Differential distribution of ERα and ERβ mRNA in intrauterine tissues of the pregnant rhesus monkey

2000 ◽  
Vol 278 (1) ◽  
pp. C190-C198 ◽  
Author(s):  
Wen Xuan Wu ◽  
Xiao Hong Ma ◽  
Gordon C. S. Smith ◽  
Peter W. Nathanielsz

Two estrogen receptor (ER) isoforms, ERα and ERβ, have been described. However, no information is available in any species regarding the comparison of ERα and ERβ levels in pregnant intrauterine tissues. We investigated 1) distribution of ERα and ERβ mRNA in myometrium, amnion, choriodecidua, and placenta; 2) their abundance in intrauterine tissues at term not in labor (NIL) and in spontaneous term labor (STL); and 3) immunolocalization of ERα and ERβ in pregnant rhesus monkey myometrium. Myometrium, amnion, choriodecidua, and placenta were obtained at cesarean section from monkeys in STL at 156–166 days gestational age (GA) ( n = 4) and from control monkeys NIL at 140–152 days GA ( n = 4). RT-PCR was conducted to determine ERα and ERβ and glyceraldehyde-3-phosphate dehydrogenase mRNA abundance in four intrauterine tissues of the pregnant rhesus monkey. The cloned ERβ PCR fragment was subjected to sequence analysis. ERα and ERβ were localized in the myometrium by immunohistochemistry. We demonstrated that 1) rhesus monkey ERβ shares >97% identity with human ERβ in the region sequenced; 2) both ERs were expressed in myometrium, amnion, and choriodecidua but not in placenta in the current study; 3) ERα and ERβ were differentially distributed in myometrium and amnion; 4) ERα and ERβ were immunolocalized in myometrial smooth cells and smooth muscle and endothelial cells of the myometrial blood vessels. The biological significance of these quantitative differences in ER subtypes merits further study.

1997 ◽  
Vol 11 ◽  
pp. 51
Author(s):  
Anton J.G. Horrevoets ◽  
Carlie J.M. de Vries ◽  
A.E. van Achterberg ◽  
Ruud D. Fontijn ◽  
Benien van Aken ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3683-3683
Author(s):  
Petra Obrtlikova ◽  
Jeffrey J. Ross ◽  
Aernout Luttun ◽  
Joan D. Beckman ◽  
Susan A. Keirstead ◽  
...  

Abstract There is now growing evidence that embryonic stem cells provide an important resource to define the cellular and molecular mechanism of vascular development, and may serve as a potential source of cells for vascular repair. In our study, human ES cells (hESCs), H9 cell line, were differentiated by stromal cell co-culture with mouse bone marrow-derived stromal cell line S17 for 10–15 days. After this time, flow cytometry analysis confirmed the presence of a cell population with expression of surface antigens typical of endothelial cells (ECs). The hESC-derived cells were sorted for specific subpopulations of CD34+, CD31+, Flk1+ and Tie2+ cells using immunomagnetic selection to enrich for endothelial precursors. These sorted cells were cultured on fibronectin-coated plates in EGM2 media. Under these conditions, the cells assume EC morphology. The putative hESC-derived ECs expressed several EC markers including Flk1, Tie2, CD143, CD146, and bound to the lectin UEA-1. Gene expression analysis by RT-PCR further confirmed expression of transcripts for endothelial genes: Flk1, CD31, CD34, Tie2, eNOs, vWF and VE-Cadherin. Furthermore, the ECs were functional as shown by their ability to take up acLDL and form capillary-like structures when replated on Matrigel. To evaluate the smooth muscle cell (SMC) potential of this hESC-derived EC population, culture conditions were changed to media containing FBS, TGF β and PDGF-BB. Under these SMC-conditions, the cell populations converted to a flatter morphology and acquired intracellular fibrils. These cells expressed smooth muscle specific markers, as determined by immunohistochemistry: α-SMC actin, calponin and SM22. Q-RT-PCR confirmed a remarkable increase in expression of transcripts specific for SMC: α-SMC actin, calponin, SM22, smoothelin, myocardin. Importantly, we also found concomitant increased expression of 2 genes APEG-1and CRP2/SmLIM, preferentially expressed in arterial SMCs. At the time when SMC-gene expression increased, there was a corresponding dramatic decrease in expression of transcripts for endothelial genes. Notably, HUVEC cells treated with the same SMC-conditions did not develop into SMCs, suggesting this transition potential is unique to hESC-derived cells. Next, we used two functional tests to further evaluate the hESC-derived SMCs. First, we examined increase in intracellular calcium concentration evoked by 9 different agonists. The majority of the SMC population responded to bradykinin, oxytocin, endothelin-1, histamine and ATP, with fewer cells demonstrating a response to serotonin, vasopressin, norephinephrine and carbachol, consistent with a smooth muscle phenotype. In contrast, the hES-derived ECs responded to endothelin-1, histamine, bradykinin, as well as carbachol, with little response to oxytocin or the other agonists. Finally, we demonstrate that co-culture of hESC-derived SMCs together with hESC-derived ECs form ordered vascular structures composed of both cell types when cultured in a 3-dimensional Matrigel. These studies demonstrate that populations of hESC-derived ECs can convert to SMCs based on defined culture conditions. Further studies are now needed to identify whether this transition is the result of bipotential progenitor cells; or, if specific differentiated cells switch between these lineages. Alternatively, differentiated hESCs may produce progenitor cells specific for each lineage that are retained within the EC population.


2000 ◽  
Vol 164 (2) ◽  
pp. 207-214 ◽  
Author(s):  
F Xiao ◽  
GP Vinson ◽  

One controversy in the field of vascular angiotensin generation has surrounded the nature and particularly the source of vascular renin. This study investigated the expression of renin protein and its mRNA in aortic endothelial cells using immunocytochemistry, Western blotting, in situ hybridization and reverse transcription PCR (RT-PCR). Using a monoclonal antibody against human renin, immunocytochemical analysis revealed positive immunoreactivity in the cytoplasm of cultured bovine aortic endothelial cells. Immunoblotting of solubilized proteins separated by SDS-PAGE from cultured aortic endothelial cells identified two immunoreactive species with molecular masses of approximately 37-40 kDa. In situ hybridization showed that renin mRNA was localized in the cytoplasm of these cells. Using RT-PCR of RNA extracted from bovine aortic endothelial cells with primers specific for human renin, a clear single band was detected, which had the predicted size of 142 bp for (pro)renin. Angiotensin II (Ang II) was assayed in conditioned medium (CM) from cultured bovine aortic endothelial cells, and in addition, the effects of Ang II and CM on the proliferation of aorta smooth muscle cells (ASMC) were also studied. The results showed that CM contained Ang II equivalent to 15.05+/-4.67 pg/10(6) cells. Assay of smooth muscle cell proliferation by cell number, and by tritiated thymidine uptake, showed that proliferative responses in the presence of Ang II at a concentration of 10(-6)M were evident within 1 day of subculture, and cell numbers were nearly twice those of controls after 2 days. Thymidine incorporation into ASMC was also increased by Ang II in a dose-dependent manner and by endothelial cell CM. In both cases, stimulated proliferation was inhibited by the Ang II type 1 (AT1) receptor selective antagonist, losartan. These findings suggest that these vascular endothelial cells are a source of locally synthesized renin that may thus be involved in vascular Ang II generation. They also suggest that Ang II produced by the endothelial cells may be secreted and stimulate ASMC proliferation via the AT1 receptor.


2014 ◽  
Vol 34 (suppl_1) ◽  
Author(s):  
Mao Luo ◽  
Min Zeng ◽  
Meiping Ren ◽  
Rong Li ◽  
Xin Deng ◽  
...  

Objective: Platelets can regulate endothelial cell genes expression through their adhesion to the subendothelium in response to vascular injury. Recently it has been reported that some endothelial cells (ECs)-secreted genes can modulate vascular smooth muscle cells (VSMCs) phenotypic transformation by ECs/VSMCs co-culture. However, little is known about the effects of platelets adhesion to ECs interaction on VSMCs phenotype. In this study, we investigated the role of some genes secreted by platelets adhesion to EC in regulating SMC phenotypic transformation. Methods and Results: By Q-RT-PCR, expression of PAI-1, MMP-2 and MMP-9 were up-regulated in ECs after platelets adhesion to ECs comparing to ECs monoculture alone. In migration (scratch) and proliferation (CCK-8) assays, platelets adhesion to ECs increased EC migration-promoting activity and proliferative activity. After adhesion of platelets to ECs/VSMCs co-culture, expression of VSMCs contractile apparatus SM-MHC, Smoothelin-B, SMA and SM22a were significantly decreased comparing to ECs/VSMCs co-culture by Q-RT-PCR, but decreasing slightly in adhesion with platelets from type 2 diabetes. Conversely, it increased the expression of synthetic marker Smemb, CCND-1, CCND-2. Further analysis found that platelets adhesion to ECs/VSMCs co-culture significantly increased VSMCs migration-promoting activity and proliferative activity. In Gelatin Zymography assays, there were high levels of MMP-2 and MMP-9 after adhesion of platelets to ECs/VSMCs co-culture comparing to ECs/VSMCs co-culture alone. Conclusion: These results offer important insights into the mechanisms controlling phenotypic modulation of VSMCs by platelets adhesion to ECs, and help to explain the effects of platelets-ECs-VSMCs interaction on VSMCs contractile/synthetic, migration, and proliferation, suggesting that the adhesion of platelets to ECs is a key process in regulating phenotypic switch of VSMCs.


2011 ◽  
Vol 301 (5) ◽  
pp. E882-E888 ◽  
Author(s):  
Xuan Yu ◽  
Handong Ma ◽  
Scott A. Barman ◽  
Alexander T. Liu ◽  
Minga Sellers ◽  
...  

Estrogens can either relax or contract arteries via rapid, nongenomic mechanisms involving classic estrogen receptors (ER). In addition to ERα and ERβ, estrogen may also stimulate G protein-coupled estrogen receptor 1 (GPER) in nonvascular tissue; however, a potential role for GPER in coronary arteries is unclear. The purpose of this study was to determine how GPER activity influenced coronary artery reactivity. In vitro isometric force recordings were performed on endothelium-denuded porcine arteries. These studies were augmented by RT-PCR and single-cell patch-clamp experiments. RT-PCR and immunoblot studies confirmed expression of GPER mRNA and protein, respectively, in smooth muscle from either porcine or human coronary arteries. G-1, a selective GPER agonist, produced a concentration-dependent relaxation of endothelium-denuded porcine coronary arteries in vitro. This response was attenuated by G15, a GPER-selective antagonist, or by inhibiting large-conductance calcium-activated potassium (BKCa) channels with iberiotoxin, but not by inhibiting NO signaling. Last, single-channel patch-clamp studies demonstrated that G-1 stimulates BKCa channel activity in intact smooth muscle cells from either porcine or human coronary arteries but had no effect on channels isolated in excised membrane patches. In summary, GPER activation relaxes coronary artery smooth muscle by increasing potassium efflux via BKCa channels and requires an intact cellular signaling mechanism. This novel action of estrogen-like compounds may help clarify some of the controversy surrounding the vascular effects of estrogens.


2021 ◽  
Vol 8 ◽  
Author(s):  
Bo Li ◽  
Zhijun Lei ◽  
You Wu ◽  
Bingyu Li ◽  
Ming Zhai ◽  
...  

Background: Serine proteinase inhibitor A3 (SERPINA3) has been discovered in the pathogenesis of many human diseases, but little is known about the role of SERPINA3 in coronary artery disease (CAD). Therefore, we aim to determine its relationship with CAD and its function in the pathogenesis of atherosclerosis.Methods: In total 86 patients with CAD and 64 patients with non-CAD were compared. The plasma SERPINA3 levels were measured using ELISA. Logistic regression analysis and receiver-operating characteristic (ROC) analysis were performed to illustrate the association between plasma SERPINA3 levels and CAD. In vitro, real-time PCR (RT-PCR) and immunofluorescence staining were used to determine the expression of SERPINA3 in atherosclerotic plaques and their component cells. Then rat aortic smooth muscle cells (RASMCs) were transfected with siRNA to knock down the expression of SERPINA3 and human umbilical vein endothelial cells (HUVECs) were stimulated by SERPINA3 protein. EdU assay and scratch assay were used for assessing the capability of proliferation and migration. The cell signaling pathway was evaluated by western blot and RT-PCR.Results: Patients with CAD [104.4(54.5–259.2) μg/mL] had higher levels of plasma SERPINA3 than non-CAD [65.3(47.5–137.3) μg/mL] (P = 0.004). After being fully adjusted, both log-transformed and tertiles of plasma SERPINA3 levels were significantly associated with CAD. While its diagnostic value was relatively low since the area under the ROC curve was 0.64 (95% CI: 0.55–0.73). Secreted SERPINA3 might increase the expression of inflammatory factors in HUVECs. Vascular smooth muscle cells had the highest SERPINA3 expression among the aorta compared to endothelial cells and inflammatory cells. The knockdown of SERPINA3 in RASMCs attenuated its proliferation and migration. The phosphorylated IκBα and its downstream pathway were inhibited when SERPINA3 was knocked down.Conclusions: Elevated plasma SERPINA3 levels were associated with CAD. SERPINA3 can increase inflammatory factors expression in HUVECs. It can regulate VSMCs proliferation, migration, and releasing of inflammatory factors through the NF-κB signaling pathway. Thus, SERPINA3 played a significant role in the pathogenesis of atherosclerosis.


1994 ◽  
Vol 72 (01) ◽  
pp. 044-053 ◽  
Author(s):  
N Chomiki ◽  
M Henry ◽  
M C Alessi ◽  
F Anfosso ◽  
I Juhan-Vague

SummaryIndividuals with elevated levels of plasminogen activator inhibitor type 1 are at risk of developing atherosclerosis. The mechanisms leading to increased plasma PAI-1 concentrations are not well understood. The link observed between increased PAI-1 levels and insulin resistance has lead workers to investigate the effects of insulin or triglyceride rich lipoproteins on PAI-1 production by cultured hepatocytes or endothelial cells. However, little is known about the contribution of these cells to PAI-1 production in vivo. We have studied the expression of PAI-1 in human liver sections as well as in vessel walls from different territories, by immunocytochemistry and in situ hybridization.We have observed that normal liver endothelial cells expressed PAI-1 while parenchymal cells did not. However, this fact does not refute the role of parenchymal liver cells in pathological states.In healthy vessels, PAI-1 mRNA and protein were detected primarily at the endothelium from the lumen as well as from the vasa vasorum. In normal arteries, smooth muscle cells were able to produce PAI-1 depending on the territory tested. In deeply altered vessels, PAI-1 expression was observed in neovessels scattering the lesions, in some intimal cells and in smooth muscle cells. Local increase PAI-1 mRNA described in atherosclerotic lesions could be due to the abundant neovascularization present in the lesion as well as a raised expression in smooth muscle cells. The increased PAI-1 in atherosclerosis could lead to fibrin deposit during plaque rupture contributing further to the development and progression of the lesion.


Sign in / Sign up

Export Citation Format

Share Document