Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
Biol Sex Differ ; 15(1): 52, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898532

ABSTRACT

BACKGROUND: Prenatal hypoxia, a common pregnancy complication, leads to impaired cardiovascular outcomes in the adult offspring. It results in impaired vasodilation in coronary and mesenteric arteries of the adult offspring, due to reduced nitric oxide (NO). Thromboxane A2 (TxA2) is a potent vasoconstrictor increased in cardiovascular diseases, but its role in the impact of prenatal hypoxia is unknown. To prevent the risk of cardiovascular disease by prenatal hypoxia, we have tested a maternal treatment using a nanoparticle-encapsulated mitochondrial antioxidant (nMitoQ). We hypothesized that prenatal hypoxia enhances vascular TxA2 responses in the adult offspring, due to decreased NO modulation, and that this might be prevented by maternal nMitoQ treatment. METHODS: Pregnant Sprague-Dawley rats received a single intravenous injection (100 µL) of vehicle (saline) or nMitoQ (125 µmol/L) on gestational day (GD)15 and were exposed to normoxia (21% O2) or hypoxia (11% O2) from GD15 to GD21 (term = 22 days). Coronary and mesenteric arteries were isolated from the 4-month-old female and male offspring, and vasoconstriction responses to U46619 (TxA2 analog) were evaluated using wire myography. In mesenteric arteries, L-NAME (pan-NO synthase (NOS) inhibitor) was used to assess NO modulation. Mesenteric artery endothelial (e)NOS, and TxA2 receptor expression, superoxide, and 3-nitrotyrosine levels were assessed by immunofluorescence. RESULTS: Prenatal hypoxia resulted in increased U46619 responsiveness in coronary and mesenteric arteries of the female offspring, and to a lesser extent in the male offspring, which was prevented by nMitoQ. In females, there was a reduced impact of L-NAME in mesenteric arteries of the prenatal hypoxia saline-treated females, and reduced 3-nitrotyrosine levels. In males, L-NAME increased U46619 responses in mesenteric artery to a similar extent, but TxA2 receptor expression was increased by prenatal hypoxia. There were no changes in eNOS or superoxide levels. CONCLUSIONS: Prenatal hypoxia increased TxA2 vasoconstrictor capacity in the adult offspring in a sex-specific manner, via reduced NO modulation in females and increased TP expression in males. Maternal placental antioxidant treatment prevented the impact of prenatal hypoxia. These findings increase our understanding of how complicated pregnancies can lead to a sex difference in the programming of cardiovascular disease in the adult offspring.


Prenatal hypoxia, when the fetus does not receive enough oxygen, is a common problem during pregnancy that impacts the developing fetus. It is associated with an increased risk of cardiovascular disease in the offspring in adulthood. While the mechanisms are not fully understood, the blood vessel function in the offspring may be impacted by prenatal hypoxia. We hypothesize that prenatal hypoxia increases the constriction of the blood vessels in the offspring. The placenta, an essential organ for fetal development, supplies oxygen and nutrients to the fetus. In prenatal hypoxia pregnancies, the placenta does not work properly. We have been studying a placental treatment (called nMitoQ) to improve placenta function and thereby the blood vessel function of the offspring. We used a rat model of prenatal hypoxia, where pregnant rats (dams) were placed in a low oxygen environment (hypoxia) during the last trimester of pregnancy. Control rats were kept in normal oxygen conditions. The dams were treated with nMitoQ, or with saline (control). Next, we studied the blood vessels of the offspring in adulthood. We found that prenatal hypoxia increases the constriction of the blood vessels, which was prevented by treating the dams with nMitoQ. Interestingly, this impact was more severe in females compared to males, and the mechanisms were different between the sexes. This study helps in the understanding of how complicated pregnancies can impair cardiovascular health in the offspring, and in a potential development of targeted and sex-specific therapies for those offspring at high risk for future cardiovascular disease.


Subject(s)
Prenatal Exposure Delayed Effects , Rats, Sprague-Dawley , Sex Characteristics , Thromboxane A2 , Vasoconstriction , Animals , Female , Pregnancy , Vasoconstriction/drug effects , Male , Thromboxane A2/metabolism , Antioxidants/pharmacology , Nitric Oxide/metabolism , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Rats , Hypoxia/metabolism , Fetal Hypoxia/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology
2.
Radiol Clin North Am ; 62(4): 619-625, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38777538

ABSTRACT

Breast cancer risk prediction models based on common clinical risk factors are used to identify women eligible for high-risk screening and prevention. Unfortunately, these models have only modest discriminatory accuracy with disparities in performance in underrepresented race and ethnicity groups. The field of artificial intelligence (AI) and deep learning are rapidly advancing the field of breast cancer risk prediction with the development of mammography-based AI breast cancer risk models. Early studies suggest mammography-based AI risk models may perform better than traditional risk factor-based models with more equitable performance.


Subject(s)
Artificial Intelligence , Breast Neoplasms , Mammography , Humans , Breast Neoplasms/diagnostic imaging , Female , Risk Assessment/methods , Mammography/methods , Breast/diagnostic imaging , Risk Factors , Early Detection of Cancer/methods
3.
Int J Mol Sci ; 24(17)2023 Sep 03.
Article in English | MEDLINE | ID: mdl-37686430

ABSTRACT

Prenatal hypoxia is associated with placental oxidative stress, leading to impaired fetal growth and an increased risk of cardiovascular disease in the adult offspring; however, the mechanisms are unknown. Alterations in mitochondrial function may result in impaired cardiac function in offspring. In this study, we hypothesized that cardiac mitochondrial function is impaired in adult offspring exposed to intrauterine hypoxia, which can be prevented by placental treatment with a nanoparticle-encapsulated mitochondrial antioxidant (nMitoQ). Cardiac mitochondrial respiration was assessed in 4-month-old rat offspring exposed to prenatal hypoxia (11% O2) from gestational day (GD)15-21 receiving either saline or nMitoQ on GD 15. Prenatal hypoxia did not alter cardiac mitochondrial oxidative phosphorylation capacity in the male offspring. In females, the NADH + succinate pathway capacity decreased by prenatal hypoxia and tended to be increased by nMitoQ. Prenatal hypoxia also decreased the succinate pathway capacity in females. nMitoQ treatment increased respiratory coupling efficiency in prenatal hypoxia-exposed female offspring. In conclusion, prenatal hypoxia impaired cardiac mitochondrial function in adult female offspring only, which was improved with prenatal nMitoQ treatment. Therefore, treatment strategies targeting placental oxidative stress in prenatal hypoxia may reduce the risk of cardiovascular disease in adult offspring by improving cardiac mitochondrial function in a sex-specific manner.


Subject(s)
Antioxidants , Cardiovascular Diseases , Female , Male , Pregnancy , Animals , Rats , Antioxidants/pharmacology , Antioxidants/therapeutic use , Placenta , Vitamins , Hypoxia/complications , Hypoxia/drug therapy , Mitochondria , Succinates
4.
Am J Physiol Heart Circ Physiol ; 325(1): H136-H141, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37235521

ABSTRACT

Prenatal hypoxia is associated with enhanced susceptibility to cardiac ischemia-reperfusion (I/R) injury in adult offspring, however, the mechanisms remain to be fully investigated. Endothelin-1 (ET-1) is a vasoconstrictor that acts via endothelin A (ETA) and endothelin B (ETB) receptors and is essential in maintaining cardiovascular (CV) function. Prenatal hypoxia alters the ET-1 system in adult offspring possibly contributing to I/R susceptibility. We previously showed that ex vivo application of ETA antagonist ABT-627 during I/R prevented the recovery of cardiac function in prenatal hypoxia-exposed males but not in normoxic males nor normoxic or prenatal hypoxia-exposed females. In this follow-up study, we examined whether placenta-targeted treatment with a nanoparticle-encapsulated mitochondrial antioxidant (nMitoQ) during hypoxic pregnancies could alleviate this hypoxic phenotype observed in adult male offspring. We used a rat model of prenatal hypoxia where pregnant Sprague-Dawley rats were exposed to hypoxia (11% O2) from gestational days (GD) 15-21 after injection with 100 µL saline or nMitoQ (125 µM) on GD15. Male offspring were aged to 4 mo and ex vivo cardiac recovery from I/R was assessed. Offspring born from hypoxic pregnancies and treated with nMitoQ had increased cardiac recovery from I/R in the presence of ABT-627 compared with their untreated counterparts where ABT-627 prevented recovery. Cardiac ETA levels were increased in males born from hypoxic pregnancies with nMitoQ treatment compared with saline controls (Western blotting). Our data indicate a profound impact of placenta-targeted treatment to prevent an ETA receptor cardiac phenotype observed in adult male offspring exposed to hypoxia in utero.NEW & NOTEWORTHY In this follow-up study, we showed a complete lack of recovery from I/R injury after the application of an ETA receptor antagonist (ABT-627) in adult male offspring exposed to hypoxia in utero while maternal treatment with nMitoQ during prenatal hypoxia exposure prevented this effect. Our data suggest that nMitoQ treatment during hypoxic pregnancies may prevent a hypoxic cardiac phenotype in adult male offspring.


Subject(s)
Hypoxia , Receptors, Endothelin , Pregnancy , Female , Rats , Male , Animals , Rats, Sprague-Dawley , Atrasentan , Follow-Up Studies , Hypoxia/complications , Placenta , Endothelin-1
5.
FASEB J ; 35(2): e21338, 2021 02.
Article in English | MEDLINE | ID: mdl-33428278

ABSTRACT

Pregnancy complications associated with prenatal hypoxia lead to increased placental oxidative stress. Previous studies suggest that prenatal hypoxia can reduce mitochondrial respiratory capacity and mitochondrial fusion, which could lead to placental dysfunction and impaired fetal development. We developed a placenta-targeted treatment strategy using a mitochondrial antioxidant, MitoQ, encapsulated into nanoparticles (nMitoQ) to reduce placental oxidative stress and (indirectly) improve fetal outcomes. We hypothesized that, in a rat model of prenatal hypoxia, nMitoQ improves placental mitochondrial function and promotes mitochondrial fusion in both male and female placentae. Pregnant rats were treated with saline or nMitoQ on gestational day (GD) 15 and exposed to normoxia (21% O2 ) or hypoxia (11% O2 ) from GD15-21. On GD21, male and female placental labyrinth zones were collected for mitochondrial respirometry assessments, mitochondrial content, and markers of mitochondrial biogenesis, fusion and fission. Prenatal hypoxia reduced complex IV activity and fusion in male placentae, while nMitoQ improved complex IV activity in hypoxic male placentae. In female placentae, prenatal hypoxia decreased respiration through the S-pathway (complex II) and increased N-pathway (complex I) respiration, while nMitoQ increased fusion in hypoxic female placentae. No changes in mitochondrial content, biogenesis or fission were found. In conclusion, nMitoQ improved placental mitochondrial function in male and female placentae from fetuses exposed to prenatal hypoxia, which may contribute to improved placental function. However, the mechanisms (ie, changes in mitochondrial respiratory capacity and mitochondrial fusion) were distinct between the sexes. Treatment strategies targeted against placental oxidative stress could improve placental mitochondrial function in complicated pregnancies.


Subject(s)
Antioxidants/therapeutic use , Fetal Hypoxia/drug therapy , Mitochondria/drug effects , Nanoparticles/chemistry , Organophosphorus Compounds/therapeutic use , Placenta/drug effects , Ubiquinone/analogs & derivatives , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Cell Respiration , Female , Male , Mitochondria/metabolism , Mitochondrial Dynamics , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/pharmacology , Placenta/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Sex Factors , Ubiquinone/administration & dosage , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
6.
Pharmacol Res ; 165: 105461, 2021 03.
Article in English | MEDLINE | ID: mdl-33513355

ABSTRACT

Offspring born from complicated pregnancies are at greater risk of cardiovascular disease in adulthood. Prenatal hypoxia is a common pregnancy complication that results in placental oxidative stress and impairs fetal development. Adult offspring exposed to hypoxia during fetal life are more susceptible to develop cardiac dysfunction, and show decreased cardiac tolerance to an ischemia/reperfusion (I/R) insult. To improve offspring cardiac outcomes, we have assessed the use of a placenta-targeted intervention during hypoxic pregnancies, by encapsulating the mitochondrial antioxidant MitoQ into nanoparticles (nMitoQ). We hypothesized that maternal nMitoQ treatment during hypoxic pregnancies improves cardiac tolerance to I/R insult in adult male and female offspring. Pregnant Sprague-Dawley rats were exposed to normoxia (21 % O2) or hypoxia (11 % O2) from gestational day 15-20, after injection with 100 µL saline or nMitoQ (125 µM) on GD15 (n=6-8/group). Male and female offspring were aged to 4 months. Both male and female offspring from hypoxic pregnancies showed reduced cardiac tolerance to I/R (assessed ex vivo using the isolated working heart technique) which was ameliorated by nMitoQ treatment. To identify potential molecular mechanisms for the changes in cardiac tolerance to I/R, cardiac levels/phosphorylation of proteins important for intracellular Ca2+ cycling were assessed with Western blotting. In prenatally hypoxic male offspring, improved cardiac recovery from I/R by nMitoQ was accompanied by increased cardiac phospholamban and phosphatase 2Ce levels, and a trend to decreased Ca2+/calmodulin-dependent protein kinase IIδ phosphorylation. In contrast, in female offspring, nMitoQ treatment in hypoxic pregnancies increased phospholamban and protein kinase Cε phosphorylation. Maternal nMitoQ treatment improves cardiac tolerance to I/R insult in adult offspring and thus has the potential to improve the later-life trajectory of cardiovascular health of adult offspring born from pregnancies complicated by prenatal hypoxia.


Subject(s)
Cardiovascular Diseases/metabolism , Hypoxia/metabolism , Organophosphorus Compounds/administration & dosage , Placenta/metabolism , Prenatal Exposure Delayed Effects/metabolism , Reperfusion Injury/metabolism , Ubiquinone/analogs & derivatives , Age Factors , Animals , Antioxidants/administration & dosage , Cardiovascular Diseases/prevention & control , Female , Hypoxia/drug therapy , Male , Nanoparticles/administration & dosage , Placenta/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/drug therapy , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Ubiquinone/administration & dosage
7.
Neurobiol Stress ; 13: 100281, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33344732

ABSTRACT

Maternal exposure to stress during pregnancy is associated with an increased risk of psychiatric disorders in the offspring in later life. The mechanisms through which the effects of maternal stress are transmitted to the fetus are unclear, however the placenta, as the interface between mother and fetus, is likely to play a key role. Using a rat model, we investigated a role for placental oxidative stress in conveying the effects of maternal social stress to the fetus and the potential for treatment using a nanoparticle-bound antioxidant to prevent adverse outcomes in the offspring. Maternal psychosocial stress increased circulating corticosterone in the mother, but not in the fetuses. Maternal stress also induced oxidative stress in the placenta, but not in the fetal brain. Blocking oxidative stress using an antioxidant prevented the prenatal stress-induced anxiety phenotype in the male offspring, and prevented sex-specific neurobiological changes, specifically a reduction in dendrite lengths in the hippocampus, as well as reductions in the number of parvalbumin-positive neurons and GABA receptor subunits in the hippocampus and basolateral amygdala of the male offspring. Importantly, many of these effects were mimicked in neuronal cultures by application of placental-conditioned medium or fetal plasma from stressed pregnancies, indicating molecules released from the placenta may mediate the effects of prenatal stress on the fetal brain. Indeed, both placenta-conditioned medium and fetal plasma contained differentially abundant microRNAs following maternal stress, and their predicted targets were enriched for genes relevant to nervous system development and psychiatric disorders. The results highlight placental oxidative stress as a key mediator in transmitting the maternal social stress effects on the offspring's brain and behavior, and offer a potential intervention to prevent stress-induced fetal programming of affective disorders.

8.
Exp Physiol ; 105(9): 1507-1514, 2020 09.
Article in English | MEDLINE | ID: mdl-32749725

ABSTRACT

NEW FINDINGS: What is the central question of this study? Does treatment of hypoxic dams with a placenta-targeted antioxidant prevent the release of placenta-derived factors that impair maturation or growth of fetal cardiomyocytes in vitro? What is the main finding and its importance? Factors released from hypoxic placentae impaired fetal cardiomyocyte maturation (induced terminal differentiation) and growth (increased cell size) in vitro, which was prevented by maternal treatment with a placenta-targeted antioxidant (nMitoQ). Moreover, there were no sex differences in the effects of placental factors on fetal cardiomyocyte maturation and growth. Overall, our data suggest that treatment targeted against placental oxidative stress could prevent fetal programming of cardiac diseases via the release of placental factors. ABSTRACT: Pregnancy complications associated with placental oxidative stress may impair fetal organ development through the release of placenta-derived factors into the fetal circulation. We assessed the effect of factors secreted from placentae previously exposed to prenatal hypoxia on fetal cardiomyocyte development and developed a treatment strategy that targets placental oxidative stress by encapsulating the antioxidant MitoQ into nanoparticles (nMitoQ). We used a rat model of prenatal hypoxia (gestational day (GD) 15-21), which was treated with saline or nMitoQ on GD15. On GD21, placentae were harvested, placed in culture, and conditioned medium (containing placenta-derived factors) was collected after 24 h. This conditioned medium was then added to cultured cardiomyocytes from control dam fetuses. Conditioned medium from prenatally hypoxic placentae increased the percentage of binucleated cardiomyocytes (marker of terminal differentiation) and the size of mononucleated and binucleated cardiomyocytes (sign of hypertrophy), effects that were prevented by nMitoQ treatment. Our data suggest that factors derived from placentae previously exposed to prenatal hypoxia lead to abnormal fetal cardiomyocyte development, and show that treatment against placental oxidative stress may prevent fetal programming of cardiac disease.


Subject(s)
Antioxidants/pharmacology , Fetal Development/drug effects , Hypoxia/drug therapy , Myocytes, Cardiac/physiology , Placenta/physiology , Animals , Cells, Cultured , Culture Media, Conditioned , Female , Male , Organophosphorus Compounds/pharmacology , Oxidative Stress , Pregnancy , Rats , Rats, Sprague-Dawley , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
9.
Front Physiol ; 10: 562, 2019.
Article in English | MEDLINE | ID: mdl-31178743

ABSTRACT

Pregnancy complications associated with chronic fetal hypoxia have been linked to the development of adult cardiovascular disease in the offspring. Prenatal hypoxia has been shown to increase placental oxidative stress and impair placental function in a sex-specific manner, thereby affecting fetal development. As oxidative stress is central to placental dysfunction, we developed a placenta-targeted treatment strategy using the antioxidant MitoQ encapsulated into nanoparticles (nMitoQ) to reduce placental oxidative/nitrosative stress and improve placental function without direct drug exposure to the fetus in order to avoid off-target effects during development. We hypothesized that, in a rat model of prenatal hypoxia, nMitoQ prevents hypoxia-induced placental oxidative/nitrosative stress, promotes angiogenesis, improves placental morphology, and ultimately improves fetal oxygenation. Additionally, we assessed whether there were sex differences in the effectiveness of nMitoQ treatment. Pregnant rats were intravenously injected with saline or nMitoQ (100 µl of 125 µM) on gestational day (GD) 15 and exposed to either normoxia (21% O2) or hypoxia (11% O2) from GD15 to 21. On GD21, placentae from both sexes were collected for detection of superoxide, nitrotyrosine, nitric oxide, CD31 (endothelial cell marker), and fetal blood spaces, Vegfa and Igf2 mRNA expression in the placental labyrinth zone. Prenatal hypoxia decreased male fetal weight, which was not changed by nMitoQ treatment; however, placental efficiency (fetal/placental weight ratio) decreased by hypoxia and was increased by nMitoQ in both males and females. nMitoQ treatment reduced the prenatal hypoxia-induced increase in placental superoxide levels in both male and female placentae but improved oxygenation in only female placentae. Nitrotyrosine levels were increased in hypoxic female placentae and were reduced by nMitoQ. Prenatal hypoxia reduced placental Vegfa and Igf2 expression in both sexes, while nMitoQ increased Vegfa and Igf2 expression only in hypoxic female placentae. In summary, our study suggests that nMitoQ treatment could be pursued as a potential preventative strategy against placental oxidative stress and programming of adult cardiovascular disease in offspring exposed to hypoxia in utero. However, sex differences need to be taken into account when developing therapeutic strategies to improve fetal development in complicated pregnancies, as nMitoQ treatment was more effective in placentae from females than males.

10.
Sci Rep ; 9(1): 4370, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867444

ABSTRACT

For many diseases with a foetal origin, the cause for the disease initiation remains unknown. Common childhood acute leukaemia is thought to be caused by two hits, the first in utero and the second in childhood in response to infection. The mechanism for the initial DNA damaging event are unknown. Here we have used in vitro, ex vivo and in vivo models to show that a placental barrier will respond to agents that are suspected of initiating childhood leukaemia by releasing factors that cause DNA damage in cord blood and bone marrow cells, including stem cells. We show that DNA damage caused by in utero exposure can reappear postnatally after an immune challenge. Furthermore, both foetal and postnatal DNA damage are prevented by prenatal exposure of the placenta to a mitochondrially-targeted antioxidant. We conclude that the placenta might contribute to the first hit towards leukaemia initiation by bystander-like signalling to foetal haematopoietic cells.


Subject(s)
DNA Damage , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , Placenta/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Signal Transduction , Carcinogens/pharmacology , Chromosome Aberrations , Culture Media, Conditioned , DNA Damage/drug effects , Female , Fibroblasts/metabolism , Humans , Infant, Newborn , Leukemia, Myeloid, Acute/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pregnancy , Signal Transduction/drug effects , Stem Cells/metabolism , Trophoblasts/drug effects , Trophoblasts/metabolism
11.
Biomaterials ; 192: 140-148, 2019 02.
Article in English | MEDLINE | ID: mdl-30448698

ABSTRACT

Fetal development may be compromised by adverse events at the placental interface between mother and fetus. However, it is still unclear how the communication between mother and fetus occurs through the placenta. In vitro - models of the human placental barrier, which could help our understanding and which recreate three-dimensional (3D) structures with biological functionalities and vasculatures, have not been reported yet. Here we present a 3D-vascularized human primary placental barrier model which can be constructed in 1 day. We illustrate the similarity of our model to first trimester human placenta, both in its structure and in its ability to respond to altered oxygen and to secrete factors that cause damage cells across the barrier including embryonic cortical neurons. We use this model to highlight the possibility that both the trophoblast and the endothelium within the placenta might play a role in the fetomaternal dialogue.


Subject(s)
Connective Tissue Cells/cytology , Endothelium, Vascular/cytology , Placenta/blood supply , Trophoblasts/cytology , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells , Humans , Neurons/cytology , Placenta/cytology , Pregnancy
12.
Pharmacol Res ; 134: 332-342, 2018 08.
Article in English | MEDLINE | ID: mdl-29778808

ABSTRACT

Intrauterine growth restriction, a common consequence of prenatal hypoxia, is a leading cause of fetal morbidity and mortality with a significant impact on population health. Hypoxia may increase placental oxidative stress and lead to an abnormal release of placental-derived factors, which are emerging as potential contributors to developmental programming. Nanoparticle-linked drugs are emerging as a novel method to deliver therapeutics targeted to the placenta and avoid risking direct exposure to the fetus. We hypothesize that placental treatment with antioxidant MitoQ loaded onto nanoparticles (nMitoQ) will prevent the development of cardiovascular disease in offspring exposed to prenatal hypoxia. Pregnant rats were intravenously injected with saline or nMitoQ (125 µM) on gestational day (GD) 15 and exposed to either normoxia (21% O2) or hypoxia (11% O2) from GD15-21 (term: 22 days). In one set of animals, rats were euthanized on GD 21 to assess fetal body weight, placental weight and placental oxidative stress. In another set of animals, dams were allowed to give birth under normal atmospheric conditions (term: GD 22) and male and female offspring were assessed at 7 and 13 months of age for in vivo cardiac function (echocardiography) and vascular function (wire myography, mesenteric artery). Hypoxia increased oxidative stress in placentas of male and female fetuses, which was prevented by nMitoQ. 7-month-old male and female offspring exposed to prenatal hypoxia demonstrated cardiac diastolic dysfunction, of which nMitoQ improved only in 7-month-old female offspring. Vascular sensitivity to methacholine was reduced in 13-month-old female offspring exposed to prenatal hypoxia, while nMitoQ treatment improved vasorelaxation in both control and hypoxia exposed female offspring. Male 13-month-old offspring exposed to hypoxia showed an age-related decrease in vascular sensitivity to phenylephrine, which was prevented by nMitoQ. In summary, placental-targeted MitoQ treatment in utero has beneficial sex- and age-dependent effects on adult offspring cardiovascular function.


Subject(s)
Antioxidants/administration & dosage , Cardiovascular Diseases/prevention & control , Fetal Hypoxia/drug therapy , Organophosphorus Compounds/administration & dosage , Oxidative Stress/drug effects , Placenta/drug effects , Prenatal Exposure Delayed Effects , Ubiquinone/analogs & derivatives , Age Factors , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Disease Models, Animal , Female , Fetal Hypoxia/metabolism , Fetal Hypoxia/physiopathology , Gestational Age , Hemodynamics/drug effects , Male , Maternal Exposure , Myocardial Contraction/drug effects , Nanoparticles , Placenta/metabolism , Placenta/physiopathology , Pregnancy , Rats, Sprague-Dawley , Sex Factors , Ubiquinone/administration & dosage , Ventricular Function, Left/drug effects
13.
Nat Nanotechnol ; 13(5): 427-433, 2018 05.
Article in English | MEDLINE | ID: mdl-29610530

ABSTRACT

The potential for maternal nanoparticle (NP) exposures to cause developmental toxicity in the fetus without the direct passage of NPs has previously been shown, but the mechanism remained elusive. We now demonstrate that exposure of cobalt and chromium NPs to BeWo cell barriers, an in vitro model of the human placenta, triggers impairment of the autophagic flux and release of interleukin-6. This contributes to the altered differentiation of human neural progenitor cells and DNA damage in the derived neurons and astrocytes. Crucially, neuronal DNA damage is mediated by astrocytes. Inhibiting the autophagic degradation in the BeWo barrier by overexpression of the dominant-negative human ATG4BC74A significantly reduces the levels of DNA damage in astrocytes. In vivo, indirect NP toxicity in mice results in neurodevelopmental abnormalities with reactive astrogliosis and increased DNA damage in the fetal hippocampus. Our results demonstrate the potential importance of autophagy to elicit NP toxicity and the risk of indirect developmental neurotoxicity after maternal NP exposure.


Subject(s)
Astrocytes/metabolism , Models, Biological , Nanoparticles/toxicity , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Placenta/pathology , Pregnancy Complications/metabolism , Animals , Astrocytes/pathology , Cell Line , Female , Humans , Male , Mice , Neurodevelopmental Disorders/chemically induced , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neurons/pathology , Neurotoxicity Syndromes/pathology , Pregnancy , Pregnancy Complications/chemically induced , Pregnancy Complications/pathology
14.
Neuronal Signal ; 2(4): NS20180139, 2018 Dec.
Article in English | MEDLINE | ID: mdl-32714596

ABSTRACT

Prenatal development is a critical period for programming of neurological disease. Preeclampsia, a pregnancy complication involving oxidative stress in the placenta, has been associated with long-term health implications for the child, including an increased risk of developing schizophrenia and autism spectrum disorders in later life. To investigate if molecules released by the placenta may be important mediators in foetal programming of the brain, we analysed if placental tissue delivered from patients with preeclampsia secreted molecules that could affect cortical cells in culture. Application of culture medium conditioned by preeclamptic placentae to mixed cortical cultures caused changes in neurons and astrocytes that were related to key changes observed in brains of patients with schizophrenia and autism, including effects on dendrite lengths, astrocyte number as well as on levels of glutamate and γ-aminobutyric acid receptors. Treatment of the placental explants with an antioxidant prevented neuronal abnormalities. Furthermore, we identified that bidirectional communication between neurons and astrocytes, potentially via glutamate, is required to produce the effects of preeclamptic placenta medium on cortical cells. Analysis of possible signalling molecules in the placenta-conditioned medium showed that the secretion profile of extracellular microRNAs, small post-transcriptional regulators, was altered in preeclampsia and partially rescued by antioxidant treatment of the placental explants. Predicted targets of these differentially abundant microRNAs were linked to neurodevelopment and the placenta. The present study provides further evidence that the diseased placenta may release factors that damage cortical cells and suggests the possibility of targeted antioxidant treatment of the placenta to prevent neurodevelopmental disorders.

15.
Sci Rep ; 7(1): 9079, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831049

ABSTRACT

Some neuropsychiatric disease, including schizophrenia, may originate during prenatal development, following periods of gestational hypoxia and placental oxidative stress. Here we investigated if gestational hypoxia promotes damaging secretions from the placenta that affect fetal development and whether a mitochondria-targeted antioxidant MitoQ might prevent this. Gestational hypoxia caused low birth-weight and changes in young adult offspring brain, mimicking those in human neuropsychiatric disease. Exposure of cultured neurons to fetal plasma or to secretions from the placenta or from model trophoblast barriers that had been exposed to altered oxygenation caused similar morphological changes. The secretions and plasma contained altered microRNAs whose targets were linked with changes in gene expression in the fetal brain and with human schizophrenia loci. Molecular and morphological changes in vivo and in vitro were prevented by a single dose of MitoQ bound to nanoparticles, which were shown to localise and prevent oxidative stress in the placenta but not in the fetus. We suggest the possibility of developing preventative treatments that target the placenta and not the fetus to reduce risk of psychiatric disease in later life.


Subject(s)
Brain/embryology , Brain/metabolism , Fetal Development , Hypoxia/metabolism , Placenta/metabolism , Pregnancy Complications/metabolism , Animals , Antioxidants/metabolism , Biomarkers , Female , Fetus/metabolism , Gene Expression , Microscopy, Confocal , Organogenesis , Oxidative Stress , Pregnancy , Rats , Reactive Oxygen Species/metabolism
16.
Sci Rep ; 5: 11694, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26170169

ABSTRACT

Maternal exposure during pregnancy to toxins can occasionally lead to miscarriage and malformation. It is currently thought that toxins pass through the placental barrier, albeit bi-layered in the first trimester, and damage the fetus directly, albeit at low concentration. Here we examined the responses of human embryonic stem (hES) cells in tissue culture to two metals at low concentration. We compared direct exposures with indirect exposures across a bi-layered model of the placenta cell barrier. Direct exposure caused increased DNA damage without apoptosis or a loss of cell number but with some evidence of altered differentiation. Indirect exposure caused increased DNA damage and apoptosis but without loss of pluripotency. This was not caused by metal ions passing through the barrier. Instead the hES cells responded to signalling molecules (including TNF-α) secreted by the barrier cells. This mechanism was dependent on connexin 43 mediated intercellular 'bystander signalling' both within and between the trophoblast barrier and the hES colonies. These results highlight key differences between direct and indirect exposure of hES cells across a trophoblast barrier to metal toxins. It offers a theoretical possibility that an indirectly mediated toxicity of hES cells might have biological relevance to fetal development.


Subject(s)
Cell Communication , Human Embryonic Stem Cells/metabolism , Signal Transduction , Trophoblasts/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Connexin 43/metabolism , Cytokines/biosynthesis , DNA Damage/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gap Junctions/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Metals, Heavy/toxicity , Trophoblasts/cytology , Trophoblasts/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
17.
Adv Colloid Interface Sci ; 218: 48-68, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25708746

ABSTRACT

Understanding interactions between nanoparticles (NPs) with biological matter, particularly cells, is becoming increasingly important due to their growing application in medicine and materials, and consequent biological and environmental exposure. For NPs to be utilised to their full potential, it is important to correlate their functional characteristics with their physical properties, which may also be used to predict any adverse cellular responses. A key mechanism for NPs to impart toxicity is to gain cellular entry directly. Many parameters affect the behaviour of nanomaterials in a cellular environment particularly their interactions with cell membranes, including their size, shape and surface chemistry as well as factors such as the cell type, location and external environment (e.g. other surrounding materials, temperature, pH and pressure). Aside from in vitro and in vivo experiments, model cell membrane systems have been used in both computer simulations and physicochemical experiments to elucidate the mechanisms for NP cellular entry. Here we present a brief overview of the effects of NPs physical parameters on their cellular uptake, with focuses on 1) related research using model membrane systems and physicochemical methodologies; and 2) proposed physical mechanisms for NP cellular entrance, with implications to their nanotoxicity. We conclude with a suggestion that the energetic process of NP cellular entry can be evaluated by studying the effects of NPs on lipid mesophase transitions, as the molecular deformations and thus the elastic energy cost are analogous between such transitions and endocytosis. This presents an opportunity for contributions to understanding nanotoxicity from a physicochemical perspective.


Subject(s)
Cell Membrane/drug effects , Nanoparticles/administration & dosage , Endocytosis , Humans , Hydrophobic and Hydrophilic Interactions , Nanoparticles/chemistry , Nanoparticles/toxicity
18.
Exp Neurol ; 261: 386-95, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24818543

ABSTRACT

Some psychiatric diseases in children and young adults are thought to originate from adverse exposures during foetal life, including hypoxia and hypoxia/reoxygenation. The mechanism is not understood. Several authors have emphasised that the placenta is likely to play an important role as the key interface between mother and foetus. Here we have explored whether a first trimester human placenta or model barrier of primary human cytotrophoblasts might secrete factors, in response to hypoxia or hypoxia/reoxygenation, that could damage neurones. We find that the secretions in conditioned media caused an increase of [Ca(2+)]i and mitochondrial free radicals and a decrease of dendritic lengths, branching complexity, spine density and synaptic activity in dissociated neurones from embryonic rat cerebral cortex. There was altered staining of glutamate and GABA receptors. We identify glutamate as an active factor within the conditioned media and demonstrate a specific release of glutamate from the placenta/cytotrophoblast barriers invitro after hypoxia or hypoxia/reoxygenation. Injection of conditioned media into developing brains of P4 rats reduced the numerical density of parvalbumin-containing neurones in cortex, hippocampus and reticular nucleus, reduced immunostaining of glutamate receptors and altered cellular turnover. These results show that the placenta is able to release factors, in response to altered oxygen, that can damage developing neurones under experimental conditions.


Subject(s)
Brain , Culture Media, Conditioned/adverse effects , Hypoxia , Neurons/drug effects , Oxygen/pharmacology , Placenta/chemistry , Animals , Animals, Newborn , Brain/cytology , Brain/growth & development , Brain/pathology , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/cytology , Culture Media, Conditioned/chemistry , Dendrites/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian , Female , Fetus , Glial Fibrillary Acidic Protein/metabolism , Humans , Hypoxia/drug therapy , Hypoxia/pathology , Hypoxia/physiopathology , Membrane Potentials/drug effects , Neurons/cytology , Neurons/physiology , Placenta/cytology , Pregnancy , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Tissue Culture Techniques
19.
Hip Int ; 24(3): 243-8, 2014.
Article in English | MEDLINE | ID: mdl-24500829

ABSTRACT

The histological specimens from 29 failed metal-on-metal (MoM) hip arthroplasties treated at our institution were reviewed. Five patients had a failed MoM total hip arthroplasty (THA), and 24 patients a failed hip resurfacing. Clinical and radiographic features of each hip were correlated with the histological findings. We report three major histological subtypes. Patients either have a macrophage response to metal debris, a lymphocytic response (ALVAL) or a mixed picture of both. In addition we observe that the ALVAL response is located deep within tissue specimens, and can occur in environments of low wear debris. The macrophage response is limited to the surface of tissue specimens, with normal underlying tissue. Patients with subsequently confirmed ALVAL underwent revision surgery sooner than patients whose histology confirms a macrophage response (3.8 vs. 6.9 years p<0.05). Both histological subtypes (ALVAL and macrophage dominant) are responsible for abnormal soft tissue swellings.


Subject(s)
Arthroplasty, Replacement, Hip , Hip Prosthesis , Metal-on-Metal Joint Prostheses , Female , Foreign-Body Reaction/pathology , Humans , Lymphocytes/pathology , Macrophages/pathology , Male , Metal-on-Metal Joint Prostheses/adverse effects , Middle Aged , Prosthesis Failure , Reoperation , Retrospective Studies
20.
Biomaterials ; 34(34): 8564-80, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23932295

ABSTRACT

Metal hip replacements generate both metal particles and ions. The biological effects of peri-articular exposure to nanometre and micron sized cobalt chrome (CoCr) wear particles were investigated in a mouse model. Mice received injections of two clinically relevant doses of nanoparticles (32 nm), one of micron sized (2.9 µm) CoCr particles or vehicle alone into the right knee joint at 0, 6, 12 and 18 weeks. Mice were analysed for genotoxic and immunological effects 1, 4 and 40 weeks post exposure. Nanoparticles but not micron particles progressively corroded at the injection site. Micron sized particles were physically removed. No increase of Co or Cr was seen in peripheral blood between 1 and 40 weeks post exposure to particles. No significant inflammatory changes were observed in the knee tissues including ALVAL or necrosis. DNA damage was increased in bone marrow at one and forty weeks and in cells isolated from frontal cortex at 40 weeks after injection with nanoparticles. Mice exposed to the micron sized, but not nanoparticles became immunologically sensitized to Cr(III), Cr (VI) and Ni(II) over the 40 week period as determined by lymphocyte transformation and ELISpot (IFN-γ and IL-2) assays. The data indicated that the response to the micron sized particles was Th1 driven, indicative of type IV hypersensitivity. This study adds to understanding of the potential adverse biological reactions to metal wear products.


Subject(s)
Bone Marrow/pathology , Chromium Alloys/adverse effects , Metal Nanoparticles/adverse effects , Prefrontal Cortex/pathology , Animals , Bone Marrow/drug effects , Chromium/metabolism , Chromosome Aberrations/drug effects , Cobalt/metabolism , Comet Assay , DNA Damage/drug effects , Female , Injections, Intra-Articular , Interferon-gamma/metabolism , Interleukin-2/metabolism , Knee Joint/drug effects , Knee Joint/pathology , Mice , Mice, Inbred C3H , Particle Size , Prefrontal Cortex/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...