Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.007.673
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Annu Rev Immunol ; 41: 127-151, 2023 04 26.
Article in English | MEDLINE | ID: mdl-36630598

ABSTRACT

The presence of granulated lymphocytes in the human uterine mucosa, known as decidua during pregnancy, or endometrium otherwise, was first noted in the nineteenth century, but it was not until 1990 that these cells were identified as a type of natural killer (NK) cell. From the outset, uterine NK (uNK) cells were found to be less cytotoxic than their circulating counterparts, peripheral NK (pNK) cells. Recently, unbiased approaches have defined three subpopulations of uNK cells, all of which cluster separately from pNK cells. Here, we review the history of research into uNK cells, including their ability to interact with placental extravillous trophoblast cells and their potential role in regulating placental implantation. We go on to review more recent advances that focus on uNK cell development and heterogeneity and their potential to defend against infection and to mediate memory effects. Finally, we consider how a better understanding of these cells could be leveraged in the future to improve outcomes of pregnancy for mothers and babies.


Subject(s)
Placenta , Uterus , Humans , Pregnancy , Female , Animals , Killer Cells, Natural/metabolism , Mucous Membrane , Decidua
2.
Cell ; 187(3): 764-781.e14, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38306985

ABSTRACT

Pregnancy induces dramatic metabolic changes in females; yet, the intricacies of this metabolic reprogramming remain poorly understood, especially in primates. Using cynomolgus monkeys, we constructed a comprehensive multi-tissue metabolome atlas, analyzing 273 samples from 23 maternal tissues during pregnancy. We discovered a decline in metabolic coupling between tissues as pregnancy progressed. Core metabolic pathways that were rewired during primate pregnancy included steroidogenesis, fatty acid metabolism, and arachidonic acid metabolism. Our atlas revealed 91 pregnancy-adaptive metabolites changing consistently across 23 tissues, whose roles we verified in human cell models and patient samples. Corticosterone and palmitoyl-carnitine regulated placental maturation and maternal tissue progenitors, respectively, with implications for maternal preeclampsia, diabetes, cardiac hypertrophy, and muscle and liver regeneration. Moreover, we found that corticosterone deficiency induced preeclampsia-like inflammation, indicating the atlas's potential clinical value. Overall, our multi-tissue metabolome atlas serves as a framework for elucidating the role of metabolic regulation in female health during pregnancy.


Subject(s)
Metabolomics , Pregnancy , Animals , Female , Humans , Pregnancy/metabolism , Corticosterone/metabolism , Metabolome/physiology , Placenta/metabolism , Pre-Eclampsia , Primates/metabolism
3.
Cell ; 187(6): 1402-1421.e21, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38428422

ABSTRACT

Neonates are highly susceptible to inflammation and infection. Here, we investigate how late fetal liver (FL) mouse hematopoietic stem and progenitor cells (HSPCs) respond to inflammation, testing the hypothesis that deficits in the engagement of emergency myelopoiesis (EM) pathways limit neutrophil output and contribute to perinatal neutropenia. We show that fetal HSPCs have limited production of myeloid cells at steady state and fail to activate a classical adult-like EM transcriptional program. Moreover, we find that fetal HSPCs can respond to EM-inducing inflammatory stimuli in vitro but are restricted by maternal anti-inflammatory factors, primarily interleukin-10 (IL-10), from activating EM pathways in utero. Accordingly, we demonstrate that the loss of maternal IL-10 restores EM activation in fetal HSPCs but at the cost of fetal demise. These results reveal the evolutionary trade-off inherent in maternal anti-inflammatory responses that maintain pregnancy but render the fetus unresponsive to EM activation signals and susceptible to infection.


Subject(s)
Inflammation , Interleukin-10 , Myelopoiesis , Animals , Mice , Pregnancy/immunology , Fetus , Hematopoiesis , Hematopoietic Stem Cells/cytology , Inflammation/immunology , Interleukin-10/immunology , Animals, Newborn , Female
4.
Cell ; 187(1): 204-215.e14, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38070508

ABSTRACT

Mounting evidence suggests metabolism instructs stem cell fate decisions. However, how fetal metabolism changes during development and how altered maternal metabolism shapes fetal metabolism remain unexplored. We present a descriptive atlas of in vivo fetal murine metabolism during mid-to-late gestation in normal and diabetic pregnancy. Using 13C-glucose and liquid chromatography-mass spectrometry (LC-MS), we profiled the metabolism of fetal brains, hearts, livers, and placentas harvested from pregnant dams between embryonic days (E)10.5 and 18.5. Our analysis revealed metabolic features specific to a hyperglycemic environment and signatures that may denote developmental transitions during euglycemic development. We observed sorbitol accumulation in fetal tissues and altered neurotransmitter levels in fetal brains isolated from hyperglycemic dams. Tracing 13C-glucose revealed disparate fetal nutrient sourcing depending on maternal glycemic states. Regardless of glycemic state, histidine-derived metabolites accumulated in late-stage fetal tissues. Our rich dataset presents a comprehensive overview of in vivo fetal tissue metabolism and alterations due to maternal hyperglycemia.


Subject(s)
Diabetes Mellitus , Diabetes, Gestational , Fetus , Animals , Female , Mice , Pregnancy , Diabetes Mellitus/metabolism , Fetus/metabolism , Glucose/metabolism , Placenta/metabolism , Diabetes, Gestational/metabolism
5.
Cell ; 187(8): 1853-1873.e15, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38574728

ABSTRACT

This study has followed a birth cohort for over 20 years to find factors associated with neurodevelopmental disorder (ND) diagnosis. Detailed, early-life longitudinal questionnaires captured infection and antibiotic events, stress, prenatal factors, family history, and more. Biomarkers including cord serum metabolome and lipidome, human leukocyte antigen (HLA) genotype, infant microbiota, and stool metabolome were assessed. Among the 16,440 Swedish children followed across time, 1,197 developed an ND. Significant associations emerged for future ND diagnosis in general and for specific ND subtypes, spanning intellectual disability, speech disorder, attention-deficit/hyperactivity disorder, and autism. This investigation revealed microbiome connections to future diagnosis as well as early emerging mood and gastrointestinal problems. The findings suggest links to immunodysregulation and metabolism, compounded by stress, early-life infection, and antibiotics. The convergence of infant biomarkers and risk factors in this prospective, longitudinal study on a large-scale population establishes a foundation for early-life prediction and intervention in neurodevelopment.


Subject(s)
Biomarkers , Gastrointestinal Microbiome , Neurodevelopmental Disorders , Child , Female , Humans , Infant , Pregnancy , Autism Spectrum Disorder/microbiology , Longitudinal Studies , Prospective Studies , Feces/microbiology , Mood Disorders/microbiology
6.
Cell ; 187(4): 981-998.e25, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38325365

ABSTRACT

The female reproductive tract (FRT) undergoes extensive remodeling during reproductive cycling. This recurrent remodeling and how it shapes organ-specific aging remains poorly explored. Using single-cell and spatial transcriptomics, we systematically characterized morphological and gene expression changes occurring in ovary, oviduct, uterus, cervix, and vagina at each phase of the mouse estrous cycle, during decidualization, and into aging. These analyses reveal that fibroblasts play central-and highly organ-specific-roles in FRT remodeling by orchestrating extracellular matrix (ECM) reorganization and inflammation. Our results suggest a model wherein recurrent FRT remodeling over reproductive lifespan drives the gradual, age-related development of fibrosis and chronic inflammation. This hypothesis was directly tested using chemical ablation of cycling, which reduced fibrotic accumulation during aging. Our atlas provides extensive detail into how estrus, pregnancy, and aging shape the organs of the female reproductive tract and reveals the unexpected cost of the recurrent remodeling required for reproduction.


Subject(s)
Aging , Genitalia, Female , Animals , Female , Mice , Pregnancy , Genitalia, Female/cytology , Genitalia, Female/metabolism , Inflammation/metabolism , Uterus/cytology , Vagina/cytology , Single-Cell Analysis
7.
Cell ; 187(12): 2952-2968.e13, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38795705

ABSTRACT

Recent studies suggest that human-associated bacteria interact with host-produced steroids, but the mechanisms and physiological impact of such interactions remain unclear. Here, we show that the human gut bacteria Gordonibacter pamelaeae and Eggerthella lenta convert abundant biliary corticoids into progestins through 21-dehydroxylation, thereby transforming a class of immuno- and metabo-regulatory steroids into a class of sex hormones and neurosteroids. Using comparative genomics, homologous expression, and heterologous expression, we identify a bacterial gene cluster that performs 21-dehydroxylation. We also uncover an unexpected role for hydrogen gas production by gut commensals in promoting 21-dehydroxylation, suggesting that hydrogen modulates secondary metabolism in the gut. Levels of certain bacterial progestins, including allopregnanolone, better known as brexanolone, an FDA-approved drug for postpartum depression, are substantially increased in feces from pregnant humans. Thus, bacterial conversion of corticoids into progestins may affect host physiology, particularly in the context of pregnancy and women's health.


Subject(s)
Gastrointestinal Microbiome , Glucocorticoids , Hydrogen , Progestins , Humans , Progestins/metabolism , Hydrogen/metabolism , Female , Glucocorticoids/metabolism , Pregnancy , Animals , Multigene Family , Feces/microbiology , Pregnanolone/metabolism , Mice
8.
Cell ; 186(23): 4996-5014.e24, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37949056

ABSTRACT

A formal demonstration that mammalian pluripotent stem cells possess preimplantation embryonic cell-like (naive) pluripotency is the generation of chimeric animals through early embryo complementation with homologous cells. Whereas such naive pluripotency has been well demonstrated in rodents, poor chimerism has been achieved in other species including non-human primates due to the inability of the donor cells to match the developmental state of the host embryos. Here, we have systematically tested various culture conditions for establishing monkey naive embryonic stem cells and optimized the procedures for chimeric embryo culture. This approach generated an aborted fetus and a live chimeric monkey with high donor cell contribution. A stringent characterization pipeline demonstrated that donor cells efficiently (up to 90%) incorporated into various tissues (including the gonads and placenta) of the chimeric monkeys. Our results have major implications for the study of primate naive pluripotency and genetic engineering of non-human primates.


Subject(s)
Embryonic Stem Cells , Genetic Engineering , Haplorhini , Animals , Female , Pregnancy , Haplorhini/genetics , Live Birth , Mammals , Pluripotent Stem Cells , Primates , Genetic Engineering/methods
9.
Cell ; 186(18): 3755-3757, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37657417

ABSTRACT

The second week of embryonic development is a critical phase of the human life cycle and one that has been largely inaccessible to scientific investigation. Recent studies of human embryo models built from stem cells promise to yield dramatic insights into the key events of cell specification and morphogenesis that occur during this brief window of embryogenesis.


Subject(s)
Embryo, Mammalian , Embryonic Development , Female , Pregnancy , Animals , Humans , Life Cycle Stages , Morphogenesis , Stem Cells
10.
Cell ; 186(9): 1930-1949.e31, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37071993

ABSTRACT

Cortical circuits are composed predominantly of pyramidal-to-pyramidal neuron connections, yet their assembly during embryonic development is not well understood. We show that mouse embryonic Rbp4-Cre cortical neurons, transcriptomically closest to layer 5 pyramidal neurons, display two phases of circuit assembly in vivo. At E14.5, they form a multi-layered circuit motif, composed of only embryonic near-projecting-type neurons. By E17.5, this transitions to a second motif involving all three embryonic types, analogous to the three adult layer 5 types. In vivo patch clamp recordings and two-photon calcium imaging of embryonic Rbp4-Cre neurons reveal active somas and neurites, tetrodotoxin-sensitive voltage-gated conductances, and functional glutamatergic synapses, from E14.5 onwards. Embryonic Rbp4-Cre neurons strongly express autism-associated genes and perturbing these genes interferes with the switch between the two motifs. Hence, pyramidal neurons form active, transient, multi-layered pyramidal-to-pyramidal circuits at the inception of neocortex, and studying these circuits could yield insights into the etiology of autism.


Subject(s)
Autistic Disorder , Neocortex , Pyramidal Cells , Animals , Female , Mice , Pregnancy , Autistic Disorder/genetics , Autistic Disorder/pathology , Mutation , Neocortex/physiology , Neurons/physiology , Pyramidal Cells/physiology
11.
Cell ; 186(18): 3776-3792.e16, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37478861

ABSTRACT

In vitro stem cell models that replicate human gastrulation have been generated, but they lack the essential extraembryonic cells needed for embryonic development, morphogenesis, and patterning. Here, we describe a robust and efficient method that prompts human extended pluripotent stem cells to self-organize into embryo-like structures, termed peri-gastruloids, which encompass both embryonic (epiblast) and extraembryonic (hypoblast) tissues. Although peri-gastruloids are not viable due to the exclusion of trophoblasts, they recapitulate critical stages of human peri-gastrulation development, such as forming amniotic and yolk sac cavities, developing bilaminar and trilaminar embryonic discs, specifying primordial germ cells, initiating gastrulation, and undergoing early neurulation and organogenesis. Single-cell RNA-sequencing unveiled transcriptomic similarities between advanced human peri-gastruloids and primary peri-gastrulation cell types found in humans and non-human primates. This peri-gastruloid platform allows for further exploration beyond gastrulation and may potentially aid in the development of human fetal tissues for use in regenerative medicine.


Subject(s)
Embryo Implantation , Gastrulation , Pluripotent Stem Cells , Animals , Female , Humans , Pregnancy , Cell Differentiation , Embryo, Mammalian , Embryonic Development , Organogenesis , Pluripotent Stem Cells/metabolism , Primates
12.
Cell ; 186(24): 5308-5327.e25, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37922900

ABSTRACT

Mammalian oocytes are filled with poorly understood structures called cytoplasmic lattices. First discovered in the 1960s and speculated to correspond to mammalian yolk, ribosomal arrays, or intermediate filaments, their function has remained enigmatic to date. Here, we show that cytoplasmic lattices are sites where oocytes store essential proteins for early embryonic development. Using super-resolution light microscopy and cryoelectron tomography, we show that cytoplasmic lattices are composed of filaments with a high surface area, which contain PADI6 and subcortical maternal complex proteins. The lattices associate with many proteins critical for embryonic development, including proteins that control epigenetic reprogramming of the preimplantation embryo. Loss of cytoplasmic lattices by knocking out PADI6 or the subcortical maternal complex prevents the accumulation of these proteins and results in early embryonic arrest. Our work suggests that cytoplasmic lattices enrich maternally provided proteins to prevent their premature degradation and cellular activity, thereby enabling early mammalian development.


Subject(s)
Oocytes , Proteins , Pregnancy , Animals , Female , Oocytes/metabolism , Proteins/metabolism , Embryo, Mammalian/metabolism , Cytoskeleton , Ribosomes , Embryonic Development , Mammals
13.
Cell ; 186(15): 3166-3181.e18, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37413989

ABSTRACT

Proper preimplantation development is essential to assemble a blastocyst capable of implantation. Live imaging has uncovered major events driving early development in mouse embryos; yet, studies in humans have been limited by restrictions on genetic manipulation and lack of imaging approaches. We have overcome this barrier by combining fluorescent dyes with live imaging to reveal the dynamics of chromosome segregation, compaction, polarization, blastocyst formation, and hatching in the human embryo. We also show that blastocyst expansion mechanically constrains trophectoderm cells, causing nuclear budding and DNA shedding into the cytoplasm. Furthermore, cells with lower perinuclear keratin levels are more prone to undergo DNA loss. Moreover, applying trophectoderm biopsy, a mechanical procedure performed clinically for genetic testing, increases DNA shedding. Thus, our work reveals distinct processes underlying human development compared with mouse and suggests that aneuploidies in human embryos may not only originate from chromosome segregation errors during mitosis but also from nuclear DNA shedding.


Subject(s)
Preimplantation Diagnosis , Pregnancy , Female , Humans , Animals , Mice , Preimplantation Diagnosis/methods , Blastocyst , Embryo Implantation , Genetic Testing/methods , Aneuploidy , Biopsy/methods
14.
Cell ; 186(6): 1212-1229.e21, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36827974

ABSTRACT

Mitochondrial activity differs markedly between organs, but it is not known how and when this arises. Here we show that cell lineage-specific expression profiles involving essential mitochondrial genes emerge at an early stage in mouse development, including tissue-specific isoforms present before organ formation. However, the nuclear transcriptional signatures were not independent of organelle function. Genetically disrupting intra-mitochondrial protein synthesis with two different mtDNA mutations induced cell lineage-specific compensatory responses, including molecular pathways not previously implicated in organellar maintenance. We saw downregulation of genes whose expression is known to exacerbate the effects of exogenous mitochondrial toxins, indicating a transcriptional adaptation to mitochondrial dysfunction during embryonic development. The compensatory pathways were both tissue and mutation specific and under the control of transcription factors which promote organelle resilience. These are likely to contribute to the tissue specificity which characterizes human mitochondrial diseases and are potential targets for organ-directed treatments.


Subject(s)
Mitochondria , Organogenesis , Animals , Female , Humans , Mice , Pregnancy , Cell Lineage , DNA, Mitochondrial/genetics , Mitochondria/metabolism , Mitochondrial Diseases , Organ Specificity , Embryonic Development , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism
15.
Cell ; 186(9): 1950-1967.e25, 2023 04 27.
Article in English | MEDLINE | ID: mdl-36996814

ABSTRACT

Little is known about the critical metabolic changes that neural cells have to undergo during development and how temporary shifts in this program can influence brain circuitries and behavior. Inspired by the discovery that mutations in SLC7A5, a transporter of metabolically essential large neutral amino acids (LNAAs), lead to autism, we employed metabolomic profiling to study the metabolic states of the cerebral cortex across different developmental stages. We found that the forebrain undergoes significant metabolic remodeling throughout development, with certain groups of metabolites showing stage-specific changes, but what are the consequences of perturbing this metabolic program? By manipulating Slc7a5 expression in neural cells, we found that the metabolism of LNAAs and lipids are interconnected in the cortex. Deletion of Slc7a5 in neurons affects the postnatal metabolic state, leading to a shift in lipid metabolism. Additionally, it causes stage- and cell-type-specific alterations in neuronal activity patterns, resulting in a long-term circuit dysfunction.


Subject(s)
Amino Acids, Neutral , Large Neutral Amino Acid-Transporter 1 , Female , Humans , Pregnancy , Amino Acids, Neutral/genetics , Amino Acids, Neutral/metabolism , Brain/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Mutation , Neurons/metabolism , Animals , Mice
16.
Cell ; 185(20): 3645-3647, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179664

ABSTRACT

Fetal human brain stem cell niches that contain multipotent neural progenitors are progressively vascularized during development. Crouch et al. (Crouch et al., 2022) report endothelial and mural lineage trajectories that build developing prenatal vascular in second trimester fetal brain. This cerebral angiogenesis in neural progenitor zones occurs simultaneously with and can promote neurogenesis.


Subject(s)
Neurogenesis , Stem Cells , Brain , Cell Lineage , Female , Humans , Pregnancy
17.
Cell ; 185(21): 3849-3853, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36174580

ABSTRACT

The 2022 Lasker-DeBakey Clinical Medical Research Award is presented to Yuk Ming Dennis Lo of the Chinese University of Hong Kong for the discovery of fetal DNA in maternal blood, leading to development of noninvasive prenatal testing for Down syndrome.


Subject(s)
Awards and Prizes , Biomedical Research , Midwifery , DNA , Female , Fetus , Humans , Pregnancy
18.
Cell ; 185(25): 4826-4840.e17, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36402135

ABSTRACT

Congenital Zika virus (ZIKV) infection results in neurodevelopmental deficits in up to 14% of infants born to ZIKV-infected mothers. Neutralizing antibodies are a critical component of protective immunity. Here, we demonstrate that plasma IgM contributes to ZIKV immunity in pregnancy, mediating neutralization up to 3 months post-symptoms. From a ZIKV-infected pregnant woman, we isolated a pentameric ZIKV-specific IgM (DH1017.IgM) that exhibited ultrapotent ZIKV neutralization dependent on the IgM isotype. DH1017.IgM targets an envelope dimer epitope within domain II. The epitope arrangement on the virion is compatible with concurrent engagement of all ten antigen-binding sites of DH1017.IgM, a solution not available to IgG. DH1017.IgM protected mice against viremia upon lethal ZIKV challenge more efficiently than when expressed as an IgG. Our findings identify a role for antibodies of the IgM isotype in protection against ZIKV and posit DH1017.IgM as a safe and effective candidate immunotherapeutic, particularly during pregnancy.


Subject(s)
Immunoglobulin M , Pregnancy , Zika Virus Infection , Zika Virus , Animals , Female , Mice , Pregnancy/immunology , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Neutralization Tests , Zika Virus Infection/immunology , Immunoglobulin M/immunology , Immunoglobulin M/isolation & purification
19.
Cell ; 185(23): 4428-4447.e28, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36318921

ABSTRACT

Human brain development is underpinned by cellular and molecular reconfigurations continuing into the third decade of life. To reveal cell dynamics orchestrating neural maturation, we profiled human prefrontal cortex gene expression and chromatin accessibility at single-cell resolution from gestation to adulthood. Integrative analyses define the dynamic trajectories of each cell type, revealing major gene expression reconfiguration at the prenatal-to-postnatal transition in all cell types followed by continuous reconfiguration into adulthood and identifying regulatory networks guiding cellular developmental programs, states, and functions. We uncover links between expression dynamics and developmental milestones, characterize the diverse timing of when cells acquire adult-like states, and identify molecular convergence from distinct developmental origins. We further reveal cellular dynamics and their regulators implicated in neurological disorders. Finally, using this reference, we benchmark cell identities and maturation states in organoid models. Together, this captures the dynamic regulatory landscape of human cortical development.


Subject(s)
Neurogenesis , Organoids , Pregnancy , Female , Humans , Adult , Chromatin , Prefrontal Cortex , Single-Cell Analysis , Gene Regulatory Networks
20.
Cell ; 185(26): 4921-4936.e15, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36563663

ABSTRACT

The perinatal period represents a critical window for cognitive and immune system development, promoted by maternal and infant gut microbiomes and their metabolites. Here, we tracked the co-development of microbiomes and metabolomes from late pregnancy to 1 year of age using longitudinal multi-omics data from a cohort of 70 mother-infant dyads. We discovered large-scale mother-to-infant interspecies transfer of mobile genetic elements, frequently involving genes associated with diet-related adaptations. Infant gut metabolomes were less diverse than maternal but featured hundreds of unique metabolites and microbe-metabolite associations not detected in mothers. Metabolomes and serum cytokine signatures of infants who received regular-but not extensively hydrolyzed-formula were distinct from those of exclusively breastfed infants. Taken together, our integrative analysis expands the concept of vertical transmission of the gut microbiome and provides original insights into the development of maternal and infant microbiomes and metabolomes during late pregnancy and early life.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Female , Humans , Infant , Pregnancy , Gastrointestinal Microbiome/genetics , Microbiota/genetics , Mothers , Breast Feeding , Feces , Interspersed Repetitive Sequences
SELECTION OF CITATIONS
SEARCH DETAIL