Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 172
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 49(3): 397-412, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231982

RESUMEN

Successful pregnancy requires carefully-coordinated communications between the mother and fetus. Immune cells and cytokine signaling pathways participate as mediators of these communications to promote healthy pregnancy. At the same time, certain infections or inflammatory conditions in pregnant mothers cause severe disease and have detrimental impacts on the developing fetus. In this review, we examine evidence for the role of maternal and fetal immune responses affecting pregnancy and fetal development, both under homeostasis and following infection. We discuss immune responses that are necessary to promote healthy pregnancy and those that lead to congenital disorders and pregnancy complications, with a particular emphasis on the role of interferons and cytokines. Understanding the contributions of the immune system in pregnancy and fetal development provides important insights into the pathogenesis underlying maternal and fetal diseases and sheds insights on possible targets for therapy.


Asunto(s)
Citocinas/metabolismo , Desarrollo Fetal/inmunología , Mediadores de Inflamación/metabolismo , Interferones/metabolismo , Complicaciones del Embarazo/inmunología , Embarazo/inmunología , Animales , Femenino , Humanos , Exposición Materna/efectos adversos
2.
Immunity ; 47(6): 1100-1113.e6, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262349

RESUMEN

Natural killer (NK) cells are present in large populations at the maternal-fetal interface during early pregnancy. However, the role of NK cells in fetal growth is unclear. Here, we have identified a CD49a+Eomes+ subset of NK cells that secreted growth-promoting factors (GPFs), including pleiotrophin and osteoglycin, in both humans and mice. The crosstalk between HLA-G and ILT2 served as a stimulus for GPF-secreting function of this NK cell subset. Decreases in this GPF-secreting NK cell subset impaired fetal development, resulting in fetal growth restriction. The transcription factor Nfil3, but not T-bet, affected the function and the number of this decidual NK cell subset. Adoptive transfer of induced CD49a+Eomes+ NK cells reversed impaired fetal growth and rebuilt an appropriate local microenvironment. These findings reveal properties of NK cells in promoting fetal growth. In addition, this research proposes approaches for therapeutic administration of NK cells in order to reverse restricted nourishments within the uterine microenvironment during early pregnancy.


Asunto(s)
Aborto Habitual/inmunología , Traslado Adoptivo , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Desarrollo Fetal/inmunología , Retardo del Crecimiento Fetal/prevención & control , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Asesinas Naturales/trasplante , Aborto Habitual/genética , Aborto Habitual/patología , Adulto , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Microambiente Celular , Citocinas/genética , Citocinas/inmunología , Decidua/inmunología , Decidua/patología , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/inmunología , Retardo del Crecimiento Fetal/patología , Feto , Regulación del Desarrollo de la Expresión Génica , Antígenos HLA-G/genética , Antígenos HLA-G/inmunología , Humanos , Integrina alfa1/genética , Integrina alfa1/inmunología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Receptor Leucocitario Tipo Inmunoglobulina B1/genética , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Ratones , Ratones Endogámicos C57BL , Embarazo , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología
3.
Immunity ; 43(2): 382-93, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26287683

RESUMEN

Macrophages are one of the most diverse cell populations in terms of their anatomical location and functional specialization during both homeostasis and disease. Although it has been shown in different fate mapping models that some macrophages present in adult tissues are already established during fetal development, their exact origins are still under debate. In the current study, we developed a fate mapping strain, based on the Kit locus, which allowed us to readdress "the origins" question. Different types of macrophages from various adult tissues were traced to their fetal or adult sources by inducing labeling in precursors at several time points either during fetal development or in adult mice. We show that all adult macrophages, resident or infiltrating, are progenies of classical hematopoietic stem cells (HSC) with the exception of microglia and, partially epidermal Langerhans cells, which are yolk sac (YS)-derived.


Asunto(s)
Desarrollo Fetal/inmunología , Células Madre Hematopoyéticas/fisiología , Macrófagos/fisiología , Microglía/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Antígenos CD/metabolismo , Diferenciación Celular , Linaje de la Célula , Embrión de Mamíferos , Femenino , Homeostasis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Proteínas Proto-Oncogénicas c-kit/genética , Saco Vitelino/fisiología
4.
J Immunol ; 208(2): 221-226, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35017211

RESUMEN

Maternal infection during pregnancy is known to alter the development and function of offspring's immune system, leading to inappropriate immune responses to common childhood infections and immunizations. Although this is an expanding field, maternal parasitic infections remain understudied. Millions of women of reproductive age are currently at risk for parasitic infection, whereas many pregnant, chronically infected women are excluded from mass drug administration due partially to a lack of resources, as well as fear of unknown adverse fetal developmental outcomes. In areas endemic for multiple parasitic infections, such as sub-Saharan Africa, there are increased rates of morbidity and mortality for various infections during early childhood in comparison with nonendemic areas. Despite evidence supporting similar immunomodulatory effects between various parasite species, there is no clear mechanistic understanding of how maternal infection reprograms offspring immunity. This brief review will compare the effects of selected maternal parasitic infections on offspring immunity.


Asunto(s)
Desarrollo Fetal/inmunología , Helmintiasis/inmunología , Malaria Falciparum/inmunología , Enfermedades Parasitarias/transmisión , Complicaciones Parasitarias del Embarazo/epidemiología , Adulto , África del Sur del Sahara/epidemiología , Animales , Femenino , Helmintiasis/parasitología , Helmintiasis/transmisión , Helmintos/patogenicidad , Humanos , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Enfermedades Parasitarias/epidemiología , Enfermedades Parasitarias/inmunología , Embarazo , Suelo/parasitología
5.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892139

RESUMEN

Maternal obesity and over/undernutrition can have a long-lasting impact on offspring health during critical periods in the first 1000 days of life. Children born to mothers with obesity have reduced immune responses to stimuli which increase susceptibility to infections. Recently, maternal western-style diets (WSDs), high in fat and simple sugars, have been associated with skewing neonatal immune cell development, and recent evidence suggests that dysregulation of innate immunity in early life has long-term consequences on metabolic diseases and behavioral disorders in later life. Several factors contribute to abnormal innate immune tolerance or trained immunity, including changes in gut microbiota, metabolites, and epigenetic modifications. Critical knowledge gaps remain regarding the mechanisms whereby these factors impact fetal and postnatal immune cell development, especially in precursor stem cells in bone marrow and fetal liver. Components of the maternal microbiota that are transferred from mothers consuming a WSD to their offspring are understudied and identifying cause and effect on neonatal innate and adaptive immune development needs to be refined. Tools including single-cell RNA-sequencing, epigenetic analysis, and spatial location of specific immune cells in liver and bone marrow are critical for understanding immune system programming. Considering the vital role immune function plays in offspring health, it will be important to understand how maternal diets can control developmental programming of innate and adaptive immunity.


Asunto(s)
Dieta Occidental , Desarrollo Fetal , Efectos Tardíos de la Exposición Prenatal , Humanos , Femenino , Embarazo , Dieta Occidental/efectos adversos , Animales , Desarrollo Fetal/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Epigénesis Genética , Microbioma Gastrointestinal/inmunología , Inmunidad Innata , Fenómenos Fisiologicos Nutricionales Maternos , Feto/inmunología
6.
Nat Immunol ; 12(2): 129-36, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21186366

RESUMEN

The nuclear adaptor Ldb1 functions as a core component of multiprotein transcription complexes that regulate differentiation in diverse cell types. In the hematopoietic lineage, Ldb1 forms a complex with the non-DNA-binding adaptor Lmo2 and the transcription factors E2A, Scl and GATA-1 (or GATA-2). Here we demonstrate a critical and continuous requirement for Ldb1 in the maintenance of both fetal and adult mouse hematopoietic stem cells (HSCs). Deletion of Ldb1 in hematopoietic progenitors resulted in the downregulation of many transcripts required for HSC maintenance. Genome-wide profiling by chromatin immunoprecipitation followed by sequencing (ChIP-Seq) identified Ldb1 complex-binding sites at highly conserved regions in the promoters of genes involved in HSC maintenance. Our results identify a central role for Ldb1 in regulating the transcriptional program responsible for the maintenance of HSCs.


Asunto(s)
Células Madre Adultas/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Desarrollo Fetal , Células Madre Hematopoyéticas/metabolismo , Traslado Adoptivo , Células Madre Adultas/citología , Células Madre Adultas/inmunología , Células Madre Adultas/trasplante , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/trasplante , Femenino , Desarrollo Fetal/genética , Desarrollo Fetal/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas con Dominio LIM , Ratones , Ratones Noqueados , Ratones Transgénicos , Embarazo , Unión Proteica , Elementos Reguladores de la Transcripción/genética , Elementos Reguladores de la Transcripción/inmunología , Activación Transcripcional/genética , Activación Transcripcional/inmunología
7.
Immunology ; 164(4): 665-676, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34287859

RESUMEN

The immunology of pregnancy has been the focus of many studies to better understand how the mother is able to tolerate the presence of a semi-allogeneic fetus. Far from the initial view of pregnancy as a state of immunosuppression, successful fetal development from implantation to birth is now known to be under the control of an intricate balance of immune cells. The balance between pro-inflammatory functions used to promote embryo implantation and placental development and immunosuppressive activity to maintain maternal tolerance of the fetus is an immunological phenotype unique to pregnancy, which is dependent on the time of gestation. Neutrophils are one of a host of innate immune cells detected at the maternal-fetal interface, but very little is known of their function. In this review, we explore the emerging functions of neutrophils during pregnancy and their interactions with and regulation of T cells, a key adaptive immune cell population essential for the establishment of fetal-maternal tolerance.


Asunto(s)
Inmunidad Adaptativa , Desarrollo Fetal/inmunología , Tolerancia Inmunológica , Inmunidad Innata , Intercambio Materno-Fetal/inmunología , Neutrófilos/fisiología , Animales , Comunicación Celular/inmunología , Femenino , Humanos , Inmunomodulación , Fenotipo , Placenta/inmunología , Placenta/metabolismo , Embarazo , Linfocitos T/inmunología , Linfocitos T/metabolismo
8.
Br J Cancer ; 124(12): 1897-1899, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33767421
9.
Int J Mol Sci ; 22(4)2021 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572203

RESUMEN

Overweight and obesity during pregnancy have been associated with increased birth weight, childhood obesity, and noncommunicable diseases in the offspring, leading to a vicious transgenerational perpetuating of metabolic derangements. Key components in intrauterine developmental programming still remain to be identified. Obesity involves chronic low-grade systemic inflammation that, in addition to physiological adaptations to pregnancy, may potentially expand to the placental interface and lead to intrauterine derangements with a threshold effect. Animal models, where maternal inflammation is mimicked by single injections with lipopolysaccharide (LPS) resembling the obesity-induced immune profile, showed increased adiposity and impaired metabolic homeostasis in the offspring, similar to the phenotype observed after exposure to maternal obesity. Cytokine levels might be specifically important for the metabolic imprinting, as cytokines are transferable from maternal to fetal circulation and have the capability to modulate placental nutrient transfer. Maternal inflammation may induce metabolic reprogramming at several levels, starting from the periconceptional period with effects on the oocyte going through early stages of embryonic and placental development. Given the potential to reduce inflammation through inexpensive, widely available therapies, examinations of the impact of chronic inflammation on reproductive and pregnancy outcomes, as well as preventive interventions, are now needed.


Asunto(s)
Desarrollo Infantil/fisiología , Desarrollo Fetal/inmunología , Obesidad Materna/inmunología , Obesidad Infantil/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Animales , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/inmunología , Intercambio Materno-Fetal/inmunología , Redes y Vías Metabólicas/inmunología , Obesidad Materna/complicaciones , Obesidad Materna/metabolismo , Obesidad Materna/terapia , Obesidad Infantil/metabolismo , Obesidad Infantil/prevención & control , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/prevención & control
10.
Front Neuroendocrinol ; 55: 100797, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31574280

RESUMEN

The maternal microbiota is positioned to regulate the development of offspring immunity, metabolism, as well as brain function and behavior. The mechanisms by which maternal microbial signals drive these processes are beginning to be elucidated. In this review, we provide a brief overview on the importance of the microbiome in brain function and behavior, define the maternal vaginal and gut microbiota as distinct influences on offspring development, and outline current concepts in microbial origins of offspring health outcomes. We propose that the maternal microbiota influences prenatal and early postnatal offspring development and health outcomes through two overlapping processes. First, during pregnancy maternal gut microbiota provide metabolites and substrates essential for fetal growth through metabolic provisioning, driving expansion and maturation of central and peripheral immune cells, and formation of neural circuits. Second, vertical transmission of maternal microbiota during birth and in the early postnatal window elicits a potent immunostimulatory effect in offspring that induces metabolic and developmental transcriptional programs, primes the immune system for subsequent microbial exposure, and provides substrates for brain metabolism. Finally, we explore the possibility that environmental factors, such as malnutrition, stress and infection, may exert programmatic effects by disrupting the functional contributions of the maternal microbiome during prenatal and postnatal development to influence offspring outcomes across the lifespan.


Asunto(s)
Encéfalo , Desarrollo Infantil/fisiología , Desarrollo Fetal/fisiología , Microbioma Gastrointestinal , Efectos Tardíos de la Exposición Prenatal , Vagina/microbiología , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/inmunología , Encéfalo/metabolismo , Femenino , Desarrollo Fetal/inmunología , Microbioma Gastrointestinal/inmunología , Humanos , Recién Nacido , Embarazo , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/microbiología
11.
Front Neuroendocrinol ; 55: 100794, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31560883

RESUMEN

The role of oxytocin (OT) as a neuropeptide that modulates social behavior has been extensively studied and reviewed, but beyond these functions, OT's adaptive functions at birth are quite numerous, as OT coordinates many physiological processes in the mother and fetus to ensure a successful delivery. In this review we explore in detail the potential adaptive roles of oxytocin as an anti-inflammatory, protective molecule at birth for the developing fetal brain and gastrointestinal system based on evidence that birth is a potent inflammatory/immune event. We discuss data with relevance for a number of neurodevelopmental disorders, as well as the emerging role of the gut-brain axis for health and disease. Finally, we discuss the potential relevance of sex differences in OT signaling present at birth in the increased male vulnerability to neurodevelopmental disabilities.


Asunto(s)
Encéfalo , Desarrollo Fetal , Microbioma Gastrointestinal , Inflamación , Trastornos del Neurodesarrollo , Oxitocina/metabolismo , Transducción de Señal , Encéfalo/crecimiento & desarrollo , Encéfalo/inmunología , Encéfalo/metabolismo , Desarrollo Fetal/inmunología , Humanos , Recién Nacido , Inflamación/inmunología , Inflamación/metabolismo , Trastornos del Neurodesarrollo/inmunología , Trastornos del Neurodesarrollo/metabolismo
12.
Brain Behav Immun ; 83: 44-55, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31493445

RESUMEN

Adverse experiences during pregnancy induce placental programming, affecting the fetus and its developmental trajectory. However, the influence of 'positive' maternal experiences on the placenta and fetus remain unclear. In animal models of early life stress, environmental enrichment (EE) has ameliorated and even prevented associated impairments in brain and behavior. Here, using a maternal immune activation (MIA) model in rats, we test whether EE attenuates maternal, placental and/or fetal responses to an inflammatory challenge, thereby offering a mechanism by which fetal programming may be prevented. Moreover, we evaluate life-long EE exposure on offspring development and examine a constellation of genes and epigenetic writers that may protect against MIA challenges. In our model, maternal plasma corticosterone and interleukin-1ß were elevated 3 h after MIA, validating the maternal inflammatory response. Evidence for developmental programming was demonstrated by a simultaneous decrease in the placental enzymes Hsd11b2 and Hsd11b2/Hsd11b1, suggesting disturbances in glucocorticoid metabolism. Reductions of Hsd11b2 in response to challenge is thought to result in excess glucocorticoid exposure to the fetus and altered glucocorticoid receptor expression, increasing susceptibility to behavioral impairments later in life. The placental, but not maternal, glucocorticoid implications of MIA were attenuated by EE. There were also sustained changes in epigenetic writers in both placenta and fetal brain as a consequence of environmental experience and sex. Following MIA, both male and female juvenile animals were impaired in social discrimination ability. Life-long EE mitigated these impairments, in addition to the sex specific MIA associated disruptions in central Fkbp5 and Oprm1. These data provide the first evidence that EE protects placental functioning during stressor exposure, underscoring the importance of addressing maternal health and well-being throughout pregnancy. Future work must evaluate critical periods of EE use to determine if postnatal EE experience is necessary, or if prenatal exposure alone is sufficient to confer protection.


Asunto(s)
Desarrollo Fetal/inmunología , Placenta/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , 11-beta-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Femenino , Feto/inmunología , Glucocorticoides/metabolismo , Masculino , Placenta/enzimología , Placenta/metabolismo , Embarazo , Ratas
13.
J Pathol ; 247(5): 686-696, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30506724

RESUMEN

Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Inmunidad Adaptativa/fisiología , Pulmón/crecimiento & desarrollo , Angiotensina II/inmunología , Angiotensina II/fisiología , Animales , Bronquios/citología , Desarrollo Fetal/inmunología , Desarrollo Fetal/fisiología , Humanos , Pulmón/embriología , Pulmón/inmunología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/fisiología , Ratones , Neovascularización Fisiológica/inmunología , Neovascularización Fisiológica/fisiología , Neumonía/inmunología , Neumonía/fisiopatología , Regeneración/inmunología , Regeneración/fisiología , Transducción de Señal/inmunología , Transducción de Señal/fisiología
14.
J Immunol ; 201(2): 325-334, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29987001

RESUMEN

Immune cells adapt their phenotypic and functional characteristics in response to the tissue microenvironment within which they traffic and reside. The fetomaternal interface, consisting of placental trophoblasts and the maternal decidua, is a highly specialized tissue with a unique and time-limited function: to nourish and support development of the semiallogeneic fetus and protect it from inflammatory or immune-mediated injury. It is therefore important to understand how immune cells within these tissues are educated and adapt to fulfill their biological functions. This review article focuses on the local regulatory mechanisms ensuring that both innate and adaptive immune cells appropriately support the early events of implantation and placental development through direct involvement in promoting immune tolerance of fetal alloantigens, suppressing inflammation, and remodeling of maternal uterine vessels to facilitate optimal placental function and fetal growth.


Asunto(s)
Microambiente Celular/inmunología , Desarrollo Fetal/inmunología , Feto/inmunología , Placentación/inmunología , Animales , Femenino , Humanos , Embarazo
15.
Cell Mol Life Sci ; 76(4): 729-743, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30374520

RESUMEN

Cannabinoids are the most commonly abused illicit drugs worldwide. While cannabis can be beneficial for certain heath conditions, abuse of potent synthetic cannabinoids has been on the rise. Exposure to cannabinoids is also prevalent in women of child-bearing age and pregnant women. These compounds can cross the placental barrier and directly affect the fetus. They mediate their effects primarily through G-protein coupled cannabinoid receptors, CB1 and CB2. In addition to significant neurological effects, cannabinoids can trigger robust immunomodulation by altering cytokine levels, causing apoptosis of lymphoid cells and inducing suppressor cells of the immune system. Profound effects of cannabinoids on the immune system as discussed in this review, suggest that maternal exposure during pregnancy could lead to dysregulation of innate and adaptive immune system of developing fetus and offspring potentially leading to weakening of immune defenses against infections and cancer later in life. Emerging evidence also indicates the underlying role of epigenetic mechanisms causing long-lasting impact following cannabinoid exposure in utero.


Asunto(s)
Cannabinoides/envenenamiento , Desarrollo Fetal/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/inmunología , Cannabinoides/química , Femenino , Desarrollo Fetal/inmunología , Humanos , Sistema Inmunológico/embriología , Sistema Inmunológico/inmunología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Estructura Molecular , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
16.
J Allergy Clin Immunol ; 144(6): 1703-1713, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31615640

RESUMEN

BACKGROUND: Individual susceptibility to allergic diseases is developmentally programmed by early-life exposures. Evidence from preclinical studies suggests that intrauterine growth restriction is protective against later inflammatory responses to allergens. OBJECTIVE: We sought to evaluate whether prenatal growth affects susceptibility to allergy in human subjects. METHODS: We systematically searched for relevant studies in 11 databases, including Web of Science, ProQuest, EMBASE, and PubMed. We included only studies that corrected for gestational age or were restricted to full-term infants to separate effects of fetal growth from those of prematurity. RESULTS: The 42 eligible studies included prospective and retrospective cohort, cross-sectional, and case-control studies. Only 2 studies reported allergic asthma. A birth weight increase of 1 kg was associated with a 44% greater risk of food allergy in children (odds ratio [OR], 1.44; 95% CI, 1.04-1.99; P = .001), a 17% greater risk of ever allergic dermatitis in children (OR, 1.17; 95% CI, 1.04-1.32; P = .008), and a 34% greater risk of ever or current allergic dermatitis in infants up to 2 years of age (OR, 1.34; 95% CI, 1.08-1.68; P = .009). Risks of allergic rhinitis were not associated with birth weight. CONCLUSIONS: The results of these meta-analyses suggest that intrauterine growth restriction protects against allergic diseases in human subjects consistent with preclinical evidence but that effects might differ between allergic diseases. The strongest evidence is available for infancy and early childhood, and additional studies in older children and adults are needed to determine whether the effects of prenatal growth on each allergic disease persist or differ between those with severe and mild phenotypes.


Asunto(s)
Peso al Nacer/inmunología , Desarrollo Fetal/inmunología , Hipersensibilidad/inmunología , Preescolar , Ensayos Clínicos como Asunto , Femenino , Humanos , Hipersensibilidad/epidemiología , Hipersensibilidad/etiología , Lactante , Recién Nacido , Masculino , Factores de Riesgo
17.
J Physiol ; 597(16): 4251-4262, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31192454

RESUMEN

KEY POINTS: Experimental maternal allergic asthma in sheep provides an experimental model in which to test impacts on progeny. Fetuses from allergic asthmatic ewes had fewer surfactant-producing cells in lungs. A greater proportion of lymphocytes from thymus were CD44 positive in fetuses from allergic asthmatic ewes than in controls. These changes to fetal development might contribute to poor neonatal lung function and increased risk of allergy seen in offspring of pregnancies complicated by asthma. ABSTRACT: Asthma is prevalent in pregnancy and increases the risk of disease in offspring, including neonatal respiratory distress and childhood asthma and allergy, but the mechanisms are not understood. We hypothesized that fetal lung structure and immune phenotype in late gestation fetal sheep would be impaired in our sheep model of maternal allergic asthma during pregnancy. Singleton-bearing ewes were either sensitized before pregnancy to house dust mite (HDM, allergic, n = 7) or were non-allergic (control, n = 5). The ewes were subsequently subjected to repeated airway challenges with HDM (allergic group) or saline (control group) throughout gestation. Tissues were collected at 140 ± 1 days gestational age (term, ∼147 days). The density of type II alveolar epithelial cells (surfactant protein C-immunostained) in the lungs was 30% lower in fetuses from allergic ewes than in controls (P < 0.001), but tissue-to-air space ratio and numbers of leucocytes and macrophages were not different between groups. The proportion of CD44+ lymphocytes in the fetal thymus was 3.5-fold higher in fetuses from allergic ewes than in control ewes (P = 0.043). Fewer surfactant-producing type II alveolar epithelial cells may contribute to the increased risk of neonatal respiratory distress in infants of asthmatic mothers, suggesting that interventions to promote lung maturation could improve their neonatal outcomes. If the elevated lymphocyte expression of CD44 persists postnatally, this would confer greater susceptibility to allergic diseases in progeny of asthmatic mothers, consistent with observations in humans. Further experiments are needed to evaluate postnatal phenotypes of progeny and investigate potential interventions.


Asunto(s)
Asma , Desarrollo Fetal/inmunología , Hipersensibilidad , Pulmón/embriología , Pulmón/inmunología , Ovinos/inmunología , Líquido Amniótico/química , Animales , Anticuerpos/sangre , Pruebas de Provocación Bronquial/métodos , Citocinas/química , Citocinas/metabolismo , Femenino , Hidrocortisona/sangre , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA