Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
2.
Placenta ; 129: 84-86, 2022 11.
Article in English | MEDLINE | ID: mdl-36270043

ABSTRACT

During the first trimester of pregnancy, there is a large decrease in systemic vascular resistance (SVR) which coincides temporally with increasing extrusion of extracellular vesicles (EVs) from the placenta. We hypothesized that placental EVs may be one of the mechanisms contributing to maternal vasodilation. Macro-, micro-, nano-EVs from human first trimester placenta, or control injections containing EVs derived from fresh culture media, were injected into pregnant mice at day 12.5. After 30 min or 24 h, second order resistance arteries assessed for their reactivity to various vasomodulators. Placental EVs induced an anti-constrictive, pro-vasodilatory effect in maternal resistance arteries compared to control injections after 24 h suggesting that placental EVs may contribute to the maternal vasodilation during pregnancy.


Subject(s)
Extracellular Vesicles , Placenta , Pregnancy , Female , Humans , Mice , Animals , Pregnancy Trimester, First , Vasodilation , Arteries
3.
Int J Pharm ; 626: 122152, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36055442

ABSTRACT

Notwithstanding the growing evidence of improved drug delivery efficiency to the brain by ligand modification of PEGylated liposomes, the comprehensive knowledge of their transport processes and payload across the BBB is yet to be revealed. Herein, this study sought to understand the glutathione (GSH) ligand effect on transcellular transport mechanisms of liposomes through the blood-brain barrier (BBB) by comparing PEGylated liposomes (PEG-L) and GSH PEGylated liposomes (GSH-PEG-L). Endocytosis and exocytosis of liposomes including the role of secreted extracellular vesicles (EVs) of brain endothelial cells (BECs) were assessed. Furthermore, pharmacokinetics and brain distribution analysis of gemcitabine loaded liposomes were carried out in healthy rats to ascertain the in vivo applicability. Our findings suggested that the presence of GSH increased the cellular uptake of liposomes by up to 3-fold in human brain microvascular endothelial cells depending on the dose but not in astrocytes. The cell exposure to liposomes particularly GSH-PEG-L dramatically increased the cell secretion of small and microvesicles with liposomal components, though different liposomes preferred different vesicles for exocytosis. This correlated with GSH-PEG-L transport efficiency of 4 % across the in vitro BBB model in 24 h, 1.7-fold higher than that of PEG-L (p < 0.05). In rats, while PEG-L and GSH-PEG-L showed similar pharmacokinetic profiles and prolonged circulation properties, 3.8 % of the total injected dose (ID) of gemcitabine was found in the brain of the GSH-PEG-L group at 8 h post-injection, compared with 2.8 % ID in the PEG-L group. A brain: blood concentration ratio of 1.27 ± 0.12 indicated that an active transport mechanism to cross the BBB for GSH-PEG-L. Overall, this study revealed that GSH augmented the transcellular transport efficiency of liposomes through BBB to improve targeted brain delivery by enhancing cellular uptake and vesicular exocytosis route of BECs.


Subject(s)
Blood-Brain Barrier , Liposomes , Animals , Brain , Endothelial Cells , Glutathione , Humans , Ligands , Polyethylene Glycols , Rats , Tissue Distribution , Transcytosis
4.
Int J Pharm ; 626: 122153, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36055444

ABSTRACT

Small extracellular vesicles (sEVs) have emerged as attractive drug delivery systems. However, the intracellular release of their cargoes is restricted. This study aimed to develop an efficient approach to re-engineer sEVs by hybridisation with pH-sensitive liposomes (PSLs) and investigate their endosome escape potential. MIA PaCa-2 cell-derived sEVs and PSLs were fused via three methods, and fusion efficiency (FE) was measured using a fluorescence resonance energy transfer assay and nanoparticle tracking analysis. Cellular uptake, intracellular trafficking, and cytotoxicity of doxorubicin-loaded vesicles (Dox@hybrids, Dox@sEVs, and Dox@PSLs) were investigated on MIA PaCa-2 cells. Among the three methods, Ca2+-mediated fusion was the simplest and led to a comparable FE with freeze-thaw method, which was significantly higher than PEG8000-mediated fusion. sEVs were more stable after hybridisation with PSLs. Confocal microscopy revealed that the hybrids internalised more efficiently than natural sEVs. While the internalised Dox@sEVs were primarily co-localised with endo/lysosomes even after 8 h, Dox from Dox@hybrids was found to escape from endosomes by 2 h and homogenously distributed in the cytosol before accumulated at nucleus, corresponding to the in vitro pH-responsive release profile. Consequently, Dox@hybrids enhanced cytotoxicity compared with Dox@sEVs, Dox@PSLs, or free drugs. Overall, the biomimetic nanosystem generated by simple Ca2+-mediated fusion was more stable and demonstrated higher efficiencies of cellular uptake and endosome escape compared to natural sEVs.


Subject(s)
Extracellular Vesicles , Liposomes , Doxorubicin/pharmacology , Drug Delivery Systems , Endosomes
5.
Elife ; 112022 08 03.
Article in English | MEDLINE | ID: mdl-35920626

ABSTRACT

Single-cell technologies (RNA-sequencing, flow cytometry) are critical tools to reveal how cell heterogeneity impacts developmental pathways. The placenta is a fetal exchange organ, containing a heterogeneous mix of mesenchymal cells (fibroblasts, myofibroblasts, perivascular, and progenitor cells). Placental mesenchymal stromal cells (pMSC) are also routinely isolated, for therapeutic and research purposes. However, our understanding of the diverse phenotypes of placental mesenchymal lineages, and their relationships remain unclear. We designed a 23-colour flow cytometry panel to assess mesenchymal heterogeneity in first-trimester human placentae. Four distinct mesenchymal subsets were identified; CD73+CD90+ mesenchymal cells, CD146+CD271+ perivascular cells, podoplanin+CD36+ stromal cells, and CD26+CD90+ myofibroblasts. CD73+CD90+ and podoplanin + CD36+ cells expressed markers consistent with cultured pMSCs, and were explored further. Despite their distinct ex-vivo phenotype, in culture CD73+CD90+ cells and podoplanin+CD36+ cells underwent phenotypic convergence, losing CD271 or CD36 expression respectively, and homogenously exhibiting a basic MSC phenotype (CD73+CD90+CD31-CD144-CD45-). However, some markers (CD26, CD146) were not impacted, or differentially impacted by culture in different populations. Comparisons of cultured phenotypes to pMSCs further suggested cultured pMSCs originate from podoplanin+CD36+ cells. This highlights the importance of detailed cell phenotyping to optimise therapeutic capacity, and ensure use of relevant cells in functional assays.


Subject(s)
Dipeptidyl Peptidase 4 , Mesenchymal Stem Cells , Adapalene/metabolism , Biomarkers/metabolism , CD146 Antigen/genetics , CD146 Antigen/metabolism , Cell Differentiation/physiology , Cells, Cultured , Dipeptidyl Peptidase 4/metabolism , Female , Flow Cytometry , Humans , Mesenchymal Stem Cells/metabolism , Phenotype , Placenta/metabolism , Pregnancy , Pregnancy Trimester, First , Thy-1 Antigens/metabolism
6.
Placenta ; 125: 1, 2022 07.
Article in English | MEDLINE | ID: mdl-35660100
7.
Placenta ; 123: 24-30, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35533511

ABSTRACT

INTRODUCTION: Extracellular vesicles are now believed to be important mediators of placental-maternal communication. However, little is known about the formation of extracellular vesicles by human placenta. This study uses nanoscale three-dimensional imaging to investigate how and where placental extracellular vesicles form. METHODS: Term and first trimester human placental villi were imaged by serial block face scanning electron microscopy. These images were analysed to quantify vesicle surface density. Segmentation was performed to reconstruct three-dimensional images of extracellular vesicles. Live imaging light microscopy of first trimester villous explants was performed. RESULTS: Vesicles were observed on the tips of placental microvilli in term and first trimester placenta. In term placenta these microvillous tip vesicles had a median size of 0.55 µm and their surface area density exceeded 22000 per mm2. Microvillous tip vesicle membranes had a lower electron density than the microvillous plasma membrane. Thirty seven percent of vesicles had a complex membrane structure including double membranes, internal vesicles and vesicle chains. Budding of smaller secondary vesicles from microvillous tip vesicle membranes was observed. Live imaging of a first trimester villus explant observed formation of vesicles which were larger but visually similar to the secondary vesicles observed by electron microscopy. DISCUSSION: These observations suggest that extracellular vesicles are forming on the tips of placental microvilli prior to release into maternal blood. However, it cannot be discounted that there are maternal extracellular vesicles that have bound to microvilli. In either case, the high surface area density of microvillous tip vesicles is consistent with an important role in placental-maternal signalling.


Subject(s)
Extracellular Vesicles , Placenta , Chorionic Villi , Female , Humans , Microvilli , Placenta/metabolism , Pregnancy , Pregnancy Trimester, First
8.
J Hum Hypertens ; 36(2): 192-200, 2022 02.
Article in English | MEDLINE | ID: mdl-33686209

ABSTRACT

Endothelial cell dysfunction in pregnancy, which can be induced by placental factors, is the fundamental component of the pathogenesis of pre-eclampsia. The dysfunctional vascular endothelium disrupts the balance of vasodilatory and vasoconstrictive factors, resulting in increasing blood pressure. There is currently no effective treatment for pre-eclampsia and effective control of hypertension may reduce neonatal morbidity and mortality by prolonging gestation, especially in cases of early onset disease. To date methyldopa, labetalol, nifedipine and metoprolol are recommended for controlling blood pressure in pre-eclampsia. All of these drugs have different mechanisms of action. In this in vitro study we investigated whether different types of anti-hypertensive drugs could have different effects on improving maternal endothelial cell dysfunction. Endothelial cells (HMEC-1) were exposed to phorbol-12-myristate-13-acetate (PMA) or pre-eclamptic sera or extracellular vesicles (EVs) derived from pre-eclamptic placentae, in the presence of each of the studied anti-hypertensive drugs (methyldopa, labetalol, nifedipine and metoprolol) or placebo for 24 h. Endothelial cell-surface adhesion molecule (ICAM-1) and monocyte adhesion were measured. The expression of cell-face ICAM-1 by HMEC-1 cells and THP-1 monocyte adherent to HMEC-1 that were exposed to three separate well-known activators of endothelial cells in the presence of four anti-hypertensive drugs was significantly reduced regardless of the dose. However, the effect on the reduction of ICAM-1 expression and monocyte adhesion was not significantly different between the four medications. Our data suggest that the beneficial effect on improving endothelial cell function by these commonly prescribed anti-hypertensive drugs is seemingly independent of the anti-hypertensive mechanisms of the medication.


Subject(s)
Labetalol , Pre-Eclampsia , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Endothelial Cells , Female , Humans , Labetalol/pharmacology , Labetalol/therapeutic use , Placenta/metabolism , Pregnancy
9.
Placenta ; 115: 115-120, 2021 11.
Article in English | MEDLINE | ID: mdl-34600275

ABSTRACT

INTRODUCTION: To investigate the role of placental extracellular vesicles (EVs), especially in pathological pregnancy, the use of freshly isolated EVs is often limited due to the sporadic and unpredictable availability of placental samples. Therefore, it is important to understand and use optimised storage conditions for placental EVs. In this study, we investigated different conditions for the short-term storage of placental micro- and nano-EVs and examined their biological activity. METHODS: Placental EVs were collected from first trimester placentae. EVs were suspended in PBS and aliquoted, and then stored for up to 14 days at room temperature, 4 °C or -20 °C. Total protein and DNA levels were measured at various time points. The ability of stored placental EVs to alter endothelial cell activation was quantified by monocyte adhesion assays. RESULTS: There was no difference in the concentration of placental micro- or nano-EVs between each time point, when stored at either room temperature or 4 °C. However, there was a significant loss of placental EVs after storage at -20 °C. There was no difference in protein or DNA levels of placental EVs when stored at either room temperature or 4 °C. Biological activity of placental EVs was retained for up to 14 days at either room temperature or 4 °C measured by monocyte adhesion assays. DISCUSSION: We have shown that placental micro- and nano-EVs are stable and retain biological activities following storage in PBS or media for 14 days at either room temperature or 4 °C.


Subject(s)
Extracellular Vesicles/physiology , Placenta/ultrastructure , Tissue Preservation/methods , Cell-Derived Microparticles/physiology , Female , Gestational Age , Humans , Pregnancy , Temperature , Time Factors
10.
Placenta ; 116: 1, 2021 12.
Article in English | MEDLINE | ID: mdl-34629159
11.
Cells ; 10(8)2021 07 27.
Article in English | MEDLINE | ID: mdl-34440672

ABSTRACT

Preeclampsia, characterised by maternal endothelial cell activation, is triggered by toxic factors, such as placental extracellular vesicles (EVs) from a dysfunctional placenta. The increased oxidative stress seen in the preeclamptic placenta links to endoplasmic reticulum (ER) stress. The ER regulates protein folding and trafficking. When the ER is stressed, proteins are misfolded, and misfolded proteins are toxic. Misfolded proteins can be exported from cells, via EVs which target to other cells where the misfolded proteins may also be toxic. Melatonin is a hormone and antioxidant produced by the pineal gland and placenta. Levels of melatonin are reduced in preeclampsia. In this study we investigated whether melatonin treatment can change the nature of placental EVs that are released from a preeclamptic placenta. EVs were collected from preeclamptic (n = 6) and normotensive (n = 6) placental explants cultured in the presence or absence of melatonin for 18 h. Misfolded proteins were measured using a fluorescent compound, Thioflavin-T (ThT). Endothelial cells were exposed to placental EVs overnight. Endothelial cell activation was measured by the quantification of cell-surface ICAM-1 using a cell-based ELISA. EVs from preeclamptic placentae carried significantly (p < 0.001) more misfolded proteins than normotensive controls. Incubating preeclamptic placental explants in the presence of melatonin (1 µM and 10 µM) significantly (p < 0.001) reduced the misfolded proteins carried by EVs. Culturing endothelial cells in the presence of preeclamptic EVs significantly increased the expression of ICAM-1. This increased ICAM-1 expression was significantly reduced when the endothelial cells were exposed to preeclamptic EVs cultured in the presence of melatonin. This study demonstrates that melatonin reduces the amount of misfolded proteins carried by EVs from preeclamptic placentae and reduces the ability of these EVs to activate endothelial cells. Our study provides further preclinical support for the use of melatonin as a treatment for preeclampsia.


Subject(s)
Endothelial Cells/metabolism , Extracellular Vesicles/drug effects , Melatonin/pharmacology , Placenta/drug effects , Pre-Eclampsia/drug therapy , Adult , Cell Line , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Female , Humans , Intercellular Adhesion Molecule-1/metabolism , Placenta/metabolism , Placenta/pathology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Protein Folding , Secretory Pathway
12.
Pharm Res ; 38(2): 179-197, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33604783

ABSTRACT

Extracellular vesicles (EVs) are a heterogeneous population of lipid bilayer membrane-enclosed vesicles and act like 'messages in a bottle' in cell-cell communication by transporting their cargoes to recipient cells. Small EVs (sEVs, < 200 nm) are highly researched recently and have been harnessed as novel delivery systems for the treatment of various diseases, including neurodegenerative disorders, cardiovascular diseases, and most importantly cancer primarily because of their non-immunogenicity, tissue penetration and cell-tropism. This review will first provide a comprehensive overview of sEVs regarding the current understanding on their properties, biogenesis, new classification by the ISEV, composition, as well as their roles in cancer development (thereby called "oncosomes"). The primary focus will be given to the current state of sEVs as natural nanocarriers for cancer drug delivery, the technologies and challenges involved in sEV isolation and characterization, therapeutic cargo loading, and surface modification to enhance tumor-targeting. We will also provide examples of sEV products under clinical trials. Furthermore, the current challenges as well as the advance in "sEV mimetics" to address some of the sEVs limitations is briefly discussed. We seek to advance our understanding of sEVs to unlock their full potential as superior drug delivery vehicles in cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Extracellular Vesicles/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Clinical Trials as Topic , Humans , Particle Size , Treatment Outcome
13.
ACS Appl Mater Interfaces ; 12(35): 39005-39013, 2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32805904

ABSTRACT

There is a significant and growing research interest in the isolation of extracellular vesicles (EVs) from large volumes of biological samples and their subsequent concentration into clean and small volumes of buffers, especially for applications in medical diagnostics. Materials that are easily incorporated into simple sampling devices and which allow the release of EVs without the need for auxiliary and hence contaminating reagents are particularly in demand. Herein, we report on the design and fabrication of a flexible, microporous, electrochemically switchable cloth that addresses the key challenges in diagnostic applications of EVs. We demonstrate the utility of our electrochemically switchable substrate for the fast, selective, nondestructive, and efficient capture and subsequent release of EVs. The substrate consists of an electrospun cloth, infused with a conducting polymer and decorated with gold particles. Utilizing gold-sulfur covalent bonding, the electrospun substrates may be functionalized with SH-terminated aptamer probes selective to EV surface proteins. We demonstrate that EVs derived from primary human dermal fibroblast (HDFa) and breast cancer (MCF-7) cell lines are selectively captured with low nonspecific adsorption using an aptamer specific to the CD63 protein expressed on the EV membranes. The specific aptamer-EV interactions enable easy removal of the nonspecifically bound material through washing steps. The conducting polymer component of the cloth provides a means for efficient (>92%) and fast (<5 min) electrochemical release of clean and intact captured EVs by cathodic cleavage of the Au-S bond. We demonstrate successful capture of diluted EVs from a large volume sample and their release into a small volume of clean phosphate-buffered saline buffer. The developed cloth can easily be incorporated into different designs for separation systems and would be adaptable to other biological entities including cells and other EVs. Furthermore, the capture/release capability holds great promise for liquid biopsies if used to targeted disease-specific markers.


Subject(s)
Electrochemical Techniques/methods , Extracellular Vesicles/chemistry , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Line , Extracellular Vesicles/metabolism , Gold/chemistry , Humans , MCF-7 Cells , Polymers/chemistry , Porosity , Sulfur/chemistry , Tetraspanin 30/metabolism
14.
Placenta ; 94: 1-12, 2020 05.
Article in English | MEDLINE | ID: mdl-32217265

ABSTRACT

BACKGROUND: It has been reported that during the culture of human placental explants, the syncytiotrophoblast dies between 3 and 24 h and is then replaced within 48 h by a new syncytiotrophoblast layer formed by the fusion of underlying cytotrophoblasts. Most frequently the death of the syncytiotrophoblast is indicated by the uptake of nuclear stains such as propidium iodide (PI). This process is reportedly similar in both early and late gestation placental explants. METHODS: We cultured first trimester placental explants for up to 48 h and tested membrane intactness by exposure to PI. Connexin and pannexin mRNAs were quantified by RT-PCR and protein levels determined by immunofluorescence. The syncytiotrophoblast membrane leak was determined by culturing explants in the presence of hemichannel blockers. Extrusion of extracellular vesicles from the syncytiotrophoblast was quantified. RESULTS: Nuclei of the syncytiotrophoblast were stained with PI following approximately 4 h of culture and this was prevented by culturing the explants with pannexin-1 blockers. Expression of pannexin-1 hemichannels increased during explant culture (p = 0.0027). Extracellular vesicles were most abundantly extruded from the explants during the first 3 h of culture and the temporal pattern of extrusion was unaltered by blocking hemichannels. DISCUSSION: We show the mechanism of uptake of nuclear non-viability stains into the syncytiotrophoblast during explant culture is via upregulation of pannexin 1 hemichannels. Contrary to suggestions by some, the production of extracellular vesicles from cultured placental explants is not an in vitro artefact resulting from the apparent death of the syncytiotrophoblast in explant cultures.


Subject(s)
Cell Death/physiology , Connexins/genetics , Nerve Tissue Proteins/genetics , Placenta/physiology , Tissue Culture Techniques , Trophoblasts/physiology , Connexin 43/antagonists & inhibitors , Connexin 43/genetics , Connexin 43/physiology , Connexins/antagonists & inhibitors , Connexins/physiology , Extracellular Vesicles/metabolism , Female , Humans , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Pregnancy , Probenecid/pharmacokinetics , Propidium/metabolism , RNA, Messenger/analysis , Time Factors , Trophoblasts/chemistry , Up-Regulation
15.
Clin Sci (Lond) ; 134(5): 459-472, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32068238

ABSTRACT

Antiphospholipid autoantibodies (aPLs), a major maternal risk factor for preeclampsia, are taken into the syncytiotrophoblast where they bind intracellular vesicles and mitochondria. Subsequently, large quantities of extracellular vesicles (EVs) extruded from syncytiotrophoblast into the maternal circulation are altered such that they cause maternal endothelial cell activation. However, the mechanism driving this change is unknown. First trimester placental explants were treated with aPL for 18 h. The EVs were then collected by different centrifugation. The levels of HSP 70, misfolded proteins, caspase 8 activity, and Mixed Lineage Kinase domain-Like (MLKL) were measured in placental explants and EVs. In addition, the levels of TNF-α and CD95 in conditioned medium were also measured. Treating placental explants with aPL caused an increase in levels of HSP 70, misfolded proteins and MLKL in placental explants and EVs. Increased activity of caspase 8 was also seen in placental explants. Higher levels of TNF-α were seen conditioned medium from aPL-treated placental explant cultures. aPLs appear to induce endoplasmic reticulum stress in the syncytiotrophoblast in a manner that involved caspase 8 and TNF-α. To avoid accumulation of the associated misfolded proteins and MLKL, the syncytiotrophoblast exports these potentially dangerous proteins in EVs. It is likely that the dangerous proteins that are loaded into placental EVs in preeclampsia contribute to dysfunction of the maternal cells.


Subject(s)
Antibodies, Antiphospholipid/pharmacology , Endoplasmic Reticulum Stress/drug effects , Extracellular Vesicles/metabolism , Placenta/drug effects , Trophoblasts/drug effects , Caspase 8/metabolism , Female , HSP70 Heat-Shock Proteins/metabolism , Humans , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Pregnancy Trimester, First , Protein Kinases/metabolism , Tissue Culture Techniques , Trophoblasts/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Placenta ; 84: 9-13, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30773233

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialized topics. At IFPA meeting 2018 there were nine themed workshops, five of which are summarised in this report. These workshops discussed new perspectives and knowledge in the following areas of research: 1) preeclampsia; 2) abnormally invasive placenta; 3) placental infection; 4) gestational trophoblastic disease; 4) drug delivery to treat placental dysfunction.


Subject(s)
Drug Delivery Systems/methods , Gestational Trophoblastic Disease , Inflammation , Placenta Diseases , Pre-Eclampsia , Pregnancy Complications, Infectious , Animals , Biomedical Research/organization & administration , Biomedical Research/trends , Education/organization & administration , Education/standards , Female , Gestational Trophoblastic Disease/drug therapy , Gestational Trophoblastic Disease/etiology , Gestational Trophoblastic Disease/pathology , Gynecology/organization & administration , Gynecology/standards , Gynecology/trends , History, 21st Century , Humans , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Japan , Obstetrics/organization & administration , Obstetrics/standards , Obstetrics/trends , Placenta/drug effects , Placenta/metabolism , Placenta Diseases/drug therapy , Placenta Diseases/etiology , Placenta Diseases/pathology , Pre-Eclampsia/drug therapy , Pre-Eclampsia/etiology , Pre-Eclampsia/pathology , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/etiology , Pregnancy Complications, Infectious/pathology , Societies, Medical/organization & administration
17.
Placenta ; 84: 4-8, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30772008

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialized topics. At IFPA meeting 2018 there were nine themed workshops, four of which are summarized in this report. These workshops discussed new knowledge and technological innovations in the following areas of research: 1) viviparity in ocean-living species; 2) placental imaging; 3) epigenetics; and 4) extracellular vesicles in pregnancy.


Subject(s)
Aquatic Organisms/physiology , Epigenesis, Genetic/physiology , Extracellular Vesicles/physiology , Placenta/diagnostic imaging , Placentation/physiology , Pregnancy, Animal , Reproduction/physiology , Animals , Biomedical Research/organization & administration , Biomedical Research/trends , Education/organization & administration , Education/standards , Epigenomics , Female , Gynecology/organization & administration , Gynecology/standards , Gynecology/trends , History, 21st Century , Japan , Obstetrics/organization & administration , Obstetrics/standards , Obstetrics/trends , Oceans and Seas , Pregnancy , Prenatal Diagnosis/methods , Prenatal Diagnosis/veterinary , Societies, Medical/organization & administration
18.
Placenta ; 72-73: 41-47, 2018 12.
Article in English | MEDLINE | ID: mdl-30501880

ABSTRACT

BACKGROUND: The human placenta extrudes a variety of extracellular vesicles (EVs) into the maternal blood daily. These vesicles may be crucial to the adaptation of the maternal cardiovascular and immune systems to pregnancy. Quantifying the EVs that are released in early gestation is important to our understanding of how placental EVs may contribute to the regulation of maternal physiology. METHODS: EVs were isolated from first trimester placental explants and separated into micro- and nano-vesicles by differential centrifugation. The numbers of each type of EVs extruded from each milligram of placentae between gestational weeks 8 and 12 was determined by Nanoparticle Tracking Analysis. The total protein or DNA content of the vesicles was determined by BCA assay or Qubit® 2.0. RESULTS: Neither the number of micro- nor nano-EVs/mg explant (n = 49), nor the total protein (n = 19) and DNA content (n = 29) of these EVs changed significantly between 8 and 12 weeks of gestation. When the increasing placental weight with gestation was accounted for, the daily number of placental EVs extruded into the maternal blood increased by more than 100 fold between 8 and 12 weeks (micro-EVs 6.23 X 1014 and nano-EVs 1.84 X 1014 at 12 weeks, p = 0.0003). DISCUSSION: Constant production of micro- and nano-EVs per-milligram placenta, regardless of gestational age, and the increased daily burden of EVs across gestational age indicate these EVs have the potential to regulate maternal physiology from early pregnancy. Since total EV protein content, like EV numbers was, constant, this is a potentially reliable surrogate for quantifying EVs.


Subject(s)
Extracellular Vesicles/physiology , Gestational Age , Placenta/ultrastructure , Adaptation, Physiological , Cardiovascular Physiological Phenomena , DNA/analysis , Extracellular Vesicles/chemistry , Extracellular Vesicles/immunology , Female , Humans , Immune System/physiology , Pregnancy , Pregnancy Trimester, First , Proteins/analysis , Tissue Culture Techniques
19.
Placenta ; 64 Suppl 1: S4-S8, 2018 04.
Article in English | MEDLINE | ID: mdl-29273272

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialized topics. At IFPA meeting 2017 there were four themed workshops, all of which are summarized in this report. These workshops discussed new knowledge and technological innovations in the following areas of research: 1) placental bed; 2) 3D structural modeling; 3) clinical placentology; 4) treatment of placental dysfunction.


Subject(s)
Models, Anatomic , Placenta Diseases/physiopathology , Placenta/physiology , Placentation/physiology , Female , Humans , Placenta/anatomy & histology , Placenta Diseases/pathology , Pregnancy
20.
Hum Reprod ; 32(11): 2188-2198, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29040541

ABSTRACT

STUDY QUESTION: How do nano-vesicles extruded from normal first trimester human placentae affect maternal vascular function? SUMMARY ANSWER: Placental nano-vesicles affect the ability of systemic mesenteric arteries to undergo endothelium- and nitric oxide- (NO-) dependent vasodilation in vivo in pregnant mice. WHAT IS KNOWN ALREADY: Dramatic cardiovascular adaptations occur during human pregnancy, including a substantial decrease in total peripheral resistance in the first trimester. The human placenta constantly extrudes extracellular vesicles that can enter the maternal circulation and these vesicles may play an important role in feto-maternal communication. STUDY DESIGN, SIZE, DURATION: Human placental nano-vesicles were administered into CD1 mice via a tail vein and their localization and vascular effects at 30 min and 24 h post-injection were investigated. PARTICIPANTS/MATERIALS, SETTING, METHODS: Nano-vesicles from normal first trimester human placentae were collected and administered into pregnant (D12.5) or non-pregnant female mice. After either 30 min or 24 h of exposure, all major organs were dissected for imaging (n = 7 at each time point) while uterine and mesenteric arteries were dissected for wire myography (n = 6 at each time point). Additional in vitro studies using HMEC-1 endothelial cells were also conducted to investigate the kinetics of interaction between placental nano-vesicles and endothelial cells. MAIN RESULTS AND THE ROLE OF CHANCE: Nano-vesicles from first trimester human placentae localized to the lungs, liver and kidneys 24 h after injection into pregnant mice (n = 7). Exposure of pregnant mice to placental nano-vesicles for 30 min in vivo increased the vasodilatory response of mesenteric arteries to acetylcholine, while exposure for 24 h had the opposite effect (P < 0.05, n = 6). These responses were prevented by L-NAME, an NO synthase inhibitor. Placental nano-vesicles did not affect the function of uterine arteries or mesenteric arteries from non-pregnant mice. Placental nano-vesicles rapidly interacted with endothelial cells via a combination of phagocytosis, endocytosis and cell surface binding in vitro. LARGE SCALE DATA: N/A. LIMITATIONS REASONS FOR CAUTION: As it is not ethical to administer labelled placental nano-vesicles to pregnant women, pregnant CD1 mice were used as a model of pregnancy. WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to report the localization of placental nano-vesicles and their vascular effects in vivo. This work provides new insight into how the dramatic maternal cardiovascular adaptations to pregnancy may occur and indicates that placental extracellular vesicles may be important mediators of feto-maternal communication in a healthy pregnancy. STUDY FUNDING/COMPETING INTEREST(S): This research was supported by the Faculty of Medical and Health Science (FMHS) School of Medicine PBRF research fund to L.W.C. M.T. is a recipient of a University of Auckland Health Research Doctoral Scholarship and the Freemasons Postgraduate Scholarship. No authors have any competing interests to disclose.


Subject(s)
Extracellular Vesicles/transplantation , Mesenteric Arteries/physiology , Placenta/physiology , Uterine Artery/physiology , Vasodilation/physiology , Animals , Female , Humans , Kidney/physiology , Liver/physiology , Lung/physiology , Mice , Myography , Pregnancy , Vascular Resistance/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...