Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.347
Filter
Add more filters

Coleção CLAP
Publication year range
1.
Cell ; 166(5): 1247-1256.e4, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565347

ABSTRACT

Zika virus (ZIKV) can be transmitted sexually between humans. However, it is unknown whether ZIKV replicates in the vagina and impacts the unborn fetus. Here, we establish a mouse model of vaginal ZIKV infection and demonstrate that, unlike other routes, ZIKV replicates within the genital mucosa even in wild-type (WT) mice. Mice lacking RNA sensors or transcription factors IRF3 and IRF7 resulted in higher levels of local viral replication. Furthermore, mice lacking the type I interferon (IFN) receptor (IFNAR) became viremic and died of infection after a high-dose vaginal ZIKV challenge. Notably, vaginal infection of pregnant dams during early pregnancy led to fetal growth restriction and infection of the fetal brain in WT mice. This was exacerbated in mice deficient in IFN pathways, leading to abortion. Our study highlights the vaginal tract as a highly susceptible site of ZIKV replication and illustrates the dire disease consequences during pregnancy.


Subject(s)
Brain Diseases/virology , Brain/virology , Fetal Growth Retardation/virology , Pregnancy Complications, Infectious/virology , Vagina/virology , Virus Replication , Zika Virus Infection/transmission , Zika Virus/physiology , Abortion, Habitual/virology , Animals , Brain Diseases/immunology , Disease Models, Animal , Female , Fetal Growth Retardation/immunology , Interferon Regulatory Factor-3/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pregnancy , Pregnancy Complications, Infectious/immunology , Receptor, Interferon alpha-beta/genetics
2.
Cell ; 165(3): 566-79, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27087445

ABSTRACT

Hepatic glucose release into the circulation is vital for brain function and survival during periods of fasting and is modulated by an array of hormones that precisely regulate plasma glucose levels. We have identified a fasting-induced protein hormone that modulates hepatic glucose release. It is the C-terminal cleavage product of profibrillin, and we name it Asprosin. Asprosin is secreted by white adipose, circulates at nanomolar levels, and is recruited to the liver, where it activates the G protein-cAMP-PKA pathway, resulting in rapid glucose release into the circulation. Humans and mice with insulin resistance show pathologically elevated plasma asprosin, and its loss of function via immunologic or genetic means has a profound glucose- and insulin-lowering effect secondary to reduced hepatic glucose release. Asprosin represents a glucogenic protein hormone, and therapeutically targeting it may be beneficial in type II diabetes and metabolic syndrome.


Subject(s)
Fasting/metabolism , Microfilament Proteins/metabolism , Peptide Fragments/metabolism , Peptide Hormones/metabolism , Adipose Tissue, White/metabolism , Amino Acid Sequence , Animals , Antibodies/administration & dosage , Circadian Rhythm , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Fasting/blood , Female , Fetal Growth Retardation/metabolism , Fibrillin-1 , Glucose/metabolism , Humans , Insulin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microfilament Proteins/blood , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Molecular Sequence Data , Peptide Fragments/blood , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Hormones/blood , Peptide Hormones/chemistry , Peptide Hormones/genetics , Progeria/metabolism , Recombinant Proteins/administration & dosage , Sequence Alignment
3.
Cell ; 155(1): 81-93, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24074862

ABSTRACT

The importance of maternal folate consumption for normal development is well established, yet the molecular mechanism linking folate metabolism to development remains poorly understood. The enzyme methionine synthase reductase (Mtrr) is necessary for utilization of methyl groups from the folate cycle. We found that a hypomorphic mutation of the mouse Mtrr gene results in intrauterine growth restriction, developmental delay, and congenital malformations, including neural tube, heart, and placental defects. Importantly, these defects were dependent upon the Mtrr genotypes of the maternal grandparents. Furthermore, we observed widespread epigenetic instability associated with altered gene expression in the placentas of wild-type grandprogeny of Mtrr-deficient maternal grandparents. Embryo transfer experiments revealed that Mtrr deficiency in mice lead to two distinct, separable phenotypes: adverse effects on their wild-type daughters' uterine environment, leading to growth defects in wild-type grandprogeny, and the appearance of congenital malformations independent of maternal environment that persist for five generations, likely through transgenerational epigenetic inheritance.


Subject(s)
Congenital Abnormalities/genetics , Embryo, Mammalian/metabolism , Epigenesis, Genetic , Ferredoxin-NADP Reductase/genetics , Fetal Growth Retardation/genetics , Folic Acid/metabolism , Animals , Crosses, Genetic , DNA Methylation , Female , Ferredoxin-NADP Reductase/metabolism , Male , Mice , Mutation
4.
Genes Dev ; 34(21-22): 1520-1533, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33060134

ABSTRACT

DNA replication is fundamental for cell proliferation in all organisms. Nonetheless, components of the replisome have been implicated in human disease, and here we report PRIM1 encoding the catalytic subunit of DNA primase as a novel disease gene. Using a variant classification agnostic approach, biallelic mutations in PRIM1 were identified in five individuals. PRIM1 protein levels were markedly reduced in patient cells, accompanied by replication fork asymmetry, increased interorigin distances, replication stress, and prolonged S-phase duration. Consequently, cell proliferation was markedly impaired, explaining the patients' extreme growth failure. Notably, phenotypic features distinct from those previously reported with DNA polymerase genes were evident, highlighting differing developmental requirements for this core replisome component that warrant future investigation.


Subject(s)
DNA Primase/genetics , Dwarfism/genetics , Fetal Growth Retardation/genetics , DNA Primase/chemistry , DNA Primase/deficiency , Dwarfism/diagnostic imaging , Dwarfism/pathology , Female , Fetal Growth Retardation/diagnostic imaging , Fetal Growth Retardation/pathology , Genetic Variation , Humans , Infant , Male , Pedigree , Syndrome
5.
Proc Natl Acad Sci U S A ; 121(44): e2409597121, 2024 Oct 29.
Article in English | MEDLINE | ID: mdl-39432779

ABSTRACT

Epidemiological studies show a strong correlation between air pollution and fetal growth restriction (FGR), but existing results are controversial due to inherent limitations, such as causality of specific pollutants, developmental origin, and maternal-fetal transmission. To address this controversy, we first conducted a retrospective analysis of 28,796 newborns and revealed that maternal nitrogen dioxide (NO2) exposure during the second trimester was positively associated with FGR, with an adjusted odds ratio of 1.075 (95% confidence interval: 1.020-1.133) per 10 µg/m3 NO2 increase for small for gestational age. Then, by establishing an animal model of prenatal NO2 exposure, we confirmed its adverse effects on embryonic growth and hematopoiesis in the yolk sac and fetal liver, primarily affecting the differentiation of hematopoietic stem and progenitor cells and erythroid maturation. By applying internal exposure analyses coupled with 15N isotope tracing, we found that maternal NO2 inhalation induced acquired methemoglobinemia through its byproducts and placental hypoxia in pregnant mice. Importantly, by combining transcriptional profiling, bioinformatics analysis, and RNA binding protein immunoprecipitation (RIP)/chromatin immunoprecipitation (CHIP), we clarified that placental-fetal hypoxia transmission activated hypoxia-inducible factors, disturbed hematopoiesis through the hypoxia-inducible factor 1ß-long noncoding RNAs-CCAAT/enhancer binding protein alpha-proinflammatory signaling pathway, ultimately contributing to FGR progression. These findings provide insights for risk prevention and clinical intervention to promote child well-being in NO2-polluted areas.


Subject(s)
Fetal Growth Retardation , Hematopoiesis , Maternal Exposure , Nitrogen Dioxide , RNA, Long Noncoding , Pregnancy , Female , Animals , Fetal Growth Retardation/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mice , Hematopoiesis/genetics , Humans , Nitrogen Dioxide/adverse effects , Nitrogen Dioxide/toxicity , Maternal Exposure/adverse effects , Inflammation/metabolism , Hypoxia , Infant, Newborn , Retrospective Studies , Male
6.
Development ; 150(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37831056

ABSTRACT

Compelling epidemiological and animal experimental data demonstrate that cardiometabolic and neuropsychiatric diseases originate in a suboptimal intrauterine environment. Here, we review evidence suggesting that altered placental function may, at least in part, mediate the link between the maternal environment and changes in fetal growth and development. Emerging evidence indicates that the placenta controls the development and function of several fetal tissues through nutrient sensing, modulation of trophoblast nutrient transporters and by altering the number and cargo of released extracellular vesicles. In this Review, we discuss the development and functions of the maternal-placental-fetal interface (in humans and mice) and how cross-talk between these compartments may be a mechanism for in utero programming, focusing on mechanistic target of rapamycin (mTOR), adiponectin and O-GlcNac transferase (OGT) signaling. We also discuss how maternal diet and stress influences fetal development and metabolism and how fetal growth restriction can result in susceptibility to developing chronic disease later in life. Finally, we speculate how interventions targeting placental function may offer unprecedented opportunities to prevent cardiometabolic disease in future generations.


Subject(s)
Fetal Development , Placenta , Pregnancy , Female , Humans , Mice , Animals , Placenta/metabolism , Trophoblasts/metabolism , Signal Transduction , Fetal Growth Retardation
7.
Nat Immunol ; 15(9): 839-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25064072

ABSTRACT

Sensors of the innate immune system that detect intracellular nucleic acids must be regulated to prevent inappropriate activation by endogenous DNA and RNA. The exonuclease Trex1 regulates the DNA-sensing pathway by metabolizing potential DNA ligands that trigger it. However, an analogous mechanism for regulating the RIG-I-like receptors (RLRs) that detect RNA remains unknown. We found here that the SKIV2L RNA exosome potently limited the activation of RLRs. The unfolded protein response (UPR), which generated endogenous RLR ligands through the cleavage of cellular RNA by the endonuclease IRE-1, triggered the production of type I interferons in cells depleted of SKIV2L. Humans with deficiency in SKIV2L had a type I interferon signature in their peripheral blood. Our findings reveal a mechanism for the intracellular metabolism of immunostimulatory RNA, with implications for specific autoimmune disorders.


Subject(s)
DEAD-box RNA Helicases/immunology , Diarrhea, Infantile/immunology , Endoribonucleases/immunology , Exosome Multienzyme Ribonuclease Complex , Fetal Growth Retardation/immunology , Hair Diseases/immunology , Immunity, Innate/immunology , Nuclear Proteins/immunology , Protein Serine-Threonine Kinases/immunology , RNA Helicases/immunology , RNA-Binding Proteins/immunology , Unfolded Protein Response/immunology , Animals , DEAD Box Protein 58 , Facies , Gene Knockdown Techniques , Humans , Interferon Type I/immunology , Mice, Inbred C57BL , Proteins/immunology
8.
Immunity ; 47(6): 1100-1113.e6, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29262349

ABSTRACT

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


Subject(s)
Abortion, Habitual/immunology , Adoptive Transfer , Carrier Proteins/metabolism , Cytokines/metabolism , Fetal Development/immunology , Fetal Growth Retardation/prevention & control , Intercellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/transplantation , Abortion, Habitual/genetics , Abortion, Habitual/pathology , Adult , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Cellular Microenvironment , Cytokines/genetics , Cytokines/immunology , Decidua/immunology , Decidua/pathology , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/immunology , Fetal Growth Retardation/pathology , Fetus , Gene Expression Regulation, Developmental , HLA-G Antigens/genetics , HLA-G Antigens/immunology , Humans , Integrin alpha1/genetics , Integrin alpha1/immunology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Leukocyte Immunoglobulin-like Receptor B1/genetics , Leukocyte Immunoglobulin-like Receptor B1/immunology , Mice , Mice, Inbred C57BL , Pregnancy , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology
9.
Proc Natl Acad Sci U S A ; 120(9): e2102569120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36802443

ABSTRACT

In the human genome, about 750 genes contain one intron excised by the minor spliceosome. This spliceosome comprises its own set of snRNAs, among which U4atac. Its noncoding gene, RNU4ATAC, has been found mutated in Taybi-Linder (TALS/microcephalic osteodysplastic primordial dwarfism type 1), Roifman (RFMN), and Lowry-Wood (LWS) syndromes. These rare developmental disorders, whose physiopathological mechanisms remain unsolved, associate ante- and post-natal growth retardation, microcephaly, skeletal dysplasia, intellectual disability, retinal dystrophy, and immunodeficiency. Here, we report bi-allelic RNU4ATAC mutations in five patients presenting with traits suggestive of the Joubert syndrome (JBTS), a well-characterized ciliopathy. These patients also present with traits typical of TALS/RFMN/LWS, thus widening the clinical spectrum of RNU4ATAC-associated disorders and indicating ciliary dysfunction as a mechanism downstream of minor splicing defects. Intriguingly, all five patients carry the n.16G>A mutation, in the Stem II domain, either at the homozygous or compound heterozygous state. A gene ontology term enrichment analysis on minor intron-containing genes reveals that the cilium assembly process is over-represented, with no less than 86 cilium-related genes containing at least one minor intron, among which there are 23 ciliopathy-related genes. The link between RNU4ATAC mutations and ciliopathy traits is supported by alterations of primary cilium function in TALS and JBTS-like patient fibroblasts, as well as by u4atac zebrafish model, which exhibits ciliopathy-related phenotypes and ciliary defects. These phenotypes could be rescued by WT but not by pathogenic variants-carrying human U4atac. Altogether, our data indicate that alteration of cilium biogenesis is part of the physiopathological mechanisms of TALS/RFMN/LWS, secondarily to defects of minor intron splicing.


Subject(s)
Ciliopathies , Spliceosomes , Female , Animals , Humans , Spliceosomes/genetics , RNA, Small Nuclear/genetics , Zebrafish/genetics , Fetal Growth Retardation/genetics , Mutation , Ciliopathies/genetics
10.
N Engl J Med ; 386(19): 1781-1792, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35363951

ABSTRACT

BACKGROUND: The benefits and safety of the treatment of mild chronic hypertension (blood pressure, <160/100 mm Hg) during pregnancy are uncertain. Data are needed on whether a strategy of targeting a blood pressure of less than 140/90 mm Hg reduces the incidence of adverse pregnancy outcomes without compromising fetal growth. METHODS: In this open-label, multicenter, randomized trial, we assigned pregnant women with mild chronic hypertension and singleton fetuses at a gestational age of less than 23 weeks to receive antihypertensive medications recommended for use in pregnancy (active-treatment group) or to receive no such treatment unless severe hypertension (systolic pressure, ≥160 mm Hg; or diastolic pressure, ≥105 mm Hg) developed (control group). The primary outcome was a composite of preeclampsia with severe features, medically indicated preterm birth at less than 35 weeks' gestation, placental abruption, or fetal or neonatal death. The safety outcome was small-for-gestational-age birth weight below the 10th percentile for gestational age. Secondary outcomes included composites of serious neonatal or maternal complications, preeclampsia, and preterm birth. RESULTS: A total of 2408 women were enrolled in the trial. The incidence of a primary-outcome event was lower in the active-treatment group than in the control group (30.2% vs. 37.0%), for an adjusted risk ratio of 0.82 (95% confidence interval [CI], 0.74 to 0.92; P<0.001). The percentage of small-for-gestational-age birth weights below the 10th percentile was 11.2% in the active-treatment group and 10.4% in the control group (adjusted risk ratio, 1.04; 95% CI, 0.82 to 1.31; P = 0.76). The incidence of serious maternal complications was 2.1% and 2.8%, respectively (risk ratio, 0.75; 95% CI, 0.45 to 1.26), and the incidence of severe neonatal complications was 2.0% and 2.6% (risk ratio, 0.77; 95% CI, 0.45 to 1.30). The incidence of any preeclampsia in the two groups was 24.4% and 31.1%, respectively (risk ratio, 0.79; 95% CI, 0.69 to 0.89), and the incidence of preterm birth was 27.5% and 31.4% (risk ratio, 0.87; 95% CI, 0.77 to 0.99). CONCLUSIONS: In pregnant women with mild chronic hypertension, a strategy of targeting a blood pressure of less than 140/90 mm Hg was associated with better pregnancy outcomes than a strategy of reserving treatment only for severe hypertension, with no increase in the risk of small-for-gestational-age birth weight. (Funded by the National Heart, Lung, and Blood Institute; CHAP ClinicalTrials.gov number, NCT02299414.).


Subject(s)
Antihypertensive Agents/therapeutic use , Hypertension, Pregnancy-Induced/drug therapy , Hypertension , Pregnancy Outcome , Abruptio Placentae/epidemiology , Abruptio Placentae/prevention & control , Birth Weight , Chronic Disease , Female , Fetal Growth Retardation/epidemiology , Fetal Growth Retardation/prevention & control , Humans , Hypertension/complications , Hypertension/drug therapy , Infant, Newborn , Pre-Eclampsia/epidemiology , Pre-Eclampsia/prevention & control , Pregnancy , Pregnancy Outcome/epidemiology , Premature Birth/epidemiology , Premature Birth/prevention & control
11.
Development ; 149(19): dev200717, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36193846

ABSTRACT

Placentas from pregnancies complicated by severe early-onset fetal growth restriction (FGR) exhibit diminished vascular development mediated by impaired angiogenesis, but underlying mechanisms remain unknown. In this study, we show that FGR endothelial cells demonstrate inherently reduced migratory capacity despite the presence of fibronectin, a matrix protein abundant in placental stroma that displays abnormal organization in FGR placentas. Thus, we hypothesized that aberrant endothelial-fibronectin interactions in FGR are a key mechanism underlying impaired FGR endothelial migration. Using human fetoplacental endothelial cells isolated from uncomplicated term control and FGR pregnancies, we assessed integrin α5ß1 and αvß3 regulation during cell migration. We show that endothelial integrin α5ß1 and αvß3 interactions with fibronectin are required for migration and that FGR endothelial cells responded differentially to integrin inhibition, indicating integrin dysregulation in FGR. Whole-cell expression was not different between groups. However, there were significantly more integrins in focal adhesions and reduced intracellular trafficking in FGR. These newly identified changes in FGR endothelial cellular processes represent previously unidentified mechanisms contributing to persistent angiogenic deficiencies in FGR.


Subject(s)
Fetal Growth Retardation , Integrin alphaVbeta3 , Endothelial Cells/metabolism , Female , Fibronectins/genetics , Fibronectins/metabolism , Humans , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Placenta/metabolism , Pregnancy
12.
Am J Pathol ; 194(9): 1636-1663, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39182948

ABSTRACT

Air pollution exposure during pregnancy may affect fetal growth. Fetal growth restriction (FGR) is associated with reduced lung function in children that can persist into adulthood. Using an established model of asymmetrical FGR in Long-Evans rats, this study investigated sex differences in effects of early life ozone exposure on lung development and maturation. Adverse health effects for i) gestational exposure (with impacts on primary alveolarization), ii) peri-adolescent exposure (with impacts on secondary alveolarization), and iii) cumulative exposure across both periods were evaluated. Notably, female offspring were most affected by gestational ozone exposure, likely because of impaired angiogenesis and corresponding decreases in primary alveolarization. Females had diminished lung capacity, fewer mature alveoli, and medial hypertrophy of small and large pulmonary arteries. Males, especially FGR-prone offspring, were more affected by peri-adolescent ozone exposure. Males had increased ductal areas, likely due to disrupted secondary alveolarization. Altered lung development may increase risk of developing diseases, such as pulmonary arterial hypertension or chronic obstructive pulmonary disease. Pulmonary arterial hypertension disproportionately affects women. In the United States, chronic obstructive pulmonary disease prevalence is increasing, especially in women; and prevalence for both men and women is highest in urbanized areas. This investigation underlines the importance of evaluating results separately by sex, and provides biologic plausibility for later consequences of early-life exposure to ozone, a ubiquitous urban air pollutant.


Subject(s)
Lung , Ozone , Prenatal Exposure Delayed Effects , Rats, Long-Evans , Animals , Ozone/toxicity , Ozone/adverse effects , Female , Male , Pregnancy , Lung/drug effects , Lung/pathology , Lung/growth & development , Rats , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Sex Characteristics , Humans , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/pathology , Air Pollutants/toxicity , Air Pollutants/adverse effects
13.
FASEB J ; 38(19): e70100, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39387608

ABSTRACT

Maternal obesity in pregnancy is strongly associated with complications such as fetal overgrowth and infants of obese mothers have an increased risk to develop obesity, diabetes, and cardiovascular disease later in life. However, the underlying mechanisms are not well established. Circulating levels of adiponectin are low in obese pregnant women and maternal circulating adiponectin is negatively associated with birth weight. We have reported that normalizing maternal adiponectin in obese pregnant mice prevents placental dysfunction, fetal overgrowth, and programming of offspring cardio-metabolic disease. However, the mechanistic link between maternal adiponectin, placental function, and fetal growth remains to be established. We hypothesized that trophoblast-specific overexpression of the adiponectin receptor 2 (Adipor2) in healthy pregnant mice inhibits placental mTORC1 signaling and nutrient transport, resulting in fetal growth restriction. Using lentiviral transduction of blastocysts with a mammalian gene expression lentiviral vector for up-regulation of Adipor2 (Adipor2-OX), we achieved a ~ 3-fold increase in placenta Adipor2 mRNA levels and a 2-fold increase of the ADIPOR2 protein in the trophoblast plasma membrane. Placenta-specific Adipor2-OX increased placental peroxisome proliferator-activated receptor-α phosphorylation, ceramide synthase expression and ceramide concentrations. Furthermore, Adipor2-OX inhibited placental mTORC1 signaling and reduced in vivo placental transport of glucose and amino acids. Lastly, Adipor2-OX reduced fetal weight by 11%. These data provide mechanistic evidence that placental Adipor2 signaling directly affects fetal growth. We propose that low circulating adiponectin in maternal obesity causes fetal overgrowth and programs the offspring for cardio-metabolic disease mediated by a direct effect on placental function.


Subject(s)
Fetal Growth Retardation , Receptors, Adiponectin , Trophoblasts , Animals , Pregnancy , Female , Receptors, Adiponectin/metabolism , Receptors, Adiponectin/genetics , Fetal Growth Retardation/metabolism , Mice , Trophoblasts/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Placenta/metabolism , Adiponectin/metabolism , Adiponectin/genetics , Signal Transduction , Mice, Inbred C57BL
14.
FASEB J ; 38(7): e23598, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38581244

ABSTRACT

The precise molecular mechanism behind fetal growth restriction (FGR) is still unclear, although there is a strong connection between placental dysfunction, inadequate trophoblast invasion, and its etiology and pathogenesis. As a new type of non-coding RNA, circRNA has been shown to play a crucial role in the development of FGR. This investigation identified the downregulation of hsa_circ_0034533 (circTHBS1) in FGR placentas through high-sequencing analysis and confirmed this finding in 25 clinical placenta samples using qRT-PCR. Subsequent in vitro functional assays demonstrated that silencing circTHBS1 inhibited trophoblast proliferation, migration, invasion, and epithelial mesenchymal transition (EMT) progression and promoted apoptosis. Furthermore, when circTHBS1 was overexpressed, cell function experiments showed the opposite result. Analysis using fluorescence in situ hybridization revealed that circTHBS1 was primarily found in the cytoplasmic region. Through bioinformatics analysis, we anticipated the involvement of miR-136-3p and IGF2R in downstream processes, which was subsequently validated through qRT-PCR and dual-luciferase assays. Moreover, the inhibition of miR-136-3p or the overexpression of IGF2R partially reinstated proliferation, migration, and invasion abilities following the silencing of circTHBS1. In summary, the circTHBS1/miR-136-3p/IGF2R axis plays a crucial role in the progression and development of FGR, offering potential avenues for the exploration of biological indicators and treatment targets.


Subject(s)
MicroRNAs , Female , Humans , Pregnancy , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Fetal Growth Retardation/metabolism , In Situ Hybridization, Fluorescence , MicroRNAs/genetics , MicroRNAs/metabolism , Placenta/metabolism , Trophoblasts/metabolism
15.
Cereb Cortex ; 34(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38879758

ABSTRACT

Placental-related fetal growth restriction, resulting from placental dysfunction, impacts 3-5% of pregnancies and is linked to elevated risk of adverse neurodevelopmental outcomes. In response, the fetus employs a mechanism known as brain-sparing, redirecting blood flow to the cerebral circuit, for adequate supply to the brain. In this study we aimed to quantitatively evaluate disparities in gyrification and brain volumes among fetal growth restriction, small for gestational age and appropriate-for gestational-age fetuses. Additionally, we compared fetal growth restriction fetuses with and without brain-sparing. The study encompassed 106 fetuses: 35 fetal growth restriction (14 with and 21 without brain-sparing), 8 small for gestational age, and 63 appropriate for gestational age. Gyrification, supratentorial, and infratentorial brain volumes were automatically computed from T2-weighted magnetic resonance images, following semi-automatic brain segmentation. Fetal growth restriction fetuses exhibited significantly reduced gyrification and brain volumes compared to appropriate for gestational age (P < 0.001). Small for gestational age fetuses displayed significantly reduced gyrification (P = 0.038) and smaller supratentorial volume (P < 0.001) compared to appropriate for gestational age. Moreover, fetal growth restriction fetuses with BS demonstrated reduced gyrification compared to those without BS (P = 0.04), with no significant differences observed in brain volumes. These findings demonstrate that brain development is affected in fetuses with fetal growth restriction, more severely than in small for gestational age, and support the concept that vasodilatation of the fetal middle cerebral artery reflects more severe hypoxemia, affecting brain development.


Subject(s)
Fetal Growth Retardation , Magnetic Resonance Imaging , Fetal Growth Retardation/diagnostic imaging , Fetal Growth Retardation/pathology , Humans , Female , Magnetic Resonance Imaging/methods , Pregnancy , Adult , Gestational Age , Brain/diagnostic imaging , Brain/growth & development , Brain/pathology , Male , Infant, Small for Gestational Age
16.
Cereb Cortex ; 34(1)2024 01 14.
Article in English | MEDLINE | ID: mdl-37885155

ABSTRACT

Normal cortical growth and the resulting folding patterns are crucial for normal brain function. Although cortical development is largely influenced by genetic factors, environmental factors in fetal life can modify the gene expression associated with brain development. As the placenta plays a vital role in shaping the fetal environment, affecting fetal growth through the exchange of oxygen and nutrients, placental oxygen transport might be one of the environmental factors that also affect early human cortical growth. In this study, we aimed to assess the placental oxygen transport during maternal hyperoxia and its impact on fetal brain development using MRI in identical twins to control for genetic and maternal factors. We enrolled 9 pregnant subjects with monochorionic diamniotic twins (30.03 ± 2.39 gestational weeks [mean ± SD]). We observed that the fetuses with slower placental oxygen delivery had reduced volumetric and surface growth of the cerebral cortex. Moreover, when the difference between placenta oxygen delivery increased between the twin pairs, sulcal folding patterns were more divergent. Thus, there is a significant relationship between placental oxygen transport and fetal brain cortical growth and folding in monochorionic twins.


Subject(s)
Placenta , Twins, Monozygotic , Female , Humans , Pregnancy , Fetal Development , Fetal Growth Retardation/metabolism , Oxygen/metabolism , Placenta/diagnostic imaging , Placenta/metabolism
17.
Cell Mol Life Sci ; 81(1): 329, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090270

ABSTRACT

Decidualisation of the endometrium is a key event in early pregnancy, which enables embryo implantation. Importantly, the molecular processes impairing decidualisation in obese mothers are yet to be characterised. We hypothesise that impaired decidualisation in obese mice is mediated by the upregulation of leptin modulators, the suppressor of cytokine signalling 3 (SOCS3) and the protein tyrosine phosphatase non-receptor type 2 (PTPN2), together with the disruption of progesterone (P4)-signal transducer and activator of transcription (STAT3) signalling. After feeding mice with chow diet (CD) or high-fat diet (HFD) for 16 weeks, we confirmed the downregulation of P4 and oestradiol (E2) steroid receptors in decidua from embryonic day (E) 6.5 and decreased proliferation of stromal cells from HFD. In vitro decidualised mouse endometrial stromal cells (MESCs) and E6.5 deciduas from the HFD showed decreased expression of decidualisation markers, followed by the upregulation of SOCS3 and PTPN2 and decreased phosphorylation of STAT3. In vivo and in vitro leptin treatment of mice and MESCs mimicked the results observed in the obese model. The downregulation of Socs3 and Ptpn2 after siRNA transfection of MESCs from HFD mice restored the expression level of decidualisation markers. Finally, DIO mice placentas from E18.5 showed decreased labyrinth development and vascularisation and fetal growth restricted embryos. The present study revealed major defects in decidualisation in obese mice, characterised by altered uterine response to E2 and P4 steroid signalling. Importantly, altered hormonal response was associated with increased expression of leptin signalling modulators SOCS3 and PTPN2. Elevated levels of SOCS3 and PTPN2 were shown to molecularly affect decidualisation in obese mice, potentially disrupting the STAT3-PR regulatory molecular hub.


Subject(s)
Decidua , Fetal Growth Retardation , Leptin , Placenta , Signal Transduction , Animals , Female , Mice , Pregnancy , Decidua/metabolism , Decidua/pathology , Diet, High-Fat/adverse effects , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Leptin/metabolism , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Obesity/pathology , Placenta/metabolism , Progesterone/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , STAT3 Transcription Factor/metabolism , Stromal Cells/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics
18.
Proteomics ; 24(7): e2300260, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38059784

ABSTRACT

Intrauterine growth restriction (IUGR) is associated with increased risk of cardiometabolic disease later in life and has been shown to affect female and male offspring differently, but the mechanisms remain unclear. The purpose of this study was to identify proteomic differences and metabolic risk markers in IUGR male and female neonates when compared to appropriate for gestational age (AGA) babies that will provide a better understanding of IUGR pathogenesis and its associated risks. Our results revealed alterations in IUGR cord plasma proteomes with most of the differentially abundant proteins implicated in peroxisome pathways. This effect was evident in females but not in males. Furthermore, we observed that catalase activity, a peroxisomal enzyme, was significantly increased in females (p < 0.05) but unchanged in males. Finally, we identified risk proteins associated with obesity, type-2 diabetes, and glucose intolerance such as EGF containing fibulin extracellular matrix protein 1 (EFEMP1), proprotein convertase subtilisin/kexin type 9 (PCSK9) and transforming growth factor beta receptor 3 (TGFBR3) proteins unique to females while coagulation factor IX (C9) and retinol binding protein 4 (RBP4) are unique in males. In conclusion, IUGR may display sexual dimorphism which may be associated with differences in lifelong risk for cardiometabolic disease between males and females.


Subject(s)
Cardiovascular Diseases , Fetal Growth Retardation , Infant, Newborn , Infant , Humans , Male , Female , Fetal Growth Retardation/etiology , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Proprotein Convertase 9/metabolism , Proteomics , Retinol-Binding Proteins, Plasma , Extracellular Matrix Proteins/metabolism
19.
Am J Respir Cell Mol Biol ; 70(5): 400-413, 2024 May.
Article in English | MEDLINE | ID: mdl-38301267

ABSTRACT

Newborns with intrauterine growth restriction (IUGR) have a higher likelihood of developing pulmonary arterial hypertension (PAH) in adulthood. Although there is increasing evidence suggesting that pericytes play a role in regulating myofibroblast transdifferentiation and angiogenesis in malignant and cardiovascular diseases, their involvement in the pathogenesis of IUGR-related pulmonary hypertension and the underlying mechanisms remain incompletely understood. To address this issue, a study was conducted using a Sprague-Dawley rat model of IUGR-related pulmonary hypertension. Our investigation revealed increased proliferation and migration of pulmonary microvascular pericytes in IUGR-related pulmonary hypertension, accompanied by weakened endothelial-pericyte interactions. Through whole-transcriptome sequencing, Ddx5 (DEAD-box protein 5) was identified as one of the hub genes in pericytes. DDX5, a member of the RNA helicase family, plays a role in the regulation of ATP-dependent RNA helicase activities and cellular function. MicroRNAs have been implicated in the pathogenesis of PAH, and microRNA-205 (miR-205) regulates cell proliferation, migration, and angiogenesis. The results of dual-luciferase reporter assays confirmed the specific binding of miR-205 to Ddx5. Mechanistically, miR-205 negatively regulates Ddx5, leading to the degradation of ß-catenin by inhibiting the phosphorylation of Gsk3ß at serine 9. In vitro experiments showed the addition of miR-205 effectively ameliorated pericyte dysfunction. Furthermore, in vivo experiments demonstrated that miR-205 agomir could ameliorate pulmonary hypertension. Our findings indicated that the downregulation of miR-205 expression mediates pericyte dysfunction through the activation of Ddx5. Therefore, targeting the miR-205/Ddx5/p-Gsk3ß/ß-catenin axis could be a promising therapeutic approach for IUGR-related pulmonary hypertension.


Subject(s)
Cell Proliferation , DEAD-box RNA Helicases , Epigenesis, Genetic , Fetal Growth Retardation , Glycogen Synthase Kinase 3 beta , Hypertension, Pulmonary , MicroRNAs , Pericytes , Rats, Sprague-Dawley , Animals , Female , Humans , Male , Rats , beta Catenin/metabolism , beta Catenin/genetics , Cell Movement/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Pericytes/metabolism , Pericytes/pathology
20.
J Physiol ; 602(12): 2697-2715, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38743350

ABSTRACT

Fetuses affected by intrauterine growth restriction have an increased risk of developing heart disease and failure in adulthood. Compared with controls, late gestation intrauterine growth-restricted (IUGR) fetal sheep have fewer binucleated cardiomyocytes, reflecting a more immature heart, which may reduce mitochondrial capacity to oxidize substrates. We hypothesized that the late gestation IUGR fetal heart has a lower capacity for mitochondrial oxidative phosphorylation. Left (LV) and right (RV) ventricles from IUGR and control (CON) fetal sheep at 90% gestation were harvested. Mitochondrial respiration (states 1-3, LeakOmy, and maximal respiration) in response to carbohydrates and lipids, citrate synthase (CS) activity, protein expression levels of mitochondrial oxidative phosphorylation complexes (CI-CV), and mRNA expression levels of mitochondrial biosynthesis regulators were measured. The carbohydrate and lipid state 3 respiration rates were lower in IUGR than CON, and CS activity was lower in IUGR LV than CON LV. However, relative CII and CV protein levels were higher in IUGR than CON; CV expression level was higher in IUGR than CON. Genes involved in lipid metabolism had lower expression in IUGR than CON. In addition, the LV and RV demonstrated distinct differences in oxygen flux and gene expression levels, which were independent from CON and IUGR status. Low mitochondrial respiration and CS activity in the IUGR heart compared with CON are consistent with delayed cardiomyocyte maturation, and CII and CV protein expression levels may be upregulated to support ATP production. These insights will provide a better understanding of fetal heart development in an adverse in utero environment. KEY POINTS: Growth-restricted fetuses have a higher risk of developing and dying from cardiovascular diseases in adulthood. Mitochondria are the main supplier of energy for the heart. As the heart matures, the substrate preference of the mitochondria switches from carbohydrates to lipids. We used a sheep model of intrauterine growth restriction to study the capacity of the mitochondria in the heart to produce energy using either carbohydrate or lipid substrates by measuring how much oxygen was consumed. Our data show that the mitochondria respiration levels in the growth-restricted fetal heart were lower than in the normally growing fetuses, and the expression levels of genes involved in lipid metabolism were also lower. Differences between the right and left ventricles that are independent of the fetal growth restriction condition were identified. These results indicate an impaired metabolic maturation of the growth-restricted fetal heart associated with a decreased capacity to oxidize lipids postnatally.


Subject(s)
Fetal Growth Retardation , Fetal Heart , Mitochondria, Heart , Animals , Fetal Growth Retardation/metabolism , Sheep , Female , Mitochondria, Heart/metabolism , Fetal Heart/metabolism , Pregnancy , Cell Respiration , Oxidative Phosphorylation , Lipid Metabolism , Citrate (si)-Synthase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL