Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
2.
Immunol Rev ; 315(1): 154-170, 2023 05.
Article in English | MEDLINE | ID: mdl-36939073

ABSTRACT

Lymphoid cells encompass the adaptive immune system, including T and B cells and Natural killer T cells (NKT), and innate immune cells (ILCs), including Natural Killer (NK) cells. During adult life, these lineages are thought to derive from the differentiation of long-term hematopoietic stem cells (HSCs) residing in the bone marrow. However, during embryogenesis and fetal development, the ontogeny of lymphoid cells is both complex and multifaceted, with a large body of evidence suggesting that lymphoid lineages arise from progenitor cell populations antedating the emergence of HSCs. Recently, the application of single cell RNA-sequencing technologies and pluripotent stem cell-based developmental models has provided new insights into lymphoid ontogeny during embryogenesis. Indeed, PSC differentiation platforms have enabled de novo generation of lymphoid immune cells independently of HSCs, supporting conclusions drawn from the study of hematopoiesis in vivo. Here, we examine lymphoid development from non-HSC progenitor cells and technological advances in the differentiation of human lymphoid cells from pluripotent stem cells for clinical translation.


Subject(s)
Pluripotent Stem Cells , Adult , Humans , Cell Differentiation , Hematopoietic Stem Cells , Killer Cells, Natural , Hematopoiesis
3.
Semin Immunopathol ; 44(6): 747-766, 2022 11.
Article in English | MEDLINE | ID: mdl-35508672

ABSTRACT

The immune system establishes during the prenatal period from distinct waves of stem and progenitor cells and continuously adapts to the needs and challenges of early postnatal and adult life. Fetal immune development not only lays the foundation for postnatal immunity but establishes functional populations of tissue-resident immune cells that are instrumental for fetal immune responses amidst organ growth and maturation. This review aims to discuss current knowledge about the development and function of tissue-resident immune populations during fetal life, focusing on the brain, lung, and gastrointestinal tract as sites with distinct developmental trajectories. While recent progress using system-level approaches has shed light on the fetal immune landscape, further work is required to describe precise roles of prenatal immune populations and their migration and adaptation to respective organ environments. Defining points of prenatal susceptibility to environmental challenges will support the search for potential therapeutic targets to positively impact postnatal health.


Subject(s)
Fetal Development , Fetus , Pregnancy , Adult , Female , Humans , Brain , Immune System , Prenatal Care
4.
Nutrients ; 14(8)2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35458152

ABSTRACT

The first nine months from conception to birth involves greater changes than at any other time in life, affecting organogenesis, endocrine, metabolic and immune programming. It has led to the concept that the "first 1000 days" from conception to the second birthday are critical in establishing long term health or susceptibility to disease. Immune ontogeny is predominantly complete within that time and is influenced by the maternal genome, health, diet and environment pre-conception and during pregnancy and lactation. Components of the immunological protection of the pregnancy is the generation of Th-2 and T-regulatory cytokines with the consequence that neonatal adaptive responses are also biased towards Th-2 (allergy promoting) and T-regulatory (tolerance promoting) responses. Normally after birth Th-1 activity increases while Th-2 down-regulates and the evolving normal human microbiome likely plays a key role. This in turn will have been affected by maternal health, diet, exposure to antibiotics, mode of delivery, and breast or cow milk formula feeding. Complex gene/environment interactions affect outcomes. Many individual nutrients affect immune mechanisms and variations in levels have been associated with susceptibility to allergic disease. However, intervention trials employing single nutrient supplementation to prevent allergic disease have not achieved the expected outcomes suggested by observational studies. Investigation of overall dietary practices including fresh fruit and vegetables, fish, olive oil, lower meat intake and home cooked foods as seen in the Mediterranean and other healthy diets have been associated with reduced prevalence of allergic disease. This suggests that the "soup" of overall nutrition is more important than individual nutrients and requires further investigation both during pregnancy and after the infant has been weaned. Amongst all the potential factors affecting allergy outcomes, modification of maternal and infant nutrition and the microbiome are easier to employ than changing other aspects of the environment but require large controlled trials before recommending changes to current practice.


Subject(s)
Hypersensitivity , Animals , Breast Feeding , Cattle , Diet , Female , Humans , Infant , Lactation , Milk , Pregnancy
5.
Viruses ; 13(10)2021 10 03.
Article in English | MEDLINE | ID: mdl-34696417

ABSTRACT

Gamma-delta (γδ) T cells are unconventional T cells that help control cytomegalovirus (CMV) infection in adults. γδ T cells develop early in gestation, and a fetal public γδ T cell receptor (TCR) clonotype is detected in congenital CMV infections. However, age-dependent γδ T cell responses to primary CMV infection are not well-understood. Flow cytometry and TCR sequencing was used to comprehensively characterize γδ T cell responses to CMV infection in a cohort of 32 infants followed prospectively from birth. Peripheral blood γδ T cell frequencies increased during infancy, and were higher among CMV-infected infants relative to uninfected. Clustering analyses revealed associations between CMV infection and activation marker expression on adaptive-like Vδ1 and Vδ3, but not innate-like Vγ9Vδ2 γδ T cell subsets. Frequencies of NKG2C+CD57+ γδ T cells were temporally associated with the quantity of CMV shed in saliva by infants with primary infection. The public γδ TCR clonotype was only detected in CMV-infected infants <120 days old and at lower frequencies than previously described in fetal infections. Our findings support the notion that CMV infection drives age-dependent expansions of specific γδ T cell populations, and provide insight for novel strategies to prevent CMV transmission and disease.


Subject(s)
Cytomegalovirus Infections/immunology , Intraepithelial Lymphocytes/metabolism , Intraepithelial Lymphocytes/physiology , Adult , Age Factors , Child , Child, Preschool , Cohort Studies , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/virology , Female , Flow Cytometry/methods , Humans , Infant , Infant, Newborn , Lymphocyte Activation/immunology , Male , Prospective Studies , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , Uganda/epidemiology
6.
Am J Phys Anthropol ; 175 Suppl 72: 57-78, 2021 08.
Article in English | MEDLINE | ID: mdl-33460467

ABSTRACT

Breastfeeding is known to be a powerful mediator of maternal and childhood health, with impacts throughout the life course. Paleodietary studies of the past 30 years have accordingly taken an enduring interest in the health and diet of young children as a potential indicator of population fertility, subsistence, and mortality patterns. While progress has been made in recent decades toward acknowledging the agency of children, many paleodietary reconstructions have failed to incorporate developments in cognate disciplines revealing synergistic dynamics between maternal and offspring biology. Paleodietary interpretation has relied heavily on the "weanling's dilemma," in which infants are thought to face a bleak choice between loss of immunity or malnutrition. Using a review of immunological and epidemiological evidence for the dynamic and supportive role that breastfeeding plays throughout the complementary feeding period, this article offers context and nuance for understanding past feeding transitions. We suggest that future interpretative frameworks for infant paleodietary and bioarchaeological research should include a broad knowledge base that keeps pace with relevant developments outside of those disciplines.


Subject(s)
Diet , Infant Nutritional Physiological Phenomena , Milk, Human , Breast Feeding , Humans , Infant , Infant Nutritional Physiological Phenomena/immunology , Infant Nutritional Physiological Phenomena/physiology , Milk, Human/immunology , Milk, Human/physiology , Paleontology , Weaning
7.
Front Immunol ; 11: 1683, 2020.
Article in English | MEDLINE | ID: mdl-32849587

ABSTRACT

Systems biology is an approach to interrogate complex biological systems through large-scale quantification of numerous biomolecules. The immune system involves >1,500 genes/proteins in many interconnected pathways and processes, and a systems-level approach is critical in broadening our understanding of the immune response to vaccination. Changes in molecular pathways can be detected using high-throughput omics datasets (e.g., transcriptomics, proteomics, and metabolomics) by using methods such as pathway enrichment, network analysis, machine learning, etc. Importantly, integration of multiple omic datasets is becoming key to revealing novel biological insights. In this perspective article, we highlight the use of protein-protein interaction (PPI) networks as a multi-omics integration approach to unravel information flow and mechanisms during complex biological events, with a focus on the immune system. This involves a combination of tools, including: InnateDB, a database of curated interactions between genes and protein products involved in the innate immunity; NetworkAnalyst, a visualization and analysis platform for InnateDB interactions; and MetaBridge, a tool to integrate metabolite data into PPI networks. The application of these systems techniques is demonstrated for a variety of biological questions, including: the developmental trajectory of neonates during the first week of life, mechanisms in host-pathogen interaction, disease prognosis, biomarker discovery, and drug discovery and repurposing. Overall, systems biology analyses of omics data have been applied to a variety of immunology-related questions, and here we demonstrate the numerous ways in which PPI network analysis can be a powerful tool in contributing to our understanding of the immune system and the study of vaccines.


Subject(s)
Genomics , Immune System/drug effects , Immunity, Innate/drug effects , Metabolomics , Systems Biology , Vaccines/pharmacology , Adaptive Immunity/drug effects , Gene Regulatory Networks , Host-Pathogen Interactions , Humans , Immune System/immunology , Immune System/metabolism , Protein Interaction Maps , Signal Transduction , Systems Integration , Transcriptome
8.
Front Immunol ; 11: 1285, 2020.
Article in English | MEDLINE | ID: mdl-32636848

ABSTRACT

Chorioamnionitis, a potentially serious inflammatory complication of pregnancy, is associated with the development of an inflammatory milieu within the amniotic fluid surrounding the developing fetus. When chorioamnionitis occurs, the fetal lung finds itself in the unique position of being constantly exposed to the consequent inflammatory meditators and/or microbial products found in the amniotic fluid. This exposure results in significant changes to the fetal lung, such as increased leukocyte infiltration, altered cytokine, and surfactant production, and diminished alveolarization. These alterations can have potentially lasting impacts on lung development and function. However, studies to date have only begun to elucidate the association between such inflammatory exposures and lifelong consequences such as lung dysfunction. In this review, we discuss the pathogenesis of and fetal immune response to chorioamnionitis, detail the consequences of chorioamnionitis exposure on the developing fetal lung, highlighting the various animal models that have contributed to our current understanding and discuss the importance of fetal exposures in regard to the development of chronic respiratory disease. Finally, we focus on the clinical, basic, and therapeutic challenges in fetal inflammatory injury to the lung, and propose next steps and future directions to improve our therapeutic understanding of this important perinatal stress.


Subject(s)
Chorioamnionitis/immunology , Fetus/immunology , Lung/embryology , Lung/pathology , Prenatal Exposure Delayed Effects/immunology , Animals , Chorioamnionitis/pathology , Female , Humans , Pregnancy , Prenatal Exposure Delayed Effects/pathology
9.
mSphere ; 4(4)2019 07 17.
Article in English | MEDLINE | ID: mdl-31315966

ABSTRACT

This report summarizes a consultation meeting convened by the National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), on 12 September 2017 to discuss the scientific rationale for selectively testing relevant HIV vaccine candidates in early life that are designed to initiate immune responses for lifelong protective immunity. The urgent need to develop interventions providing durable protective immunity to HIV before sexual debut coupled with the practicality of infant vaccine schedules supports optimizing infant HIV vaccines as a high priority. The panelists discussed the unique opportunities and challenges of testing candidate HIV vaccines in the context of distinct early-life immunity. Key developments providing rationale and grounds for cautious optimism regarding evaluation of early-life HIV vaccines include recent studies of early-life immune ontogeny, studies of HIV-infected infants demonstrating relatively rapid generation of broadly neutralizing antibodies (bNAbs), discovery of novel adjuvants active in early life, and cutting-edge sample-sparing systems biology and immunologic assays promising deep insight into vaccine action in infants. Multidisciplinary efforts toward the goal of an infant HIV vaccine are under way and should be nurtured and amplified.IMPORTANCE Young adults represent one of the highest-risk groups for new HIV infections and the only group in which morbidity continues to increase. Therefore, an HIV vaccine to prevent HIV acquisition in adolescence is a top priority. The introduction of any vaccine during adolescence is challenging. This meeting discussed the opportunities and challenges of testing HIV vaccine candidates in the context of the infant immune system given recent advances in our knowledge of immune ontogeny and adjuvant design and studies demonstrating that HIV-infected infants generate broadly neutralizing antibodies, a main target of HIV vaccines, more rapidly than adults. Considering the global success of pediatric vaccines, the concept of an HIV vaccine introduced in early life holds merit and warrants testing.


Subject(s)
AIDS Vaccines , HIV Infections/prevention & control , Immunization Schedule , Adjuvants, Immunologic , Adolescent , Child , Clinical Trials as Topic , Congresses as Topic , Female , HIV Infections/immunology , HIV-1/immunology , Humans , Infant , Referral and Consultation , United States
10.
Immunity ; 50(2): 462-476.e8, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30770246

ABSTRACT

Although the fetal immune system is considered tolerogenic, preterm infants can suffer from severe intestinal inflammation, including necrotizing enterocolitis (NEC). Here, we demonstrate that human fetal intestines predominantly contain tumor necrosis factor-α (TNF-α)+CD4+CD69+ T effector memory (Tem) cells. Single-cell RNA sequencing of fetal intestinal CD4+ T cells showed a T helper 1 phenotype and expression of genes mediating epithelial growth and cell cycling. Organoid co-cultures revealed a dose-dependent, TNF-α-mediated effect of fetal intestinal CD4+ T cells on intestinal stem cell (ISC) development, in which low T cell numbers supported epithelial development, whereas high numbers abrogated ISC proliferation. CD4+ Tem cell frequencies were higher in inflamed intestines from preterm infants with NEC than in healthy infant intestines and showed enhanced TNF signaling. These findings reveal a distinct population of TNF-α-producing CD4+ T cells that promote mucosal development in fetal intestines but can also mediate inflammation upon preterm birth.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fetus/immunology , Immunologic Memory/immunology , Intestines/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Fetus/metabolism , Humans , Infant, Newborn , Intestinal Mucosa/embryology , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Intestines/embryology , Intestines/growth & development , Mice, Inbred C57BL , Pregnancy , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Cell ; 174(1): 117-130.e14, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29909981

ABSTRACT

Heterogeneity is a hallmark feature of the adaptive immune system in vertebrates. Following infection, naive T cells differentiate into various subsets of effector and memory T cells, which help to eliminate pathogens and maintain long-term immunity. The current model suggests there is a single lineage of naive T cells that give rise to different populations of effector and memory T cells depending on the type and amounts of stimulation they encounter during infection. Here, we have discovered that multiple sub-populations of cells exist in the naive CD8+ T cell pool that are distinguished by their developmental origin, unique transcriptional profiles, distinct chromatin landscapes, and different kinetics and phenotypes after microbial challenge. These data demonstrate that the naive CD8+ T cell pool is not as homogeneous as previously thought and offers a new framework for explaining the remarkable heterogeneity in the effector and memory T cell subsets that arise after infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Genes, Developmental , Listeria monocytogenes/pathogenicity , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Line, Tumor , Chromatin/metabolism , Cytokines/pharmacology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Immunologic Memory , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Listeria monocytogenes/metabolism , Mice , Mice, Inbred C57BL , Principal Component Analysis , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/transplantation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
12.
Semin Immunopathol ; 38(6): 739-763, 2016 11.
Article in English | MEDLINE | ID: mdl-27465226

ABSTRACT

Endogenous levels of glucocorticoids rise during pregnancy to warrant development and maturation of the fetal organs close to birth. However, during most of the gestation, the fetus is protected from excessive biologically active endogenous glucocorticoids by placental and fetal expression of 11ß-hydroxysteroid dehydrogenase 2 (11ß-HSD2). Maternal stress, which may overwhelm placental 11ß-HSD2 activity with high glucocorticoid levels, or administration of synthetic glucocorticoids to improve the survival chances of the premature newborn, are associated to postnatal increased risk for immune diseases. Fetal exposure to excessive glucocorticoids may underlie this altered postnatal immunity. Here, we revise the role that placental and fetal 11ß-HSD2, fetal glucocorticoid exposure, and programming of the offspring's the hypothalamic-pituitary-adrenal (HPA) axis play on concerted steps in immune fetal development. We could identify gaps in knowledge about glucocorticoid-induced programming of immune diseases. Finally, based on current evidence about glucocorticoid and HPA axis-mediated immune regulation, we hypothesize on mechanisms that could drive the enhanced risk for atopies, infections, and type I diabetes in offspring that were prenatally exposed to glucocorticoids.


Subject(s)
Glucocorticoids/administration & dosage , Glucocorticoids/metabolism , Immunity/drug effects , 11-beta-Hydroxysteroid Dehydrogenases/genetics , 11-beta-Hydroxysteroid Dehydrogenases/metabolism , Female , Fetus/drug effects , Fetus/metabolism , Gene Expression Regulation/drug effects , Glucocorticoids/adverse effects , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Immune System/drug effects , Immune System/immunology , Immune System/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Placenta/metabolism , Pregnancy , Prenatal Care , Prenatal Exposure Delayed Effects , Reproductive Physiological Phenomena
13.
F1000Res ; 5: 414, 2016.
Article in English | MEDLINE | ID: mdl-27347375

ABSTRACT

Compendia of large-scale datasets made available in public repositories provide an opportunity to identify and fill gaps in biomedical knowledge. But first, these data need to be made readily accessible to research investigators for interpretation. Here we make available a collection of transcriptome datasets to investigate the functional programming of human hematopoietic cells in early life. Thirty two datasets were retrieved from the NCBI Gene Expression Omnibus (GEO) and loaded in a custom web application called the Gene Expression Browser (GXB), which was designed for interactive query and visualization of integrated large-scale data. Quality control checks were performed. Multiple sample groupings and gene rank lists were created allowing users to reveal age-related differences in transcriptome profiles, changes in the gene expression of neonatal hematopoietic cells to a variety of immune stimulators and modulators, as well as during cell differentiation. Available demographic, clinical, and cell phenotypic information can be overlaid with the gene expression data and used to sort samples. Web links to customized graphical views can be generated and subsequently inserted in manuscripts to report novel findings. GXB also enables browsing of a single gene across projects, thereby providing new perspectives on age- and developmental stage-specific expression of a given gene across the human hematopoietic system. This dataset collection is available at: http://developmentalimmunology.gxbsidra.org/dm3/geneBrowser/list.

14.
Cytokine ; 83: 99-109, 2016 07.
Article in English | MEDLINE | ID: mdl-27081760

ABSTRACT

Most infections occur in early life, prompting development of novel adjuvanted vaccines to protect newborns and infants. Several Toll-like receptor (TLR) agonists (TLRAs) are components of licensed vaccine formulations or are in development as candidate adjuvants. However, the type and magnitude of immune responses to TLRAs may vary with the TLR activated as well as age and geographic location. Most notably, in newborns, as compared to adults, the immune response to TLRAs is polarized with lower Th1 cytokine production and robust Th2 and anti-inflammatory cytokine production. The ontogeny of TLR-mediated cytokine responses in international cohorts has been reported, but no study has compared cytokine responses to TLRAs between U.S. neonates and infants at the age of 6months. Both are critical age groups for the currently pediatric vaccine schedule. In this study, we report quantitative differences in the production of a panel of 14 cytokines and chemokines after in vitro stimulation of newborn cord blood and infant and adult peripheral blood with agonists of TLR4, including monophosphoryl lipid A (MPLA) and glucopyranosyl lipid Adjuvant aqueous formulation (GLA-AF), as well as agonists of TLR7/8 (R848) and TLR9 (CpG). Both TLR4 agonists, MPLA and GLA-AF, induced greater concentrations of Th1 cytokines CXCL10, TNF and Interleukin (IL)-12p70 in infant and adult blood compared to newborn blood. All the tested TLRAs induced greater infant IFN-α2 production compared to newborn and adult blood. In contrast, CpG induced greater IFN-γ, IL-1ß, IL-4, IL-12p40, IL-10 and CXCL8 in newborn than in infant and adult blood. Overall, to the extent that these in vitro studies mirror responses in vivo, our study demonstrates distinct age-specific effects of TLRAs that may inform their development as candidate adjuvants for early life vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aging/immunology , Cytokines/immunology , Oligodeoxyribonucleotides/pharmacology , Th1 Cells/immunology , Th2 Cells/immunology , Toll-Like Receptors/immunology , Adult , Female , Humans , Infant , Infant, Newborn , Male
15.
R Soc Open Sci ; 2(4): 150108, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26064646

ABSTRACT

Variations in immune function can arise owing to trade-offs, that is, the allocation of limited resources among costly competing physiological functions. Nevertheless, there is little information regarding the ontogeny of the immune system within an ecological context, and it is still unknown whether development affects the way in which resources are allocated to different immune effectors. We investigated changes in the inflammatory response during early development of the California sea lion (Zalophus californianus) and examined its association with body condition, as a proxy for the availability of energetic resources. We found that the relationship between inflammation and body condition varied according to developmental stage and circulating levels of leucocyte populations, a proxy for current infection. Body condition was related to the magnitude of the inflammatory response during two of the three developmental periods assessed, allowing for the possibility that the availability of pup energetic reserves can limit immune function. For older pups, the ability to mount an inflammatory response was related to their circulating levels of neutrophils and the neutrophil to lymphocyte ratio, implying that the infection status of an individual will influence its ability to respond to a new challenge. Our results suggest that trade-offs may occur within the immune system and highlight the importance of taking into account ontogeny in ecoimmunological studies.

16.
Philos Trans R Soc Lond B Biol Sci ; 370(1671)2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25964459

ABSTRACT

Despite significant progress in reducing the burden of mortality in children under the age of five, reducing mortality in newborns remains a major challenge. Infection plays a significant role in infant deaths and interventions such as early vaccination or antenatal immunization could make a significant contribution to prevention of such deaths. In the last few years, we have gained new insights into immune ontogeny and are now beginning to understand the impact of vaccines, nutrition and environmental factors on 'training' of the immune response in early life. This review article sets out to explain why vaccine responses can be heterogeneous between populations and individuals by providing examples chosen to illustrate the impact of host, pathogen and environmental factors on shaping the immune 'interactome' in young children.


Subject(s)
Immunity, Innate/physiology , Vaccines/immunology , Humans , Infant , Infant, Newborn
18.
J Reprod Immunol ; 108: 12-25, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25638482

ABSTRACT

The presence of maternal cells in the organs of the offspring is referred to as maternal microchimerism (MMc). MMc is physiologically acquired during pregnancy and lactation and can persist until adulthood. The detection of MMc in a variety of human diseases has raised interest in the short- and long-term functional consequences for the offspring. Owing to limited availability and access to human tissue, mouse models have become an essential tool in elucidating the functional role of MMc. This review compiles the detection techniques and experimental settings used in murine MMc research. It aims to summarize the potential mechanisms of migration of MMc, pre- and postnatal tissue distribution, phenotype and concatenated function, as well as factors modulating its occurrence. In this context, we propose MMc to be a materno-fetal messenger with the capacity to critically shape the development of the offspring's immunity.


Subject(s)
Chimerism , Disease Models, Animal , Immunity, Maternally-Acquired , Animals , Chimerism/embryology , Female , Humans , Immunomodulation , Maternal Exposure/adverse effects , Maternal-Fetal Exchange , Mice , Pregnancy
19.
Front Immunol ; 5: 387, 2014.
Article in English | MEDLINE | ID: mdl-25165466

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of granulocytic or monocytic cells that suppress innate as well as adaptive immune responses. In healthy adults, immature myeloid cells differentiate into macrophages, dendritic cells, and granulocytes in the bone marrow and MDSC are rarely detected in peripheral blood. However, in certain pathologies, in particular malignancies and chronic infection, differentiation of these cells is altered resulting in accumulation of circulating suppressive myeloid cells. MDSC express suppressive factors such as arginase-1, reactive oxygen species, and inducible nitric oxide synthase, which have the ability to inhibit T cell proliferation and cytoxicity, induce the expansion of regulatory T cells, and block natural killer cell activation. It is increasingly recognized that MDSC alter the immune response to several cancers, and perhaps chronic viral infections, in clinically important ways. In this review, we outline the potential contribution of MDSC to the generation of feto-maternal tolerance and to the ineffective immune responses to many infections and vaccines observed in early post-natal life. Granulocytic MDSC are present in large numbers in pregnant women and in cord blood, and wane rapidly during infancy. Furthermore, cord blood MDSC suppress in vitro T cell and NK responses, suggesting that they may play a significant role in human immune ontogeny. However, there are currently no data that demonstrate in vivo effects of MDSC on feto-maternal tolerance or immune ontogeny. Studies are ongoing to evaluate the functional importance of MDSC, including their effects on control of infection and response to vaccination in infancy. Importantly, several pharmacologic interventions have the potential to reverse MDSC function. Understanding the role of MDSC in infant ontogeny and their mechanisms of action could lead to interventions that reduce mortality due to early-life infections.

20.
Trends Immunol ; 35(7): 299-310, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880460

ABSTRACT

The human immune system comprises cellular and molecular components designed to coordinately prevent infection while avoiding potentially harmful inflammation and autoimmunity. Immunity varies with age, reflecting unique age-dependent challenges including fetal gestation, the neonatal phase, and infancy. Here, we review novel mechanistic insights into early life immunity, with an emphasis on emerging models of human immune ontogeny, which may inform age-specific translational development of novel anti-infectives, immunomodulators, and vaccines.


Subject(s)
Aging/immunology , Immune System/embryology , Infections/immunology , Anti-Infective Agents/therapeutic use , Humans , Immune System/growth & development , Immunity , Immunomodulation , Infant , Infant, Newborn , Infections/therapy , Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL