Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 737
Filtrar
1.
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
2.
Immunol Rev ; 315(1): 11-30, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36929134

RESUMEN

It has been over three decades since Drs. Herzenberg and Herzenberg proposed the layered immune system hypothesis, suggesting that different types of stem cells with distinct hematopoietic potential produce specific immune cells. This layering of immune system development is now supported by recent studies showing the presence of fetal-derived immune cells that function in adults. It has been shown that various immune cells arise at different embryonic ages via multiple waves of hematopoiesis from special endothelial cells (ECs), referred to as hemogenic ECs. However, it remains unknown whether these fetal-derived immune cells are produced by hematopoietic stem cells (HSCs) during the fetal to neonatal period. To address this question, many advanced tools have been used, including lineage-tracing mouse models, cellular barcoding techniques, clonal assays, and transplantation assays at the single-cell level. In this review, we will review the history of the search for the origins of HSCs, B-1a progenitors, and mast cells in the mouse embryo. HSCs can produce both B-1a and mast cells within a very limited time window, and this ability declines after embryonic day (E) 14.5. Furthermore, the latest data have revealed that HSC-independent adaptive immune cells exist in adult mice, which implies more complicated developmental pathways of immune cells. We propose revised road maps of immune cell development.


Asunto(s)
Sistema Inmunológico , Sistema Inmunológico/citología , Sistema Inmunológico/crecimiento & desarrollo , Humanos , Animales , Hematopoyesis , Embrión de Mamíferos/citología , Células Madre Hematopoyéticas/citología , Linfocitos/citología , Linaje de la Célula
3.
Immunol Rev ; 315(1): 108-125, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36653953

RESUMEN

Historically, the immune system was believed to develop along a linear axis of maturity from fetal life to adulthood. Now, it is clear that distinct layers of immune cells are generated from unique waves of hematopoietic progenitors during different windows of development. This model, known as the layered immune model, has provided a useful framework for understanding why distinct lineages of B cells and γδ T cells arise in succession and display unique functions in adulthood. However, the layered immune model has not been applied to CD8+ T cells, which are still often viewed as a uniform population of cells belonging to the same lineage, with functional differences between cells arising from environmental factors encountered during infection. Recent studies have challenged this idea, demonstrating that not all CD8+ T cells are created equally and that the functions of individual CD8+ T cells in adults are linked to when they were created in the host. In this review, we discuss the accumulating evidence suggesting there are distinct ontogenetic subpopulations of CD8+ T cells and propose that the layered immune model be extended to the CD8+ T cell compartment.


Asunto(s)
Linfocitos T CD8-positivos , Sistema Inmunológico , Subgrupos de Linfocitos T , Humanos , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Desarrollo Humano/fisiología , Sistema Inmunológico/citología , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/inmunología , Sistema Inmunológico/fisiología , Inmunidad/inmunología , Inmunidad/fisiología , Subgrupos de Linfocitos T/inmunología
4.
Am J Clin Nutr ; 115(2): 570-587, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-34634105

RESUMEN

Bioactive ingredients for infant formula have been sought to reduce disparities in health outcomes between breastfed and formula-fed infants. Traditional food safety methodologies have limited ability to assess some bioactive ingredients. It is difficult to assess the effects of nutrition on the infant immune system because of coincident developmental adaptations to birth, establishment of the microbiome and introduction to solid foods, and perinatal environmental factors. An expert panel was convened to review information on immune system development published since the 2004 Institute of Medicine report on evaluating the safety of new infant formula ingredients and to recommend measurements that demonstrate the safety of bioactive ingredients intended for that use. Panel members participated in a 2-d virtual symposium in November 2020 and in follow-up discussions throughout early 2021. Key topics included identification of immune system endpoints from nutritional intervention studies, effects of human milk feeding and human milk substances on infant health outcomes, ontologic development of the infant immune system, and microbial influences on tolerance. The panel explored how "nonnormal" conditions such as preterm birth, allergy, and genetic disorders could help define developmental immune markers for healthy term infants. With consideration of breastfed infants as a reference, ensuring proper control groups, and attention to numerous potential confounders, the panel recommended a set of standard clinical endpoints including growth, response to vaccination, infection and other adverse effects related to inflammation, and allergy and atopic diseases. It compiled a set of candidate markers to characterize stereotypical patterns of immune system development during infancy, but absence of reference ranges, variability in methods and populations, and unreliability of individual markers to predict disease prevented the panel from including many markers as safety endpoints. The panel's findings and recommendations are applicable for industry, regulatory, and academic settings, and will inform safety assessments for immunomodulatory ingredients in foods besides infant formula.


Asunto(s)
Ingredientes Alimentarios/análisis , Sistema Inmunológico/crecimiento & desarrollo , Fórmulas Infantiles/análisis , Fenómenos Fisiológicos Nutricionales del Lactante/inmunología , Fitoquímicos/inmunología , Femenino , Humanos , Lactante , Recién Nacido , Masculino
5.
Front Immunol ; 12: 774780, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899730

RESUMEN

Background and Aims: Congenital heart diseases (CHDs) are diagnosed in approximately 9 in 1,000 newborns, and early cardiac corrective surgery often requires partial or complete thymectomy. As the long-term effect of early thymectomy on the subsequent development of the immune system in humans has not been completely elucidated, the present study aimed to evaluate the effects of thymus removal on the functional capacity of the immune system after different periods. Methods: A systematic review of the literature was performed using MEDLINE, EMBASE, LILACS and Scopus. The inclusion criteria were original studies that analyzed any component of the immune system in patients with CHD who had undergone thymectomy during cardiac surgery in the first years of life. The results were evaluated for the quality of evidence. Results: Twenty-three studies were selected and showed that patients who underwent a thymectomy in the first years of life tended to exhibit important alterations in the T cell compartment, such as fewer total T cells, CD4+, CD8+, naïve and CD31+ T cells, lower TRECs, decreased diversity of the TCR repertoire and higher peripheral proliferation (increased Ki-67 expression) than controls. However, the numbers of memory T cells and Treg cells differed across the selected studies. Conclusions: Early thymectomy, either partial or complete, may be associated with a reduction in many T cell subpopulations and TCR diversity, and these alterations may persist during long-term follow-up. Alternative solutions should be studied, either in the operative technique with partial preservation of the thymus or through the autograft of fragments of the gland. Systematic Review Registration: Prospero [157188].


Asunto(s)
Desarrollo Infantil , Sistema Inmunológico/inmunología , Subgrupos de Linfocitos T/inmunología , Timectomía/efectos adversos , Timo/cirugía , Factores de Edad , Variación Antigénica , Proliferación Celular , Niño , Preescolar , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Memoria Inmunológica , Lactante , Recién Nacido , Fenotipo , Receptores de Antígenos de Linfocitos T/inmunología , Timo/inmunología , Resultado del Tratamiento
6.
Physiol Res ; 70(S2): S209-S225, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34913353

RESUMEN

The SARS-CoV-2 pandemic has indeed been one of the most significant problems facing the world in the last decade. It has affected (directly or indirectly) the entire population and all age groups. Children have accounted for 1.7 % to 2 % of the diagnosed cases of COVID-19. COVID-19 in children is usually associated with a mild course of the disease and a better survival rate than in adults. In this review, we investigate the different mechanisms which underlie this observation. Generally, we can say that the innate immune response of children is strong because they have a trained immunity, allowing the early control of infection at the site of entry. Suppressed adaptive immunity and a dysfunctional innate immune response is seen in adult patients with severe infections but not in children. This may relate to immunosenescence in the elderly. Another proposed factor is the different receptors for SARS-CoV-2 and their differences in expression between these age groups. In infants and toddlers, effective immune response to viral particles can be modulated by the pre-existing non-specific effect of live attenuated vaccines on innate immunity and vitamin D prophylaxis. However, all the proposed mechanisms require verification in larger cohorts of patients. Our knowledge about SARS-CoV-2 is still developing.


Asunto(s)
Desarrollo del Adolescente , COVID-19/fisiopatología , Desarrollo Infantil , Sistema Inmunológico/crecimiento & desarrollo , SARS-CoV-2/patogenicidad , Adolescente , Adulto , Factores de Edad , COVID-19/inmunología , COVID-19/terapia , COVID-19/virología , Niño , Preescolar , Interacciones Huésped-Patógeno , Humanos , Sistema Inmunológico/fisiopatología , Sistema Inmunológico/virología , Lactante , Recién Nacido , Pronóstico , Medición de Riesgo , Factores de Riesgo , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Adulto Joven
7.
Front Immunol ; 12: 791081, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868080

RESUMEN

Dysregulation of the immune system is associated with many pathologies, including cardiovascular diseases, diabetes, and cancer. To date, the most commonly used models in biomedical research are rodents, and despite the various advantages they offer, their use also raises numerous drawbacks. Recently, another in vivo model, the chicken embryo and its chorioallantoic membrane, has re-emerged for various applications. This model has many benefits compared to other classical models, as it is cost-effective, time-efficient, and easier to use. In this review, we explain how the chicken embryo can be used as a model for immune-based studies, as it gradually develops an embryonic immune system, yet which is functionally similar to humans'. We mainly aim to describe the avian immune system, highlighting the differences and similarities with the human immune system, including the repertoire of lymphoid tissues, immune cells, and other key features. We also describe the general in ovo immune ontogeny. In conclusion, we expect that this review will help future studies better tailor their use of the chicken embryo model for testing specific experimental hypotheses or performing preclinical testing.


Asunto(s)
Embrión de Pollo/inmunología , Membrana Corioalantoides/inmunología , Sistema Inmunológico/inmunología , Animales , Embrión de Pollo/metabolismo , Membrana Corioalantoides/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/metabolismo , Mediadores de Inflamación/metabolismo , Modelos Animales , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Transducción de Señal , Especificidad de la Especie
8.
Cells ; 10(12)2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34944105

RESUMEN

Emerging evidence indicates that perinatal infection and inflammation can influence the developing immune system and may ultimately affect long-term health and disease outcomes in offspring by perturbing tissue and immune homeostasis. We posit that perinatal inflammation influences immune outcomes in offspring by perturbing (1) the development and function of fetal-derived immune cells that regulate tissue development and homeostasis, and (2) the establishment and function of developing hematopoietic stem cells (HSCs) that continually generate immune cells across the lifespan. To disentangle the complexities of these interlinked systems, we propose the cochlea as an ideal model tissue to investigate how perinatal infection affects immune, tissue, and stem cell development. The cochlea contains complex tissue architecture and a rich immune milieu that is established during early life. A wide range of congenital infections cause cochlea dysfunction and sensorineural hearing loss (SNHL), likely attributable to early life inflammation. Furthermore, we show that both immune cells and bone marrow hematopoietic progenitors can be simultaneously analyzed within neonatal cochlear samples. Future work investigating the pathogenesis of SNHL in the context of congenital infection will therefore provide critical information on how perinatal inflammation drives disease susceptibility in offspring.


Asunto(s)
Cóclea/patología , Hematopoyesis , Sistema Inmunológico/crecimiento & desarrollo , Inflamación/patología , Animales , Feto/inmunología , Pérdida Auditiva Sensorineural/inmunología , Humanos
9.
Biomolecules ; 11(12)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34944387

RESUMEN

Breastfeeding-or lactation-is a unique and defining reproductive trait of mammals that nourishes offspring by supplying nutrient-rich breast milk [...].


Asunto(s)
Factores Biológicos/química , Lactancia Materna/estadística & datos numéricos , Leche Humana/química , Factores Biológicos/inmunología , Lactancia Materna/tendencias , Desarrollo Infantil , Femenino , Homeostasis , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Lactante , Leche Humana/inmunología
10.
Nutrients ; 13(12)2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34959752

RESUMEN

Intestinal colonization of the neonate is highly dependent on the term of pregnancy, the mode of delivery, the type of feeding [breast feeding or formula feeding]. Postnatal immune maturation is dependent on the intestinal microbiome implementation and composition and type of feeding is a key issue in the human gut development, the diversity of microbiome, and the intestinal function. It is well established that exclusive breastfeeding for 6 months or more has several benefits with respect to formula feeding. The composition of the new generation of infant formulas aims in mimicking HM by reproducing its beneficial effects on intestinal microbiome and on the gut associated immune system (GAIS). Several approaches have been developed currently for designing new infant formulas by the addition of bioactive ingredients such as human milk oligosaccharides (HMOs), probiotics, prebiotics [fructo-oligosaccharides (FOSs) and galacto-oligosaccharides (GOSs)], or by obtaining the so-called post-biotics also known as milk fermentation products. The aim of this article is to guide the practitioner in the understanding of these different types of Microbiota Influencing Formulas by listing and summarizing the main concepts and characteristics of these different models of enriched IFs with bioactive ingredients.


Asunto(s)
Ingestión de Alimentos/inmunología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/microbiología , Fórmulas Infantiles/química , Fenómenos Fisiológicos Nutricionales del Lactante/inmunología , Femenino , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Fórmulas Infantiles/microbiología , Recién Nacido , Intestinos/crecimiento & desarrollo , Intestinos/inmunología , Masculino , Leche Humana/química , Leche Humana/microbiología , Oligosacáridos/administración & dosificación , Prebióticos/administración & dosificación
11.
Front Immunol ; 12: 754589, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34707617

RESUMEN

In many countries where tuberculosis (TB) is endemic, the Bacillus Calmette-Guérin (BCG) vaccine is given as close to birth as possible to protect infants and children from severe forms of TB. However, BCG has variable efficacy and is not as effective against adult pulmonary TB. At present, most animal models used to study novel TB vaccine candidates rely on the use of adult animals. Human studies show that the infant immune system is different to that of an adult. Understanding how the phenotypic profile and functional ability of the immature host immune system compares to that of a mature adult, together with the subsequent BCG immune response, is critical to ensuring that new TB vaccines are tested in the most appropriate models. BCG-specific immune responses were detected in macaques vaccinated within a week of birth from six weeks after immunization indicating that neonatal macaques are able to generate a functional cellular response to the vaccine. However, the responses measured were significantly lower than those typically observed following BCG vaccination in adult rhesus macaques and infant profiles were skewed towards the activation and attraction of macrophages and monocytes and the synthesis in addition to release of pro-inflammatory cytokines such as IL-1, IL-6 and TNF-α. The frequency of specific immune cell populations changed significantly through the first three years of life as the infants developed into young adult macaques. Notably, the CD4:CD8 ratio significantly declined as the macaques aged due to a significant decrease in the proportion of CD4+ T-cells relative to a significant increase in CD8+ T-cells. Also, the frequency of both CD4+ and CD8+ T-cells expressing the memory marker CD95, and memory subset populations including effector memory, central memory and stem cell memory, increased significantly as animals matured. Infant macaques, vaccinated with BCG within a week of birth, possessed a significantly higher frequency of CD14+ classical monocytes and granulocytes which remained different throughout the first three years of life compared to unvaccinated age matched animals. These findings, along with the increase in monokines following vaccination in infants, may provide an insight into the mechanism by which vaccination with BCG is able to provide non-specific immunity against non-mycobacterial organisms.


Asunto(s)
Envejecimiento/inmunología , Vacuna BCG/inmunología , Sistema Inmunológico/crecimiento & desarrollo , Inmunogenicidad Vacunal , Macaca mulatta/inmunología , Animales , Animales Recién Nacidos/inmunología , Antígenos Bacterianos/inmunología , Biomarcadores , Relación CD4-CD8 , Citocinas/sangre , Femenino , Inmunidad Innata , Esquemas de Inmunización , Memoria Inmunológica , Péptidos y Proteínas de Señalización Intercelular/sangre , Interferón gamma/sangre , Macaca mulatta/crecimiento & desarrollo , Macrófagos/inmunología , Masculino , Monocitos/inmunología , Mycobacterium tuberculosis/inmunología , Especificidad de la Especie , Tuberculina/inmunología
12.
Nutrients ; 13(10)2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34684311

RESUMEN

Infant formulas offer an alternative to breast milk for both normal birth weight (NBW) and immunocompromised intrauterine growth restricted (IUGR) infants. Although the lipid fraction in formulas is often derived from vegetable oils, it is unclear if this alters immunological outcomes relative to milk fats or whether these effects differ between IUGR and NBW infants. We hypothesized that replacing vegetable oil with bovine milk fat in infant formula would improve immune development in IUGR and NBW neonates. Two-day old piglets were selected (NBW, n = 18, IUGR, n = 18) and each group of animals were fed formula based on either vegetable oil (VEG) or bovine milk fat (MILK). Animals were reared until day 23/24 and systemic immune parameters were evaluated. Milk-fat feeding decreased blood neutrophil counts and improved neutrophil function while transiently reducing leucocytes' expression of genes related to adaptive and innate immunity as well as energy metabolism, following in vitro stimulation by live Staphylococcus epidermidis (whole blood, 2 h). However, there were only a few interactions between milk-fat type and birthweight status. Thus, piglets fed milk-fat-based formula had improved neutrophil maturation and suppressed pro-inflammatory responses, compared to those fed vegetable-oil-based formula.


Asunto(s)
Peso al Nacer , Grasas/química , Retardo del Crecimiento Fetal/patología , Sistema Inmunológico/crecimiento & desarrollo , Fórmulas Infantiles , Leche/química , Inmunidad Adaptativa , Animales , Animales Recién Nacidos , Retardo del Crecimiento Fetal/genética , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/genética , Recién Nacido , Monocitos/metabolismo , Neutrófilos/metabolismo , Linfocitos T/metabolismo
13.
Nutrients ; 13(9)2021 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-34578971

RESUMEN

Human breast milk (HBM) is not only an indispensable source of nutrients for early human growth and development, supplying components that support infant growth and development, but also contains various essential immunologic components with anti-infectious activities and critical roles in the formation of immunity. It is also known that HBM contains its own unique microbiome, including beneficial, commensal, and potentially probiotic bacteria, that can contribute to infant gut colonization. In addition, HBM-derived extracellular vesicles, exosomes, and microRNA are attracting increasing interest for their potential to transfer to the infant and their role in infant development. In this article, we examine some of the various constituents in HBM and review the evidence supporting their associated health effects and their potential applications in human health.


Asunto(s)
Sistema Inmunológico/crecimiento & desarrollo , MicroARNs/fisiología , Microbiota/fisiología , Leche Humana/química , Leche Humana/fisiología , Valor Nutritivo/fisiología , Desarrollo Infantil/fisiología , Femenino , Estado de Salud , Humanos , Lactante , Recién Nacido , Leche Humana/inmunología
14.
Cell Host Microbe ; 29(10): 1558-1572.e6, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34480872

RESUMEN

Premature infants are at substantial risk for suffering from perinatal white matter injury. Though the gut microbiota has been implicated in early-life development, a detailed understanding of the gut-microbiota-immune-brain axis in premature neonates is lacking. Here, we profiled the gut microbiota, immunological, and neurophysiological development of 60 extremely premature infants, which received standard hospital care including antibiotics and probiotics. We found that maturation of electrocortical activity is suppressed in infants with severe brain damage. This is accompanied by elevated γδ T cell levels and increased T cell secretion of vascular endothelial growth factor and reduced secretion of neuroprotectants. Notably, Klebsiella overgrowth in the gut is highly predictive for brain damage and is associated with a pro-inflammatory immunological tone. These results suggest that aberrant development of the gut-microbiota-immune-brain axis may drive or exacerbate brain injury in extremely premature neonates and represents a promising target for novel intervention strategies.


Asunto(s)
Lesiones Encefálicas/inmunología , Lesiones Encefálicas/microbiología , Microbioma Gastrointestinal , Recien Nacido Prematuro/crecimiento & desarrollo , Bacterias/clasificación , Bacterias/genética , Bacterias/crecimiento & desarrollo , Bacterias/aislamiento & purificación , Encéfalo/crecimiento & desarrollo , Lesiones Encefálicas/fisiopatología , Femenino , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Recién Nacido , Recien Nacido Prematuro/inmunología , Masculino , Linfocitos T/inmunología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/inmunología
16.
Immunity ; 54(8): 1633-1635, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34380062

RESUMEN

Immune-system maturation starts early in life, but studies investigating immune-system education in human infants remain scarce. In a recent issue of Cell, Henrick et al. study early gut microbiota and immune-system development in two infant cohorts. The authors describe that Bifidobacteria can use milk sugars to produce immunoregulatory compounds that induce immune tolerance and reduce intestinal inflammation.


Asunto(s)
Bifidobacterium/metabolismo , Sistema Inmunológico/crecimiento & desarrollo , Intestinos/inmunología , Intestinos/microbiología , Leche Humana/química , Oligosacáridos/metabolismo , Animales , Lactancia Materna , Microbioma Gastrointestinal/fisiología , Humanos , Tolerancia Inmunológica/inmunología , Factores Inmunológicos/química , Inmunomodulación/inmunología , Lactante , Suecia , Estados Unidos
17.
Front Immunol ; 12: 672853, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248954

RESUMEN

Through the release of hormones, the neuro-endocrine system regulates the immune system function promoting adaptation of the organism to the external environment and to intrinsic physiological changes. Glucocorticoids (GCs) and sex hormones not only regulate immune responses, but also control the hematopoietic stem cell (HSC) differentiation and subsequent maturation of immune cell subsets. During the development of an organism, this regulation has long-term consequences. Indeed, the effects of GC exposure during the perinatal period become evident in the adulthood. Analogously, in the context of HSC transplantation (HSCT), the immune system development starts de novo from the donor HSCs. In this review, we summarize the effects of GCs and sex hormones on the regulation of HSC, as well as of adaptive and innate immune cells. Moreover, we discuss the short and long-term implications on hematopoiesis of sex steroid ablation and synthetic GC administration upon HSCT.


Asunto(s)
Glucocorticoides/inmunología , Hormonas Esteroides Gonadales/inmunología , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/inmunología , Animales , Humanos
18.
Cell ; 184(15): 3884-3898.e11, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34143954

RESUMEN

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


Asunto(s)
Bifidobacterium/fisiología , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/microbiología , Antibacterianos/farmacología , Biomarcadores/metabolismo , Lactancia Materna , Linfocitos T CD4-Positivos/inmunología , Polaridad Celular , Proliferación Celular , Citocinas/metabolismo , Heces/química , Heces/microbiología , Galectina 1/metabolismo , Microbioma Gastrointestinal , Humanos , Indoles/metabolismo , Recién Nacido , Inflamación/sangre , Inflamación/genética , Mucosa Intestinal/inmunología , Metaboloma , Leche Humana/química , Oligosacáridos/metabolismo , Células Th17/inmunología , Células Th2/inmunología , Agua
19.
Elife ; 102021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34032570

RESUMEN

At the transition from intrauterine to postnatal life, drastic alterations are mirrored by changes in cellular immunity. These changes are in part immune cell intrinsic, originate in the replacement of fetal cells, or result from global regulatory mechanisms and adaptation to changes in the tissue microenvironment. Overall, longer developmental trajectories are intersected by events related to mother-infant separation, birth cues, acquisition of microbiota and metabolic factors. Perinatal alterations particularly affect immune niches, where structures with discrete functions meet, the intestinal mucosa, epidermis and lung. Accordingly, the following questions will be addressed in this review.How does the preprogrammed development supported by endogenous cues, steer innate immune cell differentiation, adaptation to tissue structures, and immunity to infection?How does the transition at birth impact on tissue immune make-up including its topology?How do postnatal cues guide innate immune cell differentiation and function at immunological niches?


Asunto(s)
Inmunidad Innata , Femenino , Homeostasis , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/embriología , Sistema Inmunológico/crecimiento & desarrollo , Recién Nacido , Inflamación/inmunología , Microbiota , Embarazo
20.
Front Immunol ; 12: 644269, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815397

RESUMEN

The first 1000 days of life, including the intrauterine period, are regarded as a fundamental stepping stone for the development of a human. Unequivocally, nutrition during this period plays a key role on the proper development of a child, both directly through the intake of essential nutrients and indirectly by affecting the composition of the gut microbiota. The gut microbiota, including bacteria, viruses, fungi, protists and other microorganisms, is a highly modifiable and adaptive system that is influenced by diet, lifestyle, medicinal products and the environment. Reversely, it affects the immune system in multiple complex ways. Many noncommunicable diseases (NCDs) associated with dysbiosis are "programmed" during childhood. Nutrition is a potent determinant of the children's microbiota composition and maturation and, therefore, a strong determinant of the NCDs' programming. In this review we explore the interplay between nutrition during the first 1000 days of life, the gut microbiota, virome and mycobiome composition and the development of NCDs.


Asunto(s)
Fenómenos Fisiológicos Nutricionales Infantiles/inmunología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico , Micobioma/inmunología , Enfermedades no Transmisibles , Viroma/inmunología , Niño , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA