Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
Front Immunol ; 13: 821542, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35185911

RESUMEN

CD49a+ natural killer (NK) cells play a critical role in promoting fetal development and maintaining immune tolerance at the maternal-fetal interface during the early stages of pregnancy. However, given their residency in human tissue, thorough studies and clinical applications are difficult to perform. It is still unclear as to how functional human CD49a+ NK cells can be induced to benefit pregnancy outcomes. In this study, we established three no-feeder cell induction systems to induce human CD49a+ NK cells from umbilical cord blood hematopoietic stem cells (HSCs), bone marrow HSCs, and peripheral blood NK cells in vitro. These induced NK cells (iNKs) from three cell induction systems display high levels of CD49a, CD9, CD39, CD151 expression, low levels of CD16 expression, and no obvious cytotoxic capability. They are phenotypically and functionally similar to decidual NK cells. Furthermore, these iNKs display a high expression of growth-promoting factors and proangiogenic factors and can promote fetal growth and improve uterine artery blood flow in a murine pregnancy model in vivo. This research demonstrates the ability of human-induced CD49a+ NK cells to promote fetal growth via three cell induction systems, which could eventually be used to treat patients experiencing adverse pregnancy outcomes.


Asunto(s)
Desarrollo Fetal/inmunología , Integrina alfa1/inmunología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Asesinas Naturales/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Integrina alfa1/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Ratones , Embarazo
2.
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
3.
Front Immunol ; 12: 771054, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745147

RESUMEN

The placenta is a fetal-derived organ whose function is crucial for both maternal and fetal health. The human placenta contains a population of fetal macrophages termed Hofbauer cells. These macrophages play diverse roles, aiding in placental development, function and defence. The outer layer of the human placenta is formed by syncytiotrophoblast cells, that fuse to form the syncytium. Adhered to the syncytium at sites of damage, on the maternal side of the placenta, is a population of macrophages termed placenta associated maternal macrophages (PAMM1a). Here we discuss recent developments that have led to renewed insight into our understanding of the ontogeny, phenotype and function of placental macrophages. Finally, we discuss how the application of new technologies within placental research are helping us to further understand these cells.


Asunto(s)
Desarrollo Fetal/inmunología , Feto/inmunología , Inmunidad Innata/inmunología , Macrófagos/inmunología , Placenta/inmunología , Animales , Movimiento Celular/inmunología , Movimiento Celular/fisiología , Vellosidades Coriónicas/inmunología , Vellosidades Coriónicas/metabolismo , Femenino , Feto/citología , Feto/fisiología , Receptor 2 de Folato/inmunología , Receptor 2 de Folato/metabolismo , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/fisiología , Fagocitosis/inmunología , Fagocitosis/fisiología , Placenta/citología , Placenta/fisiología , Embarazo
4.
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
5.
Sci Immunol ; 6(58)2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893173

RESUMEN

Accumulating evidence suggests that the mouse embryonic thymus produces distinct waves of innate effector γδ T cells. However, it is unclear whether this process occurs similarly in humans and whether it comprises a dedicated subset of innate-like type 3 effector γδ T cells. Here, we present a protocol for high-throughput sequencing of TRG and TRD pairs that comprise the clonal γδTCR. In combination with single-cell RNA sequencing, multiparameter flow cytometry, and TCR sequencing, we reveal a high heterogeneity of γδ T cells sorted from neonatal and adult blood that correlated with TCR usage. Immature γδ T cell clusters displayed mixed and diverse TCRs, but effector cell types segregated according to the expression of either highly expanded individual Vδ1+ TCRs or moderately expanded semi-invariant Vγ9Vδ2+ TCRs. The Vγ9Vδ2+ T cells shared expression of genes that mark innate-like T cells, including ZBTB16 (encoding PLZF), KLRB1, and KLRC1, but consisted of distinct clusters with unrelated Vγ9Vδ2+ TCR clones characterized either by TBX21, FCGR3A, and cytotoxicity-associated gene expression (type 1) or by CCR6, RORC, IL23R, and DPP4 expression (type 3). Effector γδ T cells with type 1 and type 3 innate T cell signatures were detected in a public dataset of early embryonic thymus organogenesis. Together, this study suggests that functionally distinct waves of human innate-like effector γδ T cells with semi-invariant Vγ9Vδ2+ TCR develop in the early fetal thymus and persist into adulthood.


Asunto(s)
Sangre Fetal/citología , Desarrollo Fetal/inmunología , Linfocitos Intraepiteliales/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/inmunología , Adulto , Diferenciación Celular/inmunología , Células Cultivadas , Femenino , Sangre Fetal/inmunología , Humanos , Linfocitos Intraepiteliales/metabolismo , Activación de Linfocitos , Masculino , RNA-Seq , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Análisis de la Célula Individual , Subgrupos de Linfocitos T/metabolismo
6.
Br J Cancer ; 124(12): 1897-1899, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33767421
7.
Front Immunol ; 12: 637975, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679795

RESUMEN

Human B-lymphopoiesis is a dynamic life-long process that starts in utero by around six post-conception weeks. A detailed understanding of human fetal B-lymphopoiesis and how it changes in postnatal life is vital for building a complete picture of normal B-lymphoid development through ontogeny, and its relevance in disease. B-cell acute lymphoblastic leukemia (B-ALL) is one of the most common cancers in children, with many of the leukemia-initiating events originating in utero. It is likely that the biology of B-ALL, including leukemia initiation, maintenance and progression depends on the developmental stage and type of B-lymphoid cell in which it originates. This is particularly important for early life leukemias, where specific characteristics of fetal B-cells might be key to determining how the disease behaves, including response to treatment. These cellular, molecular and/or epigenetic features are likely to change with age in a cell intrinsic and/or microenvironment directed manner. Most of our understanding of fetal B-lymphopoiesis has been based on murine data, but many recent studies have focussed on characterizing human fetal B-cell development, including functional and molecular assays at a single cell level. In this mini-review we will give a short overview of the recent advances in the understanding of human fetal B-lymphopoiesis, including its relevance to infant/childhood leukemia, and highlight future questions in the field.


Asunto(s)
Linfocitos B/inmunología , Desarrollo Fetal/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Células Precursoras de Linfocitos B/inmunología , Carcinogénesis , Diferenciación Celular , Humanos , Activación de Linfocitos
8.
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
9.
Nat Rev Endocrinol ; 17(4): 235-245, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33526907

RESUMEN

Intrauterine growth restriction (IUGR) is a common complication of pregnancy and increases the risk of the offspring developing type 2 diabetes mellitus (T2DM) later in life. Alterations in the immune system are implicated in the pathogenesis of IUGR-induced T2DM. The development of the fetal immune system is a delicate balance as it must remain tolerant of maternal antigens whilst also preparing for the post-birth environment. In addition, the fetal immune system is susceptible to an altered intrauterine milieu caused by maternal and placental inflammatory mediators or secondary to nutrient and oxygen deprivation. Pancreatic-resident macrophages populate the pancreas during fetal development, and their phenotype is dynamic through the neonatal period. Furthermore, macrophages in the islets are instrumental in islet development as they influence ß-cell proliferation and islet neogenesis. In addition, cytokines, derived from ß-cells and macrophages, are important to islet homeostasis in the fetus and adult and, when perturbed, can cause islet dysfunction. Several activated immune pathways have been identified in the islets of people who experienced IUGR, with alternations in the levels of IL-1ß and IL-4 as well as changes in TGFß signalling. Leptin levels are also altered. Immunomodulation has shown therapeutic benefit in T2DM and might be particularly useful in IUGR-induced T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/inmunología , Desarrollo Fetal/inmunología , Retardo del Crecimiento Fetal/inmunología , Animales , Humanos , Sistema Inmunológico/inmunología , Lesiones Prenatales/inmunología
10.
Toxicol Lett ; 339: 1-11, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33301788

RESUMEN

Despite numerous reports that ambient particulate matter is a key determinant for human health, toxicity data produced based on physicochemical properties of particulate matters is very lack, suggesting lack of scientific evidence for regulation. In this study, we sampled inhalable particulate matters (PM10) in northern Seoul, Korea. PM10 showed atypical- and fiber-type particles with the average size and the surface charge of 1,598.1 ± 128.7 nm and -27.5 ± 2.8, respectively, and various toxic elements were detected in the water extract. On day 90 after the first pulmonary exposure, total cell number dose-dependently increased in the lungs of both sexes of mice. PM10 induced Th1-dominant immune response with pathological changes in both sexes of mice. Meanwhile, composition of total cells and expression of proteins which functions in cell-to-cell communication showed different trends between sexes. Following, male and female mice were mated to identify effects of PM10 to the next generation. PM10 remained in the lung of dams until day 21 after birth, and the levels of IgA and IgE increased in the blood of dams exposed to the maximum dose compared to control. In addition, the interval between births of fetuses, the number of offspring, the neonatal survival rate (day 4 after birth) and the sex ratio seemed to be affected at the maximum dose, and particularly, all offspring from one dam were stillborn. In addition, expression of HIF-1α protein increased in the lung tissue of dams exposed to PM10, and level of hypoxia-related proteins was notably enhanced in PM10-exposed bronchial epithelial cells compared to control. Taken together, we suggest that inhaled PM10 may induce Th1-shifting immune response in the lung, and that it may affect reproduction (fetus development) by causing lung hypoxia. Additionally, we propose that further study is needed to identify particle-size-dependent effects on development of the next generation.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Desarrollo Fetal/efectos de los fármacos , Desarrollo Fetal/inmunología , Inmunidad/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Material Particulado/toxicidad , Contaminantes Atmosféricos/inmunología , Animales , Femenino , Humanos , Masculino , Ratones , Modelos Animales , Material Particulado/inmunología , República de Corea
11.
Medicine (Baltimore) ; 99(46): e22722, 2020 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-33181648

RESUMEN

In general terms, fetal growth restriction (FGR) is considered the impossibility of achieving the genetically determined potential size. In the vast majority of cases, it is related to uteroplacental insufficiency. Although its origin remains unknown and causes are only known in 30% of cases, it is believed to be related to an interaction of environmental and genetic factors with either a fetal or maternal origin. One hypothesis is that alterations in the gastrointestinal microbiota composition, and thus alteration in the immune response, could play a role in FGR development. We performed an observational, prospective study in a subpopulation affected with FGR to elucidate the implications of this microbiota on the FGR condition.A total of 63 fetuses with FGR diagnosed in the third trimester as defined by the Delphi consensus, and 63 fetuses with fetal growth appropriate for gestational age will be recruited. Obstetric and nutritional information will be registered by means of specific questionnaires. We will collect maternal fecal samples between 30 to 36 weeks, intrapartum samples (maternal feces, maternal and cord blood) and postpartum samples (meconium and new-born feces at 6 weeks of life). Samples will be analyzed in the Department of Biochemistry and Molecular Biology II, Nutrition and Food Technology Institute of the University of Granada (UGR), for the determination of the gastrointestinal microbiota composition and its relationship with inflammatory biomarkers.This study will contribute to a better understanding of the influence of gastrointestinal microbiota and related inflammatory biomarkers in the development of FGR.Trial registration: NCT04047966. Registered August 7, 2019, during the recruitment stage. Retrospectively registered. Ongoing research.


Asunto(s)
Retardo del Crecimiento Fetal/inmunología , Feto/inmunología , Microbiota/inmunología , Mujeres Embarazadas , Adulto , Biomarcadores/análisis , Estudios de Casos y Controles , Cordocentesis/métodos , Técnica Delphi , Femenino , Desarrollo Fetal/inmunología , Desarrollo Fetal/fisiología , Feto/fisiopatología , Edad Gestacional , Humanos , Microbiota/fisiología , Embarazo , Estudios Prospectivos , España
12.
Sci Rep ; 10(1): 20288, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33219314

RESUMEN

Maternal stress during pregnancy is widespread and is associated with poor offspring outcomes, including long-term mental health issues. Prenatal stress-induced fetal neuroinflammation is thought to underlie aberrant neurodevelopment and to derive from a disruption in intrauterine immune homeostasis, though the exact origins are incompletely defined. We aimed to identify divergent immune and microbial metagenome profiles of stressed gestating mice that may trigger detrimental inflammatory signaling at the maternal-fetal interface. In response to stress, maternal glucocorticoid circuit activation corresponded with indicators of systemic immunosuppression. At the maternal-fetal interface, density of placental mononuclear leukocytes decreased with stress, yet maternal whole blood leukocyte analysis indicated monocytosis and classical M1 phenotypic shifts. Genome-resolved microbial metagenomic analyses revealed reductions in genes, microbial strains, and metabolic pathways in stressed dams that are primarily associated with pro-inflammatory function. In particular, disrupted Parasutterella excrementihominis appears to be integral to inflammatory and metabolic dysregulation during prenatal stress. Overall, these perturbations in maternal immunological and microbial regulation during pregnancy may displace immune equilibrium at the maternal-fetal interface. Notably, the absence of and reduction in overt maternal inflammation during stress indicates that the signaling patterns driving fetal outcomes in this context are more nuanced and complex than originally anticipated.


Asunto(s)
Encéfalo/embriología , Desarrollo Fetal/inmunología , Microbioma Gastrointestinal/inmunología , Complicaciones del Embarazo/inmunología , Estrés Psicológico/inmunología , Animales , Encéfalo/inmunología , Burkholderiales/genética , Burkholderiales/inmunología , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal/genética , Glucocorticoides/metabolismo , Humanos , Leucocitos Mononucleares/inmunología , Intercambio Materno-Fetal/inmunología , Salud Mental , Metagenómica , Ratones , Neuroinmunomodulación/inmunología , Placenta/citología , Placenta/inmunología , Embarazo , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/psicología , Efectos Tardíos de la Exposición Prenatal/inmunología , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología
13.
J Reprod Immunol ; 142: 103185, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32853845

RESUMEN

Preeclampsia (PE) and human immunodeficiency virus (HIV) have been linked with marked increases in maternal stress, resulting in a significant change in placental function ranging from alterations in placental structure to the precise and delicate transformations in placental gene expression. Such changes may lead to altered transport of essential signals to the fetus, which can have long-term impacts on offspring health and consequently affect fetal neurodevelopment. Therefore, this work investigated the role of placental 11ß-hydroxysteroid dehydrogenase types 2 (11ß-HSD2) in HIV associated preeclampsia. The placenta were obtained from 76 pregnant women, which were stratified based on pregnancy type and HIV status into; Normotensive HIV negative, normotensive HIV positive, PE HIV negative and PE HIV positive. The placental tissue was processed for immunocytochemistry and stained with rabbit polyclonal to 11ß-HSD2 Our results showed significant downregulation in the placental expression of 11ß-HSD2 in both the conducting and exchange villi of PE and HIV-positive patients when compared with Normotensive and HIV-negative individuals, respectively. Our results provide inferential evidence for comorbidity of PE and HIV in the downregulation of placental 11ß-HSD2 enzyme function, which mediates the programmed outcomes of an adverse maternal environment during pregnancy and long-term impacts on offspring health and consequently affects fetal neurodevelopment.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Desarrollo Fetal/inmunología , Infecciones por VIH/complicaciones , Trastornos del Neurodesarrollo/inmunología , Preeclampsia/inmunología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/análisis , Adolescente , Adulto , Encéfalo/embriología , Estudios de Casos y Controles , Regulación hacia Abajo , Femenino , Glucocorticoides/inmunología , Glucocorticoides/metabolismo , Infecciones por VIH/diagnóstico , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/aislamiento & purificación , Humanos , Edad Materna , Trastornos del Neurodesarrollo/patología , Placenta/enzimología , Placenta/inmunología , Placenta/patología , Preeclampsia/patología , Preeclampsia/virología , Embarazo , Estudios Retrospectivos , Sudáfrica , Adulto Joven
14.
Front Immunol ; 11: 1672, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733490

RESUMEN

Pregnancy comprises a unique immunological condition, to allow fetal development and to protect the host from pathogenic infections. Viral infections during pregnancy can disrupt immunological tolerance and may generate deleterious effects on the fetus. Despite these possible links between pregnancy and infection-induced morbidity, it is unclear how pregnancy interferes with maternal response to some viral pathogens. In this context, the novel coronavirus (SARS-CoV-2) can induce the coronavirus diseases-2019 (COVID-19) in pregnant women. The potential risk of vertical transmission is unclear, babies born from COVID-19-positive mothers seems to have no serious clinical symptoms, the possible mechanisms are discussed, which highlights that checking the children's outcome and more research is warranted. In this review, we investigate the reports concerning viral infections and COVID-19 during pregnancy, to establish a correlation and possible implications of COVID-19 during pregnancy and neonatal's health.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/transmisión , Neumonía Viral/inmunología , Neumonía Viral/transmisión , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , COVID-19 , Preescolar , Infecciones por Coronavirus/sangre , Infecciones por Coronavirus/virología , Citocinas/sangre , Femenino , Desarrollo Fetal/inmunología , Humanos , Lactante , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Madres , Pandemias , Neumonía Viral/sangre , Neumonía Viral/virología , Embarazo , Complicaciones Infecciosas del Embarazo/sangre , SARS-CoV-2
15.
Front Immunol ; 11: 1681, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849586

RESUMEN

The complement cascade was identified over 100 years ago, yet investigation of its role in pregnancy remains an area of intense research. Complement inhibitors at the maternal-fetal interface prevent inappropriate complement activation to protect the fetus. However, this versatile proteolytic cascade also favorably influences numerous stages of pregnancy, including implantation, fetal development, and labor. Inappropriate complement activation in pregnancy can have adverse lifelong sequelae for both mother and child. This review summarizes the current understanding of complement activation during all stages of pregnancy. In addition, consequences of complement dysregulation during adverse pregnancy outcomes from miscarriage, preeclampsia, and pre-term birth are examined. Finally, future research directions into complement activation during pregnancy are considered.


Asunto(s)
Activación de Complemento/inmunología , Implantación del Embrión/inmunología , Parto/inmunología , Complicaciones del Embarazo/inmunología , Embarazo/inmunología , Proteínas del Sistema Complemento/inmunología , Femenino , Desarrollo Fetal/inmunología , Humanos
16.
Front Immunol ; 11: 1035, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582165

RESUMEN

Preterm infants born before 32 weeks gestational age (GA) have high rates of late onset sepsis (LOS) and necrotizing enterocolitis (NEC) despite recent improvements in infection control and nutrition. Breast milk has a clear protective effect against both these outcomes likely due to multiple mechanisms which are not fully understood but may involve effects on both the infant's immune system and the developing gut microbiota. Congregating at the interface between the mucosal barrier and the microbiota, innate and adaptive T lymphocytes (T cells) participate in this interaction but few studies have explored their development after preterm delivery. We conducted a literature review of T cell development that focuses on fetal development, postnatal maturation and the influence of milk diet. The majority of circulating T cells in the preterm infant display a naïve phenotype but are still able to initiate functional responses similar to those seen in term infants. T cells from preterm infants display a skew toward a T-helper 2(Th2) phenotype and have an increased population of regulatory cells (Tregs). There are significant gaps in knowledge in this area, particularly in regards to innate-like T cells, but work is emerging: transcriptomics and mass cytometry are currently being used to map out T cell development, whilst microbiomic approaches may help improve understanding of events at mucosal surfaces. A rapid rise in organoid models will allow robust exploration of host-microbe interactions and may support the development of interventions that modulate T-cell responses for improved infant health.


Asunto(s)
Recien Nacido Prematuro/inmunología , Leche Humana/inmunología , Subgrupos de Linfocitos T/inmunología , Inmunidad Adaptativa , Susceptibilidad a Enfermedades , Enterocolitis Necrotizante/inmunología , Femenino , Desarrollo Fetal/inmunología , Microbioma Gastrointestinal/inmunología , Interacciones Microbiota-Huesped/inmunología , Humanos , Inmunidad Innata , Inmunidad Mucosa , Fórmulas Infantiles , Fenómenos Fisiológicos Nutricionales del Lactante/inmunología , Recién Nacido , Leche Humana/citología , Modelos Inmunológicos , Embarazo , Sepsis/inmunología
17.
Science ; 368(6491): 608-612, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32381717

RESUMEN

Viral infections during pregnancy can have devastating consequences on pregnancy outcomes, fetal development, and maternal health. In this review, we examine fetal and maternal immune defense mechanisms that mediate resistance against viral infections and discuss the range of syndromes that ensue when such mechanisms fail, from fetal developmental defects to establishment of chronic infection. Further, we highlight the role of maternal immune activation, or uncontrolled inflammation triggered by viral infections during pregnancy, and its potential downstream pathological effects, including tissue damage and fetal demise. Insights into the respective contributions of direct viral toxicity versus fetal and maternal immune responses that underlie the pathogenesis of congenital disease will guide future treatment strategies.


Asunto(s)
Enfermedades Fetales/virología , Feto/anomalías , Intercambio Materno-Fetal/inmunología , Complicaciones Infecciosas del Embarazo/virología , Virosis/inmunología , Femenino , Desarrollo Fetal/inmunología , Enfermedades Fetales/inmunología , Feto/inmunología , Feto/virología , Humanos , Inmunidad Innata , Embarazo , Resultado del Embarazo
18.
Dev Neurobiol ; 80(5-6): 149-159, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32333505

RESUMEN

Fetuses exposed to an inflammatory environment are predisposed to long-term adverse neurological outcomes. However, the mechanism by which intrauterine inflammation (IUI) is responsible for abnormal fetal brain development is not fully understood. The mechanistic target of rapamycin (mTOR) signaling pathway is closely associated with fetal brain development. We hypothesized that mTOR signaling might be involved in fetal brain injury and malformation when fetuses are exposed to the IUI environment. A well-established IUI model was utilized by intrauterine injection of lipopolysaccharide (LPS) to explore the effect of IUI on mTOR signaling in mouse fetal brains. We found that microglia activation in LPS fetal brains was increased, as demonstrated by elevated Iba-1 protein level and immunofluorescence density. LPS fetal brains also showed reduced neuronal cell counts, decreased cell proliferation demonstrated by low Ki67-positive density, and elevated neuron apoptosis evidenced by high expression of cleaved Caspase 3. Furthermore, we found that mTOR signaling in LPS fetal brains was elevated at 2 hr after LPS treatment, declined at 6 hr and showed overall inhibition at 24 hr. In summary, our study revealed that LPS-induced IUI leads to increased activation of microglia cells, neuronal damage, and dynamic alterations in mTOR signaling in the mouse fetal brain. Our findings indicate that abnormal changes in mTOR signaling may underlie the development of future neurological complications in offspring exposed to prenatal IUI.


Asunto(s)
Encefalopatías , Corteza Cerebral , Desarrollo Fetal/fisiología , Enfermedades Fetales , Inflamación , Microglía , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Encefalopatías/inmunología , Encefalopatías/metabolismo , Encefalopatías/patología , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Desarrollo Fetal/inmunología , Enfermedades Fetales/inmunología , Enfermedades Fetales/metabolismo , Enfermedades Fetales/patología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Ratones , Microglía/inmunología , Microglía/metabolismo , Embarazo , Transducción de Señal/inmunología
19.
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
20.
Nat Rev Immunol ; 20(5): 279-293, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31853049

RESUMEN

Much of our knowledge regarding the interactions between epithelial tissues and the immune system has been gathered from animal models and co-cultures with cell lines. However, unique features of human cells cannot be modelled in mice, and cell lines are often transformed or genetically immortalized. Organoid technology has emerged as a powerful tool to maintain epithelial cells in a near-native state. In this Review, we discuss how organoids are being used in immunological research to understand the role of epithelial cell-immune cell interactions in tissue development and homeostasis, as well as in diseases such as cancer.


Asunto(s)
Células Epiteliales/inmunología , Epitelio/inmunología , Desarrollo Fetal/inmunología , Infecciones/inmunología , Linfopoyesis/inmunología , Organoides/inmunología , Regeneración/inmunología , Alergia e Inmunología , Investigación Biomédica , Diferenciación Celular/inmunología , Homeostasis , Humanos , Linfocitos T/inmunología , Timo/inmunología , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA