Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 95(2): 43, 2016 08.
Article in English | MEDLINE | ID: mdl-27335074

ABSTRACT

The purpose of this study was to establish the time course and hemodynamic significance of de novo formed and enlarged uteroplacental arteries during pregnancy. Using x-ray microcomputed tomography (n = 4-7 placentas from 2-4 dams/gestational group), uteroplacental arterial vascular dimensions were measured at individual implantation sites. Dimensions and topology were used to compute total and vessel-specific resistances and cross-sectional areas. Diameter enlargement of the uterine artery (+55% by Embryonic Day 5.5 [E5.5]) and preplacental radial arteries (+30% by E8.5) was significant only in early gestation. Formation of spiral arteries (E9.5-E11.5), maternal canals, and canal branches (E11.5-E13.5) during midgestation was followed by enlargement of these vessels such that, from E9.5 to E17.5 (near term), spiral artery resistance dropped 9-fold, and canal resistance became negligible. A 12-fold increase in terminal vessel cross-sectional area was nearly sufficient to offset known increases in flow so that blood velocity entering the exchange region was predicted to increase by only 2-fold. The calculated 47% decrease in total resistance downstream of the uterine artery, determined from vascular geometry, was in accord with prior uterine blood flow data in vivo and was due to enlarging spiral artery diameters. Interestingly, radial artery resistance was unchanged after E9.5 so that radial arteries accounted for 91% of resistance and pressure drop in the uteroplacental arterial network by E17.5. These findings led us to propose functional roles for the three morphologically defined vessel types: radial arteries to reduce pressure, spiral artery enlargement to increase flow with gestation, and maternal canal elaboration and enlargement to maintain low exit velocities into the exchange region.


Subject(s)
Hemodynamics/physiology , Placenta/blood supply , Placental Circulation/physiology , Uterus/blood supply , Animals , Female , Mice , Pregnancy , Radial Artery/diagnostic imaging , Radial Artery/physiology , Uterine Artery/diagnostic imaging , Uterine Artery/physiology , Uterus/diagnostic imaging , Vascular Resistance/physiology , X-Ray Microtomography
2.
Biol Reprod ; 92(2): 48, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25519187

ABSTRACT

The sites of elevated vascular resistance that impede placental perfusion in pathological pregnancies are unknown. In the current study, we identified these sites in a knockout mouse model (eNOS(-/-)) with reduced uterine (-55%) and umbilical (-29%) artery blood flows caused by endothelial nitric oxide synthase deficiency. Uteroplacental and fetoplacental arterial vascular trees of pregnant mice near term were imaged using x-ray microcomputed tomography (n = 5-10 placentas from 3-5 dams/group). The resulting three-dimensional images were analyzed to assess vessel geometry and vascular resistance. In control and eNOS(-/-) trees, ∼90% of total uteroplacental vascular resistance was located in the radial arteries. Changes in eNOS(-/-) vessel geometry, including 30% reductions in uterine, radial, and spiral artery diameters, were calculated to increase arterial resistance downstream of the uterine artery by 2.3-fold, predicting a 57% decrease in uterine blood flow. Despite large reductions in eNOS(-/-) spiral arteries (-55% by volume) and maternal canals (-67% by volume), these vessels were relatively minor contributors to resistance. In the eNOS(-/-) fetoplacental tree, the number of arterioles (50-75 µm diameter) increased by 26%. Nevertheless, calculated resistance rose by 19%, predominantly because arteries near the periphery of the tree selectively exhibited a 7%-9% diameter reduction. We conclude that previously observed decreases in uterine and umbilical blood flows in eNOS(-/-) pregnancies are associated with markedly divergent structural changes in the uteroplacental versus fetoplacental circulations. Results showed the radial arteries were critical determinants of uteroplacental resistance in mice and therefore warrant greater attention in future studies in pathological human pregnancies.


Subject(s)
Nitric Oxide Synthase Type III/genetics , Placenta/blood supply , Placental Circulation/genetics , Radial Artery/diagnostic imaging , Uterine Artery/diagnostic imaging , Vascular Resistance/genetics , Animals , Female , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Placenta/metabolism , Pregnancy , Radial Artery/metabolism , Radiography , Uterine Artery/metabolism , Uterus/blood supply , Uterus/metabolism
3.
Biol Reprod ; 91(4): 87, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25122061

ABSTRACT

Given the angiogenic function of vascular endothelial growth factor A (VEGFA), the function of its expression by trophoblast in the avascular placental junctional zone is unknown. In mice, cells from the trophoblast-specific protein alpha (Tpbpa) lineage populate this zone and, in late gestation, some of these cells invade the decidual layer. To diminish Vegfa expression in Tpbpa cells, we crossed Vegfa(flox/flox) females with males carrying Tpbpa-Cre. For single deletion (sd) of Vegfa in Tpbpa cells in 100% of conceptuses (SD100 pregnancies, sd conceptuses) we crossed homozygous lines. For double deletion (dd) of both Vegfa alleles in 50% of the conceptuses (DD50 pregnancies, 50% dd conceptuses and 50% no deletion [nd]), we crossed homozygous Vegfa(flox/flox) females with males heterozygous for Tpbpa-Cre and homozygous for Vegfa(flox/flox). Controls were Vegfa(flox/flox) females bred to wild-type males (V-CTRL pregnancies). In SD100 pregnancies, maternal plasma immunoreactive VEGFA significantly increased and arterial blood pressure decreased, whereas fetal body weight and placental Flt1, sFlt1, and Prl3b1 mRNA were unchanged. In DD50, maternal immunoreactive VEGFA and arterial pressures were unaltered, but both dd and nd conceptuses exhibited significantly increased embryonic lethality, altered expression of Flt1, sFlt1, and Prl3b1 mRNA in the decidual layer, and decreased fetal body weight relative to V-CTRL. Maternal cardiac output significantly increased in proportion to dd conceptuses in the pregnancy. In DD50, results are consistent with altered maternal function beginning in early gestation and adversely impacting both conceptus genotypes. We conclude that maternal function is influenced by Vegfa expression in trophoblast cells at the maternal-fetal interface, likely via an endocrine mechanism.


Subject(s)
Placenta/metabolism , Trophoblasts/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Lineage , Female , Gene Deletion , Gene Expression Regulation/physiology , Integrases/genetics , Integrases/metabolism , Maternal-Fetal Exchange , Mice , Mice, Transgenic , Placenta/cytology , Placental Circulation/physiology , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics
4.
Hypertension ; 61(1): 259-66, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23150513

ABSTRACT

It is not known whether eNOS deficiency in the mother or the conceptus (ie, placenta and fetus) causes fetal growth restriction in mice lacking the endothelial NO synthase gene (eNOS knockout [KO]). We hypothesized that eNOS sustains fetal growth by maintaining low fetoplacental vascular tone and promoting fetoplacental vascularity and that this is a conceptus effect and is independent of maternal genotype. We found that eNOS deficiency blunted fetal growth, and blunted the normal increase in umbilical blood flow and umbilical venous diameter and the decrease in umbilical arterial Resistance Index in late gestation (14.5-17.5 days) in eNOS KO relative to C57Bl/6J controls. On day 17.5, fetoplacental capillary lobule length and capillary density in vascular corrosion casts were reduced in eNOS KO placentas. Reduced vascularization may be a result of decreased vascular endothelial growth factor mRNA and protein expression in eNOS KO placentas at this stage. These factors, combined with significant anemia found in eNOS KO fetuses, would be anticipated to reduce fetal oxygen delivery and contribute to the fetal tissue hypoxia that was detected in the heart, lung, kidney, and liver by immunohistochemistry using pimonidazole. Although maternal eNOS deficiency impairs uteroplacental adaptations to pregnancy, maternal genotype was not a significant factor affecting growth in heterozygous conceptuses. This indicates that fetal growth restriction was primarily caused by conceptus eNOS deficiency. In mice, placental hemodynamic and vascular changes with gestation and growth restriction showed strong parallels with human pregnancy. Thus, the eNOS KO model could provide insights into the pathogenesis of human intrauterine growth restriction.


Subject(s)
Fetal Development/genetics , Fetal Growth Retardation/metabolism , Fetus/blood supply , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/genetics , Placenta/blood supply , Animals , Female , Fetus/metabolism , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Placenta/metabolism , Pregnancy
5.
Hypertension ; 60(1): 231-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22615111

ABSTRACT

Preeclampsia is associated with impaired uteroplacental adaptations during pregnancy and abnormalities in the endothelial NO synthase (eNOS)-NO pathway, but whether eNOS deficiency plays a causal role is unknown. Thus, the objective of the current study was to determine the role of eNOS in the mother and/or conceptus in uteroplacental changes during pregnancy using eNOS knockout mice. We quantified uterine artery blood flow using microultrasound, visualized the uteroplacental vasculature using vascular corrosion casts, and used pimonidazole and hypoxia-inducible factor 1α immunohistochemistry as markers of hypoxia in the placentas of eNOS knockout mice versus the background strain, C57Bl/6J (wild type). We found that increases in uteroplacental blood flow, uterine artery diameter, and spiral artery length were reduced, and markers of placental hypoxia in the junctional zone were elevated in late gestation in eNOS knockout mice. Both maternal and conceptus genotypes contributed to changes in uterine artery diameter and flow. Despite placental hypoxia, placental soluble fms-like tyrosine kinase 1 and tumor necrosis factor-α mRNA, and in maternal plasma, soluble fms-like tyrosine kinase 1 were not elevated in eNOS knockout mice. Thus, our results show that both eNOS in the mother and the conceptus contribute to uteroplacental vascular changes and increased uterine arterial blood flow in normal pregnancy.


Subject(s)
Nitric Oxide Synthase Type III/deficiency , Placenta/metabolism , Uterine Artery/physiology , Uterus/metabolism , Animals , Blood Flow Velocity , Enzyme-Linked Immunosorbent Assay , Female , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Nitric Oxide Synthase Type III/genetics , Oxygen/metabolism , Placenta/blood supply , Placenta/immunology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Uterine Artery/ultrastructure , Uterus/blood supply , Vascular Endothelial Growth Factor Receptor-1/blood , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
6.
Hypertension ; 59(3): 732-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22275534

ABSTRACT

Preeclampsia is a life-threatening disorder characterized by maternal gestational hypertension and proteinuria that results from placental dysfunction. Placental abnormalities include abnormal syncytiotrophoblast and a 50% reduction in placental expression of the transcription factor Gcm1. In mice, homozygous deletion of Gcm1 prevents syncytiotrophoblast differentiation and is embryonic lethal. We used heterozygous Gcm1 mutants (Gcm1(+/-)) to test the hypothesis that hypomorphic expression of placental Gcm1 causes defective syncytiotrophoblast differentiation and maternal and placental phenotypes that resemble preeclampsia. We mated wild-type female mice with Gcm1(+/-) fathers to obtain wild-type mothers carrying ≈50% Gcm1(+/-) conceptuses. Gcm1(+/-) placentas had syncytiotrophoblast abnormalities including reduced gene expression of Gcm1-regulated SynB, elevated expression of sFlt1, a thickened interhemal membrane separating maternal and fetal circulations, and electron microscopic evidence in syncytiotrophoblast of necrosis and impaired maternal-fetal transfer. Fetoplacental vascularity was quantified by histomorphometry and microcomputed tomography imaging. In Gcm1(+/-), it was ≈30% greater than wild-type littermates, whereas placental vascular endothelial growth factor A (Vegfa) expression and fetal and placental weights did not differ. Wild-type mothers carrying Gcm1(+/-) conceptuses developed late gestational hypertension (118±2 versus 109.6±0.7 mm Hg in controls; P<0.05). We next correlated fetoplacental vascularity with placental Gcm1 expression in human control and pathological pregnancies and found that, as in mice, fetoplacental vascularity increased when GCM1 protein expression decreased (R(2)=-0.45; P<0.05). These results support a role for reduced placental Gcm1 expression as a causative factor in defective syncytiotrophoblast differentiation and maternal and placental phenotypes in preeclampsia in humans.


Subject(s)
Fetus/blood supply , Gene Expression Regulation , Nuclear Proteins/genetics , Placenta/blood supply , Pre-Eclampsia/genetics , Pregnancy, Animal , Transcription Factors/genetics , Trophoblasts/metabolism , Animals , DNA/genetics , DNA-Binding Proteins , Disease Models, Animal , Female , Fetus/metabolism , Gestational Age , Humans , Male , Mice , Neuropeptides/biosynthesis , Neuropeptides/genetics , Nuclear Proteins/biosynthesis , Phenotype , Placenta/metabolism , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Pregnancy Outcome , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis , Trophoblasts/cytology
7.
Am J Physiol Heart Circ Physiol ; 302(6): H1261-73, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22268107

ABSTRACT

How the fetoplacental arterial tree grows and expands during late gestational development is largely unknown. In this study, we quantified changes in arterial branching in the fetal exchange region of the mouse placenta during late gestation, when capillarization increases rapidly. We studied two commonly used mouse strains, CD1 and C57Bl/6 (B6), at embryonic days (E)13.5, 15.5, and 17.5. B6 mice differ from CD1 mice by exhibiting a blunted fetal weight gain in late gestation. We found that B6 capillarization and interhemal membrane thinning were reduced and placental hypoxia-inducible factor-1α and VEGF-A expression were higher than CD1 near term. Automated vascular segmentation of microcomputed tomography data sets revealed that the number of arterial vessels ≥50 µm remained constant during late gestation in both strains, despite large increases in downstream capillary volume quantified by stereology (+65% in B6 mice and +200% in CD1 mice). Arterial diameters expanded in both strains from E13.5 to E15.5; however, diameters continued to expand to E17.5 in B6 mice only. The diameter scaling coefficient at branch sites was near optimal (-3.0) and remained constant in CD1 mice, whereas it decreased, becoming abnormal, in B6 mice at term (-3.5 ± 0.2). Based on arterial tree geometry, resistance remained constant throughout late gestation (∼0.45 mmHg·s·µl(-1)) in CD1 mice, whereas it decreased by 50% in late gestation in B6 mice. Quantification of the fetoplacental vasculature revealed significant strain-dependent differences in arterial and capillary expansion in late gestation. In both strains, enlargement of the fetoplacental arterial tree occurred primarily by increased arterial diameters with no change in segment numbers in late gestation.


Subject(s)
Capillaries/embryology , Fetus/blood supply , Neovascularization, Physiologic , Placenta/blood supply , Placental Circulation , Animals , Arteries/embryology , Blotting, Western , Capillaries/diagnostic imaging , Capillaries/ultrastructure , Female , Fetal Weight , Genotype , Gestational Age , Hemodynamics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Litter Size , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Neovascularization, Physiologic/genetics , Phenotype , Pregnancy , Species Specificity , Vascular Endothelial Growth Factor A/metabolism , X-Ray Microtomography
8.
Am J Physiol Heart Circ Physiol ; 300(2): H675-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21148768

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous environmental pollutants and the main toxicants found in cigarettes. Women are often exposed to PAHs before pregnancy, typically via prepregnancy smoking. To determine how prepregnancy exposure affects the fetoplacental vasculature of the placenta, we exposed female mice to PAHs before conception, perfused the fetoplacental arterial trees with X-ray contrast agent, and imaged the vasculature ex vivo by microcomputed tomography (micro-CT) at embryonic day 15.5. Automated vascular segmentation and flow calculations revealed that in control trees, <40 chorionic plate vessels (diameter>180 µm) gave rise to ∼1,300 intraplacental arteries (50-180 µm), predicting an arterial vascular resistance of 0.37±0.04 mmHg·s·µl(-1). PAH exposure increased vessel curvature of chorionic plate vessels and significantly increased the tortuousity ratio of the tree. Intraplacental arteries were reduced by 17%, primarily due to a 27% decrease in the number of arteriole-sized (50-100 µm) vessels. There were no changes in the number of chorionic vessels, the depth or span of the tree, the diameter scaling coefficient, or the segment length-to-diameter ratio. PAH exposure resulted in a tree with a similar size and dichotomous branching structure, but one that was comparatively sparse so that arterial vascular resistance was increased by 30%. Assuming the same pressure gradient, blood flow would be 19% lower. Low flow may contribute to the 23% reduction observed in fetal weight. New insights into the specific effects of PAH exposure on a developing arterial tree were achieved using micro-CT imaging and automated vascular segmentation analysis.


Subject(s)
Arteries/drug effects , Placental Circulation/drug effects , Polycyclic Aromatic Hydrocarbons/toxicity , Animals , Arteries/pathology , Arterioles/drug effects , Arterioles/pathology , Body Weight/drug effects , Chorion/blood supply , Contrast Media , Female , Fetal Development/drug effects , Hemodynamics/physiology , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Models, Anatomic , Neovascularization, Pathologic , Phenotype , Placenta/drug effects , Placentation , Pregnancy , Receptors, Aryl Hydrocarbon/metabolism , Smoking/pathology , Tomography, X-Ray Computed , Vascular Resistance/drug effects
9.
Am J Physiol Endocrinol Metab ; 295(2): E519-30, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18559983

ABSTRACT

Maternal cigarette smoking is considered an important risk factor associated with fetal intrauterine growth restriction (IUGR). Polycyclic aromatic hydrocarbons (PAHs) are well-known constituents of cigarette smoke, and the effects of acute exposure to these chemicals at different gestational stages have been well established in a variety of laboratory animals. In addition, many PAHs are known ligands of the aryl hydrocarbon receptor (AhR), a cellular xenobiotic sensor responsible for activating the metabolic machinery. In this study, we have applied a chronic, low-dose regimen of PAH exposure to C57Bl/6 female mice before conception. This treatment caused IUGR in day 15.5 post coitum (d15.5) fetuses and yielded abnormalities in the placental vasculature, resulting in significantly reduced arterial surface area and volume of the fetal arterial vasculature of the placenta. However, examination of the small vasculature within the placental labyrinth of PAH-exposed dams revealed extensive branching and enlargement of these vessels, indicating a possible compensatory mechanism. These alterations in vascularization were accompanied by reduced placental cell death rates, increased expression levels of antiapoptotic Xiap, and decreased expression of proapoptotic Bax, cleaved poly(ADP-ribose) polymerase-1, and active caspase-3. AhR-deficient fetuses were rescued from PAH-induced growth restriction and exhibited no changes in the labyrinthine cell death rate. The results of this investigation suggest that chronic exposure to PAHs is a contributing factor to the development of IUGR in human smokers and that the AhR pathway is involved.


Subject(s)
Fetal Growth Retardation/chemically induced , Maternal Exposure/adverse effects , Placenta/blood supply , Placenta/drug effects , Polycyclic Aromatic Hydrocarbons/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Caspase 3/metabolism , Cell Death/drug effects , Female , Fetal Growth Retardation/metabolism , Histocytochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Poly(ADP-ribose) Polymerases/metabolism , Pregnancy , Random Allocation , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-2-Associated X Protein/metabolism
10.
ILAR J ; 47(2): 103-17, 2006.
Article in English | MEDLINE | ID: mdl-16547367

ABSTRACT

Considerable progress has been made in adapting existing and developing new technologies to enable increasingly detailed phenotypic information to be obtained in embryonic and newborn mice. Sophisticated methods for imaging mouse embryos and newborns are available and include ultrasound and magnetic resonance imaging (MRI) for in vivo imaging, and MRI, vascular corrosion casts, micro-computed tomography, and optical projection tomography (OPT) for postmortem imaging. In addition, Doppler and M-mode ultrasound are useful noninvasive tools to monitor cardiac and vascular hemodynamics in vivo in embryos and newborns. The developmental stage of the animals being phenotyped is an important consideration when selecting the appropriate technique for anesthesia or euthanasia and for labeling animals in longitudinal studies. Study design also needs to control for possible differences between inter- and intralitter variability, and for possible long-term developmental effects caused by anesthesia and/or procedures. Noninvasive or minimally invasive intravenous or intracardiac injections or blood sampling, and arterial pressure and electrocardiography (ECG) measurements are feasible in newborns. Whereas microinjection techniques are available for embryos as young as 6.5 days of gestation, further advances are required to enable minimally invasive fluid or tissue samples, or blood pressure or ECG measurements, to be obtained from mouse embryos in utero. The growing repertoire of techniques available for phenotyping mouse embryos and newborns promises to accelerate knowledge gained from studies using genetically engineered mice to understand molecular regulation of morphogenesis and the etiology of congenital diseases.


Subject(s)
Animals, Newborn/physiology , Mice, Transgenic/embryology , Mice, Transgenic/physiology , Animals , Diagnostic Imaging/methods , Embryo, Mammalian/diagnostic imaging , Female , Mice , Phenotype , Pregnancy , Ultrasonography
11.
Methods Mol Med ; 121: 371-92, 2006.
Article in English | MEDLINE | ID: mdl-16251755

ABSTRACT

This chapter describes methods for making vascular corrosion casts of the uteroplacental and fetoplacental vasculature of the mouse placenta. A catheter placed in the ascending thoracic aorta of a pregnant mouse permits the introduction of a methyl methacrylate casting compound into the lower body vasculature, including the uterus and placenta. A fine-tipped glass cannula attached to a double-lumen catheter is used to instill the same casting compound in the fetoplacental vessels of mouse placentas. Following polymerization of the casting compound, tissue is digested off of the placental casts using 20% KOH. The washed and dried casts are then available for light or scanning electron microscopy. The methods described have been used to cast the mouse uteroplacental vasculature from 5.5-18.5 d gestation and the fetoplacental vasculature from 12.5 d gestation to term.


Subject(s)
Corrosion Casting/methods , Placenta/blood supply , Placental Circulation , Animals , Catheterization , Female , Mice , Placenta/cytology , Pregnancy
12.
Dev Biol ; 250(2): 358-73, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12376109

ABSTRACT

Mammalian embryos have an intimate relationship with their mothers, particularly with the placental vasculature from which embryos obtain nutrients essential for growth. It is an interesting vascular bed because maternal vessel number and diameter change dramatically during gestation and, in rodents and primates, the terminal blood space becomes lined by placental trophoblast cells rather than endothelial cells. Molecular genetic studies in mice aimed at identifying potential regulators of these processes have been hampered by lack of understanding of the anatomy of the vascular spaces in the placenta and the general nature of maternal-fetal vascular interactions. To address this problem, we examined the anatomy of the mouse placenta by preparing plastic vascular casts and serial histological sections of implantation sites from embryonic day (E) 10.5 to term. We found that each radial artery carrying maternal blood into the uterus branched into 5-10 dilated spiral arteries located within the metrial triangle, populated by uterine natural killer (uNK) cells, and the decidua basalis. The endothelial-lined spiral arteries converged together at the trophoblast giant cell layer and emptied into a few straight, trophoblast-lined "canals" that carried maternal blood to the base of the placenta. Maternal blood then percolated back through the intervillous space of the labyrinth toward the maternal side of the placenta in a direction that is countercurrent to the direction of the fetal capillary blood flow. Trophoblast cells were found invading the uterus in two patterns. Large cells that expressed the trophoblast giant cell-specific gene Plf (encoding Proliferin) invaded during the early postimplantation period in a pattern tightly associated with spiral arteries. These peri/endovascular trophoblast were detected only approximately 150-300 microm upstream of the main giant cell layer. A second type of widespread interstitial invasion in the decidua basalis by glycogen trophoblast cells was detected after E12.5. These cells did not express Plf, but rather expressed the spongiotrophoblast-specific gene Tpbp. Dilation of the spiral arteries was obvious between E10.5 and E14.5 and was associated with a lack of elastic lamina and smooth muscle cells. These features were apparent even in the metrial triangle, a site far away from the invading trophoblast cells. By contrast, the transition from endothelium-lined artery to trophoblast-lined (hemochorial) blood space was associated with trophoblast giant cells. Moreover, the shaping of the maternal blood spaces within the labyrinth was dependent on chorioallantoic morphogenesis and therefore disrupted in Gcm1 mutants. These studies provide important insights into how the fetoplacental unit interacts with the maternal intrauterine vascular system during pregnancy in mice.


Subject(s)
Maternal-Fetal Exchange/physiology , Placenta/blood supply , Placental Circulation/physiology , Trophoblasts/cytology , Trophoblasts/physiology , Animals , Arteries/anatomy & histology , Corrosion Casting , DNA-Binding Proteins , Female , Glycoproteins/genetics , Glycoproteins/physiology , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred ICR , Mice, Mutant Strains , Neovascularization, Physiologic , Neuropeptides/genetics , Neuropeptides/physiology , Nuclear Proteins , Placenta/embryology , Pregnancy , Prolactin , Species Specificity , Transcription Factors
13.
Am J Physiol Heart Circ Physiol ; 282(3): H918-25, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11834487

ABSTRACT

Genetically altered mice may provide useful models for exploring cardiovascular regulation during pregnancy and postpartum if changes in mice mimic humans. We found in awake ICR (CD-1) mice at 17.5 days gestation that hematocrit was reduced 18%, and the pressor response to intravenous angiotensin II was reduced ~33%. Arterial pressure in awake mice was 12% lower in early pregnancy (3.5 days) than late pregnancy (17.5 days) and postpartum (3 and 17 days after delivery), whereas heart rate was 10-20% higher in the peripartum period (17.5 days gestation and 3 days postpartum). In late pregnancy, cardiac output under isoflurane anesthesia was 64% higher than in nonpregnant mice, due to a 37% increase in stroke volume and a 17% increase in heart rate. All changes P < 0.05. We conclude that, as in humans, mice exhibit hypotension in early pregnancy, and a blunted pressor response to angiotensin II, a decrease in hematocrit, and a marked increase in cardiac output in late pregnancy.


Subject(s)
Cardiovascular Physiological Phenomena , Hemodynamics/physiology , Postpartum Period/physiology , Pregnancy, Animal/physiology , Animals , Blood Flow Velocity , Blood Pressure , Cardiac Output , Female , Heart Rate , Hematocrit , Mice , Mice, Inbred ICR , Pregnancy , Reference Values , Stroke Volume
SELECTION OF CITATIONS
SEARCH DETAIL
...