1
|
Croucher NJ, Campo JJ, Le TQ, Pablo JV, Hung C, Teng AA, Turner C, Nosten F, Bentley SD, Liang X, Turner P, Goldblatt D. Genomic and panproteomic analysis of the development of infant immune responses to antigenically-diverse pneumococci. Nat Commun 2024; 15:355. [PMID: 38191887 PMCID: PMC10774285 DOI: 10.1038/s41467-023-44584-2] [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: 08/03/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is a nasopharyngeal commensal and respiratory pathogen. This study characterises the immunoglobulin G (IgG) repertoire recognising pneumococci from birth to 24 months old (mo) in a prospectively-sampled cohort of 63 children using a panproteome array. IgG levels are highest at birth, due to transplacental transmission of maternal antibodies. The subsequent emergence of responses to individual antigens exhibit distinct kinetics across the cohort. Stable differences in the strength of individuals' responses, correlating with maternal IgG concentrations, are established by 6 mo. By 12 mo, children develop unique antibody profiles that are boosted by re-exposure. However, some proteins only stimulate substantial responses in adults. Integrating genomic data on nasopharyngeal colonisation demonstrates rare pneumococcal antigens can elicit strong IgG levels post-exposure. Quantifying such responses to the diverse core loci (DCL) proteins is complicated by cross-immunity between variants. In particular, the conserved N terminus of DCL protein zinc metalloprotease B provokes the strongest early IgG responses. DCL proteins' ability to inhibit mucosal immunity likely explains continued pneumococcal carriage despite hosts' polyvalent antibody repertoire. Yet higher IgG levels are associated with reduced incidence, and severity, of pneumonia, demonstrating the importance of the heterogeneity in response strength and kinetics across antigens and individuals.
Collapse
Affiliation(s)
- Nicholas J Croucher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, W12 0BZ, UK.
| | - Joseph J Campo
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Timothy Q Le
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Jozelyn V Pablo
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Christopher Hung
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Andy A Teng
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Claudia Turner
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, 9V54+8FQ, Cambodia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - François Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, 63110, Thailand
| | - Stephen D Bentley
- Parasites & Microbes, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Xiaowu Liang
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Paul Turner
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, 9V54+8FQ, Cambodia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health, University College London, London, WC1N 1EH, UK
| |
Collapse
|
2
|
Hu Y, Hu Q, Li Y, Lu L, Xiang Z, Yin Z, Kabelitz D, Wu Y. γδ T cells: origin and fate, subsets, diseases and immunotherapy. Signal Transduct Target Ther 2023; 8:434. [PMID: 37989744 PMCID: PMC10663641 DOI: 10.1038/s41392-023-01653-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 11/23/2023] Open
Abstract
The intricacy of diseases, shaped by intrinsic processes like immune system exhaustion and hyperactivation, highlights the potential of immune renormalization as a promising strategy in disease treatment. In recent years, our primary focus has centered on γδ T cell-based immunotherapy, particularly pioneering the use of allogeneic Vδ2+ γδ T cells for treating late-stage solid tumors and tuberculosis patients. However, we recognize untapped potential and optimization opportunities to fully harness γδ T cell effector functions in immunotherapy. This review aims to thoroughly examine γδ T cell immunology and its role in diseases. Initially, we elucidate functional differences between γδ T cells and their αβ T cell counterparts. We also provide an overview of major milestones in γδ T cell research since their discovery in 1984. Furthermore, we delve into the intricate biological processes governing their origin, development, fate decisions, and T cell receptor (TCR) rearrangement within the thymus. By examining the mechanisms underlying the anti-tumor functions of distinct γδ T cell subtypes based on γδTCR structure or cytokine release, we emphasize the importance of accurate subtyping in understanding γδ T cell function. We also explore the microenvironment-dependent functions of γδ T cell subsets, particularly in infectious diseases, autoimmune conditions, hematological malignancies, and solid tumors. Finally, we propose future strategies for utilizing allogeneic γδ T cells in tumor immunotherapy. Through this comprehensive review, we aim to provide readers with a holistic understanding of the molecular fundamentals and translational research frontiers of γδ T cells, ultimately contributing to further advancements in harnessing the therapeutic potential of γδ T cells.
Collapse
Affiliation(s)
- Yi Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Qinglin Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Zheng Xiang
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zhinan Yin
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany.
| | - Yangzhe Wu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China.
| |
Collapse
|
3
|
Liang S, Yao J, Liu D, Rao L, Chen X, Wang Z. Harnessing Nanomaterials for Cancer Sonodynamic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211130. [PMID: 36881527 DOI: 10.1002/adma.202211130] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/12/2023] [Indexed: 06/18/2023]
Abstract
Immunotherapy has made remarkable strides in cancer therapy over the past decade. However, such emerging therapy still suffers from the low response rates and immune-related adverse events. Various strategies have been developed to overcome these serious challenges. Therein, sonodynamic therapy (SDT), as a non-invasive treatment, has received ever-increasing attention especially in the treatment of deep-seated tumors. Significantly, SDT can effectively induce immunogenic cell death to trigger systemic anti-tumor immune response, termed sonodynamic immunotherapy. The rapid development of nanotechnology has revolutionized SDT effects with robust immune response induction. As a result, more and more innovative nanosonosensitizers and synergistic treatment modalities are established with superior efficacy and safe profile. In this review, the recent advances in cancer sonodynamic immunotherapy are summarized with a particular emphasis on how nanotechnology can be explored to harness SDT for amplifying anti-tumor immune response. Moreover, the current challenges in this field and the prospects for its clinical translation are also presented. It is anticipated that this review can provide rational guidance and facilitate the development of nanomaterials-assisted sonodynamic immunotherapy, helping to pave the way for next-generation cancer therapy and eventually achieve a durable response in patients.
Collapse
Affiliation(s)
- Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jianjun Yao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
4
|
Povroznik JM, Akhter H, Vance JK, Annamanedi M, Dziadowicz SA, Wang L, Divens AM, Hu G, Robinson CM. Interleukin-27-dependent transcriptome signatures during neonatal sepsis. Front Immunol 2023; 14:1124140. [PMID: 36891292 PMCID: PMC9986606 DOI: 10.3389/fimmu.2023.1124140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Human newborns exhibit increased vulnerability and risk of mortality from infection that is consistent with key differences in the innate and adaptive immune responses relative to those in adult cells. We have previously shown an increase in the immune suppressive cytokine, IL-27, in neonatal cells and tissues from mice and humans. In a murine model of neonatal sepsis, mice deficient in IL-27 signaling exhibit reduced mortality, increased weight gain, and better control of bacteria with reduced systemic inflammation. To explore a reprogramming of the host response in the absence of IL-27 signaling, we profiled the transcriptome of the neonatal spleen during Escherichia coli-induced sepsis in wild-type (WT) and IL-27Rα-deficient (KO) mice. We identified 634 genes that were differentially expressed, and those most upregulated in WT mice were associated with inflammation, cytokine signaling, and G protein coupled receptor ligand binding and signaling. These genes failed to increase in the IL-27Rα KO mice. We further isolated an innate myeloid population enriched in macrophages from the spleens of control and infected WT neonates and observed similar changes in gene expression aligned with changes in chromatin accessibility. This supports macrophages as an innate myeloid population contributing to the inflammatory profile in septic WT pups. Collectively, our findings highlight the first report of improved pathogen clearance amidst a less inflammatory environment in IL-27Rα KO. This suggests a direct relationship between IL-27 signaling and bacterial killing. An improved response to infection that is not reliant upon heightened levels of inflammation offers new promise to the potential of antagonizing IL-27 as a host-directed therapy for neonates.
Collapse
Affiliation(s)
- Jessica M. Povroznik
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Halima Akhter
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Jordan K. Vance
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Madhavi Annamanedi
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Lei Wang
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Ashley M. Divens
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Gangqing Hu
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Cory M. Robinson
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| |
Collapse
|
5
|
Miller NW, Seman BG, Akers SM, Povroznik JM, Brundage K, Fang W, Robinson CM. The impact of opioid exposure during pregnancy on the human neonatal immune profile. Pediatr Res 2022; 92:1566-1574. [PMID: 35288639 PMCID: PMC8920062 DOI: 10.1038/s41390-022-02014-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/19/2022] [Accepted: 02/04/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The increasing magnitude of the opioid crisis and rising rates of neonatal abstinence syndrome (NAS) diagnoses highlight the need for increased research into how maternal substance use during pregnancy can impact the neonatal immune profile and its functionality. We hypothesized that neonates with opioid exposure would have reduced proportions of some immune cells, an anti-inflammatory cytokine profile, reduced T cell proliferation, and monocyte bacterial killing activity compared to the control population. METHODS The present study compares immune cell populations, inflammatory and anti-inflammatory cytokine and chemokine levels in the serum, and monocyte and T cell functional activity using umbilical cord samples from neonates with known opioid exposure during gestation and from control neonates without known exposure. RESULTS Our findings demonstrated a significant reduction in neutrophils, decreased levels of inflammatory cytokines in the serum, and reduced IL-2 production during in vitro CD4+ T cell proliferation in neonates exposed to opioids compared to controls. The neutrophil findings were supported by retrospective analysis of an extended network of deidentified patient records. CONCLUSIONS This study is the first of its kind to evaluate differences in neonatal immunity as a result of opioid exposure in the human population that will inform continued mechanistic studies. IMPACT The opioid epidemic has become a public health crisis in the United States, and the corresponding incidence of neonatal abstinence syndrome (NAS) have risen accordingly. New research is required to understand the short and long-term health impacts of opioid exposure to the neonate. This is the first human study to investigate the immunologic profile and functionality in neonates with known opioid exposure in utero. The abundance of neutrophils and the ratio of neutrophils to lymphocytes is significantly reduced along with inflammatory cytokines and chemokines following opioid exposure during pregnancy. The immune profile in opioid-exposed neonates may promote susceptibility to infection.
Collapse
Affiliation(s)
- Nicholas W Miller
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Brittany G Seman
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Stephen M Akers
- Department of Pediatrics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Jessica M Povroznik
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA
| | - Kathleen Brundage
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Wei Fang
- West Virginia Clinical and Translational Science Institute, Morgantown, WV, 26506, USA
| | - Cory M Robinson
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA.
| |
Collapse
|
6
|
Maheshwari A. The Phylogeny, Ontogeny, and Organ-specific Differentiation of Macrophages in the Developing Intestine. NEWBORN (CLARKSVILLE, MD.) 2022; 1:340-355. [PMID: 36698382 PMCID: PMC9872774 DOI: 10.5005/jp-journals-11002-0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Macrophages are large highly motile phagocytic leukocytes that appear early during embryonic development and have been conserved during evolution. The developmental roles of macrophages were first described nearly a century ago, at about the time these cells were being identified as central effectors in phagocytosis and elimination of microbes. Since then, we have made considerable progress in understanding the development of various subsets of macrophages and the diverse roles these cells play in both physiology and disease. This article reviews the phylogeny and the ontogeny of macrophages with a particular focus on the gastrointestinal tract, and the role of these mucosal macrophages in immune surveillance, innate immunity, homeostasis, tissue remodeling, angiogenesis, and repair of damaged tissues. We also discuss the importance of these macrophages in the inflammatory changes in neonatal necrotizing enterocolitis (NEC). This article presents a combination of our own peer-reviewed clinical and preclinical studies, with an extensive review of the literature using the databases PubMed, EMBASE, and Scopus.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Global Newborn Society, Clarksville, Maryland, United States of America
| |
Collapse
|
7
|
Antimicrobial Peptides in Early-Life Host Defense, Perinatal Infections, and Necrotizing Enterocolitis—An Update. J Clin Med 2022; 11:jcm11175074. [PMID: 36079001 PMCID: PMC9457252 DOI: 10.3390/jcm11175074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
Host defense against early-life infections such as chorioamnionitis, neonatal sepsis, or necrotizing enterocolitis (NEC) relies primarily on innate immunity, in which antimicrobial peptides (AMPs) play a major role. AMPs that are important for the fetus and neonate include α and β defensins, cathelicidin LL-37, antiproteases (elafin, SLPI), and hepcidin. They can be produced by the fetus or neonate, the placenta, chorioamniotic membranes, recruited neutrophils, and milk-protein ingestion or proteolysis. They possess antimicrobial, immunomodulating, inflammation-regulating, and tissue-repairing properties. AMPs are expressed as early as the 13th week and increase progressively through gestation. Limited studies are available on AMP expression and levels in the fetus and neonate. Nevertheless, existing evidence supports the role of AMPs in pathogenesis of chorioamnionitis, neonatal sepsis, and NEC, and their association with disease severity. This suggests a potential role of AMPs in diagnosis, prevention, prognosis, and treatment of sepsis and NEC. Herein, we present an overview of the antimicrobial and immunomodulating properties of human AMPs, their sources in the intrauterine environment, fetus, and neonate, and their changes during pre- and post-natal infections and NEC. We also discuss emerging data regarding the potential utility of AMPs in early-life infections, as diagnostic or predictive biomarkers and as therapeutic alternatives or adjuncts to antibiotic therapy considering the increase of antibiotic resistance in neonatal intensive care units.
Collapse
|
8
|
TLR agonists induce sustained IgG to hemagglutinin stem and modulate T cells following newborn vaccination. NPJ Vaccines 2022; 7:102. [PMID: 36038596 PMCID: PMC9424286 DOI: 10.1038/s41541-022-00523-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
The newborn immune system is characterized by diminished immune responses that leave infants vulnerable to virus-mediated disease and make vaccination more challenging. Optimal vaccination strategies for influenza A virus (IAV) in newborns should result in robust levels of protective antibodies, including those with broad reactivity to combat the variability in IAV strains across seasons. The stem region of the hemagglutinin (HA) molecule is a target of such antibodies. Using a nonhuman primate model, we investigate the capacity of newborns to generate and maintain antibodies to the conserved stem region following vaccination. We find adjuvanting an inactivated vaccine with the TLR7/8 agonist R848 is effective in promoting sustained HA stem-specific IgG. Unexpectedly, HA stem-specific antibodies were generated with a distinct kinetic pattern compared to the overall response. Administration of R848 was associated with increased influenza-specific T follicular helper cells as well as Tregs with a less suppressive phenotype, suggesting adjuvant impacts multiple cell types that have the potential to contribute to the HA-stem response.
Collapse
|
9
|
Sarraf TR, Sen M. Wnt5A signaling supports antigen processing and CD8 T cell activation. Front Immunol 2022; 13:960060. [PMID: 36091060 PMCID: PMC9459031 DOI: 10.3389/fimmu.2022.960060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Antigen processing and antigen-specific CD8 T cell activation form part and parcel of cell-mediated immunity to infections. Yet, several lacunae remain in our understanding of how antigen processing and CD8 T cell response are coordinated. In this study, using mouse bone marrow-derived dendritic cells (BMDC) as antigen-presenting cells and Ovalbumin (OVA)/DQ-Ovalbumin (DQ-OVA) as model antigen we demonstrated that Wnt5A signaling in BMDC supports antigen processing/presentation and concomitant CD8 T cell activation through regulation of actin and proteasome dynamics. Recombinant Wnt5A conditioning of BMDC and associated actin assembly facilitated DQ-OVA processing, which was inhibited by the proteasome inhibitor MG132. Moreover, Wnt5A depletion led to a significant reduction in OVA processing and presentation. Impaired DQ-OVA processing in Wnt5A depleted BMDC correlated with altered dynamics of both actin and the proteasome regulator PA28α-PA28β, and reduced association of DQ-OVA with actin and proteasome subunits. Inhibited OVA processing/presentation in the Wnt5A depleted BMDC also resulted in subdued activation of OVA-sensitized CD8 T cells in co-culture with the BMDC. In concurrence with these findings, we demonstrated reduced OVA processing and impaired CD8 T cell response to OVA immunization in Wnt5A heterozygous mice lacking a copy of the Wnt5A gene in comparison to the wild-type cohorts. Taken together, our results reveal a crucial requirement of Wnt5A signaling in antigen processing/presentation and CD8 T cell activation, thus unveiling a vital regulatory node of cell-mediated immunity, unidentified thus far.
Collapse
|
10
|
Jalbert E, Ghosh T, Smith C, Amaral FR, Mussi-Pinhata MM, Weinberg A. Impaired functionality of antigen presenting cells in HIV- exposed uninfected infants in the first six months of life. Front Immunol 2022; 13:960313. [PMID: 36032106 PMCID: PMC9411519 DOI: 10.3389/fimmu.2022.960313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
HIV-exposed uninfected infants (HEU) have increased morbidity and mortality due to infections in the first 6 months of life that tapers down to 2 years of life. The underlying immunologic defects remain undefined. We investigated antigen-presenting cells (APC) by comparing the phenotype of unstimulated APC, responses to toll-like receptor (TLR) stimulation, and ability to activate natural killer (NK) cells in 24 HEU and 64 HIV-unexposed infants (HUU) at 1-2 days of life (birth) and 28 HEU and 45 HUU at 6 months of life. At birth, unstimulated APC showed higher levels of activation and cytokine production in HEU than HUU and stimulation with TLR agonists revealed lower expression of inflammatory cytokines and activation markers, but similar expression of IL10 regulatory cytokine, in APC from HEU compared to HUU. Differences were still present at 6 months of life. From birth to 6 months, APC underwent extensive phenotypic and functional changes in HUU and minimal changes in HEU. TLR stimulation also generated lower NK cell expression of CD69 and/or IFNγ in HEU compared with HUU at birth and 6 months. In vitro experiments showed that NK IFNγ expression depended on APC cytokine secretion in response to TLR stimulation. Ex vivo IL10 supplementation decreased APC-mediated NK cell activation measured by IFNγ expression. We conclude that APC maturation was stunted or delayed in the first 6 months of life in HEU compared with HUU. Deficient inflammatory APC responses and/or the imbalance between inflammatory and regulatory responses in HEU may play an important role in their increased susceptibility to severe infections.
Collapse
Affiliation(s)
- Emilie Jalbert
- Department of Pediatrics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Tusharkanti Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Christiana Smith
- Department of Pediatrics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Fabiana R. Amaral
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marisa M. Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Adriana Weinberg
- Department of Pediatrics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine and Pathology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: Adriana Weinberg,
| |
Collapse
|
11
|
Ucciferri CC, Dunn SE. Effect of puberty on the immune system: Relevance to multiple sclerosis. Front Pediatr 2022; 10:1059083. [PMID: 36533239 PMCID: PMC9755749 DOI: 10.3389/fped.2022.1059083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
Puberty is a dynamic period marked by changing levels of sex hormones, the development of secondary sexual characteristics and reproductive maturity. This period has profound effects on various organ systems, including the immune system. The critical changes that occur in the immune system during pubertal onset have been shown to have implications for autoimmune conditions, including Multiple Sclerosis (MS). MS is rare prior to puberty but can manifest in children after puberty. This disease also has a clear female preponderance that only arises following pubertal onset, highlighting a potential role for sex hormones in autoimmunity. Early onset of puberty has also been shown to be a risk factor for MS. The purpose of this review is to overview the evidence that puberty regulates MS susceptibility and disease activity. Given that there is a paucity of studies that directly evaluate the effects of puberty on the immune system, we also discuss how the immune system is different in children and mice of pre- vs. post-pubertal ages and describe how gonadal hormones may regulate these immune mechanisms. We present evidence that puberty enhances the expression of co-stimulatory molecules and cytokine production by type 2 dendritic cells (DC2s) and plasmacytoid dendritic cells (pDCs), increases T helper 1 (Th1), Th17, and T follicular helper immunity, and promotes immunoglobulin (Ig)G antibody production. Overall, this review highlights how the immune system undergoes a functional maturation during puberty, which has the potential to explain the higher prevalence of MS and other autoimmune diseases seen in adolescence.
Collapse
Affiliation(s)
- Carmen C Ucciferri
- Department of Immunology, The University of Toronto, Toronto, ON, Canada
| | - Shannon E Dunn
- Department of Immunology, The University of Toronto, Toronto, ON, Canada.,Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
| |
Collapse
|
12
|
Mthembu N, Ikwegbue P, Brombacher F, Hadebe S. Respiratory Viral and Bacterial Factors That Influence Early Childhood Asthma. FRONTIERS IN ALLERGY 2021; 2:692841. [PMID: 35387053 PMCID: PMC8974778 DOI: 10.3389/falgy.2021.692841] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
Asthma is a chronic respiratory condition characterised by episodes of shortness of breath due to reduced airway flow. The disease is triggered by a hyperreactive immune response to innocuous allergens, leading to hyper inflammation, mucus production, changes in structural cells lining the airways, and airway hyperresponsiveness. Asthma, although present in adults, is considered as a childhood condition, with a total of about 6.2 million children aged 18 and below affected globally. There has been progress in understanding asthma heterogeneity in adults, which has led to better patient stratification and characterisation of multiple asthma endotypes with distinct, but overlapping inflammatory features. The asthma inflammatory profile in children is not well-defined and heterogeneity of the disease is less described. Although many factors such as genetics, food allergies, antibiotic usage, type of birth, and cigarette smoke exposure can influence asthma development particularly in children, respiratory infections are thought to be the major contributing factor in poor lung function and onset of the disease. In this review, we focus on viral and bacterial respiratory infections in the first 10 years of life that could influence development of asthma in children. We also review literature on inflammatory immune heterogeneity in asthmatic children and how this overlaps with early lung development, poor lung function and respiratory infections. Finally, we review animal studies that model early development of asthma and how these studies could inform future therapies and better understanding of this complex disease.
Collapse
Affiliation(s)
- Nontobeko Mthembu
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Paul Ikwegbue
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Immunology, Health Science Faculty, International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Sabelo Hadebe
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
13
|
Lineweaver CH, Bussey KJ, Blackburn AC, Davies PCW. Cancer progression as a sequence of atavistic reversions. Bioessays 2021; 43:e2000305. [PMID: 33984158 DOI: 10.1002/bies.202000305] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/27/2022]
Abstract
It has long been recognized that cancer onset and progression represent a type of reversion to an ancestral quasi-unicellular phenotype. This general concept has been refined into the atavistic model of cancer that attempts to provide a quantitative analysis and testable predictions based on genomic data. Over the past decade, support for the multicellular-to-unicellular reversion predicted by the atavism model has come from phylostratigraphy. Here, we propose that cancer onset and progression involve more than a one-off multicellular-to-unicellular reversion, and are better described as a series of reversionary transitions. We make new predictions based on the chronology of the unicellular-eukaryote-to-multicellular-eukaryote transition. We also make new predictions based on three other evolutionary transitions that occurred in our lineage: eukaryogenesis, oxidative phosphorylation and the transition to adaptive immunity. We propose several modifications to current phylostratigraphy to improve age resolution to test these predictions. Also see the video abstract here: https://youtu.be/3unEu5JYJrQ.
Collapse
Affiliation(s)
- Charles H Lineweaver
- Planetary Science Institute, Research School of Astronomy and Astrophysics & Research School of Earth Sciences, The Australian National University, Canberra, ACT, Australia.,Mt Stromlo Observatory, Canberra, ACT, Australia
| | - Kimberly J Bussey
- Beyond Center for Fundamental Concepts in Science, Arizona State University, Tempe, Arizona, USA.,Precision Medicine, Midwestern University, Glendale, Arizona, USA
| | - Anneke C Blackburn
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Paul C W Davies
- Beyond Center for Fundamental Concepts in Science, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
14
|
Lauro R, Irrera N, Eid AH, Bitto A. Could Antigen Presenting Cells Represent a Protective Element during SARS-CoV-2 Infection in Children? Pathogens 2021; 10:476. [PMID: 33920011 PMCID: PMC8071032 DOI: 10.3390/pathogens10040476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 12/12/2022] Open
Abstract
Antigen Presenting Cells (APC) are immune cells that recognize, process, and present antigens to lymphocytes. APCs are among the earliest immune responders against an antigen. Thus, in patients with COVID-19, a disease caused by the newly reported SARS-CoV-2 virus, the role of APCs becomes increasingly important. In this paper, we dissect the role of these cells in the fight against SARS-CoV-2. Interestingly, this virus appears to cause a higher mortality among adults than children. This may suggest that the immune system, particularly APCs, of children may be different from that of adults, which may then explain differences in immune responses between these two populations, evident as different pathological outcome. However, the underlying molecular mechanisms that differentiate juvenile from other APCs are not well understood. Whether juvenile APCs are one reason why children are less susceptible to SARS-CoV-2 requires much attention. The goal of this review is to examine the role of APCs, both in adults and children. The molecular mechanisms governing APCs, especially against SARS-CoV-2, may explain the differential immune responsiveness in the two populations.
Collapse
Affiliation(s)
- Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (R.L.); (N.I.)
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (R.L.); (N.I.)
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (R.L.); (N.I.)
| |
Collapse
|
15
|
Beijnen EMS, van Haren SD. Vaccine-Induced CD8 + T Cell Responses in Children: A Review of Age-Specific Molecular Determinants Contributing to Antigen Cross-Presentation. Front Immunol 2020; 11:607977. [PMID: 33424857 PMCID: PMC7786054 DOI: 10.3389/fimmu.2020.607977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Infections are most common and most severe at the extremes of age, the young and the elderly. Vaccination can be a key approach to enhance immunogenicity and protection against pathogens in these vulnerable populations, who have a functionally distinct immune system compared to other age groups. More than 50% of the vaccine market is for pediatric use, yet to date vaccine development is often empiric and not tailored to molecular distinctions in innate and adaptive immune activation in early life. With modern vaccine development shifting from whole-cell based vaccines to subunit vaccines also comes the need for formulations that can elicit a CD8+ T cell response when needed, for example, by promoting antigen cross-presentation. While our group and others have identified many cellular and molecular determinants of successful activation of antigen-presenting cells, B cells and CD4+ T cells in early life, much less is known about the ontogeny of CD8+ T cell induction. In this review, we summarize the literature pertaining to the frequency and phenotype of newborn and infant CD8+ T cells, and any evidence of induction of CD8+ T cells by currently licensed pediatric vaccine formulations. In addition, we review the molecular determinants of antigen cross-presentation on MHC I and successful CD8+ T cell induction and discuss potential distinctions that can be made in children. Finally, we discuss recent advances in development of novel adjuvants and provide future directions for basic and translational research in this area.
Collapse
Affiliation(s)
- Elisabeth M. S. Beijnen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Simon D. van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Challenges for the Newborn Immune Response to Respiratory Virus Infection and Vaccination. Vaccines (Basel) 2020; 8:vaccines8040558. [PMID: 32987691 PMCID: PMC7712002 DOI: 10.3390/vaccines8040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
The initial months of life reflect an extremely challenging time for newborns as a naïve immune system is bombarded with a large array of pathogens, commensals, and other foreign entities. In many instances, the immune response of young infants is dampened or altered, resulting in increased susceptibility and disease following infection. This is the result of both qualitative and quantitative changes in the response of multiple cell types across the immune system. Here we provide a review of the challenges associated with the newborn response to respiratory viral pathogens as well as the hurdles and advances for vaccine-mediated protection.
Collapse
|
17
|
Rackaityte E, Halkias J. Mechanisms of Fetal T Cell Tolerance and Immune Regulation. Front Immunol 2020; 11:588. [PMID: 32328065 PMCID: PMC7160249 DOI: 10.3389/fimmu.2020.00588] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/13/2020] [Indexed: 12/19/2022] Open
Abstract
The developing human fetus generates both tolerogenic and protective immune responses in response to the unique requirements of gestation. Thus, a successful human pregnancy depends on a fine balance between two opposing immunological forces: the semi-allogeneic fetus learns to tolerate both self- and maternal- antigens and, in parallel, develops protective immunity in preparation for birth. This critical window of immune development bridges prenatal immune tolerance with the need for postnatal environmental protection, resulting in a vulnerable neonatal period with heightened risk of infection. The fetal immune system is highly specialized to mediate this transition and thus serves a different function from that of the adult. Adaptive immune memory is already evident in the fetal intestine. Fetal T cells with pro-inflammatory potential are born in a tolerogenic environment and are tightly controlled by both cell-intrinsic and -extrinsic mechanisms, suggesting that compartmentalization and specialization, rather than immaturity, define the fetal immune system. Dysregulation of fetal tolerance generates an inflammatory response with deleterious effects to the pregnancy. This review aims to discuss the recent advances in our understanding of the cellular and molecular composition of fetal adaptive immunity and the mechanisms that govern T cell development and function. We also discuss the tolerance promoting environment that impacts fetal immunity and the consequences of its breakdown. A greater understanding of fetal mechanisms of immune activation and regulation has the potential to uncover novel paradigms of immune balance which may be leveraged to develop therapies for transplantation, autoimmune disease, and birth-associated inflammatory pathologies.
Collapse
Affiliation(s)
- Elze Rackaityte
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Joanna Halkias
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA, United States
| |
Collapse
|
18
|
Type I IFN ineffectively activates neonatal dendritic cells limiting respiratory antiviral T-cell responses. Mucosal Immunol 2020; 13:371-380. [PMID: 31797910 PMCID: PMC7044048 DOI: 10.1038/s41385-019-0234-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/04/2023]
Abstract
Insufficient T-cell responses contribute to the increased burden of viral respiratory disease in infancy. Neonatal dendritic cells (DCs) often provide defective activation of pathogen-specific T cells through mechanisms that are incompletely understood, which hinders vaccine design for this vulnerable age group. Enhancing our characterization of neonatal DC sub-specialization and function is therefore critical to developing their potential for immunomodulation of T-cell responses. In this study, we engineered respiratory syncytial virus (RSV) to express a model protein, ovalbumin, to track antigen-presenting DCs in vivo. We found that murine neonatal conventional DC1s (cDC1s) efficiently migrated and presented RSV-derived antigen, challenging the paradigm that neonatal DCs are globally immature. In a key observation, however, we discovered that during infection neonatal cDC1s presenting viral antigen were unable to upregulate costimulatory molecules in response to type I interferons (IFN-I), contributing to poor antiviral T-cell responses. Importantly, we showed that the deficient response to IFN-I was also exhibited by human neonatal cDC1s, independent of infection. These findings reveal a functionally distinct response to IFN-I by neonatal cDC1s that may leave young infants susceptible to viral infections, and provide a new target for exploration, in light of failed efforts to design neonatal RSV vaccines.
Collapse
|
19
|
Lack of Activation Marker Induction and Chemokine Receptor Switch in Human Neonatal Myeloid Dendritic Cells in Response to Human Respiratory Syncytial Virus. J Virol 2019; 93:JVI.01216-19. [PMID: 31484754 DOI: 10.1128/jvi.01216-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 08/25/2019] [Indexed: 02/05/2023] Open
Abstract
Respiratory syncytial virus (RSV) infects and causes disease in infants and reinfects with reduced disease throughout life without significant antigenic change. In contrast, reinfection by influenza A virus (IAV) largely requires antigenic change. The adaptive immune response depends on antigen presentation by dendritic cells (DC), which may be too immature in young infants to induce a fully protective immune response against RSV reinfections. We therefore compared the ability of RSV and IAV to activate primary human cord blood (CB) and adult blood (AB) myeloid DC (mDC). While RSV and IAV infected with similar efficiencies, RSV poorly induced maturation and cytokine production in CB and AB mDC. This difference between RSV and IAV was more profound in CB mDC. While IAV activated CB mDC to some extent, RSV did not induce CB mDC to increase the maturation markers CD38 and CD86 or CCR7, which directs DC migration to lymphatic tissue. Low CCR7 surface expression was associated with high expression of CCR5, which keeps DC in inflamed peripheral tissues. To evaluate a possible inhibition by RSV, we subjected RSV-inoculated AB mDC to secondary IAV inoculation. While RSV-inoculated AB mDC responded to secondary IAV inoculation by efficiently upregulating activation markers and cytokine production, IAV-induced CCR5 downregulation was slightly inhibited in cells exhibiting robust RSV infection. Thus, suboptimal stimulation and weak and mostly reversible inhibition seem to be responsible for inefficient mDC activation by RSV. The inefficient mDC stimulation and immunological immaturity in young infants may contribute to reduced immune responses and incomplete protection against RSV reinfection.IMPORTANCE Respiratory syncytial virus (RSV) causes disease early in life and can reinfect symptomatically throughout life without undergoing significant antigenic change. In contrast, reinfection by influenza A virus (IAV) requires antigenic change. The adaptive immune response depends on antigen presentation by dendritic cells (DC). We used myeloid DC (mDC) from cord blood and adult blood donors to evaluate whether immunological immaturity contributes to the inability to mount a fully protective immune response to RSV. While IAV induced some activation and chemokine receptor switching in cord blood mDC, RSV did not. This appeared to be due to a lack of activation and a weak and mostly reversible inhibition of DC functions. Both viruses induced a stronger activation of mDC from adults than mDC from cord blood. Thus, inefficient stimulation of mDC by RSV and immunological immaturity may contribute to reduced immune responses and increased susceptibility to RSV disease and reinfection in young infants.
Collapse
|
20
|
Catania VD, Boscarelli A, Lauriti G, Morini F, Zani A. Risk Factors for Surgical Site Infection in Neonates: A Systematic Review of the Literature and Meta-Analysis. Front Pediatr 2019; 7:101. [PMID: 30984722 PMCID: PMC6449628 DOI: 10.3389/fped.2019.00101] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose: Surgical site infections (SSI) contribute to postoperative morbidity and mortality in children. Our aim was to evaluate the prevalence and identify risk factors for SSI in neonates. Methods: Using a defined strategy, three investigators searched articles on neonatal SSI published since 2000. Studies on neonates and/or patients admitted to neonatal intensive care unit following cervical/thoracic/abdominal surgery were included. Risk factors were identified from comparative studies. Meta-analysis was conducted according to PRISMA guidelines using RevMan 5.3. Data are (mean ± SD) prevalence. Results: Systematic review-of 885 abstracts screened, 48 studies (27,760 neonates) were included. The incidence of SSI was 5.6% (1,564 patients). SSI was more frequent in males (61.8%), premature babies (77.4%), and following gastrointestinal surgery (95.4%). Meta-analysis-10 comparative studies (16,442 neonates; 946 SSI 5.7%) showed that predictive factors for SSI development were gestational age, birth weight, age at surgery, length of surgical procedure, number of procedure per patient, length of preoperative hospital stay, and preoperative sepsis. Conversely, preoperative antibiotic use was not significantly associated with development of SSI. Conclusions: Younger neonates and those undergoing abdominal procedures are at higher risk for SSI. Given the lack of evidence-based literature, prospective studies may help determine the risk factors for SSI in neonates.
Collapse
Affiliation(s)
- Vincenzo Davide Catania
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Alessandro Boscarelli
- Neonatal Surgery Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giuseppe Lauriti
- Department of Pediatric Surgery, Spirito Santo Hospital and G. d'Annunzio University of Chieti and Pescara, Chieti, Italy
| | - Francesco Morini
- Neonatal Surgery Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Augusto Zani
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Wu J, Gong RL, Hu QF, Chen XT, Zhao W, Chen TX. Immunoregulatory effect of human β-defensin 1 on neonatal cord blood monocyte-derived dendritic cells and T cells. Mol Immunol 2019; 109:99-107. [PMID: 30921683 DOI: 10.1016/j.molimm.2019.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/11/2019] [Accepted: 03/15/2019] [Indexed: 12/31/2022]
Abstract
The relationship between breastfeeding and infant health has been well elucidated in past decades. Our previous study has shown that human β-defensin 1 (hBD-1) in human breast milk plays a protective role in reducing the incidence of upper respiratory infection in infants younger than 6 months. In the present study, we aim to reveal the mechanism underlying the protective role of hBD-1 by focusing on its immunoregulatory function in neonates. Cord blood (CB) from newborns' umbilical cords, which can simulate many of the neonatal symptoms, was used to study the immunomodulatory role of hBD-1 in neonates in vitro. Our results showed that hBD-1 promotes the GM-CSF- and IL-4-driven differentiation of neonatal umbilical CB monocytes to immature dendritic cells (DCs) and the final maturation of CB monocyte-derived DCs (moDCs) induced by LPS but not inflammatory cytokine production. In addition, hBD-1 inhibits apoptosis in neonatal moDCs through CCR6, which might be a possible mechanism of the hBD-1-induced phenotypes in moDCs. Furthermore, we found that hBD-1 promotes the proliferation and activation, but not the maturation, of neonatal CB CD4 + T cells. These results extend the immunoregulatory effects of hBD-1 and provide a potential mechanism for the protective role of hBD-1 in early infants, which will inform the development of infant nutrition, novel vaccines and anti-infective strategies in the future.
Collapse
Affiliation(s)
- Jing Wu
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ruo-Lan Gong
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qing-Feng Hu
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xu-Ting Chen
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Wei Zhao
- Division of Allergy and Immunology, Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Tong-Xin Chen
- Department of Rheumatology/Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
22
|
Lopez BS, Hurley DJ, Giancola S, Giguère S, Felippe MJB, Hart KA. The effect of age on foal monocyte-derived dendritic cell (MoDC) maturation and function after exposure to killed bacteria. Vet Immunol Immunopathol 2019; 210:38-45. [PMID: 30947978 DOI: 10.1016/j.vetimm.2018.11.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/25/2018] [Accepted: 11/01/2018] [Indexed: 02/06/2023]
Abstract
Neonatal foals are uniquely susceptible to certain infections early in life. Dendritic cells (DC) are vital in the transition between the innate and adaptive immune response to infection, but DC biology in foals is not fully characterized. Monocyte-derived DC represent a suitable in vitro model similar to DC that differentiate from monocytes recruited from circulation. We hypothesized that foal monocyte-derived DC (MoDC) would exhibit age-dependent phenotypic and functional differences compared to adult horse MoDC. MoDC generated from 9 horses (collected once) and from 8 foals (collected at 1, 7, and 30 days-of-age) were exposed to killed whole cell Escherichia coli or Staphylococcus aureus bacteria. MoDC expression of MHC class II (MHC class-II), CD86, and CD14 were measured by flow cytometry, and supernatant cytokine concentrations of IL-4, IL-17, IFN-γ, and IL-10 were quantified with a validated immunoassay. The percentage of MoDC expressing MHC class-II and CD86 was lower and CD14 was higher for cells generated from 1-day-old foals compared to cells generated from adult horses (P < 0.0001). Bacterial exposure increased the percentage of cells expressing CD86 at all ages (P < 0.0001). Bacteria-exposed MoDC from 1-day-old foals produced significantly less IL-4, IL-17, and IFN-γ than adult MoDC produced in response to bacterial exposure (P ≤ 0.04). Following bacterial exposure, foal MoDC phenotype and cytokine secretion were different than those of mature horses. These differences could reduce the ability of foals to generate a protective immune response against bacterial infection.
Collapse
Affiliation(s)
- Brina S Lopez
- From the Department of Large Animal Medicine, 2200 College Station Road, University of Georgia College of Veterinary Medicine, Athens, GA, 30602, USA
| | - David J Hurley
- From the Department of Population Health, 2200 College Station Road, University of Georgia College of Veterinary Medicine, Athens, GA, 30602, USA
| | - Shyla Giancola
- From the Department of Large Animal Medicine, 2200 College Station Road, University of Georgia College of Veterinary Medicine, Athens, GA, 30602, USA
| | - Steeve Giguère
- From the Department of Large Animal Medicine, 2200 College Station Road, University of Georgia College of Veterinary Medicine, Athens, GA, 30602, USA
| | - M Julia B Felippe
- Equine Immunology Laboratory, Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Kelsey A Hart
- From the Department of Large Animal Medicine, 2200 College Station Road, University of Georgia College of Veterinary Medicine, Athens, GA, 30602, USA.
| |
Collapse
|
23
|
Verhoeven D. Immunometabolism and innate immunity in the context of immunological maturation and respiratory pathogens in young children. J Leukoc Biol 2019; 106:301-308. [DOI: 10.1002/jlb.mr0518-204rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- David Verhoeven
- Department of Veterinary Microbiology and Preventative MedicineIowa State University Ames Iowa USA
| |
Collapse
|
24
|
Vreman S, Auray G, Savelkoul HFJ, Rebel A, Summerfield A, Stockhofe-Zurwieden N. Neonatal porcine blood derived dendritic cell subsets show activation after TLR2 or TLR9 stimulation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 84:361-370. [PMID: 29555549 DOI: 10.1016/j.dci.2018.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/07/2018] [Accepted: 03/15/2018] [Indexed: 06/08/2023]
Abstract
The present study investigated the innate immune response in vitro to determine porcine neonate responses with Toll-like receptor (TLR)2 ligand (Pam3Cys) or TLR9 ligand (CpG) and compared these with adults. We identified the same phenotypically defined dendritic cell (DC) subsets and DC proportions in porcine neonate and adult blood by flow cytometry, which were plasmacytoid DCs (pDCs): CD14-CD4+CD172a+CADM1-) and conventional DCs (cDCs), being further divided into a cDC1 (CD14-CD4-CD172alowCADM1+) and a cDC2 (CD14-CD4-CD172a+CADM1+) subset. With neonatal cells, the TLR2 ligand induced a stronger TNF expression in monocytes and pDCs, and a stronger CD80/86 upregulation in cDC1, when compared to adult cells. Furthermore, in neonatal mononuclear cells TLR9 ligand was more potent at inducing IL12p40 mRNA expression. These results indicate clear responses of porcine neonatal antigen presenting cells after TLR2 and TLR9 stimulation, suggesting that corresponding ligands could be promising candidates for neonatal adjuvant application.
Collapse
Affiliation(s)
- Sandra Vreman
- Wageningen Bioveterinary Research, Wageningen University & Research, P.O. Box 29703, 2502 LS, The Hague, The Netherlands.
| | - Gael Auray
- Institute of Virology and Immunology, Sensemattstrasse 293, 3147, Mittelhausern, Switzerland; Department of Infectious Diseases and Pathobiology, University of Bern, Switzerland
| | - Huub F J Savelkoul
- Cell Biology & Immunology Group, Wageningen University & Research P.O. Box 338, 6700 HA, Wageningen, The Netherlands
| | - Annemarie Rebel
- Wageningen Bioveterinary Research, Wageningen University & Research, P.O. Box 29703, 2502 LS, The Hague, The Netherlands
| | - Artur Summerfield
- Institute of Virology and Immunology, Sensemattstrasse 293, 3147, Mittelhausern, Switzerland; Department of Infectious Diseases and Pathobiology, University of Bern, Switzerland
| | - Norbert Stockhofe-Zurwieden
- Wageningen Bioveterinary Research, Wageningen University & Research, P.O. Box 29703, 2502 LS, The Hague, The Netherlands
| |
Collapse
|
25
|
Surendran N, Simmons A, Pichichero ME. TLR agonist combinations that stimulate Th type I polarizing responses from human neonates. Innate Immun 2018; 24:240-251. [PMID: 29673285 PMCID: PMC6830928 DOI: 10.1177/1753425918771178] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Each year millions of neonates die due to vaccine preventable infectious diseases. Our study seeks to develop novel neonatal vaccines and improve immunogenicity of early childhood vaccines by incorporating TLR agonist-adjuvant combinations that overcome the inherent neonatal Th2 bias and stimulate Th1 polarizing response from neonatal APCs. We systematically stimulated cord blood mononuclear cells with single and multiple combinations of TLR agonists and measured levels of IL-12p70, IFN-γ, IFN-α, IL-10, IL-13, TNF-α, IL-6 and IL-1β from cell culture supernatants. APC-specific surface expression levels of costimulatory markers CD40, CD83 and PD-L1 were assessed by flow cytometry. Whole blood assays were included to account for the effect of plasma inhibitory factors and APC intracellular TNF-α and IL-12p40 secretions were measured. We found robust Th1 polarizing IL-12p70, IFN-γ and IFN-α responses when cord blood APCs were stimulated with TLR agonist combinations that contained Poly I:C, Monophosphoryl Lipid A (MPLA) or R848. Addition of class A CpG oligonucleotide (ODN) to Th1 polarizing TLR agonist combinations significantly reduced cord blood IL-12p70 and IFN-γ levels and addition of a TLR2 agonist induced significantly high Th2 polarizing IL-13. Multi-TLR agonist combinations that included R848 induced lower inhibitory PD-L1 expression on cord blood classical dendritic cells than CpG ODN-containing combinations. Incorporation of combination adjuvants containing TLR3, TLR4 and TLR7/8 agonists to neonatal vaccines may be an effective strategy to overcome neonatal Th2 bias.
Collapse
Affiliation(s)
- Naveen Surendran
- Center for Infectious Diseases and Immunology, Rochester General Hospital Research Institute, Rochester, NY, USA
| | - Andrea Simmons
- Center for Infectious Diseases and Immunology, Rochester General Hospital Research Institute, Rochester, NY, USA
| | - Michael E Pichichero
- Center for Infectious Diseases and Immunology, Rochester General Hospital Research Institute, Rochester, NY, USA
| |
Collapse
|
26
|
Brook B, Harbeson D, Ben-Othman R, Viemann D, Kollmann TR. Newborn susceptibility to infection vs. disease depends on complex in vivo interactions of host and pathogen. Semin Immunopathol 2017; 39:615-625. [PMID: 29098373 DOI: 10.1007/s00281-017-0651-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 09/01/2017] [Indexed: 02/07/2023]
Abstract
The burden of newborn infectious disease has long been recognized as the highest across the entire human life span. The precise underlying cause is unfortunately still far from clear. A substantial body of data derived mostly from in vitro experimentation indicates "lower" host immune responses in early vs. adult life and is briefly summarized within this review. However, emerging data derived mostly from in vivo experimentation reveal that the newborn host also exhibits an exuberant immune and inflammatory response following infection when compared to the adult. In this context, it is important to emphasize that "infection" does not equate "infectious disease," as for many infections it is the host response to the infection that causes disease. This simple insight readily arranges existing evidence into cause-effect relationships that explain much of the increase in clinical suffering from infection in early life. We here briefly summarize the evidence in support of this paradigm and highlight the important implications it has for efforts to ameliorate the suffering and dying from infection in early life.
Collapse
Affiliation(s)
- Byron Brook
- Department of Experimental Medicine, University of British Columbia, UBC, BCCHRI A5-175, 950 W 28th Ave, Vancouver, BC, V5Z4H4, Canada
| | - Danny Harbeson
- Department of Experimental Medicine, University of British Columbia, UBC, BCCHRI A5-175, 950 W 28th Ave, Vancouver, BC, V5Z4H4, Canada
| | - Rym Ben-Othman
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, Canada
| | - Dorothee Viemann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Tobias R Kollmann
- Department of Experimental Medicine, University of British Columbia, UBC, BCCHRI A5-175, 950 W 28th Ave, Vancouver, BC, V5Z4H4, Canada. .,Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
27
|
Surgical site infections in neonates are independently associated with longer hospitalizations. J Perinatol 2017; 37:1130-1134. [PMID: 28749483 DOI: 10.1038/jp.2017.107] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/17/2017] [Accepted: 05/22/2017] [Indexed: 11/09/2022]
Abstract
OBJECTIVE There is limited data characterizing the risk and impact of surgical site infection (SSI) in neonates; this makes it difficult to identify factors that increase neonatal SSI risk and to determine how SSI affects outcomes in this special population. STUDY DESIGN The American College of Surgeons National Surgical Improvement Program Pediatric (NSQIP-P) collected data on children undergoing surgery at children's hospitals from 2012 to 2014. Neonates undergoing general surgical procedures were characterized with regard to demographic characteristics and comorbidities. Perioperative variables such as wound class, type of surgery and length of operation were also evaluated. RESULTS Seven thousand three hundred and seventy-nine neonates were identified in the NSQIP-P participant user file. The overall SSI rate was 2.6%. Only wound class and length of surgery were significantly associated with SSI. Furthermore, SSI was independently associated with longer length of stay, even after adjusting for covariates. CONCLUSIONS This is the largest study to date analyzing SSI in neonates. We found that perioperative variables have a more significant impact on SSI than patient factors, suggesting that operation-related characteristics are influencing SSI. Furthermore, neonates with SSI are more likely to have prolonged hospitalizations even after adjusting for patient comorbidities.
Collapse
|
28
|
Georgountzou A, Papadopoulos NG. Postnatal Innate Immune Development: From Birth to Adulthood. Front Immunol 2017; 8:957. [PMID: 28848557 PMCID: PMC5554489 DOI: 10.3389/fimmu.2017.00957] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/26/2017] [Indexed: 12/20/2022] Open
Abstract
It is well established that adaptive immune responses are deficient in early life, contributing to increased mortality and morbidity. The developmental trajectories of different components of innate immunity are only recently being explored. Individual molecules, cells, or pathways of innate recognition and signaling, within different compartments/anatomical sites, demonstrate variable maturation patterns. Despite some discrepancies among published data, valuable information is emerging, showing that the developmental pattern of cytokine responses during early life is age and toll-like receptor specific, and may be modified by genetic and environmental factors. Interestingly, specific environmental exposures have been linked both to innate function modifications and the occurrence of chronic inflammatory disorders, such as respiratory allergies. As these conditions are on the rise, our knowledge on innate immune development and its modulating factors needs to be expanded. Improved understanding of the sequence of events associated with disease onset and persistence will lead toward meaningful interventions. This review describes the state-of-the-art on normal postnatal innate immune ontogeny and highlights research areas that are currently explored or should be further addressed.
Collapse
Affiliation(s)
- Anastasia Georgountzou
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos G Papadopoulos
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Division of Infection, Inflammation and Respiratory Medicine, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
Hardy A, Benford D, Halldorsson T, Jeger MJ, Knutsen HK, More S, Naegeli H, Noteborn H, Ockleford C, Ricci A, Rychen G, Schlatter JR, Silano V, Solecki R, Turck D, Bresson JL, Dusemund B, Gundert-Remy U, Kersting M, Lambré C, Penninks A, Tritscher A, Waalkens-Berendsen I, Woutersen R, Arcella D, Court Marques D, Dorne JL, Kass GE, Mortensen A. Guidance on the risk assessment of substances present in food intended for infants below 16 weeks of age. EFSA J 2017; 15:e04849. [PMID: 32625502 PMCID: PMC7010120 DOI: 10.2903/j.efsa.2017.4849] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Following a request from the European Commission to EFSA, the EFSA Scientific Committee (SC) prepared a guidance for the risk assessment of substances present in food intended for infants below 16 weeks of age. In its approach to develop this guidance, the EFSA SC took into account, among others, (i) an exposure assessment based on infant formula as the only source of nutrition; (ii) knowledge of organ development in human infants, including the development of the gut, metabolic and excretory capacities, the brain and brain barriers, the immune system, the endocrine and reproductive systems; (iii) the overall toxicological profile of the substance identified through the standard toxicological tests, including critical effects; (iv) the relevance for the human infant of the neonatal experimental animal models used. The EFSA SC notes that during the period from birth up to 16 weeks, infants are expected to be exclusively fed on breast milk and/or infant formula. The EFSA SC views this period as the time where health-based guidance values for the general population do not apply without further considerations. High infant formula consumption per body weight is derived from 95th percentile consumption. The first weeks of life is the time of the highest relative consumption on a body weight basis. Therefore, when performing an exposure assessment, the EFSA SC proposes to use the high consumption value of 260 mL/kg bw per day. A decision tree approach is proposed that enables a risk assessment of substances present in food intended for infants below 16 weeks of age. The additional information needed when testing substances present in food for infants below 16 weeks of age and the approach to be taken for the risk assessment are on a case-by-case basis, depending on whether the substance is added intentionally to food and is systemically available.
Collapse
|
30
|
Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J. A critical assessment of the "sterile womb" and "in utero colonization" hypotheses: implications for research on the pioneer infant microbiome. MICROBIOME 2017; 5:48. [PMID: 28454555 PMCID: PMC5410102 DOI: 10.1186/s40168-017-0268-4] [Citation(s) in RCA: 614] [Impact Index Per Article: 87.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/21/2017] [Indexed: 05/10/2023]
Abstract
After more than a century of active research, the notion that the human fetal environment is sterile and that the neonate's microbiome is acquired during and after birth was an accepted dogma. However, recent studies using molecular techniques suggest bacterial communities in the placenta, amniotic fluid, and meconium from healthy pregnancies. These findings have led many scientists to challenge the "sterile womb paradigm" and propose that microbiome acquisition instead begins in utero, an idea that would fundamentally change our understanding of gut microbiota acquisition and its role in human development. In this review, we provide a critical assessment of the evidence supporting these two opposing hypotheses, specifically as it relates to (i) anatomical, immunological, and physiological characteristics of the placenta and fetus; (ii) the research methods currently used to study microbial populations in the intrauterine environment; (iii) the fecal microbiome during the first days of life; and (iv) the generation of axenic animals and humans. Based on this analysis, we argue that the evidence in support of the "in utero colonization hypothesis" is extremely weak as it is founded almost entirely on studies that (i) used molecular approaches with an insufficient detection limit to study "low-biomass" microbial populations, (ii) lacked appropriate controls for contamination, and (iii) failed to provide evidence of bacterial viability. Most importantly, the ability to reliably derive axenic animals via cesarean sections strongly supports sterility of the fetal environment in mammals. We conclude that current scientific evidence does not support the existence of microbiomes within the healthy fetal milieu, which has implications for the development of clinical practices that prevent microbiome perturbations after birth and the establishment of future research priorities.
Collapse
Affiliation(s)
- Maria Elisa Perez-Muñoz
- Department of Agriculture, Food and Nutritional Sciences, 4-126 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta T6G 2E1 Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1 Canada
- Department of Pediatrics, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1 Canada
| | - Amanda E. Ramer-Tait
- Department of Food Science and Technology, 260 Food Innovation Center, University of Nebraska-Lincoln, 1901 N 21st Street, Lincoln, Nebraska 68588-6205 USA
| | - Jens Walter
- Department of Agriculture, Food and Nutritional Sciences, 4-126 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta T6G 2E1 Canada
- Department of Biological Sciences, 7-142 Katz Group Centre, University of Alberta, Edmonton, Alberta T6G 2E1 Canada
| |
Collapse
|
31
|
Heninger AK, Eugster A, Kuehn D, Buettner F, Kuhn M, Lindner A, Dietz S, Jergens S, Wilhelm C, Beyerlein A, Ziegler AG, Bonifacio E. A divergent population of autoantigen-responsive CD4+T cells in infants prior to β cell autoimmunity. Sci Transl Med 2017; 9:9/378/eaaf8848. [DOI: 10.1126/scitranslmed.aaf8848] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 12/30/2016] [Indexed: 12/14/2022]
|
32
|
Dinan TG, Cryan JF. Microbes, Immunity, and Behavior: Psychoneuroimmunology Meets the Microbiome. Neuropsychopharmacology 2017; 42:178-192. [PMID: 27319972 PMCID: PMC5143479 DOI: 10.1038/npp.2016.103] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/26/2016] [Accepted: 06/13/2016] [Indexed: 02/07/2023]
Abstract
There is now a large volume of evidence to support the view that the immune system is a key communication pathway between the gut and brain, which plays an important role in stress-related psychopathologies and thus provides a potentially fruitful target for psychotropic intervention. The gut microbiota is a complex ecosystem with a diverse range of organisms and a sophisticated genomic structure. Bacteria within the gut are estimated to weigh in excess of 1 kg in the adult human and the microbes within not only produce antimicrobial peptides, short chain fatty acids, and vitamins, but also most of the common neurotransmitters found in the human brain. That the microbial content of the gut plays a key role in immune development is now beyond doubt. Early disruption of the host-microbe interplay can have lifelong consequences, not just in terms of intestinal function but in distal organs including the brain. It is clear that the immune system and nervous system are in continuous communication in order to maintain a state of homeostasis. Significant gaps in knowledge remain about the effect of the gut microbiota in coordinating the immune-nervous systems dialogue. However, studies using germ-free animals, infective models, prebiotics, probiotics, and antibiotics have increased our understanding of the interplay. Early life stress can have a lifelong impact on the microbial content of the intestine and permanently alter immune functioning. That early life stress can also impact adult psychopathology has long been appreciated in psychiatry. The challenge now is to fully decipher the molecular mechanisms that link the gut microbiota, immune, and central nervous systems in a network of communication that impacts behavior patterns and psychopathology, to eventually translate these findings to the human situation both in health and disease. Even at this juncture, there is evidence to pinpoint key sites of communication where gut microbial interventions either with drugs or diet or perhaps fecal microbiota transplantation may positively impact mental health.
Collapse
Affiliation(s)
- Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Psychiatry & Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
33
|
|
34
|
Senthamaraikannan P, Presicce P, Rueda CM, Maneenil G, Schmidt AF, Miller LA, Waites KB, Jobe AH, Kallapur SG, Chougnet CA. Intra-amniotic Ureaplasma parvum-Induced Maternal and Fetal Inflammation and Immune Responses in Rhesus Macaques. J Infect Dis 2016; 214:1597-1604. [PMID: 27601620 DOI: 10.1093/infdis/jiw408] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/22/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Although Ureaplasma species are the most common organisms associated with prematurity, their effects on the maternal and fetal immune system remain poorly characterized. METHODS Rhesus macaque dams at approximately 80% gestation were injected intra-amniotically with 107 colony-forming units of Ureaplasma parvum or saline (control). Fetuses were delivered surgically 3 or 7 days later. We performed comprehensive assessments of inflammation and immune effects in multiple fetal and maternal tissues. RESULTS Although U. parvum grew well in amniotic fluid, there was minimal chorioamnionitis. U. parvum colonized the fetal lung, but fetal systemic microbial invasion was limited. Fetal lung inflammation was mild, with elevations in CXCL8, tumor necrosis factor (TNF) α, and CCL2 levels in alveolar washes at day 7. Inflammation was not detected in the fetal brain. Significantly, U. parvum decreased regulatory T cells (Tregs) and activated interferon γ production in these Tregs in the fetus. It was detected in uterine tissue by day 7 and induced mild inflammation and increased expression of connexin 43, a gap junction protein involved with labor. CONCLUSIONS U. parvum colonized the amniotic fluid and caused uterine inflammation, but without overt chorioamnionitis. It caused mild fetal lung inflammation but had a more profound effect on the fetal immune system, decreasing Tregs and polarizing them toward a T-helper 1 phenotype.
Collapse
Affiliation(s)
| | | | - Cesar M Rueda
- Perinatal Institute.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Ohio
| | - Gunlawadee Maneenil
- Perinatal Institute.,Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | | | - Lisa A Miller
- California National Primate Research Center.,Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis
| | - Ken B Waites
- Department of Pathology, University of Alabama at Birmingham
| | | | | | - Claire A Chougnet
- Perinatal Institute.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Ohio
| |
Collapse
|
35
|
Sins JW, Biemond BJ, van den Bersselaar SM, Heijboer H, Rijneveld AW, Cnossen MH, Kerkhoffs JLH, van Meurs AH, von Ronnen F, Zalpuri S, de Rijke YB, Ellen van der Schoot C, de Haas M, van der Bom JG, Fijnvandraat K. Early occurrence of red blood cell alloimmunization in patients with sickle cell disease. Am J Hematol 2016; 91:763-9. [PMID: 27102719 DOI: 10.1002/ajh.24397] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/14/2022]
Abstract
Red blood cell (RBC) alloimmunization is a major complication of transfusion therapy in sickle cell disease (SCD). Identification of high-risk patients is hampered by lack of studies that take the cumulative transfusion exposure into account. In this retrospective cohort study among previously non-transfused SCD patients in the Netherlands, we aimed to elucidate the association between the cumulative transfusion exposure, first alloimmunization and independent risk factors. A total of 245 patients received 11 952 RBC units. Alloimmunization occurred in 43 patients (18%), half of them formed their first alloantibody before the 8th unit. In patients with exposure to non-extended matched transfusions (ABO and RhD) the cumulative alloimmunization risk increased up to 35% after 60 transfused units. This was significantly higher compared to a general transfused population (HR 6.6, CI 4.2-10.6). Receiving the first transfusion after the age of 5 was an independent risk factor for alloimmunization (HR 2.3, CI 1.0-5.1). Incidental, episodic transfusions in comparison to chronic scheme transfusions (HR 2.3, CI 0.9-6.0), and exposure to non-extended matched units in comparison to extended matching (HR 2.0, CI 0.9-4.6) seemed to confer a higher alloimmunization risk. The majority of first alloantibodies are formed after minor transfusion exposure, substantiating suggestions of a responder phenotype in SCD and stressing the need for risk factor identification. In this study, older age at first transfusion, episodic transfusions and non-extended matched transfusions appeared to be risk factors for alloimmunization. Am. J. Hematol. 91:763-769, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joep W.R. Sins
- Department of Pediatric Hematology; Emma Children's Hospital, Academic Medical Center; Amsterdam The Netherlands
- Department of Hematology; Academic Medical Center; Amsterdam The Netherlands
| | - Bart J. Biemond
- Department of Hematology; Academic Medical Center; Amsterdam The Netherlands
| | - Sil M. van den Bersselaar
- Department of Pediatric Hematology; Emma Children's Hospital, Academic Medical Center; Amsterdam The Netherlands
- Department of Hematology; Academic Medical Center; Amsterdam The Netherlands
| | - H. Heijboer
- Department of Pediatric Hematology; Emma Children's Hospital, Academic Medical Center; Amsterdam The Netherlands
| | - Anita W. Rijneveld
- Department of Hematology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Marjon H. Cnossen
- Department of Pediatric Hematology; Erasmus MC, University Medical Center-Sophia Children's Hospital; Rotterdam The Netherlands
| | | | | | - F.B. von Ronnen
- Department of Pediatric Hematology; Emma Children's Hospital, Academic Medical Center; Amsterdam The Netherlands
- Department of Hematology; Academic Medical Center; Amsterdam The Netherlands
| | - Saurabh Zalpuri
- Center for Clinical Transfusion; Sanquin Blood Supply Foundation; Leiden and Department of Clinical Epidemiology; Leiden University Medical Center; Leiden The Netherlands
| | - Yolanda B. de Rijke
- Department of Clinical Chemistry; Erasmus MC, University Medical Center Rotterdam; The Netherlands
| | - C. Ellen van der Schoot
- Department of Experimental Immunohematology; Sanquin Blood Supply Foundation; Amsterdam The Netherlands
| | - Masja de Haas
- Department of Experimental Immunohematology; Sanquin Blood Supply Foundation; Amsterdam The Netherlands
| | - Johanna G. van der Bom
- Center for Clinical Transfusion; Sanquin Blood Supply Foundation; Leiden and Department of Clinical Epidemiology; Leiden University Medical Center; Leiden The Netherlands
| | - Karin Fijnvandraat
- Department of Pediatric Hematology; Emma Children's Hospital, Academic Medical Center; Amsterdam The Netherlands
- Unit of Clinical Transfusion Medicine; Sanquin Blood Supply Foundation; Amsterdam The Netherlands
| |
Collapse
|
36
|
Saliganti V, Kapila R, Kapila S. Consumption of probioticLactobacillus rhamnosus(MTCC: 5897) containing fermented milk plays a key role in development of the immune system in newborn mice during the suckling-weaning transition. Microbiol Immunol 2016; 60:261-7. [DOI: 10.1111/1348-0421.12342] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/12/2015] [Accepted: 11/16/2015] [Indexed: 11/27/2022]
Affiliation(s)
- Vamshi Saliganti
- Animal Biochemistry Division; National Dairy Research Institute; Karnal 132001 India
| | - Rajeev Kapila
- Animal Biochemistry Division; National Dairy Research Institute; Karnal 132001 India
| | - Suman Kapila
- Animal Biochemistry Division; National Dairy Research Institute; Karnal 132001 India
| |
Collapse
|
37
|
Pan H, Gazarian A, Dubernard JM, Belot A, Michallet MC, Michallet M. Transplant Tolerance Induction in Newborn Infants: Mechanisms, Advantages, and Potential Strategies. Front Immunol 2016; 7:116. [PMID: 27092138 PMCID: PMC4823304 DOI: 10.3389/fimmu.2016.00116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/14/2016] [Indexed: 12/26/2022] Open
Abstract
Although several tolerance induction protocols have been successfully implemented in adult renal transplantation, no tolerance induction approach has, as yet, been defined for solid organ transplantations in young infants. Pediatric transplant recipients have a pressing demand for the elaboration of tolerance induction regimens. Indeed, since they display a longer survival time, they are exposed to a higher level of risks linked to long-term immunosuppression (IS) and to chronic rejection. Interestingly, central tolerance induction may be of great interest in newborns, because of their immunological immaturity and the important role of the thymus at this early stage in life. The present review aims to clarify mechanisms and strategies of tolerance induction in these immunologically premature recipients. We first introduce the discovery and mechanisms of neonatal tolerance in murine experimental models and subsequently analyze tolerance induction in human newborn infants. Hematopoietic mixed chimerism in neonates is also discussed based on in utero hematopoietic stem cell (HSC) transplant studies. Then, we review the recent advances in tolerance induction approaches in adults, including the infusion of HSCs associated with less toxic conditioning regimens, regulatory T cells/facilitating cells/mesenchymal stem cells transplantation, costimulatory blockade, and thymus manipulation. Finally, IS withdrawal in pediatric solid organ transplant is discussed. In conclusion, the establishment of transplant tolerance induction in infants is promising and deserves further investigations. Future studies could focus on the selection of patients, on less toxic conditioning regimens, and on biomarkers for IS minimization or withdrawal.
Collapse
Affiliation(s)
- Hua Pan
- Chair of Transplantation, VetAgro Sup-Campus Vétérinaire de Lyon, Marcy l'Etoile, France; Plastic and Reconstructive Surgery Department, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Aram Gazarian
- Chair of Transplantation, VetAgro Sup-Campus Vétérinaire de Lyon, Marcy l'Etoile, France; Department of Hand Surgery, Clinique du Parc, Lyon, France
| | - Jean-Michel Dubernard
- Chair of Transplantation, VetAgro Sup-Campus Vétérinaire de Lyon, Marcy l'Etoile, France; Department of Transplantation, Hôpital Edouard Herriot, Lyon, France
| | - Alexandre Belot
- International Center for Infectiology Research (CIRI), Université de Lyon , Lyon , France
| | - Marie-Cécile Michallet
- Chair of Transplantation, VetAgro Sup-Campus Vétérinaire de Lyon, Marcy l'Etoile, France; Cancer Research Center Lyon (CRCL), UMR INSERM 1052 CNRS 5286, Centre Leon Berard, Lyon, France
| | - Mauricette Michallet
- Chair of Transplantation, VetAgro Sup-Campus Vétérinaire de Lyon, Marcy l'Etoile, France; Department of Hematology, Centre Hospitalier Lyon-Sud, Pierre Benite, France
| |
Collapse
|
38
|
Verhoeven D, Perry S, Pryharski K. Control of influenza infection is impaired by diminished interferon-γ secretion by CD4 T cells in the lungs of toddler mice. J Leukoc Biol 2016; 100:203-12. [PMID: 26823488 DOI: 10.1189/jlb.4a1014-497rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/04/2016] [Indexed: 11/24/2022] Open
Abstract
Respiratory viral infections, such as influenza, can lead to delayed viral clearance in toddlers, possibly exacerbating disease morbidity. We hypothesized that defective CD4 T cells in toddlers may contribute to a failure to clear virus at a similar rate to adults. Thus, we developed a young mouse model to examine potential divergent responses between toddlers and adults. We determined that young mice (toddler mice, 21 d old) were actively generating and recruiting effector/memory T cells, whereas memory populations were firmly established in older, adult mice (8-10 wk old). We infected toddler and adult mice with influenza A/PR8/34 (H1N1) and found young mice had elevated morbidity, as measured by enhanced weight loss and lower partial pressure of oxygen levels, throughout the infection, thus, modeling the higher morbidity observed in children (<2 y old) during infection. Early viral loads were comparable to adult mice, but toddler mice failed to clear virus by 10 d postinfection. This delayed clearance corresponded to poor lung recruitment of CD4 T cells, lower antiviral T cell responses, and lower B cell/antibodies in the lungs. Mechanistically, diminished interferon-γ was detected in the lungs of toddler mice throughout the infection and corresponded to intrinsic, rather than extrinsic, CD4 T cell limitations in interferon-γ transcription. Moreover, defects in interferon-γ production appeared downstream from signal transducer and activator of transcription 4 in the interleukin-12 signaling pathway, suggesting maturational delays different from neonates. Importantly, recombinant interferon-γ supplementation rescued CD4 T cell numbers in the lungs and influenza-specific antibody formation. This study highlights the intrinsic limitations in CD4 T cell effector functions that may arise in toddlers and contribute to disease pathology.
Collapse
Affiliation(s)
- David Verhoeven
- Rochester General Hospital Research Institute, Rochester General Hospital, Rochester, New York, USA; and Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Sheldon Perry
- Rochester General Hospital Research Institute, Rochester General Hospital, Rochester, New York, USA; and
| | - Karin Pryharski
- Rochester General Hospital Research Institute, Rochester General Hospital, Rochester, New York, USA; and
| |
Collapse
|
39
|
Antas PRZ, Pedro TQC, Santiago EA, Lima JR, Silva FC, Melca LAV, Ponte CGG. Human neonates display altered ex vivo monokine production related to healthy adults. Immunol Lett 2015; 170:64-7. [PMID: 26687810 DOI: 10.1016/j.imlet.2015.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/20/2015] [Accepted: 12/09/2015] [Indexed: 11/29/2022]
Abstract
The inflammatory response plays an important role during the induction of several neonatal diseases. Previous studies have shown that during newborn infections, the natural imbalance between pro- and anti-inflammatory responses shifts toward the production of pro-inflammatory cytokines. In this study, we employed an array system to detect 9 pro- and anti-inflammatory cytokines, and performed ELISA for 6 other cytokines. We then compared the immune response profiling in umbilical cord blood (UV) plasma samples with circulating levels in otherwise healthy donors (HD). Concentrations of ex vivo monokine levels, such as interleukins (IL)-18, IL-23 and IL-27, were profoundly reduced in the UV in relation to the HD group (p-values of 0.003, 0.009 and <0.0001, respectively). Conversely, UV-plasmatic TGF-β1 levels displayed marked enhancement (p-value=0.005) in relation to HD. Several factors may be implicated in these neonatal alterations, and additional characterization of a broader cytokine panel is warranted to reveal other possible candidates.
Collapse
Affiliation(s)
- Paulo R Z Antas
- Clinical Immunology Laboratory, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro, Brazil.
| | - Thaíze Q C Pedro
- Clinical Immunology Laboratory, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro, Brazil
| | - Eliana A Santiago
- Clinical Immunology Laboratory, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro, Brazil
| | - Jessica R Lima
- Clinical Immunology Laboratory, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro, Brazil
| | - Fernanda C Silva
- Gaffree Guinle State University Hospital of Rio de Janeiro, Brazil
| | - Luis A V Melca
- Gaffree Guinle State University Hospital of Rio de Janeiro, Brazil
| | - Carlos G G Ponte
- Clinical Immunology Laboratory, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
40
|
El Aidy S, Dinan TG, Cryan JF. Gut Microbiota: The Conductor in the Orchestra of Immune-Neuroendocrine Communication. Clin Ther 2015; 37:954-67. [PMID: 25846319 DOI: 10.1016/j.clinthera.2015.03.002] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 02/23/2015] [Accepted: 03/04/2015] [Indexed: 01/01/2023]
Abstract
PURPOSE It is well established that mammals are so-called super-organisms that coexist with a complex microbiota. Growing evidence points to the delicacy of this host-microbe interplay and how disruptive interventions could have lifelong consequences. The goal of this article was to provide insights into the potential role of the gut microbiota in coordinating the immune-neuroendocrine cross-talk. METHODS Literature from a range of sources, including PubMed, Google Scholar, and MEDLINE, was searched to identify recent reports regarding the impact of the gut microbiota on the host immune and neuroendocrine systems in health and disease. FINDINGS The immune system and nervous system are in continuous communication to maintain a state of homeostasis. Significant gaps in knowledge remain regarding the effect of the gut microbiota in coordinating the immune-nervous systems dialogue. Recent evidence from experimental animal models found that stimulation of subsets of immune cells by the gut microbiota, and the subsequent cross-talk between the immune cells and enteric neurons, may have a major impact on the host in health and disease. IMPLICATIONS Data from rodent models, as well as from a few human studies, suggest that the gut microbiota may have a major role in coordinating the communication between the immune and neuroendocrine systems to develop and maintain homeostasis. However, the underlying mechanisms remain unclear. The challenge now is to fully decipher the molecular mechanisms that link the gut microbiota, the immune system, and the neuroendocrine system in a network of communication to eventually translate these findings to the human situation, both in health and disease.
Collapse
Affiliation(s)
- Sahar El Aidy
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Industrial Biotechnology, Genetic Engineering and Biotechnology Research Institute, Sadat City University, Sadat City, Egypt
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Psychiatry, University College Cork, Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
41
|
Patil RS, Bhat SA, Dar AA, Chiplunkar SV. The Jekyll and Hyde story of IL17-Producing γδT Cells. Front Immunol 2015; 6:37. [PMID: 25699053 PMCID: PMC4316782 DOI: 10.3389/fimmu.2015.00037] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/20/2015] [Indexed: 12/19/2022] Open
Abstract
In comparison to conventional αβT cells, γδT cells are considered as specialized T cells based on their contributions in regulating immune response. γδT cells sense early environmental signals and initiate local immune-surveillance. The development of functional subtypes of γδT cells takes place in the thymus but they also exhibit plasticity in response to the activating signals and cytokines encountered in the extrathymic region. Thymic development of Tγδ1 requires strong TCR, CD27, and Skint-1 signals. However, differentiation of IL17-producing γδT cells (Tγδ17) is independent of Skint-1 or CD27 but requires notch signaling along with IL6 and TGFβ cytokines in the presence of weak TCR signal. In response to cytokines like IL23, IL6, and IL1β, Tγδ17 outshine Th17 cells for early activation and IL17 secretion. Despite expressing similar repertoire of lineage transcriptional factors, cytokines, and chemokine receptors, Tγδ17 cells differ from Th17 in spatial and temporal fashion. There are compelling reasons to consider significant role of Tγδ17 cells in regulating inflammation and thereby disease outcome. Tγδ17 cells regulate mobilization of innate immune cells and induce keratinocytes to secrete anti-microbial peptides thus exhibiting protective functions in anti-microbial immunity. In contrast, dysregulated Tγδ17 cells inhibit Treg cells, exacerbate autoimmunity, and are also known to support carcinogenesis by enhancing angiogenesis. The mechanism associated with this dual behavior of Tγδ17 is not clear. To exploit, Tγδ17 cells for beneficial use requires comprehensive analysis of their biology. Here, we summarize the current understanding on the characteristics, development, and functions of Tγδ17 cells in various pathological scenarios.
Collapse
Affiliation(s)
- Rushikesh S Patil
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre , Kharghar , India
| | - Sajad A Bhat
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre , Kharghar , India
| | - Asif A Dar
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre , Kharghar , India
| | - Shubhada V Chiplunkar
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre , Kharghar , India
| |
Collapse
|
42
|
Tchakoute CT, Hesseling AC, Kidzeru EB, Gamieldien H, Passmore JAS, Jones CE, Gray CM, Sodora DL, Jaspan HB. Delaying BCG vaccination until 8 weeks of age results in robust BCG-specific T-cell responses in HIV-exposed infants. J Infect Dis 2015; 211:338-46. [PMID: 25108027 PMCID: PMC4318913 DOI: 10.1093/infdis/jiu434] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 07/14/2014] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND BCG vaccination prevents disseminated tuberculosis in children, but it is contraindicated for persons with human immunodeficiency virus (HIV) infection because it can result in severe disease in this population. In tuberculosis-endemic regions, BCG vaccine is administered soon after birth, before in utero and peripartum HIV infection is excluded. We therefore assessed the immunogenicity of BCG vaccine in HIV-exposed infants who received BCG at birth or at 8 weeks of age. METHODS HIV-exposed, uninfected infants were randomly assigned to receive BCG vaccination at birth (the early vaccination arm) or 8 weeks of age (the delayed vaccination arm). BCG-specific proliferative and intracellular cytokine responses were assessed in 28 infants per arm at 6, 8, and 14 weeks of life. RESULTS There was no difference in BCG-specific T-cell proliferation between the study arms 6 weeks after vaccination. However, at 14 weeks of age, the frequency of interferon γ-expressing CD4(+) T cells and multifunctional BCG-specific responses in the delayed vaccinated arm were significantly higher than those in the early vaccination arm (P = .021 and P = .011, respectively). CONCLUSIONS The immunogenicity of BCG vaccination in HIV-exposed, uninfected infants is not compromised when delayed until 8 weeks of age and results in robust BCG-specific T-cell responses at 14 weeks of age. These findings support further evaluation of this modified BCG vaccination strategy for HIV-exposed infants. CLINICAL TRIALS REGISTRATION NCT02062580.
Collapse
Affiliation(s)
| | - Anneke C. Hesseling
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town
| | | | | | - Jo-Ann S. Passmore
- Division of Medical Virology
- National Health Laboratory Services, South Africa
| | - Christine E. Jones
- Institute of Infectious Disease and Molecular Medicine, Department of Clinical Laboratory Sciences, University of Cape Town
- Paediatric Infectious Diseases Research Group, St George's, University of London, United Kingdom
| | - Clive M. Gray
- Division of Immunology
- National Health Laboratory Services, South Africa
| | | | - Heather B. Jaspan
- Division of Immunology
- Seattle Biomedical Research Institute, Washington
| |
Collapse
|
43
|
Pan H, Gazarian A, Buff S, Solla F, Gagnieu MC, Leveneur O, Watrelot-Virieux D, Morisset S, Sobh M, Michallet MC, Roger T, Dubernard JM, Michallet M. Oral cyclosporine A in neonatal swines for transplantation studies. Immunopharmacol Immunotoxicol 2014; 37:19-25. [PMID: 25358107 DOI: 10.3109/08923973.2014.975818] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The purpose of this study is to define the optimal dose of oral cyclosporine A (CsA) microemulsion in newborn swine for transplantation studies and to describe its pharmacokinetics and acute renal effects in short-term administration. Thirteen neonatal pigs were randomized into four groups: one control and three groups with CsA administration at 4, 8 and 12 mg/kg/d for 15 days (D). Blood samples were collected on D 0, 2, 4, 9 and 14 to determine the changes of the CsA trough concentrations, the creatinine (Cr) and blood urea nitrogen (BUN) serum concentrations. On D 14, blood samples were collected every hour from 1 h to 10 h after CsA administration to determine the area under the curve (AUC). On D 15, kidneys were removed for histological analysis. We observed a stabilization of CsA trough concentrations from D 4 to D 14. On D 14, in the three treated groups, CsA trough concentrations were 687 ± 7, 1200 ± 77 and 2211 ± 1030 ng/ml, respectively; AUC (0-10 h) were 6721 ± 51 ng·h/ml in group 4 mg/kg/d, 13431 ± 988 ng·h/ml in group 8 mg/kg/d and 28264 ± 9430 ng·h/ml in group 12 mg/kg/d. Cr concentrations were not significantly different among the four groups; but compared to control group, BUN concentrations of the three treated groups increased significantly. CsA was well tolerated; neither acute, severe adverse event nor renal histological abnormality was observed. In conclusion, a 15-d course of oral CsA treatment ranged from 4 to 12 mg/kg/d is safe for newborn pigs, which need much lower CsA dose than adult pigs to reach comparable trough level and AUC. As immunosuppressive therapy in newborn pigs, we recommend a CsA dose of 4 mg/kg/d to achieve a trough blood concentration between 400 and 800 ng/ml.
Collapse
Affiliation(s)
- Hua Pan
- Department of Transplantation, Hôpital Edouard Herriot , Lyon , France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kai-Larsen Y, Gudmundsson GH, Agerberth B. A review of the innate immune defence of the human foetus and newborn, with the emphasis on antimicrobial peptides. Acta Paediatr 2014; 103:1000-8. [PMID: 24861898 DOI: 10.1111/apa.12700] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/09/2014] [Accepted: 05/19/2014] [Indexed: 12/30/2022]
Abstract
UNLABELLED At birth, the foetus makes the transition from the uterus to a world full of microbes. The newborn baby needs protection against potential invading pathogens and needs to establish a normal microbiota. CONCLUSION Antimicrobial peptides and proteins are key effector molecules of innate immunity and are also important immunomodulators. Their presence in the cells and tissues of the uterus, foetus and the neonate indicates an important role in immunity during pregnancy and in early life.
Collapse
Affiliation(s)
- Ylva Kai-Larsen
- Department of Laboratory Medicine; Karolinska Institutet; Karolinska University Hospital Huddinge; Stockholm Sweden
| | | | - Birgitta Agerberth
- Department of Laboratory Medicine; Karolinska Institutet; Karolinska University Hospital Huddinge; Stockholm Sweden
| |
Collapse
|
45
|
Parra M, Herrera D, Jácome MF, Mesa MC, Rodríguez LS, Guzmán C, Angel J, Franco MA. Circulating rotavirus-specific T cells have a poor functional profile. Virology 2014; 468-470:340-350. [PMID: 25238642 DOI: 10.1016/j.virol.2014.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 05/12/2014] [Accepted: 08/21/2014] [Indexed: 10/24/2022]
Abstract
Frequencies of circulating T cells producing IFN-γ, TNF-α, and IL-2, and percentages of T cells proliferating after stimulation with rotavirus (RV), tetanus toxoid, and influenza were evaluated in PBMC derived from healthy adults and children. In addition, the potential anergic state of RV-specific T cells was analyzed by stimulation of PBMC with RV antigen in the presence of three anergy inhibitors (rIL-2, rIL-12, or DGKα-i). The quality and magnitude of RV-T cell responses were significantly lower than those of tetanus toxoid and influenza antigens. RV-CD4 T cell response was enriched in monofunctional IFN-γ(+) cells, while influenza-CD4 and tetanus toxoid-CD4 T cell responses were enriched in multifunctional T cells. Moreover, rIL-2--unlike rIL-12 or DGKα-i--increased the frequencies of RV-CD4 TNF-α(+), CD4 IFN-γ(+), and CD8 IFN-γ(+) cells. Thus, circulating RV-T cells seem to have a relatively poor functional profile that may be partially reversed in vitro by the addition of rIL-2.
Collapse
Affiliation(s)
- Miguel Parra
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Pontificia Universidad Javeriana, Carrera 7 # 40-62, Bogotá, Colombia
| | - Daniel Herrera
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Pontificia Universidad Javeriana, Carrera 7 # 40-62, Bogotá, Colombia
| | - María Fernanda Jácome
- Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Martha C Mesa
- Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Luz-Stella Rodríguez
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Pontificia Universidad Javeriana, Carrera 7 # 40-62, Bogotá, Colombia
| | - Carolina Guzmán
- Departamento de Pediatría, Hospital Universitario San Ignacio, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Juana Angel
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Pontificia Universidad Javeriana, Carrera 7 # 40-62, Bogotá, Colombia
| | - Manuel A Franco
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Pontificia Universidad Javeriana, Carrera 7 # 40-62, Bogotá, Colombia
| |
Collapse
|
46
|
Dar AA, Patil RS, Chiplunkar SV. Insights into the Relationship between Toll Like Receptors and Gamma Delta T Cell Responses. Front Immunol 2014; 5:366. [PMID: 25132835 PMCID: PMC4116803 DOI: 10.3389/fimmu.2014.00366] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/15/2014] [Indexed: 01/26/2023] Open
Abstract
The tumor microenvironment is an important aspect of cancer biology that contributes to tumor initiation, tumor progression and responses to therapy. The composition and characteristics of the tumor microenvironment vary widely and are important in determining the anti-tumor immune response. Successful immunization requires activation of both innate and adaptive immunity. Generally, immune system is compromised in patients with cancer due to immune suppression, loss of tumor antigen expression and dysfunction of antigen presenting cells (APC). Thus, therapeutic immunization leading to cancer regression remains a significant challenge. Certain cells of the immune system, including dendritic cells (DCs) and gamma delta (γδ) T cells are capable of driving potent anti-tumor responses. The property of MHC-unrestricted cytotoxicity, high potential of cytokine release, tissue tropism and early activation in infections and malignant disease makes γδ T cells as an emerging candidate for immunotherapy. Various strategies are being developed to enhance anti-tumor immune responses of γδ T cells and DCs one of them is the use of novel adjuvants like toll like receptors (TLR) agonists, which enhance γδ T cell function directly or through DC activation, which has ability to prime γδ T cells. TLR agonists are being used clinically either alone or in combination with tumor antigens and has shown initial success in both enhancing immune responses and eliciting anti-tumor activity. TLR activated γδ T cells and DCs nurture each other's activation. This provides a potent base for first line of defense and manipulation of the adaptive response against pathogens and cancer. The available data provides a strong rationale for initiating combinatorial therapy for the treatment of diseases and this review will summarize the application of adjuvants (TLRs) for boosting immune response of γδ T cells to treat cancer and infectious diseases and their use in combinatorial therapy.
Collapse
Affiliation(s)
- Asif Amin Dar
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre , Navi Mumbai , India
| | - Rushikesh Sudam Patil
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre , Navi Mumbai , India
| | - Shubhada Vivek Chiplunkar
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre , Navi Mumbai , India
| |
Collapse
|
47
|
Clay CC, Reader JR, Gerriets JE, Wang TT, Harrod KS, Miller LA. Enhanced viral replication and modulated innate immune responses in infant airway epithelium following H1N1 infection. J Virol 2014; 88:7412-25. [PMID: 24741104 PMCID: PMC4054429 DOI: 10.1128/jvi.00188-14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 04/12/2014] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Influenza is the cause of significant morbidity and mortality in pediatric populations. The contribution of pulmonary host defense mechanisms to viral respiratory infection susceptibility in very young children is poorly understood. As a surrogate to compare mucosal immune responses of infant and adult lungs, rhesus monkey primary airway epithelial cell cultures were infected with pandemic influenza A/H1N1 virus in vitro. Virus replication, cytokine secretion, cell viability, and type I interferon (IFN) pathway PCR array profiles were evaluated for both infant and adult cultures. In comparison with adult cultures, infant cultures showed significantly increased levels of H1N1 replication, reduced alpha interferon (IFN-α) protein synthesis, and no difference in cell death following infection. Age-dependent differences in expression levels of multiple genes associated with the type I IFN pathway were observed in H1N1-infected cultures. To investigate the pulmonary and systemic responses to H1N1 infection in early life, infant monkeys were inoculated with H1N1 by upper airway administration. Animals were monitored for virus and parameters of inflammation over a 14-day period. High H1N1 titers were recovered from airways at day 1, with viral RNA remaining detectable until day 9 postinfection. Despite viral clearance, bronchiolitis and alveolitis persisted at day 14 postinfection; histopathological analysis revealed alveolar septal thickening and intermittent type II pneumocyte hyperplasia. Our overall findings are consistent with the known susceptibility of pediatric populations to respiratory virus infection and suggest that intrinsic developmental differences in airway epithelial cell immune function may contribute to the limited efficacy of host defense during early childhood. IMPORTANCE To the best of our knowledge, this study represents the first report of intrinsic developmental differences in infant airway epithelial cells that may contribute to the increased susceptibility of the host to respiratory virus infections. Despite the global burden of influenza, there are currently no vaccine formulations approved for children <6 months of age. Given the challenges of conducting experimental studies involving pediatric patients, rhesus monkeys are an ideal laboratory animal model to investigate the maturation of pulmonary mucosal immune mechanisms during early life because they are most similar to those of humans with regard to postnatal maturation of the lung structure and the immune system. Thus, our findings are highly relevant to translational medicine, and these data may ultimately lead to novel approaches that enhance airway immunity in very young children.
Collapse
Affiliation(s)
- Candice C Clay
- California National Primate Research Center, University of California, Davis, California, USA
| | - J Rachel Reader
- California National Primate Research Center, University of California, Davis, California, USA
| | - Joan E Gerriets
- California National Primate Research Center, University of California, Davis, California, USA
| | - Theodore T Wang
- California National Primate Research Center, University of California, Davis, California, USA
| | - Kevin S Harrod
- Infectious Disease Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | - Lisa A Miller
- California National Primate Research Center, University of California, Davis, California, USA Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, California, USA
| |
Collapse
|
48
|
Pichichero ME. Challenges in vaccination of neonates, infants and young children. Vaccine 2014; 32:3886-94. [PMID: 24837502 PMCID: PMC4135535 DOI: 10.1016/j.vaccine.2014.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/18/2014] [Accepted: 05/01/2014] [Indexed: 12/14/2022]
Abstract
All neonates, infants and young children receive multiple priming doses and booster vaccinations in the 1st and 2nd year of life to prevent infections by viral and bacterial pathogens. Despite high vaccine compliance, outbreaks of vaccine-preventable infections are occurring worldwide. These data strongly argue for an improved understanding of the immune responses of neonates, infants and young children to vaccine antigens and further study of the exploitable mechanisms to achieve more robust and prolonged immunity with fewer primary and booster vaccinations in the pediatric population. This review will focus on our recent work involving infant and young child immunity following routine recommended vaccinations. The discussion will address vaccine responses with respect to four areas: (1) systemic antibody responses, (2) memory B-cell generation, (3) CD4 T-cell responses, and (4) APC function.
Collapse
Affiliation(s)
- Michael E Pichichero
- Rochester General Hospital Research Institute, Rochester, NY 14621, United States of America.
| |
Collapse
|
49
|
Quinello C, Silveira-Lessa AL, Ceccon MEJR, Cianciarullo MA, Carneiro-Sampaio M, Palmeira P. Phenotypic Differences in Leucocyte Populations among Healthy Preterm and Full-Term Newborns. Scand J Immunol 2014; 80:57-70. [DOI: 10.1111/sji.12183] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/07/2014] [Indexed: 01/12/2023]
Affiliation(s)
- C. Quinello
- Department of Pediatrics; Faculdade de Medicina da Universidade de São Paulo; São Paulo Brazil
- Laboratory of Medical Investigation (LIM-36); Instituto da Criança; Hospital das Clínicas; São Paulo Brazil
| | - A. L. Silveira-Lessa
- Laboratory of Medical Investigation (LIM-36); Instituto da Criança; Hospital das Clínicas; São Paulo Brazil
- Department of Parasitology; Instituto de Ciências Biomédicas da Universidade de São Paulo; São Paulo Brazil
| | - M. E. J. R. Ceccon
- Department of Pediatrics; Faculdade de Medicina da Universidade de São Paulo; São Paulo Brazil
| | - M. A. Cianciarullo
- Department of Pediatrics; Faculdade de Medicina da Universidade de São Paulo; São Paulo Brazil
| | - M. Carneiro-Sampaio
- Department of Pediatrics; Faculdade de Medicina da Universidade de São Paulo; São Paulo Brazil
- Laboratory of Medical Investigation (LIM-36); Instituto da Criança; Hospital das Clínicas; São Paulo Brazil
| | - P. Palmeira
- Department of Pediatrics; Faculdade de Medicina da Universidade de São Paulo; São Paulo Brazil
- Laboratory of Medical Investigation (LIM-36); Instituto da Criança; Hospital das Clínicas; São Paulo Brazil
| |
Collapse
|
50
|
López MC, Palmer BE, Lawrence DA. Naïve T cells, unconventional NK and NKT cells, and highly responsive monocyte-derived macrophages characterize human cord blood. Immunobiology 2014; 219:756-65. [PMID: 24986635 DOI: 10.1016/j.imbio.2014.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 04/22/2014] [Accepted: 06/03/2014] [Indexed: 10/25/2022]
Abstract
This study compares the human immune systems of neonates and adults. Flow cytometric analysis was used to study the cellular phenotypes of cord blood (CB) and adult peripheral blood (APB). Luminex analysis was used to determine the levels of cytokines in cell culture supernatants. Our findings indicate that T cells in CB were mainly naïve and thus less responsive to PMA/ionomycin with the synthesis of cytokines. The percentages of CD3(+)CD4(+)CD25(high) and of CD3(+)CD4(+)CD25(dim) cells expressing chemokine receptors were different between CB and APB. TLR1, TLR6 and TLR9 expressions on NK and NKT cells also differed between CB and APB. CB monocyte-derived macrophages responded better than APB macrophages to TLR ligands with increased secretion of inflammatory cytokines, especially IL-6. The high levels of the inflammatory cytokines in cell culture supernatants of CB were mainly due to higher numbers of responsive macrophages, since dendritic cell numbers were lower in CB than APB.
Collapse
Affiliation(s)
- María C López
- Wadsworth Center, New York State Department of Health, Empire State Plaza, PO Box 0509, Albany, NY 12201-0509, USA.
| | - Brent E Palmer
- Department of Medicine, University of Colorado, Denver, CO 80262, USA
| | - David A Lawrence
- Wadsworth Center, New York State Department of Health, Empire State Plaza, PO Box 0509, Albany, NY 12201-0509, USA
| |
Collapse
|