1
|
Faas MM, Smink AM. Shaping immunity: the influence of the maternal gut bacteria on fetal immune development. Semin Immunopathol 2025; 47:13. [PMID: 39891756 PMCID: PMC11787218 DOI: 10.1007/s00281-025-01039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025]
Abstract
The development of the fetal immune response is a highly complex process. In the present review, we describe the development of the fetal immune response and the role of the maternal gut bacteria in this process. In contrast to the previous belief that the fetal immune response is inert, it is now thought that the fetal immune response is uniquely tolerant to maternal and allo-antigens, but able to respond to infectious agents, such as bacteria. This is accomplished by the development of T cells toward regulatory T cells rather than toward effector T cells, but also by the presence of functional innate immune cells, such as monocytes and NK cells. Moreover, in fetuses there is different programming of CD8 + T cells and memory T cells toward innate immune cells rather than to adaptive immune cells. The maternal gut bacteria are important in shaping the fetal immune response by producing bacterial products and metabolites that pass the placenta into the fetus and influence development of the fetal immune response. Insight into how and when these products affect the fetal immune response may open new treatment options with pre- or probiotics to affect the maternal gut bacteria and therewith the fetal immune response.
Collapse
Affiliation(s)
- Marijke M Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen and University of Groningen, Hanzeplein 1, Groningen, 9713 GZ, The Netherlands.
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen and University of Groningen, Hanzeplein 1, Groningen, 9713 GZ, The Netherlands
| |
Collapse
|
2
|
Rezai T, Fell-Hakai S, Guleria S, Toldi G. The Role of Breast Milk Cell-Free DNA in the Regulation of the Neonatal Immune Response. Nutrients 2024; 16:4373. [PMID: 39770994 PMCID: PMC11678730 DOI: 10.3390/nu16244373] [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: 11/26/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The neonatal period is a critical phase for the development of the intestinal immune system, marked by rapid adaptation to the external environment and unique nutritional demands. Breast milk plays a pivotal role in this transition, yet the mechanisms by which it influences neonatal mucosal immunity remain unclear. This review examines the potential mechanisms by which cell-free DNA (cfDNA) in breast milk may impact neonatal immune development, particularly through Toll-like receptor 9 (TLR9) signalling and gut microbiota interactions. We propose that cfDNA in breast milk interacts with TLR9 on the apical surface of neonatal intestinal epithelial cells, potentially serving as an initial anti-inflammatory stimulus before the establishment of commensal bacteria. This hypothesis is supported by the high concentration and stability of cfDNA in breast milk, as well as the known activation of TLR9 by mitochondrial DNA in breast milk. The review emphasises the need for further empirical research to validate these interactions and their implications for neonatal health, suggesting that understanding these dynamics could lead to improved strategies for neonatal care and disease prevention.
Collapse
Affiliation(s)
| | | | | | - Gergely Toldi
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand; (T.R.); (S.F.-H.); (S.G.)
| |
Collapse
|
3
|
Kawajiri A, Li J, Koinuma K, Yang Z, Yoon HJ, Yi J, Nagashima H, Ishii M, Gao F, Sato K, Tayama S, Harigae H, Iwakura Y, Ishii N, Sher A, Ishigaki K, Zhu J, Kim KS, Kawabe T. Naturally arising memory-phenotype CD4 + T lymphocytes contain an undifferentiated population that can generate T H1, T H17, and T reg cells. SCIENCE ADVANCES 2024; 10:eadq6618. [PMID: 39630890 PMCID: PMC11619248 DOI: 10.1126/sciadv.adq6618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
Memory-phenotype (MP) CD4+ T lymphocytes develop from naïve cells via self-recognition at homeostasis. While previous studies defined MP cells as a heterogeneous population that comprises T helper 1 (TH1)/17-like subsets, functional significance of the T-bet- Rorγt- subpopulation remains unknown. Here we show that MP lymphocytes as a whole population can differentiate into TH1/17/regulatory T (Treg) cells to mediate mild and persistent inflammation in lymphopenic environments, whereas naïve cells exhibit strong, TH1-dominated responses. Moreover, we demonstrate that MP lymphocytes comprise not only TH1/17-differentiated subsets but a polyclonal, transcriptomically immature "undifferentiated" subpopulation at homeostasis. Furthermore, our data argue that while the T-bet+ Rorγt- MP subset is terminally TH1-differentiated, its undifferentiated counterpart retains the capacity to rapidly proliferate to differentiate into TH1/17/Treg cells, with the latter response tonically constrained by preexisting Treg cells. Together, our results identify undifferentiated MP CD4+ T lymphocytes as a unique precursor that has a diverse differentiation potential to generate TH1/17/Treg cells to contribute to pathogenesis of inflammation.
Collapse
Affiliation(s)
- Akihisa Kawajiri
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Hematology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jing Li
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Keita Koinuma
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ziying Yang
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hye Jin Yoon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jaeu Yi
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biological Science, Ajou University, Suwon, Republic of Korea
| | - Hiroyuki Nagashima
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Minami Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Feng Gao
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kosuke Sato
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hideo Harigae
- Department of Hematology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Alan Sher
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Li J, Yang Z, Kawajiri A, Sato K, Tayama S, Ishii N, Zhu J, Kawabe T. Excess generation and activation of naturally arising memory-phenotype CD4 + T lymphocytes are inhibited by regulatory T cells in steady state. Front Immunol 2024; 15:1429954. [PMID: 39221254 PMCID: PMC11361994 DOI: 10.3389/fimmu.2024.1429954] [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: 05/09/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Conventional CD4+ T lymphocytes consist of naïve, foreign antigen-specific memory, and self-antigen-driven memory-phenotype (MP) cell compartments at homeostasis. We recently showed that MP cells tonically proliferate in response to self-antigens and differentiate into the T-bet+ subset in steady state. How excess proliferation and differentiation of MP cells are inhibited remains unclear. Given immunosuppressive function of regulatory T cells (Tregs), it is possible that they are also involved in inhibition of spontaneous MP cell activation. Here we show using Foxp3-diphtheria toxin receptor-transgenic mice that both MP and naïve CD4+ T cells spontaneously proliferate and differentiate into Th1 cells upon acute Treg depletion. At an early time point post Treg depletion, MP as compared to naïve CD4+ T cells are preferentially activated while at a later stage, the response is dominated by activated cells originated from the naïve pool. Moreover, we argue that MP cell proliferation is driven by TCR and CD28 signaling whereas Th1 differentiation mediated by IL-2. Furthermore, our data indicate that such activation of MP and naïve CD4+ T lymphocytes contribute to development of multi-organ inflammation at early and later time points, respectively, after Treg ablation. Together our findings reveal that Tregs tonically inhibit early, spontaneous proliferation and Th1 differentiation of MP CD4+ T lymphocytes as well as late activation of naïve cells, thereby contributing to maintenance of T cell homeostasis.
Collapse
Affiliation(s)
- Jing Li
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ziying Yang
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Akihisa Kawajiri
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kosuke Sato
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
5
|
Borghesi A. Life-threatening infections in human newborns: Reconciling age-specific vulnerability and interindividual variability. Cell Immunol 2024; 397-398:104807. [PMID: 38232634 DOI: 10.1016/j.cellimm.2024.104807] [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: 10/29/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/19/2024]
Abstract
In humans, the interindividual variability of clinical outcome following exposure to a microorganism is immense, ranging from silent infection to life-threatening disease. Age-specific immune responses partially account for the high incidence of infection during the first 28 days of life and the related high mortality at population level. However, the occurrence of life-threatening disease in individual newborns remains unexplained. By contrast, inborn errors of immunity and their immune phenocopies are increasingly being discovered in children and adults with life-threatening viral, bacterial, mycobacterial and fungal infections. There is a need for convergence between the fields of neonatal immunology, with its in-depth population-wide characterization of newborn-specific immune responses, and clinical immunology, with its investigations of infections in patients at the cellular and molecular levels, to facilitate identification of the mechanisms of susceptibility to infection in individual newborns and the design of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Borghesi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia, EU, Italy; School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland.
| |
Collapse
|
6
|
Ria F, Delogu G, Ingrosso L, Sali M, Di Sante G. Secrets and lies of host-microbial interactions: MHC restriction and trans-regulation of T cell trafficking conceal the role of microbial agents on the edge between health and multifactorial/complex diseases. Cell Mol Life Sci 2024; 81:40. [PMID: 38216734 PMCID: PMC11071949 DOI: 10.1007/s00018-023-05040-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/04/2023] [Accepted: 11/06/2023] [Indexed: 01/14/2024]
Abstract
Here we critically discuss data supporting the view that microbial agents (pathogens, pathobionts or commensals alike) play a relevant role in the pathogenesis of multifactorial diseases, but their role is concealed by the rules presiding over T cell antigen recognition and trafficking. These rules make it difficult to associate univocally infectious agents to diseases' pathogenesis using the paradigm developed for canonical infectious diseases. (Cross-)recognition of a variable repertoire of epitopes leads to the possibility that distinct infectious agents can determine the same disease(s). There can be the need for sequential infection/colonization by two or more microorganisms to develop a given disease. Altered spreading of infectious agents can determine an unwanted activation of T cells towards a pro-inflammatory and trafficking phenotype, due to differences in the local microenvironment. Finally, trans-regulation of T cell trafficking allows infectious agents unrelated to the specificity of T cell to modify their homing to target organs, thereby driving flares of disease. The relevant role of microbial agents in largely prevalent diseases provides a conceptual basis for the evaluation of more specific therapeutic approaches, targeted to prevent (vaccine) or cure (antibiotics and/or Biologic Response Modifiers) multifactorial diseases.
Collapse
Affiliation(s)
- F Ria
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - G Delogu
- Mater Olbia Hospital, 07026, Olbia, Italy
- Department of Biotechnological, Basic, Intensivological and Perioperatory Sciences-Section of Microbiology, Università Cattolica del S Cuore, 00168, Rome, Italy
| | - L Ingrosso
- Department Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
- European Program for Public Health Microbiology Training (EUPHEM), European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| | - M Sali
- Department of Biotechnological, Basic, Intensivological and Perioperatory Sciences-Section of Microbiology, Università Cattolica del S Cuore, 00168, Rome, Italy
- Department of Laboratory and Infectivology Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - G Di Sante
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132, Perugia, Italy.
| |
Collapse
|
7
|
Koren O, Konnikova L, Brodin P, Mysorekar IU, Collado MC. The maternal gut microbiome in pregnancy: implications for the developing immune system. Nat Rev Gastroenterol Hepatol 2024; 21:35-45. [PMID: 38097774 DOI: 10.1038/s41575-023-00864-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 01/04/2024]
Abstract
The gut microbiome has important roles in host metabolism and immunity, and microbial dysbiosis affects human physiology and health. Maternal immunity and microbial metabolites during pregnancy, microbial transfer during birth, and transfer of immune factors, microorganisms and metabolites via breastfeeding provide critical sources of early-life microbial and immune training, with important consequences for human health. Only a few studies have directly examined the interactions between the gut microbiome and the immune system during pregnancy, and the subsequent effect on offspring development. In this Review, we aim to describe how the maternal microbiome shapes overall pregnancy-associated maternal, fetal and early neonatal immune systems, focusing on the existing evidence and highlighting current gaps to promote further research.
Collapse
Affiliation(s)
- Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Liza Konnikova
- Department of Paediatrics and Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Petter Brodin
- Department of Immunology and Inflammation, Imperial College London, London, UK
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
8
|
Kennedy KM, de Goffau MC, Perez-Muñoz ME, Arrieta MC, Bäckhed F, Bork P, Braun T, Bushman FD, Dore J, de Vos WM, Earl AM, Eisen JA, Elovitz MA, Ganal-Vonarburg SC, Gänzle MG, Garrett WS, Hall LJ, Hornef MW, Huttenhower C, Konnikova L, Lebeer S, Macpherson AJ, Massey RC, McHardy AC, Koren O, Lawley TD, Ley RE, O'Mahony L, O'Toole PW, Pamer EG, Parkhill J, Raes J, Rattei T, Salonen A, Segal E, Segata N, Shanahan F, Sloboda DM, Smith GCS, Sokol H, Spector TD, Surette MG, Tannock GW, Walker AW, Yassour M, Walter J. Questioning the fetal microbiome illustrates pitfalls of low-biomass microbial studies. Nature 2023; 613:639-649. [PMID: 36697862 PMCID: PMC11333990 DOI: 10.1038/s41586-022-05546-8] [Citation(s) in RCA: 171] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/09/2022] [Indexed: 01/26/2023]
Abstract
Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.
Collapse
Affiliation(s)
- Katherine M Kennedy
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Marcus C de Goffau
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Wellcome Sanger Institute, Cambridge, UK
| | - Maria Elisa Perez-Muñoz
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marie-Claire Arrieta
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, South Korea
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Thorsten Braun
- Department of Obstetrics and Experimental Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frederic D Bushman
- Department of Microbiology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel Dore
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jonathan A Eisen
- Department of Evolution and Ecology, University of California, Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Michal A Elovitz
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephanie C Ganal-Vonarburg
- Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michael G Gänzle
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
- Department of Medicine and Division of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Chair of Intestinal Microbiome, ZIEL-Institute for Food and Health, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Curtis Huttenhower
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liza Konnikova
- Departments of Pediatrics and Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Andrew J Macpherson
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Ruth C Massey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Alice Carolyn McHardy
- Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), Hannover Braunschweig site, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Trevor D Lawley
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Eric G Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jeroen Raes
- VIB Center for Microbiology, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Thomas Rattei
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eran Segal
- Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Harry Sokol
- Gastroenterology Department, AP-HP, Saint Antoine Hospital, Centre de Recherche Saint-Antoine, CRSA, INSERM and Sorbonne Université, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Tim D Spector
- Department of Twin Research, King's College London, London, UK
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gerald W Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Alan W Walker
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Moran Yassour
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland.
- Department of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|
9
|
Kim AE, Nelson A, Stimpert K, Flyckt RL, Thirumavalavan N, Baker KC, Weinmann SC, Hoimes CJ. Minding the Bathwater: Fertility and Reproductive Toxicity in the Age of Immuno-Oncology. JCO Oncol Pract 2022; 18:815-822. [PMID: 36174117 PMCID: PMC10166412 DOI: 10.1200/op.22.00469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/29/2022] [Indexed: 01/05/2023] Open
Abstract
Immune checkpoint inhibition has resulted in significant efficacy across many cancer types, including melanoma. Melanoma is the second most common cancer among those of reproductive age, yet the reproductive toxicities of adjuvant and first-line immunotherapy are largely unknown.The normal innate and adaptive immune systems play a vital role in reproductive organ homeostasis of men and women and are essential for implantation, fertility, and a successful pregnancy. The programmed cell death-1 receptor/programmed cell death receptor ligand-1 (PD-1/PD-L1) pathway is essential in several aspects of fertility and pregnancy. Recent studies have largely focused on the role of the PD-1/PD-L1 pathway in fetomaternal tolerance, highlighting the importance of intact immune regulation in promoting a successful pregnancy.In this review, we describe a case of a reproductive-aged female with stage IIIC melanoma who sought guidance on family planning after pembrolizumab therapy. We discuss the known fertility-related toxicities of immune checkpoint inhibitors, the potential targets for reproductive toxicity in males and nonpregnant females, and the implications of anti-PD-1 therapy in relation to fetomaternal tolerance. Informed decision making will benefit from data and consensus.
Collapse
Affiliation(s)
- Anne E. Kim
- Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, PA
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Ariel Nelson
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
- Department of Medicine, Division of Hematology and Oncology, The Medical College of Wisconsin, Milwaukee, WI
| | - Kyle Stimpert
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
- VA Northeast Ohio Healthcare System, Cleveland, OH
| | | | - Nannan Thirumavalavan
- Department of Urology, University Hospitals, Case Western Reserve University, Cleveland, OH
| | | | - Sophia C. Weinmann
- Division of Rheumatology and Immunology, Duke University, Durham, NC
- Center for Cancer Immunotherapy, Duke University, Durham, NC
| | - Christopher J. Hoimes
- Center for Cancer Immunotherapy, Duke University, Durham, NC
- Duke Cancer Institute, Duke University, Durham, NC
| |
Collapse
|
10
|
Homeostatic cytokines tune naivety and stemness of cord blood-derived transgenic T cells. Cancer Gene Ther 2022; 29:961-972. [PMID: 34645974 DOI: 10.1038/s41417-021-00395-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/28/2021] [Accepted: 09/28/2021] [Indexed: 12/18/2022]
Abstract
Engineered T-cell therapies have proven to be successful in cancer and their clinical effectiveness is directly correlated with the infused T-cell differentiation profile. Indeed, stem cell memory and central memory T cells proliferate and persist longer in vivo compared with more-differentiated T cells, while conferring enhanced antitumor activity. Here, we propose an optimized process using cord blood (CB) to generate minimally differentiated T-cell products in terms of phenotype, function, gene expression, and metabolism, using peripheral blood (PB)-derived T cells cultured with IL-2 as a standard. Phenotypically, CB-derived T cells, particularly CD4 T cells, are less differentiated than their PB counterparts when cultured with IL-2 or with IL-7 and IL-15. Furthermore, culture with IL-7 and IL-15 enables better preservation of less-differentiated CB-derived T cells compared with IL-2. In addition, transcriptomic and metabolic assessments of CB-derived transgenic T cells cultured with IL-7 and IL-15 point out their naivety and stemness signature. These relatively quiescent transgenic T cells are nevertheless primed for secondary stimulation and cytokine production. In conclusion, our study indicates that CB may be used as a source of early differentiated T cells to develop more effective adoptive cancer immunotherapy.
Collapse
|
11
|
Reitermaier R, Ayub T, Staller J, Kienzl P, Fortelny N, Vieyra-Garcia PA, Worda C, Fiala C, Staud C, Eppel W, Scharrer A, Krausgruber T, Elbe-Bürger A. The molecular and phenotypic makeup of fetal human skin T lymphocytes. Development 2022; 149:dev199781. [PMID: 34604909 PMCID: PMC8601710 DOI: 10.1242/dev.199781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
The adult human skin contains a vast number of T cells that are essential for skin homeostasis and pathogen defense. T cells are first observed in the skin at the early stages of gestation; however, our understanding of their contribution to early immunity has been limited by their low abundance and lack of comprehensive methodologies for their assessment. Here, we describe a new workflow for isolating and expanding significant amounts of T cells from fetal human skin. Using multiparametric flow cytometry and in situ immunofluorescence, we found a large population with a naive phenotype and small populations with a memory and regulatory phenotype. Their molecular state was characterized using single-cell transcriptomics and TCR repertoire profiling. Importantly, culture of total fetal skin biopsies facilitated T cell expansion without a substantial impact on their phenotype, a major prerequisite for subsequent functional assays. Collectively, our experimental approaches and data advance the understanding of fetal skin immunity and potential use in future therapeutic interventions.
Collapse
Affiliation(s)
- René Reitermaier
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Tanya Ayub
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Julia Staller
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Philip Kienzl
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Nikolaus Fortelny
- Department of Biosciences, University of Salzburg, Salzburg 5020, Austria
| | | | - Christof Worda
- Department of Obstetrics & Gynecology, Medical University of Vienna, Vienna 1090, Austria
| | - Christian Fiala
- Gynmed Clinic, Vienna 1150, Austria
- Department of Women's and Children's Health, Division of Obstetrics and Gynaecology, Karolinska Institute and Karolinska University Hospital, Stockholm 171 77, Sweden
| | - Clement Staud
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna 1090, Austria
| | - Wolfgang Eppel
- Department of Obstetrics & Gynecology, Medical University of Vienna, Vienna 1090, Austria
| | - Anke Scharrer
- Department of Pathology, Medical University of Vienna, Vienna 1090, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | |
Collapse
|
12
|
Matute JD, Finander B, Pepin D, Ai X, Smith NP, Li JZ, Edlow AG, Villani AC, Lerou PH, Kalish BT. Single-cell immunophenotyping of the fetal immune response to maternal SARS-CoV-2 infection in late gestation. Pediatr Res 2022; 91:1090-1098. [PMID: 34750520 PMCID: PMC8573077 DOI: 10.1038/s41390-021-01793-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/31/2021] [Accepted: 09/28/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND During the COVID-19 pandemic, thousands of pregnant women have been infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The implications of maternal SARS-CoV-2 infection on fetal and childhood well-being need to be characterized. We aimed to characterize the fetal immune response to maternal SARS-CoV-2 infection. METHODS We performed single-cell RNA-sequencing and T cell receptor sequencing on cord blood mononuclear cells (CBMCs) from newborns of mothers infected with SARS-CoV-2 in the third trimester (cases) or without SARS-CoV-2 infection (controls). RESULTS We identified widespread gene expression changes in CBMCs from cases, including upregulation of interferon-stimulated genes and major histocompatibility complex genes in CD14+ monocytes, transcriptional changes suggestive of activation of plasmacytoid dendritic cells, and activation and exhaustion of natural killer cells. Lastly, we observed fetal T cell clonal expansion in cases compared to controls. CONCLUSIONS As none of the infants were infected with SARS-CoV-2, our results suggest that maternal SARS-CoV-2 infection might modulate the fetal immune system in the absence of vertical transmission. IMPACT The implications of maternal SARS-CoV-2 infection in the absence of vertical transmission on fetal and childhood well-being are poorly understood. Maternal SARS-CoV-2 infection might modulate the fetal immune system in the absence of vertical transmission. This study raises important questions about the untoward effects of maternal SARS-CoV-2 on the fetus, even in the absence of vertical transmission.
Collapse
Affiliation(s)
- Juan D Matute
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital (MGH), Boston, MA, USA.
| | - Benjamin Finander
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - David Pepin
- Department of Pediatric Surgery, MGH, Boston, MA, USA
| | - Xingbin Ai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital (MGH), Boston, MA, USA
| | | | - Jonathan Z Li
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, MGH, Boston, MA, USA
| | | | - Paul H Lerou
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Brian T Kalish
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
- Division of Neonatology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
13
|
Toothaker JM, Olaloye O, McCourt BT, McCourt CC, Silva TN, Case RM, Liu P, Yimlamai D, Tseng G, Konnikova L. Immune landscape of human placental villi using single-cell analysis. Development 2022; 149:274057. [PMID: 35050308 PMCID: PMC8935213 DOI: 10.1242/dev.200013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 12/30/2021] [Indexed: 12/19/2022]
Abstract
Maintenance of a healthy pregnancy is reliant on a successful balance between the fetal and maternal immune systems. Although the maternal mechanisms responsible have been well studied, those used by the fetal immune system remain poorly understood. Using suspension mass cytometry and various imaging modalities, we report a complex immune system within the mid-gestation (17-23 weeks) human placental villi (PV). Consistent with recent reports in other fetal organs, T cells with memory phenotypes, although rare in abundance, were detected within the PV tissue and vasculature. Moreover, we determined that T cells isolated from PV samples may be more proliferative after T cell receptor stimulation than adult T cells at baseline. Collectively, we identified multiple subtypes of fetal immune cells within the PV and specifically highlight the enhanced proliferative capacity of fetal PV T cells.
Collapse
Affiliation(s)
- Jessica M. Toothaker
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA,Department of Pediatrics, Yale University, New Haven, CT 06510, USA
| | | | - Blake T. McCourt
- Department of Pediatrics, Yale University, New Haven, CT 06510, USA
| | - Collin C. McCourt
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15219, USA
| | - Tatiana N. Silva
- Department of Pediatrics, Yale University, New Haven, CT 06510, USA
| | - Rebecca M. Case
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15219, USA
| | - Peng Liu
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Dean Yimlamai
- Department of Pediatrics, Yale University, New Haven, CT 06510, USA
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Liza Konnikova
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA,Department of Pediatrics, Yale University, New Haven, CT 06510, USA,Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT 06519, USA,Program in Human and Translational Immunology, Yale University, New Haven, CT 06519, USA,Author for correspondence ()
| |
Collapse
|
14
|
Zhang L, Chu CQ. Gut Microbiota-Medication Interaction in Rheumatic Diseases. Front Immunol 2021; 12:796865. [PMID: 34925383 PMCID: PMC8678121 DOI: 10.3389/fimmu.2021.796865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023] Open
Abstract
Besides its contribution to the development of rheumatic diseases, the gut microbiota interact with anti-rheumatic drugs. The intestinal microbiota can directly metabolize many drugs and indirectly change drug metabolism through a complex multi-dimensional interaction with the host, thus affecting individual response to drug therapy and adverse effects. The focus of the current review is to address recent advances and important progress in our understanding of how the gut microbiota interact with anti-rheumatic drugs and provide perspectives on promoting precision treatment, drug discovery, and better therapy for rheumatic diseases.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States.,Section of Rheumatology, Veterans Affairs (VA) Portland Healthcare System, Portland, OR, United States
| |
Collapse
|
15
|
Twisselmann N, Pagel J, Künstner A, Weckmann M, Hartz A, Glaser K, Hilgendorff A, Göpel W, Busch H, Herting E, Weinberg JB, Härtel C. Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation. Front Immunol 2021; 12:762789. [PMID: 34868007 PMCID: PMC8637891 DOI: 10.3389/fimmu.2021.762789] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/26/2021] [Indexed: 11/15/2022] Open
Abstract
Preterm infants are highly susceptible to sustained lung inflammation, which may be triggered by exposure to multiple environmental cues such as supplemental oxygen (O2) and infections. We hypothesized that dysregulated macrophage (MФ) activation is a key feature leading to inflammation-mediated development of bronchopulmonary dysplasia (BPD) in preterm infants. Therefore, we aimed to determine age-dependent differences in immune responses of monocyte-derived MФ comparing cord blood samples derived from preterm (n=14) and term (n=19) infants as well as peripheral blood samples from healthy adults (n=17) after lipopolysaccharide (LPS) exposure. Compared to term and adult MФ, LPS-stimulated preterm MФ showed an enhanced and sustained pro-inflammatory immune response determined by transcriptome analysis, cytokine release inducing a RORC upregulation due to T cell polarization of neonatal T cells, and TLR4 surface expression. In addition, a double-hit model was developed to study pulmonary relevant exposure factors by priming MФ with hyperoxia (O2 = 65%) or hypoxia (O2 = 3%) followed by lipopolysaccharide (LPS, 100ng/ml). When primed by 65% O2, subsequent LPS stimulation in preterm MФ led to an exaggerated pro-inflammatory response (e.g. increased HLA-DR expression and cytokine release) compared to LPS stimulation alone. Both, exposure to 65% or 3% O2 together with subsequent LPS stimulation, resulted in an exaggerated pro-inflammatory response of preterm MФ determined by transcriptome analysis. Downregulation of two major transcriptional factors, early growth response gene (Egr)-2 and growth factor independence 1 (Gfi1), were identified to play a role in the exaggerated pro-inflammatory response of preterm MФ to LPS insult after priming with 65% or 3% O2. Preterm MФ responses to LPS and hyperoxia/hypoxia suggest their involvement in excessive inflammation due to age-dependent differences, potentially mediated by downregulation of Egr2 and Gfi1 in the developing lung.
Collapse
Affiliation(s)
- Nele Twisselmann
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Julia Pagel
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany.,Department of Infectious Diseases and Microbiology, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Institute of Experimental Dermatology, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Markus Weckmann
- Department of Pediatrics Pneumology & Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany.,Airway Research Center North (ARCN) , Member of the German Center for Lung Research (DZL), Lübeck, Germany
| | - Annika Hartz
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Kirsten Glaser
- Center for Pediatric Research, Division of Neonatology, Department of Women's and Children's Health, University of Leipzig Medical Centre, Leipzig, Germany
| | - Anne Hilgendorff
- Center for Comprehensive Developmental Care (CDeCLMU), Member of the German Center for Lung Research (DZL), Hospital of the Ludwig-Maximilians University (LMU), CPC-M bioArchive, Munich, Germany
| | - Wolfgang Göpel
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Institute of Experimental Dermatology, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Egbert Herting
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Jason B Weinberg
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States.,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States
| | - Christoph Härtel
- Department of Pediatrics, University of Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Taylor M, Pillaye J, Horsnell WGC. Inherent maternal type 2 immunity: Consequences for maternal and offspring health. Semin Immunol 2021; 53:101527. [PMID: 34838445 DOI: 10.1016/j.smim.2021.101527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023]
Abstract
An inherent elevation in type 2 immunity is a feature of maternal and offspring immune systems. This has diverse implications for maternal and offspring biology including influencing success of pregnancy, offspring immune development and maternal and offspring ability to control infection and diseases such as allergies. In this review we provide a broad insight into how this immunological feature of pregnancy and early life impacts both maternal and offspring biology. We also suggest how understanding of this axis of immune influence is and may be utilised to improve maternal and offspring health.
Collapse
Affiliation(s)
- Matthew Taylor
- Institute of Immunology and Infection Research, Ashworth Laboratories, The Kings Buildings, University of Edinburgh, West Mains Road, Edinburgh, EH9 3JT, UK.
| | - Jamie Pillaye
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - William Gordon Charles Horsnell
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK; Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Science, University of Cape Town, Cape Town, 7925, South Africa.
| |
Collapse
|
17
|
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: 0.8] [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
|
18
|
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: 1.0] [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
|
19
|
Oh JH, Rehermann B. Natural versus Laboratory World: Incorporating Wild-Derived Microbiota into Preclinical Rodent Models. THE JOURNAL OF IMMUNOLOGY 2021; 207:1703-1709. [PMID: 34544812 DOI: 10.4049/jimmunol.2100426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/28/2021] [Indexed: 01/12/2023]
Abstract
Advances in data collection (high-throughput shotgun metagenomics, transcriptomics, and metabolomics) and analysis (bioinformatics and multiomics) led to the realization that all mammals are metaorganisms, shaped not only by their own genome but also by the genomes of the microbes that colonize them. To date, most studies have focused on the bacterial microbiome, whereas curated databases for viruses, fungi, and protozoa are still evolving. Studies on the interdependency of microbial kingdoms and their combined effects on host physiology are just starting. Although it is clear that past and present exposure to commensals and pathogens profoundly affect human physiology, such exposure is lacking in standard preclinical models such as laboratory mice. Laboratory mouse colonies are repeatedly rederived in germ-free status and subjected to restrictive, pathogen-free housing conditions. This review summarizes efforts to bring the wild microbiome into the laboratory setting to improve preclinical models and their translational research value.
Collapse
Affiliation(s)
- Ji Hoon Oh
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| |
Collapse
|
20
|
Mishra A, Lai GC, Yao LJ, Aung TT, Shental N, Rotter-Maskowitz A, Shepherdson E, Singh GSN, Pai R, Shanti A, Wong RMM, Lee A, Khyriem C, Dutertre CA, Chakarov S, Srinivasan KG, Shadan NB, Zhang XM, Khalilnezhad S, Cottier F, Tan ASM, Low G, Chen P, Fan Y, Hor PX, Lee AKM, Choolani M, Vermijlen D, Sharma A, Fuks G, Straussman R, Pavelka N, Malleret B, McGovern N, Albani S, Chan JKY, Ginhoux F. Microbial exposure during early human development primes fetal immune cells. Cell 2021; 184:3394-3409.e20. [PMID: 34077752 PMCID: PMC8240556 DOI: 10.1016/j.cell.2021.04.039] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/09/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
The human fetal immune system begins to develop early during gestation; however, factors responsible for fetal immune-priming remain elusive. We explored potential exposure to microbial agents in utero and their contribution toward activation of memory T cells in fetal tissues. We profiled microbes across fetal organs using 16S rRNA gene sequencing and detected low but consistent microbial signal in fetal gut, skin, placenta, and lungs in the 2nd trimester of gestation. We identified several live bacterial strains including Staphylococcus and Lactobacillus in fetal tissues, which induced in vitro activation of memory T cells in fetal mesenteric lymph node, supporting the role of microbial exposure in fetal immune-priming. Finally, using SEM and RNA-ISH, we visualized discrete localization of bacteria-like structures and eubacterial-RNA within 14th weeks fetal gut lumen. These findings indicate selective presence of live microbes in fetal organs during the 2nd trimester of gestation and have broader implications toward the establishment of immune competency and priming before birth.
Collapse
Affiliation(s)
- Archita Mishra
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Ghee Chuan Lai
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Leong Jing Yao
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore
| | - Thet Tun Aung
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore
| | - Noam Shental
- Department of Mathematics and Computer Science, Open University of Israel, Ra'anana 4353701, Israel
| | - Aviva Rotter-Maskowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Edwin Shepherdson
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Gurmit Singh Naranjan Singh
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Rhea Pai
- Genome Institute of Singapore (GIS), A(∗)STAR, 60 Biopolis Street, Singapore 138672, Singapore
| | - Adhika Shanti
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Regina Men Men Wong
- Genome Institute of Singapore (GIS), A(∗)STAR, 60 Biopolis Street, Singapore 138672, Singapore
| | - Andrea Lee
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore
| | - Costerwell Khyriem
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Charles Antoine Dutertre
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore; Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore; Program in Emerging Infectious Disease, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Svetoslav Chakarov
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - K G Srinivasan
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Nurhidaya Binte Shadan
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Xiao-Meng Zhang
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Shabnam Khalilnezhad
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Fabien Cottier
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Alrina Shin Min Tan
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Gillian Low
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Phyllis Chen
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore
| | - Yiping Fan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Pei Xiang Hor
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Avery Khoo May Lee
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Mahesh Choolani
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, 1E Kent Ridge Road, Singapore 119228, Singpore
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Institute for Medical Immunology, ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels 1050, Belgium
| | - Ankur Sharma
- Genome Institute of Singapore (GIS), A(∗)STAR, 60 Biopolis Street, Singapore 138672, Singapore; Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, PO Box 7214, 6 Verdun Street, Nedlands, Perth, WA 6009, Australia; Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Garold Fuks
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Norman Pavelka
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Benoit Malleret
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore; Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore
| | - Naomi McGovern
- Department of Pathology and Centre for Trophoblast Research, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Salvatore Albani
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore.
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; OBGYN-Academic Clinical Program, Duke-NUS, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore 119077, Singapore.
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore; Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore; Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
21
|
Razzaghian HR, Sharafian Z, Sharma AA, Boyce GK, Lee K, Da Silva R, Orban PC, Sekaly RP, Ross CJ, Lavoie PM. Neonatal T Helper 17 Responses Are Skewed Towards an Immunoregulatory Interleukin-22 Phenotype. Front Immunol 2021; 12:655027. [PMID: 34012439 PMCID: PMC8126652 DOI: 10.3389/fimmu.2021.655027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 01/03/2023] Open
Abstract
Newborns are frequently affected by mucocutaneous candidiasis. Th17 cells essentially limit mucosal invasion by commensal Candida spp. Here, we sought to understand the molecular basis for the developmental lack of Th17 cell responses in circulating blood neonatal T cells. Naive cord blood CD4 T cells stimulated in Th17-differentiating conditions inherently produced high levels of the interleukin-22 immunoregulatory cytokine, particularly in the presence of neonatal antigen-presenting cells. A genome-wide transcriptome analysis comparing neonatal and adult naïve CD4 T cells ex vivo revealed major developmental differences in gene networks regulating Small Drosophila Mothers Against Decapentaplegic (SMAD) and Signal Transducer and Activator of Transcription 3 (STAT3) signaling. These changes were functionally validated by experiments showing that the requirement for TGF-β in human Th17 cell differentiation is age-dependent. Moreover, STAT3 activity was profoundly diminished while overexpression of the STAT3 gene restored Th17 cell differentiation capacity in neonatal T cells. These data reveal that Th17 cell responses are developmentally regulated at the gene expression level in human neonates. These developmental changes may protect newborns against pathological Th17 cell responses, at the same time increasing their susceptibility to mucocutaneous candidiasis.
Collapse
Affiliation(s)
- Hamid R. Razzaghian
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Zohreh Sharafian
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Experimental Medicine Program, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ashish A. Sharma
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Experimental Medicine Program, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology & Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Guilaine K. Boyce
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Experimental Medicine Program, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kelsey Lee
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rachel Da Silva
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Paul C. Orban
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rafick-Pierre Sekaly
- Department of Pathology & Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Colin J. Ross
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Pascal M. Lavoie
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Experimental Medicine Program, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
22
|
Callaway PC, Farrington LA, Feeney ME. Malaria and Early Life Immunity: Competence in Context. Front Immunol 2021; 12:634749. [PMID: 33679787 PMCID: PMC7933008 DOI: 10.3389/fimmu.2021.634749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/02/2021] [Indexed: 12/31/2022] Open
Abstract
Childhood vaccines have been the cornerstone tool of public health over the past century. A major barrier to neonatal vaccination is the “immaturity” of the infant immune system and the inefficiency of conventional vaccine approaches at inducing immunity at birth. While much of the literature on fetal and neonatal immunity has focused on the early life propensity toward immune tolerance, recent studies indicate that the fetus is more immunologically capable than previously thought, and can, in some circumstances, mount adaptive B and T cell responses to perinatal pathogens in utero. Although significant hurdles remain before these findings can be translated into vaccines and other protective strategies, they should lend optimism to the prospect that neonatal and even fetal vaccination is achievable. Next steps toward this goal should include efforts to define the conditions for optimal stimulation of infant immune responses, including antigen timing, dose, and route of delivery, as well as antigen presentation pathways and co-stimulatory requirements. A better understanding of these factors will enable optimal deployment of vaccines against malaria and other pathogens to protect infants during their period of greatest vulnerability.
Collapse
Affiliation(s)
- Perri C Callaway
- Infectious Diseases and Immunity Graduate Group, University of California, Berkeley, Berkeley, CA, United States.,Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Lila A Farrington
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Margaret E Feeney
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
23
|
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: 9.5] [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
|
24
|
Gaimann MU, Nguyen M, Desponds J, Mayer A. Early life imprints the hierarchy of T cell clone sizes. eLife 2020; 9:e61639. [PMID: 33345776 PMCID: PMC7870140 DOI: 10.7554/elife.61639] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/20/2020] [Indexed: 12/30/2022] Open
Abstract
The adaptive immune system responds to pathogens by selecting clones of cells with specific receptors. While clonal selection in response to particular antigens has been studied in detail, it is unknown how a lifetime of exposures to many antigens collectively shape the immune repertoire. Here, using mathematical modeling and statistical analyses of T cell receptor sequencing data, we develop a quantitative theory of human T cell dynamics compatible with the statistical laws of repertoire organization. We find that clonal expansions during a perinatal time window leave a long-lasting imprint on the human T cell repertoire, which is only slowly reshaped by fluctuating clonal selection during adult life. Our work provides a mechanism for how early clonal dynamics imprint the hierarchy of T cell clone sizes with implications for pathogen defense and autoimmunity.
Collapse
Affiliation(s)
- Mario U Gaimann
- Lewis-Sigler Institute for Integrative Genomics, Princeton UniversityPrincetonUnited States
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of Physics, Ludwig-Maximilians-Universität MünchenMünchenGermany
| | - Maximilian Nguyen
- Lewis-Sigler Institute for Integrative Genomics, Princeton UniversityPrincetonUnited States
| | - Jonathan Desponds
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
| | - Andreas Mayer
- Lewis-Sigler Institute for Integrative Genomics, Princeton UniversityPrincetonUnited States
| |
Collapse
|
25
|
Dhariwala MO, Karthikeyan D, Vasquez KS, Farhat S, Weckel A, Taravati K, Leitner EG, Clancy S, Pauli M, Piper ML, Cohen JN, Ashouri JF, Lowe MM, Rosenblum MD, Scharschmidt TC. Developing Human Skin Contains Lymphocytes Demonstrating a Memory Signature. CELL REPORTS MEDICINE 2020; 1:100132. [PMID: 33294857 PMCID: PMC7691438 DOI: 10.1016/j.xcrm.2020.100132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/20/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022]
Abstract
Lymphocytes in barrier tissues play critical roles in host defense and homeostasis. These cells take up residence in tissues during defined developmental windows, when they may demonstrate distinct phenotypes and functions. Here, we utilized mass and flow cytometry to elucidate early features of human skin immunity. Although most conventional αβ T (Tconv) cells in fetal skin have a naive, proliferative phenotype, a subset of CD4+ Tconv and CD8+ cells demonstrate memory-like features and a propensity for interferon (IFN)γ production. Skin regulatory T cells dynamically accumulate over the second trimester in temporal and regional association with hair follicle development. These fetal skin regulatory T cells (Tregs) demonstrate an effector memory phenotype while differing from their adult counterparts in expression of key effector molecules. Thus, we identify features of prenatal skin lymphocytes that may have key implications for understanding antigen and allergen encounters in utero and in infancy. CyTOF reveals a complex lymphocyte landscape in developing human skin Developing skin contains CD45RO+ conventional T cells with propensity to produce IFNγ Regulatory T cells (Tregs) in skin before birth display effector memory properties Skin Tregs increase in conjunction with initial hair follicle morphogenesis
Collapse
Affiliation(s)
- Miqdad O Dhariwala
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dhuvarakesh Karthikeyan
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kimberly S Vasquez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sepideh Farhat
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Antonin Weckel
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Keyon Taravati
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elizabeth G Leitner
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sean Clancy
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mariela Pauli
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Merisa L Piper
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jarish N Cohen
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Judith F Ashouri
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Margaret M Lowe
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
26
|
Vannella KM, Stein S, Connelly M, Swerczek J, Amaro-Carambot E, Coyle EM, Babyak A, Winkler CW, Saturday G, Gai ND, Hammoud DA, Dowd KA, Valencia LP, Ramos-Benitez MJ, Kindrachuk J, Pierson TC, Peterson KE, Brenchley JM, Whitehead SS, Khurana S, Herbert R, Chertow DS. Nonhuman primates exposed to Zika virus in utero are not protected against reinfection at 1 year postpartum. Sci Transl Med 2020; 12:eaaz4997. [PMID: 33115950 PMCID: PMC11256112 DOI: 10.1126/scitranslmed.aaz4997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
There is limited information about the impact of Zika virus (ZIKV) exposure in utero on the anti-ZIKV immune responses of offspring. We infected six rhesus macaque dams with ZIKV early or late in pregnancy and studied four of their offspring over the course of a year postpartum. Despite evidence of ZIKV exposure in utero, we observed no structural brain abnormalities in the offspring. We detected infant-derived ZIKV-specific immunoglobulin A antibody responses and T cell memory responses during the first year postpartum in the two offspring born to dams infected with ZIKV early in pregnancy. Critically, although the infants had acquired some immunological memory of ZIKV, it was not sufficient to protect them against reinfection with ZIKV at 1 year postpartum. The four offspring reexposed to ZIKV at 1 year postpartum all survived but exhibited acute viremia and viral tropism to lymphoid tissues; three of four reexposed offspring exhibited spinal cord pathology. These data suggest that macaque infants born to dams infected with ZIKV during pregnancy remain susceptible to postnatal infection and consequent neuropathology.
Collapse
Affiliation(s)
- Kevin M Vannella
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sydney Stein
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark Connelly
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joanna Swerczek
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, MD 20837, USA
| | - Emerito Amaro-Carambot
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth M Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Ashley Babyak
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Clayton W Winkler
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Neville D Gai
- Center for Infectious Disease Imaging, Radiology and Imaging Services, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Services, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kimberly A Dowd
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luis Perez Valencia
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcos J Ramos-Benitez
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jason Kindrachuk
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Theodore C Pierson
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karin E Peterson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steve S Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Richard Herbert
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, MD 20837, USA
| | - Daniel S Chertow
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Abstract
Purpose of Review Detail normal adaptive immune maturation during fetal and neonatal life and review the clinical implications of arrested immune development. Recent Findings Advancements in the field of immunology have enabled investigations of the adaptive immunity starting during fetal life. New insights have drawn important distinctions between the neonatal and adult immune systems. The presence of diverse immunologic responses in the perinatal period suggests the importance of in utero immune development. Disruption of immune maturation due to premature birth may have significant implications for clinical pathology. Summary Establishing protective adaptive immunity during the perinatal period is critical for effective immune responses later in life. Preterm infants are susceptible to aberrant immune system maturation and inflammatory immune responses have been associated with the development of necrotizing enterocolitis (NEC) and bronchopulmonary dysplasia (BPD). Improving our understanding of how immune responses contribute to the pathogenesis of NEC and BPD may offer new opportunities for future treatment and prevention of these diseases.
Collapse
|
28
|
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: 48] [Impact Index Per Article: 9.6] [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
|
29
|
Sproat T, Payne RP, Embleton ND, Berrington J, Hambleton S. T Cells in Preterm Infants and the Influence of Milk Diet. Front Immunol 2020; 11:1035. [PMID: 32582165 PMCID: PMC7280433 DOI: 10.3389/fimmu.2020.01035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 04/29/2020] [Indexed: 12/22/2022] Open
Abstract
Preterm infants born before 32 weeks gestational age (GA) have high rates of late onset sepsis (LOS) and necrotizing enterocolitis (NEC) despite recent improvements in infection control and nutrition. Breast milk has a clear protective effect against both these outcomes likely due to multiple mechanisms which are not fully understood but may involve effects on both the infant's immune system and the developing gut microbiota. Congregating at the interface between the mucosal barrier and the microbiota, innate and adaptive T lymphocytes (T cells) participate in this interaction but few studies have explored their development after preterm delivery. We conducted a literature review of T cell development that focuses on fetal development, postnatal maturation and the influence of milk diet. The majority of circulating T cells in the preterm infant display a naïve phenotype but are still able to initiate functional responses similar to those seen in term infants. T cells from preterm infants display a skew toward a T-helper 2(Th2) phenotype and have an increased population of regulatory cells (Tregs). There are significant gaps in knowledge in this area, particularly in regards to innate-like T cells, but work is emerging: transcriptomics and mass cytometry are currently being used to map out T cell development, whilst microbiomic approaches may help improve understanding of events at mucosal surfaces. A rapid rise in organoid models will allow robust exploration of host-microbe interactions and may support the development of interventions that modulate T-cell responses for improved infant health.
Collapse
Affiliation(s)
- Thomas Sproat
- Neonatal Intensive Care Unit, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
- Immunity and Inflammation Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rebecca Pamela Payne
- Immunity and Inflammation Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nicholas D. Embleton
- Neonatal Intensive Care Unit, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
- Population Health Science Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Janet Berrington
- Neonatal Intensive Care Unit, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
- Immunity and Inflammation Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Sophie Hambleton
- Immunity and Inflammation Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
30
|
Abstract
The blood and immune systems develop in parallel during early prenatal life. Waves of hematopoiesis separated in anatomical space and time give rise to circulating and tissue-resident immune cells. Previous observations have relied on animal models, which differ from humans in both their developmental timeline and exposure to microorganisms. Decoding the composition of the human immune system is now tractable using single-cell multi-omics approaches. Large-scale single-cell genomics, imaging technologies, and the Human Cell Atlas initiative have together enabled a systems-level mapping of the developing human immune system and its emergent properties. Although the precise roles of specific immune cells during development require further investigation, the system as a whole displays malleable and responsive properties according to developmental need and environmental challenge.
Collapse
Affiliation(s)
- Jong-Eun Park
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Faculty of Medical Sciences, Newcastle upon Tyne NE2 4HH, UK
| | - Berthold Gottgens
- Department of Haematology, University of Cambridge, Cambridge CB2 2XY, UK
- Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 2XY, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK.
- Department of Physics/Cavendish Laboratory, University of Cambridge, JJ Thomson Ave., Cambridge CB3 0HE, UK
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK.
- Biosciences Institute, Newcastle University, Faculty of Medical Sciences, Newcastle upon Tyne NE2 4HH, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK
| |
Collapse
|
31
|
Jardine L, Haniffa M. Reconstructing human DC, monocyte and macrophage development in utero using single cell technologies. Mol Immunol 2020; 123:1-6. [PMID: 32380279 DOI: 10.1016/j.molimm.2020.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/10/2020] [Accepted: 04/22/2020] [Indexed: 12/27/2022]
Abstract
The repertoire of dendritic cells (DCs), monocytes and macrophages in adult humans is diverse and we are appreciating this to a greater extent as high throughput methods, such a single-cell RNA sequencing, become widely adopted and scalable. This powerful lens of analysis is also beginning to shed light on prenatal immunology, allowing us to chart the emergence, tissue distribution and developmental regulation of DCs, monocytes and macrophages during early human life. In this review, we will integrate recent insights from studies of the developing immune system into our understanding of adult DC, monocyte and macrophage organization, illustrating where insights from early life both affirm and challenge current understanding.
Collapse
Affiliation(s)
- Laura Jardine
- Biosciences Institute, Newcastle University, Faculty of Medical Sciences, Newcastle upon Tyne, NE2 4HH, UK; Department of Haematology, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK.
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Faculty of Medical Sciences, Newcastle upon Tyne, NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE2 4LP, UK.
| |
Collapse
|
32
|
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: 91] [Impact Index Per Article: 18.2] [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
|
33
|
Spidale NA, Malhotra N, Frascoli M, Sylvia K, Miu B, Freeman C, Stadinski BD, Huseby E, Kang J. Neonatal-derived IL-17 producing dermal γδ T cells are required to prevent spontaneous atopic dermatitis. eLife 2020; 9:e51188. [PMID: 32065580 PMCID: PMC7025821 DOI: 10.7554/elife.51188] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 02/11/2020] [Indexed: 01/01/2023] Open
Abstract
Atopic Dermatitis (AD) is a T cell-mediated chronic skin disease and is associated with altered skin barrier integrity. Infants with mutations in genes involved in tissue barrier fitness are predisposed towards inflammatory diseases, but most do not develop or sustain the diseases, suggesting that there exist regulatory immune mechanisms to prevent aberrant inflammation. The absence of one single murine dermal cell type, the innate neonatal-derived IL-17 producing γδ T (Tγδ17) cells, from birth resulted in spontaneous, highly penetrant AD with many of the major hallmarks of human AD. In Tγδ17 cell-deficient mice, basal keratinocyte transcriptome was altered months in advance of AD induction. Tγδ17 cells respond to skin commensal bacteria and the fulminant disease in their absence was driven by skin commensal bacteria dysbiosis. AD in this model was characterized by highly expanded dermal αβ T clonotypes that produce the type three cytokines, IL-17 and IL-22. These results demonstrate that neonatal Tγδ17 cells are innate skin regulatory T cells that are critical for skin homeostasis, and that IL-17 has dual homeostatic and inflammatory function in the skin.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Autoantigens/genetics
- Cell Differentiation
- Dermatitis, Atopic/genetics
- Dermatitis, Atopic/immunology
- Dermatitis, Atopic/prevention & control
- Disease Models, Animal
- Gene Expression
- Interleukin-17/biosynthesis
- Interleukins/biosynthesis
- Keratinocytes/cytology
- Keratinocytes/metabolism
- Lymphocyte Activation
- Mice
- Mice, Knockout
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Skin/metabolism
- Skin/microbiology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Interleukin-22
Collapse
Affiliation(s)
- Nicholas A Spidale
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Nidhi Malhotra
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Michela Frascoli
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Katelyn Sylvia
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Bing Miu
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Coral Freeman
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Brian D Stadinski
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Eric Huseby
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Joonsoo Kang
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| |
Collapse
|
34
|
Emiloju OE, Potdar R, Jorge V, Gupta S, Varadi G. Clinical Advancement and Challenges of ex vivo Expansion of Human Cord Blood Cells. Clin Hematol Int 2019; 2:18-26. [PMID: 34595439 PMCID: PMC8432338 DOI: 10.2991/chi.d.191121.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/16/2019] [Indexed: 02/02/2023] Open
Abstract
Apart from peripheral blood stem cell (PBSC), umbilical cord blood (UCB) is now a recognized source of stem cells for transplantation. UCB is an especially important source of stem cells for minority populations, which would otherwise be unable to find appropriately matched adult donors. UCB has fewer mature T lymphocytes compared with peripheral blood, thus making a UCB transplantation (UCBT) with a greater degree of HLA mismatch possible. The limited cell dose per UCB sample is however associated with delayed engraftment and a higher risk of graft failure, especially in adult recipients. This lower cell dose can be optimized by performing double unit UCBT, ex vivo UCB expansion prior to transplant and enhancement of the capabilities of the stem cells to home to the bone marrow. UCB contains naïve and immature T cells, thus posing significant challenges with increased risk of infections, graft versus host diseases (GVHD) and relapse following UCBT. Cell engineering techniques have been developed to circumnavigate the immaturity of the T cells, and include virus-specific cytotoxic T cells (VSTs), T cells transduced with disease-specific chimeric antigen receptor (CAR T cells) and regulatory T cell (Tregs) engineering. In this article, we review the advances in UCB ex vivo expansion and engineering to improve engraftment and reduce complications. As further research continues to find ways to overcome the current challenges, outcomes from UCBT will likely improve.
Collapse
Affiliation(s)
| | - Rashmika Potdar
- Hematology and Oncology Department, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Vinicius Jorge
- Hematology and Oncology Department, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Sorab Gupta
- Hematology and Oncology Department, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Gabor Varadi
- Hematology and Oncology Department, Albert Einstein Medical Center, Philadelphia, PA, USA
| |
Collapse
|
35
|
Yegorov S, Joag V, Galiwango RM, Good SV, Okech B, Kaul R. Impact of Endemic Infections on HIV Susceptibility in Sub-Saharan Africa. TROPICAL DISEASES TRAVEL MEDICINE AND VACCINES 2019; 5:22. [PMID: 31798936 PMCID: PMC6884859 DOI: 10.1186/s40794-019-0097-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Human immunodeficiency virus (HIV) remains a leading cause of global morbidity with the highest burden in Sub-Saharan Africa (SSA). For reasons that are incompletely understood, the likelihood of HIV transmission is several fold higher in SSA than in higher income countries, and most of these infections are acquired by young women. Residents of SSA are also exposed to a variety of endemic infections, such as malaria and various helminthiases that could influence mucosal and systemic immunology. Since these immune parameters are important determinants of HIV acquisition and progression, this review explores the possible effects of endemic infections on HIV susceptibility and summarizes current knowledge of the epidemiology and underlying immunological mechanisms by which endemic infections could impact HIV acquisition. A better understanding of the interaction between endemic infections and HIV may enhance HIV prevention programs in SSA.
Collapse
Affiliation(s)
- Sergey Yegorov
- 1Departments of Immunology and Medicine, University of Toronto, Toronto, Canada.,2Department of Pedagogical Mathematics and Natural Science, Faculty of Education and Humanities, Suleyman Demirel University, Almaty, Kazakhstan
| | - Vineet Joag
- 3Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN USA
| | - Ronald M Galiwango
- 1Departments of Immunology and Medicine, University of Toronto, Toronto, Canada
| | - Sara V Good
- 4Genetics & Genome Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON Canada.,5Community Health Sciences, University of Manitoba, Winnipeg, MB Canada
| | | | - Rupert Kaul
- 1Departments of Immunology and Medicine, University of Toronto, Toronto, Canada.,7Department of Medicine, University Health Network, Toronto, Canada
| |
Collapse
|
36
|
Stras SF, Werner L, Toothaker JM, Olaloye OO, Oldham AL, McCourt CC, Lee YN, Rechavi E, Shouval DS, Konnikova L. Maturation of the Human Intestinal Immune System Occurs Early in Fetal Development. Dev Cell 2019; 51:357-373.e5. [PMID: 31607651 DOI: 10.1016/j.devcel.2019.09.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 06/16/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023]
Abstract
There are limited data on fetal and early life development of human intestinal immunity. Using mass cytometry (CyTOF) and next-generation sequencing of B and T cell receptor (BCR and TCR) repertoires, we demonstrate complex intestinal immunity from 16 weeks' gestational age (GA). Both BCR and TCR repertoires are diverse with CDRH and CDR3β length increasing with advancing GA. The difference-from-germline, CDR insertions and/or deletions, similarly occur in utero for TCR but not BCR, suggesting earlier mucosal T than B cell maturity. Innate immunity is dominated by macrophages, dendritic cells (DCs), innate lymphoid cells (ILCs), and natural killer (NK) cells. Follicular and transitional B cells are enriched in fetuses while CD69+IgM+ B cells are abundant in infants. Both CD4+ and CD8+ T cells are abundant, capable of secreting cytokines and are phenotypically of the tissue resident memory state in utero. Our data provide the foundation for a 2nd trimester and infant intestinal immune atlas and suggest that a complex innate and adaptive immune landscape exists significantly earlier than previously reported.
Collapse
Affiliation(s)
- Stephanie F Stras
- Division of Newborn Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Lael Werner
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Jessica M Toothaker
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Oluwabunmi O Olaloye
- Division of Newborn Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Austin L Oldham
- Division of Newborn Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Collin C McCourt
- Division of Newborn Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Yu Nee Lee
- Pediatric Department A, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer 52621, Israel; Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer 52621, Israel; Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Erez Rechavi
- Pediatric Department A, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer 52621, Israel; Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer 52621, Israel; Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dror S Shouval
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Liza Konnikova
- Division of Newborn Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
37
|
Odorizzi PM, Jagannathan P, McIntyre TI, Budker R, Prahl M, Auma A, Burt TD, Nankya F, Nalubega M, Sikyomu E, Musinguzi K, Naluwu K, Kakuru A, Dorsey G, Kamya MR, Feeney ME. In utero priming of highly functional effector T cell responses to human malaria. Sci Transl Med 2019; 10:10/463/eaat6176. [PMID: 30333241 DOI: 10.1126/scitranslmed.aat6176] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/10/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022]
Abstract
Malaria remains a significant cause of morbidity and mortality worldwide, particularly in infants and children. Some studies have reported that exposure to malaria antigens in utero results in the development of tolerance, which could contribute to poor immunity to malaria in early life. However, the effector T cell response to pathogen-derived antigens encountered in utero, including malaria, has not been well characterized. Here, we assessed the frequency, phenotype, and function of cord blood T cells from Ugandan infants born to mothers with and without placental malaria. We found that infants born to mothers with active placental malaria had elevated frequencies of proliferating effector memory fetal CD4+ T cells and higher frequencies of CD4+ and CD8+ T cells that produced inflammatory cytokines. Fetal CD4+ and CD8+ T cells from placental malaria-exposed infants exhibited greater in vitro proliferation to malaria antigens. Malaria-specific CD4+ T cell proliferation correlated with prospective protection from malaria during childhood. These data demonstrate that placental malaria is associated with the generation of proinflammatory malaria-responsive fetal T cells. These findings add to our current understanding of fetal immunity and indicate that a functional and protective pathogen-specific T cell response can be generated in utero.
Collapse
Affiliation(s)
- Pamela M Odorizzi
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA
| | | | - Tara I McIntyre
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA
| | - Rachel Budker
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA
| | - Mary Prahl
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Ann Auma
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Trevor D Burt
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | - Esther Sikyomu
- Infectious Disease Research Collaboration, Kampala, Uganda
| | | | - Kate Naluwu
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Abel Kakuru
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA
| | - Moses R Kamya
- School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Margaret E Feeney
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA. .,Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
38
|
Feeney ME. The immune response to malaria in utero. Immunol Rev 2019; 293:216-229. [PMID: 31553066 DOI: 10.1111/imr.12806] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022]
Abstract
Malaria causes tremendous early childhood morbidity and mortality, providing an urgent impetus for the development of a vaccine that is effective in neonates. However, the infant immune response to malaria may be influenced by events that occur well before birth. Placental malaria infection complicates one quarter of all pregnancies in Africa and frequently results in exposure of the fetus to malaria antigens in utero, while the immune system is still developing. Some data suggest that in utero exposure to malaria may induce immunologic tolerance that interferes with the development of protective immunity during childhood. More recently, however, a growing body of evidence suggests that fetal malaria exposure can prime highly functional malaria-specific T- and B-cells, which may contribute to postnatal protection from malaria. In utero exposure to malaria also impacts the activation and maturation of fetal antigen presenting cells and innate lymphocytes, which could have implications for global immunity in the infant. Here, we review recent advances in our understanding of how various components of the fetal immune system are altered by in utero exposure to malaria, discuss factors that may tilt the critical balance between tolerance and adaptive immunity, and consider the implications of these findings for malaria prevention strategies.
Collapse
Affiliation(s)
- Margaret E Feeney
- Departments of Pediatrics and Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
39
|
Frascoli M, Coniglio L, Witt R, Jeanty C, Fleck-Derderian S, Myers DE, Lee TH, Keating S, Busch MP, Norris PJ, Tang Q, Cruz G, Barcellos LF, Gomez-Lopez N, Romero R, MacKenzie TC. Alloreactive fetal T cells promote uterine contractility in preterm labor via IFN-γ and TNF-α. Sci Transl Med 2019; 10:10/438/eaan2263. [PMID: 29695455 DOI: 10.1126/scitranslmed.aan2263] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 12/13/2017] [Accepted: 04/05/2018] [Indexed: 12/15/2022]
Abstract
Healthy pregnancy is the most successful form of graft tolerance, whereas preterm labor (PTL) may represent a breakdown in maternal-fetal tolerance. Although maternal immune responses have been implicated in pregnancy complications, fetal immune responses against maternal antigens are often not considered. To examine the fetal immune system in the relevant clinical setting, we analyzed maternal and cord blood in patients with PTL and healthy term controls. We report here that the cord blood of preterm infants has higher amounts of inflammatory cytokines and a greater activation of dendritic cells. Moreover, preterm cord blood is characterized by the presence of a population of central memory cells with a type 1 T helper phenotype, which is absent in term infants, and an increase in maternal microchimerism. T cells from preterm infants mount a robust proliferative, proinflammatory response to maternal antigens compared to term infants yet fail to respond to third-party antigens. Furthermore, we show that T cells from preterm infants stimulate uterine myometrial contractility through interferon-γ and tumor necrosis factor-α. In parallel, we found that adoptive transfer of activated T cells directly into mouse fetuses resulted in pregnancy loss. Our findings indicate that fetal inflammation and rejection of maternal antigens can contribute to the signaling cascade that promotes uterine contractility and that aberrant fetal immune responses should be considered in the pathogenesis of PTL.
Collapse
Affiliation(s)
- Michela Frascoli
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Lacy Coniglio
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Russell Witt
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Cerine Jeanty
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Surgery, University of California, San Francisco, CA 94143, USA
| | | | - Dana E Myers
- Obstetrics and Gynecology, University of California, San Francisco, CA 94143, USA
| | - Tzong-Hae Lee
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Sheila Keating
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Michael P Busch
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Philip J Norris
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Giovanna Cruz
- Division of Epidemiology, Genetic Epidemiology and Genomics Laboratory, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Lisa F Barcellos
- Division of Epidemiology, Genetic Epidemiology and Genomics Laboratory, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)/National Institutes of Health (NIH)/U.S. Department of Health and Human Services (DHHS), Bethesda, MD 20892, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA.,Department of Microbiology, Immunology, and Biochemistry, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)/National Institutes of Health (NIH)/U.S. Department of Health and Human Services (DHHS), Bethesda, MD 20892, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Tippi C MacKenzie
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA. .,Department of Surgery, University of California, San Francisco, CA 94143, USA.,Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
40
|
Saglani S, Gregory LG, Manghera AK, Branchett WJ, Uwadiae F, Entwistle LJ, Oliver RA, Vasiliou JE, Sherburn R, Lui S, Puttur F, Vöhringer D, Walker SA, Buckley J, Grychtol R, Fainardi V, Denney L, Byrne A, von Mutius E, Bush A, Lloyd CM. Inception of early-life allergen-induced airway hyperresponsiveness is reliant on IL-13 +CD4 + T cells. Sci Immunol 2019; 3:3/27/eaan4128. [PMID: 30194239 DOI: 10.1126/sciimmunol.aan4128] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/09/2018] [Indexed: 12/15/2022]
Abstract
Airway hyperresponsiveness (AHR) is a critical feature of wheezing and asthma in children, but the initiating immune mechanisms remain unconfirmed. We demonstrate that both recombinant interleukin-33 (rIL-33) and allergen [house dust mite (HDM) or Alternaria alternata] exposure from day 3 of life resulted in significantly increased pulmonary IL-13+CD4+ T cells, which were indispensable for the development of AHR. In contrast, adult mice had a predominance of pulmonary LinnegCD45+CD90+IL-13+ type 2 innate lymphoid cells (ILC2s) after administration of rIL-33. HDM exposure of neonatal IL-33 knockout (KO) mice still resulted in AHR. However, neonatal CD4creIL-13 KO mice (lacking IL-13+CD4+ T cells) exposed to allergen from day 3 of life were protected from AHR despite persistent pulmonary eosinophilia, elevated IL-33 levels, and IL-13+ ILCs. Moreover, neonatal mice were protected from AHR when inhaled Acinetobacter lwoffii (an environmental bacterial isolate found in cattle farms, which is known to protect from childhood asthma) was administered concurrent with HDM. A. lwoffii blocked the expansion of pulmonary IL-13+CD4+ T cells, whereas IL-13+ ILCs and IL-33 remained elevated. Administration of A. lwoffii mirrored the findings from the CD4creIL-13 KO mice, providing a translational approach for disease protection in early life. These data demonstrate that IL-13+CD4+ T cells, rather than IL-13+ ILCs or IL-33, are critical for inception of allergic AHR in early life.
Collapse
Affiliation(s)
- Sejal Saglani
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK. .,Respiratory Paediatrics, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| | - Lisa G Gregory
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Avneet K Manghera
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - William J Branchett
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Faith Uwadiae
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Lewis J Entwistle
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - R A Oliver
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jessica E Vasiliou
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Rebekah Sherburn
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Stephen Lui
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - F Puttur
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - David Vöhringer
- Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg Wasserturmstrasse 3-5, 91054 Erlangen, Germany
| | - Simone A Walker
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - James Buckley
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Ruth Grychtol
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Valentina Fainardi
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Laura Denney
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Adam Byrne
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Erika von Mutius
- Dr Von Hauner Childrens Hospital of Ludwig-Maximilians-Universitaet of Munich and Helmholtz Zentrum Munchen-German Research Center for Environmental Health, Institute for Asthma and Allergy Prevention, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Andrew Bush
- Respiratory Paediatrics, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| | - Clare M Lloyd
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
41
|
Sureshchandra S, Marshall NE, Messaoudi I. Impact of pregravid obesity on maternal and fetal immunity: Fertile grounds for reprogramming. J Leukoc Biol 2019; 106:1035-1050. [PMID: 31483523 DOI: 10.1002/jlb.3ri0619-181r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022] Open
Abstract
Maternal pregravid obesity results in several adverse health outcomes during pregnancy, including increased risk of gestational diabetes, preeclampsia, placental abruption, and complications at delivery. Additionally, pregravid obesity and in utero exposure to high fat diet have been shown to have detrimental effects on fetal programming, predisposing the offspring to adverse cardiometabolic, endocrine, and neurodevelopmental outcomes. More recently, a deeper appreciation for the modulation of offspring immunity and infectious disease-related outcomes by maternal pregravid obesity has emerged. This review will describe currently available animal models for studying the impact of maternal pregravid obesity on fetal immunity and review the data from clinical and animal model studies. We also examine the burden of pregravid obesity on the maternal-fetal interface and the link between placental and systemic inflammation. Finally, we discuss future studies needed to identify key mechanistic underpinnings that link maternal inflammatory changes and fetal cellular reprogramming events.
Collapse
Affiliation(s)
- Suhas Sureshchandra
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Nicole E Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
42
|
Lloyd CM, Saglani S. Opening the Window of Immune Opportunity: Treating Childhood Asthma. Trends Immunol 2019; 40:786-798. [PMID: 31420279 DOI: 10.1016/j.it.2019.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022]
Abstract
Asthma is an increasingly common childhood disease and although most patients can control their symptoms with medication, a proportion experience life-threatening symptoms. The advent of novel biologic therapies represents a giant leap forward for asthma treatment, but efficacy is rarely tested in children. Recent mechanistic work in mice suggests that early life is a key period for immune development and, therefore, allergen sensitization. Although children with severe asthma experience significant comorbidities and are at increased risk for serious diseases such as chronic obstructive pulmonary disease as adults, no specific investigation into tailored treatment for young children with severe asthma exists. Here, we propose how new information regarding early life immunity could be used to inform modified treatments for children.
Collapse
Affiliation(s)
- Clare M Lloyd
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.
| | - Sejal Saglani
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.
| |
Collapse
|
43
|
Li N, van Unen V, Guo N, Abdelaal T, Somarakis A, Eggermont J, Mahfouz A, Chuva de Sousa Lopes SM, Lelieveldt BPF, Koning F. Early-Life Compartmentalization of Immune Cells in Human Fetal Tissues Revealed by High-Dimensional Mass Cytometry. Front Immunol 2019; 10:1932. [PMID: 31474997 PMCID: PMC6703141 DOI: 10.3389/fimmu.2019.01932] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 07/30/2019] [Indexed: 12/21/2022] Open
Abstract
The human fetal immune system must protect the infant against the sudden exposure to a large variety of pathogens upon birth. While it is known that the fetal immune system develops in sequential waves, relatively little is known about the composition of the innate and adaptive immune system in the tissues. Here, we applied high-dimensional mass cytometry to profile the immune system in human fetal liver, spleen, and intestine. With Hierarchical Stochastic Neighbor Embedding (HSNE) we distinguished 177 distinct immune cell clusters, including both previously identified and novel cell clusters. PCA analysis indicated substantial differences between the compositions of the immune system in the different organs. Through dual t-SNE we identified tissue-specific cell clusters, which were found both in the innate and adaptive compartment. To determine the spatial location of tissue-specific subsets we developed a 31-antibody panel to reveal both the immune compartment and surrounding stromal elements through analysis of snap-frozen tissue samples with imaging mass cytometry. Imaging mass cytometry reconstructed the tissue architecture and allowed both the characterization and determination of the location of the various immune cell clusters within the tissue context. Moreover, it further underpinned the distinctness of the immune system in the tissues. Thus, our results provide evidence for early compartmentalization of the adaptive and innate immune compartment in fetal spleen, liver, and intestine. Together, our data provide a unique and comprehensive overview of the composition and organization of the human fetal immune system in several tissues.
Collapse
Affiliation(s)
- Na Li
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Vincent van Unen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Nannan Guo
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Tamim Abdelaal
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, Netherlands.,Department of Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, Netherlands
| | - Antonios Somarakis
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Jeroen Eggermont
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Ahmed Mahfouz
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Boudewijn P F Lelieveldt
- Department of Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Frits Koning
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
44
|
Halkias J, Rackaityte E, Hillman SL, Aran D, Mendoza VF, Marshall LR, MacKenzie TC, Burt TD. CD161 contributes to prenatal immune suppression of IFNγ-producing PLZF+ T cells. J Clin Invest 2019; 129:3562-3577. [PMID: 31145102 DOI: 10.1172/jci125957] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND While the human fetal immune system defaults to a program of tolerance, there is concurrent need for protective immunity to meet the antigenic challenges encountered after birth. Activation of T cells in utero is associated with the fetal inflammatory response with broad implications for the health of the fetus and of the pregnancy. However, the characteristics of the fetal effector T cells that contribute to this process are largely unknown. METHODS We analyzed primary human fetal lymphoid and mucosal tissues and performed phenotypic, functional, and transcriptional analysis to identify T cells with pro-inflammatory potential. The frequency and function of fetal-specific effector T cells was assessed in the cord blood of infants with localized and systemic inflammatory pathologies and compared to healthy term controls. RESULTS We identified a transcriptionally distinct population of CD4+ T cells characterized by expression of the transcription factor Promyelocytic Leukemia Zinc Finger (PLZF). PLZF+ CD4+ T cells were specifically enriched in the fetal intestine, possessed an effector memory phenotype, and rapidly produced pro-inflammatory cytokines. Engagement of the C-type lectin CD161 on these cells inhibited TCR-dependent production of IFNγ in a fetal-specific manner. IFNγ-producing PLZF+ CD4+ T cells were enriched in the cord blood of infants with gastroschisis, a natural model of chronic inflammation originating from the intestine, as well as in preterm birth, suggesting these cells contribute to fetal systemic immune activation. CONCLUSION Our work reveals a fetal-specific program of protective immunity whose dysregulation is associated with fetal and neonatal inflammatory pathologies.
Collapse
Affiliation(s)
| | - Elze Rackaityte
- Biomedical Sciences Program, UCSF, San Francisco, California, USA
| | - Sara L Hillman
- Maternal and Fetal Medicine Department, Institute for Women's Health, University College London, London, United Kingdom
| | - Dvir Aran
- Institute for Computational Health Sciences, UCSF, San Francisco, California, USA
| | - Ventura F Mendoza
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
| | - Lucy R Marshall
- Division of Infection Immunity and Inflammation, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Tippi C MacKenzie
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA.,Department of Surgery, UCSF, San Francisco, California, USA
| | - Trevor D Burt
- Division of Neonatology, Department of Pediatrics, and.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
| |
Collapse
|
45
|
Affiliation(s)
- Ai Ing Lim
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Oliver J Harrison
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
- Center for Human Immunology, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
46
|
Schreurs RRCE, Baumdick ME, Sagebiel AF, Kaufmann M, Mokry M, Klarenbeek PL, Schaltenberg N, Steinert FL, van Rijn JM, Drewniak A, The SMML, Bakx R, Derikx JPM, de Vries N, Corpeleijn WE, Pals ST, Gagliani N, Friese MA, Middendorp S, Nieuwenhuis EES, Reinshagen K, Geijtenbeek TBH, van Goudoever JB, Bunders MJ. Human Fetal TNF-α-Cytokine-Producing CD4 + Effector Memory T Cells Promote Intestinal Development and Mediate Inflammation Early in Life. Immunity 2019; 50:462-476.e8. [PMID: 30770246 DOI: 10.1016/j.immuni.2018.12.010] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 10/01/2018] [Accepted: 12/05/2018] [Indexed: 11/29/2022]
Abstract
Although the fetal immune system is considered tolerogenic, preterm infants can suffer from severe intestinal inflammation, including necrotizing enterocolitis (NEC). Here, we demonstrate that human fetal intestines predominantly contain tumor necrosis factor-α (TNF-α)+CD4+CD69+ T effector memory (Tem) cells. Single-cell RNA sequencing of fetal intestinal CD4+ T cells showed a T helper 1 phenotype and expression of genes mediating epithelial growth and cell cycling. Organoid co-cultures revealed a dose-dependent, TNF-α-mediated effect of fetal intestinal CD4+ T cells on intestinal stem cell (ISC) development, in which low T cell numbers supported epithelial development, whereas high numbers abrogated ISC proliferation. CD4+ Tem cell frequencies were higher in inflamed intestines from preterm infants with NEC than in healthy infant intestines and showed enhanced TNF signaling. These findings reveal a distinct population of TNF-α-producing CD4+ T cells that promote mucosal development in fetal intestines but can also mediate inflammation upon preterm birth.
Collapse
Affiliation(s)
- Renée R C E Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Martin E Baumdick
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Adrian F Sagebiel
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Michal Mokry
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Paul L Klarenbeek
- Department of Clinical Immunology and Rheumatology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Nicola Schaltenberg
- Department of General, Visceral, and Thoracic Surgery and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Fenja L Steinert
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Jorik M van Rijn
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Agata Drewniak
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Kiadis Pharma B.V., Amsterdam 1105 BV, the Netherlands
| | - Sarah-May M L The
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Roel Bakx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Joep P M Derikx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Niek de Vries
- Department of Clinical Immunology and Rheumatology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Willemijn E Corpeleijn
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Nicola Gagliani
- Department of General, Visceral, and Thoracic Surgery and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm 17176, Sweden
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Sabine Middendorp
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Edward E S Nieuwenhuis
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Johannes B van Goudoever
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam 1081 HV, the Netherlands
| | - Madeleine J Bunders
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany.
| |
Collapse
|
47
|
Li N, van Unen V, Abdelaal T, Guo N, Kasatskaya SA, Ladell K, McLaren JE, Egorov ES, Izraelson M, Chuva de Sousa Lopes SM, Höllt T, Britanova OV, Eggermont J, de Miranda NFCC, Chudakov DM, Price DA, Lelieveldt BPF, Koning F. Memory CD4 + T cells are generated in the human fetal intestine. Nat Immunol 2019; 20:301-312. [PMID: 30664737 PMCID: PMC6420108 DOI: 10.1038/s41590-018-0294-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/04/2018] [Indexed: 12/31/2022]
Abstract
The fetus is thought to be protected from exposure to foreign antigens,
yet CD45RO+ T cells reside in the fetal intestine. Here we combined
functional assays with mass cytometry, single-cell RNA-sequencing and
high-throughput T cell antigen receptor (TCR) sequencing to characterize the
CD4+ T cell compartment in the human fetal intestine. We
identified 22 CD4+ T cell clusters, including naive-like,
regulatory-like and memory-like subpopulations, which were confirmed and further
characterized at the transcriptional level. Memory-like CD4+ T cells
had high expression of Ki-67, indicative of cell division, and CD5, a surrogate
marker of TCR avidity, and produced the cytokines IFN-γ and IL-2. Pathway
analysis revealed a differentiation trajectory associated with cellular
activation and proinflammatory effector functions, and TCR repertoire analysis
indicated clonal expansions, distinct repertoire characteristics and
interconnections between subpopulations of memory-like CD4+ T cells.
Imaging-mass cytometry indicated that memory-like CD4+ T cells
colocalized with antigen-presenting cells. Collectively, these results provide
evidence for the generation of memory-like CD4+ T cells in the human
fetal intestine that is consistent with exposure to foreign antigens.
Collapse
Affiliation(s)
- Na Li
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent van Unen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Tamim Abdelaal
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands.,Department of Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, the Netherlands
| | - Nannan Guo
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Sofya A Kasatskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Centre for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - James E McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Evgeny S Egorov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Mark Izraelson
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Thomas Höllt
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands.,Computer Graphics and Visualization Group, Delft University of Technology, Delft, the Netherlands
| | - Olga V Britanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Jeroen Eggermont
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Dmitriy M Chudakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Centre for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia.,Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Department of Molecular Technologies, Pirogov Russian National Research Medical University, Moscow, Russia.,MiLaboratory LLC, Skolkovo Innovation Centre, Moscow, Russia.,Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK.,Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - Boudewijn P F Lelieveldt
- Department of Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits Koning
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
48
|
Abstract
The growth and maturity of the peripheral immune system and subsequent development of pulmonary immunity in early life is dictated by host, environmental and microbial factors. Dysregulation during the critical window of immune development in the postnatal years results in disease which impacts on lifelong lung health. Asthma is a common disease in childhood and is often preceded by wheezing illnesses during the preschool years. However, the mechanisms underlying development of wheeze and how and why only some children progress to asthma is unknown. Human studies to date have generally focused on peripheral immune development, with little assessment of local tissue pathology in young children. Moreover, mechanisms underlying the interactions between inflammation and tissue repair at mucosal surfaces in early life remain unknown. Disappointingly, mechanistic studies in mice have predominantly used adult models. This review will consider the aspects of the neonatal immune system which might contribute to the development of early life wheezing disorders and asthma, and discuss the external environmental factors which may influence this process.
Collapse
Affiliation(s)
- Clare M Lloyd
- Inflammation, Repair & Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK
| | - Sejal Saglani
- Inflammation, Repair & Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, Royal Brompton Harefield NHS Foundation Trust, London, UK
| |
Collapse
|
49
|
Scheible KM, Emo J, Laniewski N, Baran AM, Peterson DR, Holden-Wiltse J, Bandyopadhyay S, Straw AG, Huyck H, Ashton JM, Tripi KS, Arul K, Werner E, Scalise T, Maffett D, Caserta M, Ryan RM, Reynolds AM, Ren CL, Topham DJ, Mariani TJ, Pryhuber GS. T cell developmental arrest in former premature infants increases risk of respiratory morbidity later in infancy. JCI Insight 2018; 3:96724. [PMID: 29467329 DOI: 10.1172/jci.insight.96724] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/17/2018] [Indexed: 12/31/2022] Open
Abstract
The inverse relationship between gestational age at birth and postviral respiratory morbidity suggests that infants born preterm (PT) may miss a critical developmental window of T cell maturation. Despite a continued increase in younger PT survivors with respiratory complications, we have limited understanding of normal human fetal T cell maturation, how ex utero development in premature infants may interrupt normal T cell development, and whether T cell development has an effect on infant outcomes. In our longitudinal cohort of 157 infants born between 23 and 42 weeks of gestation, we identified differences in T cells present at birth that were dependent on gestational age and differences in postnatal T cell development that predicted respiratory outcome at 1 year of age. We show that naive CD4+ T cells shift from a CD31-TNF-α+ bias in mid gestation to a CD31+IL-8+ predominance by term gestation. Former PT infants discharged with CD31+IL8+CD4+ T cells below a range similar to that of full-term born infants were at an over 3.5-fold higher risk for respiratory complications after NICU discharge. This study is the first to our knowledge to identify a pattern of normal functional T cell development in later gestation and to associate abnormal T cell development with health outcomes in infants.
Collapse
Affiliation(s)
| | | | | | - Andrea M Baran
- Department of Biostatistics and Computational Biology, and
| | | | | | | | - Andrew G Straw
- Department of Biostatistics and Computational Biology, and
| | | | | | | | - Karan Arul
- Undergraduate Campus, University of Rochester, Rochester, New York, USA
| | | | | | | | | | - Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anne Marie Reynolds
- Department of Pediatrics, State University of New York, University at Buffalo, Buffalo, New York, USA
| | - Clement L Ren
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | |
Collapse
|
50
|
Lawrence SM, Wynn JL. Chorioamnionitis, IL-17A, and fetal origins of neurologic disease. Am J Reprod Immunol 2017; 79:e12803. [PMID: 29271527 DOI: 10.1111/aji.12803] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/28/2017] [Indexed: 12/23/2022] Open
Abstract
The Centers for Disease Control and Prevention estimate that 1 in 323 infants have cerebral palsy. Highly correlated to intrauterine infection and inflammation, the incidence of cerebral palsy has remained constant over the last few decades despite significant advances in neonatal intensive care including improved ventilator techniques, surfactant therapy, maternal steroid administration, and use of intrapartum empiric antimicrobials. Recent advances in our understanding of immune responses to infection and inflammation have identified the cytokine IL-17A as a crucial component of early proinflammatory mediators that cause brain injury associated with neurologic impairment. Remarkably, maternal inflammatory responses to in utero inflammation and infection can also lead to potentially debilitating neurologic conditions in the offspring, which often become clinically apparent during childhood and/or early adulthood. This review details the role of IL-17A in fetal and maternal proinflammatory responses that lead to fetal brain injury and neurologic sequelae, including cerebral palsy. Recent findings regarding the role of maternal inflammatory responses in the development of childhood and adult neurologic conditions, such as autism, schizophrenia, and multiple sclerosis, will also be highlighted.
Collapse
Affiliation(s)
- Shelley M Lawrence
- College of Medicine, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of California, San Diego, CA, USA.,Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, CA, USA
| | - James L Wynn
- College of Medicine, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Florida, Gainesville, FL, USA.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|