Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 711.373
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Cell ; 185(3): 485-492.e10, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35051367

ABSTRACT

An outbreak of over 1,000 COVID-19 cases in Provincetown, Massachusetts (MA), in July 2021-the first large outbreak mostly in vaccinated individuals in the US-prompted a comprehensive public health response, motivating changes to national masking recommendations and raising questions about infection and transmission among vaccinated individuals. To address these questions, we combined viral genomic and epidemiological data from 467 individuals, including 40% of outbreak-associated cases. The Delta variant accounted for 99% of cases in this dataset; it was introduced from at least 40 sources, but 83% of cases derived from a single source, likely through transmission across multiple settings over a short time rather than a single event. Genomic and epidemiological data supported multiple transmissions of Delta from and between fully vaccinated individuals. However, despite its magnitude, the outbreak had limited onward impact in MA and the US overall, likely due to high vaccination rates and a robust public health response.


Subject(s)
COVID-19/epidemiology , COVID-19/immunology , COVID-19/transmission , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/virology , Child , Child, Preschool , Contact Tracing/methods , Disease Outbreaks , Female , Genome, Viral , Humans , Infant , Infant, Newborn , Male , Massachusetts/epidemiology , Middle Aged , Molecular Epidemiology , Phylogeny , SARS-CoV-2/classification , Vaccination , Whole Genome Sequencing , Young Adult
2.
Cell ; 184(3): 628-642.e10, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33476549

ABSTRACT

SARS-CoV-2 infection causes more severe disease in pregnant women compared to age-matched non-pregnant women. Whether maternal infection causes changes in the transfer of immunity to infants remains unclear. Maternal infections have previously been associated with compromised placental antibody transfer, but the mechanism underlying this compromised transfer is not established. Here, we used systems serology to characterize the Fc profile of influenza-, pertussis-, and SARS-CoV-2-specific antibodies transferred across the placenta. Influenza- and pertussis-specific antibodies were actively transferred. However, SARS-CoV-2-specific antibody transfer was significantly reduced compared to influenza- and pertussis-specific antibodies, and cord titers and functional activity were lower than in maternal plasma. This effect was only observed in third-trimester infection. SARS-CoV-2-specific transfer was linked to altered SARS-CoV-2-antibody glycosylation profiles and was partially rescued by infection-induced increases in IgG and increased FCGR3A placental expression. These results point to unexpected compensatory mechanisms to boost immunity in neonates, providing insights for maternal vaccine design.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Immunoglobulin G/immunology , Maternal-Fetal Exchange/immunology , Placenta/immunology , Pregnancy Complications, Infectious/immunology , SARS-CoV-2/immunology , Adult , Female , Humans , Infant, Newborn , Pregnancy , Pregnancy Trimester, Third/immunology , Receptors, IgG/immunology , THP-1 Cells
3.
Cell ; 184(7): 1858-1864.e10, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33631096

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread within the human population. Although SARS-CoV-2 is a novel coronavirus, most humans had been previously exposed to other antigenically distinct common seasonal human coronaviruses (hCoVs) before the coronavirus disease 2019 (COVID-19) pandemic. Here, we quantified levels of SARS-CoV-2-reactive antibodies and hCoV-reactive antibodies in serum samples collected from 431 humans before the COVID-19 pandemic. We then quantified pre-pandemic antibody levels in serum from a separate cohort of 251 individuals who became PCR-confirmed infected with SARS-CoV-2. Finally, we longitudinally measured hCoV and SARS-CoV-2 antibodies in the serum of hospitalized COVID-19 patients. Our studies indicate that most individuals possessed hCoV-reactive antibodies before the COVID-19 pandemic. We determined that ∼20% of these individuals possessed non-neutralizing antibodies that cross-reacted with SARS-CoV-2 spike and nucleocapsid proteins. These antibodies were not associated with protection against SARS-CoV-2 infections or hospitalizations, but they were boosted upon SARS-CoV-2 infection.


Subject(s)
Alphacoronavirus/immunology , Antibodies, Viral , Betacoronavirus/immunology , COVID-19/immunology , Adolescent , Adult , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19 Serological Testing , Child , Child, Preschool , Chlorocebus aethiops , Cross Protection , Cross Reactions , Disease Susceptibility , HEK293 Cells , Humans , Infant , Infant, Newborn , Vero Cells
4.
Cell ; 184(15): 3884-3898.e11, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34143954

ABSTRACT

Immune-microbe interactions early in life influence the risk of allergies, asthma, and other inflammatory diseases. Breastfeeding guides healthier immune-microbe relationships by providing nutrients to specialized microbes that in turn benefit the host's immune system. Such bacteria have co-evolved with humans but are now increasingly rare in modern societies. Here we show that a lack of bifidobacteria, and in particular depletion of genes required for human milk oligosaccharide (HMO) utilization from the metagenome, is associated with systemic inflammation and immune dysregulation early in life. In breastfed infants given Bifidobacterium infantis EVC001, which expresses all HMO-utilization genes, intestinal T helper 2 (Th2) and Th17 cytokines were silenced and interferon ß (IFNß) was induced. Fecal water from EVC001-supplemented infants contains abundant indolelactate and B. infantis-derived indole-3-lactic acid (ILA) upregulated immunoregulatory galectin-1 in Th2 and Th17 cells during polarization, providing a functional link between beneficial microbes and immunoregulation during the first months of life.


Subject(s)
Bifidobacterium/physiology , Immune System/growth & development , Immune System/microbiology , Anti-Bacterial Agents/pharmacology , Biomarkers/metabolism , Breast Feeding , CD4-Positive T-Lymphocytes/immunology , Cell Polarity , Cell Proliferation , Cytokines/metabolism , Feces/chemistry , Feces/microbiology , Galectin 1/metabolism , Gastrointestinal Microbiome , Humans , Indoles/metabolism , Infant, Newborn , Inflammation/blood , Inflammation/genetics , Intestinal Mucosa/immunology , Metabolome , Milk, Human/chemistry , Oligosaccharides/metabolism , Th17 Cells/immunology , Th2 Cells/immunology , Water
5.
Nat Immunol ; 24(11): 1890-1907, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37749325

ABSTRACT

CD8+ T cells provide robust antiviral immunity, but how epitope-specific T cells evolve across the human lifespan is unclear. Here we defined CD8+ T cell immunity directed at the prominent influenza epitope HLA-A*02:01-M158-66 (A2/M158) across four age groups at phenotypic, transcriptomic, clonal and functional levels. We identify a linear differentiation trajectory from newborns to children then adults, followed by divergence and a clonal reset in older adults. Gene profiles in older adults closely resemble those of newborns and children, despite being clonally distinct. Only child-derived and adult-derived A2/M158+CD8+ T cells had the potential to differentiate into highly cytotoxic epitope-specific CD8+ T cells, which was linked to highly functional public T cell receptor (TCR)αß signatures. Suboptimal TCRαß signatures in older adults led to less proliferation, polyfunctionality, avidity and recognition of peptide mutants, although displayed no signs of exhaustion. These data suggest that priming T cells at different stages of life might greatly affect CD8+ T cell responses toward viral infections.


Subject(s)
CD8-Positive T-Lymphocytes , Longevity , Infant, Newborn , Humans , Aged , Epitopes, T-Lymphocyte/genetics , T-Lymphocytes, Cytotoxic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell/genetics
6.
Cell ; 183(2): 324-334.e5, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33007265

ABSTRACT

Infants born by vaginal delivery are colonized with maternal fecal microbes. Cesarean section (CS) birth disturbs mother-to-neonate transmission. In this study (NCT03568734), we evaluated whether disturbed intestinal microbiota development could be restored in term CS-born infants by postnatal, orally delivered fecal microbiota transplantation (FMT). We recruited 17 mothers, of whom seven were selected after careful screening. Their infants received a diluted fecal sample from their own mothers, taken 3 weeks prior to delivery. All seven infants had an uneventful clinical course during the 3-month follow-up and showed no adverse effects. The temporal development of the fecal microbiota composition of FMT-treated CS-born infants no longer resembled that of untreated CS-born infants but showed significant similarity to that of vaginally born infants. This proof-of-concept study demonstrates that the intestinal microbiota of CS-born infants can be restored postnatally by maternal FMT. However, this should only be done after careful clinical and microbiological screening.


Subject(s)
Fecal Microbiota Transplantation/methods , Feces/microbiology , Gastrointestinal Microbiome/physiology , Adult , Cesarean Section/adverse effects , Delivery, Obstetric , Female , Humans , Infant , Infant, Newborn , Male , Microbiota/physiology , Mothers , Pregnancy , Proof of Concept Study , Vagina/microbiology
7.
Cell ; 183(4): 982-995.e14, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32991843

ABSTRACT

Initially, children were thought to be spared from disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, a month into the epidemic, a novel multisystem inflammatory syndrome in children (MIS-C) emerged. Herein, we report on the immune profiles of nine MIS-C cases. All MIS-C patients had evidence of prior SARS-CoV-2 exposure, mounting an antibody response with intact neutralization capability. Cytokine profiling identified elevated signatures of inflammation (IL-18 and IL-6), lymphocytic and myeloid chemotaxis and activation (CCL3, CCL4, and CDCP1), and mucosal immune dysregulation (IL-17A, CCL20, and CCL28). Immunophenotyping of peripheral blood revealed reductions of non-classical monocytes, and subsets of NK and T lymphocytes, suggesting extravasation to affected tissues. Finally, profiling the autoantigen reactivity of MIS-C plasma revealed both known disease-associated autoantibodies (anti-La) and novel candidates that recognize endothelial, gastrointestinal, and immune-cell antigens. All patients were treated with anti-IL-6R antibody and/or IVIG, which led to rapid disease resolution.


Subject(s)
Inflammation/pathology , Systemic Inflammatory Response Syndrome/pathology , Adolescent , Antibodies, Viral/blood , Autoantibodies/blood , Betacoronavirus/immunology , Betacoronavirus/isolation & purification , COVID-19 , Chemokine CCL3/metabolism , Child , Child, Preschool , Coronavirus Infections/complications , Coronavirus Infections/pathology , Coronavirus Infections/virology , Female , Humans , Immunity, Humoral , Infant , Infant, Newborn , Inflammation/metabolism , Interleukin-17/metabolism , Interleukin-18/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Male , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , SARS-CoV-2 , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Young Adult
8.
Nat Immunol ; 23(7): 1109-1120, 2022 07.
Article in English | MEDLINE | ID: mdl-35761081

ABSTRACT

Nonimmune cells can have immunomodulatory roles that contribute to healthy development. However, the molecular and cellular mechanisms underlying the immunomodulatory functions of erythroid cells during human ontogenesis remain elusive. Here, integrated, single-cell transcriptomic studies of erythroid cells from the human yolk sac, fetal liver, preterm umbilical cord blood (UCB), term UCB and adult bone marrow (BM) identified classical and immune subsets of erythroid precursors with divergent differentiation trajectories. Immune-erythroid cells were present from the yolk sac to the adult BM throughout human ontogenesis but failed to be generated in vitro from human embryonic stem cells. Compared with classical-erythroid precursors, these immune-erythroid cells possessed dual erythroid and immune regulatory networks, showed immunomodulatory functions and interacted more frequently with various innate and adaptive immune cells. Our findings provide important insights into the nature of immune-erythroid cells and their roles during development and diseases.


Subject(s)
Erythroid Precursor Cells , Transcriptome , Adult , Cell Differentiation/genetics , Erythroid Cells , Fetal Blood , Humans , Infant, Newborn , Yolk Sac
9.
Nat Immunol ; 23(1): 75-85, 2022 01.
Article in English | MEDLINE | ID: mdl-34937930

ABSTRACT

We report a pleiotropic disease due to loss-of-function mutations in RHBDF2, the gene encoding iRHOM2, in two kindreds with recurrent infections in different organs. One patient had recurrent pneumonia but no colon involvement, another had recurrent infectious hemorrhagic colitis but no lung involvement and the other two experienced recurrent respiratory infections. Loss of iRHOM2, a rhomboid superfamily member that regulates the ADAM17 metalloproteinase, caused defective ADAM17-dependent cleavage and release of cytokines, including tumor-necrosis factor and amphiregulin. To understand the diverse clinical phenotypes, we challenged Rhbdf2-/- mice with Pseudomonas aeruginosa by nasal gavage and observed more severe pneumonia, whereas infection with Citrobacter rodentium caused worse inflammatory colitis than in wild-type mice. The fecal microbiota in the colitis patient had characteristic oral species that can predispose to colitis. Thus, a human immunodeficiency arising from iRHOM2 deficiency causes divergent disease phenotypes that can involve the local microbial environment.


Subject(s)
ADAM17 Protein/genetics , Carrier Proteins/genetics , Primary Immunodeficiency Diseases/genetics , A549 Cells , Animals , Child , Child, Preschool , Citrobacter rodentium/pathogenicity , Colitis/genetics , Cytokines/genetics , Enterobacteriaceae Infections/genetics , Female , HEK293 Cells , Humans , Infant, Newborn , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/pathogenicity , Signal Transduction/genetics
10.
Cell ; 177(3): 587-596.e9, 2019 04 18.
Article in English | MEDLINE | ID: mdl-31002795

ABSTRACT

Severe obesity is a rapidly growing global health threat. Although often attributed to unhealthy lifestyle choices or environmental factors, obesity is known to be heritable and highly polygenic; the majority of inherited susceptibility is related to the cumulative effect of many common DNA variants. Here we derive and validate a new polygenic predictor comprised of 2.1 million common variants to quantify this susceptibility and test this predictor in more than 300,000 individuals ranging from middle age to birth. Among middle-aged adults, we observe a 13-kg gradient in weight and a 25-fold gradient in risk of severe obesity across polygenic score deciles. In a longitudinal birth cohort, we note minimal differences in birthweight across score deciles, but a significant gradient emerged in early childhood and reached 12 kg by 18 years of age. This new approach to quantify inherited susceptibility to obesity affords new opportunities for clinical prevention and mechanistic assessment.


Subject(s)
Body Weight , Multifactorial Inheritance/genetics , Obesity/pathology , Adolescent , Body Mass Index , Child , Databases, Factual , Female , Genome-Wide Association Study , Humans , Infant, Newborn , Longitudinal Studies , Male , Middle Aged , Obesity/genetics , Risk Factors , Severity of Illness Index
11.
Cell ; 178(1): 202-215.e14, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31204102

ABSTRACT

Despite the worldwide success of vaccination, newborns remain vulnerable to infections. While neonatal vaccination has been hampered by maternal antibody-mediated dampening of immune responses, enhanced regulatory and tolerogenic mechanisms, and immune system immaturity, maternal pre-natal immunization aims to boost neonatal immunity via antibody transfer to the fetus. However, emerging data suggest that antibodies are not transferred equally across the placenta. To understand this, we used systems serology to define Fc features associated with antibody transfer. The Fc-profile of neonatal and maternal antibodies differed, skewed toward natural killer (NK) cell-activating antibodies. This selective transfer was linked to digalactosylated Fc-glycans that selectively bind FcRn and FCGR3A, resulting in transfer of antibodies able to efficiently leverage innate immune cells present at birth. Given emerging data that vaccination may direct antibody glycosylation, our study provides insights for the development of next-generation maternal vaccines designed to elicit antibodies that will most effectively aid neonates.


Subject(s)
Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Placenta/metabolism , Polysaccharides/metabolism , Receptors, Fc/immunology , Receptors, Fc/metabolism , Adolescent , Adult , Belgium , Cell Degranulation , Cohort Studies , Female , Glycosylation , Humans , Infant, Newborn , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Male , Pregnancy , Receptors, IgG/metabolism , THP-1 Cells , United States , Vaccination , Young Adult
12.
Cell ; 178(1): 190-201.e11, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31204101

ABSTRACT

The placental transfer of maternal IgG is critical for infant protection against infectious pathogens. However, factors that modulate the placental transfer of IgG remain largely undefined. HIV-infected women have impaired placental IgG transfer, presenting a unique "disruption model" to define factors that modulate placental IgG transfer. We measured the placental transfer efficiency of maternal HIV and pathogen-specific IgG in US and Malawian HIV-infected mothers and their HIV-exposed uninfected and infected infants. We examined the role of maternal HIV disease progression, infant factors, placental Fc receptor expression, IgG subclass, and glycan signatures and their association with placental IgG transfer efficiency. Maternal IgG characteristics, such as binding to placentally expressed Fc receptors FcγRIIa and FcγRIIIa, and Fc region glycan profiles were associated with placental IgG transfer efficiency. Our findings suggest that Fc region characteristics modulate the selective placental transfer of IgG, with implications for maternal vaccine design and infant health.


Subject(s)
HIV Infections/transmission , HIV/genetics , Immunoglobulin G/blood , Infectious Disease Transmission, Vertical , Placenta/metabolism , Pregnancy Complications, Infectious/virology , Receptors, IgG/metabolism , Cohort Studies , Disease Progression , Female , Glycosylation , HIV Infections/immunology , HIV Infections/virology , Humans , Immunoglobulin Fc Fragments/metabolism , Infant , Infant, Newborn , Malawi , Pregnancy , Pregnancy Complications, Infectious/immunology , United States , Viral Load/genetics
13.
Nat Immunol ; 22(6): 723-734, 2021 06.
Article in English | MEDLINE | ID: mdl-33958784

ABSTRACT

Continuous supply of immune cells throughout life relies on the delicate balance in the hematopoietic stem cell (HSC) pool between long-term maintenance and meeting the demands of both normal blood production and unexpected stress conditions. Here we identified distinct subsets of human long-term (LT)-HSCs that responded differently to regeneration-mediated stress: an immune checkpoint ligand CD112lo subset that exhibited a transient engraftment restraint (termed latency) before contributing to hematopoietic reconstitution and a primed CD112hi subset that responded rapidly. This functional heterogeneity and CD112 expression are regulated by INKA1 through direct interaction with PAK4 and SIRT1, inducing epigenetic changes and defining an alternative state of LT-HSC quiescence that serves to preserve self-renewal and regenerative capacity upon regeneration-mediated stress. Collectively, our data uncovered the molecular intricacies underlying HSC heterogeneity and self-renewal regulation and point to latency as an orchestrated physiological response that balances blood cell demands with preserving a stem cell reservoir.


Subject(s)
Cell Self Renewal/immunology , Hematopoietic Stem Cells/physiology , Immune Reconstitution , Multipotent Stem Cells/physiology , Stress, Physiological/immunology , Adult , Animals , Cell Self Renewal/genetics , Cells, Cultured , Epigenesis, Genetic/immunology , Female , Fetal Blood/cytology , Flow Cytometry , Gene Knockdown Techniques , Hematopoiesis , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunomagnetic Separation , Infant, Newborn , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Middle Aged , Nectins/metabolism , Primary Cell Culture , RNA-Seq , Single-Cell Analysis , Sirtuin 1/metabolism , Stress, Physiological/genetics , Transplantation, Heterologous , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
14.
Nat Immunol ; 22(12): 1490-1502, 2021 12.
Article in English | MEDLINE | ID: mdl-34616036

ABSTRACT

Despite extensive studies into severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the effect of maternal infection on the neonate is unclear. To investigate this, we characterized the immunology of neonates born to mothers with confirmed SARS-CoV-2 infection during pregnancy. Here we show that maternal SARS-CoV-2 infection affects the neonatal immune system. Despite similar proportions of B cells, CD4+ T cells and CD8+ T cells, increased percentages of natural killer cells, Vδ2+ γδ T cells and regulatory T cells were detected in neonates born to mothers with recent or ongoing infection compared with those born to recovered or uninfected mothers. Increased plasma cytokine levels were also evident in neonates and mothers within the recent or ongoing infection group. Cytokine functionality was enhanced in neonates born to SARS-CoV-2-exposed mothers, compared to those born to uninfected mothers. In most neonates, this immune imprinting was nonspecific, suggesting vertical transmission of SARS-CoV-2 is limited, a finding supported by a lack of SARS-CoV-2-specific IgM in neonates despite maternal IgG transfer.


Subject(s)
COVID-19/immunology , Infant, Newborn, Diseases/immunology , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious/immunology , SARS-CoV-2/immunology , Adult , Antibodies, Viral/immunology , COVID-19/diagnosis , COVID-19/virology , Cytokines/blood , Cytokines/immunology , Cytokines/metabolism , Female , Humans , Immunity, Innate/immunology , Immunoglobulin G/immunology , Infant, Newborn , Infant, Newborn, Diseases/diagnosis , Infant, Newborn, Diseases/virology , Killer Cells, Natural/immunology , Pregnancy , Pregnancy Complications, Infectious/virology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , SARS-CoV-2/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology
15.
Immunity ; 57(2): 200-202, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38354699

ABSTRACT

Disease-associated microglia (DAMs) are a unique microglial state in development and various CNS pathologies. In this issue of Immunity, Lan and colleagues provide novel insights into the diversity of DAMs in CNS diseases, revealing their terminal fate following juvenile stroke verses their reversible fate following neonatal stroke and their ability to maintain immune memory upon return to homeostatic states.


Subject(s)
Central Nervous System Diseases , Stroke , Infant, Newborn , Humans , Microglia
16.
Immunity ; 57(8): 1728-1730, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39142273

ABSTRACT

The immune system is imprinted by gut microbes in early life. In this issue of Immunity, Perdijk et al. show that dysregulation of airway epithelial function by neonatal antibiotic treatment can be reversed by supplementation with a depleted microbial metabolite.


Subject(s)
Anti-Bacterial Agents , Gastrointestinal Microbiome , Humans , Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/immunology , Infant, Newborn
17.
Immunity ; 57(7): 1681-1695.e4, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38876099

ABSTRACT

Respiratory syncytial virus (RSV) is among the most common causes of lower respiratory tract infection (LRTI) and hospitalization in infants. However, the mechanisms of immune control in infants remain incompletely understood. Antibody profiling against attachment (G) and fusion (F) proteins in children less than 2 years of age, with mild (outpatients) or severe (inpatients) RSV disease, indicated substantial age-dependent differences in RSV-specific immunity. Maternal antibodies were detectable for the first 3 months of life, followed by a long window of immune vulnerability between 3 and 6 months and a rapid evolution of FcγR-recruiting immunity after 6 months of age. Acutely ill hospitalized children exhibited lower G-specific antibodies compared with healthy controls. With disease resolution, RSV-infected infants generated broad functional RSV strain-specific G-responses and evolved cross-reactive F-responses, with minimal maternal imprinting. These data suggest an age-independent RSV G-specific functional humoral correlate of protection, and the evolution of RSV F-specific functional immunity with disease resolution.


Subject(s)
Antibodies, Viral , Cross Reactions , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus Infections/immunology , Infant , Antibodies, Viral/immunology , Cross Reactions/immunology , Respiratory Syncytial Virus, Human/immunology , Female , Immunity, Humoral/immunology , Viral Fusion Proteins/immunology , Longitudinal Studies , Male , Immunoglobulin G/immunology , Immunoglobulin G/blood , Infant, Newborn , Immunity, Maternally-Acquired
18.
Cell ; 174(5): 1051-1053, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30142343

ABSTRACT

The study of neonatal immunology has been hampered by lack of access to infant samples. Novel sample sparing methods and systems-wide approaches have uniquely expanded this field, demonstrating that newborn immunity varies widely but converges over the first 3 months of life. During this important time window, environmental and genetic factors impact the infant immune system and can influence lifelong immunity.


Subject(s)
Immune System , Immunity , Adolescent , Child , Humans , Infant, Newborn , Middle Aged
19.
Cell ; 174(5): 1277-1292.e14, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30142345

ABSTRACT

Epidemiological data suggest that early life exposures are key determinants of immune-mediated disease later in life. Young children are also particularly susceptible to infections, warranting more analyses of immune system development early in life. Such analyses mostly have been performed in mouse models or human cord blood samples, but these cannot account for the complex environmental exposures influencing human newborns after birth. Here, we performed longitudinal analyses in 100 newborn children, sampled up to 4 times during their first 3 months of life. From 100 µL of blood, we analyze the development of 58 immune cell populations by mass cytometry and 267 plasma proteins by immunoassays, uncovering drastic changes not predictable from cord blood measurements but following a stereotypic pattern. Preterm and term children differ at birth but converge onto a shared trajectory, seemingly driven by microbial interactions and hampered by early gut bacterial dysbiosis.


Subject(s)
Fetal Blood/immunology , Immune System/physiology , Infant, Premature/immunology , Inflammation , Cell Lineage , Dysbiosis , Female , Gastrointestinal Microbiome , Humans , Immunoassay , Infant, Newborn , Leukocytes, Mononuclear/metabolism , Longitudinal Studies , Male , Parents , Phenotype , Premature Birth/immunology , Transcriptome
20.
Immunity ; 56(3): 485-499, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36921575

ABSTRACT

The mucosal immune system of neonates goes through successive, non-redundant phases that support the developmental needs of the infant and ultimately establish immune homeostasis. These phases are informed by environmental cues, including dietary and microbial stimuli, but also evolutionary developmental programming that functions independently of external stimuli. The immune response to exogenous stimuli is tightly regulated during early life; thresholds are set within this neonatal "window of opportunity" that govern how the immune system will respond to diet, the microbiota, and pathogenic microorganisms in the future. Thus, changes in early-life exposure, such as breastfeeding or environmental and microbial stimuli, influence immunological and metabolic homeostasis and the risk of developing diseases such as asthma/allergy and obesity.


Subject(s)
Asthma , Hypersensitivity , Microbiota , Infant , Infant, Newborn , Humans , Immune System/physiology , Mucous Membrane
SELECTION OF CITATIONS
SEARCH DETAIL