levels of serum IL-10. CONCLUSION Our clinical and experimental findings underscore the association between Treg frequency and graft rejection after ITx, advocating for strategies that promote their expansion within the gut mucosa to enhance long-term outcomes.
Collapse
Affiliation(s)
- Rodrigo Papa-Gobbi
- Institute for Immunological and Pathophysiological Studies (IIFP), School of Exact Sciences, National University of La Plata, National Council of Scientific and Technical Research (CONICET), La Plata, Argentina
| | - Pablo Stringa
- Institute for Immunological and Pathophysiological Studies (IIFP), School of Exact Sciences, National University of La Plata, National Council of Scientific and Technical Research (CONICET), La Plata, Argentina
| | - Maria Virginia Gentilini
- Intestinal Failure, Rehabilitation and Transplant Unit, University Hospital Foundation Favaloro; Institute of Translational Medicine, Transplantation and Bioengineering (ImeTTyB), University Favaloro-CONICET, Buenos Aires, Argentina
| | - Ivana Ivanoff
- Institute for Immunological and Pathophysiological Studies (IIFP), School of Exact Sciences, National University of La Plata, National Council of Scientific and Technical Research (CONICET), La Plata, Argentina
| | - Mariana Machuca
- Special Pathology Laboratory, Faculty of Veterinary Sciences, National University of La Plata, La Plata, Buenos Aires, Argentina
| | - Nidia Monserrat Arreola
- Transplant Group, La Paz University Hospital Health Research Institute (IdiPAZ), Madrid, Spain
- Department of Pediatric Surgery, La Paz University Hospital, Madrid, Spain
| | - Javier Serradilla
- Transplant Group, La Paz University Hospital Health Research Institute (IdiPAZ), Madrid, Spain
- Department of Pediatric Surgery, La Paz University Hospital, Madrid, Spain
| | - Karla Estefanía-Fernández
- Transplant Group, La Paz University Hospital Health Research Institute (IdiPAZ), Madrid, Spain
- Department of Pediatric Surgery, La Paz University Hospital, Madrid, Spain
| | - Paloma Talayero
- Immunology Department, 12 de Octubre University Hospital, Madrid, Spain
| | - María Velayos
- Transplant Group, La Paz University Hospital Health Research Institute (IdiPAZ), Madrid, Spain
- Department of Pediatric Surgery, La Paz University Hospital, Madrid, Spain
| | | | - Gabriel Gondolesi
- Intestinal Failure, Rehabilitation and Transplant Unit, University Hospital Foundation Favaloro; Institute of Translational Medicine, Transplantation and Bioengineering (ImeTTyB), University Favaloro-CONICET, Buenos Aires, Argentina
| | - Ane Andrés-Moreno
- Transplant Group, La Paz University Hospital Health Research Institute (IdiPAZ), Madrid, Spain
- Department of Pediatric Surgery, La Paz University Hospital, Madrid, Spain
| | - Martin Rumbo
- Institute for Immunological and Pathophysiological Studies (IIFP), School of Exact Sciences, National University of La Plata, National Council of Scientific and Technical Research (CONICET), La Plata, Argentina
| | - Francisco Hernández-Oliveros
- Transplant Group, La Paz University Hospital Health Research Institute (IdiPAZ), Madrid, Spain
- Department of Pediatric Surgery, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
4
|
Filoni J, Ferrari A, Jofra T, Putignano AR, Da Dalt L, Cesarano S, Di Dedda C, Bonacina F, Marchesi F, Norata GD, Bonini C, Piemonti L, Monti P. Metabolic reprogramming of naïve regulatory T cells by IL-7 and IL-15 promotes their persistence and performance upon adoptive transfer. Commun Biol 2025; 8:99. [PMID: 39838096 PMCID: PMC11751088 DOI: 10.1038/s42003-024-07381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Tregs for adoptive therapy are traditionally expanded ex vivo using high doses of IL-2. However, the final Treg product has limited survival once infused in patients, potentially affecting therapeutic effectiveness. Here, we tested a novel expansion protocol in which highly purified naïve Tregs were expanded with a combination of IL-7 and IL-15, in the absence of IL-2. The final Treg product was enriched with cells displaying an immature CD45RA+CD62L+CD95+ phenotype, reminiscent of conventional memory stem T cells. The combination of IL-7 and IL-15 confers Tregs a glycolytic metabolism and improved metabolic fitness, characterized by an increased capacity to adapt metabolism according to glucose and oxygen availability. Tregs expanded with IL-7 and IL-15 showed longer persistence and an improved capacity to control xeno-GvHD in NSG mice. This work suggests that metabolic reprogramming induced by IL-7 and IL-15 provides better Treg performance for adoptive therapy.
Collapse
Affiliation(s)
- Jessica Filoni
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Arianna Ferrari
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Tatiana Jofra
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Anna Rita Putignano
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital Rozzano, Rozzano, Italy
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Susanna Cesarano
- Experimental Hematology Unit, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Carla Di Dedda
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Federica Marchesi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, IRCCS Ospedale San Raffaele Milan, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Monti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy.
| |
Collapse
|
5
|
Singer M, Elsayed AM, Husseiny MI. Regulatory T-cells: The Face-off of the Immune Balance. FRONT BIOSCI-LANDMRK 2024; 29:377. [PMID: 39614434 DOI: 10.31083/j.fbl2911377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Regulatory T-cells (Tregs) play a crucial role in maintaining immune homeostasis, ensuring a balanced immune response. Tregs primarily operate in an antigen-specific fashion, facilitated by their distinct distribution within discrete niches. Tregs have been studied extensively, from their point of origin in the thymus origin to their fate in the periphery or organs. Signals received from antigen-presenting cells (APCs) stimulate Tregs to dampen inflammation. Almost all tumors are characterized by a pathological abundance of immune suppression in their microenvironment. Conversely, the lack thereof proves detrimental to immunological disorders. Achieving a balanced expression of Tregs in relation to other immune compartments is important in establishing an effective and adaptable immune tolerance towards cancer cells and autoantigens. In the context of cancer, it is essential to decrease the frequency of Tregs to overcome tumor suppression. A lower survival rate is associated with the presence of excessive exhausted effector immune cells and an increased frequency of regulatory cells. However, when it comes to treating graft rejection and autoimmune diseases, the focus lies on immune tolerance and the transfer of Tregs. Here, we explore the complex mechanisms that Tregs use in human disease to balance effector immune cells.
Collapse
Affiliation(s)
- Mahmoud Singer
- School of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - Ahmed M Elsayed
- Division of Infectious Diseases, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
6
|
Jin X, Pirenne J, Vos R, Hooft C, Kaes J, Van Slambrouck J, Kortleven P, Vandervelde C, Beeckmans H, Kerckhof P, Carlon MS, Van Raemdonck D, Looney MR, Vanaudenaerde BM, Ceulemans LJ. Donor-Specific Blood Transfusion in Lung Transplantation. Transpl Int 2024; 37:12822. [PMID: 39553536 PMCID: PMC11565953 DOI: 10.3389/ti.2024.12822] [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: 02/08/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024]
Abstract
Lung transplantation is still hindered by a high rate of chronic rejection necessitating profound immunosuppression with its associated complications. Donor-specific blood transfusion is a pre-transplant strategy aimed at improving graft acceptance. In contrast with standard stored blood or donor-specific regulatory T cells transfusions, this approach utilizes fresh whole blood from the donor prior to allograft transplantation, encompassing all cell types and plasma. The precise mechanisms underlying donor-specific blood transfusion-induced tolerance remain incompletely understood. Associations with regulatory/helper T cells, modulation of mononuclear phagocytic cells or microchimerism have been suggested. While numerous (pre-)clinical studies have explored its application in solid organ transplants like liver, kidney, and intestine, limited attention has been given to the setting of lung transplantation. This comprehensive review summarizes existing knowledge on the mechanisms and outcomes of donor-specific blood transfusion in solid organ transplants both in preclinical and clinical settings. We also address the potential benefits and risks associated with donor-specific blood transfusion in the field of lung transplantation, offering insights into future research directions.
Collapse
Affiliation(s)
- Xin Jin
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Department of Microbiology, Immunology and Transplantation, Transplantation Research Group, Lab of Abdominal Transplantation, KU Leuven, Leuven, Belgium
- Department of Abdominal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Charlotte Hooft
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Janne Kaes
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism (VIB-KU Leuven), KU Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Phéline Kortleven
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Christelle Vandervelde
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Hanne Beeckmans
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Pieterjan Kerckhof
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Marianne S. Carlon
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Dirk Van Raemdonck
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Bart M. Vanaudenaerde
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Laurens J. Ceulemans
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Li H, Xian Y, Ma M, Zhang K, Hong L. Prospective study on immune function in renal transplant patients during perioperative period: A prospective cohort study. Medicine (Baltimore) 2024; 103:e40070. [PMID: 39432644 PMCID: PMC11495714 DOI: 10.1097/md.0000000000040070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024] Open
Abstract
Delayed graft function (DGF) is a type of acute renal failure that is closely linked to the immune system. The objective of this study is to investigate immune trends during the perioperative period of renal transplantation and compare the variations between patients with DGF and immediate graft function (IGF). A total of 48 kidney transplant patients were enrolled. Parameters including stimulated adenosine triphosphatase concentrations (sATP), nonstimulated ATP concentrations, white blood cells, and lymphocyte count were assessed. Patients were categorized into the DGF or IGF group. Clinical information and changes in immune markers were compared. Receiver operating characteristic analysis was performed to determine the sensitivity and specificity in predicting DGF. Additionally, separate immune function analyses were conducted for the 3 infection cases. Following induction immunosuppressive therapy, white blood cells, and neutrophil count showed a significant initial increase followed by a gradual decline. Lymphocyte count, nonstimulated ATP concentrations, and sATP exhibited an initial significant decrease followed by a slow recovery. Immune markers between the DGF and IGF groups were significantly different at day 4 after renal transplantation. Only sATP levels at day 4 after renal transplantation (area under the curve = 0.731, sensitivity = 0.864, specificity = 0.684) demonstrated predictive value for DGF occurrence. Among the 3 infection cases, 2 cases exhibited persistently decreased sATP levels and died within the first month and 6 months, while the remaining case showed a recovery of sATP levels at D9 and survived. These findings indicate that sATP level can potentially serve as a biomarker reflecting the impact of immunosuppressants. Poor recovery of sATP may be associated with DGF, infection, or even mortality.
Collapse
Affiliation(s)
- Heng Li
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying Xian
- General Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Maolin Ma
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kouxing Zhang
- General Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liangqing Hong
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Iesari S, Nava FL, Zais IE, Coubeau L, Ferraresso M, Favi E, Lerut J. Advancing immunosuppression in liver transplantation: A narrative review. Hepatobiliary Pancreat Dis Int 2024; 23:441-448. [PMID: 38523030 DOI: 10.1016/j.hbpd.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Immunosuppression is essential to ensure recipient and graft survivals after liver transplantation (LT). However, our understanding and management of the immune system remain suboptimal. Current immunosuppressive therapy cannot selectively inhibit the graft-specific immune response and entails a significant risk of serious side effects, i.e., among others, de novo cancers, infections, cardiovascular events, renal failure, metabolic syndrome, and late graft fibrosis, with progressive loss of graft function. Pharmacological research, aimed to develop alternative immunosuppressive agents in LT, is behind other solid-organ transplantation subspecialties, and, therefore, the development of new compounds and strategies should get priority in LT. The research trajectories cover mechanisms to induce T-cell exhaustion, to inhibit co-stimulation, to mitigate non-antigen-specific inflammatory response, and, lastly, to minimize the development and action of donor-specific antibodies. Moreover, while cellular modulation techniques are complex, active research is underway to foster the action of T-regulatory cells, to induce tolerogenic dendritic cells, and to promote the function of B-regulatory cells. We herein discuss current lines of research in clinical immunosuppression, particularly focusing on possible applications in the LT setting.
Collapse
Affiliation(s)
- Samuele Iesari
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Francesca Laura Nava
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Ilaria Elena Zais
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Laurent Coubeau
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium; Service de Chirurgie et Transplantation Abdominale, Cliniques Universitaires Saint-Luc, 55 Avenue Hippocrate, 1200 Brussels, Belgium
| | - Mariano Ferraresso
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 19 Via della Commenda, 20122 Milan, Italy
| | - Evaldo Favi
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 19 Via della Commenda, 20122 Milan, Italy.
| | - Jan Lerut
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
| |
Collapse
|
9
|
Xu Y, Ren S, Wang H, Qin Y, Liu T, Sun C, Xiao Y, Shao B, Zhang J, Chen Q, Zhao P, Yang G, Liu X, Wang H. Endometrial regeneration cell-derived exosomes loaded with siSLAMF6 inhibit cardiac allograft rejection through the suppression of desialylation modification. Cell Mol Biol Lett 2024; 29:128. [PMID: 39354345 PMCID: PMC11443917 DOI: 10.1186/s11658-024-00645-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUNDS Acute transplant rejection is a major component of poor prognoses for organ transplantation. Owing to the multiple complex mechanisms involved, new treatments are still under exploration. Endometrial regenerative cells (ERCs) have been widely used in various refractory immune-related diseases, but the role of ERC-derived exosomes (ERC-Exos) in alleviating transplant rejection has not been extensively studied. Signaling lymphocyte activation molecule family 6 (SLAMF6) plays an important role in regulating immune responses. In this study, we explored the main mechanism by which ERC-Exos loaded with siSLAMF6 can alleviate allogeneic transplant rejection. METHODS C57BL/6 mouse recipients of BALB/c mouse kidney transplants were randomly divided into four groups and treated with exosomes. The graft pathology was evaluated by H&E staining. Splenic and transplanted heart immune cell populations were analyzed by flow cytometry. Recipient serum cytokine profiles were determined by enzyme-linked immunosorbent assay (ELISA). The proliferation and differentiation capacity of CD4+ T cell populations were evaluated in vitro. The α-2,6-sialylation levels in the CD4+ T cells were determined by SNA blotting. RESULTS In vivo, mice treated with ERC-siSLAMF6 Exo achieved significantly prolonged allograft survival. The serum cytokine profiles of the recipients were significantly altered in the ERC-siSLAMF6 Exo-treated recipients. In vitro, we found that ERC-siSLAMF6-Exo considerably downregulated α-2,6-sialyltransferase (ST6GAL1) expression in CD4+ T cells, and significantly reduced α-2,6-sialylation levels. Through desialylation, ERC-siSLAMF6 Exo therapy significantly decreased CD4+ T cell proliferation and inhibited CD4+ T cell differentiation into Th1 and Th17 cells while promoting regulatory T cell (Treg) differentiation. CONCLUSIONS Our study indicated that ERC-Exos loaded with siSLAMF6 reduce the amount of sialic acid connected to α-2,6 at the end of the N-glycan chain on the CD4+ T cell surface, increase the number of therapeutic exosomes endocytosed into CD4+ T cells, and inhibit the activation of T cell receptor signaling pathways, which prolongs allograft survival. This study confirms the feasibility of using ERC-Exos as natural carriers combined with gene therapy, which could be used as a potential therapeutic strategy to alleviate allograft rejection.
Collapse
Affiliation(s)
- Yini Xu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shaohua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hongda Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yafei Qin
- Department of Vascular Surgery, Henan Provincial People's Hospital, The Affiliated People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chenglu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yiyi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jingyi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Qiang Chen
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Pengyu Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Guangmei Yang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xu Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
10
|
Blinova VG, Zhdanov DD. Many Faces of Regulatory T Cells: Heterogeneity or Plasticity? Cells 2024; 13:959. [PMID: 38891091 PMCID: PMC11171907 DOI: 10.3390/cells13110959] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining the immune balance in normal and pathological conditions. In autoimmune diseases and transplantation, they restrain the loss of self-tolerance and promote engraftment, whereas in cancer, an increase in Treg numbers is mostly associated with tumor growth and poor prognosis. Numerous markers and their combinations have been used to identify Treg subsets, demonstrating the phenotypic diversity of Tregs. The complexity of Treg identification can be hampered by the unstable expression of some markers, the decrease in the expression of a specific marker over time or the emergence of a new marker. It remains unclear whether such phenotypic shifts are due to new conditions or whether the observed changes are due to initially different populations. In the first case, cellular plasticity is observed, whereas in the second, cellular heterogeneity is observed. The difference between these terms in relation to Tregs is rather blurred. Considering the promising perspectives of Tregs in regenerative cell-based therapy, the existing confusing data on Treg phenotypes require further investigation and analysis. In our review, we introduce criteria that allow us to distinguish between the heterogeneity and plasticity of Tregs normally and pathologically, taking a closer look at their diversity and drawing the line between two terms.
Collapse
Affiliation(s)
- Varvara G. Blinova
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia;
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia;
- Department of Biochemistry, People’s Friendship University of Russia Named after Patrice Lumumba (RUDN University), Miklukho-Maklaya st. 6, 117198 Moscow, Russia
| |
Collapse
|
11
|
Ravella S. Association between oral nutrition and inflammation after intestinal transplantation. Hum Immunol 2024; 85:110809. [PMID: 38724327 DOI: 10.1016/j.humimm.2024.110809] [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: 01/18/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024]
Abstract
Intestinal transplantation (Itx) can be a life-saving treatment for certain patient populations, including those patients with intestinal failure (IF) who develop life-threatening complications due to the use of parenteral nutrition (PN). Most patients who have undergone Itx are eventually able to tolerate a full oral diet. However, little guidance or consensus exists regarding optimizing the specific components of an oral diet for Itx patients, including macronutrients, micronutrients and dietary patterns. While oral dietary prescriptions have moved to the forefront of primary and preventive care, this movement has yet to occur across the field of organ transplantation. Evidence to date points to the role of systemic chronic inflammation (SCI) in a wide variety of chronic diseases as well as post-transplant graft dysfunction. This review will discuss current trends in oral nutrition for Itx patients and also offer novel insights into nutritional management techniques that may help to decrease SCI and chronic disease risk as well as optimize graft function.
Collapse
|
12
|
Chen W, Toda E, Takeuchi K, Sawa Y, Wakamatsu K, Kuwahara N, Ishikawa A, Igarashi Y, Terasaki M, Kunugi S, Terasaki Y, Yamada K, Terashima Y, Shimizu A. Disulfiram treatment suppresses antibody-producing reactions by inhibiting macrophage activation and B cell pyrimidine metabolism. Commun Biol 2024; 7:488. [PMID: 38649462 PMCID: PMC11035657 DOI: 10.1038/s42003-024-06183-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Antibody responses, involving B cells, CD4 + T cells, and macrophages, are implicated in autoimmune diseases and organ transplant rejection. We have previously shown that inhibiting FROUNT with disulfiram (DSF) suppresses macrophage activation and migration, effectively treating inflammatory diseases. In this study, we investigated the effectiveness of DSF in antibody-producing reactions. Using a heart transplantation mouse model with antibody-mediated rejection, we administered anti-CD8 antibody to exclude cellular rejection. DSF directly inhibited B cell responses in vitro and significantly reduced plasma donor-specific antibodies and graft antibody deposition in vivo, resulting in prolonged survival of the heart graft. DSF also mediated various effects, including decreased macrophage infiltration and increased Foxp3+ regulatory T-cells in the grafts. Additionally, DSF inhibited pyrimidine metabolism-related gene expression induced by B-cell stimulation. These findings demonstrate that DSF modulates antibody production in the immune response complexity by regulating B-cell and macrophage responses.
Collapse
Affiliation(s)
- Weili Chen
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Etsuko Toda
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan.
- Laboratory for Morphological and Biomolecular Imaging, Nippon Medical School, Tokyo, Japan.
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan.
| | - Kazuhiro Takeuchi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Yurika Sawa
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Kyoko Wakamatsu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Naomi Kuwahara
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Arimi Ishikawa
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Yuri Igarashi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Mika Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Shinobu Kunugi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | | | - Kazuhiko Yamada
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Yuya Terashima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan.
| |
Collapse
|
13
|
Yang TT, Liu PJ, Sun QY, Wang ZY, Yuan GB, Fan ZX, Ma L, Lu JF, Yuan BY, Zou WL, Zhao LM, Li Q, Liu GZ. CD4 +CD25 + regulatory T cells ex vivo generated from autologous naïve CD4 + T cells suppress EAE progression. Sci Rep 2024; 14:6262. [PMID: 38491084 PMCID: PMC10943184 DOI: 10.1038/s41598-024-56739-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/11/2024] [Indexed: 03/18/2024] Open
Abstract
CD4+CD25+ regulatory T cells (Tregs) play an important role in maintaining immune homeostasis in multiple sclerosis (MS). Hence, we aimed to explore the therapeutic efficacy and safety of adoptive cell therapy (ACT) utilizing induced antigen-specific Tregs in an animal model of MS, that is, in an experimental autoimmune encephalomyelitis (EAE) model. B cells from EAE model that were activated with soluble CD40L were used as antigen-presenting cells (APCs) to induce the differentiation of antigen-specific Tregs from naïve CD4 precursors, and then, a stepwise isolation of CD4+CD25highCD127low Tregs was performed using a flow sorter. All EAE mice were divided into Treg-treated group (2 × 104 cells in 0.2 mL per mouse, n = 14) and sham-treated group (0.2 mL normal saline (NS), n = 20), which were observed daily for clinical assessment, and for abnormal appearance for 6 weeks. Afterward, histological analysis, immunofluorescence and real-time PCR were performed. Compared to sham-treated mice, Treg-treated mice exhibited a significant decrease in disease severity scores and reduced inflammatory infiltration and demyelination in the spinal cord. Additionally, Tregs-treated mice demonstrated higher CCN3 protein and mRNA levels than sham-treated mice. The results of this preclinical study further support the therapeutic potential of this ACT approach in the treatment of MS.
Collapse
Affiliation(s)
- Ting-Ting Yang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Pen-Ju Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qing-Yu Sun
- Department of Anesthesiology, Chang Hai Hospital, Naval Military Medical University, Shanghai, China
| | - Ze-Yi Wang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guo-Bin Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ze-Xin Fan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lin Ma
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jian-Feng Lu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bo-Yi Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wen-Long Zou
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Li-Min Zhao
- Experimental Center, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Qian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Guang-Zhi Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Lee S, Carrow JK, Fraser LA, Yan J, Jeyamogan S, Sambandam Y, Clemons TD, Kolberg-Edelbrock AN, He J, Mathew J, Zhang ZJ, Leventhal JP, Gallon L, Palmer LC, Stupp SI. Single-cell coating with biomimetic extracellular nanofiber matrices. Acta Biomater 2024; 177:50-61. [PMID: 38331132 DOI: 10.1016/j.actbio.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/17/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Cell therapies offer great promise in the treatment of diseases and tissue regeneration, but their clinical use has many challenges including survival, optimal performance in their intended function, or localization at sites where they are needed for effective outcomes. We report here on a method to coat a biodegradable matrix of biomimetic nanofibers on single cells that could have specific functions ranging from cell signaling to targeting and helping cells survive when used for therapies. The fibers are composed of peptide amphiphile (PA) molecules that self-assemble into supramolecular nanoscale filaments. The PA nanofibers were able to create a mesh-like coating for a wide range of cell lineages with nearly 100 % efficiency, without interrupting the natural cellular phenotype or functions. The targeting abilities of this system were assessed in vitro using human primary regulatory T (hTreg) cells coated with PAs displaying a vascular cell adhesion protein 1 (VCAM-1) targeting motif. This approach provides a biocompatible method for single-cell coating that does not negatively alter cellular phenotype, binding capacity, or immunosuppressive functionality, with potential utility across a broad spectrum of cell therapies. STATEMENT OF SIGNIFICANCE: Cell therapies hold great promise in the treatment of diseases and tissue regeneration, but their clinical use has been limited by cell survival, targeting, and function. We report here a method to coat single cells with a biodegradable matrix of biomimetic nanofibers composed of peptide amphiphile (PA) molecules. The nanofibers were able to coat cells, such as human primary regulatory T cells, with nearly 100 % efficiency, without interrupting the natural cellular phenotype or functions. The approach provides a biocompatible method for single-cell coating that does not negatively alter cellular phenotype, binding capacity, or immunosuppressive functionality, with potential utility across a broad spectrum of cell therapies.
Collapse
Affiliation(s)
- Slgirim Lee
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - James K Carrow
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Lewis A Fraser
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Jianglong Yan
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Shareni Jeyamogan
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Yuvaraj Sambandam
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Tristan D Clemons
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States; Department of Chemistry, Northwestern University, Evanston, IL 60208, United States
| | - Alexandra N Kolberg-Edelbrock
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, United States
| | - Jie He
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - James Mathew
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States; Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Zheng Jenny Zhang
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States; Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Joseph P Leventhal
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States; Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Lorenzo Gallon
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States; Department of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Liam C Palmer
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States; Department of Chemistry, Northwestern University, Evanston, IL 60208, United States.
| | - Samuel I Stupp
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States; Department of Chemistry, Northwestern University, Evanston, IL 60208, United States; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, United States; Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, United States; Department of Medicine, Northwestern University, Chicago, IL 60611, United States.
| |
Collapse
|
15
|
Yao M, Henriksson J, Fahlander H, Guisti Coitinho P, Lundgren T, Ågren N, Ericzon BG, Kumagai-Braesch M. Evaluation of Methods to Obtain Peripheral Blood Mononuclear Cells From Deceased Donors for Tolerance-Induction Protocols. Cell Transplant 2024; 33:9636897241256462. [PMID: 38808671 PMCID: PMC11143843 DOI: 10.1177/09636897241256462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024] Open
Abstract
Regulatory cell therapies have shown promise in tolerance-induction protocols in living donor organ transplantation. These protocols should be pursued in deceased donor transplantation. Donor peripheral mononuclear cells (PBMCs) are an optimal source of donor antigens for the induction of donor-specific regulatory cells. During the development of a regulatory cell tolerance-induction protocol with organs from deceased donors, we compared 3 methods of obtaining PBMCs from deceased donors focusing on cell yield, viability, and contamination of unwanted cell types. PBMC procurement methods: 1. During organ procurement at the time of cold perfusion, blood was collected from the vena cava and placed into a 10-liter blood collection bag, and thereafter transported to Karolinska University Hospital, where leukapheresis was performed (BCL). 2. Blood was collected via the vena cava into blood donation bags before cold perfusion. The bags underwent buffy coat separation and thereafter automated leukocyte isolation system (BCS). 3. To collect PBMCs, leukapheresis was performed via a central dialysis catheter on deceased donors in the intensive care unit (ICU) prior to the organ procurement procedure (LEU).All 3 methods to obtain PBMC from deceased donors were safe and did not affect the procurement of organs. BCL contained around 50% of NK cells in lymphocytes population. LEU had a highest yield of donor PBMC among 3 groups. LEU had the lower amount of granulocyte contamination, compared to BCS and BCL. Based on these results, we choose LEU as the preferred method to obtain donor PBMC in the development of our tolerance-induction protocol.
Collapse
Affiliation(s)
- Ming Yao
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Jarmo Henriksson
- Centre for Apheresis and Stem Cell Laboratory, KITM, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Henrik Fahlander
- Centre for Apheresis and Stem Cell Laboratory, KITM, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Pablo Guisti Coitinho
- Centre for Apheresis and Stem Cell Laboratory, KITM, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Torbjörn Lundgren
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Nils Ågren
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Bo-Göran Ericzon
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Makiko Kumagai-Braesch
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|
16
|
Baron KJ, Turnquist HR. Clinical Manufacturing of Regulatory T Cell Products For Adoptive Cell Therapy and Strategies to Improve Therapeutic Efficacy. Organogenesis 2023; 19:2164159. [PMID: 36681905 PMCID: PMC9870008 DOI: 10.1080/15476278.2022.2164159] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Based on successes in preclinical animal transplant models, adoptive cell therapy (ACT) with regulatory T cells (Tregs) is a promising modality to induce allograft tolerance or reduce the use of immunosuppressive drugs to prevent rejection. Extensive work has been done in optimizing the best approach to manufacture Treg cell products for testing in transplant recipients. Collectively, clinical evaluations have demonstrated that large numbers of Tregs can be expanded ex vivo and infused safely. However, these trials have failed to induce robust drug-free tolerance and/or significantly reduce the level of immunosuppression needed to prevent solid organ transplant (SOTx) rejection. Improving Treg therapy effectiveness may require increasing Treg persistence or orchestrating Treg migration to secondary lymphatic tissues or places of inflammation. In this review, we describe current clinical Treg manufacturing methods used for clinical trials. We also highlight current strategies being implemented to improve delivered Treg ACT persistence and migration in preclinical studies.
Collapse
Affiliation(s)
- Kassandra J. Baron
- Departments of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Department of Infectious Disease and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Hēth R. Turnquist
- Departments of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,CONTACT Hēth R. Turnquist Departments of Surgery, University of Pittsburgh School of Medicine, Thomas E. Starzl Transplantation Institute 200 Lothrop Street, BST W1542, PittsburghPA 15213, USA
| |
Collapse
|
17
|
Elalouf A, Elalouf H, Rosenfeld A. Modulatory immune responses in fungal infection associated with organ transplant - advancements, management, and challenges. Front Immunol 2023; 14:1292625. [PMID: 38143753 PMCID: PMC10748506 DOI: 10.3389/fimmu.2023.1292625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Organ transplantation stands as a pivotal achievement in modern medicine, offering hope to individuals with end-stage organ diseases. Advancements in immunology led to improved organ transplant survival through the development of immunosuppressants, but this heightened susceptibility to fungal infections with nonspecific symptoms in recipients. This review aims to establish an intricate balance between immune responses and fungal infections in organ transplant recipients. It explores the fundamental immune mechanisms, recent advances in immune response dynamics, and strategies for immune modulation, encompassing responses to fungal infections, immunomodulatory approaches, diagnostics, treatment challenges, and management. Early diagnosis of fungal infections in transplant patients is emphasized with the understanding that innate immune responses could potentially reduce immunosuppression and promise efficient and safe immuno-modulating treatments. Advances in fungal research and genetic influences on immune-fungal interactions are underscored, as well as the potential of single-cell technologies integrated with machine learning for biomarker discovery. This review provides a snapshot of the complex interplay between immune responses and fungal infections in organ transplantation and underscores key research directions.
Collapse
Affiliation(s)
- Amir Elalouf
- Department of Management, Bar-Ilan University, Ramat Gan, Israel
| | - Hadas Elalouf
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| | - Ariel Rosenfeld
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
18
|
Cosorich I, Filoni J, Di Dedda C, Ferrari A, Jofra T, Cesarano S, Bonini C, Piemonti L, Monti P. Interleukin-7 improves the fitness of regulatory T cells for adoptive transfer. Immunology 2023; 170:540-552. [PMID: 37671510 DOI: 10.1111/imm.13690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/18/2023] [Indexed: 09/07/2023] Open
Abstract
Adoptive regulatory T-cell (Treg) transfer has emerged as a promising therapeutic strategy for regulating immune responses in organ transplantation, graft versus host disease, and autoimmunity, including Type 1 diabetes. Traditionally, Treg for adoptive therapy have been sorted and expanded in vitro using high doses of IL-2, demonstrating stability and suppressive capabilities. However, limitations in their long-term survival post-infusion into patients have been observed. To address this challenge, we investigated a novel expansion protocol incorporating interleukin-7 (IL-7) alongside the traditional method utilizing IL-2 (referred to as IL-7 method, IL-7M). Our study revealed that naïve Treg express significant levels of CD127 and display robust responsiveness to IL-7, characterized by STAT-5 phosphorylation. Expanding naïve Treg with the IL-7M protocol led to a substantial enrichment of CD45RA+ CD62L+ CD95+ Treg but showing a reduction in the final cell yield and suppressive function. Moreover, Treg expanded with the IL-7M exhibited preserved telomere length and demonstrated enhanced resistance to cytokine withdrawal and fas-mediated apoptosis. When transferred into NSG mice IL-7M-Treg persisted longer and reduced the expansion of T cells, but did not significantly reduce the severity of xenoGvHD. In conclusion, our data demonstrate the feasibility of expanding naïve Treg in the presence of IL-7 to generate a Treg product enriched in poorly differentiated CD45RA+ cells with enhanced survival capabilities.
Collapse
Affiliation(s)
- Ilaria Cosorich
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Jessica Filoni
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Carla Di Dedda
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Arianna Ferrari
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Tatiana Jofra
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Susanna Cesarano
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Monti
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| |
Collapse
|
19
|
Guinan EC, Contreras-Ruiz L, Crisalli K, Rickert C, Rosales I, Makar R, Colvin R, Geissler EK, Sawitzki B, Harden P, Tang Q, Blancho G, Turka LA, Markmann JF. Donor antigen-specific regulatory T cell administration to recipients of live donor kidneys: A ONE Study consortium pilot trial. Am J Transplant 2023; 23:1872-1881. [PMID: 37422112 DOI: 10.1016/j.ajt.2023.06.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 07/10/2023]
Abstract
Regulatory T cells (Tregs) can inhibit cellular immunity in diverse experimental models and have entered early phase clinical trials in autoimmunity and transplantation to assess safety and efficacy. As part of the ONE Study consortium, we conducted a phase I-II clinical trial in which purified donor antigen reactive (dar)-Tregs (CD4+CD25+CD127lo) were administered to 3 patients, 7 to 11 days after live donor renal transplant. Recipients received a modified immunosuppression regimen, without induction therapy, consisting of maintenance tacrolimus, mycophenolate mofetil, and steroids. Steroids were weaned off over 14 weeks. No rejection was seen on any protocol biopsy. Therefore, all patients discontinued mycophenolate mofetil 11 to 13 months posttransplant, per protocol. An early for-cause biopsy in 1 patient, 5 days after dar-Treg infusion, revealed absence of rejection and accumulation of Tregs in the kidney allograft. All patients had Treg-containing lymphoid aggregates evident on protocol biopsies performed 8 months posttransplant. The patients are now all >6 years posttransplant on tacrolimus monotherapy with excellent graft function. None experienced rejection episodes. No serious adverse events were attributable to Treg administration. These results support a favorable safety profile of dar-Tregs administered early after renal transplant, suggest early biopsy might be an instructive research endpoint and provide preliminary evidence of potential immunomodulatory activity.
Collapse
Affiliation(s)
- Eva C Guinan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.
| | - Laura Contreras-Ruiz
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.
| | - Kerry Crisalli
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | - Charles Rickert
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | - Ivy Rosales
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | - Robert Makar
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | - Robert Colvin
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | - Edward K Geissler
- University Hospital Regensburg, Department of Surgery, Regensburg, Germany.
| | - Birgit Sawitzki
- Institute of Medical Immunology, Virchow - Klinikum, Berlin, Germany.
| | - Paul Harden
- Oxford Transplant Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Qizhi Tang
- Division of Transplantation, Department of Surgery, University of California, San Francisco, California, USA.
| | - Giles Blancho
- Centre of Research in Transplantation and Immunology, Nantes University, Nantes, France.
| | - Laurence A Turka
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | - James F Markmann
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
20
|
Vafadar A, Vosough P, Jahromi HK, Tajbakhsh A, Savardshtaki A, Butler AE, Sahebkar A. The role of efferocytosis and transplant rejection: Strategies in promoting transplantation tolerance using apoptotic cell therapy and/or synthetic particles. Cell Biochem Funct 2023; 41:959-977. [PMID: 37787641 DOI: 10.1002/cbf.3852] [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: 06/05/2023] [Revised: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023]
Abstract
Recently, efforts have been made to recognize the precise reason(s) for transplant failure and the process of rejection utilizing the molecular signature. Most transplant recipients do not appreciate the unknown length of survival of allogeneic grafts with the existing standard of care. Two noteworthy immunological pathways occur during allogeneic transplant rejection. A nonspecific innate immune response predominates in the early stages of the immune reaction, and allogeneic antigens initiate a donor-specific adaptive reaction. Though the adaptive response is the major cause of allograft rejection, earlier pro-inflammatory responses that are part of the innate immune response are also regarded as significant in graft loss. The onset of the innate and adaptive immune response causes chronic and acute transplant rejection. Currently employed immunosuppressive medications have shown little or no influence on chronic rejection and, as a result, on overall long-term transplant survival. Furthermore, long-term pharmaceutical immunosuppression is associated with side effects, toxicity, and an increased risk of developing diseases, both infectious and metabolic. As a result, there is a need for the development of innovative donor-specific immunosuppressive medications to regulate the allorecognition pathways that induce graft loss and to reduce the side effects of immunosuppression. Efferocytosis is an immunomodulatory mechanism with fast and efficient clearance of apoptotic cells (ACs). As such, AC therapy strategies have been suggested to limit transplant-related sequelae. Efferocytosis-based medicines/treatments can also decrease the use of immunosuppressive drugs and have no detrimental side effects. Thus, this review aims to investigate the impact of efferocytosis on transplant rejection/tolerance and identify approaches using AC clearance to increase transplant viability.
Collapse
Affiliation(s)
- Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Kargar Jahromi
- Research Center for Non-Communicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Amir Tajbakhsh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardshtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland - Bahrain, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Chang SH, Park CG. Comparing the Benefits and Drawbacks of Stem Cell Therapy Based on the Cell Origin or Manipulation Process: Addressing Immunogenicity. Immune Netw 2023; 23:e44. [PMID: 38188600 PMCID: PMC10767552 DOI: 10.4110/in.2023.23.e44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/12/2023] [Indexed: 01/09/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are effective in treating autoimmune diseases and managing various conditions, such as engraftment of allogeneic islets. Additionally, autologous and HLA-matched allogeneic MSCs can aid in the engraftment of human allogeneic kidneys with or without low doses of tacrolimus, respectively. However, HLA alloantigens are problematic because cell therapy uses more HLA-mismatched allogeneic cells than autologous for convenience and standardization. In particular, HLA-mismatched MSCs showed increased Ag-specific T/B cells and reduced viability faster than HLA-matched MSCs. In CRISPR/Cas9-based cell therapy, Cas9 induce T cell activation in the recipient's immune system. Interestingly, despite their immunogenicity being limited to the cells with foreign Ags, the accumulation of HLA alloantigen-sensitized T/B cells may lead to allograft rejection, suggesting that alloantigens may have a greater scope of adverse effects than foreign Ags. To avoid alloantigen recognition, the β2-microglobulin knockout (B2MKO) system, eliminating class-I MHC, was able to avoid rejection by alloreactive CD8 T cells compared to controls. Moreover, universal donor cells in which both B2M and Class II MHC transactivator (CIITA) were knocked out was more effective in avoiding immune rejection than single KO. However, B2MKO and CIITA KO system remain to be controlled and validated for adverse effects such as the development of tumorigenicity due to deficient Ag recognition by CD8 T and CD4 T cells, respectively. Overall, better HLA-matching or depletion of HLA alloantigens prior to cell therapy can reduce repetitive transplantation through the long-term survival of allogeneic cell therapy, which may be especially important for patients seeking allogeneic transplantation.
Collapse
Affiliation(s)
- Sung-Ho Chang
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Korea
| | - Chung Gyu Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Transplantation Research Institute, Medical Research center, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
22
|
Menasché P. Human PSC-derived cardiac cells and their products: therapies for cardiac repair. J Mol Cell Cardiol 2023; 183:14-21. [PMID: 37595498 DOI: 10.1016/j.yjmcc.2023.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/20/2023]
Abstract
Despite the dramatic improvements in the management of patients with chronic heart failure which have occurred over the last decades, some of them still exhaust conventional drug-based therapies without being eligible for more aggressive options like heart transplantation or implantation of a left ventricular assist device. Cell therapy has thus emerged as a possible means of filling this niche. Multiple cell types have now been tested both in the laboratory but also in the clinics and it is fair to acknowledge that none of the clinical trials have yet conclusively proven the efficacy of cell-based approaches. These clinical studies, however, have entailed the use of cells from various sources but of non-cardiac lineage origins. Although this might not be the main reason for their failures, the discovery of pluripotent stem cells capable of generating cardiomyocytes now raises the hope that such cardiac-committed cells could be therapeutically more effective. In this review, we will first describe where we currently are with regard to the clinical trials using PSC-differentiated cells and discuss the main issues which remain to be addressed. In parallel, because the capacity of cells to stably engraft in the recipient heart has increasingly been questioned, it has been hypothesized that a major mechanism of action could be the cell-triggered release of biomolecules that foster host-associated reparative pathways. Thus, in the second part of this review, we will discuss the rationale, clinically relevant advantages and pitfalls associated with the use of these PSC "products".
Collapse
Affiliation(s)
- Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Université Paris Cité, Inserm, PARCC, F-75015 Paris, France.
| |
Collapse
|
23
|
Jovisic M, Mambetsariev N, Singer BD, Morales-Nebreda L. Differential roles of regulatory T cells in acute respiratory infections. J Clin Invest 2023; 133:e170505. [PMID: 37463441 PMCID: PMC10348770 DOI: 10.1172/jci170505] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
Acute respiratory infections trigger an inflammatory immune response with the goal of pathogen clearance; however, overexuberant inflammation causes tissue damage and impairs pulmonary function. CD4+FOXP3+ regulatory T cells (Tregs) interact with cells of both the innate and the adaptive immune system to limit acute pulmonary inflammation and promote its resolution. Tregs also provide tissue protection and coordinate lung tissue repair, facilitating a return to homeostatic pulmonary function. Here, we review Treg-mediated modulation of the host response to respiratory pathogens, focusing on mechanisms underlying how Tregs promote resolution of inflammation and repair of acute lung injury. We also discuss potential strategies to harness and optimize Tregs as a cellular therapy for patients with severe acute respiratory infection and discuss open questions in the field.
Collapse
Affiliation(s)
- Milica Jovisic
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Simpson Querrey Lung Institute for Translational Science
| | | | - Benjamin D. Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Simpson Querrey Lung Institute for Translational Science
- Department of Biochemistry and Molecular Genetics, and
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Luisa Morales-Nebreda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Simpson Querrey Lung Institute for Translational Science
| |
Collapse
|
24
|
Abhishek K, Louis Sam Titus ASC, Dinh MTP, Mukhamedshin A, Mohan C, Gifford SC, Shevkoplyas SS. Red blood cell rosetting enables size-based separation of specific lymphocyte subsets from blood in a microfluidic device. LAB ON A CHIP 2023; 23:1804-1815. [PMID: 36723024 PMCID: PMC10050098 DOI: 10.1039/d2lc00817c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The isolation of a specific lymphocyte subset from blood is the required first step in the manufacturing of many novel cellular immunotherapies. Microfluidic size-based separation methods are poised to significantly simplify this process because they require neither centrifugation nor magnetic or fluorescent labeling to operate. Lymphocytes can be separated from red blood cells (RBCs) and platelets as well as monocytes and granulocytes because their size differs from each of these cell types. However, further separation of a specific lymphocyte subset from other unwanted lymphocytes using size-based methods is impossible because all lymphocytes have approximately the same size and can only be distinguished by surface markers. This paper describes a new approach that made it possible for a size-based separation method to isolate a desired subset of lymphocytes by making unwanted lymphocytes as well as other blood cells artificially larger. The separation was enabled by selectively binding multiple RBCs to each unwanted cell to create 'rosettes' with an effective size significantly larger than the diameter of a typical lymphocyte. The desired lymphocytes remained unaffected by rosetting and were separated from the rosettes by passing the mixture through a microfluidic size-based separation device based on controlled incremental filtration (CIF). This new rosette-enabled size-based (RESIZE) separation approach demonstrated recovery of 80-90% for all lymphocyte subsets tested (CD3+, CD4+, CD56+) which was ∼2.5-fold higher than that for the standard immunodensity method (RBC rosetting followed by density gradient centrifugation). The purity of separation was >90% for CD3+ cells but declined with increasing cell rarity. Unlike the immunodensity approach, RESIZE required neither centrifugation nor cell washing after the separation and was ∼2.5-fold faster when processing the same sample volume. The results of this study suggest that integration of the RESIZE approach for high-yield isolation of lymphocyte subsets from blood could significantly streamline the manufacturing workflow and thus have a potentially transformative impact on the cost and availability of novel cellular immunotherapies.
Collapse
Affiliation(s)
- Kumar Abhishek
- Department of Biomedical Engineering, University of Houston, 3605 Cullen Blvd, Houston, TX 77204-5060, USA.
| | | | - Mai T P Dinh
- Department of Biomedical Engineering, University of Houston, 3605 Cullen Blvd, Houston, TX 77204-5060, USA.
| | - Anton Mukhamedshin
- Department of Biomedical Engineering, University of Houston, 3605 Cullen Blvd, Houston, TX 77204-5060, USA.
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, 3605 Cullen Blvd, Houston, TX 77204-5060, USA.
| | - Sean C Gifford
- Halcyon Biomedical Incorporated, Friendswood, TX 77546, USA
| | - Sergey S Shevkoplyas
- Department of Biomedical Engineering, University of Houston, 3605 Cullen Blvd, Houston, TX 77204-5060, USA.
| |
Collapse
|
25
|
Jiang J, Huang H, Chen R, Lin Y, Ling Q. Immunotherapy for hepatocellular carcinoma recurrence after liver transplantation, can we harness the power of immune checkpoint inhibitors? Front Immunol 2023; 14:1092401. [PMID: 36875077 PMCID: PMC9978931 DOI: 10.3389/fimmu.2023.1092401] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death globally and liver transplantation (LT) can serve as the best curative treatment option. However, HCC recurrence after LT remains the major obstacle to the long-term survival of recipients. Recently, immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many cancers and provided a new treatment strategy for post-LT HCC recurrence. Evidence has been accumulated with the real-world application of ICIs in patients with post-LT HCC recurrence. Notably, the use of these agents as immunity boosters in recipients treated with immunosuppressors is still controversial. In this review, we summarized the immunotherapy for post-LT HCC recurrence and conducted an efficacy and safety evaluation based on the current experience of ICIs for post-LT HCC recurrence. In addition, we further discussed the potential mechanism of ICIs and immunosuppressive agents in regulating the balance between immune immunosuppression and lasting anti-tumor immunity.
Collapse
Affiliation(s)
- Jingyu Jiang
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Haitao Huang
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ruihan Chen
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yimou Lin
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Ling
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Pérez-Escobar J, Jimenez JV, Rodríguez-Aguilar EF, Servín-Rojas M, Ruiz-Manriquez J, Safar-Boueri L, Carrillo-Maravilla E, Navasa M, García-Juárez I. Immunotolerance in liver transplantation: a primer for the clinician. Ann Hepatol 2023; 28:100760. [PMID: 36179797 DOI: 10.1016/j.aohep.2022.100760] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/08/2022] [Indexed: 02/04/2023]
Abstract
The use of immunosuppressive medications for solid organ transplantation is associated with cardiovascular, metabolic, and oncologic complications. On the other hand, the development of graft rejection is associated with increased mortality and graft dysfunction. Liver transplant recipients can withdraw from immunosuppression without developing graft injury while preserving an adequate antimicrobial response - a characteristic known as immunotolerance. Immunotolerance can be spontaneously or pharmacologically achieved. Contrary to the classic dogma, clinical studies have elucidated low rates of true spontaneous immunotolerance (no serologic or histological markers of immune injury) among liver transplant recipients. However, clinical, serologic, and tissue biomarkers can aid in selecting patients in whom immunosuppression can be safely withdrawn. For those who failed an immunosuppression withdrawal trial or are at high risk of rejection, pharmacological interventions for immunotolerance induction are under development. In this review, we provide an overview of the mechanisms of immunotolerance, the clinical studies investigating predictors and biomarkers of spontaneous immunotolerance, as well as the potential pharmacological interventions for inducing it.
Collapse
Affiliation(s)
- Juanita Pérez-Escobar
- Department of Hepatology and Liver Transplant, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jose Victor Jimenez
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Erika Faride Rodríguez-Aguilar
- Department of Hepatology and Liver Transplant, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Maximiliano Servín-Rojas
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jesus Ruiz-Manriquez
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luisa Safar-Boueri
- Comprehensive Transplant Center, Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Eduardo Carrillo-Maravilla
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miquel Navasa
- Liver Transplant Unit, Hepatology Service, Hospital Clínic de Barcelona, IDIBAPS, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Ignacio García-Juárez
- Department of Hepatology and Liver Transplant, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
27
|
McGovern KE, Sonar SA, Watanabe M, Coplen CP, Bradshaw CM, Nikolich JŽ. The aging of the immune system and its implications for transplantation. GeroScience 2023:10.1007/s11357-022-00720-2. [PMID: 36626019 PMCID: PMC9838392 DOI: 10.1007/s11357-022-00720-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
By the last third of life, most mammals, including humans, exhibit a decline in immune cell numbers, immune organ structure, and immune defense of the organism, commonly known as immunosenescence. This decline leads to clinical manifestations of increased susceptibility to infections, particularly those caused by emerging and reemerging microorganisms, which can reach staggering levels-infection with SARS-CoV-2 has been 270-fold more lethal to older adults over 80 years of age, compared to their 18-39-year-old counterparts. However, while this would be expected to be beneficial to situations where hyporeactivity of the immune system may be desirable, this is not always the case. Here, we discuss the cellular and molecular underpinnings of immunosenescence as they pertain to outcomes of solid organ and hematopoietic transplantation.
Collapse
Affiliation(s)
- Kathryn E McGovern
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Sandip A Sonar
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Christopher P Coplen
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Christine M Bradshaw
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA.
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA.
- BIO5 Institute, University of Arizona, Tucson, AZ, USA.
- The Aegis Consortium for Pandemic-free Future, University of Arizona Health Sciences, University of Arizona, Tucson, 85719, USA.
| |
Collapse
|
28
|
Wing JB, Sakaguchi S. Regulatory Immune Cells. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
29
|
Abstract
CD4+ regulatory T (Treg) cells play an important role in maintaining immune homeostasis. Although these cells were initially studied as a homogenous cohort, we now know that they have unprecedented underlying heterogeneity. This heterogeneity is reflected in their phenotype and functions. As human Treg subpopulations are very small in numbers, it is necessary to develop novel ways of isolating and manipulating these cell populations. In this chapter, we discuss immunoassays established to this effect.
Collapse
Affiliation(s)
- Mo Atif
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Immunology Department Hôpital Pitié-Salpêtrière, Paris, France
| | - Mustapha Cherai
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Immunology Department Hôpital Pitié-Salpêtrière, Paris, France
| | - Makoto Miyara
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Immunology Department Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
30
|
Alrashidi HE, Alotiby AA. Zinc Modulates the Priming of T Helper 1, T Helper 17, and T Regulatory Cells in Allogeneic and Autologous in vitro Models. J Inflamm Res 2022; 15:6931-6939. [PMID: 36605132 PMCID: PMC9809170 DOI: 10.2147/jir.s391407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
Introduction Zinc is essential for the growth and differentiation of immune cells. Zinc insufficiency affects immune system function, thereby increasing infection susceptibility, autoimmunity, and allergies. Here, we aimed to determine the effects of zinc supplementation on T cell subpopulations, regulatory T (Tregs), T helper 1 (Th1), and T helper 17 (Th17) cells, in mixed lymphocyte cultures (MLC). Methods Allogeneic immune reactions were imitative using mixed lymphocyte cultures, followed by incubation with zinc to further monitor their effects. Cells were analyzed by flow cytometry. Production of Interferon-gamma (IFNγ), Interleukin-17 A (IL17A), and IL10 were analyzed by enzyme-linked immunosorbent assay. Th1 cell-specific Tbet, Th17 cell-specific RORC2, and Tregs-specific Foxp3 expression levels were determined by quantitative real-time PCR. Results Zinc supplementation at a physiological dose significantly increased CD4+ Foxp3+ Tregs and CD25+ Foxp3+ Tregs numbers and slightly decreased CD4+ RORC2+ and CD25+ RORC2+ Th17 cell numbers. A significant reduction in IFNγ production was observed in both restimulated T cells with autologous peripheral blood mononuclear cell (PBMC) and allogeneic PBMC compared to that in untreated T cells. Zinc significantly reduced IL17 expression, but the increase in IL10 expression was insignificant. In zinc-supplemented MLC, a non-significant decrease in Th1 or Th17 cell-specific transcription factors expression was observed, whereas there was a significant increase in Tregs-specific transcription factor expression. Conclusion Zinc can stabilize Tregs participating in adverse immune reactions or in an in vitro transplantation model.
Collapse
Affiliation(s)
- Hanan E Alrashidi
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Amna A Alotiby
- Haematology and Immunology Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia,Correspondence: Amna A Alotiby, Haematology and Immunology Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia, Email
| |
Collapse
|
31
|
Abdeladhim M, Karnell JL, Rieder SA. In or out of control: Modulating regulatory T cell homeostasis and function with immune checkpoint pathways. Front Immunol 2022; 13:1033705. [PMID: 36591244 PMCID: PMC9799097 DOI: 10.3389/fimmu.2022.1033705] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/16/2022] [Indexed: 12/16/2022] Open
Abstract
Regulatory T cells (Tregs) are the master regulators of immunity and they have been implicated in different disease states such as infection, autoimmunity and cancer. Since their discovery, many studies have focused on understanding Treg development, differentiation, and function. While there are many players in the generation and function of truly suppressive Tregs, the role of checkpoint pathways in these processes have been studied extensively. In this paper, we systematically review the role of different checkpoint pathways in Treg homeostasis and function. We describe how co-stimulatory and co-inhibitory pathways modulate Treg homeostasis and function and highlight data from mouse and human studies. Multiple checkpoint pathways are being targeted in cancer and autoimmunity; therefore, we share insights from the clinic and discuss the effect of experimental and approved therapeutics on Treg biology.
Collapse
|
32
|
Clarkson BDS, Johnson RK, Bingel C, Lothaller C, Howe CL. Preservation of antigen-specific responses in cryopreserved CD4 + and CD8 + T cells expanded with IL-2 and IL-7. J Transl Autoimmun 2022; 5:100173. [PMID: 36467614 PMCID: PMC9713293 DOI: 10.1016/j.jtauto.2022.100173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/31/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022] Open
Abstract
Objectives We sought to develop medium throughput standard operating procedures for screening cryopreserved human peripheral blood mononuclear cells (PBMCs) for CD4+ and CD8+ T cell responses to potential autoantigens. Methods Dendritic cells were loaded with a peptide cocktail from ubiquitous viruses or full-length viral protein antigens and cocultured with autologous T cells. We measured expression of surface activation markers on T cells by flow cytometry and cytometry by time of flight 24-72 h later. We tested responses among T cells freshly isolated from healthy control PBMCs, cryopreserved T cells, and T cells derived from a variety of T cell expansion protocols. We also compared the transcriptional profile of CD8+ T cells rested with interleukin (IL)7 for 48 h after 1) initial thawing, 2) expansion, and 3) secondary cryopreservation/thawing of expanded cells. To generate competent antigen presenting cells from PBMCs, we promoted differentiation of PBMCs into dendritic cells with granulocyte macrophage colony stimulating factor and IL-4. Results We observed robust dendritic cell differentiation from human PBMCs treated with 50 ng/mL GM-CSF and 20 ng/mL IL-4 in as little as 3 days. Dendritic cell purity was substantially increased by magnetically enriching for CD14+ monocytes prior to differentiation. We also measured antigen-dependent T cell activation in DC-T cell cocultures. However, polyclonal expansion of T cells with anti-CD3/antiCD28 abolished antigen-dependent upregulation of CD69 in our assay despite minimal transcriptional differences between rested CD8+ T cells before and after expansion. Furthermore, resting these expanded T cells in IL-2, IL-7 or IL-15 did not restore the antigen dependent responses. In contrast, T cells that were initially expanded with IL-2 + IL-7 rather than plate bound anti-CD3 + anti-CD28 retained responsiveness to antigen stimulation and these responses strongly correlated with responses measured at initial thawing. Significance While screening techniques for potential pathological autoantibodies have come a long way, comparable full-length protein target assays for screening patient T cells at medium throughput are noticeably lacking due to technical hurdles. Here we advance techniques that should have broad applicability to translational studies investigating cell mediated immunity in infectious or autoimmune diseases. Future studies are aimed at investigating possible CD8+ T cell autoantigens in MS and other CNS autoimmune diseases.
Collapse
Affiliation(s)
- Benjamin DS. Clarkson
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA,Corresponding author. Mayo Clinic, Guggenheim 1521C, 200 First Street SW, Rochester, MN, 55905.
| | | | - Corinna Bingel
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center, Heidelberg, Germany
| | | | - Charles L. Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Division of Experimental Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
33
|
Sanders JM, Jeyamogan S, Mathew JM, Leventhal JR. Foxp3+ regulatory T cell therapy for tolerance in autoimmunity and solid organ transplantation. Front Immunol 2022; 13:1055466. [PMID: 36466912 PMCID: PMC9714335 DOI: 10.3389/fimmu.2022.1055466] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/02/2022] [Indexed: 08/03/2023] Open
Abstract
Regulatory T cells (Tregs) are critical for tolerance in humans. The exact mechanisms by which the loss of peripheral tolerance leads to the development of autoimmunity and the specific role Tregs play in allograft tolerance are not fully understood; however, this population of T cells presents a unique opportunity in the development of targeted therapeutics. In this review, we discuss the potential roles of Foxp3+ Tregs in the development of tolerance in transplantation and autoimmunity, and the available data regarding their use as a treatment modality.
Collapse
Affiliation(s)
- Jes M. Sanders
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Shareni Jeyamogan
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - James M. Mathew
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Joseph R. Leventhal
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
34
|
Gupta PK, Allocco JB, Fraipont JM, McKeague ML, Wang P, Andrade MS, McIntosh C, Chen L, Wang Y, Li Y, Andrade J, Conejo-Garcia JR, Chong AS, Alegre ML. Reduced Satb1 expression predisposes CD4 + T conventional cells to Treg suppression and promotes transplant survival. Proc Natl Acad Sci U S A 2022; 119:e2205062119. [PMID: 36161903 PMCID: PMC9546564 DOI: 10.1073/pnas.2205062119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Limiting CD4+ T cell responses is important to prevent solid organ transplant rejection. In a mouse model of costimulation blockade-dependent cardiac allograft tolerance, we previously reported that alloreactive CD4+ conventional T cells (Tconvs) develop dysfunction, losing proliferative capacity. In parallel, induction of transplantation tolerance is dependent on the presence of regulatory T cells (Tregs). Whether susceptibility of CD4+ Tconvs to Treg suppression is modulated during tolerance induction is unknown. We found that alloreactive Tconvs from transplant tolerant mice had augmented sensitivity to Treg suppression when compared with memory T cells from rejector mice and expressed a transcriptional profile distinct from these memory T cells, including down-regulated expression of the transcription factor Special AT-rich sequence-binding protein 1 (Satb1). Mechanistically, Satb1 deficiency in CD4+ T cells limited their expression of CD25 and IL-2, and addition of Tregs, which express higher levels of CD25 than Satb1-deficient Tconvs and successfully competed for IL-2, resulted in greater suppression of Satb1-deficient than wild-type Tconvs in vitro. In vivo, Satb1-deficient Tconvs were more susceptible to Treg suppression, resulting in significantly prolonged skin allograft survival. Overall, our study reveals that transplantation tolerance is associated with Tconvs' susceptibility to Treg suppression, via modulated expression of Tconv-intrinsic Satb1. Targeting Satb1 in the context of Treg-sparing immunosuppressive therapies might be exploited to improve transplant outcomes.
Collapse
Affiliation(s)
- Pawan K. Gupta
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jennifer B. Allocco
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jane M. Fraipont
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Michelle L. McKeague
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Peter Wang
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Michael S. Andrade
- Section of Transplantation, Department of Surgery, University of Chicago, Chicago, IL 60637
| | - Christine McIntosh
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Luqiu Chen
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Ying Wang
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Yan Li
- Center for Research Informatics, University of Chicago, Chicago, IL 60637
| | - Jorge Andrade
- Center for Research Informatics, University of Chicago, Chicago, IL 60637
| | - José R. Conejo-Garcia
- Department of Immunology, Moffitt Cancer Center & Research Institute, University of South Florida, Tampa, FL 33612
| | - Anita S. Chong
- Section of Transplantation, Department of Surgery, University of Chicago, Chicago, IL 60637
| | - Maria-Luisa Alegre
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| |
Collapse
|
35
|
Iglesias M, Brennan DC, Larsen CP, Raimondi G. Targeting inflammation and immune activation to improve CTLA4-Ig-based modulation of transplant rejection. Front Immunol 2022; 13:926648. [PMID: 36119093 PMCID: PMC9478663 DOI: 10.3389/fimmu.2022.926648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
For the last few decades, Calcineurin inhibitors (CNI)-based therapy has been the pillar of immunosuppression for prevention of organ transplant rejection. However, despite exerting effective control of acute rejection in the first year post-transplant, prolonged CNI use is associated with significant side effects and is not well suited for long term allograft survival. The implementation of Costimulation Blockade (CoB) therapies, based on the interruption of T cell costimulatory signals as strategy to control allo-responses, has proven potential for better management of transplant recipients compared to CNI-based therapies. The use of the biologic cytotoxic T-lymphocyte associated protein 4 (CTLA4)-Ig is the most successful approach to date in this arena. Following evaluation of the BENEFIT trials, Belatacept, a high-affinity version of CTLA4-Ig, has been FDA approved for use in kidney transplant recipients. Despite its benefits, the use of CTLA4-Ig as a monotherapy has proved to be insufficient to induce long-term allograft acceptance in several settings. Multiple studies have demonstrated that events that induce an acute inflammatory response with the consequent release of proinflammatory cytokines, and an abundance of allograft-reactive memory cells in the recipient, can prevent the induction of or break established immunomodulation induced with CoB regimens. This review highlights advances in our understanding of the factors and mechanisms that limit CoB regimens efficacy. We also discuss recent successes in experimentally designing complementary therapies that favor CTLA4-Ig effect, affording a better control of transplant rejection and supporting their clinical applicability.
Collapse
Affiliation(s)
- Marcos Iglesias
- Vascularized and Composite Allotransplantation (VCA) Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Giorgio Raimondi, ; Marcos Iglesias,
| | - Daniel C. Brennan
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Christian P. Larsen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Giorgio Raimondi
- Vascularized and Composite Allotransplantation (VCA) Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Giorgio Raimondi, ; Marcos Iglesias,
| |
Collapse
|
36
|
Silva RCMC, Vasconcelos LR, Travassos LH. The different facets of heme-oxygenase 1 in innate and adaptive immunity. Cell Biochem Biophys 2022; 80:609-631. [PMID: 36018440 DOI: 10.1007/s12013-022-01087-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 07/20/2022] [Indexed: 11/26/2022]
Abstract
Heme oxygenase (HO) enzymes are responsible for the main oxidative step in heme degradation, generating equimolar amounts of free iron, biliverdin and carbon monoxide. HO-1 is induced as a crucial stress response protein, playing protective roles in physiologic and pathological conditions, due to its antioxidant, anti-apoptotic and anti-inflammatory effects. The mechanisms behind HO-1-mediated protection are being explored by different studies, affecting cell fate through multiple ways, such as reduction in intracellular levels of heme and ROS, transcriptional regulation, and through its byproducts generation. In this review we focus on the interplay between HO-1 and immune-related signaling pathways, which culminate in the activation of transcription factors important in immune responses and inflammation. We also discuss the dual interaction of HO-1 and inflammatory mediators that govern resolution and tissue damage. We highlight the dichotomy of HO-1 in innate and adaptive immune cells development and activation in different disease contexts. Finally, we address different known anti-inflammatory pharmaceuticals that are now being described to modulate HO-1, and the possible contribution of HO-1 in their anti-inflammatory effects.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Luiz Ricardo Vasconcelos
- Cellular Signaling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Leonardo Holanda Travassos
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
37
|
Vandermeulen M, Erpicum P, Bletard N, Poma L, Jouret F, Detry O. Effect of the Combination of Everolimus and Mesenchymal Stromal Cells on Regulatory T Cells Levels and in a Liver Transplant Rejection Model in Rats. Front Immunol 2022; 13:877953. [PMID: 35757737 PMCID: PMC9226583 DOI: 10.3389/fimmu.2022.877953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction Mesenchymal stromal cells (MSCs) have particular properties that are of interest in organ transplantation, including the expansion of regulatory T cells (Tregs), a key factor in transplant tolerance induction. However, the most effective immunosuppressive drug to associate with MSCs has yet to be defined. Additionally, the impact of the association of everolimus with MSCs on Treg expansion, and on the induction of liver graft tolerance, has never been studied. The aim of this study was to evaluate the effects of MSCs in combination, or not, with everolimus on Treg expansion and in a model of rejection after liver transplantation (LT) in the rat. Methods Firstly, 24 Lewis rats were assigned to 4 groups (n=6 in each group) receiving intravenous MSCs or saline injection at day (D)9 with/without subcutaneous everolimus from D0 to D14. Analysis of circulating Tregs was performed at D0, D14 and D28. In a second set of experiment, 30 Lewis rats were randomized in 3 groups 48hours after LT with a Dark Agouti rat liver: everolimus (subcutaneous for 14 days), MSCs (intravenous injection at post-operative day 2 and 9), or both everolimus and MSCs. Rejection of the liver graft was assessed by liver tests, histology and survival. Results Individually, MSC infusion and everolimus promoted Treg expansion in rats, and everolimus had no negative impact on Treg expansion in combination with MSCs. However, in the LT model, injections of MSCs two and nine days following LT were not effective at preventing acute rejection, and the combination of MSCs with everolimus failed to show any synergistic effect when compared to everolimus alone. Conclusion Everolimus may be used in association with MSCs. However, in our model of LT in the rat, post-transplant MSC injections did not prevent acute rejection, and the association of MSCs with everolimus did not show any synergistic effect.
Collapse
Affiliation(s)
- Morgan Vandermeulen
- Department of Abdominal Surgery and Transplantation, University of Liege Hospital [Centre Hospitalier Universitaire (CHU) ULiege], Liege, Belgium.,Centre de Recherche et de Développement du Département de Chirurgie (CREDEC), Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liege (ULiege), Liege, Belgium.,Laboratory of Translational Research in Nephrology (LTRN), Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liege (ULiege), Liege, Belgium
| | - Pauline Erpicum
- Laboratory of Translational Research in Nephrology (LTRN), Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liege (ULiege), Liege, Belgium.,Division of Nephrology, University of Liege Hospital [Centre Hospitalier Universitaire (CHU) ULiege], Liege, Belgium
| | - Noella Bletard
- Department of Pathology, University of Liege Hospital [Centre Hospitalier Universitaire (CHU) ULiege], Liege, Belgium
| | - Laurence Poma
- Laboratory of Translational Research in Nephrology (LTRN), Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liege (ULiege), Liege, Belgium
| | - François Jouret
- Laboratory of Translational Research in Nephrology (LTRN), Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liege (ULiege), Liege, Belgium.,Division of Nephrology, University of Liege Hospital [Centre Hospitalier Universitaire (CHU) ULiege], Liege, Belgium
| | - Olivier Detry
- Department of Abdominal Surgery and Transplantation, University of Liege Hospital [Centre Hospitalier Universitaire (CHU) ULiege], Liege, Belgium.,Centre de Recherche et de Développement du Département de Chirurgie (CREDEC), Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liege (ULiege), Liege, Belgium
| |
Collapse
|
38
|
Bernaldo-de-Quirós E, Cózar B, López-Esteban R, Clemente M, Gil-Jaurena JM, Pardo C, Pita A, Pérez-Caballero R, Camino M, Gil N, Fernández-Santos ME, Suarez S, Pion M, Martínez-Bonet M, Correa-Rocha R. A Novel GMP Protocol to Produce High-Quality Treg Cells From the Pediatric Thymic Tissue to Be Employed as Cellular Therapy. Front Immunol 2022; 13:893576. [PMID: 35651624 PMCID: PMC9148974 DOI: 10.3389/fimmu.2022.893576] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022] Open
Abstract
Due to their suppressive capacity, the adoptive transfer of regulatory T cells (Treg) has acquired a growing interest in controlling exacerbated inflammatory responses. Limited Treg recovery and reduced quality remain the main obstacles in most current protocols where differentiated Treg are obtained from adult peripheral blood. An alternate Treg source is umbilical cord blood, a promising source of Treg cells due to the higher frequency of naïve Treg and lower frequency of memory T cells present in the fetus’ blood. However, the Treg number isolated from cord blood remains limiting. Human thymuses routinely discarded during pediatric cardiac surgeries to access the retrosternal operative field has been recently proposed as a novel source of Treg for cellular therapy. This strategy overcomes the main limitations of current Treg sources, allowing the obtention of very high numbers of undifferentiated Treg. We have developed a novel good manufacturing practice (GMP) protocol to obtain large Treg amounts, with very high purity and suppressive capacity, from the pediatric thymus (named hereafter thyTreg). The total amount of thyTreg obtained at the end of the procedure, after a short-term culture of 7 days, reach an average of 1,757 x106 (range 50 x 106 – 13,649 x 106) cells from a single thymus. The thyTreg product obtained with our protocol shows very high viability (mean 93.25%; range 83.35% – 97.97%), very high purity (mean 92.89%; range 70.10% – 98.41% of CD25+FOXP3+ cells), stability under proinflammatory conditions and a very high suppressive capacity (inhibiting in more than 75% the proliferation of activated CD4+ and CD8+ T cells in vitro at a thyTreg:responder cells ratio of 1:1). Our thyTreg product has been approved by the Spanish Drug Agency (AEMPS) to be administered as cell therapy. We are recruiting patients in the first-in-human phase I/II clinical trial worldwide that evaluates the safety, feasibility, and efficacy of autologous thyTreg administration in children undergoing heart transplantation (NCT04924491). The high quality and amount of thyTreg and the differential features of the final product obtained with our protocol allow preparing hundreds of doses from a single thymus with improved therapeutic properties, which can be cryopreserved and could open the possibility of an “off-the-shelf” allogeneic use in another individual.
Collapse
Affiliation(s)
| | - Beatriz Cózar
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Rocío López-Esteban
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Maribel Clemente
- Cell Culture Unit, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | | | - Carlos Pardo
- Pediatric Cardiac Surgery Unit, Hospital Materno Infantil Gregorio Marañón, Madrid, Spain
| | - Ana Pita
- Pediatric Cardiac Surgery Unit, Hospital Materno Infantil Gregorio Marañón, Madrid, Spain
| | - Ramón Pérez-Caballero
- Pediatric Cardiac Surgery Unit, Hospital Materno Infantil Gregorio Marañón, Madrid, Spain
| | - Manuela Camino
- Pediatric Heart Transplant Unit, Hospital Materno Infantil Gregorio Marañón, Madrid, Spain
| | - Nuria Gil
- Pediatric Heart Transplant Unit, Hospital Materno Infantil Gregorio Marañón, Madrid, Spain
| | | | - Susana Suarez
- Cell Production Unit, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Marjorie Pion
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Marta Martínez-Bonet
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- *Correspondence: Rafael Correa-Rocha, ; Marta Martínez-Bonet,
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- *Correspondence: Rafael Correa-Rocha, ; Marta Martínez-Bonet,
| |
Collapse
|
39
|
Lapp MM, Lin G, Komin A, Andrews L, Knudson M, Mossman L, Raimondi G, Arciero JC. Modeling the Potential of Treg-Based Therapies for Transplant Rejection: Effect of Dose, Timing, and Accumulation Site. Transpl Int 2022; 35:10297. [PMID: 35479106 PMCID: PMC9035492 DOI: 10.3389/ti.2022.10297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/17/2022] [Indexed: 02/04/2023]
Abstract
Introduction: The adoptive transfer of regulatory T cells (Tregs) has emerged as a method to promote graft tolerance. Clinical trials have demonstrated the safety of adoptive transfer and are now assessing their therapeutic efficacy. Strategies that generate large numbers of antigen specific Tregs are even more efficacious. However, the combinations of factors that influence the outcome of adoptive transfer are too numerous to be tested experimentally. Here, mathematical modeling is used to predict the most impactful treatment scenarios. Methods: We adapted our mathematical model of murine heart transplant rejection to simulate Treg adoptive transfer and to correlate therapeutic efficacy with Treg dose and timing, frequency of administration, and distribution of injected cells. Results: The model predicts that Tregs directly accumulating to the graft are more protective than Tregs localizing to draining lymph nodes. Inhibiting antigen-presenting cell maturation and effector functions at the graft site was more effective at modulating rejection than inhibition of T cell activation in lymphoid tissues. These complex dynamics define non-intuitive relationships between graft survival and timing and frequency of adoptive transfer. Conclusion: This work provides the framework for better understanding the impact of Treg adoptive transfer and will guide experimental design to improve interventions.
Collapse
Affiliation(s)
- Maya M. Lapp
- Department of Mathematics, The College of Wooster, Wooster, OH, United States
| | - Guang Lin
- Department of Mathematics, Purdue University, West Lafayette, IN, United States
| | - Alexander Komin
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Leah Andrews
- Department of Mathematics, St. Olaf College, Northfield, MN, United States
| | - Mei Knudson
- Department of Mathematics, Carleton College, Northfield, MN, United States
| | - Lauren Mossman
- Department of Mathematics, St. Olaf College, Northfield, MN, United States
| | - Giorgio Raimondi
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, United States,*Correspondence: Giorgio Raimondi, ; Julia C. Arciero,
| | - Julia C. Arciero
- Department of Mathematical Sciences, Indiana University-Purdue University of Indianapolis, Indianapolis, IN, United States,*Correspondence: Giorgio Raimondi, ; Julia C. Arciero,
| |
Collapse
|
40
|
Xiong X, Luo Z, Zhou H, Duan Z, Niu L, Zhang K, Huang G, Li W. Downregulation of TIGIT Expression in FOXP3+Regulatory T Cells in Acute Coronary Syndrome. J Inflamm Res 2022; 15:1195-1207. [PMID: 35228811 PMCID: PMC8882028 DOI: 10.2147/jir.s351364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
Objective Little is currently known on the role of T-cell immunoglobulin and ITIM domain (TIGIT) expression in Foxp3+ regulatory T cells (TIGIT+Tregs) in acute coronary syndrome (ACS) patients. The aim of this study was to investigate the role and alterations of TIGIT+Tregs in ACS patients. Methods We enrolled 117 subjects, including 61 ACS patients, 26 chronic coronary syndrome (CCS) patients, and 30 control subjects without coronary artery disease. The quantification of TIGIT+Tregs was determined by flow cytometry; serum interleukin-6 (IL-6) and transforming growth factor-β (TGF-β) were also measured. Results TIGIT+Tregs expression was significantly lower in ACS patients compared with CCS and control patients (P<0.05). The expression of TIGIT+Tregs was comparable in patients with and without traditional risk factors (P>0.05). Logistic regression analysis revealed that TIGIT+Tregs levels are independent predictors of ACS (P<0.01). Receiver-operating characteristic (ROC) curve analysis showed the expression levels of TIGIT+Tregs had a discriminative power for ACS (P<0.01). IL-6 levels were increased (P<0.01), while TGF-β was decreased in ACS patients compared with CCS and control patients (P<0.01). Meanwhile, an inverse correlation between IL-6 and TIGIT+Tregs was observed (P<0.01), while a positive correlation between TGF-β and TIGIT+Tregs was found (P<0.05). Conclusion TIGIT+Tregs levels are significantly reduced in ACS, accompanied by upregulated IL-6 and downregulated TGF-β expression. The downregulated TIGIT+Tregs are independent predictors of ACS. These findings suggest that TIGIT+Tregs may have an anti-inflammatory and protective effect on ACS, and its decreased expression may be associated with atherosclerotic plaque destabilization.
Collapse
Affiliation(s)
- Xinlin Xiong
- Clinical College, Guizhou Medical University, Guiyang City, Guizhou Province, People’s Republic of China
- Department of Cardiology, Chengdu University Affiliated Hospital, Chengdu City, Sichuan Province, People’s Republic of China
- Department of Cardiology, Guizhou Medical University Affiliated Hospital, Guiyang City, Guizhou Province, People’s Republic of China
| | - Zhenhua Luo
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People’s Hospital, Guiyang City, Guizhou Province, People’s Republic of China
- Department of Central Lab, Guizhou Provincial People’s Hospital, Guiyang City, Guizhou Province, People’s Republic of China
| | - Haiyan Zhou
- Department of Cardiology, Guizhou Medical University Affiliated Hospital, Guiyang City, Guizhou Province, People’s Republic of China
| | - Zonggang Duan
- Clinical College, Guizhou Medical University, Guiyang City, Guizhou Province, People’s Republic of China
| | - Li Niu
- Department of Cardiology, Guizhou Medical University Affiliated Hospital, Guiyang City, Guizhou Province, People’s Republic of China
| | - Kai Zhang
- Clinical College, Guizhou Medical University, Guiyang City, Guizhou Province, People’s Republic of China
| | - Guangwei Huang
- Clinical College, Guizhou Medical University, Guiyang City, Guizhou Province, People’s Republic of China
| | - Wei Li
- Clinical College, Guizhou Medical University, Guiyang City, Guizhou Province, People’s Republic of China
- Department of Cardiology, Guizhou Medical University Affiliated Hospital, Guiyang City, Guizhou Province, People’s Republic of China
- Correspondence: Wei Li, Email
| |
Collapse
|
41
|
Bernaldo-de-Quirós E, Pion M, Martínez-Bonet M, Correa-Rocha R. A New Generation of Cell Therapies Employing Regulatory T Cells (Treg) to Induce Immune Tolerance in Pediatric Transplantation. Front Pediatr 2022; 10:862807. [PMID: 35633970 PMCID: PMC9130702 DOI: 10.3389/fped.2022.862807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Kidney transplantation is the most common solid organ transplant and the preferred treatment for pediatric patients with end-stage renal disease, but it is still not a definitive solution due to immune graft rejection. Regulatory T cells (Treg) and their control over effector T cells is a crucial and intrinsic tolerance mechanism in limiting excessive immune responses. In the case of transplants, Treg are important for the survival of the transplanted organ, and their dysregulation could increase the risk of rejection in transplanted children. Chronic immunosuppression to prevent rejection, for which Treg are especially sensitive, have a detrimental effect on Treg counts, decreasing the Treg/T-effector balance. Cell therapy with Treg cells is a promising approach to restore this imbalance, promoting tolerance and thus increasing graft survival. However, the strategies used to date that employ peripheral blood as a Treg source have shown limited efficacy. Moreover, it is not possible to use this approach in pediatric patients due to the limited volume of blood that can be extracted from children. Here, we outline our innovative strategy that employs the thymus removed during pediatric cardiac surgeries as a source of therapeutic Treg that could make this therapy accessible to transplanted children. The advantageous properties and the massive amount of Treg cells obtained from pediatric thymic tissue (thyTreg) opens a new possibility for Treg therapies to prevent rejection in pediatric kidney transplants. We are recruiting patients in a clinical trial to prevent rejection in heart-transplanted children through the infusion of autologous thyTreg cells (NCT04924491). If its efficacy is confirmed, thyTreg therapy may establish a new paradigm in preventing organ rejection in pediatric transplants, and their allogeneic use would extend its application to other solid organ transplantation.
Collapse
Affiliation(s)
- Esther Bernaldo-de-Quirós
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marjorie Pion
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marta Martínez-Bonet
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
42
|
Silva RCMC, Panis C, Pires BRB. Lessons from transmissible cancers for immunotherapy and transplant. Immunol Med 2021; 45:146-161. [PMID: 34962854 DOI: 10.1080/25785826.2021.2018783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The emergence of horizontal transmission of cancer between vertebrates is an issue that interests scientists and medical society. Transmission requires: (i) a mechanism by which cancer cells can transfer to another organism and (ii) a repressed immune response on the part of the recipient. Transmissible tumors are unique models to comprehend the responses and mechanisms mediated by the major histocompatibility complex (MHC), which can be transposed for transplant biology. Here, we discuss the mechanisms involved in immune-mediated tissue rejection, making a parallel with transmissible cancers. We also discuss cellular and molecular mechanisms involved in cancer immunotherapy and anti-rejection therapies.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio De Janeiro, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of West Paraná, UNIOESTE, Francisco Beltrão, Brazil
| | | |
Collapse
|
43
|
Hann A, Oo YH, Perera MTPR. Regulatory T-Cell Therapy in Liver Transplantation and Chronic Liver Disease. Front Immunol 2021; 12:719954. [PMID: 34721383 PMCID: PMC8552037 DOI: 10.3389/fimmu.2021.719954] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/24/2021] [Indexed: 12/29/2022] Open
Abstract
The constant exposure of the liver to gut derived foreign antigens has resulted in this organ attaining unique immunological characteristics, however it remains susceptible to immune mediated injury. Our understanding of this type of injury, in both the native and transplanted liver, has improved significantly in recent decades. This includes a greater awareness of the tolerance inducing CD4+ CD25+ CD127low T-cell lineage with the transcription factor FoxP3, known as regulatory T-Cells (Tregs). These cells comprise 5-10% of CD4+ T cells and are known to function as an immunological "braking" mechanism, thereby preventing immune mediated tissue damage. Therapies that aim to increase Treg frequency and function have proved beneficial in the setting of both autoimmune diseases and solid organ transplantations. The safety and efficacy of Treg therapy in liver disease is an area of intense research at present and has huge potential. Due to these cells possessing significant plasticity, and the potential for conversion towards a T-helper 1 (Th1) and 17 (Th17) subsets in the hepatic microenvironment, it is pre-requisite to modify the microenvironment to a Treg favourable atmosphere to maintain these cells' function. In addition, implementation of therapies that effectively increase Treg functional activity in the liver may result in the suppression of immune responses and will hinder those that destroy tumour cells. Thus, fine adjustment is crucial to achieve this immunological balance. This review will describe the hepatic microenvironment with relevance to Treg function, and the role these cells have in both native diseased and transplanted livers.
Collapse
Affiliation(s)
- Angus Hann
- The Liver Unit, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ye H Oo
- The Liver Unit, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Centre for Rare Disease (ERN-Rare Liver Centre), University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - M Thamara P R Perera
- The Liver Unit, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
44
|
Saini KS, Svane IM, Juan M, Barlesi F, André F. Manufacture of adoptive cell therapies at academic cancer centers: scientific, safety and regulatory challenges. Ann Oncol 2021; 33:6-12. [PMID: 34655734 DOI: 10.1016/j.annonc.2021.09.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- K S Saini
- Labcorp Drug Development Inc., Princeton, USA
| | - I M Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - M Juan
- Department of Immunology, Hospital Clinic, IDIBAPS, Immunotherapy Platform Hospital Sant Joan de Déu, Universidad de Barcelona, Barcelona, Spain
| | - F Barlesi
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France; Aix Marseille University, CNRS, INSERM, CRCM, Marseille, France
| | - F André
- Institut Gustave Roussy, INSERM UMR981, Université Paris Saclay, Paris, France.
| |
Collapse
|
45
|
Pham MN, Khoryati L, Jamison BL, Hayes E, Sullivan JM, Campbell DJ, Gavin MA. In Vivo Expansion of Antigen-Specific Regulatory T Cells through Staggered Fc.IL-2 Mutein Dosing and Antigen-Specific Immunotherapy. Immunohorizons 2021; 5:782-791. [PMID: 34583939 PMCID: PMC11034776 DOI: 10.4049/immunohorizons.2100051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/30/2021] [Indexed: 11/19/2022] Open
Abstract
In mice, Ag administration in the absence of adjuvant typically elicits tolerogenic immune responses through the deletion or inactivation of conventional CD4 T cells and the formation or expansion of regulatory CD4 T cells (Treg). Although these "Ag-specific immunotherapy" (ASI) approaches are currently under clinical development to treat autoinflammatory conditions, efficacy and safety may be variable and unpredictable because of the diverse activation states of immune cells in subjects with autoimmune and allergic diseases. To reliably induce Ag-specific tolerance in patients, novel methods to control T cell responses during ASI are needed, and strategies that permanently increase Treg frequencies among Ag-specific CD4 T cells may provide long-lasting immunosuppression between treatments. In this study, we present an approach to durably increase the frequency of Ag-specific Treg in mice by administering ASI when Treg numbers are transiently increased with individual doses of a half-life-extended Treg-selective IL-2 mutein. Repeated weekly cycles of IL-2 mutein doses (day 0) followed by ASI (day 3) resulted in a 3- to 5-fold enrichment in Treg among Ag-responsive CD4 T cells. Expanded Ag-specific Treg persisted for more than 3 wk following treatment cessation, as well as through an inflammatory T cell response to an Ag-expressing virus. Combining Treg enrichment with ASI has the potential to durably treat autoimmune disease or allergy by increasing the Treg/conventional CD4 T cell ratio among autoantigen- or allergen-specific T cells.
Collapse
Affiliation(s)
- Minh N Pham
- Benaroya Research Institute, Seattle, WA; and
| | | | | | - Erika Hayes
- Benaroya Research Institute, Seattle, WA; and
| | | | | | - Marc A Gavin
- Benaroya Research Institute, Seattle, WA; and
- Omeros Corp., Seattle, WA
| |
Collapse
|
46
|
Pilat N, Lefsihane K, Brouard S, Kotsch K, Falk C, Steiner R, Thaunat O, Fusil F, Montserrat N, Amarelli C, Casiraghi F. T- and B-cell therapy in solid organ transplantation: current evidence and future expectations. Transpl Int 2021; 34:1594-1606. [PMID: 34448274 DOI: 10.1111/tri.13972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/08/2021] [Indexed: 01/13/2023]
Abstract
Cell therapy has emerged as an attractive therapeutic option in organ transplantation. During the last decade, the therapeutic potency of Treg immunotherapy has been shown in various preclinical animal models and safety was demonstrated in first clinical trials. However, there are still critical open questions regarding specificity, survival, and migration to the target tissue so the best Treg population for infusion into patients is still under debate. Recent advances in CAR technology hold the promise for Treg-functional superiority. Another exciting strategy is the generation of B-cell antibody receptor (BAR) Treg/cytotoxic T cells to specifically regulate or deplete alloreactive memory B cells. Finally, B cells are also capable of immune regulation, making them promising candidates for immunomodulatory therapeutic strategies. This article summarizes available literature on cell-based innovative therapeutic approaches aiming at modulating alloimmune response for transplantation. Crucial areas of investigation that need a joined effort of the transplant community for moving the field toward successful achievement of tolerance are highlighted.
Collapse
Affiliation(s)
- Nina Pilat
- Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Katia Lefsihane
- International Center of Infectiology Research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Sophie Brouard
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | - Katja Kotsch
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Department for General and Visceral Surgery, Berlin Institute of Health, Berlin, Germany
| | - Christine Falk
- Institute of Transplant Immunology, Hannover Medical School, MHH, Hannover, Germany
| | - Romy Steiner
- Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Olivier Thaunat
- International Center of Infectiology Research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Floriane Fusil
- International Center of Infectiology Research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Cristiano Amarelli
- Department of Cardiac Surgery and Transplants Monaldi, A.O. dei Colli, Naples, Italy
| | | |
Collapse
|
47
|
Robinson S, Thomas R. Potential for Antigen-Specific Tolerizing Immunotherapy in Systematic Lupus Erythematosus. Front Immunol 2021; 12:654701. [PMID: 34335564 PMCID: PMC8322693 DOI: 10.3389/fimmu.2021.654701] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic complex systemic autoimmune disease characterized by multiple autoantibodies and clinical manifestations, with the potential to affect nearly every organ. SLE treatments, including corticosteroids and immunosuppressive drugs, have greatly increased survival rates, but there is no curative therapy and SLE management is limited by drug complications and toxicities. There is an obvious clinical need for safe, effective SLE treatments. A promising treatment avenue is to restore immunological tolerance to reduce inflammatory clinical manifestations of SLE. Indeed, recent clinical trials of low-dose IL-2 supplementation in SLE patients showed that in vivo expansion of regulatory T cells (Treg cells) is associated with dramatic but transient improvement in SLE disease markers and clinical manifestations. However, the Treg cells that expanded were short-lived and unstable. Alternatively, antigen-specific tolerance (ASIT) approaches that establish long-lived immunological tolerance could be deployed in the context of SLE. In this review, we discuss the potential benefits and challenges of nanoparticle ASIT approaches to induce prolonged immunological tolerance in SLE.
Collapse
Affiliation(s)
- Sean Robinson
- School of Medicine, Faculty of Medicine and Biomedical Sciences, University of Queensland, St Lucia, QLD, Australia
| | - Ranjeny Thomas
- University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| |
Collapse
|
48
|
The Presence of a Marked Imbalance Between Regulatory T Cells and Effector T Cells Reveals That Tolerance Mechanisms Could Be Compromised in Heart Transplant Children. Transplant Direct 2021; 7:e693. [PMID: 33928185 PMCID: PMC8078462 DOI: 10.1097/txd.0000000000001152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 12/19/2022] Open
Abstract
Regulatory T cells (Treg) are crucial for the induction and maintenance of graft tolerance. In pediatric heart transplant procedures, the thymus is routinely excised, removing the primary source of T-cell replenishment. Consequently, thymectomy joined to the effects of immunosuppression on the T-cell compartment may have a detrimental impact on Treg values, compromising the intrinsic tolerance mechanisms and the protective role of Treg preventing graft rejection in heart transplant children.
Collapse
|
49
|
Aghabi YO, Yasin A, Kennedy JI, Davies SP, Butler AE, Stamataki Z. Targeting Enclysis in Liver Autoimmunity, Transplantation, Viral Infection and Cancer. Front Immunol 2021; 12:662134. [PMID: 33953725 PMCID: PMC8089374 DOI: 10.3389/fimmu.2021.662134] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Persistent liver inflammation can lead to cirrhosis, which associates with significant morbidity and mortality worldwide. There are no curative treatments beyond transplantation, followed by long-term immunosuppression. The global burden of end stage liver disease has been increasing and there is a shortage of donor organs, therefore new therapies are desperately needed. Harnessing the power of the immune system has shown promise in certain autoimmunity and cancer settings. In the context of the liver, regulatory T cell (Treg) therapies are in development. The hypothesis is that these specialized lymphocytes that dampen inflammation may reduce liver injury in patients with chronic, progressive diseases, and promote transplant tolerance. Various strategies including intrinsic and extracorporeal expansion of Treg cells, aim to increase their abundance to suppress immune responses. We recently discovered that hepatocytes engulf and delete Treg cells by enclysis. Herein, we propose that inhibition of enclysis may potentiate existing regulatory T cell therapeutic approaches in patients with autoimmune liver diseases and in patients receiving a transplant. Moreover, in settings where the abundance of Treg cells could hinder beneficial immunity, such us in chronic viral infection or liver cancer, enhancement of enclysis could result in transient, localized reduction of Treg cell numbers and tip the balance towards antiviral and anti-tumor immunity. We describe enclysis as is a natural process of liver immune regulation that lends itself to therapeutic targeting, particularly in combination with current Treg cell approaches.
Collapse
Affiliation(s)
| | | | | | | | | | - Zania Stamataki
- College of Medical and Dental Sciences, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
50
|
Marfil-Garza BA, Hefler J, Bermudez De Leon M, Pawlick R, Dadheech N, Shapiro AMJ. Progress in Translational Regulatory T Cell Therapies for Type 1 Diabetes and Islet Transplantation. Endocr Rev 2021; 42:198-218. [PMID: 33247733 DOI: 10.1210/endrev/bnaa028] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Regulatory T cells (Tregs) have become highly relevant in the pathophysiology and treatment of autoimmune diseases, such as type 1 diabetes (T1D). As these cells are known to be defective in T1D, recent efforts have explored ex vivo and in vivo Treg expansion and enhancement as a means for restoring self-tolerance in this disease. Given their capacity to also modulate alloimmune responses, studies using Treg-based therapies have recently been undertaken in transplantation. Islet transplantation provides a unique opportunity to study the critical immunological crossroads between auto- and alloimmunity. This procedure has advanced greatly in recent years, and reports of complete abrogation of severe hypoglycemia and long-term insulin independence have become increasingly reported. It is clear that cellular transplantation has the potential to be a true cure in T1D, provided the remaining barriers of cell supply and abrogated need for immune suppression can be overcome. However, the role that Tregs play in islet transplantation remains to be defined. Herein, we synthesize the progress and current state of Treg-based therapies in T1D and islet transplantation. We provide an extensive, but concise, background to understand the physiology and function of these cells and discuss the clinical evidence supporting potency and potential Treg-based therapies in the context of T1D and islet transplantation. Finally, we discuss some areas of opportunity and potential research avenues to guide effective future clinical application. This review provides a basic framework of knowledge for clinicians and researchers involved in the care of patients with T1D and islet transplantation.
Collapse
Affiliation(s)
| | - Joshua Hefler
- Department of Surgery, University of Alberta, Edmonton, Canada
| | - Mario Bermudez De Leon
- Department of Molecular Biology, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey, Nuevo Leon, Mexico
| | - Rena Pawlick
- Department of Surgery, University of Alberta, Edmonton, Canada
| | | | - A M James Shapiro
- Department of Surgery, University of Alberta, Edmonton, Canada.,Clinical Islet Transplant Program, University of Alberta, Edmonton, Canada
| |
Collapse
|