1
|
Mureanu N, Bowman AM, Porter-Wright IA, Verma P, Efthymiou A, Nicolaides KH, Scotta C, Lombardi G, Tribe RM, Shangaris P. The Immunomodulatory Role of Regulatory T Cells in Preterm Birth and Associated Pregnancy Outcomes. Int J Mol Sci 2024; 25:11878. [PMID: 39595948 PMCID: PMC11593591 DOI: 10.3390/ijms252211878] [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: 10/03/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Spontaneous preterm birth (sPTB), defined as live birth before 37 weeks of gestational age, is associated with immune dysregulation and pro-inflammatory conditions that profoundly impact newborn health. The question of immune integrity at the maternal-foetal interface is a focus of recent studies centring not only sPTB but the conditions often affiliated with this outcome. Regulatory T cells (Tregs) play a critical anti-inflammatory role in pregnancy, promoting foetal tolerance and placentation. Due to this gestational role, it is hypothesised that decreased or dysfunctional Tregs may be implicated in cases of sPTB. This review examines studies comparing Treg presence in healthy term pregnancies and those with sPTB-associated conditions. Conflicting findings across different conditions and within sPTB itself have been identified. However, notable findings from the research indicate increased proinflammatory cytokines in pregnancies suffering from premature rupture of membranes (pPROM), chorioamnionitis, infection, preeclampsia, and gestational diabetes (GDM). Additionally, reduced Treg levels were identified in preeclampsia, GDM, and pPROM as well as chorioamnionitis presenting with increased Treg dysfunctionality. Treg deficiencies may contribute to health issues in preterm newborns. Current sPTB treatments are limited, underscoring the potential of in utero therapies targeting inflammation, including T cell interventions. Future research aims to establish consensus on the role of Tregs in sPTB and associated conditions and advancing understanding of mechanisms leading to Treg deficiencies in adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Nicoleta Mureanu
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
- Faculty of Medicine, Department of Obstetrics and Gynaecology, Carol Davila University of Medicine and Pharmacy, Bulevardul Eroii Sanitari 8, 050474 Bucharest, Romania
| | - Amanda M. Bowman
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Imogen A. Porter-Wright
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Priya Verma
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Athina Efthymiou
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
| | - Kypros H. Nicolaides
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
| | - Cristiano Scotta
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine, Brunel University London, London UB8 3PH, UK
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Rachel M. Tribe
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
| | - Panicos Shangaris
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| |
Collapse
|
2
|
Gardell JL, Maurer ME, Childs MM, Pham MN, Meengs B, Julien SH, Tan C, Boster DR, Quach P, Therriault JH, Hermansky G, Patton DT, Bowser J, Chen A, Morgan NN, Gilbertson EA, Bogatzki L, Encarnacion K, McMahan CJ, Crane CA, Swiderek KM. Preclinical characterization of MTX-101: a novel bispecific CD8 Treg modulator that restores CD8 Treg functions to suppress pathogenic T cells in autoimmune diseases. Front Immunol 2024; 15:1452537. [PMID: 39559361 PMCID: PMC11570885 DOI: 10.3389/fimmu.2024.1452537] [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: 06/21/2024] [Accepted: 09/23/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction Regulatory CD8 T cells (CD8 Treg) are responsible for the selective killing of self-reactive and pathogenic CD4 T cells. In autoimmune disease, CD8 Treg may accumulate in the peripheral blood but fail to control the expansion of pathogenic CD4 T cells that subsequently cause tissue destruction. This CD8 Treg dysfunction is due in part to the expression of inhibitory killer immunoglobulin-like receptors (KIR; KIR2DL isoforms [KIR2DL1, KIR2DL2, and KIR2DL3]); these molecules serve as autoimmune checkpoints and limit CD8 Treg activation. Methods Here we describe the pre-clinical characterization of MTX-101, a bispecific antibody targeting inhibitory KIR and CD8. Using human peripheral blood mononuculear cells (PBMC) derived from healthy donors and autoimmune patients, humanized mouse models, and human derived tissue organoids, we evaluated the molecular mechanisms and functional effects of MTX-101. Results By binding to KIR, MTX-101 inhibited KIR signaling that can restore CD8 Treg ability to eliminate pathogenic CD4 T cells. MTX-101 bound and activated CD8 Treg in human peripheral blood mononuclear cells (PBMC), resulting in increased CD8 Treg cytolytic capacity, activation, and prevalence. Enhancing CD8 Treg function with MTX-101 reduced pathogenic CD4 T cell expansion and inflammation, without increasing pro-inflammatory cytokines or activating immune cells that express either target alone. MTX-101 reduced antigen induced epithelial cell death in disease affected tissues, including in tissue biopsies from individuals with autoimmune disease (i.e., celiac disease, Crohn's disease). The effects of MTX-101 were specific to autoreactive CD4 T cells and did not suppress responses to viral and bacterial antigens. In a human PBMC engrafted Graft versus Host Disease (GvHD) mouse model of acute inflammation, MTX-101 bound CD8 Treg and delayed onset of disease. MTX-101 induced dose dependent binding, increased prevalence and cytolytic capacity of CD8 Treg, as well as increased CD4 T cell death. MTX-101 selectively bound CD8 Treg without unwanted immune cell activation or increase of pro-inflammatory serum cytokines and exhibited an antibody-like half-life in pharmacokinetic and exploratory tolerability studies performed using IL-15 transgenic humanized mice with engrafted human lymphocytes, including CD8 Treg at physiologic ratios. Conclusion Collectively, these data support the development of MTX-101 for the treatment of autoimmune diseases.
Collapse
|
3
|
Rui X, Alvarez Calderon F, Wobma H, Gerdemann U, Albanese A, Cagnin L, McGuckin C, Michaelis KA, Naqvi K, Blazar BR, Tkachev V, Kean LS. Human OX40L-CAR-T regs target activated antigen-presenting cells and control T cell alloreactivity. Sci Transl Med 2024; 16:eadj9331. [PMID: 39413160 DOI: 10.1126/scitranslmed.adj9331] [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: 07/26/2023] [Revised: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/18/2024]
Abstract
Regulatory T cells (Tregs) make major contributions to immune homeostasis. Because Treg dysfunction can lead to both allo- and autoimmunity, there is interest in correcting these disorders through Treg adoptive transfer. Two of the central challenges in clinically deploying Treg cellular therapies are ensuring phenotypic stability and maximizing potency. Here, we describe an approach to address both issues through the creation of OX40 ligand (OX40L)-specific chimeric antigen receptor (CAR)-Tregs under the control of a synthetic forkhead box P3 (FOXP3) promoter. The creation of these CAR-Tregs enabled selective Treg stimulation by engagement of OX40L, a key activation antigen in alloimmunity, including both graft-versus-host disease and solid organ transplant rejection, and autoimmunity, including rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus. We demonstrated that OX40L-CAR-Tregs were robustly activated in the presence of OX40L-expressing cells, leading to up-regulation of Treg suppressive proteins without induction of proinflammatory cytokine production. Compared with control Tregs, OX40L-CAR-Tregs more potently suppressed alloreactive T cell proliferation in vitro and were directly inhibitory toward activated monocyte-derived dendritic cells (DCs). We identified trogocytosis as one of the central mechanisms by which these CAR-Tregs effectively decrease extracellular display of OX40L, resulting in decreased DC stimulatory capacity. OX40L-CAR-Tregs demonstrated an enhanced ability to control xenogeneic graft-versus-host disease compared with control Tregs without abolishing the graft-versus-leukemia effect. These results suggest that OX40L-CAR-Tregs may have wide applicability as a potent cellular therapy to control both allo- and autoimmune diseases.
Collapse
Affiliation(s)
- Xianliang Rui
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Francesca Alvarez Calderon
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Holly Wobma
- Harvard Medical School, Boston, MA 02115, USA
- Division of Immunology, Boston Children's Hospital, Boston, MA 02215, USA
| | - Ulrike Gerdemann
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Alexandre Albanese
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Lorenzo Cagnin
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Connor McGuckin
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Kisa Naqvi
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Bruce R Blazar
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Victor Tkachev
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Leslie S Kean
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Li J, Zhang C, Xu Y, Yang L. Efficacy and safety of berberine plus 5-ASA for ulcerative colitis: A systematic review and meta-analysis. PLoS One 2024; 19:e0309144. [PMID: 39241013 PMCID: PMC11379390 DOI: 10.1371/journal.pone.0309144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/05/2024] [Indexed: 09/08/2024] Open
Abstract
PURPOSE This study aimed to assess the efficacy and safety of berberine(BBR) plus 5-aminosalicylic acid (5-ASA) for treating ulcerative colitis (UC). METHODS A comprehensive search was conducted in electronic databases, including Medline/PubMed, Sinomed, Embase, CNKI, Wanfang, and VIP, through January 2024 to identify all randomized controlled trials (RCTs) that administered BBR conjunction in standard therapy(5-ASA) for to support the treatment of UC. The data were synthesized using a meta-analysis approach with RevMan 5.4.1. The primary endpoint was the clinical efficacy rate. In contrast, the secondary endpoints included the Baron score, disease activity index (DAI) score, symptom relief latency, inflammatory markers, immunological indicators, and adverse events. RESULTS In this analysis, 10 RCTs comprising 952 patients with UC were examined. BBR considerably improved the clinical efficacy rate (RR = 1.22, 95% CI [1.15, 1.30], P < 0.00001), attenuated the Baron score (SMD = -1.72, 95% CI [-2.30, -1.13], P < 0.00001) and reduced the DAI score (SMD = -2.93, 95% CI [-4.42, -1.43], P < 0.00001). Additionally, it ameliorated clinical symptoms (SMD = -2.74, 95% CI [-3.45, 2.02], P < 0.00001), diminished inflammatory responses (SMD = -1.59, 95% CI [-2.14, 1.04], P < 0.00001), and modulated immune reactions (SMD = 1.06,95% CI [0.24,1.87], P <0.00001). Nonetheless, the impact of BBR on reducing adverse reactions was not statistically significant (RR = 0.75, 95% CI [0.42, 1.33], P > 0.05). CONCLUSION BBR demonstrates substantial efficacy in treating UC without causing severe adverse reactions and may serve as a viable complementary therapy. However, its clinical application warrants confirmation by additional high-quality, low-bias RCTs.
Collapse
Affiliation(s)
- Jilei Li
- Department of Oncology Diseases, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Chenchen Zhang
- Graduate School Department, Beijing University of Chinese Medicine, Beijing, China
| | - Yanchao Xu
- Department of Oncology Diseases, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Lili Yang
- Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Zou Y, Liu Y, Liu H, Feng J, Gao P, Ma H. Genetic mutation and immune infiltration in embryonal tumor with multilayered rosettes. Childs Nerv Syst 2024; 40:2685-2696. [PMID: 38802706 DOI: 10.1007/s00381-024-06461-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE Genetic mutations stand as pivotal factors leading to the occurrence of embryonal tumor with multilayered rosettes (ETMR). This study aims to identify improved treatment approaches by unraveling the genetic drivers and immune infiltration in ETMR. METHODS Two siblings with ETMR, treated at the General Hospital of Ningxia Medical University, were enrolled. Diagnosis involved MRI, Hematoxylin and Eosin (HE), and immunohistochemical (IHC) staining. Differentially expressed genes (DEGs) in ETMR were identified using GSE122077 and GSE14296 datasets. GO and KEGG analyses were used to determine ETMR-related pathways. Whole exome sequencing (WES) was utilized to annotate genetic variations in ETMR. Core genes, identified by protein-protein interaction (PPI), formed a diagnostic model evaluated by Logistic Regression. Single-sample Gene Set Enrichment Analysis (ssGSEA) assessed immune infiltration in ETMR, examining correlations between immune cells and core genes. RESULTS Two siblings were diagnosed with ETMR. In ETMR, 135 DEGs were identified, of which 25 genes were annotated with 28 mutation sites. Moreover, ETMR-related pathways included cell cycle, synaptic functions, and neurodegeneration. Three ETMR-related core genes (ALB, PSMD1, and PAK2) were screened by protein-protein interaction (PPI). The diagnostic model constructed using these genes demonstrated an AUC value of 0.901 (95% CI: 0.811-0.991) in the training set, indicating accurate predictions in ETMR. Enhanced ssGSEA scores for 16 immune cells in ETMR tissues suggested a strong immune response. CONCLUSION This study identifies diagnostic models associated with three core variant genes (ALB, PSMD1, PAK2) and enhanced immune cell activity in ETMR. It reveals crucial genetic features and significant immune responses in ETMR.
Collapse
Affiliation(s)
- Yourui Zou
- Department of Neurosurgery, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan, 750004, China
| | - Yang Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan, 750004, China
| | - Haibo Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan, 750004, China
| | - Jin Feng
- Department of Neurosurgery, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan, 750004, China
| | - Peng Gao
- Department of Neurosurgery, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan, 750004, China
| | - Hui Ma
- Department of Neurosurgery, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan, 750004, China.
| |
Collapse
|
6
|
Lepore MT, Bruzzaniti S, La Rocca C, Fusco C, Carbone F, Mottola M, Zuccarelli B, Lanzillo R, Brescia Morra V, Maniscalco GT, De Simone S, Procaccini C, Porcellini A, De Rosa V, Galgani M, Cassano S, Matarese G. Deciphering the role of protein kinase A in the control of FoxP3 expression in regulatory T cells in health and autoimmunity. Sci Rep 2024; 14:17571. [PMID: 39080325 PMCID: PMC11289137 DOI: 10.1038/s41598-024-68098-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
The molecular mechanisms that govern differential T cell development from CD4+CD25-conventional T (Tconv) into CD4+CD25+ forkhead-box-P3+ (FoxP3+) inducible regulatory T (iTreg) cells remain unclear. Herein, we investigated the relative contribution of protein kinase A (PKA) in this process. Mechanistically, we found that PKA controlled the efficiency of human iTreg cell generation through the expression of different FoxP3 splicing variants containing or not the exon 2. We found that transient PKA inhibition reduced the recruitment of cAMP-responsive element-binding protein (CREB) on regulatory regions of the FoxP3 gene, a condition that is associated with an impaired acquisition of their suppressive capacity in vitro. To corroborate our findings in a human model of autoimmunity, we measured CREB phosphorylation and FoxP3 levels in iTreg cells from treatment-naïve relapsing-remitting (RR)-multiple sclerosis (MS) subjects. Interestingly, both phospho-CREB and FoxP3 induction directly correlated and were significantly reduced in RR-MS patients, suggesting a previously unknown mechanism involved in the induction and function of human iTreg cells.
Collapse
Affiliation(s)
- Maria Teresa Lepore
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Sara Bruzzaniti
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Claudia La Rocca
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Clorinda Fusco
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Fortunata Carbone
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
- Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Mottola
- UOC di Medicina Trasfusionale, AORN Ospedale dei Colli, Ospedale Monaldi, Naples, Italy
| | - Bruno Zuccarelli
- UOC di Medicina Trasfusionale, AORN Ospedale dei Colli, Ospedale Monaldi, Naples, Italy
| | - Roberta Lanzillo
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Vincenzo Brescia Morra
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Giorgia Teresa Maniscalco
- Dipartimento di Neurologia, Centro Regionale Sclerosi Multipla, Azienda Ospedaliera "A. Cardarelli", Naples, Italy
| | - Salvatore De Simone
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Claudio Procaccini
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
- Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Porcellini
- Dipartimento di Biologia, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Mario Galgani
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Silvana Cassano
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy.
| |
Collapse
|
7
|
Sharma A, Jasrotia S, Kumar A. Effects of Chemotherapy on the Immune System: Implications for Cancer Treatment and Patient Outcomes. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2551-2566. [PMID: 37906273 DOI: 10.1007/s00210-023-02781-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023]
Abstract
Chemotherapy is a cornerstone of cancer treatment, but it can also induce immune suppression, which can have significant implications for patient outcomes. This review paper aims to give a general overview of how chemotherapy affects the immune system and how it affects cancer treatment. Chemotherapy can directly affect immune cells, leading to cytotoxic effects, cell differentiation and function alterations, and cell communication and signaling pathways disruptions. Such immune suppression can weaken the anti-tumor immune response and increase the risk of immune-related toxicities. Understanding the mechanisms of chemotherapy-induced immune suppression is crucial for optimizing treatment strategies. Strategies to mitigate immune suppression include immunomodulatory agents as adjuvants to chemotherapy, combination therapies to enhance immune function, and supportive care measures of the immune system. Additionally, identifying potential biomarkers to predict immune suppression and guide treatment decisions holds promise for personalized cancer medicine. Future directions in this field involve further elucidating underlying mechanisms, exploring novel combination therapies, and developing targeted interventions to minimize immune suppression. By understanding and addressing chemotherapy-induced immune suppression, we can optimize cancer treatment strategies, enhance the anti-tumor immune response, and improve patient outcomes.
Collapse
Affiliation(s)
- Anirudh Sharma
- Department of Biosciences (UIBT), Chandigarh University, Mohali, Punjab, 140413, India
| | - Shivam Jasrotia
- Department of Biosciences (UIBT), Chandigarh University, Mohali, Punjab, 140413, India.
| | - Ajay Kumar
- University Center for Research & Development (UCRD), Chandigarh University, Gharuan, Mohali, Punjab, 140413, India
| |
Collapse
|
8
|
Pacini CP, Soares MVD, Lacerda JF. The impact of regulatory T cells on the graft-versus-leukemia effect. Front Immunol 2024; 15:1339318. [PMID: 38711496 PMCID: PMC11070504 DOI: 10.3389/fimmu.2024.1339318] [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: 11/15/2023] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Allogeneic Hematopoietic Stem Cell Transplantation (allo-HSCT) is the only curative therapy for many hematologic malignancies, whereby the Graft-versus-Leukemia (GVL) effect plays a pivotal role in controlling relapse. However, the success of GVL is hindered by Graft-versus-Host Disease (GVHD), where donor T cells attack healthy tissues in the recipient. The ability of natural regulatory T cells (Treg) to suppress immune responses has been exploited as a therapeutical option against GVHD. Still, it is crucial to evaluate if the ability of Treg to suppress GVHD does not compromise the benefits of GVL. Initial studies in animal models suggest that Treg can attenuate GVHD while preserving GVL, but results vary according to tumor type. Human trials using Treg as GVHD prophylaxis or treatment show promising results, emphasizing the importance of infusion timing and Treg/Tcon ratios. In this review, we discuss strategies that can be used aiming to enhance GVL post-Treg infusion and the proposed mechanisms for the maintenance of the GVL effect upon the adoptive Treg transfer. In order to optimize the therapeutic outcomes of Treg administration in allo-HSCT, future efforts should focus on refining Treg sources for infusion and evaluating their specificity for antigens mediating GVHD while preserving GVL responses.
Collapse
Affiliation(s)
- Carolina P. Pacini
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria V. D. Soares
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João F. Lacerda
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, ULS Santa Maria, Lisbon, Portugal
| |
Collapse
|
9
|
Dittmar DJ, Pielmeier F, Strieder N, Fischer A, Herbst M, Stanewsky H, Wenzl N, Röseler E, Eder R, Gebhard C, Schwarzfischer-Pfeilschifter L, Albrecht C, Herr W, Edinger M, Hoffmann P, Rehli M. Donor regulatory T cells rapidly adapt to recipient tissues to control murine acute graft-versus-host disease. Nat Commun 2024; 15:3224. [PMID: 38622133 PMCID: PMC11018811 DOI: 10.1038/s41467-024-47575-z] [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: 04/03/2023] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
The adoptive transfer of regulatory T cells is a promising strategy to prevent graft-versus-host disease after allogeneic bone marrow transplantation. Here, we use a major histocompatibility complex-mismatched mouse model to follow the fate of in vitro expanded donor regulatory T cells upon migration to target organs. Employing comprehensive gene expression and repertoire profiling, we show that they retain their suppressive function and plasticity after transfer. Upon entering non-lymphoid tissues, donor regulatory T cells acquire organ-specific gene expression profiles resembling tissue-resident cells and activate hallmark suppressive and cytotoxic pathways, most evidently in the colon, when co-transplanted with graft-versus-host disease-inducing conventional T cells. Dominant T cell receptor clonotypes overlap between organs and across recipients and their relative abundance correlates with protection efficacy. Thus, this study reveals donor regulatory T cell selection and adaptation mechanisms in target organs and highlights protective features of Treg to guide the development of improved graft-versus-host disease prevention strategies.
Collapse
Affiliation(s)
- David J Dittmar
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
- BioNTech SE, 82061, Neuried, Germany
| | - Franziska Pielmeier
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | | | - Alexander Fischer
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Michael Herbst
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
- Institute of Experimental Immunology, Research Unit Tumorimmunology, University of Zurich, Zurich, Switzerland
| | - Hanna Stanewsky
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Niklas Wenzl
- Leibniz Institute for Immunotherapy, 93053, Regensburg, Germany
| | - Eveline Röseler
- Leibniz Institute for Immunotherapy, 93053, Regensburg, Germany
| | - Rüdiger Eder
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Claudia Gebhard
- Leibniz Institute for Immunotherapy, 93053, Regensburg, Germany
| | | | - Christin Albrecht
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Matthias Edinger
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany.
- Leibniz Institute for Immunotherapy, 93053, Regensburg, Germany.
| | - Petra Hoffmann
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany.
- Leibniz Institute for Immunotherapy, 93053, Regensburg, Germany.
| | - Michael Rehli
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany.
- Leibniz Institute for Immunotherapy, 93053, Regensburg, Germany.
| |
Collapse
|
10
|
Wardell CM, Fung VC, Chen E, Haque M, Gillies J, Spanier JA, Mojibian M, Fife BT, Levings MK. Short Report: CAR Tregs mediate linked suppression and infectious tolerance in islet transplantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.06.588414. [PMID: 38645184 PMCID: PMC11030375 DOI: 10.1101/2024.04.06.588414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Regulatory T cells (Tregs) have potential as a cell-based therapy to prevent or treat transplant rejection and autoimmunity. Using an HLA-A2-specific chimeric antigen receptor (A2-CAR), we previously showed that adoptive transfer of A2-CAR Tregs limited anti-HLA-A2 alloimmunity. However, it was unknown if A2-CAR Tregs could also limit immunity to autoantigens. Using a model of HLA-A2+ islet transplantation into immunodeficient non-obese diabetic mice, we investigated if A2-CAR Tregs could control diabetes induced by islet-autoreactive (BDC2.5) T cells. In mice transplanted with HLA-A2+ islets, A2-CAR Tregs reduced BDC2.5 T cell engraftment, proliferation and cytokine production, and protected mice from diabetes. Tolerance to islets was systemic, including protection of the HLA-A2negative endogenous pancreas. In tolerant mice, a significant proportion of BDC2.5 T cells gained FOXP3 expression suggesting that long-term tolerance is maintained by de novo Treg generation. Thus, A2-CAR Tregs mediate linked suppression and infectious tolerance and have potential therapeutic use to simultaneously control both allo- and autoimmunity in islet transplantation.
Collapse
Affiliation(s)
- Christine M. Wardell
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Vivian C.W. Fung
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Eleanor Chen
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Manjurul Haque
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Jana Gillies
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Justin A. Spanier
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN, USA
- Center for Autoimmune Disease Research, Department of Medicine, University of Minnesota Medical School; Minneapolis, MN, USA
- Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School; Minneapolis, MN, USA
| | - Majid Mojibian
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
| | - Brian T. Fife
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN, USA
- Center for Autoimmune Disease Research, Department of Medicine, University of Minnesota Medical School; Minneapolis, MN, USA
- Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School; Minneapolis, MN, USA
| | - Megan K. Levings
- BC Children’s Hospital Research Institute, University of British Columbia; Vancouver, BC, Canada
- Dept of Surgery, University of British Columbia; Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia; Vancouver, BC, Canada
| |
Collapse
|
11
|
López Ruiz A, Slaughter ED, Kloxin AM, Fromen CA. Bridging the gender gap in autoimmunity with T-cell-targeted biomaterials. Curr Opin Biotechnol 2024; 86:103075. [PMID: 38377884 PMCID: PMC11578274 DOI: 10.1016/j.copbio.2024.103075] [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: 12/10/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/22/2024]
Abstract
Autoimmune diseases are caused by malfunctions of the immune system and generally impact women at twice the frequency of men. Many of the most serious autoimmune diseases are accompanied by a dysregulation of T-cell phenotype, both regarding the ratio of CD4+ to CD8+ T-cells and proinflammatory versus regulatory phenotypes. Biomaterials, in the form of particles and hydrogels, have shown promise in ameliorating this dysregulation both in vivo and ex vivo. In this review, we explore the role of T-cells in autoimmune diseases, particularly those with high incidence rates in women, and evaluate the promise and efficacy of innovative biomaterial-based approaches for targeting T-cells.
Collapse
Affiliation(s)
- Aida López Ruiz
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
| | - Eric D Slaughter
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States; Material Science and Engineering, University of Delaware, Newark, DE, United States.
| | - Catherine A Fromen
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States.
| |
Collapse
|
12
|
Peng S, Lin A, Jiang A, Zhang C, Zhang J, Cheng Q, Luo P, Bai Y. CTLs heterogeneity and plasticity: implications for cancer immunotherapy. Mol Cancer 2024; 23:58. [PMID: 38515134 PMCID: PMC10956324 DOI: 10.1186/s12943-024-01972-6] [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: 12/25/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs) play critical antitumor roles, encompassing diverse subsets including CD4+, NK, and γδ T cells beyond conventional CD8+ CTLs. However, definitive CTLs biomarkers remain elusive, as cytotoxicity-molecule expression does not necessarily confer cytotoxic capacity. CTLs differentiation involves transcriptional regulation by factors such as T-bet and Blimp-1, although epigenetic regulation of CTLs is less clear. CTLs promote tumor killing through cytotoxic granules and death receptor pathways, but may also stimulate tumorigenesis in some contexts. Given that CTLs cytotoxicity varies across tumors, enhancing this function is critical. This review summarizes current knowledge on CTLs subsets, biomarkers, differentiation mechanisms, cancer-related functions, and strategies for improving cytotoxicity. Key outstanding questions include refining the CTLs definition, characterizing subtype diversity, elucidating differentiation and senescence pathways, delineating CTL-microbe relationships, and enabling multi-omics profiling. A more comprehensive understanding of CTLs biology will facilitate optimization of their immunotherapy applications. Overall, this review synthesizes the heterogeneity, regulation, functional roles, and enhancement strategies of CTLs in antitumor immunity, highlighting gaps in our knowledge of subtype diversity, definitive biomarkers, epigenetic control, microbial interactions, and multi-omics characterization. Addressing these questions will refine our understanding of CTLs immunology to better leverage cytotoxic functions against cancer.
Collapse
Affiliation(s)
- Shengkun Peng
- Department of Radiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and ImmunologySchool of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South University, Hunan, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
13
|
Talvard-Balland N, Lambert M, Chevalier MF, Minet N, Salou M, Tourret M, Bohineust A, Milo I, Parietti V, Yvorra T, Socié G, Lantz O, Caillat-Zucman S. Human MAIT cells inhibit alloreactive T cell responses and protect against acute graft-versus-host disease. JCI Insight 2024; 9:e166310. [PMID: 38300704 PMCID: PMC11143928 DOI: 10.1172/jci.insight.166310] [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: 10/14/2022] [Accepted: 01/30/2024] [Indexed: 02/03/2024] Open
Abstract
Adoptive transfer of immunoregulatory cells can prevent or ameliorate graft-versus-host disease (GVHD), which remains the main cause of nonrelapse mortality after allogeneic hematopoietic stem cell transplantation. Mucosal-associated invariant T (MAIT) cells were recently associated with tissue repair capacities and with lower rates of GVHD in humans. Here, we analyzed the immunosuppressive effect of MAIT cells in an in vitro model of alloreactivity and explored their adoptive transfer in a preclinical xenogeneic GVHD model. We found that MAIT cells, whether freshly purified or short-term expanded, dose-dependently inhibited proliferation and activation of alloreactive T cells. In immunodeficient mice injected with human PBMCs, MAIT cells greatly delayed GVHD onset and decreased severity when transferred early after PBMC injection but could also control ongoing GVHD when transferred at delayed time points. This effect was associated with decreased proliferation and effector function of human T cells infiltrating tissues of diseased mice and was correlated with lower circulating IFN-γ and TNF-α levels and increased IL-10 levels. MAIT cells acted partly in a contact-dependent manner, which likely required direct interaction of their T cell receptor with MHC class I-related molecule (MR1) induced on host-reactive T cells. These results support the setup of clinical trials using MAIT cells as universal therapeutic tools to control severe GVHD or mucosal inflammatory disorders.
Collapse
Affiliation(s)
- Nana Talvard-Balland
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
| | - Marion Lambert
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
| | - Mathieu F. Chevalier
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
| | - Norbert Minet
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
| | - Marion Salou
- Institut Curie, Université PSL, INSERM U932, Immunity and Cancer, Paris, France
| | - Marie Tourret
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
| | - Armelle Bohineust
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
| | - Idan Milo
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
| | - Véronique Parietti
- Université Paris Cité, INSERM, CNRS, UMS Saint-Louis (US53/UAR2030), Paris, France
| | - Thomas Yvorra
- Institut Curie, Université PSL, CNRS UMR3666, INSERM U1143, Paris, France
| | - Gérard Socié
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
- Hematology Transplantation, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Olivier Lantz
- Institut Curie, Université PSL, INSERM U932, Immunity and Cancer, Paris, France
- Clinical Immunology Laboratory, Institut Curie, Paris, France
- Centre d’investigation Clinique en Biothérapie Gustave-Roussy Institut Curie (CIC-BT1428), Paris, France
| | - Sophie Caillat-Zucman
- INSERM UMR-976 HIPI, Saint Louis Research Institute, Université Paris Cité, Paris, France
- Immunology Laboratory, Hôpital Saint-Louis, AP-HP, Université Paris Cité, Paris, France
| |
Collapse
|
14
|
Klug N, Burke J, Scott E. Rational Engineering of Islet Tolerance via Biomaterial-Mediated Immune Modulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:216-224. [PMID: 38166244 PMCID: PMC10766078 DOI: 10.4049/jimmunol.2300527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/17/2023] [Indexed: 01/04/2024]
Abstract
Type 1 diabetes (T1D) onset is characterized by an autoimmune attack on β islet cells within the pancreas, preventing the insulin secretion required to maintain glucose homeostasis. Targeted modulation of key immunoregulatory cell populations is a promising strategy to restore tolerance to β cells. This strategy can be used to prevent T1D onset or reverse T1D with transplanted islets. To this end, drug delivery systems can be employed to transport immunomodulatory cargo to specific cell populations that inhibit autoreactive T cell-mediated destruction of the β cell mass. The rational engineering of biomaterials into nanoscale and microscale drug carriers can facilitate targeted interactions with immune cells. The physicochemical properties of the biomaterial, the delivered immunomodulatory agent, and the target cell populations are critical variables in the design of these delivery systems. In this review, we discuss recent biomaterials-based drug delivery approaches to induce islet tolerance and the need to consider both immune and metabolic markers of disease progression.
Collapse
Affiliation(s)
- Natalie Klug
- Department of Biomedical Engineering, Robert R. McCormick School of Engineering and Applied Science, Northwestern University, Evanston, IL
| | - Jacqueline Burke
- Department of Biomedical Engineering, Robert R. McCormick School of Engineering and Applied Science, Northwestern University, Evanston, IL
| | - Evan Scott
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
15
|
Kennedy-Batalla R, Acevedo D, Luo Y, Esteve-Solé A, Vlagea A, Correa-Rocha R, Seoane-Reula ME, Alsina L. Treg in inborn errors of immunity: gaps, knowns and future perspectives. Front Immunol 2024; 14:1278759. [PMID: 38259469 PMCID: PMC10800401 DOI: 10.3389/fimmu.2023.1278759] [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: 08/16/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
Regulatory T cells (Treg) are essential for immune balance, preventing overreactive responses and autoimmunity. Although traditionally characterized as CD4+CD25+CD127lowFoxP3hi, recent research has revealed diverse Treg subsets such as Tr1, Tr1-like, and CD8 Treg. Treg dysfunction leads to severe autoimmune diseases and immune-mediated inflammatory disorders. Inborn errors of immunity (IEI) are a group of disorders that affect correct functioning of the immune system. IEI include Tregopathies caused by genetic mutations affecting Treg development or function. In addition, Treg dysfunction is also observed in other IEIs, whose underlying mechanisms are largely unknown, thus requiring further research. This review provides a comprehensive overview and discussion of Treg in IEI focused on: A) advances and controversies in the evaluation of Treg extended subphenotypes and function; B) current knowledge and gaps in Treg disturbances in Tregopathies and other IEI including Treg subpopulation changes, genotype-phenotype correlation, Treg changes with disease activity, and available therapies, and C) the potential of Treg cell-based therapies for IEI with immune dysregulation. The aim is to improve both the diagnostic and the therapeutic approaches to IEI when there is involvement of Treg. We performed a non-systematic targeted literature review with a knowledgeable selection of current, high-quality original and review articles on Treg and IEI available since 2003 (with 58% of the articles within the last 6 years) in the PubMed database.
Collapse
Affiliation(s)
- Rebeca Kennedy-Batalla
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Daniel Acevedo
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Yiyi Luo
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Ana Esteve-Solé
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Alexandru Vlagea
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Immunology Department, Biomedic Diagnostic Center (CDB), Hospital Clínic of Barcelona, Clinical Immunology Unit Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Ma Elena Seoane-Reula
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatric Immuno-Allergy Unit, Allergy Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Primary Immunodeficiencies Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
16
|
Ou Q, Power R, Griffin MD. Revisiting regulatory T cells as modulators of innate immune response and inflammatory diseases. Front Immunol 2023; 14:1287465. [PMID: 37928540 PMCID: PMC10623442 DOI: 10.3389/fimmu.2023.1287465] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Regulatory T cells (Treg) are known to be critical for the maintenance of immune homeostasis by suppressing the activation of auto- or allo-reactive effector T cells through a diverse repertoire of molecular mechanisms. Accordingly, therapeutic strategies aimed at enhancing Treg numbers or potency in the setting of autoimmunity and allogeneic transplants have been energetically pursued and are beginning to yield some encouraging outcomes in early phase clinical trials. Less well recognized from a translational perspective, however, has been the mounting body of evidence that Treg directly modulate most aspects of innate immune response under a range of different acute and chronic disease conditions. Recognizing this aspect of Treg immune modulatory function provides a bridge for the application of Treg-based therapies to common medical conditions in which organ and tissue damage is mediated primarily by inflammation involving myeloid cells (mononuclear phagocytes, granulocytes) and innate lymphocytes (NK cells, NKT cells, γδ T cells and ILCs). In this review, we comprehensively summarize pre-clinical and human research that has revealed diverse modulatory effects of Treg and specific Treg subpopulations on the range of innate immune cell types. In each case, we emphasize the key mechanistic insights and the evidence that Treg interactions with innate immune effectors can have significant impacts on disease severity or treatment. Finally, we discuss the opportunities and challenges that exist for the application of Treg-based therapeutic interventions to three globally impactful, inflammatory conditions: type 2 diabetes and its end-organ complications, ischemia reperfusion injury and atherosclerosis.
Collapse
Affiliation(s)
- Qifeng Ou
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Rachael Power
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
- Nephrology Department, Galway University Hospitals, Saolta University Healthcare Group, Galway, Ireland
| |
Collapse
|
17
|
Jeyamogan S, Leventhal JR, Mathew JM, Zhang ZJ. CD4 +CD25 +FOXP3 + regulatory T cells: a potential "armor" to shield "transplanted allografts" in the war against ischemia reperfusion injury. Front Immunol 2023; 14:1270300. [PMID: 37868962 PMCID: PMC10587564 DOI: 10.3389/fimmu.2023.1270300] [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: 07/31/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Despite the advances in therapeutic interventions, solid organ transplantation (SOT) remains the "gold standard" treatment for patients with end-stage organ failure. Recently, vascularized composite allotransplantation (VCA) has reemerged as a feasible treatment option for patients with complex composite tissue defects. In both SOT and VCA, ischemia reperfusion injury (IRI) is inevitable and is a predominant factor that can adversely affect transplant outcome by potentiating early graft dysfunction and/or graft rejection. Restoration of oxygenated blood supply to an organ which was previously hypoxic or ischemic for a period of time triggers cellular oxidative stress, production of both, pro-inflammatory cytokines and chemokines, infiltration of innate immune cells and amplifies adaptive alloimmune responses in the affected allograft. Currently, Food and Drug Administration (FDA) approved drugs for the treatment of IRI are unavailable, therefore an efficacious therapeutic modality to prevent, reduce and/or alleviate allograft damages caused by IRI induced inflammation is warranted to achieve the best-possible transplant outcome among recipients. The tolerogenic capacity of CD4+CD25+FOXP3+ regulatory T cells (Tregs), have been extensively studied in the context of transplant rejection, autoimmunity, and cancer. It was not until recently that Tregs have been recognized as a potential cell therapeutic candidate to be exploited for the prevention and/or treatment of IRI, owing to their immunomodulatory potential. Tregs can mitigate cellular oxidative stress, produce anti-inflammatory cytokines, promote wound healing, and tissue repair and prevent the infiltration of pro-inflammatory immune cells in injured tissues. By using strategic approaches to increase the number of Tregs and to promote targeted delivery, the outcome of SOT and VCA can be improved. This review focuses on two sections: (a) the therapeutic potential of Tregs in preventing and mitigating IRI in the context of SOT and VCA and (b) novel strategies on how Tregs could be utilized for the prevention and/or treatment of IRI.
Collapse
Affiliation(s)
- Shareni Jeyamogan
- Department of Surgery, Comprehensive Transplant Center 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
| | - James M. Mathew
- 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
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Zheng Jenny Zhang
- 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
- Microsurgery and Pre-Clinical Research Core, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
18
|
Venkatesh H, Tracy SI, Farrar MA. Cytotoxic CD4 T cells in the mucosa and in cancer. Front Immunol 2023; 14:1233261. [PMID: 37654482 PMCID: PMC10466411 DOI: 10.3389/fimmu.2023.1233261] [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: 06/01/2023] [Accepted: 07/24/2023] [Indexed: 09/02/2023] Open
Abstract
CD4 T cells were initially described as helper cells that promote either the cellular immune response (Th1 cells) or the humoral immune response (Th2 cells). Since then, a plethora of functionally distinct helper and regulatory CD4 T cell subsets have been described. CD4 T cells with cytotoxic function were first described in the setting of viral infections and autoimmunity, and more recently in cancer and gut dysbiosis. Regulatory CD4 T cell subsets such as Tregs and T-regulatory type 1 (Tr1) cells have also been shown to have cytotoxic potential. Indeed, Tr1 cells have been shown to be important for maintenance of stem cell niches in the bone marrow and the gut. This review will provide an overview of cytotoxic CD4 T cell development, and discuss the role of inflammatory and Tr1-like cytotoxic CD4 T cells in maintenance of intestinal stem cells and in anti-cancer immune responses.
Collapse
Affiliation(s)
- Hrishi Venkatesh
- Center for Immunology, Masonic Cancer Center, Minneapolis, MN, United States
- University of Minnesota, Department of Laboratory Medicine and Pathology, Minneapolis, MN, United States
| | - Sean I. Tracy
- Center for Immunology, Masonic Cancer Center, Minneapolis, MN, United States
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Michael A. Farrar
- Center for Immunology, Masonic Cancer Center, Minneapolis, MN, United States
- University of Minnesota, Department of Laboratory Medicine and Pathology, Minneapolis, MN, United States
| |
Collapse
|
19
|
Doglio M, Crossland RE, Alho AC, Penack O, Dickinson AM, Stary G, Lacerda JF, Eissner G, Inngjerdingen M. Cell-based therapy in prophylaxis and treatment of chronic graft-versus-host disease. Front Immunol 2022; 13:1045168. [PMID: 36466922 PMCID: PMC9714556 DOI: 10.3389/fimmu.2022.1045168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/31/2022] [Indexed: 08/31/2023] Open
Abstract
Hematopoietic allogeneic stem cell transplantation (allo-SCT) is a curative option for patients with hematological malignancies. However, due to disparities in major and minor histocompatibility antigens between donor and recipient, severe inflammatory complications can occur, among which chronic graft-versus-host disease (cGVHD) can be life-threatening. A classical therapeutic approach to the prevention and treatment of cGVHD has been broad immunosuppression, but more recently adjuvant immunotherapies have been tested. This review summarizes and discusses immunomodulatory approaches with T cells, including chimeric antigen receptor (CAR) and regulatory T cells, with natural killer (NK) cells and innate lymphoid cells (ILCs), and finally with mesenchymal stromal cells (MSC) and extracellular vesicles thereof. Clinical studies and pre-clinical research results are presented likewise.
Collapse
Affiliation(s)
- Matteo Doglio
- Experimental Haematology Unit, Division of Immunology Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, Milan, Italy
| | - Rachel E. Crossland
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ana C. Alho
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | - Olaf Penack
- Department of Hematology, Oncology, and Cancer Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anne M. Dickinson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Alcyomics Ltd, Newcastle upon Tyne, United Kingdom
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - João F. Lacerda
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | - Günther Eissner
- Systems Biology Ireland, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Marit Inngjerdingen
- Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
20
|
Bolivar-Wagers S, Loschi ML, Jin S, Thangavelu G, Larson JH, McDonald-Hyman CS, Aguilar EG, Saha A, Koehn BH, Hefazi M, Osborn MJ, Jensen MC, Wagner JE, Pennell CA, Blazar BR. Murine CAR19 Tregs suppress acute graft-versus-host disease and maintain graft-versus-tumor responses. JCI Insight 2022; 7:e160674. [PMID: 35917188 PMCID: PMC9536261 DOI: 10.1172/jci.insight.160674] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/21/2022] [Indexed: 02/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) efficacy is complicated by graft-versus-host disease (GVHD), a leading cause of morbidity and mortality. Regulatory T cells (Tregs) have shown efficacy in preventing GVHD. However, high Treg doses are often required, necessitating substantial ex vivo or in vivo expansion that may diminish suppressor function. To enhance in vivo suppressor function, murine Tregs were transduced to express an anti-human CD19 chimeric antigen receptor (hCAR19) and infused into lethally irradiated, hCD19-transgenic recipients for allo-HSCT. Compared with recipients receiving control transduced Tregs, those receiving hCAR19 Tregs had a marked decrease in acute GVHD lethality. Recipient hCD19 B cells and murine hCD19 TBL12-luciferase (TBL12luc) lymphoma cells were both cleared by allogeneic hCAR19 Tregs, which was indicative of graft-versus-tumor (GVT) maintenance and potentiation. Mechanistically, hCAR19 Tregs killed syngeneic hCD19+ but not hCD19- murine TBL12luc cells in vitro in a perforin-dependent, granzyme B-independent manner. Importantly, cyclophosphamide-treated, hCD19-transgenic mice given hCAR19 cytotoxic T lymphocytes without allo-HSCT experienced rapid lethality due to systemic toxicity that has been associated with proinflammatory cytokine release; in contrast, hCAR19 Treg suppressor function enabled avoidance of this severe complication. In conclusion, hCAR19 Tregs are a potentially novel and effective strategy to suppress GVHD without loss of GVT responses.
Collapse
Affiliation(s)
- Sara Bolivar-Wagers
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Michael L. Loschi
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Sujeong Jin
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Govindarajan Thangavelu
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Jemma H. Larson
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Cameron S. McDonald-Hyman
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Ethan G. Aguilar
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Asim Saha
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Brent H. Koehn
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Mehrdad Hefazi
- Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark J. Osborn
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Michael C. Jensen
- Department of Pediatrics, Division of Hematology and Oncology, University of Washington, Seattle, Washington, USA
| | - John E. Wagner
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| | - Christopher A. Pennell
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, and
| |
Collapse
|