1
|
Kruchen A, Fehse B, Müller I. Clinical relevance of feto-maternal microchimerism in (hematopoietic stem cell) transplantation. Semin Immunopathol 2024; 47:4. [PMID: 39644358 PMCID: PMC11625077 DOI: 10.1007/s00281-024-01028-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 11/14/2024] [Indexed: 12/09/2024]
Abstract
Toleration of a semi-allogeneic fetus in the mother's uterus as well as tolerance after allogeneic hematopoietic stem cell transplantation (HSCT) appear to share some immunologic concepts. The existence of microchimeric cells, and the original idea of a bidirectional cell trafficking between mother and child during pregnancy have been known for decades. Today, origins and mechanisms of persistence of microchimeric cells are intensively being elucidated. Both, the translation of the phenomenon of feto-maternal immune tolerance to donor choice or prevention of graft-versus-host disease (GvHD) in HSCT, and the implications of microchimeric cells in and for HSCT are highly intriguing. Yet, differences in detection methods of microchimeric cells, as well as in transplantation protocols impede the comparison of larger cohorts, and limit potential clinical advice. Still, matching of non-inherited maternal antigens (NIMA), which are expressed on maternal microchimeric cells, demonstrated a strong association with decreased risk for the development of acute GvHD in the context of various transplantation strategies. Despite the fact that advances in graft manipulation and immunosuppression ameliorated the safety and outcome after HSCT, NIMA-matching retained a beneficial role in selection of sibling, child, or maternal donors, as well as for cord blood units. Recent findings indicate the existence of a microchimeric stem cell niche, in which only one dominant microchimeric cell population of only one semi-allogeneic origin persists at a time. This implies that studies regarding the impact of (maternal and fetal) microchimerism (MC) on clinical outcome of HSCT should combine analysis of NIMA and direct detection of microchimeric cells from donor and recipient on the verge of HSCT to be efficiently conclusive.
Collapse
Affiliation(s)
- Anne Kruchen
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- German Center for Child and Adolescent Health (DZKJ), Hamburg, Germany.
| | - Ingo Müller
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, 20246, Hamburg, Germany
- German Center for Child and Adolescent Health (DZKJ), Hamburg, Germany
| |
Collapse
|
2
|
Grant MT, Nelvagal HR, Tecos M, Hamed A, Swanson K, Cooper JD, Vrecenak JD. Cellular trafficking and fate mapping of cells within the nervous system after in utero hematopoietic cell transplantation. Commun Biol 2024; 7:1624. [PMID: 39638879 PMCID: PMC11621337 DOI: 10.1038/s42003-024-06847-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 09/05/2024] [Indexed: 12/07/2024] Open
Abstract
In utero hematopoietic cell transplantation (IUHCT) utilizes fetal immune tolerance to achieve durable chimerism without conditioning or immunosuppression during a unique window in fetal development. Though donor cells have been observed within the nervous system following in utero injection, the timeline and distribution of cellular trafficking across the blood-brain barrier following IUHCT is not well understood. We injected 20 × 106 adult bone marrow mononuclear cells intravenously at gestational age (GA) 12-17 days and found that donor cells were maximally concentrated in the brain with treatment between GA 13-14. Donor cell engraftment persisted within the brain at every timepoint analyzed and concentrated within the hindbrain with significantly more grafted cells than in the forebrain. Additionally, transplanted cells terminally differentiated into various nervous system cellular morphologies and also populated the enteric nervous system. This study is the first to document the timeline and distribution of donor cell trafficking into the immune-protected nervous system and serves as a foundation for the application of IUHCT to treat neurogenetic diseases.
Collapse
Affiliation(s)
- Matthew T Grant
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Hemanth Ramesh Nelvagal
- Washington University in St. Louis School of Medicine, Department of Pediatrics, Division of Genetics and Genomics, St. Louis, MO, USA
- University College London, School of Pharmacy, Department of Pharmacology, London, UK
| | - Maria Tecos
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Amal Hamed
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Kerry Swanson
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Jonathan D Cooper
- Washington University in St. Louis School of Medicine, Department of Pediatrics, Division of Genetics and Genomics, St. Louis, MO, USA
| | - Jesse D Vrecenak
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA.
| |
Collapse
|
3
|
Riley JS, Berkowitz CL, Luks VL, Dave A, Cyril-Olutayo MC, Pogoriler J, Flake AW, Abdulmalik O, Peranteau WH. Immune modulation permits tolerance and engraftment in a murine model of late-gestation transplantation. Blood Adv 2024; 8:4523-4538. [PMID: 38941538 PMCID: PMC11395771 DOI: 10.1182/bloodadvances.2023012247] [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: 11/21/2023] [Revised: 05/08/2024] [Accepted: 06/15/2024] [Indexed: 06/30/2024] Open
Abstract
ABSTRACT In utero hematopoietic cell transplantation is an experimental nonmyeloablative therapy with potential applications in hematologic disorders, including sickle cell disease (SCD). Its clinical utility has been limited due to the early acquisition of T-cell immunity beginning at ∼14 weeks gestation, posing significant technical challenges and excluding treatment fetuses evaluated after the first trimester. Using murine neonatal transplantation at 20 days postcoitum (DPC) as a model for late-gestation transplantation (LGT) in humans, we investigated whether immune modulation with anti-CD3 monoclonal antibody (mAb) could achieve donor-specific tolerance and sustained allogeneic engraftment comparable with that of the early-gestation fetal recipient at 14 DPC. In allogeneic wild-type strain combinations, administration of anti-CD3 mAb with transplantation resulted in transient T-cell depletion followed by central tolerance induction confirmed by donor-specific clonal deletion and skin graft tolerance. Normal immune responses to third-party major histocompatibility complex and viral pathogens were preserved, and graft-versus-host disease did not occur. We further demonstrated the successful application of this approach in the Townes mouse model of SCD. These findings confirm the developing fetal T-cell response as a barrier to LGT and support transient T-cell depletion as a safe and effective immunomodulatory strategy to overcome it.
Collapse
Affiliation(s)
- John S. Riley
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Cara L. Berkowitz
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Valerie L. Luks
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Apeksha Dave
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Mojisola C. Cyril-Olutayo
- Department of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Drug Research and Production Unit, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Jennifer Pogoriler
- Department of Pathology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Alan W. Flake
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Osheiza Abdulmalik
- Department of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - William H Peranteau
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
4
|
Zong Y, Deng K, Chong WP. Regulation of Treg cells by cytokine signaling and co-stimulatory molecules. Front Immunol 2024; 15:1387975. [PMID: 38807592 PMCID: PMC11131382 DOI: 10.3389/fimmu.2024.1387975] [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: 02/19/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
CD4+CD25+Foxp3+ regulatory T cells (Tregs), a vital component of the immune system, are responsible for maintaining immune homeostasis and preventing excessive immune responses. This review explores the signaling pathways of the cytokines that regulate Treg cells, including transforming growth factor beta (TGF-β), interleukin (IL)-2, IL-10, and IL-35, which foster the differentiation and enhance the immunosuppressive capabilities of Tregs. It also examines how, conversely, signals mediated by IL-6 and tumor necrosis factor -alpha (TNF-α) can undermine Treg suppressive functions or even drive their reprogramming into effector T cells. The B7 family comprises indispensable co-stimulators for T cell activation. Among its members, this review focuses on the capacity of CTLA-4 and PD-1 to regulate the differentiation, function, and survival of Tregs. As Tregs play an essential role in maintaining immune homeostasis, their dysfunction contributes to the pathogenesis of autoimmune diseases. This review delves into the potential of employing Treg-based immunotherapy for the treatment of autoimmune diseases, transplant rejection, and cancer. By shedding light on these topics, this article aims to enhance our understanding of the regulation of Tregs by cytokines and their therapeutic potential for various pathological conditions.
Collapse
Affiliation(s)
- Yuan Zong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, China
| | - Kaihang Deng
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Wai Po Chong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, China
| |
Collapse
|
5
|
Bader CS, Pavlova A, Lowsky R, Muffly LS, Shiraz P, Arai S, Johnston LJ, Rezvani AR, Weng WK, Miklos DB, Frank MJ, Tamaresis JS, Agrawal V, Bharadwaj S, Sidana S, Shizuru JA, Fernhoff NB, Putnam A, Killian S, Xie BJ, Negrin RS, Meyer EH. Single-center randomized trial of T-reg graft alone vs T-reg graft plus tacrolimus for the prevention of acute GVHD. Blood Adv 2024; 8:1105-1115. [PMID: 38091578 PMCID: PMC10907400 DOI: 10.1182/bloodadvances.2023011625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/27/2023] [Indexed: 02/29/2024] Open
Abstract
ABSTRACT Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for hematological malignancies for which graft-versus-host disease (GVHD) remains a major complication. The use of donor T-regulatory cells (Tregs) to prevent GVHD appears promising, including in our previous evaluation of an engineered graft product (T-reg graft) consisting of the timed, sequential infusion of CD34+ hematopoietic stem cells and high-purity Tregs followed by conventional T cells. However, whether immunosuppressive prophylaxis can be removed from this protocol remains unclear. We report the results of the first stage of an open-label single-center phase 2 study (NCT01660607) investigating T-reg graft in myeloablative HCT of HLA-matched and 9/10-matched recipients. Twenty-four patients were randomized to receive T-reg graft alone (n = 12) or T-reg graft plus single-agent GVHD prophylaxis (n = 12) to determine whether T-reg graft alone was noninferior in preventing acute GVHD. All patients developed full-donor myeloid chimerism. Patients with T-reg graft alone vs with prophylaxis had incidences of grade 3 to 4 acute GVHD of 58% vs 8% (P = .005) and grade 3 to 4 of 17% vs 0% (P = .149), respectively. The incidence of moderate-to-severe chronic GVHD was 28% in the T-reg graft alone arm vs 0% with prophylaxis (P = .056). Among patients with T-reg graft and prophylaxis, CD4+ T-cell-to-Treg ratios were reduced after transplantation, gene expression profiles showed reduced CD4+ proliferation, and the achievement of full-donor T-cell chimerism was delayed. This study indicates that T-reg graft with single-agent tacrolimus is preferred over T-reg graft alone for the prevention of acute GVHD. This trial was registered at www.clinicaltrials.gov as #NCT01660607.
Collapse
Affiliation(s)
- Cameron S. Bader
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Anna Pavlova
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Robert Lowsky
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| | - Lori S. Muffly
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Parveen Shiraz
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Sally Arai
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| | - Laura J. Johnston
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Andrew R. Rezvani
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Wen-Kai Weng
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| | - David B. Miklos
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Matthew J. Frank
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | | | - Vaibhav Agrawal
- Department of Hematology and Hematopoietic Stem Cell Transplantation, City of Hope, Duarte, CA
| | - Sushma Bharadwaj
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Surbhi Sidana
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Judith A. Shizuru
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | | | | | | | | | - Robert S. Negrin
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| | - Everett H. Meyer
- Stanford Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
- Cellular Immune Tolerance Program, Stanford Department of Medicine, Stanford University, Stanford, CA
| |
Collapse
|
6
|
Palanki R, Bose SK, Dave A, White BM, Berkowitz C, Luks V, Yaqoob F, Han E, Swingle KL, Menon P, Hodgson E, Biswas A, Billingsley MM, Li L, Yiping F, Carpenter M, Trokhan A, Yeo J, Johana N, Wan TY, Alameh MG, Bennett FC, Storm PB, Jain R, Chan J, Weissman D, Mitchell MJ, Peranteau WH. Ionizable Lipid Nanoparticles for Therapeutic Base Editing of Congenital Brain Disease. ACS NANO 2023; 17:13594-13610. [PMID: 37458484 PMCID: PMC11025390 DOI: 10.1021/acsnano.3c02268] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Delivery of mRNA-based therapeutics to the perinatal brain holds great potential in treating congenital brain diseases. However, nonviral delivery platforms that facilitate nucleic acid delivery in this environment have yet to be rigorously studied. Here, we screen a diverse library of ionizable lipid nanoparticles (LNPs) via intracerebroventricular (ICV) injection in both fetal and neonatal mice and identify an LNP formulation with greater functional mRNA delivery in the perinatal brain than an FDA-approved industry standard LNP. Following in vitro optimization of the top-performing LNP (C3 LNP) for codelivery of an adenine base editing platform, we improve the biochemical phenotype of a lysosomal storage disease in the neonatal mouse brain, exhibit proof-of-principle mRNA brain transfection in vivo in a fetal nonhuman primate model, and demonstrate the translational potential of C3 LNPs ex vivo in human patient-derived brain tissues. These LNPs may provide a clinically translatable platform for in utero and postnatal mRNA therapies including gene editing in the brain.
Collapse
Affiliation(s)
- Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sourav K Bose
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Apeksha Dave
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brandon M. White
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Cara Berkowitz
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Valerie Luks
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pallavi Menon
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Emily Hodgson
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Arijit Biswas
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | | | - Li Li
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fan Yiping
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | - Marco Carpenter
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandra Trokhan
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Julie Yeo
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | | | - Tan Yi Wan
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | - Mohamad-Gabriel Alameh
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frederick Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phillip B. Storm
- Division of Neurosurgery, Children’s Hospital of Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jerry Chan
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore, 229899, SG
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - William H. Peranteau
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, PA, USA
| |
Collapse
|
7
|
Riley JS, McClain LE, Stratigis JD, Coons BE, Bose SK, Dave A, White BM, Li H, Loukogeorgakis SP, Fachin CG, Dias AIBS, Flake AW, Peranteau WH. Fetal allotransplant recipients are resistant to graft-versus-host disease. Exp Hematol 2023; 118:31-39.e3. [PMID: 36535408 PMCID: PMC9898145 DOI: 10.1016/j.exphem.2022.12.004] [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: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
In utero hematopoietic cell transplantation (IUHCT) is an experimental treatment for congenital hemoglobinopathies, including Sickle cell disease and thalassemias. One of the principal advantages of IUHCT is the predisposition of the developing fetus toward immunologic tolerance. This allows for engraftment across immune barriers without immunosuppression and, potentially, decreased susceptibility to graft-versus-host disease (GVHD). We demonstrate fetal resistance to GVHD following T cell-replete allogeneic hematopoietic cell transplantation compared with the neonate. We show that this resistance is associated with elevated fetal serum interleukin-10 conducive to the induction of regulatory T cells (Tregs). Finally, we demonstrate that the adoptive transfer of Tregs from IUHCT recipients to neonates uniformly prevents GVHD, recapitulating the predisposition to tolerance observed after fetal allotransplantation. These findings demonstrate fetal resistance to GVHD following hematopoietic cell transplantation and elucidate Tregs as important contributors.
Collapse
Affiliation(s)
- John S Riley
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Lauren E McClain
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - John D Stratigis
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Barbara E Coons
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Sourav K Bose
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Apeksha Dave
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Brandon M White
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Haiying Li
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Camila G Fachin
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Andre I B S Dias
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Alan W Flake
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - William H Peranteau
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA.
| |
Collapse
|
8
|
Fasoulakis Z, Koutras A, Ntounis T, Antsaklis P, Theodora M, Valsamaki A, Daskalakis G, Kontomanolis EN. Inflammatory Molecules Responsible for Length Shortening and Preterm Birth. Cells 2023; 12:cells12020209. [PMID: 36672145 PMCID: PMC9856720 DOI: 10.3390/cells12020209] [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: 11/13/2022] [Revised: 11/28/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
It is estimated that inflammation at the placental-maternal interface is directly responsible for or contributes to the development of 50% of all premature deliveries. Chorioamnionitis, also known as the premature rupture of the amniotic membrane in the mother, is the root cause of persistent inflammation that preterm newborns experience. Beyond contributing to the onset of early labor, inflammation is a critical element in advancing several conditions in neonates, including necrotizing enterocolitis, retinopathy of prematurity, bronchopulmonary dysplasia, intraventricular hemorrhage, retinopathy of prematurity and periventricular leukomalacia. Notably, the immune systems of preterm infants are not fully developed; immune defense mechanisms and immunosuppression (tolerance) have a delicate balance that is easily upset in this patient category. As a result, premature infants are exposed to different antigens from elements such as hospital-specific microbes, artificial devices, medications, food antigens and hypoxia/hyperoxia. This has detrimental implications for preterm deliveries of less than 28 weeks because they have not yet evolved the mechanisms to tolerate maternal and self-antigens.
Collapse
Affiliation(s)
- Zacharias Fasoulakis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, General Hospital Alexandra, 11528 Athens, Greece
- Correspondence:
| | - Antonios Koutras
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, General Hospital Alexandra, 11528 Athens, Greece
| | - Thomas Ntounis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, General Hospital Alexandra, 11528 Athens, Greece
| | - Panos Antsaklis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, General Hospital Alexandra, 11528 Athens, Greece
| | - Marianna Theodora
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, General Hospital Alexandra, 11528 Athens, Greece
| | - Asimina Valsamaki
- Department of Internal Medicine, Koutlimbaneio and Triantafylleio General Hospital of Larissa, 41221 Larissa, Greece
| | - George Daskalakis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, General Hospital Alexandra, 11528 Athens, Greece
| | - Emmanuel N. Kontomanolis
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
9
|
Shi C, Pan L, Hu Z. Experimental and clinical progress of in utero hematopoietic cell transplantation therapy for congenital disorders. Front Pharmacol 2022; 13:851375. [PMID: 36120324 PMCID: PMC9478511 DOI: 10.3389/fphar.2022.851375] [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: 01/09/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
In utero hematopoietic cell transplantation (IUHCT) is considered a potentially efficient therapeutic approach with relatively few side effects, compared to adult hematopoietic cell transplantation, for various hematological genetic disorders. The principle of IUHCT has been extensively studied in rodent models and in some large animals with close evolutionary similarities to human beings. However, IUHCT has only been used to rebuild human T cell immunity in certain patients with inherent immunodeficiencies. This review will first summarize the animal models utilized for IUHCT investigations and describe the associated outcomes. Recent advances and potential barriers for successful IUHCT are discussed, followed by possible strategies to overcome these barriers experimentally. Lastly, we will outline the progress made towards utilizing IUHCT to treat inherent disorders for patients, list out associated limitations and propose feasible means to promote the efficacy of IUHCT clinically.
Collapse
Affiliation(s)
- Chunyu Shi
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lu Pan
- Department of Pediatric Immunology, Allergy and Rheumatology, The First Hospital of Jilin University, Changchun, China
| | - Zheng Hu
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Zheng Hu,
| |
Collapse
|
10
|
de Coppi P, Loukogeorgakis S, Götherström C, David AL, Almeida-Porada G, Chan JKY, Deprest J, Wong KKY, Tam PKH. Regenerative medicine: prenatal approaches. THE LANCET. CHILD & ADOLESCENT HEALTH 2022; 6:643-653. [PMID: 35963269 PMCID: PMC10664288 DOI: 10.1016/s2352-4642(22)00192-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 10/15/2022]
Abstract
This two-paper Series focuses on recent advances and applications of regenerative medicine that could benefit paediatric patients. Innovations in genomic, stem-cell, and tissue-based technologies have created progress in disease modelling and new therapies for congenital and incurable paediatric diseases. Prenatal approaches present unique opportunities associated with substantial biotechnical, medical, and ethical obstacles. Maternal plasma fetal DNA analysis is increasingly adopted as a noninvasive prenatal screening or diagnostic test for chromosomal and monogenic disorders. The molecular basis for cell-free DNA detection stimulated the development of circulating tumour DNA testing for adult cancers. In-utero stem-cell, gene, gene-modified cell (and to a lesser extent, tissue-based) therapies have shown early clinical promise in a wide range of paediatric disorders. Fetal cells for postnatal treatment and artificial placenta for ex-utero fetal therapies are new frontiers in this exciting field.
Collapse
Affiliation(s)
- Paolo de Coppi
- Stem Cell and Regenerative Medicine Section, Department of Developmental Biology and Cancer Research and Teaching, Great Ormond Street Institute of Child Health, University College London, London, UK; Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Stavros Loukogeorgakis
- Stem Cell and Regenerative Medicine Section, Department of Developmental Biology and Cancer Research and Teaching, Great Ormond Street Institute of Child Health, University College London, London, UK; Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Cecilia Götherström
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Womens Health, University College London, London, UK
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem NC, USA
| | - Jerry K Y Chan
- Academic Clinical Program in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore; Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
| | - Jan Deprest
- Clinical Department of Obstetrics and Gynaecology, UZ Leuven, Leuven, Belgium
| | - Kenneth Kak Yuen Wong
- Division of Paediatric Surgery, Department of Surgery, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, Special Administrative Region, China
| | - Paul Kwong Hang Tam
- Division of Paediatric Surgery, Department of Surgery, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, Special Administrative Region, China; Faculty of Medicine, Macau University of Science and Technology, Macau Special Administrative Region, China.
| |
Collapse
|
11
|
White BM, Morrisey EE, Peranteau WH. In Utero Gene Editing for Inherited Lung Diseases. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-021-00205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
12
|
Kandasamy K, Tan LG, B Johana N, Tan YW, Foo W, Yeo JSL, Ravikumar V, Ginhoux F, Choolani M, Chan JKY, Mattar CNZ. Maternal microchimerism and cell-mediated immune-modulation enhance engraftment following semi-allogenic intrauterine transplantation. FASEB J 2021; 35:e21413. [PMID: 33570785 DOI: 10.1096/fj.202002185rr] [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: 10/06/2020] [Revised: 01/04/2021] [Accepted: 01/20/2021] [Indexed: 11/11/2022]
Abstract
Successful intrauterine hematopoietic cell transplantation (IUT) for congenital hemoglobinopathies is hampered by maternal alloresponsiveness. We investigate these interactions in semi-allogenic murine IUT. E14 fetuses (B6 females × BALB/c males) were each treated with 5E+6 maternal (B6) or paternal (BALB/c) bone marrow cells and serially monitored for chimerism (>1% engraftment), trafficked maternal immune cells, and immune responsiveness to donor cells. A total of 41.0% of maternal IUT recipients (mIUT) were chimeras (mean donor chimerism 3.0 ± 1.3%) versus 75.0% of paternal IUT recipients (pIUT, 3.6 ± 1.1%). Chimeras showed higher maternal microchimerism of CD4, CD8, and CD19 than non-chimeras. These maternal cells showed minimal responsiveness to B6 or BALB/c stimulation. To interrogate tolerance, mIUT were injected postnatally with 5E+6 B6 cells/pup; pIUT received BALB/c cells. IUT-treated pups showed no changes in trafficked maternal or fetal immune cell levels compared to controls. Donor-specific IgM and IgG were expressed by 1%-3% of recipients. mIUT splenocytes showed greater proliferation of regulatory T cells (Treg) upon BALB/c stimulation, while B6 stimulation upregulated the pro-inflammatory cytokines more than BALB/c. pIUT splenocytes produced identical Treg and cytokine responses to BALB/c and B6 cells, with higher Treg activity and lower pro-inflammatory cytokine expression upon exposure to BALB/c. In contrast, naïve fetal splenocytes demonstrated greater alloresponsiveness to BALB/c compared to B6 cells. Thus pIUT, associated with increased maternal cell trafficking, modulates fetal Treg, and cytokine responsiveness to donor cells more efficiently than mIUT, resulting in improved engraftment. Paternal donor cells may be considered alternatively to maternal donor cells for intrauterine and postnatal transplantation to induce tolerance and maintain engraftment.
Collapse
Affiliation(s)
- Karthikeyan Kandasamy
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lay Geok Tan
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Obstetrics and Gynaecology, National University Hospital, National University Health System, Singapore, Singapore
| | - Nuryanti B Johana
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yi Wan Tan
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Wanling Foo
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Julie S L Yeo
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Vikashini Ravikumar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mahesh Choolani
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Obstetrics and Gynaecology, National University Hospital, National University Health System, Singapore, Singapore
| | - Jerry K Y Chan
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Citra N Z Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Obstetrics and Gynaecology, National University Hospital, National University Health System, Singapore, Singapore
| |
Collapse
|
13
|
Sayitoglu EC, Freeborn RA, Roncarolo MG. The Yin and Yang of Type 1 Regulatory T Cells: From Discovery to Clinical Application. Front Immunol 2021; 12:693105. [PMID: 34177953 PMCID: PMC8222711 DOI: 10.3389/fimmu.2021.693105] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022] Open
Abstract
Regulatory T cells are essential players of peripheral tolerance and suppression of inflammatory immune responses. Type 1 regulatory T (Tr1) cells are FoxP3- regulatory T cells induced in the periphery under tolerogenic conditions. Tr1 cells are identified as LAG3+CD49b+ mature CD4+ T cells that promote peripheral tolerance through secretion of IL-10 and TGF-β in addition to exerting perforin- and granzyme B-mediated cytotoxicity against myeloid cells. After the initial challenges of isolation were overcome by surface marker identification, ex vivo expansion of antigen-specific Tr1 cells in the presence of tolerogenic dendritic cells (DCs) and IL-10 paved the way for their use in clinical trials. With one Tr1-enriched cell therapy product already in a Phase I clinical trial in the context of allogeneic hematopoietic stem cell transplantation (allo-HSCT), Tr1 cell therapy demonstrates promising results so far in terms of efficacy and safety. In the current review, we identify developments in phenotypic and molecular characterization of Tr1 cells and discuss the potential of engineered Tr1-like cells for clinical applications of Tr1 cell therapies. More than 3 decades after their initial discovery, Tr1 cell therapy is now being used to prevent graft versus host disease (GvHD) in allo-HSCT and will be an alternative to immunosuppression to promote graft tolerance in solid organ transplantation in the near future.
Collapse
Affiliation(s)
- Ece Canan Sayitoglu
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
| | - Robert Arthur Freeborn
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
| | - Maria Grazia Roncarolo
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, United States.,Center for Definitive and Curative Medicine (CDCM), Stanford School of Medicine, Stanford, CA, United States
| |
Collapse
|
14
|
Song Y, Wang N, Chen L, Fang L. Tr1 Cells as a Key Regulator for Maintaining Immune Homeostasis in Transplantation. Front Immunol 2021; 12:671579. [PMID: 33981317 PMCID: PMC8109434 DOI: 10.3389/fimmu.2021.671579] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/09/2021] [Indexed: 12/30/2022] Open
Abstract
The immune system is composed of effectors and regulators. Type 1 regulatory T (Tr1) cells are classified as a distinct subset of T cells, and they secret high levels of IL-10 but lack the expression of the forkhead box P3 (Foxp3). Tr1 cells act as key regulators in the immune network, and play a central role in maintaining immune homeostasis. The regulatory capacity of Tr1 cells depends on many mechanisms, including secretion of suppressive cytokines, cell-cell contacts, cytotoxicity and metabolic regulation. A breakdown of Tr1-cell-mediated tolerance is closely linked with the pathogenesis of various diseases. Based on this observation, Tr1-cell therapy has emerged as a successful treatment option for a number of human diseases. In this review, we describe an overview of Tr1 cell identification, functions and related molecular mechanisms. We also discuss the current protocols to induce/expand Tr1 cells in vitro for clinical application, and summarize the recent progress of Tr1 cells in transplantation.
Collapse
Affiliation(s)
- Yun Song
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Ning Wang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China.,Department of Immunology, Xi'an Medical University, Xi'an, China
| | - Lihua Chen
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Liang Fang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
15
|
Camacho V, Matkins VR, Patel SB, Lever JM, Yang Z, Ying L, Landuyt AE, Dean EC, George JF, Yang H, Ferrell PB, Maynard CL, Weaver CT, Turnquist HR, Welner RS. Bone marrow Tregs mediate stromal cell function and support hematopoiesis via IL-10. JCI Insight 2020; 5:135681. [PMID: 33208555 PMCID: PMC7710301 DOI: 10.1172/jci.insight.135681] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 10/07/2020] [Indexed: 12/31/2022] Open
Abstract
The nonimmune roles of Tregs have been described in various tissues, including the BM. In this study, we comprehensively phenotyped marrow Tregs, elucidating their key features and tissue-specific functions. We show that marrow Tregs are migratory and home back to the marrow. For trafficking, marrow Tregs use S1P gradients, and disruption of this axis allows for specific targeting of the marrow Treg pool. Following Treg depletion, the function and phenotype of both mesenchymal stromal cells (MSCs) and hematopoietic stem cells (HSCs) was impaired. Transplantation also revealed that a Treg-depleted niche has a reduced capacity to support hematopoiesis. Finally, we found that marrow Tregs are high producers of IL-10 and that Treg-secreted IL-10 has direct effects on MSC function. This is the first report to our knowledge revealing that Treg-secreted IL-10 is necessary for stromal cell maintenance, and our work outlines an alternative mechanism by which this cytokine regulates hematopoiesis.
Collapse
Affiliation(s)
| | | | | | - Jeremie M. Lever
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, and
| | - Zhengqin Yang
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Li Ying
- Cancer Science Institute of Singapore & Department of Biochemistry, National University of Singapore, Singapore
| | - Ashley E. Landuyt
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Emma C. Dean
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James F. George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Henry Yang
- Cancer Science Institute of Singapore & Department of Biochemistry, National University of Singapore, Singapore
| | - Paul Brent Ferrell
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig L. Maynard
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Casey T. Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Heth R. Turnquist
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|