Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Article in English | MEDLINE | ID: mdl-38782369

ABSTRACT

Pregnancy is a remarkable event where the semi-allogeneic fetus develops in the mother's uterus, despite genetic and immunological differences. The antigen handling and processing at the maternal-fetal interface during pregnancy appear to be crucial for the adaptation of the maternal immune system and for tolerance to the developing fetus and placenta. Maternal antigen-presenting cells (APCs), such as macrophages (Mφs) and dendritic cells (DCs), are present at the maternal-fetal interface throughout pregnancy and are believed to play a crucial role in this process. Despite numerous studies focusing on the significance of Mφs, there is limited knowledge regarding the contribution of DCs in fetomaternal tolerance during pregnancy, making it a relatively new and growing field of research. This review focuses on how the behavior of DCs at the maternal-fetal interface adapts to pregnancy's unique demands. Moreover, it discusses how DCs interact with other cells in the decidual leukocyte network to regulate uterine and placental homeostasis and the local maternal immune responses to the fetus. The review particularly examines the different cell lineages of DCs with specific surface markers, which have not been critically reviewed in previous publications. Additionally, it emphasizes the impact that even minor disruptions in DC functions can have on pregnancy-related complications and proposes further research into the potential therapeutic benefits of targeting DCs to manage these complications.


Subject(s)
Dendritic Cells , Immune Tolerance , Maternal-Fetal Exchange , Placenta , Humans , Pregnancy , Dendritic Cells/immunology , Female , Maternal-Fetal Exchange/immunology , Placenta/immunology , Fetus/immunology , Animals , Macrophages/immunology , Pregnancy Complications/immunology
2.
Methods Mol Biol ; 2728: 173-180, 2024.
Article in English | MEDLINE | ID: mdl-38019401

ABSTRACT

A major challenge in developing potential treatments for pregnancy complications is minimizing adverse effects to the fetus and mother. Placenta-targeted drug delivery could reduce the risks of drug treatments in pregnancy by targeting tissue where most pregnancy complications originate and decreasing dosages. We previously developed a tool for the targeted delivery of drug-carrying nanoparticles to the placenta using a synthetic placental chondroitin sulfate A-binding peptide (plCSA-BP) derived from the malarial protein VAR2CSA, which binds a distinct type of chondroitin sulfate A (CSA) exclusively expressed by placental trophoblasts. Liposomes are a type of nanoparticle already approved for use in humans by the Food and Drug Administration (FDA) and used successfully for the treatment of a wide range of diseases. Here, we present a detailed method to create plCSA-BP-decorated liposomes that can be used to deliver drugs specifically to placental trophoblasts. Liposomes are first generated by the standard film method and then conjugated to plCSA-BPs using the 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride/N-hydroxysulfosuccinimide (EDC/NHS) bioconjugate technique. This protocol may facilitate bench-to-bedside translation of drug discovery for the treatment of pregnancy disorders by reducing risks of side effects, and enabling rapid and scalable production.


Subject(s)
Liposomes , Pregnancy Complications , Pregnancy , United States , Humans , Female , Chondroitin Sulfates , Trophoblasts , Placenta
3.
Cells ; 12(16)2023 08 18.
Article in English | MEDLINE | ID: mdl-37626906

ABSTRACT

The aromatase-Cre recombinase (Cyp19-Cre) transgenic mouse model has been extensively used for placenta-specific gene inactivation. In a pilot study, we observed unexpected phenotypes using this mouse strain, which prompted an extensive characterization of Cyp19-Cre placental phenotypes using ROSAmT/mG transgenic reporter mice. The two strains were mated to generate bi-transgenic Cyp19-Cre;ROSAmT/mG mice following a standard transgenic breeding scheme, and placental and fetal tissues were analyzed on embryonic day 17.5. Both maternal and paternal Cre inheritance were analyzed by mating the respective Cyp19-Cre and ROSAmT/mG males and females. The genotype results showed the expected percentage of Cyp19-Cre;ROSAmT/mG fetuses (73%) and Cre mRNA was expressed in all of the Cyp19-Cre placentas. However, surprisingly, only about 50% of the Cyp19-Cre;ROSAmT/mG placentas showed Cre-mediated recombinase activity as demonstrated by placental enhanced green fluorescent protein (EGFP) expression. Further genetic excision analysis of the placentas revealed consistent results showing the absence of excision of the tdTomato in all of the Cyp19-Cre;ROSAmT/mG placentas lacking EGFP expression. Moreover, among the EGFP-expressing placentas, there was wide variability in recombination efficiency, even in placentas from the same litter, leading to a mosaic pattern of EGFP expression in different zones and cell types of the placentas. In addition, we observed a significantly higher percentage of Cre recombination activity in placentas with maternal Cre inheritance. Our results show frequent mosaicism, inconsistent recombination activity, and parent-of-origin effects in placentas from Cyp19-Cre;ROSAmT/mG mice, suggesting that tail-biopsy genotype results may not necessarily indicate the excision of floxed genes in Cyp19-Cre positive placentas. Thus, placenta-specific mutagenesis studies using the Cyp19-Cre model require extensive characterization and careful interpretation of the placental phenotypes for each floxed allele.


Subject(s)
Rosa , Female , Pregnancy , Male , Mice , Animals , Mice, Transgenic , Aromatase/genetics , Pilot Projects , Placenta , Plant Breeding , Mosaicism
4.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36835124

ABSTRACT

The purpose of this editorial is to highlight the various observations made in this Special Issue in the International Journal of Molecular Sciences [...].


Subject(s)
Pregnancy Complications , Vascular Remodeling , Pregnancy , Female , Humans
5.
Arch Immunol Ther Exp (Warsz) ; 71(1): 3, 2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36681768

ABSTRACT

Preeclampsia and HIV are a significant burden to maternal health globally, especially in low-middle income countries such as South Africa. In the KwaZulu-Natal province, SA antenatal HIV prevalence is 41.1%, while PE is 12%. PE and HIV infections are maternal stress and inflammation that impact placental function and fetal development. Therefore, this study investigated the impact of the comorbidity of PE and HIV on placental stress and neurodevelopment. Placentae were obtained from four cohorts of pregnant women: normotensive HIV negative, normotensive HIV positive, preeclamptic HIV negative, and preeclamptic HIV positive. The placental tissue sections were immunostained for OGT and T4. Our findings showed that the maternal weight, diastolic, and systolic blood pressures (BP) were higher in PE vs. the normotensive groups, irrespective of HIV status. In addition, significant changes were noticed in the placental weight, fetoplacental ratio, and placental efficiency coefficient. Our findings showed that the maternal weight, diastolic, and systolic blood pressures (BP) were statistically higher in the PE compared to the normotensive. No significant differences were observed between HIV positive and HIV negative groups. In addition, significant changes were noticed in the placental weight, fetoplacental ratio, and placental coefficient. Furthermore, considerable upregulation in the placental expression of OGT in both the conducting and exchange villi of PE and concomitant downregulation in HIV-positive patients compared with Normotensive and HIV-negative individuals, respectively. Our results provide inferential evidence on the dysregulation of OGT in the comorbidity of PE and HIV. This may mediate a compromised programmed outcome of an adverse maternal environment during pregnancy and consequently affect fetal development.


Subject(s)
HIV Infections , Pre-Eclampsia , Female , Pregnancy , Humans , Placenta , Pre-Eclampsia/metabolism , South Africa/epidemiology
6.
Eur J Obstet Gynecol Reprod Biol ; 282: 133-139, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36716536

ABSTRACT

OBJECTIVE: This study investigates the association of C1q gene (rs292001 and rs294183) polymorphisms in HIV infected and uninfected preeclamptic women of African ancestry. MATERIALS AND METHODS: The study population consisted of 325 pregnant women of African ancestry grouped into 145 normotensive pregnant women (72 HIV uninfected normotensive, 73 HIV infected normotensive) and 180 preeclamptic pregnant women (103 HIV uninfected preeclamptics, 77 HIV infected preeclamptics). Preeclamptic pregnant women were further sub-grouped into 79 early-onset preeclampsia (EOPE) (40 HIV uninfected EOPE, 39 HIV infected EOPE) and 101 late-onset preeclampsia (LOPE) (63 HIV uninfected LOPE, 38 HIV infected LOPE). Genotyping of complement C1q gene polymorphisms (rs292001 and rs294183) was detected using a TaqMan® SNP Genotyping assay from purified DNA. RESULTS: No significant differences in allelic and genotype frequencies of rs292001 and rs294183 between preeclamptic and normotensive women were observed. Likewise, there were no significant differences in allelic and genotype frequencies between HIV infected normotensive vs HIV infected preeclampsia and HIV uninfected normotensive vs HIV uninfected preeclampsia for both SNPs. However, the odds ratio of preeclamptic women having the GA genotype was 1:2. CONCLUSION: We demonstrate that SNPs of the C1q gene (rs292001 and rs294183) are not associated with the pathogenesis of PE development in women of African ancestry. The role ofC1qrs292001 heterozygous GA is highlighted (with and without HIV infection) may affect susceptibility to PE development. Notably, this dysregulation may affect C1q translation and protein output thus influencing the downstream role of the complement system and functional immunology in HIV infection comorbid with PE.


Subject(s)
HIV Infections , Pre-Eclampsia , Humans , Female , Pregnancy , HIV Infections/genetics , HIV Infections/complications , Complement C1q/genetics , Polymorphism, Single Nucleotide , Case-Control Studies
7.
Comput Intell Neurosci ; 2022: 4690936, 2022.
Article in English | MEDLINE | ID: mdl-35571706

ABSTRACT

A key challenge in clinical recommendation systems is the problem of aberrant patient profiles in social networks. As a result of a person's abnormal profile, numerous vests might be used to make fake remarks about them, cyber bullying, or cyber-attacks. Many clinical researchers have done extensive study on this topic. The most recent studies on this topic are summarized, and an overarching framework is provided. When it comes to the methods and datasets that make up the data collection, the feature presentation and algorithm selection layers provide an overview of the various types of algorithm selections available. The categorization and evaluation of diseases and disorders has been one of the major advantages of machine learning in medical. Because it was harder to predict, it rendered it more controllable. It might range from difficult-to-find cancers in the early stages to certain other illnesses spread through the bloodstream. In healthcare, we may pick methods in machine learning depending on reliable outcomes. To do so, we must run the findings through each method. The major issue arises during information training and validation. Because the dataset is so large, eliminating mistakes might be difficult. The providers, other characteristics, various algorithms, data labelling techniques, and assessment criteria are all presented and contrasted in depth. Detecting anomalous users in medical social networks, on the other hand, is a work in progress. The result evaluation layer provides an explanation of how to evaluate and mark up the results of the various algorithm selection layers. Finally, it looks forward to more study in this area.


Subject(s)
Machine Learning , Privacy , Algorithms , Delivery of Health Care , Humans , Social Networking
8.
Genes (Basel) ; 13(5)2022 04 29.
Article in English | MEDLINE | ID: mdl-35627186

ABSTRACT

Although metabolic acidosis is associated with numerous pathophysiological conditions and its vasorelaxation effects have been well described in different animal and culture models, the molecular mechanisms of acidosis-induced vasorelaxation are not fully understood. Mesenteric artery models have been used extensively to examine the vascular response to various pathophysiological conditions. Our previous studies and several other reports have suggested the vascular responses of goat mesenteric arteries and human arteries to various stimuli, including acidic stress, are highly similar. In this study, to further identify the signaling molecules responsible for altered vasoreactivity in response to acidic pH, we examined the proteomic profile of acid stress-induced vasorelaxation using a goat mesenteric artery model. The vascular proteomes under acidic pH were compared using 2D-GE with 7 cm IPG strips and mini gels, LC-MS/MS, and MALDI TOF MS. The unique proteins identified by mass spectroscopy were actin, transgelin, WD repeat-containing protein 1, desmin, tropomyosin, ATP synthase ß, Hsp27, aldehyde dehydrogenase, pyruvate kinase, and vitamin K epoxide reductase complex subunit 1-like protein. Out of five protein spots identified as actin, three were upregulated > 2-fold. ATP synthase ß was also upregulated (2.14-fold) under acid stress. Other actin-associated proteins upregulated were transgelin, desmin, and WD repeat-containing protein 1. Isometric contraction studies revealed that both receptor-mediated (histamine) and non-receptor-mediated (KCl) vasocontraction were attenuated, whereas acetylcholine-induced vasorelaxation was augmented under acidosis. Overall, the altered vasoreactivity under acidosis observed in the functional studies could possibly be attributed to the increase in expression of actin and ATP synthase ß.


Subject(s)
Acidosis , Vasodilation , Acidosis/metabolism , Actins/metabolism , Adenosine Triphosphate/metabolism , Animals , Chromatography, Liquid , Desmin/metabolism , Mesenteric Arteries/metabolism , Nitric Oxide Synthase , Proteomics , Tandem Mass Spectrometry
9.
Genes (Basel) ; 13(3)2022 03 12.
Article in English | MEDLINE | ID: mdl-35328060

ABSTRACT

Hypoxic ischemic injury to the fetal and neonatal brain is a leading cause of death and disability worldwide. Although animal and culture studies suggest that glutamate excitotoxicity is a primary contributor to neuronal death following hypoxia, the molecular mechanisms, and roles of various neural cells in the development of glutamate excitotoxicity in humans, is not fully understood. In this study, we developed a culture model of human fetal neural stem cell (FNSC)-derived astrocytes and examined their glutamate uptake in response to hypoxia. We isolated, established, and characterized cultures of FNSCs from aborted fetal brains and differentiated them into astrocytes, characterized by increased expression of the astrocyte markers glial fibrillary acidic protein (GFAP), excitatory amino acid transporter 1 (EAAT1) and EAAT2, and decreased expression of neural stem cell marker Nestin. Differentiated astrocytes were exposed to various oxygen concentrations mimicking normoxia (20% and 6%), moderate and severe hypoxia (2% and 0.2%, respectively). Interestingly, no change was observed in the expression of the glutamate transporter EAAT2 or glutamate uptake by astrocytes, even after exposure to severe hypoxia for 48 h. These results together suggest that human FNSC-derived astrocytes can maintain glutamate uptake after hypoxic injury and thus provide evidence for the possible neuroprotective role of astrocytes in hypoxic conditions.


Subject(s)
Astrocytes , Glutamic Acid , Neural Stem Cells , Astrocytes/metabolism , Cell Hypoxia , Cells, Cultured , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Glutamic Acid/metabolism , Humans , Neural Stem Cells/metabolism
10.
Mater Today Proc ; 56: 3556-3560, 2022.
Article in English | MEDLINE | ID: mdl-34900608

ABSTRACT

Past couple of years, the world is going through one of the biggest pandemic named COVID-19. In the mid of year 2019, it is a very difficult process to predict the COVID-19 just by viewing the images. Later on AI based technology has done a significant role in the prediction of COVID-19 through biomedical images such as CT scan, X ray etc. This study also implemented the deep learning model for the prediction of COVID-19 through X-ray images. The implemented model is termed as XR-CAPS which consist of two models such as U-Net model and the capsule network. The U Net model is used for performing the segmentation of the images and the capsule networks are applied for performing the feature extraction. The XR-CAPS model is applied on the X-ray images for the prediction of COVID-19 and the evaluation of the model is done by three parameters that are accuracy, sensitivity and specificity. The model is compared with other existing models like ResNet50, DenseNet121 and DenseCapsNet, this has achieved an accuracy of 93.2%, sensitivity of 94% and specificity of 97.1% which is better than other states of the art algorithms.

11.
Biomolecules ; 11(7)2021 07 20.
Article in English | MEDLINE | ID: mdl-34356686

ABSTRACT

Vascular endothelial growth factor (VEGF) is an angiogenic growth factor that acts primarily on endothelial cells, but numerous studies suggest that VEGF also acts on non-endothelial cells, including trophoblast cells. Inhibition of VEGF signaling by excess production of the endogenous soluble VEGF receptor sFlt1 in trophoblast cells has been implicated in several pregnancy complications. Our previous studies and other reports have shown that VEGF directly regulates placental vascular development and functions and that excess VEGF production adversely affects placental vascular development. Trophoblast giant cells (TGCs) line the maternal side of the placental vasculature in mice and function like endothelial cells. In this study, we specifically examined the effect of excess VEGF signaling on TGC development associated with defective placental vascular development using two mouse models an endometrial VEGF overexpression model and a placenta-specific sFlt1 knockdown model. Placentas of endometrial VEGF-overexpressing dams at embryonic days (E) 11.5 and 14.5 showed dramatic enlargement of the venous maternal spaces in junctional zones. The size and number of the parietal TGCs that line these venous spaces in the placenta were also significantly increased. Although junctional zone venous blood spaces from control and VEGF-overexpressing dams were not markedly different in size at E17.5, the number and size of P-TGCs were both significantly increased in the placentas from VEGF-overexpressing dams. In sFlt1 knockdown placentas, however, there was a significant increase in the size of the sinusoidal TGC-lined, alkaline phosphatase-positive maternal blood spaces in the labyrinth. These results suggest that VEGF signaling plays an important role in maintaining the homeostasis of the maternal vascular space in the mouse placenta through modulation of TGC development and differentiation, similar to the effect of VEGF on endothelial cells in other vascular beds.


Subject(s)
Placenta/blood supply , Placenta/cytology , Trophoblasts/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Differentiation , Endometrium/metabolism , Female , Giant Cells , Homeostasis , Male , Mice, Inbred Strains , Pregnancy , Trophoblasts/cytology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
12.
Biomolecules ; 10(6)2020 06 24.
Article in English | MEDLINE | ID: mdl-32599856

ABSTRACT

Preeclampsia (PE) is a serious pregnancy complication, affecting about 5-7% of pregnancies worldwide and is characterized by hypertension and damage to multiple maternal organs, primarily the liver and kidneys. PE usually begins after 20 weeks' gestation and, if left untreated, can lead to serious complications and lifelong disabilities-even death-in both the mother and the infant. As delivery is the only cure for the disease, treatment is primarily focused on the management of blood pressure and other clinical symptoms. The pathogenesis of PE is still not clear. Abnormal spiral artery remodeling, placental ischemia and a resulting increase in the circulating levels of vascular endothelial growth factor receptor-1 (VEGFR-1), also called soluble fms-like tyrosine kinase-1 (sFlt-1), are believed to be among the primary pathologies associated with PE. sFlt-1 is produced mainly in the placenta during pregnancy and acts as a decoy receptor, binding to free VEGF (VEGF-A) and placental growth factor (PlGF), resulting in the decreased bioavailability of each to target cells. Despite the pathogenic effects of increased sFlt-1 on the maternal vasculature, recent studies from our laboratory and others have strongly indicated that the increase in sFlt-1 in PE may fulfill critical protective functions in preeclamptic pregnancies. Thus, further studies on the roles of sFlt-1 in normal and preeclamptic pregnancies are warranted for the development of therapeutic strategies targeting VEGF signaling for the treatment of PE. Another impediment to the treatment of PE is the lack of suitable methods for delivery of cargo to placental cells, as PE is believed to be of placental origin and most available therapies for PE adversely impact both the mother and the fetus. The present review discusses the pathogenesis of PE, the complex role of sFlt-1 in maternal disease and fetal protection, and the recently developed placenta-targeted drug delivery system for the potential treatment of PE with candidate therapeutic agents.


Subject(s)
Placenta/drug effects , Placenta/pathology , Pre-Eclampsia/drug therapy , Pre-Eclampsia/pathology , Female , Humans , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Vascular Endothelial Growth Factor Receptor-1/metabolism
13.
Biomolecules ; 10(1)2020 01 14.
Article in English | MEDLINE | ID: mdl-31947662

ABSTRACT

The human endometrium undergoes sequential phases of shedding of the upper functionalis zone during menstruation, followed by regeneration of the functionalis zone from the remaining basalis zone cells, and secretory differentiation under the influence of the ovarian steroid hormones estradiol (E2) and progesterone (P4). This massive tissue regeneration after menstruation is believed to arise from endometrial stromal and epithelial stem cells residing in the basal layer of the endometrium. Although many endometrial pathologies are thought to be associated with defects in these stem cells, studies on their identification and regulation are limited, primarily due to lack of easily accessible animal models, as these processes are unique to primates. Here we describe a robust new method to study endometrial regeneration and differentiation processes using human endometrial tissue slice cultures incorporating an air-liquid interface into a 3D matrix scaffold of type I collagen gel, allowing sustained tissue viability over three weeks. The 3D collagen gel-embedded endometrial tissue slices in a double-dish culture system responded to ovarian steroid hormones, mimicking the endometrial changes that occur in vivo during the menstrual cycle. These changes included the E2-induced upregulation of Ki-67, estrogen receptor (ER), and progesterone receptor (PR) in all endometrial compartments and were markedly suppressed by both P4 and E2 plus P4 treatments. There were also distinct changes in endometrial morphology after E2 and P4 treatments, including subnuclear vacuolation and luminal secretions in glands as well as decidualization of stromal cells, typical characteristics of a progestational endometrium in vivo. This long-term slice culture method provides a unique in vivo-like microenvironment for the study of human endometrial functions and remodeling during early pregnancy and experiments on stem cell populations involved in endometrial regeneration and remodeling. Furthermore, this model has the potential to enable studies on several endometrial diseases, including endometrial cancers and pregnancy complications associated with defects in endometrial remodeling.


Subject(s)
Endometrium/physiology , Tissue Culture Techniques/methods , Cell Differentiation , Cell Survival , Endometrium/cytology , Endometrium/ultrastructure , Equipment Design , Female , Humans , Regeneration , Tissue Culture Techniques/instrumentation
14.
Arch Immunol Ther Exp (Warsz) ; 67(5): 295-309, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31286151

ABSTRACT

Macrophages (MФs) are the leukocytes produced from differentiation of monocytes and are located in almost all tissues of human body. They are involved in various processes, such as phagocytosis, innate and adaptive immunity, proinflammatory (M1) and anti-inflammatory (M2) activity, depending on the tissue microenvironment. They play a crucial role in pregnancy, and their dysfunction or alteration of polarity is involved in pregnancy disorders, like preeclampsia, recurrent spontaneous abortion, infertility, intrauterine growth restriction, and preterm labor. About 50-60% of decidual leukocytes are natural killer (NK) cells followed by MФs (the second largest population). MФs are actively involved in trophoblast invasion, tissue and vascular remodeling during early pregnancy, besides their role as major antigen-presenting cells in the decidua. These cells have different phenotypes and polarities in different stages of pregnancy. They have also been observed to enhance tumor growth by their anti-inflammatory activity (M2 type) and prevent immunogenic rejection. Targeted alteration of polarity (M1-M2 or vice versa) could be a major focus in the future treatment of pregnancy complications. This review is focused on the role of MФs in pregnancy, their involvement in pregnancy disorders, and decidual MФs as possible therapeutic targets for the treatment of pregnancy complications.


Subject(s)
Decidua/immunology , Macrophages/physiology , Pregnancy Complications/immunology , Pregnancy/immunology , Animals , Embryo Implantation/immunology , Female , Humans , Immune Tolerance , Macrophages/immunology , Macrophages/pathology , Pregnancy Complications/pathology , Vascular Endothelial Growth Factors/metabolism
15.
Placenta ; 75: 1-8, 2019 01.
Article in English | MEDLINE | ID: mdl-30712660

ABSTRACT

INTRODUCTION: Statins induce heme oxygenase-1 (HO-1) expression in vitro and in vivo. Low HO-1 expression is associated with pregnancy complications, e.g. preeclampsia and recurrent miscarriages. Here, we investigated the effects of pravastatin on HO-1 expression, placental development, and fetal survival in mice with a partial HO-1 deficiency. METHODS: At E14.5, untreated pregnant wild-type (WT, n=13-18), untreated HO-1+/- (Het, n=6-9), and Het mice treated with pravastatin (Het+Pravastatin, n=12-14) were sacrificed. Numbers of viable fetuses/resorbed concepti were recorded. Maternal livers and placentas were harvested for HO activity. Hematoxylin and eosin (H&E) and CD31 immunohistochemical staining were performed on whole placentas. RESULTS: Compared with WT, HO activity in Het livers (65±18%, P<0.001) and placentas (74±7%, P<0.001) were significantly decreased. Number of viable fetuses per dam was significantly lower in Untreated Het dams (6.0±2.2) compared with WT (9.1±1.4, P<0.01), accompanied by a higher relative risk (RR) for concepti resorption (17.1, 95% CI 4.0-73.2). In Hets treated with pravastatin, maternal liver and placental HO activity increased, approaching levels of WT controls (to 83±7% and 87±14%, respectively). The number of viable fetuses per dam increased to 7.7±2.5 with a decreased RR for concepti resorption (2.7, 95% CI 1.2-5.9). In some surviving Untreated Het placentas, there were focal losses of cellular architecture and changes suggestive of reduced blood flow in the labyrinth. These findings were absent in Het+Pravastatin placentas. DISCUSSION: Pravastatin induces maternal liver and placental HO activity, may affect placental function and improve fetal survival in the context of a partial deficiency of HO-1.


Subject(s)
Heme Oxygenase-1/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Membrane Proteins/metabolism , Placentation/drug effects , Pravastatin/therapeutic use , Pre-Eclampsia/prevention & control , Animals , Drug Evaluation, Preclinical , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver/drug effects , Liver/enzymology , Mice , Pravastatin/pharmacology , Pregnancy , Random Allocation
16.
J Vis Exp ; (139)2018 09 10.
Article in English | MEDLINE | ID: mdl-30247484

ABSTRACT

No effective treatments currently exist for placenta-associated pregnancy complications, and developing strategies for the targeted delivery of drugs to the placenta while minimizing fetal and maternal side effects remains challenging. Targeted nanoparticle carriers provide new opportunities to treat placental disorders. We recently demonstrated that a synthetic placental chondroitin sulfate A binding peptide (plCSA-BP) could be used to guide nanoparticles to deliver drugs to the placenta. In this protocol, we describe in detail a system for assessing the efficiency of drug delivery to the placenta by plCSA-BP that employs three separate methods used in combination: in vivo imaging, high-frequency ultrasound (HFUS), and high-performance liquid chromatography (HPLC). Using in vivo imaging, plCSA-BP-guided nanoparticles were visualized in the placentas of live animals, while HFUS and HPLC demonstrated that plCSA-BP-conjugated nanoparticles efficiently and specifically delivered methotrexate to the placenta. Thus, a combination of these methods can be used as an effective tool for the targeted delivery of drugs to the placenta and development of new treatment strategies for several pregnancy complications.


Subject(s)
Drug Delivery Systems/adverse effects , Drug Delivery Systems/methods , Placenta/metabolism , Safety , Chondroitin Sulfates/metabolism , Drug Carriers/chemistry , Female , Humans , Methotrexate/metabolism , Nanoparticles/chemistry , Peptides/chemistry , Peptides/metabolism , Pregnancy
17.
Theranostics ; 8(10): 2765-2781, 2018.
Article in English | MEDLINE | ID: mdl-29774074

ABSTRACT

Rationale: The availability of therapeutics to treat pregnancy complications is severely lacking, mainly due to the risk of harm to the fetus. In placental malaria, Plasmodium falciparum-infected erythrocytes (IEs) accumulate in the placenta by adhering to chondroitin sulfate A (CSA) on the surfaces of trophoblasts. Based on this principle, we have developed a method for targeted delivery of payloads to the placenta using a synthetic placental CSA-binding peptide (plCSA-BP) derived from VAR2CSA, a CSA-binding protein expressed on IEs. Methods: A biotinylated plCSA-BP was used to examine the specificity of plCSA-BP binding to mouse and human placental tissue in tissue sections in vitro. Different nanoparticles, including plCSA-BP-conjugated nanoparticles loaded with indocyanine green (plCSA-INPs) or methotrexate (plCSA-MNPs), were administered intravenously to pregnant mice to test their efficiency at drug delivery to the placenta in vivo. The tissue distribution and localization of the plCSA-INPs were monitored in live animals using an IVIS imaging system. The effect of plCSA-MNPs on fetal and placental development and pregnancy outcome were examined using a small-animal high-frequency ultrasound (HFUS) imaging system, and the concentrations of methotrexate in fetal and placental tissues were measured using high-performance liquid chromatography (HPLC). Results: plCSA-BP binds specifically to trophoblasts and not to other cell types in the placenta or to CSA-expressing cells in other tissues. Moreover, we found that intravenously administered plCSA-INPs accumulate in the mouse placenta, and ex vivo analysis of the fetuses and placentas confirmed placenta-specific delivery of these nanoparticles. We also demonstrate successful delivery of methotrexate specifically to placental cells by plCSA-BP-conjugated nanoparticles, resulting in dramatic impairment of placental and fetal development. Importantly, plCSA-MNPs treatment had no apparent adverse effects on maternal tissues. Conclusion: These results demonstrate that plCSA-BP-guided nanoparticles could be used for the targeted delivery of payloads to the placenta and serve as a novel placenta-specific drug delivery option.


Subject(s)
Nanoparticles/metabolism , Trophoblasts/metabolism , Animals , Antigens, Protozoan/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Methotrexate/administration & dosage , Methotrexate/pharmacokinetics , Mice , Nanoparticles/adverse effects , Pregnancy
18.
Proc Natl Acad Sci U S A ; 115(17): E4120-E4129, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29632208

ABSTRACT

Seeds employ sensory systems that assess various environmental cues over time to maximize the successful transition from embryo to seedling. Here we show that the Arabidopsis F-BOX protein COLD TEMPERATURE-GERMINATING (CTG)-10, identified by activation tagging, is a positive regulator of this process. When overexpressed (OE), CTG10 hastens aspects of seed germination. CTG10 is expressed predominantly in the hypocotyl, and the protein is localized to the nucleus. CTG10 interacts with PHYTOCHROME-INTERACTING FACTOR 1 (PIF1) and helps regulate its abundance in plantaCTG10-OE accelerates the loss of PIF1 in light, increasing germination efficiency, while PIF1-OE lines fail to complete germination in darkness, which is reversed by concurrent CTG10-OE Double-mutant (pif1 ctg10) lines demonstrated that PIF1 is epistatic to CTG10. Both CTG10 and PIF1 amounts decline during seed germination in the light but reaccumulate in the dark. PIF1 in turn down-regulates CTG10 transcription, suggesting a feedback loop of CTG10/PIF1 control. The genetic, physiological, and biochemical evidence, when taken together, leads us to propose that PIF1 and CTG10 coexist, and even accumulate, in the nucleus in darkness, but that, following illumination, CTG10 assists in reducing PIF1 amounts, thus promoting the completion of seed germination and subsequent seedling development.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Germination/physiology , Seeds/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Kelch Repeat , Seeds/genetics
19.
PLoS One ; 13(1): e0191040, 2018.
Article in English | MEDLINE | ID: mdl-29324807

ABSTRACT

It is increasingly evident that cytokines and growth factors produced in the decidua play a pivotal role in the regulation of the local immune microenvironment and the establishment of pregnancy. One of the major growth factors produced in the decidua is vascular endothelial growth factor (VEGF), which acts not only on endothelial cells, but also on multiple other cell types, including macrophages. We sought to determine whether decidua-derived VEGF affects macrophage recruitment and polarization using human endometrial/decidual tissue samples, primary human endometrial stromal cells (ESCs), and the human monocyte cell line THP1. In situ hybridization was used for assessment of local VEGF expression and immunohistochemistry was used for identification and localization of CD68-positive endometrial macrophages. Macrophage migration in culture was assessed using a transwell migration assay, and the various M1/M2 phenotypic markers and VEGF expression were assessed using quantitative real-time PCR (qRT-PCR). We found dramatic increases in both VEGF levels and macrophage numbers in the decidua during early pregnancy compared to the secretory phase endometrium (non-pregnant), with a significant increase in M2 macrophage markers, suggesting that M2 is the predominant macrophage phenotype in the decidua. However, decidual samples from preeclamptic pregnancies showed a significant shift in macrophage phenotype markers, with upregulation of M1 and downregulation of M2 markers. In THP1 cultures, VEGF treatment significantly enhanced macrophage migration and induced M1 macrophages to shift to an M2 phenotype. Moreover, treatment with conditioned media from decidualized ESCs induced changes in macrophage migration and polarization similar to that of VEGF treatment. These effects were abrogated by the addition of a potent VEGF inhibitor. Together these results suggest that decidual VEGF plays a significant role in macrophage recruitment and M2 polarization, and that inhibition of VEGF signaling may contribute to the shift in macrophage polarity observed in different pregnancy disorders, including preeclampsia.


Subject(s)
Cell Polarity , Decidua/cytology , Macrophages/cytology , Vascular Endothelial Growth Factor A/physiology , Adult , Cell Line , Female , Humans , Real-Time Polymerase Chain Reaction
20.
BMC Genomics ; 18(1): 617, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28810828

ABSTRACT

BACKGROUND: High ambient temperature is known to affect fish gonadal development and physiology in a variety of ways depending on the severity and duration of exposure; however, the underlying molecular mechanisms are poorly understood. Gonadal gene expression influence the gonadal development, physiology and the quality of egg/sperm produced in teleosts and the mechanistic understanding of spatio-temporal changes in the gonadal gene expression could be instrumental in controlling the fate of egg/sperm and the quality of seed produced. Real time-quantititative polymerase chain reaction (RT-qCR), is a high throughput, sensitive and reproducible methodology used for understanding gene expression patterns by measuring the relative abundance of mRNA transcripts. However, its accuracy relies upon a suitable reference gene whose expression levels remain stable across various experimental conditions. In the present study, we evaluated the suitability of ten potential reference genes to be used as internal controls in RT-qPCR analysis in gonadal tissues (ovary and testis) of minnow Puntius sophore exposed to high temperature stress for different time periods (7 days, 60 days). Expression analysis of ten different constitutively expressed genes viz. 18S ribosomal RNA (18S rRNA), beta actin (ßactin), ß-2 microglobulin (b2mg), eukaryotic elongation factor-1 (eef1), glyceraldehyde-3phosphate dehydrogenase (gapdh), glucose-6-phosphate dehydrogenase (g6pd), ribosomal binding protein L13 (rpl13), tubulin (tub), tata box binding protein (tbp), ubiquitin (ubi) was carried out by using RT-qPCR and the stability in their expressions were evaluated by using four different algorithms; namely, delta Ct, BestKeeper, geNorm and NormFinder. RESULTS: In ovary, eef1 was found to be the most suitable reference gene in all the algorithms used. In testis, b2mg was found to be the most suitable reference gene in delta Ct, BestKeeper, NormFinder analysis while tbp and eef1 were found to be the most suitable reference genes in geNorm analysis. CONCLUSIONS: In conclusion, eef1 and b2mg were found to be the most suitable reference genes in ovary and testis, respectively, of Puntius sophore exposed to high temperature stress, and could be used as internal controls for gene expression analysis in gonadal tissues of Puntius sophore.


Subject(s)
Cyprinidae/genetics , Gene Expression Profiling/standards , Heat-Shock Response/genetics , Ovary/metabolism , Real-Time Polymerase Chain Reaction/standards , Testis/metabolism , Animals , Cyprinidae/physiology , Female , Male , Reference Standards , Reproduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL