Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 685
Filtrar
Más filtros

Intervalo de año de publicación
1.
Immunol Rev ; 315(1): 31-53, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36752151

RESUMEN

Mast cells (MCs) are evolutionarily ancient innate immune cells with important roles in protective immunity against bacteria, parasites, and venomous animals. They can be found in most organs of the body, where they also contribute to normal tissue functioning, for example by engaging in crosstalk with nerves. Despite this, they are most widely known for their detrimental roles in allergy, anaphylaxis, and atopic disease. Just like macrophages, mast cells were conventionally thought to originate from the bone marrow. However, they are already present in fetal tissues before the onset of bone marrow hematopoiesis, questioning this dogma. In recent years, our view of myeloid cell ontogeny has been revised. We now know that the first mast cells originate from progenitors made in the extra-embryonic yolk sac, and later get supplemented with mast cells produced from subsequent waves of hematopoiesis. In most connective tissues, sizeable populations of fetal-derived mast cells persist into adulthood, where they self-maintain largely independently from the bone marrow. These developmental origins are highly reminiscent of macrophages, which are known to have critical functions in development. Mast cells too may thus support healthy development. Their fetal origins and longevity also make mast cells susceptible to genetic and environmental perturbations, which may render them pathological. Here, we review our current understanding of mast cell biology from a developmental perspective. We first summarize how mast cell populations are established from distinct hematopoietic progenitor waves, and how they are subsequently maintained throughout life. We then discuss what functions mast cells may normally have at early life stages, and how they may be co-opted to cause, worsen, or increase susceptibility to disease.


Asunto(s)
Células Madre Hematopoyéticas , Mastocitos , Animales , Humanos , Macrófagos , Médula Ósea , Hematopoyesis/genética , Desarrollo Fetal , Diferenciación Celular
2.
Trends Immunol ; 44(11): 877-889, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37852863

RESUMEN

Early-life environmental exposures play a significant role in shaping long-lasting immune phenotypes and disease susceptibility. Nevertheless, comprehensive understanding of the developmental programming of immunity is limited. We propose that the vertebrate immune system contains durable programmable components established through early environmental interactions and maintained in a stable and homeostatic manner. Some immune components, such as immunological memory, are intrinsically programmable. Others are influenced by conditions during critical developmental windows in early life, including microbiota, hormones, metabolites, and environmental stress, which impact programming. Developmental immune programming can promote adaptation to an anticipated future environment. However, mismatches between predicted and actual environments can result in disease. This is relevant because understanding programming mechanisms can offer insights into the origin of inflammatory diseases, ideally enabling effective prevention and treatment strategies.


Asunto(s)
Sistema Inmunológico , Microbiota , Humanos , Fenotipo , Exposición a Riesgos Ambientales
3.
EMBO Rep ; 25(4): 1752-1772, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38491313

RESUMEN

Emerging evidence indicates that parental diseases can impact the health of subsequent generations through epigenetic inheritance. Recently, it was shown that maternal diabetes alters the metaphase II oocyte transcriptome, causing metabolic dysfunction in offspring. However, type 1 diabetes (T1D) mouse models frequently utilized in previous studies may be subject to several confounding factors due to severe hyperglycemia. This limits clinical translatability given improvements in glycemic control for T1D subjects. Here, we optimize a T1D mouse model to investigate the effects of appropriately managed maternal glycemic levels on oocytes and intrauterine development. We show that diabetic mice with appropriate glycemic control exhibit better long-term health, including maintenance of the oocyte transcriptome and chromatin accessibility. We further show that human oocytes undergoing in vitro maturation challenged with mildly increased levels of glucose, reflecting appropriate glycemic management, also retain their transcriptome. However, fetal growth and placental function are affected in mice despite appropriate glycemic control, suggesting the uterine environment rather than the germline as a pathological factor in developmental programming in appropriately managed diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Hiperglucemia , Humanos , Femenino , Embarazo , Ratones , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Placenta , Hiperglucemia/genética , Hiperglucemia/metabolismo , Oocitos/metabolismo , Modelos Animales de Enfermedad
4.
J Physiol ; 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38985827

RESUMEN

Fetal growth restriction (FGR) is a common outcome in human suboptimal gestation and is related to prenatal origins of cardiovascular dysfunction in offspring. Despite this, therapy of human translational potential has not been identified. Using human umbilical and placental vessels and the chicken embryo model, we combined cellular, molecular, and functional studies to determine whether N-acetylcysteine (NAC) and hydrogen sulphide (H2S) protect cardiovascular function in growth-restricted unborn offspring. In human umbilical and placental arteries from control or FGR pregnancy and in vessels from near-term chicken embryos incubated under normoxic or hypoxic conditions, we determined the expression of the H2S gene CTH (i.e. cystathionine γ-lyase) (via quantitative PCR), the production of H2S (enzymatic activity), the DNA methylation profile (pyrosequencing) and vasodilator reactivity (wire myography) in the presence and absence of NAC treatment. The data show that FGR and hypoxia increased CTH expression in the embryonic/fetal vasculature in both species. NAC treatment increased aortic CTH expression and H2S production and enhanced third-order femoral artery dilator responses to the H2S donor sodium hydrosulphide in chicken embryos. NAC treatment also restored impaired endothelial relaxation in human third-to-fourth order chorionic arteries from FGR pregnancies and in third-order femoral arteries from hypoxic chicken embryos. This NAC-induced protection against endothelial dysfunction in hypoxic chicken embryos was mediated via nitric oxide independent mechanisms. Both developmental hypoxia and NAC promoted vascular changes in CTH DNA and NOS3 methylation patterns in chicken embryos. Combined, therefore, the data support that the effects of NAC and H2S offer a powerful mechanism of human translational potential against fetal cardiovascular dysfunction in complicated pregnancy. KEY POINTS: Gestation complicated by chronic fetal hypoxia and fetal growth restriction (FGR) increases a prenatal origin of cardiovascular disease in offspring, increasing interest in antenatal therapy to prevent against a fetal origin of cardiovascular dysfunction. We investigated the effects between N-acetylcysteine (NAC) and hydrogen sulphide (H2S) in the vasculature in FGR human pregnancy and in chronically hypoxic chicken embryos. Combining cellular, molecular, epigenetic and functional studies, we show that the vascular expression and synthesis of H2S is enhanced in hypoxic and FGR unborn offspring in both species and this acts to protect their vasculature. Therefore, the NAC/H2S pathway offers a powerful therapeutic mechanism of human translational potential against fetal cardiovascular dysfunction in complicated pregnancy.

5.
Am J Physiol Renal Physiol ; 326(5): F727-F736, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38511219

RESUMEN

Although obesity is recognized as a risk factor for cardiorenal and metabolic diseases, the impact of parental obesity on the susceptibility of their offspring to renal injury at adulthood is unknown. We examined the impact of parental obesity on offspring kidney function, morphology, and markers of kidney damage after acute kidney injury (AKI). Offspring from normal (N) diet-fed C57BL/6J parents were fed either N (NN) or a high-fat (H) diet (NH) from weaning until adulthood. Offspring from obese H diet-fed parents were fed N (HN) or H diet (HH) after weaning. All offspring groups were submitted to bilateral AKI by clamping the left and right renal pedicles for 30 min. Compared with male NH and NN offspring from lean parents, male HH and HN offspring from obese parents exhibited higher kidney injury markers such as urinary, renal osteopontin, plasma creatinine, urinary albumin excretion, and neutrophil gelatinase-associated lipocalin (NGAL) levels, and worse histological injury score at 22 wk of age. Only albumin excretion and NGAL were elevated in female HH offspring from obese parents compared with lean and obese offspring from lean parents. We also found an increased mortality rate and worse kidney injury scores after AKI in male offspring from obese parents, regardless of the diet consumed after weaning. Female offspring were protected from major kidney injury after AKI. These results indicate that parental obesity leads to increased kidney injury in their offspring after ischemia-reperfusion in a sex-dependent manner, even when their offspring remain lean.NEW & NOTEWORTHY Offspring from obese parents are more susceptible to kidney injury and worse outcomes following an acute ischemia-reperfusion insult. Male, but not female, offspring from obese parents exhibit increased blood pressure early in life. Female offspring are partially protected against major kidney injury induced by ischemia-reperfusion.


Asunto(s)
Lesión Renal Aguda , Riñón , Ratones Endogámicos C57BL , Daño por Reperfusión , Animales , Masculino , Femenino , Daño por Reperfusión/patología , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/patología , Riñón/fisiopatología , Riñón/patología , Riñón/metabolismo , Factores Sexuales , Obesidad/complicaciones , Obesidad/fisiopatología , Dieta Alta en Grasa , Embarazo , Lipocalina 2/metabolismo , Obesidad Materna/metabolismo , Obesidad Materna/complicaciones , Obesidad Materna/fisiopatología , Efectos Tardíos de la Exposición Prenatal , Ratones , Factores de Riesgo , Modelos Animales de Enfermedad , Biomarcadores/sangre
6.
Biol Reprod ; 110(2): 339-354, 2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-37971364

RESUMEN

Entering pregnancy with a history of adversity, including adverse childhood experiences and racial discrimination stress, is a predictor of negative maternal and fetal health outcomes. Little is known about the biological mechanisms by which preconception adverse experiences are stored and impact future offspring health outcomes. In our maternal preconception stress (MPS) model, female mice underwent chronic stress from postnatal days 28-70 and were mated 2 weeks post-stress. Maternal preconception stress dams blunted the pregnancy-induced shift in the circulating extracellular vesicle proteome and reduced glucose tolerance at mid-gestation, suggesting a shift in pregnancy adaptation. To investigate MPS effects at the maternal:fetal interface, we probed the mid-gestation placental, uterine, and fetal brain tissue transcriptome. Male and female placentas differentially regulated expression of genes involved in growth and metabolic signaling in response to gestation in an MPS dam. We also report novel offspring sex- and MPS-specific responses in the uterine tissue apposing these placentas. In the fetal compartment, MPS female offspring reduced expression of neurodevelopmental genes. Using a ribosome-tagging transgenic approach we detected a dramatic increase in genes involved in chromatin regulation in a PVN-enriched neuronal population in females at PN21. While MPS had an additive effect on high-fat-diet (HFD)-induced weight gain in male offspring, both MPS and HFD were necessary to induce significant weight gain in female offspring. These data highlight the preconception period as a determinant of maternal health in pregnancy and provides novel insights into mechanisms by which maternal stress history impacts offspring developmental programming.


Asunto(s)
Placenta , Aumento de Peso , Humanos , Embarazo , Ratones , Femenino , Masculino , Animales , Placenta/metabolismo , Feto/metabolismo , Transducción de Señal , Dieta Alta en Grasa/efectos adversos
7.
Biol Reprod ; 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38857381

RESUMEN

Choline is a vital micronutrient that can be utilized in the formation of betaine and multiple phospholipids. In this study, we aimed to confirm, and expand on previous findings, how choline impacts embryos from the first 7 days of development to affect postnatal phenotype. Bos indicus embryos were cultured in a choline-free medium (termed vehicle) or medium supplemented with 1.8 mM choline Blastocyst-stage embryos were transferred into crossbred recipients. Once born, calves were evaluated at birth, 94 d, 178 d and at weaning (average age = 239 d). Following weaning, all calves were enrolled into a feed efficiency trial before being separated by sex, with males being slaughtered at approximately 580 d of age and females followed until their first pregnancy check. Results confirm that exposure of 1.8 mM choline chloride during the first 7 d of development alters postnatal characteristics of the resultant calves. Calves of both sexes from choline-treated embryos were consistently heavier through weaning and males had heavier testes at 3 mo of age. There were sex-dependent alterations in DNA methylation in whole blood caused by choline treatment. After weaning, feed efficiency was affected by an interaction with sex, with choline calves being more efficient for females and less efficient for males. Calves from choline-treated embryos were heavier, or tended to be heavier, than calves from vehicle embryos at all observations after weaning. Carcass weight was heavier for choline calves and the cross-sectional area of the Longissumus thoracis muscle was increased by choline. Few females became pregnant during the experiment although numerically more choline females were pregnant than vehicle females. Results confirm that exposure of the preimplantation embryo to 1.8 mM choline can alter phenotypes of the resultant calves through the first 19 months after birth.

8.
Eur J Clin Invest ; : e14266, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38864773

RESUMEN

BACKGROUND: Pregnancy represents a window of vulnerability to fetal development. Disruptions in the prenatal environment during this crucial period can increase the risk of the offspring developing diseases over the course of their lifetime. The central nervous system (CNS) has been shown to be particularly susceptible to changes during crucial developmental windows. To date, research focused on disruptions in the development of the CNS has predominantly centred on the brain, revealing a correlation between exposure to prenatal risk factors and the onset of neuropsychiatric disorders. Nevertheless, some studies indicate that the retina, which is part of the CNS, is also vulnerable to in utero alterations during pregnancy. Such changes may affect neuronal, glial and vascular components of the retina, compromising retinal structure and function and possibly impairing visual function. METHODS: A search in the PubMed database was performed, and any literature concerning prenatal risk factors (drugs, diabetes, unbalanced diet, infection, glucocorticoids) affecting the offspring retina were included. RESULTS: This review collects evidence on the cellular, structural and functional changes occurring in the retina triggered by maternal risk factors during pregnancy. We highlight the adverse impact on retinal development and its long-lasting effects, providing a critical analysis of the current knowledge while underlining areas for future research. CONCLUSIONS: Appropriate recognition of the prenatal risk factors that negatively impact the developing retina may provide critical clues for the design of preventive strategies and for early therapeutic intervention that could change retinal pathology in the progeny.

9.
J Exp Biol ; 227(Suppl_1)2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38449325

RESUMEN

Although the long-lasting effects of variation in early-life environment have been well documented across organisms, the underlying causal mechanisms are only recently starting to be unraveled. Yet understanding the underlying mechanisms of long-lasting effects can help us predict how organisms will respond to changing environments. Birds offer a great system in which to study developmental plasticity and its underlying mechanisms owing to the production of large external eggs and variation in developmental trajectories, combined with a long tradition of applied, physiological, ecological and evolutionary research. Epigenetic changes (such as DNA methylation) have been suggested to be a key mechanism mediating long-lasting effects of the early-life environment across taxa. More recently, changes in the early-life gut microbiome have been identified as another potential mediator of developmental plasticity. As a first step in understanding whether these mechanisms contribute to developmental plasticity in birds, this Review summarizes how changes in early-life environment (both prenatal and postnatal) influence epigenetic markers and the gut microbiome. The literature shows how both early-life biotic (such as resources and social environment) and abiotic (thermal environment and various anthropogenic stressors) factors modify epigenetic markers and the gut microbiome in birds, yet data concerning many other environmental factors are limited. The causal links of these modifications to lasting phenotypic changes are still scarce, but changes in the hypothalamic-pituitary-adrenal axis have been identified as one putative pathway. This Review identifies several knowledge gaps, including data on the long-term effects, stability of the molecular changes, and lack of diversity in the systems studied, and provides directions for future research.


Asunto(s)
Aves , Epigénesis Genética , Microbiota , Animales , Aves/genética , Aves/crecimiento & desarrollo , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal
10.
Environ Res ; 249: 118323, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38336161

RESUMEN

Telomere length (TL) is a biomarker for cellular senescence and TL erosion is predictive of the risk for age-related diseases. Despite being genetically determined at birth, TL may be susceptible to modifications through epigenetic mechanisms. Pollutant agents are considered one of the major threats to both human and planetary health. Their ability to cross the placental barrier and induce oxidative stress in fetal cells is particularly concerning and it may be associated with early TL erosion. In consideration of the timely relevance of this topic, we conducted a literature review on the impact of prenatal exposure to pollutant agents on newborn TL. The search yielded a total of 1099 records, of which only 32 met the inclusion criteria for the review. These criteria included the participation of human subjects, a longitudinal design or collection of longitudinal data, reporting of original TL data, and a focus on exposure to pollutant agents. The majority of the studies reported a significant inverse association between prenatal exposure to pollutant agents and TL. Furthermore, the second trimester of pregnancy emerged as a special sensitive period for the occurrence of pollutant agent-driven TL modifications. Sex differences were inconsistently reported across studies. This review contributes to highlighting biochemical pathways for the threats of environmental pollution to human health. Future research is warranted to further highlight potential buffering mechanisms.


Asunto(s)
Contaminantes Ambientales , Humanos , Embarazo , Femenino , Contaminantes Ambientales/toxicidad , Telómero/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Exposición a Riesgos Ambientales/efectos adversos , Recién Nacido , Exposición Materna/efectos adversos , Contaminación Ambiental/efectos adversos
11.
J Dairy Sci ; 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38608953

RESUMEN

The performance of an adult dairy cow may be influenced by events that occur before her birth. The present study investigated potential effects of 2 prenatal groups of factors, Assisted Reproductive Techniques (ART) and maternal characteristics (e.g., dam parity), on offspring performance during their first lactation, in populations of 2 dairy cow breeds: French Holstein and Montbéliarde. The different ART studied included the type of semen (conventional or X-sorted) used for Artificial Insemination (AI) and the technology of conception used (AI, embryo transfer, or in vitro fertilization). Three maternal characteristics were considered: (1) the dam age at first calving, (2) dam parity number, and (3) indicators of dam udder health during gestation (somatic cell score and events of clinical mastitis). First, we investigated whether heifer survival from 3d to 18 mo old was associated with any of the prenatal factors considered. We then estimated the associations of these prenatal factors with 8 traits of commercial interest: (1) stature, (2-4) milk, fat, and protein yields, (5) somatic cell score, (6) clinical mastitis, and (7-8) heifer and cow conception rate, all measured on genotyped cows. Linear models were used for this study with the prenatal factors as covariates in the model, and for the 8 traits, phenotypes were adjusted for their corresponding genomic estimated breeding value. The results indicated that the survival rate of heifers born from embryo transfer was significantly higher than that of heifers born from AI (probably due to preferential management practices), while the other prenatal factors did not explain differences in heifer survival. Among the Montbéliarde cows born from AI, those born from X-sorted semen showed a lightly but significantly lower milk yield than those born without X-sorting of the semen (-52 kg of milk in the first lactation). Among the Holstein cows, those born from embryo transfer presented significantly lower milk performance than cows born from AI. Regarding the maternal characteristics, none or very weak associations were found between the dam age at first calving and the offspring performance in both breeds. Dam parity, on the other hand, was associated with offspring performance for milk, fat, and protein yield in both breeds, however not in the same direction. In the Holstein breed, an increase in dam parity was favorable for offspring performance for milk, fat, and protein yield, whereas in the Montbéliarde breed, an increase in dam parity was associated with lower milk and protein yield and no association was found for fat yield. The udder health of the dam during gestation was not or only weakly associated with the traits studied in the offspring. Although some significant associations were identified due to the large sample size, the effects were modest, typically less than 1% of the phenotypic mean, and were not consistently observed across the 2 breeds.

12.
Dev Psychobiol ; 66(5): e22491, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38698633

RESUMEN

Developmental plasticity is particularly important for humans and other primates because of our extended period of growth and maturation, during which our phenotypes adaptively respond to environmental cues. The hypothalamus-pituitary-gonadal (HPG) and hypothalamus-pituitary-adrenal (HPA) axes are likely to be principal targets of developmental "programming" given their roles in coordinating fitness-relevant aspects of the phenotype, including sexual development, adult reproductive and social strategies, and internal responses to the external environment. In social animals, including humans, the social environment is believed to be an important source of cues to which these axes may adaptively respond. The effects of early social environments on the HPA axis have been widely studied in humans, and to some extent, in other primates, but there are still major gaps in knowledge specifically relating to males. There has also been relatively little research examining the role that social environments play in developmental programming of the HPG axis or the HPA/HPG interface, and what does exist disproportionately focuses on females. These topics are likely understudied in males in part due to the difficulty of identifying developmental milestones in males relative to females and the general quiescence of the HPG axis prior to maturation. However, there are clear indicators that early life social environments matter for both sexes. In this review, we examine what is known about the impact of social environments on HPG and HPA axis programming during male development in humans and nonhuman primates, including the role that epigenetic mechanisms may play in this programming. We conclude by highlighting important next steps in this research area.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Primates , Medio Social , Animales , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiología , Masculino , Primates/fisiología , Humanos , Femenino
13.
Int J Mol Sci ; 25(13)2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-39000422

RESUMEN

Maternal obesity, caused by diets rich in fats and sugars during pregnancy, can predispose offspring to metabolic diseases such as diabetes. We hypothesized that obesity during pregnancy leads to increased DNA methylation and reduced protein expression in factors regulating ß-cell function and apoptosis. Female C57BL/6J mice were fed a high-fat diet (HFD; 42% fat content; n = 3) or a control diet (CON; 16% fat content; n = 3) for fourteen weeks before and during pregnancy. Offspring were euthanized at 8 weeks and pancreatic tissue was collected. Isolated DNA was analyzed using whole-genome bisulfite sequencing. Protein expression was quantified using LC-MS. No significant differences in body weight were observed between HFD and control pups (p = 0.10). Whole-genome bisulfite sequencing identified 91,703 and 88,415 differentially methylated regions (DMRs) in CON vs. HFD male and female offspring. A total of 34 and 4 proteins were determined to have changes in expression that correlated with changes in DNA methylation in CON vs. HFD males and females, respectively. The majority of these factors were grouped into the metabolic function category via pathway analyses. This study illustrates the complex relationship between epigenetics, diet, and sex-specific responses, therefore offering insights into potential therapeutic targets and areas for further research.


Asunto(s)
Metilación de ADN , Dieta Alta en Grasa , Ratones Endogámicos C57BL , Páncreas , Animales , Femenino , Dieta Alta en Grasa/efectos adversos , Embarazo , Ratones , Masculino , Páncreas/metabolismo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Obesidad/metabolismo , Obesidad/genética , Obesidad/etiología , Epigénesis Genética , Multiómica
14.
Am J Physiol Lung Cell Mol Physiol ; 325(1): L66-L73, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37280517

RESUMEN

Children born to obese mothers are prone to develop asthma and airway hyperresponsiveness, but the mechanisms behind this are unclear. Here we developed a mouse model of maternal diet-induced obesity that recapitulates metabolic abnormalities seen in humans born to obese mothers. Offspring of dams fed a high-fat diet (HFD) showed increased adiposity, hyperinsulinemia, and insulin resistance at 16 wk of age despite being fed only a regular diet (RD). Bronchoconstriction induced by inhaled 5-hydroxytriptamine was also significantly increased in offspring of HFD-fed versus RD-fed dams. Increased bronchoconstriction was blocked by vagotomy, indicating this reflex was mediated by airway nerves. Three-dimensional (3-D) confocal imaging of tracheas collected from 16-wk-old offspring showed that both epithelial sensory innervation and substance P expression were increased in the offspring of HFD-fed dams compared with offspring of RD-fed dams. For the first time, we show that maternal high-fat diet increases airway sensory innervation in offspring, leading to reflex airway hyperresponsiveness.NEW & NOTEWORTHY Our study reveals a novel potential mechanism, by which maternal high-fat diet increases the risk and severity of asthma in offspring. We found that exposure to maternal high-fat diet in mice leads to hyperinnervation of airway sensory nerves and increased reflex bronchoconstriction in offspring fed a regular diet only. These findings have important clinical implications and provide new insights into the pathophysiology of asthma, highlighting the need for preventive strategies in this patient population.


Asunto(s)
Asma , Efectos Tardíos de la Exposición Prenatal , Hipersensibilidad Respiratoria , Humanos , Femenino , Niño , Animales , Ratones , Dieta Alta en Grasa/efectos adversos , Hijos Adultos , Broncoconstricción , Obesidad , Reflejo , Asma/etiología
15.
Am J Physiol Endocrinol Metab ; 324(6): E577-E588, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37134140

RESUMEN

Maternal overnutrition is associated with increased susceptibility to type 2 diabetes in the offspring. Rodent models have shown that maternal overnutrition influences islet function in offspring. To determine whether maternal Western-style diet (WSD) alters prejuvenile islet function in a model that approximates that of human offspring, we utilized a well-characterized Japanese macaque model. We compared islet function from offspring exposed to WSD throughout pregnancy and lactation and weaned to WSD (WSD/WSD) compared with islets from offspring exposed only to postweaning WSD (CD/WSD) at 1 yr of age. WSD/WSD offspring islets showed increased basal insulin secretion and an exaggerated increase in glucose-stimulated insulin secretion, as assessed by dynamic ex vivo perifusion assays, relative to CD/WSD-exposed offspring. We probed potential mechanisms underlying insulin hypersecretion using transmission electron microscopy to evaluate ß-cell ultrastructure, qRT-PCR to quantify candidate gene expression, and Seahorse assay to assess mitochondrial function. Insulin granule density, mitochondrial density, and mitochondrial DNA ratio were similar between groups. However, islets from WSD/WSD male and female offspring had increased expression of transcripts known to facilitate stimulus-secretion coupling and changes in the expression of cell stress genes. Seahorse assay revealed increased spare respiratory capacity in islets from WSD/WSD male offspring. Overall, these results show that maternal WSD feeding confers changes to genes governing insulin secretory coupling and results in insulin hypersecretion as early as the postweaning period. The results suggest a maternal diet leads to early adaptation and developmental programming in offspring islet genes that may underlie future ß-cell dysfunction.NEW & NOTEWORTHY Programed adaptations in islets in response to maternal WSD exposure may alter ß-cell response to metabolic stress in offspring. We show that islets from maternal WSD-exposed offspring hypersecrete insulin, possibly due to increased components of stimulus-secretion coupling. These findings suggest that islet hyperfunction is programed by maternal diet, and changes can be detected as early as the postweaning period in nonhuman primate offspring.


Asunto(s)
Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , Embarazo , Animales , Masculino , Femenino , Humanos , Insulina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Occidental/efectos adversos , Primates/metabolismo , Expresión Génica , Islotes Pancreáticos/metabolismo
16.
Am J Physiol Gastrointest Liver Physiol ; 325(6): G528-G538, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37724979

RESUMEN

Perinatal exposure to selective serotonin reuptake inhibitors (SSRIs) has been shown to disrupt the development of serotonergic signaling pathways in the brain and enteric nervous system. Serotonin (5-hydroxytryptamine; 5-HT) signaling is critical for gastrointestinal homeostasis; changes in 5-HT expression and regulation have been associated with gastrointestinal diseases of motility and inflammation. We tested the hypothesis that perinatal exposure to the SSRI fluoxetine can influence the development of the gastrointestinal tract in exposed offspring. Female nulliparous Wistar rats were given fluoxetine (10 mg/kg) or vehicle control from 2 wk before mating until weaning; small and large intestines of female and male offspring were collected at postnatal days 1, 21 (P1, P21, respectively), and 6 mo of age. In histological preparations, the proportion of serotonergic neurons significantly increased in the colons of both female and male fluoxetine-exposed compared with control offspring at P21, a time point that signifies maximal exposure to fluoxetine. At 6 mo of age, male but not female fluoxetine-exposed offspring had a significant increase in circulating 5-HT, with a significant decrease in transcripts encoding the 5-HT2A receptor and monoamine oxidase as compared with control offspring. Measurement of spatiotemporal mapping of contractile activity of the small and large intestine at 6 mo of age revealed no changes in motility in the small bowel of fluoxetine-exposed offspring but revealed a significant increase in the frequency of colonic contractions in the female fluoxetine-exposed compared with control animals. Susceptibility to inflammation was examined at 6 mo using the dextran sulfate sodium model of acute colitis. In utero exposure to fluoxetine was not found to exacerbate colitis severity. These findings suggest that fluoxetine exposure during fetal and early postnatal development can lead to changes in serotonergic neurons at the peak of exposure with sex-specific changes in 5-HT signaling and colonic motility in adulthood.NEW & NOTEWORTHY There is increasing recognition of the relevance of in utero and early postnatal exposures in the developmental programming of the gastrointestinal tract. Perinatal exposure to selective serotonin reuptake inhibitors and antidepressant medications is of particular relevance as they are commonly prescribed during pregnancy, and serotonergic pathways play key roles during gastrointestinal development and in postnatal homeostasis. Here, we provide a comprehensive evaluation of clinically relevant outcomes of gastrointestinal motility and susceptibility to colitis in fluoxetine-exposed offspring and highlight changes in colonic serotonergic neurons at the peak of perinatal fluoxetine exposure with sex-dependent changes in serotonin signaling and colonic motility in adulthood.


Asunto(s)
Colitis , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Ratas , Animales , Masculino , Femenino , Fluoxetina/toxicidad , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Serotonina/metabolismo , Ratas Sprague-Dawley , Ratas Wistar , Efectos Tardíos de la Exposición Prenatal/metabolismo , Inflamación , Colitis/inducido químicamente
17.
FASEB J ; 36(12): e22644, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36415994

RESUMEN

Maternal obesity (MO) during pregnancy is linked to increased and premature risk of age-related metabolic diseases in the offspring. However, the underlying molecular mechanisms still remain not fully understood. Using a well-established nonhuman primate model of MO, we analyzed tissue biopsies and plasma samples obtained from post-pubertal offspring (3-6.5 y) of MO mothers (n = 19) and from control animals born to mothers fed a standard diet (CON, n = 13). All offspring ate a healthy chow diet after weaning. Using untargeted gas chromatography-mass spectrometry metabolomics analysis, we quantified a total of 351 liver, 316 skeletal muscle, and 423 plasma metabolites. We identified 58 metabolites significantly altered in the liver and 46 in the skeletal muscle of MO offspring, with 8 metabolites shared between both tissues. Several metabolites were changed in opposite directions in males and females in both liver and skeletal muscle. Several tissue-specific and 4 shared metabolic pathways were identified from these dysregulated metabolites. Interestingly, none of the tissue-specific metabolic changes were reflected in plasma. Overall, our study describes characteristic metabolic perturbations in the liver and skeletal muscle in MO offspring, indicating that metabolic programming in utero persists postnatally, and revealing potential novel mechanisms that may contribute to age-related metabolic diseases later in life.


Asunto(s)
Obesidad Materna , Humanos , Animales , Masculino , Femenino , Embarazo , Destete , Obesidad/metabolismo , Dieta , Músculo Esquelético/metabolismo , Hígado/metabolismo , Estilo de Vida , Pubertad
18.
J Exp Biol ; 226(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36714994

RESUMEN

The early-life environment is known to affect later-life health and disease, which could be mediated by the early-life programming of telomere length, a key hallmark of ageing. According to the fetal programming of telomere biology hypothesis, variation in prenatal exposure to hormones is likely to influence telomere length. Yet, the contribution of key metabolic hormones, i.e. thyroid hormones (THs), has been largely ignored. We recently showed that in contrast to predictions, exposure to elevated prenatal THs increased postnatal telomere length in wild collared flycatchers, but the generality of such effect, the underlying proximate mechanisms and consequences for survival have not been investigated. We therefore conducted a comprehensive study evaluating the impact of THs on potential drivers of telomere dynamics (growth, post-natal THs, mitochondria and oxidative stress), telomere length and medium-term survival using wild great tits as a model system. While prenatal THs did not significantly affect telomere length a week after hatching (i.e. day 7), they influenced postnatal telomere shortening (i.e. shorter telomeres at day 14 and the following winter) but not apparent survival. Circulating THs, mitochondrial density or oxidative stress biomarkers were not significantly influenced, whereas the TH-supplemented group showed accelerated growth, which may explain the observed delayed effect on telomeres. We discuss several alternative hypotheses that may explain the contrast with our previous findings in flycatchers. Given that shorter telomeres in early life tend to be carried until adulthood and are often associated with decreased survival prospects, the effects of prenatal THs on telomeres may have long-lasting effects on senescence.


Asunto(s)
Passeriformes , Pájaros Cantores , Embarazo , Animales , Femenino , Acortamiento del Telómero , Envejecimiento , Desarrollo Fetal , Vitaminas , Telómero , Hormonas Tiroideas , Hormonas
19.
Oecologia ; 201(3): 637-648, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36894790

RESUMEN

Understanding the consequences of heat exposure on mitochondrial function is crucial as mitochondria lie at the core of metabolic processes, also affecting population dynamics. In adults, mitochondrial metabolism varies with temperature but can also depend on thermal conditions experienced during development. We exposed zebra finches to two alternative heat treatments during early development: "constant", maintained birds at ambient 35 °C from parental pair formation to fledglings' independence, while "periodic" heated broods at 40 °C, 6 h daily at nestling stage. Two years later, we acclimated birds from both experiments at 25 °C for 21 days, before exposing them to artificial heat (40 °C, 5 h daily for 10 days). After both conditions, we measured red blood cells' mitochondrial metabolism using a high-resolution respirometer. We found significantly decreased mitochondrial metabolism for Routine, Oxidative Phosphorylation (OxPhos) and Electron Transport System maximum capacity (ETS) after the heat treatments. In addition, the birds exposed to "constant" heat in early life showed lower oxygen consumption at the Proton Leak (Leak) stage after the heat treatment as adults. Females showed higher mitochondrial respiration for Routine, ETS and Leak independent of the treatments, while this pattern was reversed for OxPhos coupling efficiency (OxCE). Our results show that short-term acclimation involved reduced mitochondrial respiration, and that the reaction of adult birds to heat depends on the intensity, pattern and duration of temperature conditions experienced at early-life stages. Our study provides insight into the complexity underlying variation in mitochondrial metabolism and raises questions on the adaptive value of long-lasting physiological adjustments triggered by the early-life thermal environment.


Asunto(s)
Pinzones , Calor , Animales , Femenino , Mitocondrias/metabolismo , Temperatura , Aclimatación/fisiología , Pinzones/fisiología
20.
Curr Osteoporos Rep ; 21(4): 433-446, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37335525

RESUMEN

PURPOSE OF REVIEW: Increasing bone mineral accrual during childhood might delay the onset of osteoporosis. We discuss the scientific evidence for early life approaches to optimising skeletal health. RECENT FINDINGS: There is an ever-growing body of evidence from observational studies suggesting associations between early life exposures, particularly during foetal development, and bone mineral density (BMD). The findings of such studies are often heterogeneous, and for some exposures, for example, maternal smoking and alcohol intake in pregnancy or age at conception, intervention studies are not feasible. The most frequently studied exposures in intervention studies are calcium or vitamin D supplementation in pregnancy, which overall suggest positive effects on offspring childhood BMD. Maternal calcium and/or vitamin D supplementation during pregnancy appear to have positive effects on offspring BMD during early childhood, but further long-term follow-up is required to demonstrate persistence of the effect into later life.


Asunto(s)
Osteoporosis , Vitamina D , Embarazo , Femenino , Humanos , Preescolar , Vitamina D/uso terapéutico , Calcio , Densidad Ósea , Calcio de la Dieta , Suplementos Dietéticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA