1
|
Rowell J, Lau CI, Ross S, Yanez DC, Peña OA, Chain B, Crompton T. Distinct T-cell receptor (TCR) gene segment usage and MHC-restriction between foetal and adult thymus. eLife 2024; 13:RP93493. [PMID: 39636212 PMCID: PMC11620746 DOI: 10.7554/elife.93493] [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] [Indexed: 12/07/2024] Open
Abstract
Here, we sequenced rearranged TCRβ and TCRα chain sequences in CD4+CD8+ double positive (DP), CD4+CD8- single positive (SP4) and CD4-CD8+ (SP8) thymocyte populations from the foetus and young adult mouse. We found that life-stage had a greater impact on TCRβ and TCRα gene segment usage than cell-type. Foetal repertoires showed bias towards 3'TRAV and 5'TRAJ rearrangements in all populations, whereas adult repertoires used more 5'TRAV gene segments, suggesting that progressive TCRα rearrangements occur less frequently in foetal DP cells. When we synchronised young adult DP thymocyte differentiation by hydrocortisone treatment the new recovering DP thymocyte population showed more foetal-like 3'TRAV and 5'TRAJ gene segment usage. In foetus we identified less influence of MHC-restriction on α-chain and β-chain combinatorial VxJ usage and CDR1xCDR2 (V region) usage in SP compared to adult, indicating weaker impact of MHC-restriction on the foetal TCR repertoire. The foetal TCRβ repertoire was less diverse, less evenly distributed, with fewer non-template insertions, and all foetal populations contained more clonotypic expansions than adult. The differences between the foetal and adult thymus TCR repertoires are consistent with the foetal thymus producing αβT-cells with properties and functions that are distinct from adult T-cells: their repertoire is less governed by MHC-restriction, with preference for particular gene segment usage, less diverse with more clonotypic expansions, and more closely encoded by genomic sequence.
Collapse
Affiliation(s)
- Jasmine Rowell
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Ching-In Lau
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Susan Ross
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Diana C Yanez
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Oscar A Peña
- School of Biochemistry, University of BristolBristolUnited Kingdom
| | - Benny Chain
- Division of Infection and Immunity, University College LondonLondonUnited Kingdom
| | - Tessa Crompton
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| |
Collapse
|
2
|
Fiordoro S, Rosano C, Pechkova E, Barocci S, Izzotti A. Epigenetic modulation of immune cells: Mechanisms and implications. Adv Biol Regul 2024; 94:101043. [PMID: 39305736 DOI: 10.1016/j.jbior.2024.101043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 12/12/2024]
Abstract
Epigenetic modulation of the immune response entails modifiable and inheritable modifications that do not modify the DNA sequence. While there have been many studies on epigenetic changes in tumor cells, there is now a growing focus on epigenetically mediated changes in immune cells of both the innate and adaptive systems. These changes have significant implications for both the body's response to tumors and the development of potential therapeutic vaccines. This study primarily discusses the key epigenetic alterations, with a specific emphasis on pseudouridination, as well as non-coding RNAs and their transportation, which can lead to the development of cancer and the acquisition of new phenotypic traits by immune cells. Furthermore, the advancement of therapeutic vaccinations targeting the tumor will be outlined.
Collapse
Affiliation(s)
- S Fiordoro
- Department of Health Sciences, University of Genova, Via Pastore 1, 16132 Genova, Italy
| | - C Rosano
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy.
| | - E Pechkova
- Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| | - S Barocci
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - A Izzotti
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| |
Collapse
|
3
|
Rehermann B, Graham AL, Masopust D, Hamilton SE. Integrating natural commensals and pathogens into preclinical mouse models. Nat Rev Immunol 2024:10.1038/s41577-024-01108-3. [PMID: 39562646 DOI: 10.1038/s41577-024-01108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 11/21/2024]
Abstract
Fundamental discoveries in many aspects of mammalian physiology have been made using laboratory mice as research models. These studies have been facilitated by the genetic tractability and inbreeding of such mice, the large set of immunological reagents that are available, and the establishment of environmentally controlled, high-throughput facilities. Such facilities typically include barriers to keep the mouse colonies free of pathogens and the frequent re-derivation of the mice severely limits their commensal flora. Because humans have co-evolved with microorganisms and are exposed to a variety of pathogens, a growing community of researchers posits that preclinical disease research can be improved by studying mice in the context of the microbiota and pathogens that they would encounter in the natural world. Here, we provide a perspective of how these different approaches can be combined and integrated to improve existing mouse models to enhance our understanding of disease mechanisms and develop new therapies for humans. We also propose that the term 'mice with natural microbiota' is more appropriate for describing these models than existing terms such as 'dirty mice'.
Collapse
Affiliation(s)
- Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Andrea L Graham
- Department of Ecology & Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - David Masopust
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
4
|
Garcia-Rodriguez P, Hidalgo L, Rodriguez-Milla MA, Somovilla-Crespo B, Garcia-Castro J. LIN28 upregulation in primary human T cells impaired CAR T antitumoral activity. Front Immunol 2024; 15:1462796. [PMID: 39478867 PMCID: PMC11521810 DOI: 10.3389/fimmu.2024.1462796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
LIN28, a highly conserved RNA-binding protein that acts as a posttranscriptional modulator, plays a vital role in the regulation of T-cell development, reprogramming, and immune activity in infectious diseases and T-cell-based immunotherapies. LIN28 inhibit the expression of let-7 miRNAs, the most prevalent family of miRNAs in lymphocytes. Recently it has been suggested that let-7 enhances murine anti-tumor immune responses. Here, we investigated the impact of LIN28 upregulation on human T cell functions, focusing on its influence on CAR T cell therapy. LIN28 lentiviral transduction of human T cells led to a stable expression of LIN28 that significantly downregulated the let-7 miRNA family without affecting cell viability or expansion potential. LIN28 overexpression maintained human T cell phenotype markers and functionality but impaired the antitumoral cytotoxicity of NKG2D-CAR T cells both in vitro and in vivo. These findings highlight the intricate relationship between LIN28/let-7 axis and human T cell functionality, including in CAR T cell therapy.
Collapse
Affiliation(s)
- Patricia Garcia-Rodriguez
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, Madrid, Spain
- Universidad Nacional de Educación a (UNED), Madrid, Spain
| | - Laura Hidalgo
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Javier Garcia-Castro
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación de Enfermedades Raras (IIER) & Departamento de Desarrollo de Medicamentos de Terapias Avanzadas (DDMTA), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Gu W, Eke C, Gonzalez Santiago E, Olaloye O, Konnikova L. Single-cell atlas of the small intestine throughout the human lifespan demonstrates unique features of fetal immune cells. Mucosal Immunol 2024; 17:599-617. [PMID: 38555026 PMCID: PMC11384551 DOI: 10.1016/j.mucimm.2024.03.011] [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: 11/02/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Proper development of mucosal immunity is critical for human health. Over the past decade, it has become evident that in humans, this process begins in utero. However, there are limited data on the unique features and functions of fetal mucosal immune cells. To address this gap, we integrated several single-cell ribonucleic acid sequencing datasets of the human small intestine (SI) to create an SI transcriptional atlas throughout the human life span, ranging from the first trimester to adulthood, with a focus on immune cells. Fetal SI displayed a complex immune landscape comprising innate and adaptive immune cells that exhibited distinct transcriptional programs from postnatal samples, especially compared with pediatric and adult samples. We identified shifts in myeloid populations across gestation and progression of memory T-cell states throughout the human lifespan. In particular, there was a marked shift of memory T cells from those with stem-like properties in the fetal samples to fully differentiated cells with a high expression of activation and effector function genes in adult samples, with neonatal samples containing both features. Finally, we demonstrate that the SI developmental atlas can be used to elucidate improper trajectories linked to mucosal diseases by implicating developmental abnormalities underlying necrotizing enterocolitis, a severe intestinal complication of prematurity. Collectively, our data provide valuable resources and important insights into intestinal immunity that will facilitate regenerative medicine and disease understanding.
Collapse
Affiliation(s)
- Weihong Gu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Chino Eke
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | | - Oluwabunmi Olaloye
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Obstetrics, Gynecology and Reproductive Science, Yale University School of Medicine, New Haven, CT, USA; Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT, USA; Program in Human Translational Immunology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
6
|
Sankaran DG, Zhu H, Maymi VI, Forlastro IM, Jiang Y, Laniewski N, Scheible KM, Rudd BD, Grimson AW. Gene Regulatory Programs that Specify Age-Related Differences during Thymocyte Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599011. [PMID: 38948840 PMCID: PMC11212896 DOI: 10.1101/2024.06.14.599011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
T cell development is fundamental to immune system establishment, yet how this development changes with age remains poorly understood. Here, we construct a transcriptional and epigenetic atlas of T cell developmental programs in neonatal and adult mice, revealing the ontogeny of divergent gene regulatory programs and their link to age-related differences in phenotype and function. Specifically, we identify a gene module that diverges with age from the earliest stages of genesis and includes programs that govern effector response and cell cycle regulation. Moreover, we reveal that neonates possess more accessible chromatin during early thymocyte development, likely establishing poised gene expression programs that manifest later in thymocyte development. Finally, we leverage this atlas, employing a CRISPR-based perturbation approach coupled with single-cell RNA sequencing as a readout to uncover a conserved transcriptional regulator, Zbtb20, that contributes to age-dependent differences in T cell development. Altogether, our study defines transcriptional and epigenetic programs that regulate age-specific differences in T cell development.
Collapse
|
7
|
Eddens T, Parks OB, Zhang Y, Manni ML, Casanova JL, Ogishi M, Williams JV. PD-1 signaling in neonates restrains CD8 + T cell function and protects against respiratory viral immunopathology. Mucosal Immunol 2024; 17:476-490. [PMID: 38176655 PMCID: PMC11180597 DOI: 10.1016/j.mucimm.2023.12.004] [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: 10/13/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Respiratory viral infections, including human metapneumovirus (HMPV), remain a leading cause of morbidity and mortality in neonates and infants. However, the mechanisms behind the increased sensitivity to those respiratory viral infections in neonates are poorly understood. Neonates, unlike adults, have several anti-inflammatory mechanisms in the lung, including elevated baseline expression of programmed death ligand 1 (PD-L1), a ligand for the inhibitory receptor programmed cell death protein 1 (PD-1). We thus hypothesized that neonates would rely on PD-1:PD-L1 signaling to restrain antiviral CD8 responses. To test this, we developed a neonatal primary HMPV infection model using wild-type C57BL/6 (B6) and Pdcd1-/- (lacking PD-1) mice. HMPV-infected neonatal mice had increased PD-L1/PD-L2 co-expression on innate immune cells but a similar number of antigen-specific CD8+ T cells and upregulation of PD-1 to that of adult B6 mice. Neonatal CD8+ T cells had reduced interferon-gamma (IFN-γ), granzyme B, and interleukin-2 production compared with B6 adults. Pdcd1-/- neonatal CD8+ T cells had markedly increased production of IFN-γ and granzyme B compared with B6 neonates. Pdcd1-/- neonates had increased acute pathology with HMPV or influenza. Pdcd1-/- neonates infected with HMPV had long-term changes in pulmonary physiology with evidence of immunopathology and a persistent CD8+ T-cell response with increased granzyme B production. Using single-cell ribonucleic acid sequencing from a child lacking PD-1 signaling, a similar activated CD8+ T-cell signature with increased granzyme B expression was observed. These data indicate that PD-1 signaling critically limits CD8+ T-cell effector functions and prevents immunopathology in response to neonatal respiratory viral infections.
Collapse
Affiliation(s)
- Taylor Eddens
- Division of Allergy and Immunology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, Pennsylvania, USA
| | - Olivia B Parks
- University of Pittsburgh Medical Scientist Training Program, Pittsburgh, Pennsylvania, USA
| | - Yu Zhang
- Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, Pennsylvania, USA
| | - Michelle L Manni
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France; University Paris Cité, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA; Howard Hughes Medical Institute, New York, New York, USA; Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - John V Williams
- University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, Pennsylvania, USA; Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
8
|
Montenegro C, Perdomo-Celis F, Franco MA. Update on Early-Life T Cells: Impact on Oral Rotavirus Vaccines. Viruses 2024; 16:818. [PMID: 38932111 PMCID: PMC11209100 DOI: 10.3390/v16060818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Rotavirus infection continues to be a significant public health problem in developing countries, despite the availability of several vaccines. The efficacy of oral rotavirus vaccines in young children may be affected by significant immunological differences between individuals in early life and adults. Therefore, understanding the dynamics of early-life systemic and mucosal immune responses and the factors that affect them is essential to improve the current rotavirus vaccines and develop the next generation of mucosal vaccines. This review focuses on the advances in T-cell development during early life in mice and humans, discussing how immune homeostasis and response to pathogens is established in this period compared to adults. Finally, the review explores how this knowledge of early-life T-cell immunity could be utilized to enhance current and novel rotavirus vaccines.
Collapse
Affiliation(s)
| | | | - Manuel A. Franco
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá 110221, Colombia; (C.M.); (F.P.-C.)
| |
Collapse
|
9
|
Maymí VI, Zhu H, Jager M, Johnson S, Getchell R, Casey JW, Grenier JK, Wherry EJ, Smith NL, Grimson A, Rudd BD. Neonatal CD8+ T Cells Resist Exhaustion during Chronic Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:834-843. [PMID: 38231127 PMCID: PMC11298781 DOI: 10.4049/jimmunol.2300396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
Chronic viral infections, such as HIV and hepatitis C virus, represent a major public health problem. Although it is well understood that neonates and adults respond differently to chronic viral infections, the underlying mechanisms remain unknown. In this study, we transferred neonatal and adult CD8+ T cells into a mouse model of chronic infection (lymphocytic choriomeningitis virus clone 13) and dissected out the key cell-intrinsic differences that alter their ability to protect the host. Interestingly, we found that neonatal CD8+ T cells preferentially became effector cells early in chronic infection compared with adult CD8+ T cells and expressed higher levels of genes associated with cell migration and effector cell differentiation. During the chronic phase of infection, the neonatal cells retained more immune functionality and expressed lower levels of surface markers and genes related to exhaustion. Because the neonatal cells protect from viral replication early in chronic infection, the altered differentiation trajectories of neonatal and adult CD8+ T cells is functionally significant. Together, our work demonstrates how cell-intrinsic differences between neonatal and adult CD8+ T cells influence key cell fate decisions during chronic infection.
Collapse
Affiliation(s)
- Viviana I. Maymí
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Hongya Zhu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Mason Jager
- Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Shawn Johnson
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Rodman Getchell
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - James W. Casey
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Jennifer K. Grenier
- Transcriptional Regulation and Expression Facility, Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - E. John Wherry
- Institute for Immunology and Immune Health, and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
10
|
Watson NB, Patel RK, Kean C, Veazey J, Oyesola OO, Laniewski N, Grenier JK, Wang J, Tabilas C, Yee Mon KJ, McNairn AJ, Peng SA, Wesnak SP, Nzingha K, Davenport MP, Tait Wojno ED, Scheible KM, Smith NL, Grimson A, Rudd BD. The gene regulatory basis of bystander activation in CD8 + T cells. Sci Immunol 2024; 9:eadf8776. [PMID: 38394230 DOI: 10.1126/sciimmunol.adf8776] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
CD8+ T cells are classically recognized as adaptive lymphocytes based on their ability to recognize specific foreign antigens and mount memory responses. However, recent studies indicate that some antigen-inexperienced CD8+ T cells can respond to innate cytokines alone in the absence of cognate T cell receptor stimulation, a phenomenon referred to as bystander activation. Here, we demonstrate that neonatal CD8+ T cells undergo a robust and diverse program of bystander activation, which corresponds to enhanced innate-like protection against unrelated pathogens. Using a multi-omics approach, we found that the ability of neonatal CD8+ T cells to respond to innate cytokines derives from their capacity to undergo rapid chromatin remodeling, resulting in the usage of a distinct set of enhancers and transcription factors typically found in innate-like T cells. We observed that the switch between innate and adaptive functions in the CD8+ T cell compartment is mediated by changes in the abundance of distinct subsets of cells. The innate CD8+ T cell subset that predominates in early life was also present in adult mice and humans. Our findings provide support for the layered immune hypothesis and indicate that the CD8+ T cell compartment is more functionally diverse than previously thought.
Collapse
Affiliation(s)
- Neva B Watson
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Ravi K Patel
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Connor Kean
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Janelle Veazey
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Oyebola O Oyesola
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Nathan Laniewski
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jennifer K Grenier
- Genomics Innovation Hub and TREx Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Jocelyn Wang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Cybelle Tabilas
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Kristel J Yee Mon
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Adrian J McNairn
- Genomics Innovation Hub and TREx Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Seth A Peng
- Department of Clinical Science, Cornell University, Ithaca, NY 14853, USA
| | - Samantha P Wesnak
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Kito Nzingha
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Miles P Davenport
- Kirby Institute for Infection and Immunity, UNSW Australia, Sydney, NSW 2052, Australia
| | - Elia D Tait Wojno
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Kristin M Scheible
- Department of Pediatrics, University of Rochester, Rochester, NY 14642, USA
| | - Norah L Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
11
|
Philpott JD, Miller J, Boribong BP, Charles S, Davis JP, Kazimierczyk S, Jimena B, Leonard MM, Shreffler WG, Fasano A, Yonker LM, Jain N. Antigen-specific T cell responses in SARS-CoV-2 mRNA-vaccinated children. Cell Rep Med 2023; 4:101298. [PMID: 38016480 PMCID: PMC10772322 DOI: 10.1016/j.xcrm.2023.101298] [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: 06/07/2023] [Revised: 08/10/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
SARS-CoV-2 mRNA vaccines elicit humoral responses in children that are comparable to those in adults. However, early-life T cell responses are distinct from adult ones, and questions remain about the nature and kinetics of mRNA vaccine-induced T cell responses in children. We report that Pfizer BNT162b2 mRNA vaccination elicits a significant antigen-specific CD4+ T cell response in the ≥12-year-old cohort. This response is weaker in magnitude in the 5- to 11-year-old cohort and is not improved by a higher vaccine dose (Moderna mRNA1273, 100 μg), suggesting distinct developmental programming that may underscore early-life T cell immunity. Increased effector phenotypes of antigen-specific T cells in younger children correspond with elevated anti-receptor binding domain antibody levels, albeit at the cost of memory generation. These studies highlight aspects of age-specific adaptive immune responses and the need for careful consideration of priming conditions including vaccine dose and adjuvant in the pediatric population.
Collapse
Affiliation(s)
- Jordan D Philpott
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA
| | - Jordan Miller
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA
| | - Brittany P Boribong
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA; Pediatric Allergy and Immunology and Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, 175 Cambridge Street, Boston, MA 02114, USA
| | - Saeina Charles
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA
| | - Jameson P Davis
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA
| | - Simon Kazimierczyk
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA
| | - Brittany Jimena
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA
| | - Maureen M Leonard
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Wayne G Shreffler
- Pediatric Allergy and Immunology and Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, 175 Cambridge Street, Boston, MA 02114, USA
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA; Pediatric Allergy and Immunology and Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, 175 Cambridge Street, Boston, MA 02114, USA
| | - Lael M Yonker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA; Pediatric Allergy and Immunology and Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, 175 Cambridge Street, Boston, MA 02114, USA.
| | - Nitya Jain
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, 114 16(th) Street, Charlestown, MA 02129, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA.
| |
Collapse
|
12
|
Reynolds HM, Bettini ML. Early-life microbiota-immune homeostasis. Front Immunol 2023; 14:1266876. [PMID: 37936686 PMCID: PMC10627000 DOI: 10.3389/fimmu.2023.1266876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
As the prevalence of allergy and autoimmune disease in industrialized societies continues to rise, improving our understanding of the mechanistic roles behind microbiota-immune homeostasis has become critical for informing therapeutic interventions in cases of dysbiosis. Of particular importance, are alterations to intestinal microbiota occurring within the critical neonatal window, during which the immune system is highly vulnerable to environmental exposures. This review will highlight recent literature concerning mechanisms of early-life microbiota-immune homeostasis as well as discuss the potential for therapeutics in restoring dysbiosis in early life.
Collapse
Affiliation(s)
| | - Matthew L. Bettini
- Department of Microbiology and Immunology, University of Utah, Salt Lake, UT, United States
| |
Collapse
|
13
|
Borrill R, Poulton K, Wynn R. Immunology of cord blood T-cells favors augmented disease response during clinical pediatric stem cell transplantation for acute leukemia. Front Pediatr 2023; 11:1232281. [PMID: 37780051 PMCID: PMC10534014 DOI: 10.3389/fped.2023.1232281] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/22/2023] [Indexed: 10/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) has been an important and efficacious treatment for acute leukemia in children for over 60 years. It works primarily through the graft-vs.-leukemia (GVL) effect, in which donor T-cells and other immune cells act to eliminate residual leukemia. Cord blood is an alternative source of stem cells for transplantation, with distinct biological and immunological characteristics. Retrospective clinical studies report superior relapse rates with cord blood transplantation (CBT), when compared to other stem cell sources, particularly for patients with high-risk leukemia. Xenograft models also support the superiority of cord blood T-cells in eradicating malignancy, when compared to those derived from peripheral blood. Conversely, CBT has historically been associated with an increased risk of transplant-related mortality (TRM) and morbidity, particularly from infection. Here we discuss clinical aspects of CBT, the unique immunology of cord blood T-cells, their role in the GVL effect and future methods to maximize their utility in cellular therapies for leukemia, honing and harnessing their antitumor properties whilst managing the risks of TRM.
Collapse
Affiliation(s)
- Roisin Borrill
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kay Poulton
- Transplantation Laboratory, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Robert Wynn
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
14
|
Wells AC, Hioki KA, Angelou CC, Lynch AC, Liang X, Ryan DJ, Thesmar I, Zhanybekova S, Zuklys S, Ullom J, Cheong A, Mager J, Hollander GA, Pobezinskaya EL, Pobezinsky LA. Let-7 enhances murine anti-tumor CD8 T cell responses by promoting memory and antagonizing terminal differentiation. Nat Commun 2023; 14:5585. [PMID: 37696797 PMCID: PMC10495470 DOI: 10.1038/s41467-023-40959-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 08/17/2023] [Indexed: 09/13/2023] Open
Abstract
The success of the CD8 T cell-mediated immune response against infections and tumors depends on the formation of a long-lived memory pool, and the protection of effector cells from exhaustion. The advent of checkpoint blockade therapy has significantly improved anti-tumor therapeutic outcomes by reversing CD8 T cell exhaustion, but fails to generate effector cells with memory potential. Here, using in vivo mouse models, we show that let-7 miRNAs determine CD8 T cell fate, where maintenance of let-7 expression during early cell activation results in memory CD8 T cell formation and tumor clearance. Conversely, let-7-deficiency promotes the generation of a terminal effector population that becomes vulnerable to exhaustion and cell death in immunosuppressive environments and fails to reject tumors. Mechanistically, let-7 restrains metabolic changes that occur during T cell activation through the inhibition of the PI3K/AKT/mTOR signaling pathway and production of reactive oxygen species, potent drivers of terminal differentiation and exhaustion. Thus, our results reveal a role for let-7 in the time-sensitive support of memory formation and the protection of effector cells from exhaustion. Overall, our data suggest a strategy in developing next-generation immunotherapies by preserving the multipotency of effector cells rather than enhancing the efficacy of differentiation.
Collapse
Affiliation(s)
- Alexandria C Wells
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Kaito A Hioki
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
- UMass Biotech Training Program (BTP), Amherst, MA, USA
| | - Constance C Angelou
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Adam C Lynch
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Xueting Liang
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Daniel J Ryan
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Iris Thesmar
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Saule Zhanybekova
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Saulius Zuklys
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Jacob Ullom
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Agnes Cheong
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Jesse Mager
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Georg A Hollander
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Elena L Pobezinskaya
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA.
| | - Leonid A Pobezinsky
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
15
|
Sturmlechner I, Jain A, Mu Y, Weyand CM, Goronzy JJ. T cell fate decisions during memory cell generation with aging. Semin Immunol 2023; 69:101800. [PMID: 37494738 PMCID: PMC10528238 DOI: 10.1016/j.smim.2023.101800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
The defense against infectious diseases, either through natural immunity or after vaccinations, relies on the generation and maintenance of protective T cell memory. Naïve T cells are at the center of memory T cell generation during primary responses. Upon activation, they undergo a complex, highly regulated differentiation process towards different functional states. Naïve T cells maintained into older age have undergone epigenetic adaptations that influence their fate decisions during differentiation. We review age-sensitive, molecular pathways and gene regulatory networks that bias naïve T cell differentiation towards effector cell generation at the expense of memory and Tfh cells. As a result, T cell differentiation in older adults is associated with release of bioactive waste products into the microenvironment, higher stress sensitivity as well as skewing towards pro-inflammatory signatures and shorter life spans. These maladaptations not only contribute to poor vaccine responses in older adults but also fuel a more inflammatory state.
Collapse
Affiliation(s)
- Ines Sturmlechner
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Abhinav Jain
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yunmei Mu
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Cornelia M Weyand
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Medicine, Division of Rheumatology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Jörg J Goronzy
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Medicine, Division of Rheumatology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA.
| |
Collapse
|
16
|
Borrill R, Poulton K, Kusyk L, Routledge A, Bonney D, Hanasoge-Nataraj R, Powys M, Mustafa O, Campbell H, Senthil S, Dillon R, Jovanovic J, Morton S, James B, Rao K, Stanworth S, Konkel J, Wynn R. Granulocyte transfusion during cord blood transplant for relapsed, refractory AML is associated with massive CD8 + T-cell expansion, significant cytokine release syndrome and induction of disease remission. Br J Haematol 2023; 202:589-598. [PMID: 37211883 DOI: 10.1111/bjh.18863] [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: 02/16/2023] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023]
Abstract
In high-risk myeloid malignancy, relapse is reduced using cord blood transplant (CBT) but remains the principal cause of treatment failure. We previously described T-cell expansion in CBT recipients receiving granulocyte transfusions. We now report the safety and tolerability of such transfusions, T-cell expansion data, immunophenotype, cytokine profiles and clinical response in children with post-transplant relapsed acute leukaemia who received T-replete, HLA-mismatched CBT and pooled granulocytes within a phase I/II trial (ClinicalTrials.Gov NCT05425043). All patients received the transfusion schedule without significant clinical toxicity. Nine of ten patients treated had detectable measurable residual disease (MRD) pre-transplant. Nine patients achieved haematological remission, and eight became MRD negative. There were five deaths: transplant complications (n = 2), disease (n = 3), including two late relapses. Five patients are alive and in remission with 12.7 months median follow up. Significant T-cell expansion occurred in nine patients with a greater median lymphocyte count than a historical cohort between days 7-13 (median 1.73 × 109 /L vs. 0.1 × 109 /L; p < 0.0001). Expanded T-cells were predominantly CD8+ and effector memory or TEMRA phenotype. They exhibited markers of activation and cytotoxicity with interferon-gamma production. All patients developed grade 1-3 cytokine release syndrome (CRS) with elevated serum IL-6 and interferon-gamma.
Collapse
Affiliation(s)
- Roisin Borrill
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kay Poulton
- Transplantation Laboratory, Manchester University NHS Foundation Trust, Manchester, UK
- Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Laura Kusyk
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Amy Routledge
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Denise Bonney
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Ramya Hanasoge-Nataraj
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Madeleine Powys
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Omima Mustafa
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Helen Campbell
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Srividhya Senthil
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Richard Dillon
- Department of Medical and Molecular Genetics, Kings College London, London, UK
| | - Jelena Jovanovic
- Department of Medical and Molecular Genetics, Kings College London, London, UK
| | | | - Beki James
- Leeds Children's Hospital, Leeds General Infirmary, Leeds, UK
| | - Kanchan Rao
- Department of Blood and Marrow Transplantation, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | - Joanne Konkel
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert Wynn
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
17
|
Abstract
Historically, the immune system was believed to develop along a linear axis of maturity from fetal life to adulthood. Now, it is clear that distinct layers of immune cells are generated from unique waves of hematopoietic progenitors during different windows of development. This model, known as the layered immune model, has provided a useful framework for understanding why distinct lineages of B cells and γδ T cells arise in succession and display unique functions in adulthood. However, the layered immune model has not been applied to CD8+ T cells, which are still often viewed as a uniform population of cells belonging to the same lineage, with functional differences between cells arising from environmental factors encountered during infection. Recent studies have challenged this idea, demonstrating that not all CD8+ T cells are created equally and that the functions of individual CD8+ T cells in adults are linked to when they were created in the host. In this review, we discuss the accumulating evidence suggesting there are distinct ontogenetic subpopulations of CD8+ T cells and propose that the layered immune model be extended to the CD8+ T cell compartment.
Collapse
Affiliation(s)
- Cybelle Tabilas
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
- Co-first author
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
- Co-first author
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
18
|
Paiola M, Dimitrakopoulou D, Pavelka MS, Robert J. Amphibians as a model to study the role of immune cell heterogeneity in host and mycobacterial interactions. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 139:104594. [PMID: 36403788 DOI: 10.1016/j.dci.2022.104594] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
Mycobacterial infections represent major concerns for aquatic and terrestrial vertebrates including humans. Although our current knowledge is mostly restricted to Mycobacterium tuberculosis and mammalian host interactions, increasing evidence suggests common features in endo- and ectothermic animals infected with non-tuberculous mycobacteria (NTMs) like those described for M. tuberculosis. Importantly, most of the pathogenic and non-pathogenic NTMs detected in amphibians from wild, farmed, and research facilities represent, in addition to the potential economic loss, a rising concern for human health. Upon mycobacterial infection in mammals, the protective immune responses involving the innate and adaptive immune systems are highly complex and therefore not fully understood. This complexity results from the versatility and resilience of mycobacteria to hostile conditions as well as from the immune cell heterogeneity arising from the distinct developmental origins according with the concept of layered immunity. Similar to the differing responses of neonates versus adults during tuberculosis development, the pathogenesis and inflammatory responses are stage-specific in Xenopus laevis during infection by the NTM M. marinum. That is, both in human fetal and neonatal development and in tadpole development, responses are characterized by hypo-responsiveness and a lower capacity to contain mycobacterial infections. Similar to a mammalian fetus and neonates, T cells and myeloid cells in Xenopus tadpoles and axolotls are different from the adult immune cells. Fetal and amphibian larval T cells, which are characterized by a lower T cell receptor (TCR) repertoire diversity, are biased toward regulatory function, and they have distinct progenitor origins from those of the adult immune cells. Some early developing T cells and likely macrophage subpopulations are conserved in adult anurans and mammals, and therefore, they likely play an important role in the host-pathogen interactions from early stages of development to adulthood. Thus, we propose the use of developing amphibians, which have the advantage of being free-living early in their development, as an alternative and complementary model to study the role of immune cell heterogeneity in host-mycobacteria interactions.
Collapse
Affiliation(s)
- Matthieu Paiola
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Dionysia Dimitrakopoulou
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Martin S Pavelka
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jacques Robert
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
19
|
Åkerstrand H, Boldrin E, Montano G, Vanhee S, Olsson K, Krausse N, Vergani S, Cieśla M, Bellodi C, Yuan J. Enhanced protein synthesis is a defining requirement for neonatal B cell development. Front Immunol 2023; 14:1130930. [PMID: 37138883 PMCID: PMC10149930 DOI: 10.3389/fimmu.2023.1130930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/21/2023] [Indexed: 05/05/2023] Open
Abstract
The LIN28B RNA binding protein exhibits an ontogenically restricted expression pattern and is a key molecular regulator of fetal and neonatal B lymphopoiesis. It enhances the positive selection of CD5+ immature B cells early in life through amplifying the CD19/PI3K/c-MYC pathway and is sufficient to reinitiate self-reactive B-1a cell output when ectopically expressed in the adult. In this study, interactome analysis in primary B cell precursors showed direct binding by LIN28B to numerous ribosomal protein transcripts, consistent with a regulatory role in cellular protein synthesis. Induction of LIN28B expression in the adult setting is sufficient to promote enhanced protein synthesis during the small Pre-B and immature B cell stages, but not during the Pro-B cell stage. This stage dependent effect was dictated by IL-7 mediated signaling, which masked the impact of LIN28B through an overpowering stimulation on the c-MYC/protein synthesis axis in Pro-B cells. Importantly, elevated protein synthesis was a distinguishing feature between neonatal and adult B cell development that was critically supported by endogenous Lin28b expression early in life. Finally, we used a ribosomal hypomorphic mouse model to demonstrate that subdued protein synthesis is specifically detrimental for neonatal B lymphopoiesis and the output of B-1a cells, without affecting B cell development in the adult. Taken together, we identify elevated protein synthesis as a defining requirement for early-life B cell development that critically depends on Lin28b. Our findings offer new mechanistic insights into the layered formation of the complex adult B cell repertoire.
Collapse
Affiliation(s)
- Hugo Åkerstrand
- Developmental Immunology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Elena Boldrin
- Developmental Immunology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Giorgia Montano
- Developmental Immunology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Stijn Vanhee
- Developmental Immunology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Karin Olsson
- Developmental Immunology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Niklas Krausse
- Developmental Immunology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Stefano Vergani
- Developmental Immunology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Maciej Cieśla
- RNA and Stem Cell Biology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Cristian Bellodi
- RNA and Stem Cell Biology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Joan Yuan
- Developmental Immunology Unit, Department of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
- *Correspondence: Joan Yuan,
| |
Collapse
|
20
|
Early microbial exposure shapes adult immunity by altering CD8+ T cell development. Proc Natl Acad Sci U S A 2022; 119:e2212548119. [PMID: 36442114 PMCID: PMC9894172 DOI: 10.1073/pnas.2212548119] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Microbial exposure during development can elicit long-lasting effects on the health of an individual. However, how microbial exposure in early life leads to permanent changes in the immune system is unknown. Here, we show that the microbial environment alters the set point for immune susceptibility by altering the developmental architecture of the CD8+ T cell compartment. In particular, early microbial exposure results in the preferential expansion of highly responsive fetal-derived CD8+ T cells that persist into adulthood and provide the host with enhanced immune protection against intracellular pathogens. Interestingly, microbial education of fetal-derived CD8+ T cells occurs during thymic development rather than in the periphery and involves the acquisition of a more effector-like epigenetic program. Collectively, our results provide a conceptual framework for understanding how microbial colonization in early life leads to lifelong changes in the immune system.
Collapse
|
21
|
Pieren DKJ, Boer MC, de Wit J. The adaptive immune system in early life: The shift makes it count. Front Immunol 2022; 13:1031924. [PMID: 36466865 PMCID: PMC9712958 DOI: 10.3389/fimmu.2022.1031924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/31/2022] [Indexed: 10/13/2023] Open
Abstract
Respiratory infectious diseases encountered early in life may result in life-threatening disease in neonates, which is primarily explained by the relatively naive neonatal immune system. Whereas vaccines are not readily available for all infectious diseases, vaccinations have greatly reduced childhood mortality. However, repeated vaccinations are required to reach protective immunity in infants and not all vaccinations are effective at young age. Moreover, protective adaptive immunity elicited by vaccination wanes more rapidly at young age compared to adulthood. The infant adaptive immune system has previously been considered immature but this paradigm has changed during the past years. Recent evidence shows that the early life adaptive immune system is equipped with a strong innate-like effector function to eliminate acute pathogenic threats. These strong innate-like effector capacities are in turn kept in check by a tolerogenic counterpart of the adaptive system that may have evolved to maintain balance and to reduce collateral damage. In this review, we provide insight into these aspects of the early life's adaptive immune system by addressing recent literature. Moreover, we speculate that this shift from innate-like and tolerogenic adaptive immune features towards formation of immune memory may underlie different efficacy of infant vaccination in these different phases of immune development. Therefore, presence of innate-like and tolerogenic features of the adaptive immune system may be used as a biomarker to improve vaccination strategies against respiratory and other infections in early life.
Collapse
Affiliation(s)
| | | | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
22
|
Rane S, Hogan T, Lee E, Seddon B, Yates AJ. Towards a unified model of naive T cell dynamics across the lifespan. eLife 2022; 11:78168. [PMID: 35678373 PMCID: PMC9348855 DOI: 10.7554/elife.78168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/08/2022] [Indexed: 12/02/2022] Open
Abstract
Naive CD4 and CD8 T cells are cornerstones of adaptive immunity, but the dynamics of their establishment early in life and how their kinetics change as they mature following release from the thymus are poorly understood. Further, due to the diverse signals implicated in naive T cell survival, it has been a long-held and conceptually attractive view that they are sustained by active homeostatic control as thymic activity wanes. Here we use multiple modelling and experimental approaches to identify a unified model of naive CD4 and CD8 T cell population dynamics in mice, across their lifespan. We infer that both subsets divide rarely, and progressively increase their survival capacity with cell age. Strikingly, this simple model is able to describe naive CD4 T cell dynamics throughout life. In contrast, we find that newly generated naive CD8 T cells are lost more rapidly during the first 3-4 weeks of life, likely due to increased recruitment into memory. We find no evidence for elevated division rates in neonates, or for feedback regulation of naive T cell numbers at any age. We show how confronting mathematical models with diverse datasets can reveal a quantitative and remarkably simple picture of naive T cell dynamics in mice from birth into old age.
Collapse
Affiliation(s)
- Sanket Rane
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States.,Irving Institute for Cancer Dynamics, Columbia University, New York, United States
| | - Thea Hogan
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Hospital, London, United Kingdom
| | - Edward Lee
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, United States
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Hospital, London, United Kingdom
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States
| |
Collapse
|
23
|
Eddens T, Parks OB, Williams JV. Neonatal Immune Responses to Respiratory Viruses. Front Immunol 2022; 13:863149. [PMID: 35493465 PMCID: PMC9047724 DOI: 10.3389/fimmu.2022.863149] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
Respiratory tract infections are a leading cause of morbidity and mortality in newborns, infants, and young children. These early life infections present a formidable immunologic challenge with a number of possibly conflicting goals: simultaneously eliminate the acute pathogen, preserve the primary gas-exchange function of the lung parenchyma in a developing lung, and limit long-term sequelae of both the infection and the inflammatory response. The latter has been most well studied in the context of childhood asthma, where multiple epidemiologic studies have linked early life viral infection with subsequent bronchospasm. This review will focus on the clinical relevance of respiratory syncytial virus (RSV), human metapneumovirus (HMPV), and rhinovirus (RV) and examine the protective and pathogenic host responses within the neonate.
Collapse
Affiliation(s)
- Taylor Eddens
- Pediatric Scientist Development Program, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Division of Allergy/Immunology, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Olivia B. Parks
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - John V. Williams
- Division of Pediatric Infectious Diseases, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
24
|
Bigley TM, Yang L, Kang LI, Saenz JB, Victorino F, Yokoyama WM. Disruption of thymic central tolerance by infection with murine roseolovirus induces autoimmune gastritis. J Exp Med 2022; 219:213039. [PMID: 35226043 PMCID: PMC8932538 DOI: 10.1084/jem.20211403] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/29/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Infections with herpesviruses, including human roseoloviruses, have been proposed to cause autoimmune disease, but defining a causal relationship and mechanism has been difficult due to the ubiquitous nature of infection and development of autoimmunity long after acute infection. Murine roseolovirus (MRV) is highly related to human roseoloviruses. Herein we show that neonatal MRV infection induced autoimmune gastritis (AIG) in adult mice in the absence of ongoing infection. MRV-induced AIG was dependent on replication during the neonatal period and was CD4+ T cell and IL-17 dependent. Moreover, neonatal MRV infection was associated with development of a wide array of autoantibodies in adult mice. Finally, neonatal MRV infection reduced medullary thymic epithelial cell numbers, thymic dendritic cell numbers, and thymic expression of AIRE and tissue-restricted antigens, in addition to increasing thymocyte apoptosis at the stage of negative selection. These findings strongly suggest that infection with a roseolovirus early in life results in disruption of central tolerance and development of autoimmune disease.
Collapse
Affiliation(s)
- Tarin M. Bigley
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO
| | - Liping Yang
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO
| | - Liang-I Kang
- Department of Pathology and Immunology, Division of Anatomic and Molecular Pathology, Washington University School of Medicine, St. Louis, MO
| | - Jose B. Saenz
- Department of Medicine, Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO
| | - Francisco Victorino
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO
| | - Wayne M. Yokoyama
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
25
|
Ucciferri CC, Dunn SE. Effect of puberty on the immune system: Relevance to multiple sclerosis. Front Pediatr 2022; 10:1059083. [PMID: 36533239 PMCID: PMC9755749 DOI: 10.3389/fped.2022.1059083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
Puberty is a dynamic period marked by changing levels of sex hormones, the development of secondary sexual characteristics and reproductive maturity. This period has profound effects on various organ systems, including the immune system. The critical changes that occur in the immune system during pubertal onset have been shown to have implications for autoimmune conditions, including Multiple Sclerosis (MS). MS is rare prior to puberty but can manifest in children after puberty. This disease also has a clear female preponderance that only arises following pubertal onset, highlighting a potential role for sex hormones in autoimmunity. Early onset of puberty has also been shown to be a risk factor for MS. The purpose of this review is to overview the evidence that puberty regulates MS susceptibility and disease activity. Given that there is a paucity of studies that directly evaluate the effects of puberty on the immune system, we also discuss how the immune system is different in children and mice of pre- vs. post-pubertal ages and describe how gonadal hormones may regulate these immune mechanisms. We present evidence that puberty enhances the expression of co-stimulatory molecules and cytokine production by type 2 dendritic cells (DC2s) and plasmacytoid dendritic cells (pDCs), increases T helper 1 (Th1), Th17, and T follicular helper immunity, and promotes immunoglobulin (Ig)G antibody production. Overall, this review highlights how the immune system undergoes a functional maturation during puberty, which has the potential to explain the higher prevalence of MS and other autoimmune diseases seen in adolescence.
Collapse
Affiliation(s)
- Carmen C Ucciferri
- Department of Immunology, The University of Toronto, Toronto, ON, Canada
| | - Shannon E Dunn
- Department of Immunology, The University of Toronto, Toronto, ON, Canada.,Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
| |
Collapse
|
26
|
Yee Mon KJ, Zhu H, Daly CWP, Vu LT, Smith NL, Patel R, Topham DJ, Scheible K, Jambo K, Le MTN, Rudd BD, Grimson A. MicroRNA-29 specifies age-related differences in the CD8+ T cell immune response. Cell Rep 2021; 37:109969. [PMID: 34758312 DOI: 10.1016/j.celrep.2021.109969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/09/2021] [Accepted: 10/19/2021] [Indexed: 11/27/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as critical regulators of cell fate in the CD8+ T cell response to infection. Although there are several examples of miRNAs acting on effector CD8+ T cells after infection, it is unclear whether differential expression of one or more miRNAs in the naive state is consequential in altering their long-term trajectory. To answer this question, we examine the role of miR-29 in neonatal and adult CD8+ T cells, which express different amounts of miR-29 only prior to infection and adopt profoundly different fates after immune challenge. We find that manipulation of miR-29 expression in the naive state is sufficient for age-adjusting the phenotype and function of CD8+ T cells, including their regulatory landscapes and long-term differentiation trajectories after infection. Thus, miR-29 acts as a developmental switch by controlling the balance between a rapid effector response in neonates and the generation of long-lived memory in adults.
Collapse
Affiliation(s)
- Kristel J Yee Mon
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Hongya Zhu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Ciarán W P Daly
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Luyen T Vu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA; Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Norah L Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Ravi Patel
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - David J Topham
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Kristin Scheible
- Department of Pediatrics, Division of Neonatology, University of Rochester, Rochester, NY 14642, USA
| | - Kondwani Jambo
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Minh T N Le
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA.
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
27
|
Holm SR, Jenkins BJ, Cronin JG, Jones N, Thornton CA. A role for metabolism in determining neonatal immune function. Pediatr Allergy Immunol 2021; 32:1616-1628. [PMID: 34170575 DOI: 10.1111/pai.13583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 01/04/2023]
Abstract
Immune responses of neonates differ markedly to those of adults, with skewed cytokine phenotypes, reduced inflammatory properties and drastically diminished memory function. Recent research efforts have started to unravel the role of cellular metabolism in determining immune cell fate and function. For studies in humans, much of the work on metabolic mechanisms underpinning innate and adaptive immune responses by different haematopoietic cell types is in adults. Studies investigating the contribution of metabolic adaptation in the unique setting of early life are just emerging, and much more work is needed to elucidate the contribution of metabolism to neonatal immune responses. Here, we discuss our current understanding of neonatal immune responses, examine some of the latest developments in neonatal immunometabolism and consider the possible role of altered metabolism to the distinctive immune phenotype of the neonate. Understanding the role of metabolism in regulating immune function at this critical stage in life has direct benefit for the child by affording opportunities to maximize immediate and long-term health. Additionally, gaining insight into the diversity of human immune function and naturally evolved immunometabolic strategies that modulate immune function could be harnessed for a wide range of opportunities including new therapeutic approaches.
Collapse
Affiliation(s)
- Sean R Holm
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea, UK
| | - Ben J Jenkins
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea, UK
| | - James G Cronin
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea, UK
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea, UK
| | - Catherine A Thornton
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea, UK
| |
Collapse
|
28
|
DNA Methylation and Immune Memory Response. Cells 2021; 10:cells10112943. [PMID: 34831166 PMCID: PMC8616503 DOI: 10.3390/cells10112943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022] Open
Abstract
The generation of memory is a cardinal feature of the adaptive immune response, involving different factors in a complex process of cellular differentiation. This process is essential for protecting the second encounter with pathogens and is the mechanism by which vaccines work. Epigenetic changes play important roles in the regulation of cell differentiation events. There are three types of epigenetic regulation: DNA methylation, histone modification, and microRNA expression. One of these epigenetic changes, DNA methylation, occurs in cytosine residues, mainly in CpG dinucleotides. This brief review aimed to analyse the literature to verify the involvement of DNA methylation during memory T and B cell development. Several studies have highlighted the importance of the DNA methyltransferases, enzymes that catalyse the methylation of DNA, during memory differentiation, maintenance, and function. The methylation profile within different subsets of naïve activated and memory cells could be an interesting tool to help monitor immune memory response.
Collapse
|
29
|
Phillips-Farfán B, Gómez-Chávez F, Medina-Torres EA, Vargas-Villavicencio JA, Carvajal-Aguilera K, Camacho L. Microbiota Signals during the Neonatal Period Forge Life-Long Immune Responses. Int J Mol Sci 2021; 22:ijms22158162. [PMID: 34360926 PMCID: PMC8348731 DOI: 10.3390/ijms22158162] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/27/2022] Open
Abstract
The microbiota regulates immunological development during early human life, with long-term effects on health and disease. Microbial products include short-chain fatty acids (SCFAs), formyl peptides (FPs), polysaccharide A (PSA), polyamines (PAs), sphingolipids (SLPs) and aryl hydrocarbon receptor (AhR) ligands. Anti-inflammatory SCFAs are produced by Actinobacteria, Bacteroidetes, Firmicutes, Spirochaetes and Verrucomicrobia by undigested-carbohydrate fermentation. Thus, fiber amount and type determine their occurrence. FPs bind receptors from the pattern recognition family, those from commensal bacteria induce a different response than those from pathogens. PSA is a capsular polysaccharide from B. fragilis stimulating immunoregulatory protein expression, promoting IL-2, STAT1 and STAT4 gene expression, affecting cytokine production and response modulation. PAs interact with neonatal immunity, contribute to gut maturation, modulate the gut–brain axis and regulate host immunity. SLPs are composed of a sphingoid attached to a fatty acid. Prokaryotic SLPs are mostly found in anaerobes. SLPs are involved in proliferation, apoptosis and immune regulation as signaling molecules. The AhR is a transcription factor regulating development, reproduction and metabolism. AhR binds many ligands due to its promiscuous binding site. It participates in immune tolerance, involving lymphocytes and antigen-presenting cells during early development in exposed humans.
Collapse
Affiliation(s)
- Bryan Phillips-Farfán
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
| | - Fernando Gómez-Chávez
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (F.G.-C.); (J.A.V.-V.)
- Cátedras CONACyT-Instituto Nacional de Pediatría, México City 04530, Mexico
- Departamento de Formación Básica Disciplinaria, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | | | | | - Karla Carvajal-Aguilera
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
| | - Luz Camacho
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
- Correspondence:
| |
Collapse
|
30
|
Dong M, Mallet Gauthier È, Fournier M, Melichar HJ. Developing the right tools for the job: Lin28 regulation of early life T-cell development and function. FEBS J 2021; 289:4416-4429. [PMID: 34077615 DOI: 10.1111/febs.16045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/29/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022]
Abstract
T cells comprise a functionally heterogeneous cell population that has important roles in the immune system. While T cells are broadly considered to be a component of the antigen-specific adaptive immune response, certain T-cell subsets display innate-like effector characteristics whereas others perform immunosuppressive functions. These functionally diverse T-cell populations preferentially arise at different stages of ontogeny and are tailored to the immunological priorities of the organism over time. Many differences in early life versus adult T-cell phenotypes can be attributed to the cell-intrinsic properties of the distinct progenitors that seed the thymus throughout development. It is becoming clear that Lin28, an evolutionarily conserved, heterochronic RNA-binding protein that is differentially expressed among early life and adult hematopoietic progenitor cells, plays a substantial role in influencing early T-cell development and function. Here, we discuss the mechanisms by which Lin28 shapes the T-cell landscape to protect the developing fetus and newborn. Manipulation of the Lin28 gene regulatory network is being considered as one means of improving hematopoietic stem cell transplant outcomes; as such, understanding the impact of Lin28 on T-cell function is of clinical relevance.
Collapse
Affiliation(s)
- Mengqi Dong
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada.,Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Ève Mallet Gauthier
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada.,Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Marilaine Fournier
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada.,Département de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
31
|
Cord blood CD8+ T-cell expansion following granulocyte transfusions eradicates refractory leukemia. Blood Adv 2021; 4:4165-4174. [PMID: 32886752 DOI: 10.1182/bloodadvances.2020001737] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/21/2020] [Indexed: 01/01/2023] Open
Abstract
The action of hematopoietic cell transplantation in controlling leukemia is principally mediated by donor T cells directed against residual recipient malignant cells. However, its utility is limited by graft-versus-host disease (GVHD), where alloreactivity is extended beyond leukemic and marrow cells. In a human/murine chimeric model, we previously showed that the preferential infiltration of cord blood (CB) CD8+ T cells eradicates an Epstein-Barr virus-driven lymphoblastoid tumor without causing xenogeneic GVHD. In the clinic, however, cord blood CD8+ T-cell reconstitution is significantly delayed, and the observation of such a robust antileukemia effect mediated by cord blood CD8+ T cells has not been reported. We describe an observation of very early T-cell expansion in 4 high-risk pediatric leukemia patients receiving third-party, pooled granulocytes after T cell-replete CB transplantation (CBT). The T-cell expansion was transient but robust, including expansion of CD8+ T cells, in contrast to the delayed CD8+ T-cell expansion ordinarily observed after T cell-replete CBT. The CD8+ T cells were polyclonal, rapidly switched to memory phenotype, and had the ability to mediate cytotoxicity. This phenomenon is reproducible, and each patient remains in long-term remission without GVHD. The results suggest that fetal-derived CB CD8+ T cells can be exploited to generate robust antileukemia effects without GVHD.
Collapse
|
32
|
Srinivasan J, Lancaster JN, Singarapu N, Hale LP, Ehrlich LIR, Richie ER. Age-Related Changes in Thymic Central Tolerance. Front Immunol 2021; 12:676236. [PMID: 33968086 PMCID: PMC8100025 DOI: 10.3389/fimmu.2021.676236] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/06/2021] [Indexed: 01/03/2023] Open
Abstract
Thymic epithelial cells (TECs) and hematopoietic antigen presenting cells (HAPCs) in the thymus microenvironment provide essential signals to self-reactive thymocytes that induce either negative selection or generation of regulatory T cells (Treg), both of which are required to establish and maintain central tolerance throughout life. HAPCs and TECs are comprised of multiple subsets that play distinct and overlapping roles in central tolerance. Changes that occur in the composition and function of TEC and HAPC subsets across the lifespan have potential consequences for central tolerance. In keeping with this possibility, there are age-associated changes in the cellular composition and function of T cells and Treg. This review summarizes changes in T cell and Treg function during the perinatal to adult transition and in the course of normal aging, and relates these changes to age-associated alterations in thymic HAPC and TEC subsets.
Collapse
Affiliation(s)
- Jayashree Srinivasan
- Department of Molecular Biosciences, Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, United States
| | | | - Nandini Singarapu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, TX, United States
| | - Laura P Hale
- Department of Pathology, Duke University School of Medicine, Durham, NC, United States
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, United States.,Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Ellen R Richie
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, TX, United States
| |
Collapse
|
33
|
Lutes LK, Steier Z, McIntyre LL, Pandey S, Kaminski J, Hoover AR, Ariotti S, Streets A, Yosef N, Robey EA. T cell self-reactivity during thymic development dictates the timing of positive selection. eLife 2021; 10:e65435. [PMID: 33884954 PMCID: PMC8116051 DOI: 10.7554/elife.65435] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
Functional tuning of T cells based on their degree of self-reactivity is established during positive selection in the thymus, although how positive selection differs for thymocytes with relatively low versus high self-reactivity is unclear. In addition, preselection thymocytes are highly sensitive to low-affinity ligands, but the mechanism underlying their enhanced T cell receptor (TCR) sensitivity is not fully understood. Here we show that murine thymocytes with low self-reactivity experience briefer TCR signals and complete positive selection more slowly than those with high self-reactivity. Additionally, we provide evidence that cells with low self-reactivity retain a preselection gene expression signature as they mature, including genes previously implicated in modulating TCR sensitivity and a novel group of ion channel genes. Our results imply that thymocytes with low self-reactivity downregulate TCR sensitivity more slowly during positive selection, and associate membrane ion channel expression with thymocyte self-reactivity and progress through positive selection.
Collapse
Affiliation(s)
- Lydia K Lutes
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Zoë Steier
- Department of Bioengineering, University of California, BerkeleyBerkeleyUnited States
| | - Laura L McIntyre
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Shraddha Pandey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - James Kaminski
- Center for Computational Biology, University of California, BerkeleyBerkeleyUnited States
| | - Ashley R Hoover
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Silvia Ariotti
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Aaron Streets
- Department of Bioengineering, University of California, BerkeleyBerkeleyUnited States
- Center for Computational Biology, University of California, BerkeleyBerkeleyUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Nir Yosef
- Department of Bioengineering, University of California, BerkeleyBerkeleyUnited States
- Center for Computational Biology, University of California, BerkeleyBerkeleyUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
- Department of Electrical Engineering and Computer Sciences, University of California, BerkeleyBerkeleyUnited States
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
| | - Ellen A Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
34
|
Vergani S, Yuan J. Developmental changes in the rules for B cell selection. Immunol Rev 2021; 300:194-202. [PMID: 33501672 DOI: 10.1111/imr.12949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Abstract
The autoimmune checkpoint during B cell maturation eliminates self-antigen reactive specificities from the mature B cell repertoire. However, an exception to this rule is illustrated by B-1 cells, an innate-like self-reactive B cell subset that is positively selected into the mature B cell pool in a self-antigen-driven fashion. The mechanisms by which B-1 cells escape central tolerance have puzzled the field for decades. A key clue comes from their restricted developmental window during fetal and neonatal life. Here we use B-1 cells as a prototypic early life derived B cell subset to explore developmental changes in the constraints of B cell selection. We discuss recent advancements in the understanding of the molecular program, centered around the RNA binding protein Lin28b, that licenses self-reactive B-1 cell output during ontogeny. Finally, we speculate on the possible link between the unique rules of early life B cell tolerance and the establishment of B cell - microbial mutualism to propose an integrated model for how developmental and environmental cues come together to create a protective layer of B cell memory involved in neonatal immune imprinting.
Collapse
Affiliation(s)
- Stefano Vergani
- Developmental Immunology Unit, Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Joan Yuan
- Developmental Immunology Unit, Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
35
|
Semmes EC, Chen JL, Goswami R, Burt TD, Permar SR, Fouda GG. Understanding Early-Life Adaptive Immunity to Guide Interventions for Pediatric Health. Front Immunol 2021; 11:595297. [PMID: 33552052 PMCID: PMC7858666 DOI: 10.3389/fimmu.2020.595297] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 12/04/2020] [Indexed: 01/16/2023] Open
Abstract
Infants are capable of mounting adaptive immune responses, but their ability to develop long-lasting immunity is limited. Understanding the particularities of the neonatal adaptive immune system is therefore critical to guide the design of immune-based interventions, including vaccines, in early life. In this review, we present a thorough summary of T cell, B cell, and humoral immunity in early life and discuss infant adaptive immune responses to pathogens and vaccines. We focus on the differences between T and B cell responses in early life and adulthood, which hinder the generation of long-lasting adaptive immune responses in infancy. We discuss how knowledge of early life adaptive immunity can be applied when developing vaccine strategies for this unique period of immune development. In particular, we emphasize the use of novel vaccine adjuvants and optimization of infant vaccine schedules. We also propose integrating maternal and infant immunization strategies to ensure optimal neonatal protection through passive maternal antibody transfer while avoiding hindering infant vaccine responses. Our review highlights that the infant adaptive immune system is functionally distinct and uniquely regulated compared to later life and that these particularities should be considered when designing interventions to promote pediatric health.
Collapse
Affiliation(s)
- Eleanor C. Semmes
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
- Medical Scientist Training Program, Duke University, Durham, NC, United States
- Children’s Health and Discovery Initiative, Department of Pediatrics, Duke University, Durham, NC, United States
| | - Jui-Lin Chen
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| | - Ria Goswami
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| | - Trevor D. Burt
- Children’s Health and Discovery Initiative, Department of Pediatrics, Duke University, Durham, NC, United States
- Division of Neonatology, Department of Pediatrics, Duke University, Durham, NC, United States
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
- Children’s Health and Discovery Initiative, Department of Pediatrics, Duke University, Durham, NC, United States
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
- Children’s Health and Discovery Initiative, Department of Pediatrics, Duke University, Durham, NC, United States
| |
Collapse
|
36
|
Abstract
T cell-mediated immune tolerance is a state of unresponsiveness of T cells towards specific self or non-self antigens. This is particularly essential during prenatal/neonatal period when T cells are exposed to dramatically changing environment and required to avoid rejection of maternal antigens, limit autoimmune responses, tolerate inert environmental and food antigens and antigens from non-harmful commensal microorganisms, promote maturation of mucosal barrier function, yet mount an appropriate response to pathogenic microorganisms. The cell-intrinsic and cell extrinsic mechanisms promote the generation of prenatal/neonatal T cells with distinct features to meet the complex and dynamic need of tolerance during this period. Reduced exposure or impaired tolerance in early life may have significant impact on allergic or autoimmune diseases in adult life. The uniqueness of conventional and regulatory T cells in human umbilical cord blood (UCB) may also provide certain advantages in UCB transplantation for hematological disorders.
Collapse
Affiliation(s)
- Lijun Yang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Rong Jin
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Dan Lu
- Institute of Systems Biomedicine, Peking University Health Science Center, Beijing, China
| | - Qing Ge
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
37
|
Jain N. The early life education of the immune system: Moms, microbes and (missed) opportunities. Gut Microbes 2020; 12:1824564. [PMID: 33043833 PMCID: PMC7781677 DOI: 10.1080/19490976.2020.1824564] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/29/2020] [Accepted: 09/08/2020] [Indexed: 02/03/2023] Open
Abstract
The early life immune system is characterized by unique developmental milestones. Functionally diverse immune cells arise from distinct waves of hematopoietic stem cells, a phenomenon referred to as 'layered' immunity. This stratified development of immune cells extends to lineages of both innate and adaptive cells. The defined time window for the development of these immune cells lends itself to the influence of specific exposures typical of the early life period. The perinatal immune system develops in a relatively sterile fetal environment but emerges into one filled with a multitude of antigenic encounters. A major burden of this comes in the form of the microbiota that is being newly established at mucosal surfaces of the newborn. Accumulating evidence suggests that early life microbial exposures, including those arising in utero, can imprint long-lasting changes in the offspring's immune system and determine disease risk throughout life. In this review, I highlight unique features of early life immunity and explore the role of intestinal bacteria in educating the developing immune system.
Collapse
Affiliation(s)
- Nitya Jain
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA, USA
| |
Collapse
|
38
|
Papadopoulou M, Sanchez Sanchez G, Vermijlen D. Innate and adaptive γδ T cells: How, when, and why. Immunol Rev 2020; 298:99-116. [PMID: 33146423 DOI: 10.1111/imr.12926] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
γδ T cells comprise the third cell lineage of lymphocytes that use, like αβ T cells and B cells, V(D)J gene rearrangement with the potential to generate a highly diverse T cell receptor (TCR) repertoire. There is no obvious conservation of γδ T cell subsets (based on TCR repertoire and/or function) between mice and human, leading to the notion that human and mouse γδ T cells are highly different. In this review, we focus on human γδ T cells, building on recent studies using high-throughput sequencing to analyze the TCR repertoire in various settings. We make then the comparison with mouse γδ T cell subsets highlighting the similarities and differences and describe the remarkable changes during lifespan of innate and adaptive γδ T cells. Finally, we propose mechanisms contributing to the generation of innate versus adaptive γδ T cells. We conclude that key elements related to the generation of the γδ TCR repertoire and γδ T cell activation/development are conserved between human and mice, highlighting the similarities between these two species.
Collapse
Affiliation(s)
- Maria Papadopoulou
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium.,ULB Center for Research in Immunology (U-CRI), Belgium
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium.,ULB Center for Research in Immunology (U-CRI), Belgium
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium.,ULB Center for Research in Immunology (U-CRI), Belgium
| |
Collapse
|
39
|
Anderson MK, Selvaratnam JS. Interaction between γδTCR signaling and the E protein-Id axis in γδ T cell development. Immunol Rev 2020; 298:181-197. [PMID: 33058287 DOI: 10.1111/imr.12924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023]
Abstract
γδ T cells acquire their functional properties in the thymus, enabling them to exert rapid innate-like responses. To understand how distinct γδ T cell subsets are generated, we have developed a Two-Stage model for γδ T cell development. This model is predicated on the finding that γδTCR signal strength impacts E protein activity through graded upregulation of Id3. Our model proposes that cells enter Stage 1 in response to a γδTCR signaling event in the cortex that activates a γδ T cell-specific gene network. Part of this program includes the upregulation of chemokine receptors that guide them to the medulla. In the medulla, Stage 1 cells receive distinct combinations of γδTCR, cytokine, and/co-stimulatory signals that induce their transit into Stage 2, either toward the γδT1 or the γδT17 lineage. The intersection between γδTCR and cytokine signals can tune Id3 expression, leading to different outcomes even in the presence of strong γδTCR signals. The thymic signaling niches required for γδT17 development are segregated in time and space, providing transient windows of opportunity during ontogeny. Understanding the regulatory context in which E proteins operate at different stages will be key in defining how their activity levels impose functional outcomes.
Collapse
Affiliation(s)
- Michele K Anderson
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Johanna S Selvaratnam
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Davenport MP, Smith NL, Rudd BD. Building a T cell compartment: how immune cell development shapes function. Nat Rev Immunol 2020; 20:499-506. [PMID: 32493982 DOI: 10.1038/s41577-020-0332-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
We are just beginning to understand the diversity of the peripheral T cell compartment, which arises from the specialization of different T cell subsets and the plasticity of individual naive T cells to adopt different fates. Although the progeny of a single T cell can differentiate into many phenotypes following infection, individual T cells are biased towards particular phenotypes. These biases are typically ascribed to random factors that occur during and after antigenic stimulation. However, the T cell compartment does not remain static with age, and shifting immune challenges during ontogeny give rise to T cells with distinct functional properties. Here, we argue that the developmental history of naive T cells creates a 'hidden layer' of diversity that persists into adulthood. Insight into this diversity can provide a new perspective on immunity and immunotherapy across the lifespan.
Collapse
Affiliation(s)
- Miles P Davenport
- Kirby Institute for Infection and Immunity, University of New South Wales Australia, Sydney, New South Wales, Australia.
| | - Norah L Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
41
|
Rackaityte E, Halkias J. Mechanisms of Fetal T Cell Tolerance and Immune Regulation. Front Immunol 2020; 11:588. [PMID: 32328065 PMCID: PMC7160249 DOI: 10.3389/fimmu.2020.00588] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/13/2020] [Indexed: 12/19/2022] Open
Abstract
The developing human fetus generates both tolerogenic and protective immune responses in response to the unique requirements of gestation. Thus, a successful human pregnancy depends on a fine balance between two opposing immunological forces: the semi-allogeneic fetus learns to tolerate both self- and maternal- antigens and, in parallel, develops protective immunity in preparation for birth. This critical window of immune development bridges prenatal immune tolerance with the need for postnatal environmental protection, resulting in a vulnerable neonatal period with heightened risk of infection. The fetal immune system is highly specialized to mediate this transition and thus serves a different function from that of the adult. Adaptive immune memory is already evident in the fetal intestine. Fetal T cells with pro-inflammatory potential are born in a tolerogenic environment and are tightly controlled by both cell-intrinsic and -extrinsic mechanisms, suggesting that compartmentalization and specialization, rather than immaturity, define the fetal immune system. Dysregulation of fetal tolerance generates an inflammatory response with deleterious effects to the pregnancy. This review aims to discuss the recent advances in our understanding of the cellular and molecular composition of fetal adaptive immunity and the mechanisms that govern T cell development and function. We also discuss the tolerance promoting environment that impacts fetal immunity and the consequences of its breakdown. A greater understanding of fetal mechanisms of immune activation and regulation has the potential to uncover novel paradigms of immune balance which may be leveraged to develop therapies for transplantation, autoimmune disease, and birth-associated inflammatory pathologies.
Collapse
Affiliation(s)
- Elze Rackaityte
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Joanna Halkias
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA, United States
| |
Collapse
|
42
|
Wells AC, Pobezinskaya EL, Pobezinsky LA. Non-coding RNAs in CD8 T cell biology. Mol Immunol 2020; 120:67-73. [PMID: 32085976 PMCID: PMC7093237 DOI: 10.1016/j.molimm.2020.01.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/17/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
CD8 T cells are among the most vigorous soldiers of the immune system that fight viral infections and cancer. CD8 T cell development, maintenance, activation and differentiation are under the tight control of multiple transcriptional and post-transcriptional networks. Over the last two decades it has become clear that non-coding RNAs (ncRNAs), which consist of microRNAs (miRNAs) and long ncRNAs (lncRNAs), have emerged as global biological regulators. While our understanding of the function of specific miRNAs has increased since the discovery of RNA interference, it is still very limited, and the field of lncRNAs is just starting to blossom. Here we will summarize our knowledge on the role of ncRNAs in CD8 T cell biology, including differentiation into memory and exhausted cells.
Collapse
Affiliation(s)
- Alexandria C Wells
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, United States.
| | - Elena L Pobezinskaya
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, United States.
| | - Leonid A Pobezinsky
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, United States.
| |
Collapse
|
43
|
Apostol AC, Jensen KDC, Beaudin AE. Training the Fetal Immune System Through Maternal Inflammation-A Layered Hygiene Hypothesis. Front Immunol 2020; 11:123. [PMID: 32117273 PMCID: PMC7026678 DOI: 10.3389/fimmu.2020.00123] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last century, the alarming surge in allergy and autoimmune disease has led to the hypothesis that decreasing exposure to microbes, which has accompanied industrialization and modern life in the Western world, has fundamentally altered the immune response. In its current iteration, the “hygiene hypothesis” suggests that reduced microbial exposures during early life restricts the production and differentiation of immune cells suited for immune regulation. Although it is now well-appreciated that the increase in hypersensitivity disorders represents a “perfect storm” of many contributing factors, we argue here that two important considerations have rarely been explored. First, the window of microbial exposure that impacts immune development is not limited to early childhood, but likely extends into the womb. Second, restricted microbial interactions by an expectant mother will bias the fetal immune system toward hypersensitivity. Here, we extend this discussion to hypothesize that the cell types sensing microbial exposures include fetal hematopoietic stem cells, which drive long-lasting changes to immunity.
Collapse
Affiliation(s)
- April C Apostol
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Kirk D C Jensen
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Anna E Beaudin
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| |
Collapse
|
44
|
Abstract
Neonatal CD4+ and CD8+ T cells have historically been characterized as immature or defective. However, recent studies prompt a reinterpretation of the functions of neonatal T cells. Rather than a population of cells always falling short of expectations set by their adult counterparts, neonatal T cells are gaining recognition as a distinct population of lymphocytes well suited for the rapidly changing environment in early life. In this review, I will highlight new evidence indicating that neonatal T cells are not inert or less potent versions of adult T cells but instead are a broadly reactive layer of T cells poised to quickly develop into regulatory or effector cells, depending on the needs of the host. In this way, neonatal T cells are well adapted to provide fast-acting immune protection against foreign pathogens, while also sustaining tolerance to self-antigens.
Collapse
Affiliation(s)
- Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York 14853, USA;
| |
Collapse
|
45
|
Tabilas C, Wang J, Liu X, Locasale JW, Smith NL, Rudd BD. Cutting Edge: Elevated Glycolytic Metabolism Limits the Formation of Memory CD8 + T Cells in Early Life. THE JOURNAL OF IMMUNOLOGY 2019; 203:2571-2576. [PMID: 31597706 DOI: 10.4049/jimmunol.1900426] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
Neonates often develop poor immunity against intracellular pathogens. Because CD8+ T cells are essential for eliminating infectious agents, it is crucial to understand why they behave differently in early life. Previous studies in mice have demonstrated that neonatal CD8+ T cells fail to form memory because of an intrinsic propensity to differentiate into short-lived effectors. However, the underlying mechanisms remain undefined. We now show that neonatal CD8+ T cells exhibit higher glycolytic activity than adult CD8+ T cells postinfection, which may be due to age-related differences in Lin28b expression. Importantly, when glycolysis is pharmacologically inhibited, the impaired formation of neonatal memory CD8+ T cells can be restored. Collectively, these data suggest that neonatal CD8+ T cells are inherently biased toward undergoing glycolytic metabolism postinfection, which compromises their ability to develop into memory CD8+ T cells in early life.
Collapse
Affiliation(s)
- Cybelle Tabilas
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Jocelyn Wang
- Department of Pediatrics, Indiana University, Indianapolis, IN 46202
| | - Xiaojing Liu
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695; and
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710
| | - Norah L Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853;
| |
Collapse
|
46
|
Vanhee S, Åkerstrand H, Kristiansen TA, Datta S, Montano G, Vergani S, Lang S, Ungerbäck J, Doyle A, Olsson K, Beneventi G, Jensen CT, Bellodi C, Soneji S, Sigvardsson M, Gyllenbäck EJ, Yuan J. Lin28b controls a neonatal to adult switch in B cell positive selection. Sci Immunol 2019; 4:4/39/eaax4453. [DOI: 10.1126/sciimmunol.aax4453] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022]
Abstract
The ability of B-1 cells to become positively selected into the mature B cell pool, despite being weakly self-reactive, has puzzled the field since its initial discovery. Here, we explore changes in B cell positive selection as a function of developmental time by exploiting a link between CD5 surface levels and the natural occurrence of self-reactive B cell receptors (BCRs) in BCR wild-type mice. We show that the heterochronic RNA binding protein Lin28b potentiates a neonatal mode of B cell selection characterized by enhanced overall positive selection in general and the developmental progression of CD5+immature B cells in particular. Lin28b achieves this by amplifying the CD19/PI3K/c-Myc positive feedback loop, and ectopic Lin28b expression restores both positive selection and mature B cell numbers in CD19−/−adult mice. Thus, the temporally restricted expression ofLin28brelaxes the rules for B cell selection during ontogeny by modulating tonic signaling. We propose that this neonatal mode of B cell selection represents a cell-intrinsic cue to accelerate the de novo establishment of the adaptive immune system and incorporate a layer of natural antibody-mediated immunity throughout life.
Collapse
|
47
|
Fike AJ, Kumova OK, Carey AJ. Dissecting the defects in the neonatal CD8 + T-cell response. J Leukoc Biol 2019; 106:1051-1061. [PMID: 31260598 DOI: 10.1002/jlb.5ru0319-105r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/24/2019] [Accepted: 06/10/2019] [Indexed: 12/26/2022] Open
Abstract
The neonatal period presents a complex scenario where the threshold of reactivity toward colonizing microbiota, maternal antigens, autoantigens, and pathogens must be carefully moderated and balanced. CD8+ T cells are critical for the response against intracellular bacteria and viruses, but this immune compartment maintains altered function relative to adult counterparts because of the unique challenges which infants face. Here, we review our current understanding of the factors which may promote the attenuation and altered function of the neonatal CD8+ T-cell response and potential avenues for future study. Specifically, we have focused on the neonatal CD8+ T-cell ontogeny, memory formation, TCR structure and repertoire, TCR inhibitory receptors, and the clinical implications of altered neonatal CD8+ T-cell function. Special emphasis has been placed on examining the response of preterm neonates relative to term neonates and adults.
Collapse
Affiliation(s)
- Adam J Fike
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Ogan K Kumova
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Alison J Carey
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
48
|
Hussain T, Quinn KM. Similar but different: virtual memory CD8 T cells as a memory-like cell population. Immunol Cell Biol 2019; 97:675-684. [PMID: 31140625 DOI: 10.1111/imcb.12277] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/26/2019] [Accepted: 05/26/2019] [Indexed: 01/02/2023]
Abstract
Immunological memory is a phenomenon where the immune system can respond more rapidly to pathogens and immunological challenges that it has previously encountered. It is defined by several key hallmarks. After an initial encounter, immune cells (1) expand and (2) differentiate to form memory cell populations. Memory cells are (3) long-lived and (4) facilitate more rapid immune responses to subsequent infection because of (i) an increase in cell number, (ii) a decrease in the signaling threshold required for entry into cell cycle or effector function and (iii) localization of cells to tissue sites for surveillance. Classically, immunological memory has been antigen specific but it is becoming apparent that mechanisms of immunological memory can be co-opted by innate or antigen-inexperienced immune cells to generate heterogeneity in immune responses. One such cell is the virtual memory CD8 T (TVM ) cell, which is a semi-differentiated but antigen-naïve CD8 T-cell population. This review will summarize current knowledge of how TVM cells are generated, their memory-like hallmarks, how they are maintained during steady state, infection and aging, and propose a model to integrate key signaling pathways during their generation.
Collapse
Affiliation(s)
- Tabinda Hussain
- Monash University Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Kylie M Quinn
- Monash University Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,RMIT University School of Biomedical and Health Sciences, Bundoora, VIC, Australia
| |
Collapse
|
49
|
For T Cells, the Child Is Father of the Man. Cell 2019; 174:16-18. [PMID: 29958105 DOI: 10.1016/j.cell.2018.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
When T cells respond to infectious agents, they differentiate into effector and memory cells. In this issue of Cell, Smith et al. use a genetic "time-stamping" method to show that the developmental time the T cell arises-near birth or as an adult-dictates what type of T effector or memory cell results.
Collapse
|
50
|
Fate mapping reveals the age structure of the peripheral T cell compartment. Proc Natl Acad Sci U S A 2019; 116:3974-3981. [PMID: 30765525 DOI: 10.1073/pnas.1811634116] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Accumulating evidence indicates that the immune system does not develop in a linear fashion, but rather as distinct developmental layers formed from sequential waves of hematopoietic stem cells, each giving rise to unique populations of immune cells at different stages of development. Although recent studies have indicated that conventional CD8+ T cells produced in early life persist into adulthood and exhibit distinct roles during infection, the developmental architecture of the peripheral T cell compartment remains undefined. In this study, we used a mouse model to permanently label CD8+ T cells produced during distinct windows of development and traced their history to generate fate maps of CD8+ T cells produced during different stages of life. We then used mathematical modeling to understand the age structure of the CD8+ T cell compartment across the lifespan. Interestingly, we found that survival rate of CD8+ T cells depends on both the age and developmental origin of the cells. Recently produced cells show an initial rapid decay rate, which slows with age of the animal at which the cells were produced. For cells produced at any age, the rate of decay also slows with the age of the cell. We derive a function to describe this and predict the "age distribution" of the CD8+ T cell pool for animals of any given age. These data provide a quantitative framework for understanding the ontogeny of the CD8+ T cell compartment and help to contextualize age-related changes in the CD8+ T cell response to infection.
Collapse
|