1
|
Lupsa N, Érsek B, Böröczky C, Kis D, Szarka E, Lumniczky K, Sáfrány G, Zádori ZS, Szöőr Á, Buzás EI, Pós Z. High sensitivity of host Helios +/Neuropilin-1 + Treg to pretransplant conditioning hampers development of OX40 bright/integrin-β7 + regulatory cells in acute gastrointestinal GvHD. Eur J Immunol 2024; 54:e2350619. [PMID: 38532599 DOI: 10.1002/eji.202350619] [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: 06/23/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
This study sought to compare the behavior of Treg subsets displaying different coexpression patterns of Neuropilin-1 (Nrp1) and Helios, under the influence of gut stress unrelated to hematopoietic stem cell transplantation, pretransplantation conditioning, and posttransplant gastrointestinal acute graft versus host disease (GI-aGvHD). Host CD4+/CD25hi/Foxp3+ Treg cells, identified by flow cytometry, were isolated from various tissues of mice affected by these stressors. Expression of CD25, CTLA-4, CD39, OX40, integrin-β7, LAG3, TGFβ/LAP, granzyme-A, -B, and interleukin-10 was compared in four Treg subsets displaying Helios or Nrp1 only, both or none. Fluorescence-activated cell sorter-sorted Treg subsets, displaying markers affected in a conditioning- and GI-aGVHD-restricted manner, were further investigated by transcriptome profiling and T-cell suppression assays. We found that conditioning by irradiation greatly diminished the relative frequency of Helios+/Nrp1+ Treg, shifting the balance toward Helios-/Nrp1- Treg in the host. Upregulation of integrin-β7 and OX40 occurred in GI-aGvHD-dependent manner in Helios+/Nrp1+ cells but not in Helios-/Nrp1- Treg. Sorted Treg subsets, confirmed to overexpress Nrp1, Helios, OX40, or integrin-β7, displayed superior immunosuppressive activity and enrichment in activation-related messenger RNA transcripts. Our data suggest that conditioning-induced shrinkage of the Nrp1+/Helios+ Treg subset may contribute to the development of GI-GvHD by impairing gut homing and decreasing the efficiency of Treg-mediated immunosuppression.
Collapse
Affiliation(s)
- Nikolett Lupsa
- Department of Genetics, Cell and Immunobiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Barbara Érsek
- Department of Genetics, Cell and Immunobiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Csenge Böröczky
- Department of Genetics, Cell and Immunobiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Dávid Kis
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Public Health Center, Budapest, Hungary
| | - Eszter Szarka
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Public Health Center, Budapest, Hungary
| | - Katalin Lumniczky
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Public Health Center, Budapest, Hungary
| | - Géza Sáfrány
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Public Health Center, Budapest, Hungary
| | - Zoltán S Zádori
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell and Immunobiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Immunproteogenomics Extracellular Vesicle Research Group of the Hungarian Academy of Sciences-Semmelweis University, Budapest, Hungary
- Extracellular Vesicle Research Group, Hungarian Center of Excellence Molecular Medicine, Budapest, Hungary
| | - Zoltán Pós
- Department of Genetics, Cell and Immunobiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
2
|
Khatun A, Wu X, Qi F, Gai K, Kharel A, Kudek MR, Fraser L, Ceicko A, Kasmani MY, Majnik A, Burns R, Chen Y, Salzman N, Taparowsky EJ, Fang D, Williams CB, Cui W. BATF is Required for Treg Homeostasis and Stability to Prevent Autoimmune Pathology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206692. [PMID: 37587835 PMCID: PMC10558681 DOI: 10.1002/advs.202206692] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/17/2023] [Indexed: 08/18/2023]
Abstract
Regulatory T (Treg) cells are inevitable to prevent deleterious immune responses to self and commensal microorganisms. Treg function requires continuous expression of the transcription factor (TF) FOXP3 and is divided into two major subsets: resting (rTregs) and activated (aTregs). Continuous T cell receptor (TCR) signaling plays a vital role in the differentiation of aTregs from their resting state, and in their immune homeostasis. The process by which Tregs differentiate, adapt tissue specificity, and maintain stable phenotypic expression at the transcriptional level is still inconclusivei. In this work, the role of BATF is investigated, which is induced in response to TCR stimulation in naïve T cells and during aTreg differentiation. Mice lacking BATF in Tregs developed multiorgan autoimmune pathology. As a transcriptional regulator, BATF is required for Treg differentiation, homeostasis, and stabilization of FOXP3 expression in different lymphoid and non-lymphoid tissues. Epigenetically, BATF showed direct regulation of Treg-specific genes involved in differentiation, maturation, and tissue accumulation. Most importantly, FOXP3 expression and Treg stability require continuous BATF expression in Tregs, as it regulates demethylation and accessibility of the CNS2 region of the Foxp3 locus. Considering its role in Treg stability, BATF should be considered an important therapeutic target in autoimmune disease.
Collapse
Affiliation(s)
- Achia Khatun
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
| | - Xiaopeng Wu
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
| | - Fu Qi
- Children's Mercy Hospital in Kansas City2401 Gillham RdKansas CityMO64108USA
| | - Kexin Gai
- Department of PathologyFeinberg School of MedicineNorthwestern University303 E Chicago AveChicagoIL60611USA
| | - Arjun Kharel
- Department of PathologyFeinberg School of MedicineNorthwestern University303 E Chicago AveChicagoIL60611USA
| | - Matthew R. Kudek
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
- Department of PediatricsMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWI53226USA
| | - Lisa Fraser
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
| | - Ashley Ceicko
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
| | - Moujtaba Y. Kasmani
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
| | - Amber Majnik
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Children's Mercy Hospital in Kansas City2401 Gillham RdKansas CityMO64108USA
| | - Robert Burns
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
| | - Yi‐Guang Chen
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Max McGee National Research Center for Juvenile DiabetesMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWI53226USA
| | - Nita Salzman
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Department of PediatricsMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWI53226USA
| | | | - Dayu Fang
- Department of PathologyFeinberg School of MedicineNorthwestern University303 E Chicago AveChicagoIL60611USA
| | - Calvin B. Williams
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Department of PediatricsMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWI53226USA
| | - Weiguo Cui
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
- Department of PathologyFeinberg School of MedicineNorthwestern University303 E Chicago AveChicagoIL60611USA
| |
Collapse
|
3
|
Marshall G, Cserny J, Wang CW, Looney B, Posgai AL, Bacher R, Keselowsky B, Brusko TM. Biomaterials-based nanoparticles conjugated to regulatory T cells provide a modular system for localized delivery of pharmacotherapeutic agents. J Biomed Mater Res A 2023; 111:185-197. [PMID: 36082558 PMCID: PMC9742177 DOI: 10.1002/jbm.a.37442] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes (T1D) presents with two therapeutic challenges: the need to correct underlying autoimmunity and restore β-cell mass. We harnessed the unique capacity of regulatory T cells (Tregs) and the T cell receptor (TCR) to direct tolerance induction along with tissue-localized delivery of therapeutic agents to restore endogenous β-cell function. Specifically, we designed a combinatorial therapy involving biomaterials-based poly(lactic-co-glycolic acid) nanoparticles co-loaded with the Treg growth factor, IL-2, and the β-cell regenerative agent, harmine (a tyrosine-regulated kinase 1A [DYRK1A] inhibitor), conjugated to the surface of Tregs. We observed continuous elution of IL-2 and harmine from nanoparticles for at least 7 days in vitro. When conjugated to primary human Tregs, IL-2 nanoparticles provided sufficient IL-2 receptor signaling to support STAT5 phosphorylation for sustained phenotypic stability and viability in culture. Inclusion of poly-L-lysine (PLL) during nanoparticle-cell coupling dramatically increased conjugation efficiency, providing sufficient IL-2 to support in vitro proliferation of IL-2-dependent CTLL-2 cells and primary murine Tregs. In 12-week-old female non-obese diabetic mice, adoptive transfer of IL-2/harmine nanoparticle-conjugated NOD.BDC2.5 Tregs, which express an islet antigen-specific TCR, significantly prevented diabetes demonstrating preserved in vivo viability. These data provide the preclinical basis to develop a biomaterials-optimized cellular therapy to restore immune tolerance and promote β-cell proliferation in T1D through receptor-targeted drug delivery within pancreatic islets.
Collapse
Affiliation(s)
| | - Judit Cserny
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32601, USA
| | | | | | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32601, USA
| | - Rhonda Bacher
- Department of Biostatistics, College of Public Health and Health Professions, and College of Medicine, University of Florida, Gainesville, FL
| | - Benjamin Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32601, USA
| | - Todd M. Brusko
- Inspira Therapeutics, Inc., Alachua, FL 32615, USA,Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32601, USA,Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32601, USA,Correspondence to: Todd M. Brusko, PhD, Department of Pathology, University of Florida, College of Medicine, Box 100275, 1600 SW Archer Road, Gainesville, FL 32610; (352) 273-9255; Fax (352) 273-9339;
| |
Collapse
|
4
|
Kurya AU, Aliyu U, Tudu AI, Usman A, Yusuf M, Gupta S, Ali A, Gulfishan M, Singh SK, Hussain I, Abubakar MG. Graft-versus-host disease: Therapeutic prospects of improving the long-term post-transplant outcomes. TRANSPLANTATION REPORTS 2022. [DOI: 10.1016/j.tpr.2022.100107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
5
|
Lentz LS, Stutz AJ, Meyer N, Schubert K, Karkossa I, von Bergen M, Zenclussen AC, Schumacher A. Human chorionic gonadotropin promotes murine Treg cells and restricts pregnancy-harmful proinflammatory Th17 responses. Front Immunol 2022; 13:989247. [PMID: 36203576 PMCID: PMC9531259 DOI: 10.3389/fimmu.2022.989247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/25/2022] [Indexed: 11/15/2022] Open
Abstract
An equilibrium between proinflammatory and anti-inflammatory immune responses is essential for maternal tolerance of the fetus throughout gestation. To study the participation of fetal tissue-derived factors in this delicate immune balance, we analyzed the effects of human chorionic gonadotropin (hCG) on murine Treg cells and Th17 cells in vitro, and on pregnancy outcomes, fetal and placental growth, blood flow velocities and remodeling of the uterine vascular bed in vivo. Compared with untreated CD4+CD25+ T cells, hCG increased the frequency of Treg cells upon activation of the LH/CG receptor. hCG, with the involvement of IL-2, also interfered with induced differentiation of CD4+ T cells into proinflammatory Th17 cells. In already differentiated Th17 cells, hCG induced an anti-inflammatory profile. Transfer of proinflammatory Th17 cells into healthy pregnant mice promoted fetal rejection, impaired fetal growth and resulted in insufficient remodeling of uterine spiral arteries, and abnormal flow velocities. Our works show that proinflammatory Th17 cells have a negative influence on pregnancy that can be partly avoided by in vitro re-programming of proinflammatory Th17 cells with hCG.
Collapse
Affiliation(s)
- Lea S. Lentz
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
| | - Annika J. Stutz
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
| | - Nicole Meyer
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Isabel Karkossa
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
- Faculty of Life Sciences, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Ana C. Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Anne Schumacher
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany
- *Correspondence: Anne Schumacher,
| |
Collapse
|
6
|
Combined treatment of graft versus host disease using donor regulatory T cells and ruxolitinib. Sci Rep 2022; 12:8348. [PMID: 35589917 PMCID: PMC9120462 DOI: 10.1038/s41598-022-12407-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Donor derived regulatory T lymphocytes and the JAK1/2 kinase inhibitor ruxolitinib are currently being evaluated as therapeutic options in the treatment of chronic graft versus host disease (cGvHD). In this work, we aimed to determine if the combined use of both agents can exert a synergistic effect in the treatment of GvHD. For this purpose, we studied the effect of this combination both in vitro and in a GvHD mouse model. Our results show that ruxolitinib favors the ratio of thymic regulatory T cells to conventional T cells in culture, without affecting the suppressive capacity of these Treg. The combination of ruxolitinib with Treg showed a higher efficacy as compared to each single treatment alone in our GvHD mouse model in terms of GvHD incidence, severity and survival without hampering graft versus leukemia effect. This beneficial effect correlated with the detection in the bone marrow of recipient mice of the infused donor allogeneic Treg after the adoptive transfer.
Collapse
|
7
|
Nakagawa Y, Egawa G, Miyake T, Nakajima S, Otsuka A, Nomura T, Kitoh A, Dainichi T, Sakabe JI, Shibaki A, Tokura Y, Honda T, Kabashima K. A phenotypic analysis of involucrin-mOVA mice following adoptive transfer of OVA-specific CD8+ T cells. JID INNOVATIONS 2022; 2:100127. [PMID: 36090298 PMCID: PMC9460514 DOI: 10.1016/j.xjidi.2022.100127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/03/2022] Open
Abstract
To investigate the mechanism of autoimmunity and peripheral tolerance in the skin, several transgenic mouse strains expressing membrane-bound ovalbumin (mOVA) as an epidermal self-antigen under the control of keratinocyte-specific promotors, such as keratin 5 and keratin 14, were employed in combination with adoptive transfer of CD8+ T cells from OT-I mice (OT-I T cells) that recognize an ovalbumin-derived peptide. However, these strains showed bodyweight loss and required additional inflammatory stimuli, such as γ-irradiation and tape-stripping, to induce skin inflammation. In this study, we generated a mouse strain expressing mOVA under the control of human involucrin promoter (involucrin-mOVA mice). In contrast to previous strains, involucrin-mOVA mice spontaneously developed skin inflammation after the transfer of OT-I T cells in the absence of external stimuli without significant bodyweight loss. We focused on the skin infiltration process of OT-I T cells and found that transferred OT-I T cells accumulated around the hair follicles in the early phase of skin inflammation, and in the later phase, the skin inflammation spontaneously resolved despite the remaining OT-I T cells in the skin. Our involucrin-mOVA mice will provide a promising tool to investigate the pathogenesis and the tolerance mechanisms of cytotoxic skin autoimmunity.
Collapse
|
8
|
Namdari H, Hosseini M, Yazdanifar M, Farajifard H, Parvizpour F, Karamigolbaghi M, Hamidieh AA, Rezaei F. Protective and pathological roles of regulatory immune cells in human cytomegalovirus infection following hematopoietic stem cell transplantation. Rev Med Virol 2021; 32:e2319. [PMID: 34914147 DOI: 10.1002/rmv.2319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/11/2022]
Abstract
Human cytomegalovirus (HCMV) is ubiquitously prevalent. Immune system in healthy individuals is capable of controlling HCMV infection; however, HCMV can be life-threatening for immunocompromised individuals, such as transplant recipients. Both innate and adaptive immune systems are critically involved in the HCMV infection. Recent studies have indicated that regulatory immune cells which play essential roles in maintaining a healthy immune environment are closely related to immune response in HCMV infection. However, the exact role of regulatory immune cells in immune regulation and homoeostasis during the battle between HCMV and host still requires further research. In this review, we highlight the protective and pathological roles of regulatory immune cells in HCMV infection following hematopoietic stem cell transplantation (HSCT).
Collapse
Affiliation(s)
- Haideh Namdari
- Iranian Tissue Bank and Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Hosseini
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Yazdanifar
- Department of Pediatrics, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Hamid Farajifard
- Iranian Tissue Bank and Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Parvizpour
- Iranian Tissue Bank and Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Karamigolbaghi
- Iranian Tissue Bank and Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Slepicka PF, Yazdanifar M, Bertaina A. Harnessing Mechanisms of Immune Tolerance to Improve Outcomes in Solid Organ Transplantation: A Review. Front Immunol 2021; 12:688460. [PMID: 34177941 PMCID: PMC8222735 DOI: 10.3389/fimmu.2021.688460] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
Survival after solid organ transplantation (SOT) is limited by chronic rejection as well as the need for lifelong immunosuppression and its associated toxicities. Several preclinical and clinical studies have tested methods designed to induce transplantation tolerance without lifelong immune suppression. The limited success of these strategies has led to the development of clinical protocols that combine SOT with other approaches, such as allogeneic hematopoietic stem cell transplantation (HSCT). HSCT prior to SOT facilitates engraftment of donor cells that can drive immune tolerance. Recent innovations in graft manipulation strategies and post-HSCT immune therapy provide further advances in promoting tolerance and improving clinical outcomes. In this review, we discuss conventional and unconventional immunological mechanisms underlying the development of immune tolerance in SOT recipients and how they can inform clinical advances. Specifically, we review the most recent mechanistic studies elucidating which immune regulatory cells dampen cytotoxic immune reactivity while fostering a tolerogenic environment. We further discuss how this understanding of regulatory cells can shape graft engineering and other therapeutic strategies to improve long-term outcomes for patients receiving HSCT and SOT.
Collapse
Affiliation(s)
- Priscila Ferreira Slepicka
- Division of Hematology, Oncology and Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Mahboubeh Yazdanifar
- Division of Hematology, Oncology and Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Alice Bertaina
- Division of Hematology, Oncology and Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
10
|
Volfson-Sedletsky V, Jones A, Hernandez-Escalante J, Dooms H. Emerging Therapeutic Strategies to Restore Regulatory T Cell Control of Islet Autoimmunity in Type 1 Diabetes. Front Immunol 2021; 12:635767. [PMID: 33815387 PMCID: PMC8015774 DOI: 10.3389/fimmu.2021.635767] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Despite many decades of investigation uncovering the autoimmune mechanisms underlying Type 1 Diabetes (T1D), translating these findings into effective therapeutics has proven extremely challenging. T1D is caused by autoreactive T cells that become inappropriately activated and kill the β cells in the pancreas, resulting in insulin insufficiency and hyperglycemia. A large body of evidence supports the idea that the unchecked activation and expansion of autoreactive T cells in T1D is due to defects in immunosuppressive regulatory T cells (Tregs) that are critical for maintaining peripheral tolerance to islet autoantigens. Hence, repairing these Treg deficiencies is a much sought-after strategy to treat the disease. To accomplish this goal in the most precise, effective and safest way possible, restored Treg functions will need to be targeted towards suppressing the autoantigen-specific immune responses only and/or be localized in the pancreas. Here we review the most recent developments in designing Treg therapies that go beyond broad activation or expansion of non-specific polyclonal Treg populations. We focus on two cutting-edge strategies namely ex vivo generation of optimized Tregs for re-introduction in T1D patients vs direct in situ stimulation and restoration of endogenous Treg function.
Collapse
Affiliation(s)
- Victoria Volfson-Sedletsky
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Albert Jones
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Jaileene Hernandez-Escalante
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Hans Dooms
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
11
|
Motwani K, Peters LD, Vliegen WH, El-sayed AG, Seay HR, Lopez MC, Baker HV, Posgai AL, Brusko MA, Perry DJ, Bacher R, Larkin J, Haller MJ, Brusko TM. Human Regulatory T Cells From Umbilical Cord Blood Display Increased Repertoire Diversity and Lineage Stability Relative to Adult Peripheral Blood. Front Immunol 2020; 11:611. [PMID: 32351504 PMCID: PMC7174770 DOI: 10.3389/fimmu.2020.00611] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 03/17/2020] [Indexed: 12/22/2022] Open
Abstract
The human T lymphocyte compartment is highly dynamic over the course of a lifetime. Of the many changes, perhaps most notable is the transition from a predominantly naïve T cell state at birth to the acquisition of antigen-experienced memory and effector subsets following environmental exposures. These phenotypic changes, including the induction of T cell exhaustion and senescence, have the potential to negatively impact efficacy of adoptive T cell therapies (ACT). When considering ACT with CD4+CD25+CD127-/lo regulatory T cells (Tregs) for the induction of immune tolerance, we previously reported ex vivo expanded umbilical cord blood (CB) Tregs remained more naïve, suppressed responder T cells equivalently, and exhibited a more diverse T cell receptor (TCR) repertoire compared to expanded adult peripheral blood (APB) Tregs. Herein, we hypothesized that upon further characterization, we would observe increased lineage heterogeneity and phenotypic diversity in APB Tregs that might negatively impact lineage stability, engraftment capacity, and the potential for Tregs to home to sites of tissue inflammation following ACT. We compared the phenotypic profiles of human Tregs isolated from CB versus the more traditional source, APB. We conducted analysis of fresh and ex vivo expanded Treg subsets at both the single cell (scRNA-seq and flow cytometry) and bulk (microarray and cytokine profiling) levels. Single cell transcriptional profiles of pre-expansion APB Tregs highlighted a cluster of cells that showed increased expression of genes associated with effector and pro-inflammatory phenotypes (CCL5, GZMK, CXCR3, LYAR, and NKG7) with low expression of Treg markers (FOXP3 and IKZF2). CB Tregs were more diverse in TCR repertoire and homogenous in phenotype, and contained fewer effector-like cells in contrast with APB Tregs. Interestingly, expression of canonical Treg markers, such as FOXP3, TIGIT, and IKZF2, were increased in CB CD4+CD127+ conventional T cells (Tconv) compared to APB Tconv, post-expansion, implying perinatal T cells may adopt a default regulatory program. Collectively, these data identify surface markers (namely CXCR3) that could be depleted to improve purity and stability of APB Tregs, and support the use of expanded CB Tregs as a potentially optimal ACT modality for the treatment of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Keshav Motwani
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Leeana D. Peters
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Willem H. Vliegen
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Ahmed Gomaa El-sayed
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Howard R. Seay
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - M. Cecilia Lopez
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Henry V. Baker
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Maigan A. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Daniel J. Perry
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Joseph Larkin
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Michael J. Haller
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
12
|
Court AC, Le-Gatt A, Luz-Crawford P, Parra E, Aliaga-Tobar V, Bátiz LF, Contreras RA, Ortúzar MI, Kurte M, Elizondo-Vega R, Maracaja-Coutinho V, Pino-Lagos K, Figueroa FE, Khoury M. Mitochondrial transfer from MSCs to T cells induces Treg differentiation and restricts inflammatory response. EMBO Rep 2020; 21:e48052. [PMID: 31984629 DOI: 10.15252/embr.201948052] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 11/11/2019] [Accepted: 11/29/2019] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have fueled ample translation for the treatment of immune-mediated diseases. They exert immunoregulatory and tissue-restoring effects. MSC-mediated transfer of mitochondria (MitoT) has been demonstrated to rescue target organs from tissue damage, yet the mechanism remains to be fully resolved. Therefore, we explored the effect of MitoT on lymphoid cells. Here, we describe dose-dependent MitoT from mitochondria-labeled MSCs mainly to CD4+ T cells, rather than CD8+ T cells or CD19+ B cells. Artificial transfer of isolated MSC-derived mitochondria increases the expression of mRNA transcripts involved in T-cell activation and T regulatory cell differentiation including FOXP3, IL2RA, CTLA4, and TGFβ1, leading to an increase in a highly suppressive CD25+ FoxP3+ population. In a GVHD mouse model, transplantation of MitoT-induced human T cells leads to significant improvement in survival and reduction in tissue damage and organ T CD4+ , CD8+ , and IFN-γ+ expressing cell infiltration. These findings point to a unique CD4+ T-cell reprogramming mechanism with pre-clinical proof-of-concept data that pave the way for the exploration of organelle-based therapies in immune diseases.
Collapse
Affiliation(s)
- Angela C Court
- Cells for Cells, Santiago, Chile.,Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | | | - Patricia Luz-Crawford
- Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Eliseo Parra
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Victor Aliaga-Tobar
- Centro de Modelamiento Molecular, Biofísica y Bioinformática (CM2B2), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases - ACCDiS, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Luis Federico Bátiz
- Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Rafael A Contreras
- Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | | | - Mónica Kurte
- Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Roberto Elizondo-Vega
- Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Vinicius Maracaja-Coutinho
- Centro de Modelamiento Molecular, Biofísica y Bioinformática (CM2B2), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases - ACCDiS, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Karina Pino-Lagos
- Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Fernando E Figueroa
- Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Maroun Khoury
- Cells for Cells, Santiago, Chile.,Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| |
Collapse
|
13
|
Li R, Johnson R, Yu G, McKenna DH, Hubel A. Preservation of cell-based immunotherapies for clinical trials. Cytotherapy 2019; 21:943-957. [PMID: 31416704 PMCID: PMC6746578 DOI: 10.1016/j.jcyt.2019.07.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/22/2019] [Indexed: 12/29/2022]
Abstract
In the unique supply chain of cellular therapies, preservation is important to keep the cell product viable. Many factors in cryopreservation affect the outcome of a cell therapy: (i) formulation and introduction of a freezing medium, (ii) cooling rate, (iii) storage conditions, (iv) thawing conditions and (v) post-thaw processing. This article surveys clinical trials of cellular immunotherapy that used cryopreserved regulatory, chimeric antigen receptor or gamma delta T cells, dendritic cells or natural killer (NK) cells. Several observations are summarized from the given information. The aforementioned cell types have been similarly frozen in media containing 5-10% dimethyl sulfoxide (DMSO) with plasma, serum or human serum albumin. Two common freezing methods are an insulated freezing container such as Nalgene Mr. Frosty and a controlled-rate freezer at a cooling rate of -1°C/min. Water baths at approximately 37°C have been commonly used for thawing. Post-thaw processing of cryopreserved cells varied greatly: some studies infused the cells immediately upon thawing; some diluted the cells in a carrier solution of varying formulation before infusion; some washed cells to remove cryoprotective agents; and others re-cultured cells to recover cell viability or functionality lost due to cryopreservation. Emerging approaches to preserving cellular immunotherapies are also described. DMSO-free formulations of the freezing media have demonstrated improved preservation of cell viability in T lymphocytes and of cytotoxic function in natural killer cells. Saccharides are a common type of molecule used as an alternative cryoprotective agent to DMSO. Improving methods of preservation will be critical to growth in the clinical use of cellular immunotherapies.
Collapse
Affiliation(s)
- Rui Li
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rachel Johnson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Guanglin Yu
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - David H McKenna
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Allison Hubel
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
14
|
Meyer EH, Laport G, Xie BJ, MacDonald K, Heydari K, Sahaf B, Tang SW, Baker J, Armstrong R, Tate K, Tadisco C, Arai S, Johnston L, Lowsky R, Muffly L, Rezvani AR, Shizuru J, Weng WK, Sheehan K, Miklos D, Negrin RS. Transplantation of donor grafts with defined ratio of conventional and regulatory T cells in HLA-matched recipients. JCI Insight 2019; 4:127244. [PMID: 31092732 DOI: 10.1172/jci.insight.127244] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUNDIn preclinical murine and early clinical studies of hematopoietic cell transplantation, engineering of donor grafts with defined ratios of CD4+CD25+FoxP3+ Tregs to conventional T cells (Tcons) results in the prevention of graft-versus-host disease and improved immune reconstitution. The use of highly purified primary graft Tregs for direct cell infusion has potential advantages over impure immunomagnetic selection or culture expansion, but has not been tested clinically. We performed a phase I study of the timed addition of CD34-selected hematopoietic stem cells and Tregs, followed by Tcons for the treatment of patients with high-risk hematological malignancies.METHODSWe present interim evaluation of a single-center open phase I/II study of administration of human leukocyte-matched Tregs and CD34-selected hematopoietic cells, followed by infusion of an equal ratio of Tcons in adult patients undergoing myeloablative hematopoietic stem cell transplantation (HCT) for high-risk or active hematological malignancies. Tregs were purified by immunomagnetic selection and high-speed cell sorting.RESULTSHere we report results for the first 12 patients who received Tregs of between 91% and 96% purity. Greater than grade II GVHD was noted in 2 patients in the first cohort of 5 patients, who received cryopreserved Tregs, but neither acute nor chronic GVHD was noted in the second cohort of 7 patients, who received fresh Tregs and single-agent GVHD prophylaxis. Patients in the second cohort appeared to have normal immune reconstitution compared with patients who underwent transplantation and did not develop GVHD.CONCLUSIONOur study shows that the use of highly purified fresh Tregs is clinically feasible and supports continued investigation of the strategy.TRIAL REGISTRATIONClinicalTrials.gov NCT01660607.FUNDINGNIH NHBLI R01 HL114591 and K08HL119590.
Collapse
Affiliation(s)
- Everett H Meyer
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA.,Cell Therapy Facility, Stanford Health Care, Stanford, California, USA
| | - Ginna Laport
- Tempest Therapeutics, San Francisco, California, USA
| | - Bryan J Xie
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Kate MacDonald
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Kartoosh Heydari
- Cell Therapy Facility, Stanford Health Care, Stanford, California, USA
| | - Bita Sahaf
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Sai-Wen Tang
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Randall Armstrong
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Keri Tate
- Laboratory for Cell and Gene Medicine, Stanford University, Palo Alto, California, USA
| | - Cynthia Tadisco
- Laboratory for Cell and Gene Medicine, Stanford University, Palo Alto, California, USA
| | - Sally Arai
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Laura Johnston
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Robert Lowsky
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Lori Muffly
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Andrew R Rezvani
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Judith Shizuru
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Kevin Sheehan
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - David Miklos
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| |
Collapse
|
15
|
Abstract
Cellular therapies, including those based on T cells, are becoming approved options for clinicians treating a range of diseases. Cytotoxic T lymphocytes (CTLs) can be modified ex vivo to express receptors such as chimeric antigen receptors (CARs) or T cell receptors, allowing them to target tumour cells when infused back into patients with particular cancers. CTLs specific for viruses can be purified ex vivo and reinfused into patients transplanted with haematopoietic stem cells to help combat viral reactivation. Regulatory T cells (Tregs) can be expanded ex vivo for infusion into patients with autoimmunity or allergy, or into those at risk of rejecting transplanted cells or tissues, or suffering graft versus host disease. Effector and regulatory T cells can also be generated by infusion of patient-derived dendritic cells (DCs) conditioned in ways to elicit anti-tumour immunity (CTLs) or Tregs. All such therapies are resource-heavy (particularly in process regulation) and so must be initially targeted to patients that have limited treatment options, but also where they have a chance of being effective.
Collapse
|
16
|
Gandelman JS, Song DJ, Chen H, Engelhardt BG, Chen YB, Clark WB, Giver CR, Waller EK, Jung DK, Jagasia M. A Prospective Trial of Extracorporeal Photopheresis for Chronic Graft-versus-Host Disease Reveals Significant Disease Response and No Association with Frequency of Regulatory T Cells. Biol Blood Marrow Transplant 2018; 24:2373-2380. [DOI: 10.1016/j.bbmt.2018.06.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
|
17
|
Ma Y, Wang Z, Zhang A, Xu F, Zhao N, Xue J, Zhang H, Luan X. Human placenta-derived mesenchymal stem cells ameliorate GVHD by modulating Th17/Tr1 balance via expression of PD-L2. Life Sci 2018; 214:98-105. [PMID: 30393022 DOI: 10.1016/j.lfs.2018.10.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/19/2018] [Accepted: 10/26/2018] [Indexed: 10/28/2022]
Abstract
AIMS To examine whether human placenta mesenchymal stem/stromal cells (hpMSCs) mitigate graft-versus-host-disease (GVHD) via regulation of Th17 and Tr1. MATERIALS AND METHODS hpMSCs or phosphate buffered saline (PBS, as a control) were injected into humanized xeno-GVHD NOD/SCID mouse model. Effects on body weights and survival times were determined. In addition, various assays, including flow cytometry (FCM) and HE stain, were performed on tissues (liver, spleen, lung and intestine) from these hpMSCs versus PBS treated GVHD mice. Th17 cell number in vitro was analyzed by FCM. KEY FINDINGS hpMSCs reduced weight loss, along with IL-6 and IL-17 production to prolong the survival of GVHD mice. Th17 cell number was down-regulated obviously in hpMSCs treated GVHD mice. Conversely, Tr1 cell number and TGF-β production were enhanced by hpMSCs. Moreover, knockdown of programmed death ligand 2 (PD-L2) increased Th17 cell number from PMA activated T cells co-cultured with hpMSCs. SIGNIFICANCE hpMSCs can modulate the balance between Th17 and Tr1 cells to alleviate GVHD. In addition, PD-L2 as expressed on hpMSCs inhibits the generation of Th17 subset from activated T cells. These data suggest that hpMSCs attenuate GVHD through inhibition of severe inflammatory responses resulting from T cell differentiation.
Collapse
Affiliation(s)
- Yanchao Ma
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, Province, 264003, People's Republic of China
| | - Zhuoya Wang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, Province, 264003, People's Republic of China
| | - Aiping Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, Province, 264003, People's Republic of China
| | - Fenghuang Xu
- Urology Department, The first affiliated hospital of Hainan Medical University, Haikou, Hainan Province 570102, People's Republic of China
| | - Nannan Zhao
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, Province, 264003, People's Republic of China
| | - Jiangnan Xue
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, Province, 264003, People's Republic of China
| | - Hongqin Zhang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong Province 264003, People's Republic of China.
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, Province, 264003, People's Republic of China.
| |
Collapse
|
18
|
Perez-Basterrechea M, Esteban MM, Vega JA, Obaya AJ. Tissue-engineering approaches in pancreatic islet transplantation. Biotechnol Bioeng 2018; 115:3009-3029. [PMID: 30144310 DOI: 10.1002/bit.26821] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
Abstract
Pancreatic islet transplantation is a promising alternative to whole-pancreas transplantation as a treatment of type 1 diabetes mellitus. This technique has been extensively developed during the past few years, with the main purpose of minimizing the complications arising from the standard protocols used in organ transplantation. By using a variety of strategies used in tissue engineering and regenerative medicine, pancreatic islets have been successfully introduced in host patients with different outcomes in terms of islet survival and functionality, as well as the desired normoglycemic control. Here, we describe and discuss those strategies to transplant islets together with different scaffolds, in combination with various cell types and diffusible factors, and always with the aim of reducing host immune response and achieving islet survival, regardless of the site of transplantation.
Collapse
Affiliation(s)
- Marcos Perez-Basterrechea
- Unidad de Terapia Celular y Medicina Regenerativa, Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain.,Plataforma de Terapias Avanzadas, Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Manuel M Esteban
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | - Jose A Vega
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Alvaro J Obaya
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW The immunosuppressive agent cyclosporine was first reported to lower daily insulin dose and improve glycemic control in patients with new-onset type 1 diabetes (T1D) in 1984. While renal toxicity limited cyclosporine's extended use, this observation ignited collaborative efforts to identify immunotherapeutic agents capable of safely preserving β cells in patients with or at risk for T1D. RECENT FINDINGS Advances in T1D prediction and early diagnosis, together with expanded knowledge of the disease mechanisms, have facilitated trials targeting specific immune cell subsets, autoantigens, and pathways. In addition, clinical responder and non-responder subsets have been defined through the use of metabolic and immunological readouts. Herein, we review emerging T1D biomarkers within the context of recent and ongoing T1D immunotherapy trials. We also discuss responder/non-responder analyses in an effort to identify therapeutic mechanisms, define actionable pathways, and guide subject selection, drug dosing, and tailored combination drug therapy for future T1D trials.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Brittney N Newby
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Michael J Haller
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA.
| |
Collapse
|