Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39.074
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(6): 1402-1421.e21, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38428422

RESUMEN

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.


Asunto(s)
Inflamación , Interleucina-10 , Mielopoyesis , Animales , Ratones , Embarazo/inmunología , Feto , Hematopoyesis , Células Madre Hematopoyéticas/citología , Inflamación/inmunología , Interleucina-10/inmunología , Animales Recién Nacidos , Femenino
2.
Cell ; 186(24): 5308-5327.e25, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37922900

RESUMEN

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.


Asunto(s)
Oocitos , Proteínas , Embarazo , Animales , Femenino , Oocitos/metabolismo , Proteínas/metabolismo , Embrión de Mamíferos/metabolismo , Citoesqueleto , Ribosomas , Desarrollo Embrionario , Mamíferos
3.
Cell ; 185(4): 654-671.e22, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35065713

RESUMEN

Sex hormones exert a profound influence on gendered behaviors. How individual sex hormone-responsive neuronal populations regulate diverse sex-typical behaviors is unclear. We performed orthogonal, genetically targeted sequencing of four estrogen receptor 1-expressing (Esr1+) populations and identified 1,415 genes expressed differentially between sexes or estrous states. Unique subsets of these genes were distributed across all 137 transcriptomically defined Esr1+ cell types, including estrous stage-specific ones, that comprise the four populations. We used differentially expressed genes labeling single Esr1+ cell types as entry points to functionally characterize two such cell types, BNSTprTac1/Esr1 and VMHvlCckar/Esr1. We observed that these two cell types, but not the other Esr1+ cell types in these populations, are essential for sex recognition in males and mating in females, respectively. Furthermore, VMHvlCckar/Esr1 cell type projections are distinct from those of other VMHvlEsr1 cell types. Together, projection and functional specialization of dimorphic cell types enables sex hormone-responsive populations to regulate diverse social behaviors.


Asunto(s)
Ciclo Estral/genética , Regulación de la Expresión Génica , Caracteres Sexuales , Conducta Sexual Animal/fisiología , Agresión , Animales , Aromatasa/metabolismo , Trastorno Autístico/genética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neuronas/metabolismo , Conducta Social
4.
Cell ; 181(6): 1276-1290.e13, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32402238

RESUMEN

At the species level, immunity depends on the selection and transmission of protective components of the immune system. A microbe-induced population of RORγ-expressing regulatory T cells (Tregs) is essential in controlling gut inflammation. We uncovered a non-genetic, non-epigenetic, non-microbial mode of transmission of their homeostatic setpoint. RORγ+ Treg proportions varied between inbred mouse strains, a trait transmitted by the mother during a tight age window after birth but stable for life, resistant to many microbial or cellular perturbations, then further transferred by females for multiple generations. RORγ+ Treg proportions negatively correlated with IgA production and coating of gut commensals, traits also subject to maternal transmission, in an immunoglobulin- and RORγ+ Treg-dependent manner. We propose a model based on a double-negative feedback loop, vertically transmitted via the entero-mammary axis. This immunologic mode of multi-generational transmission may provide adaptability and modulate the genetic tuning of gut immune responses and inflammatory disease susceptibility.


Asunto(s)
Sistema Digestivo/inmunología , Linfocitos T Reguladores/inmunología , Animales , Susceptibilidad a Enfermedades/inmunología , Femenino , Microbioma Gastrointestinal/inmunología , Homeostasis/inmunología , Inmunoglobulina A/inmunología , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos NOD , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología
5.
Cell ; 182(5): 1125-1139.e18, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32822574

RESUMEN

Maternal decidual NK (dNK) cells promote placentation, but how they protect against placental infection while maintaining fetal tolerance is unclear. Here we show that human dNK cells highly express the antimicrobial peptide granulysin (GNLY) and selectively transfer it via nanotubes to extravillous trophoblasts to kill intracellular Listeria monocytogenes (Lm) without killing the trophoblast. Transfer of GNLY, but not other cell death-inducing cytotoxic granule proteins, strongly inhibits Lm in human placental cultures and in mouse and human trophoblast cell lines. Placental and fetal Lm loads are lower and pregnancy success is greatly improved in pregnant Lm-infected GNLY-transgenic mice than in wild-type mice that lack GNLY. This immune defense is not restricted to pregnancy; peripheral NK (pNK) cells also transfer GNLY to kill bacteria in macrophages and dendritic cells without killing the host cell. Nanotube transfer of GNLY allows dNK to protect against infection while leaving the maternal-fetal barrier intact.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Bacterias/inmunología , Movimiento Celular/inmunología , Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Células HeLa , Humanos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Placenta/inmunología , Placenta/microbiología , Embarazo , Ratas , Células THP-1 , Trofoblastos/microbiología
6.
Cell ; 183(2): 324-334.e5, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33007265

RESUMEN

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.


Asunto(s)
Trasplante de Microbiota Fecal/métodos , Heces/microbiología , Microbioma Gastrointestinal/fisiología , Adulto , Cesárea/efectos adversos , Parto Obstétrico , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Microbiota/fisiología , Madres , Embarazo , Prueba de Estudio Conceptual , Vagina/microbiología
7.
Cell ; 178(1): 202-215.e14, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31204102

RESUMEN

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.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina G/metabolismo , Placenta/metabolismo , Polisacáridos/metabolismo , Receptores Fc/inmunología , Receptores Fc/metabolismo , Adolescente , Adulto , Bélgica , Degranulación de la Célula , Estudios de Cohortes , Femenino , Glicosilación , Humanos , Recién Nacido , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Masculino , Embarazo , Receptores de IgG/metabolismo , Células THP-1 , Estados Unidos , Vacunación , Adulto Joven
8.
Cell ; 178(1): 190-201.e11, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31204101

RESUMEN

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.


Asunto(s)
Infecciones por VIH/transmisión , VIH/genética , Inmunoglobulina G/sangre , Transmisión Vertical de Enfermedad Infecciosa , Placenta/metabolismo , Complicaciones Infecciosas del Embarazo/virología , Receptores de IgG/metabolismo , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Glicosilación , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Lactante , Recién Nacido , Malaui , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Estados Unidos , Carga Viral/genética
9.
Cell ; 178(1): 44-59.e7, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31104844

RESUMEN

Hypothalamic Agrp neurons regulate food ingestion in adult mice. Whether these neurons are functional before animals start to ingest food is unknown. Here, we studied the functional ontogeny of Agrp neurons during breastfeeding using postnatal day 10 mice. In contrast to adult mice, we show that isolation from the nursing nest, not milk deprivation or ingestion, activated Agrp neurons. Non-nutritive suckling and warm temperatures blunted this effect. Using in vivo fiber photometry, neonatal Agrp neurons showed a rapid increase in activity upon isolation from the nest, an effect rapidly diminished following reunion with littermates. Neonates unable to release GABA from Agrp neurons expressed blunted emission of isolation-induced ultrasonic vocalizations. Chemogenetic overactivation of these neurons further increased emission of these ultrasonic vocalizations, but not milk ingestion. We uncovered important functional properties of hypothalamic Agrp neurons during mouse development, suggesting these neurons facilitate offspring-to-caregiver bonding.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Conducta Alimentaria/fisiología , Hipotálamo/citología , Neuronas/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Animales Recién Nacidos , Ingestión de Alimentos/fisiología , Conducta Materna/fisiología , Ratones , Ratones Noqueados , Leche , Proteínas Proto-Oncogénicas c-fos/metabolismo , Aislamiento Social , Conducta en la Lactancia/fisiología , Temperatura , Vocalización Animal/fisiología , Ácido gamma-Aminobutírico/metabolismo
10.
Annu Rev Cell Dev Biol ; 36: 441-468, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32722920

RESUMEN

Fetal neurodevelopment in utero is profoundly shaped by both systemic maternal immunity and local processes at the maternal-fetal interface. Immune pathways are a critical participant in the normal physiology of pregnancy and perturbations of maternal immunity due to infections during this period have been increasingly linked to a diverse array of poor neurological outcomes, including diseases that manifest much later in postnatal life. While experimental models of maternal immune activation (MIA) have provided groundbreaking characterizations of the maternal pathways underlying pathogenesis, less commonly examined are the immune factors that serve pathogen-independent developmental functions in the embryo and fetus. In this review, we explore what is known about the in vivo role of immune factors in fetal neurodevelopment during normal pregnancy and provide an overview of how MIA perturbs the proper orchestration of this sequence of events. Finally, we discuss how the dysregulation of immune factors may contribute to the manifestation of a variety of neurological disorders.


Asunto(s)
Feto/embriología , Feto/inmunología , Factores Inmunológicos/metabolismo , Sistema Nervioso/embriología , Sistema Nervioso/inmunología , Animales , Citocinas/metabolismo , Femenino , Humanos , Intercambio Materno-Fetal/inmunología , Modelos Biológicos , Embarazo
11.
Cell ; 169(2): 216-228.e19, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28388407

RESUMEN

Chromatin architecture is fundamental in regulating gene expression. To investigate when spatial genome organization is first established during development, we examined chromatin conformation during Drosophila embryogenesis and observed the emergence of chromatin architecture within a tight time window that coincides with the onset of transcription activation in the zygote. Prior to zygotic genome activation, the genome is mostly unstructured. Early expressed genes serve as nucleation sites for topologically associating domain (TAD) boundaries. Activation of gene expression coincides with the establishment of TADs throughout the genome and co-localization of housekeeping gene clusters, which remain stable in subsequent stages of development. However, the appearance of TAD boundaries is independent of transcription and requires the transcription factor Zelda for locus-specific TAD boundary insulation. These results offer insight into when spatial organization of the genome emerges and identify a key factor that helps trigger this architecture.


Asunto(s)
Cromatina/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Genoma de los Insectos , Activación Transcripcional , Cigoto/metabolismo , Animales , Proteínas de Drosophila/metabolismo , Embrión no Mamífero/metabolismo , Genes Esenciales , Proteínas Nucleares , ARN Polimerasa II/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcripción Genética
12.
Genes Dev ; 38(3-4): 131-150, 2024 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-38453481

RESUMEN

Maternal inactivation of genes encoding components of the subcortical maternal complex (SCMC) and its associated member, PADI6, generally results in early embryo lethality. In humans, SCMC gene variants were found in the healthy mothers of children affected by multilocus imprinting disturbances (MLID). However, how the SCMC controls the DNA methylation required to regulate imprinting remains poorly defined. We generated a mouse line carrying a Padi6 missense variant that was identified in a family with Beckwith-Wiedemann syndrome and MLID. If homozygous in female mice, this variant resulted in interruption of embryo development at the two-cell stage. Single-cell multiomic analyses demonstrated defective maturation of Padi6 mutant oocytes and incomplete DNA demethylation, down-regulation of zygotic genome activation (ZGA) genes, up-regulation of maternal decay genes, and developmental delay in two-cell embryos developing from Padi6 mutant oocytes but little effect on genomic imprinting. Western blotting and immunofluorescence analyses showed reduced levels of UHRF1 in oocytes and abnormal localization of DNMT1 and UHRF1 in both oocytes and zygotes. Treatment with 5-azacytidine reverted DNA hypermethylation but did not rescue the developmental arrest of mutant embryos. Taken together, this study demonstrates that PADI6 controls both nuclear and cytoplasmic oocyte processes that are necessary for preimplantation epigenetic reprogramming and ZGA.


Asunto(s)
Oocitos , Cigoto , Animales , Niño , Femenino , Humanos , Ratones , Proteínas Potenciadoras de Unión a CCAAT/genética , Citoplasma/genética , Citoplasma/metabolismo , Metilación de ADN/genética , Desarrollo Embrionario/genética , Impresión Genómica/genética , Ubiquitina-Proteína Ligasas/metabolismo
13.
Immunity ; 55(1): 145-158.e7, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34879222

RESUMEN

Children with autism spectrum disorders often display dysregulated immune responses and related gastrointestinal symptoms. However, the underlying mechanisms leading to the development of both phenotypes have not been elucidated. Here, we show that mouse offspring exhibiting autism-like phenotypes due to prenatal exposure to maternal inflammation were more susceptible to developing intestinal inflammation following challenges later in life. In contrast to its prenatal role in neurodevelopmental phenotypes, interleukin-17A (IL-17A) generated immune-primed phenotypes in offspring through changes in the maternal gut microbiota that led to postnatal alterations in the chromatin landscape of naive CD4+ T cells. The transfer of stool samples from pregnant mice with enhanced IL-17A responses into germ-free dams produced immune-primed phenotypes in offspring. Our study provides mechanistic insights into why children exposed to heightened inflammation in the womb might have an increased risk of developing inflammatory diseases in addition to neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista/inmunología , Linfocitos T CD4-Positivos/inmunología , Cromatina/metabolismo , Microbioma Gastrointestinal/inmunología , Inflamación/inmunología , Interleucina-17/metabolismo , Intestinos/inmunología , Trastornos del Neurodesarrollo/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Animales , Trastorno del Espectro Autista/microbiología , Niño , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Femenino , Humanos , Inmunización , Inflamación/microbiología , Ratones , Trastornos del Neurodesarrollo/microbiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/microbiología
14.
Cell ; 165(2): 396-409, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27020753

RESUMEN

Multiple division cycles without growth are a characteristic feature of early embryogenesis. The female germline loads proteins and RNAs into oocytes to support these divisions, which lack many quality control mechanisms operating in somatic cells undergoing growth. Here, we describe a small RNA-Argonaute pathway that ensures early embryonic divisions in C. elegans by employing catalytic slicing activity to broadly tune, instead of silence, germline gene expression. Misregulation of one target, a kinesin-13 microtubule depolymerase, underlies a major phenotype associated with pathway loss. Tuning of target transcript levels is guided by the density of homologous small RNAs, whose generation must ultimately be related to target sequence. Thus, the tuning action of a small RNA-catalytic Argonaute pathway generates oocytes capable of supporting embryogenesis. We speculate that the specialized nature of germline chromatin led to the emergence of small RNA-catalytic Argonaute pathways in the female germline as a post-transcriptional control layer to optimize oocyte composition.


Asunto(s)
Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Embrión no Mamífero/metabolismo , Redes y Vías Metabólicas , Oocitos/metabolismo , Animales , Proteínas Argonautas/metabolismo , Secuencia de Bases , Caenorhabditis elegans/citología , Proteínas de Caenorhabditis elegans/metabolismo , División Celular , Embrión no Mamífero/citología , Desarrollo Embrionario , Femenino , Cinesinas/metabolismo , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Procesamiento Postranscripcional del ARN
15.
Genes Dev ; 37(9-10): 418-431, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37257918

RESUMEN

Translation of maternal mRNAs is detected before transcription of zygotic genes and is essential for mammalian embryo development. How certain maternal mRNAs are selected for translation instead of degradation and how this burst of translation affects zygotic genome activation remain unknown. Using gene-edited mice, we document that the oocyte-specific eukaryotic translation initiation factor 4E family member 1b (eIF4E1b) is the regulator of maternal mRNA expression that ensures subsequent reprogramming of the zygotic genome. In oocytes, eIF4E1b binds to transcripts encoding translation machinery proteins, chromatin remodelers, and reprogramming factors to promote their translation in zygotes and protect them from degradation. The protein products are thought to establish an open chromatin landscape in one-cell zygotes to enable transcription of genes required for cleavage stage development. Our results define a program for rapid resetting of the zygotic epigenome that is regulated by maternal mRNA expression and provide new insights into the mammalian maternal-to-zygotic transition.


Asunto(s)
ARN Mensajero Almacenado , Cigoto , Animales , Ratones , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Oocitos , Biosíntesis de Proteínas , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Cigoto/metabolismo
16.
Immunity ; 54(11): 2611-2631.e8, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758338

RESUMEN

Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.


Asunto(s)
Hipocampo/metabolismo , Interleucina-6/biosíntesis , Exposición Materna , Neuronas/metabolismo , Efectos Tardíos de la Exposición Prenatal , Sinapsis/metabolismo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Hipocampo/fisiopatología , Mediadores de Inflamación/metabolismo , Ratones , Embarazo , Transducción de Señal , Transmisión Sináptica
17.
Mol Cell ; 82(14): 2650-2665.e12, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35662397

RESUMEN

Coenzyme A (CoA) is essential for metabolism and protein acetylation. Current knowledge holds that each cell obtains CoA exclusively through biosynthesis via the canonical five-step pathway, starting with pantothenate uptake. However, recent studies have suggested the presence of additional CoA-generating mechanisms, indicating a more complex system for CoA homeostasis. Here, we uncovered pathways for CoA generation through inter-organismal flows of CoA precursors. Using traceable compounds and fruit flies with a genetic block in CoA biosynthesis, we demonstrate that progeny survive embryonal and early larval development by obtaining CoA precursors from maternal sources. Later in life, the microbiome can provide the essential CoA building blocks to the host, enabling continuation of normal development. A flow of stable, long-lasting CoA precursors between living organisms is revealed. This indicates the presence of complex strategies to maintain CoA homeostasis.


Asunto(s)
Coenzima A , Microbiota , Animales , Coenzima A/genética , Coenzima A/metabolismo , Drosophila/metabolismo , Femenino , Humanos , Madres , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Cigoto/metabolismo
18.
Annu Rev Neurosci ; 44: 359-381, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-33823654

RESUMEN

Oxytocin regulates parturition, lactation, parental nurturing, and many other social behaviors in both sexes. The circuit mechanisms by which oxytocin modulates social behavior are receiving increasing attention. Here, we review recent studies on oxytocin modulation of neural circuit function and social behavior, largely enabled by new methods of monitoring and manipulating oxytocin or oxytocin receptor neurons in vivo. These studies indicate that oxytocin can enhance the salience of social stimuli and increase signal-to-noise ratios by modulating spiking and synaptic plasticity in the context of circuits and networks. We highlight oxytocin effects on social behavior in nontraditional organisms such as prairie voles and discuss opportunities to enhance the utility of these organisms for studying circuit-level modulation of social behaviors. We then discuss recent insights into oxytocin neuron activity during social interactions. We conclude by discussing some of the major questions and opportunities in the field ahead.


Asunto(s)
Oxitocina , Conducta Social , Animales , Arvicolinae , Femenino , Masculino , Plasticidad Neuronal , Receptores de Oxitocina
19.
Annu Rev Cell Dev Biol ; 30: 581-613, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25150012

RESUMEN

Embryogenesis depends on a highly coordinated cascade of genetically encoded events. In animals, maternal factors contributed by the egg cytoplasm initially control development, whereas the zygotic nuclear genome is quiescent. Subsequently, the genome is activated, embryonic gene products are mobilized, and maternal factors are cleared. This transfer of developmental control is called the maternal-to-zygotic transition (MZT). In this review, we discuss recent advances toward understanding the scope, timing, and mechanisms that underlie zygotic genome activation at the MZT in animals. We describe high-throughput techniques to measure the embryonic transcriptome and explore how regulation of the cell cycle, chromatin, and transcription factors together elicits specific patterns of embryonic gene expression. Finally, we illustrate the interplay between zygotic transcription and maternal clearance and show how these two activities combine to reprogram two terminally differentiated gametes into a totipotent embryo.


Asunto(s)
Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , ARN Mensajero Almacenado/genética , Transcripción Genética , Cigoto/metabolismo , Animales , Ciclo Celular , Cromatina/genética , Cromatina/ultraestructura , Proteínas de Drosophila/fisiología , Proteínas del Huevo/genética , Embrión no Mamífero , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Histonas/fisiología , Humanos , Modelos Genéticos , Oocitos/metabolismo , Embarazo , Estabilidad del ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Transcriptoma , Proteínas de Xenopus/fisiología , Proteínas de Pez Cebra/fisiología
20.
Immunol Rev ; 323(1): 288-302, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38445769

RESUMEN

Maternal environmental exposures, particularly during gestation and lactation, significantly influence the immunological development and long-term immunity of offspring. Mammalian immune systems develop through crucial inputs from the environment, beginning in utero and continuing after birth. These critical developmental windows are essential for proper immune system development and, once closed, may not be reopened. This review focuses on the mechanisms by which maternal exposures, particularly to pathogens, diet, and microbiota, impact offspring immunity. Mechanisms driving maternal-offspring immune crosstalk include transfer of maternal antibodies, changes in the maternal microbiome and microbiota-derived metabolites, and transfer of immune cells and cytokines via the placenta and breastfeeding. We further discuss the role of transient maternal infections, which are common during pregnancy, in providing tissue-specific immune education to offspring. We propose a "maternal-driven immune education" hypothesis, which suggests that offspring can use maternal encounters that occur during a critical developmental window to develop optimal immune fitness against infection and inflammation.


Asunto(s)
Exposición Materna , Humanos , Femenino , Embarazo , Animales , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/inmunología , Inmunidad Materno-Adquirida , Microbiota/inmunología , Sistema Inmunológico/inmunología , Sistema Inmunológico/crecimiento & desarrollo , Intercambio Materno-Fetal/inmunología , Placenta/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA