Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
J Obstet Gynaecol ; 41(1): 66-72, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32148119

ABSTRACT

The aim of the study was to investigate foetal cardiac function using the modified myocardial performance index (Mod-MPI) in poorly controlled gestational diabetics and its link with intrauterine markers for hypoxia and to an adverse outcome. In a prospective, cross sectional study, 44 consecutive women with severe or poorly controlled gestational diabetic pregnancies in their third trimester on insulin therapy were recruited and matched with 44 women with normal pregnancies which served as the control group. Using Doppler echocardiography the foetal Mod-MPI was calculated. The foetal Mod-MPI was significantly higher in the diabetic group compared to the controls indicating significant myocardial dysfunction. The Mod-MPI served as an excellent marker of adverse outcomes. Foetal myocardial function was significantly impaired in poorly controlled gestational diabetics and there was a significant link of Mod-MPI to intrauterine markers of hypoxia, as well as to an adverse outcome. Mod-MPI has the potential to improve foetal surveillance in gestational diabetes.IMPACT STATEMENTWhat is already known on this subject? Abnormal foetal cardiac function, as reflected in the modified myocardial performance index, has been reported to be significantly increased in foetuses of poorly controlled diabetics managed on insulin.What do the results of this study add? There is a significant link between abnormal foetal cardiac function to intrauterine markers of hypoxia, as well as to an adverse outcome; and that development of myocardial dysfunction could be one of the main mechanisms, inducing foetal compromise in poorly controlled gestational diabetes.What are the implications of these findings for clinical practice and/or further research? This study explores an interesting concept of foetal pathophysiology in gestational diabetes, namely the concept of "pseudo-hypoxia" in a foetus of a gestational diabetic mother, and this intrauterine "hypoxic stress" in turn leading to myocardial dysfunction. The Mod-MPI, a clinical marker for cardiac dysfunction, can therefore be used in the clinical setting to track a deteriorating metabolic state.


Subject(s)
Diabetes, Gestational/physiopathology , Echocardiography, Doppler/methods , Fetal Hypoxia/diagnostic imaging , Glycemic Control/adverse effects , Ultrasonography, Prenatal/methods , Adult , Biomarkers/analysis , Cross-Sectional Studies , Diabetes, Gestational/therapy , Female , Fetal Distress/diagnostic imaging , Fetal Distress/embryology , Fetal Distress/etiology , Fetal Heart/diagnostic imaging , Fetal Heart/embryology , Fetal Hypoxia/embryology , Fetal Hypoxia/etiology , Humans , Pregnancy , Prospective Studies
2.
Taiwan J Obstet Gynecol ; 59(6): 842-847, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33218399

ABSTRACT

OBJECTIVE: This study aimed to evaluate whether state and trait anxiety among pregnant women were associated with fetoplacental Doppler findings, abnormal placental pathology, and placental angiogenic factors. MATERIALS AND METHODS: A total of 102 pregnant women at 32-35 gestational weeks were recruited and examined prospectively. State and trait anxiety were measured using the State-Trait Anxiety Inventory. Using Doppler ultrasound, pulsatility index (PI) of the umbilical artery (UA), middle cerebral artery (MCA), and uterine artery (UtA) and cerebroplacental ratio (CPR) were determined. Doppler parameters were converted into multiples of the median (MoM). Abnormal placental pathology was classified into 2 groups: vascular underperfusion (VU) and histological chorioamnionitis (HCA). Immunohistochemical analysis was performed to examine placental cells staining positive for placental growth factor (PLGF) and hypoxia-inducible factor-1-α (HIF-1α), which are markers for angiogenesis and hypoxic status, respectively. RESULTS: Women with high state anxiety scores had low MCA-PI MoM and CPR MoM, while those with high trait anxiety scores had low MCA-PI MoM. VU was associated with a higher incidence of high trait anxiety scores, and HCA was associated with a higher incidence of high state and trait anxiety scores. Regression analysis showed a relationship between maternal state anxiety on MCA-PI MoM and HCA after controlling for covariates. Maternal trait anxiety exhibited relationships with VU and HCA after adjustment. CONCLUSION: Our results demonstrated that maternal anxiety is associated with altered fetal cerebral blood flow and abnormal placental pathology but is not associated with uteroplacental insufficiency and placental angiogenic factors.


Subject(s)
Anxiety/diagnostic imaging , Fetus/blood supply , Placenta/diagnostic imaging , Pregnancy Complications/diagnostic imaging , Ultrasonography, Doppler , Ultrasonography, Prenatal/methods , Adult , Angiogenesis Inducing Agents/analysis , Anxiety/pathology , Biomarkers/analysis , Cerebrovascular Circulation , Chorioamnionitis/diagnostic imaging , Chorioamnionitis/psychology , Female , Fetal Hypoxia/diagnostic imaging , Fetal Hypoxia/embryology , Fetal Hypoxia/psychology , Fetus/diagnostic imaging , Gestational Age , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Middle Cerebral Artery/diagnostic imaging , Placenta/pathology , Placenta Growth Factor/analysis , Pregnancy , Pregnancy Complications/pathology , Pregnancy Complications/psychology , Prospective Studies , Pulsatile Flow , Umbilical Arteries/diagnostic imaging , Uterine Artery/diagnostic imaging
3.
Cells ; 9(3)2020 03 17.
Article in English | MEDLINE | ID: mdl-32192015

ABSTRACT

Chronic fetal hypoxia and infection are examples of adverse conditions during complicated pregnancy, which impact cardiac myogenesis and increase the lifetime risk of heart disease. However, the effects that chronic hypoxic or inflammatory environments exert on cardiac pacemaker cells are poorly understood. Here, we review the current evidence and novel avenues of bench-to-bed research in this field of perinatal cardiogenesis as well as its translational significance for early detection of future risk for cardiovascular disease.


Subject(s)
Cardiovascular Diseases/etiology , Fetal Hypoxia/metabolism , Heart/embryology , Inflammation/complications , Fetal Hypoxia/embryology , Humans , Hypoxia/complications , Risk
4.
Am J Physiol Regul Integr Comp Physiol ; 312(4): R492-R500, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28100476

ABSTRACT

Fetal insulin secretion is inhibited by acute hypoxemia. The relationship between prolonged hypoxemia and insulin secretion, however, is less well defined. To test the hypothesis that prolonged fetal hypoxemia impairs insulin secretion, studies were performed in sheep fetuses that were bled to anemic conditions for 9 ± 0 days (anemic, n = 19) and compared with control fetuses (n = 15). Arterial hematocrit and oxygen content were 34% and 52% lower, respectively, in anemic vs. control fetuses (P < 0.0001). Plasma glucose concentrations were 21% higher in the anemic group (P < 0.05). Plasma norepinephrine and cortisol concentrations increased 70% in the anemic group (P < 0.05). Glucose-, arginine-, and leucine-stimulated insulin secretion all were lower (P < 0.05) in anemic fetuses. No differences in pancreatic islet size or ß-cell mass were found. In vitro, isolated islets from anemic fetuses secreted insulin in response to glucose and leucine as well as control fetal islets. These findings indicate a functional islet defect in anemic fetuses, which likely involves direct effects of low oxygen and/or increased norepinephrine on insulin release. In pregnancies complicated by chronic fetal hypoxemia, increasing fetal oxygen concentrations may improve insulin secretion.


Subject(s)
Fetal Hypoxia/embryology , Fetal Hypoxia/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/embryology , Islets of Langerhans/metabolism , Anemia/embryology , Anemia/metabolism , Animals , Chronic Disease , Down-Regulation , Female , Insulin Secretion , Male , Sheep
5.
J Cereb Blood Flow Metab ; 37(4): 1362-1373, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27317658

ABSTRACT

Magnesium sulfate is now widely recommended for neuroprotection for preterm birth; however, this has been controversial because there is little evidence that magnesium sulfate is neuroprotective. Preterm fetal sheep (104 days gestation; term is 147 days) were randomly assigned to receive sham occlusion (n = 7), i.v. magnesium sulfate (n = 10) or saline (n = 8) starting 24 h before asphyxia until 24 h after asphyxia. Sheep were killed 72 h after asphyxia. Magnesium sulfate infusion reduced electroencephalograph power and fetal movements before asphyxia. Magnesium sulfate infusion did not affect electroencephalograph power during recovery, but was associated with marked reduction of the post-asphyxial seizure burden (mean ± SD: 34 ± 18 min vs. 107 ± 74 min, P < 0.05). Magnesium sulfate infusion did not affect subcortical neuronal loss. In the intragyral and periventricular white matter, magnesium sulfate was associated with reduced numbers of all (Olig-2+ve) oligodendrocytes in the intragyral (125 ± 23 vs. 163 ± 38 cells/field) and periventricular white matter (162 ± 39 vs. 209 ± 44 cells/field) compared to saline-treated controls ( P < 0.05), but no effect on microglial induction or astrogliosis. In conclusion, a clinically comparable dose of magnesium sulfate showed significant anticonvulsant effects after asphyxia in preterm fetal sheep, but did not reduce asphyxia-induced brain injury and exacerbated loss of oligodendrocytes.


Subject(s)
Brain/drug effects , Electroencephalography/drug effects , Fetal Hypoxia/drug therapy , Magnesium Sulfate/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Brain/embryology , Brain/pathology , Disease Models, Animal , Fetal Hypoxia/embryology , Fetal Hypoxia/pathology , Gestational Age , Magnesium Sulfate/administration & dosage , Magnesium Sulfate/blood , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/blood , Sheep
6.
Reprod Sci ; 22(8): 932-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25656500

ABSTRACT

This study was designed to determine the role of the MEK/ERK1/2 and PI3K/Akt pathways in cortisol production and endothelial nitric oxide synthase (eNOS) phosphorylation (peNOS) in the ovine fetal adrenal in response to long-term hypoxia (LTH). Pregnant ewes were maintained at high altitude (3820 m) for the last 100 days of gestation (dGa). At 138 to 142 dGa, fetal adrenal cortical cells (FACs) were collected from LTH and age-matched normoxic fetuses. Cortisol production and peNOS were measured in response to pretreatment with the MEK/ERK1/2 pathway inhibitor UO126 (UO) and adrenocorticotropic hormone (ACTH) stimulation. UO126 reduced ACTH-stimulated cortisol in both normoxic and LTH FACs. UO126 alone or in combination with ACTH reduced peNOS in the normoxic group, while ACTH alone or ACTH + UO inhibited peNOS in LTH FACs. Additionally, cortisol was measured in response to pretreatment with UO and treatment with 22R-hydroxycholesterol (22R-OHC) or water-soluble cholesterol (WSC) with and without ACTH stimulation. UO126 had no effect on 22R-OHC-treated cells, but reduced cortisol in cells treated with WSC and/or ACTH. Cortisol and peNOS were also measured in response to pretreatment with PI3K/Akt pathway inhibitor Wortmannin (WT) and ACTH stimulation. Wortmannin further increased cortisol under ACTH-stimulated conditions and, like ACTH, reduced peNOS in LTH but not normoxic FACs. Together, these data suggest that in LTH FACs MEK/ERK1/2 does not regulate peNOS but that UO acts downstream from eNOS, possibly at cholesterol transport, to affect cortisol production in LTH FACs, while the PI3K/Akt pathway, along with ACTH, regulates peNOS and plays a role in the fetal adaptation to LTH in FACs.


Subject(s)
Adrenal Cortex/drug effects , Adrenal Cortex/enzymology , Adrenocorticotropic Hormone/pharmacology , Fetal Hypoxia/enzymology , Hydrocortisone/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Adaptation, Physiological , Adrenal Cortex/embryology , Adrenal Cortex/physiopathology , Altitude , Animals , Cells, Cultured , Disease Models, Animal , Female , Fetal Hypoxia/embryology , Fetal Hypoxia/physiopathology , Gestational Age , Hydroxycholesterols/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Sheep , Signal Transduction/drug effects , Time Factors
7.
Comp Biochem Physiol A Mol Integr Physiol ; 163(3-4): 278-85, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22892476

ABSTRACT

Environmental stresses such as hypoxia can alter the development of the fetus that are manifested later in life, but the impact of early maternal hypoxia (MH) on cardiac performance, coronary flow and catecholamine responsiveness in adult offspring is less clear. The effects of exposure to chronic hypoxia (FIO(2)=0.12) in early intrauterine development (days E1-10) on cardiac performance of the adult offspring were estimated using the Langendorff-perfused rat heart. Cardiac dysfunction is presented as increased end-diastolic volume, with decreased ventricular stiffness in both male and female adult offspring (P<0.01 for both). While developed pressures were preserved in female MH rats, males demonstrated a decrease in systolic function, estimated as peak developed pressure (P<0.01). Challenge with dobutamine (300 nM), an adrenergic positive inotrope, increased cardiac work for control rats (P<0.01 for male and female rats) but not in MH-male rats. Coronary flow was reduced (P<0.01) and SERCA2 protein expression increased (2-fold, P<0.05) in female offspring, while eNOS protein levels were increased (2.5-fold, P<0.05) in females. This suggests gender-specific differences in compensatory responses to early MH, with female rats increasing calcium turnover to improve contractility and increasing coronary flow through increased expression of eNOS protein, partially restoring coronary perfusion while male rats show little compensation.


Subject(s)
Coronary Circulation , Fetal Hypoxia/physiopathology , Prenatal Exposure Delayed Effects , Animals , Capillaries/pathology , Coronary Vessels/pathology , Female , Fetal Hypoxia/embryology , Heart Rate , In Vitro Techniques , Liver/enzymology , Male , Maternal Exposure , Myocardial Contraction , Nitric Oxide Synthase Type III/metabolism , Pregnancy , Rats , Rats, Wistar , Regional Blood Flow
8.
Pediatr Dev Pathol ; 13(3): 192-201, 2010.
Article in English | MEDLINE | ID: mdl-19968489

ABSTRACT

Multiple congenital contractures, also known as fetal akinesia deformation sequence (FADS) and related terms, result from decreased fetal movement. The underlying etiologies are diverse and include central nervous system (CNS) dysgeneses and primary myopathies. Persistent central nuclei or the presence of myotubes is often regarded as evidence of a primary myopathic etiology; however, these findings are also associated with impaired fetal innervation. We report 7 fetuses, estimated gestational age 20 to 23 weeks, with persistent myotubular morphology, a change that could be (mis)interpreted as a primary myopathy. In 4 of the patients, CNS histology showed hypoxic/ischemic injury, polymicrogyria, mineralized neurons, and microinfarcts with or without loss of anterior horn neurons. FADS cases with polymicrogyria have frequently been interpreted as a consequence of a primary brain malformation. Only a few descriptions of FADS associate polymicrogyria with CNS hypoxic/ischemic injury, however, and do not describe skeletal muscle maturation delay. We hypothesize that this combination of neural and muscular pathology is an under-recognized pattern in FADS, which results from diffuse hypoxic/ischemic injury involving the brain and spinal cord during early to middle gestation.


Subject(s)
Contracture/pathology , Fetal Hypoxia/pathology , Hypoxia-Ischemia, Brain/pathology , Limb Deformities, Congenital/pathology , Malformations of Cortical Development/pathology , Muscle, Skeletal/abnormalities , Brain/abnormalities , Contracture/congenital , Diagnosis, Differential , Female , Fetal Hypoxia/embryology , Gestational Age , Humans , Hypoxia-Ischemia, Brain/embryology , Male , Malformations of Cortical Development/embryology , Muscle, Skeletal/embryology , Spinal Cord/abnormalities
9.
Neurotoxicology ; 30(2): 290-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19459226

ABSTRACT

Prenatal exposure to nicotine is associated with a variety of adverse outcomes. The present study investigated the effect of low doses of nicotine during pregnancy on fetal blood gases, cardiovascular system, and cellular activation in the brain. Intravenous administration of nicotine 10 or 25 microg/kg into ewe did not affect maternal blood gases, blood pressure, and heart rate. Maternal administration of nicotine also had no effect on fetal blood electrolyte concentrations, osmolality levels, and lactic acid levels. However, it significantly reduced fetal blood pO2 levels and oxygen saturation, increased fetal arterial blood pressure and decreased heart rate in utero. In addition, exposure to low doses of nicotine increased the expression of Fos in the paraventricular nucleus (PVN) and subfornic organ (SFO) in the fetal brain. The data demonstrated that even low doses of nicotine could impact significantly on fetal cardiovascular and central nervous systems, as well as oxygen status, and suggested a toxic risk to fetuses of exposure to low levels nicotine or second-hand smoking during pregnancy.


Subject(s)
Brain/cytology , Brain/drug effects , Fetal Hypoxia/chemically induced , Heart Rate, Fetal/drug effects , Nicotine/administration & dosage , Animals , Brain/embryology , Cardiovascular System/drug effects , Cardiovascular System/embryology , Female , Fetal Blood/drug effects , Fetal Blood/metabolism , Fetal Hypoxia/embryology , Fetal Hypoxia/metabolism , Fetus/drug effects , Fetus/embryology , Fetus/metabolism , Heart Rate, Fetal/physiology , Maternal-Fetal Exchange/drug effects , Maternal-Fetal Exchange/physiology , Nicotine/toxicity , Pregnancy , Sheep/embryology , Sheep/metabolism
10.
BMC Cardiovasc Disord ; 9: 1, 2009 Jan 06.
Article in English | MEDLINE | ID: mdl-19126206

ABSTRACT

BACKGROUND: Chronic hypoxia in utero (CHU) is one of the most common insults to fetal development and may be associated with poor cardiac recovery from ischaemia-reperfusion injury, yet the effects on normal cardiac mechanical performance are poorly understood. METHODS: Pregnant female wistar rats were exposed to hypoxia (12% oxygen, balance nitrogen) for days 10-20 of pregnancy. Pups were born into normal room air and weaned normally. At 10 weeks of age, hearts were excised under anaesthesia and underwent retrograde 'Langendorff' perfusion. Mechanical performance was measured at constant filling pressure (100 cm H2O) with intraventricular balloon. Left ventricular free wall was dissected away and capillary density estimated following alkaline phosphatase staining. Expression of SERCA2a and Nitric Oxide Synthases (NOS) proteins were estimated by immunoblotting. RESULTS: CHU significantly increased body mass (P < 0.001) compared with age-matched control rats but was without effect on relative cardiac mass. For incremental increases in left ventricular balloon volume, diastolic pressure was preserved. However, systolic pressure was significantly greater following CHU for balloon volume = 50 microl (P < 0.01) and up to 200 microl (P < 0.05). For higher balloon volumes systolic pressure was not significantly different from control. Developed pressures were correspondingly increased relative to controls for balloon volumes up to 250 microl (P < 0.05). Left ventricular free wall capillary density was significantly decreased in both epicardium (18%; P < 0.05) and endocardium (11%; P < 0.05) despite preserved coronary flow. Western blot analysis revealed no change to the expression of SERCA2a or nNOS but immuno-detectable eNOS protein was significantly decreased (P < 0.001) in cardiac tissue following chronic hypoxia in utero. CONCLUSION: These data offer potential mechanisms for poor recovery following ischaemia, including decreased coronary flow reserve and impaired angiogenesis with subsequent detrimental effects of post-natal cardiac performance.


Subject(s)
Fetal Hypoxia/embryology , Myocardium/enzymology , Nitric Oxide Synthase Type III/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Capillaries/metabolism , Capillaries/pathology , Coronary Circulation/physiology , Coronary Vessels/pathology , Female , Fetal Hypoxia/enzymology , Fetal Hypoxia/pathology , Fetal Hypoxia/physiopathology , In Vitro Techniques , Male , Myocardial Contraction/physiology , Myocardial Reperfusion Injury/congenital , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Ventricular Pressure/physiology
11.
Pediatr Res ; 65(2): 188-92, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047955

ABSTRACT

Intrauterine hypoxia impacts fetal growth and organ function. Inducible nitric oxide synthase (iNOS) and neuronal NOS (nNOS) expression was measured to assess the response of fetal hearts to hypoxic (HPX) stress. Pregnant guinea pigs were housed in a hypoxic chamber (10.5% O2 for 14 d, n = 17) or room air [normoxic (NMX), n = 17]. Hearts of anesthetized near-term fetuses were removed. mRNA [hypoxia-inducible factor, (HIF)-1alpha, 1beta, 2alpha, 3alpha, iNOS, and nNOS] and protein levels (HIF-1alpha, iNOS, and nNOS) of fetal cardiac left ventricles were quantified by real time polymerase chain reaction (PCR) and Western analysis, respectively. Cardiac nitrite/nitrate levels were measured in the presence/absence of L-N6-(1-iminoethyl)-lysine (L-NIL), an iNOS inhibitor, administered to pregnant sows. Hypoxia significantly increased fetal cardiac HIF-1alpha and -2alpha mRNA, HIF-1alpha protein but not HIF-3alpha or -1beta mRNA levels. Hypoxia increased both iNOS mRNA (by 5x) and protein (by 23%) levels but had no effect on nNOS levels. Nitrite/nitrate levels were increased in HPX hearts by 2.5x and decreased with L-NIL by 67 +/- 14%. Thus, up-regulation of iNOS-derived nitric oxide (NO) generation is an important mechanism by which fetal hearts respond to chronic hypoxic stress.


Subject(s)
Fetal Hypoxia/enzymology , Heart/embryology , Myocardium/enzymology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chronic Disease , Disease Models, Animal , Enzyme Induction , Enzyme Inhibitors/pharmacology , Female , Fetal Hypoxia/embryology , Gestational Age , Guinea Pigs , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysine/analogs & derivatives , Lysine/pharmacology , Nitrates/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitrites/metabolism , Pregnancy , RNA, Messenger/metabolism , Up-Regulation
12.
Izv Akad Nauk Ser Biol ; (4): 464-70, 2008.
Article in Russian | MEDLINE | ID: mdl-18771031

ABSTRACT

The effect of acute hypoxia (10% O2 for 30 min) on the rate of amnion rhythmic contractions and heart rate (HR) was studied in two age groups of European pond turtle (Emys orbicularis) embryos, on days 19-27 and 37-43 of incubation (30-40 and 60-70% of the period until hatching). Under the control conditions, the two age groups of embryos did not differ from each other in either parameter. Hypoxia did not affect significantly the amnion contraction rate but decreased the HR. The time course of the HR during hypoxia depended on the embryo age. The mean HR in the first group of embryos was 8% decreased by minutes 10-14 of hypoxia and did not change afterwards; in the second group, it was 18% decreased by minutes 3-8 and then partly or completely restored before the end of hypoxic exposure. It has been assumed that the capacity of European pond turtle embryos for restoring the normal HR when exposed to acute hypoxia during the second half of embryogenesis is related to the development of neurohumoral control mechanisms.


Subject(s)
Amnion/physiology , Fetal Hypoxia/physiopathology , Heart Rate/physiology , Myocardial Contraction/physiology , Turtles/physiology , Amnion/embryology , Animals , Embryo, Nonmammalian/physiology , Female , Fetal Hypoxia/embryology , Oxygen Consumption/physiology , Turtles/embryology
13.
Morfologiia ; 127(2): 31-6, 2005.
Article in Russian | MEDLINE | ID: mdl-16201329

ABSTRACT

Formation of the structure of striatum during two postnatal weeks in rats subjected to acute hypoxia during various periods of their embryonic development was studied using light microscopic (Nissl's stain and Golgi's silver nitrate impregnation) methods and electron microscopy. This study was supplemented by a simultaneous investigation of physiological development of the same population of rats. The data obtained demonstrated that prenatal hypoxia on day 13.5 of embryonic development (E13.5) led to a delayed neurogenesis (retardation in the development of neuropil elements and cell differentiation) as well as to the malformation of the structure of striatum (degeneration, in particular, chromatolysis of neurons and glial nodule formation). Morphometric analysis demonstrated that prenatal hypoxia on E13.5 resulted in a statistically significant decrease in cell number in the striatum, these changes being especially pronounced in large neurons. Prenatal hypoxia on E18.5, however, caused no significant changes in striatum. Structural changes in the striatum were shown to be accompanied by significant changes in the physiological development of animals. The data obtained demonstrated that the alteration of the conditions of embryogenesis (hypoxia) during the period of most intensive proliferation of forebrain neuroblasts resulted in the disturbances of the formation of both striatum nervous tissue of the organism as a whole during early postnatal ontogenesis.


Subject(s)
Embryonic Development/physiology , Fetal Hypoxia/physiopathology , Neostriatum/ultrastructure , Neurons/ultrastructure , Animals , Animals, Newborn , Cell Count , Female , Fetal Hypoxia/embryology , Gestational Age , Microscopy, Electron , Neostriatum/embryology , Neostriatum/growth & development , Neurons/cytology , Pregnancy , Rats , Rats, Wistar
14.
J Physiol ; 565(Pt 1): 35-41, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15760943

ABSTRACT

The intrauterine environment plays a powerful role in determining the life-long risk of cardiovascular disease. A number of stressors are well known to affect the development of the cardiovascular system in utero including over/under maternal nutrition, excess glucocorticoid and chronic hypoxia. Chronic fetal anaemia in sheep is a complex stressor that alters cardiac loading conditions, causes hypoxic stress and stimulates large changes in flow to specific tissues, including large increases in resting coronary blood flow and conductance. Decreased viscosity can account for approximately half of the increased flow. It appears that immature hearts are 'plastic' in that increases in coronary conductance with fetal anaemia persist into adulthood even if the anaemia is corrected before birth. These large changes in conductance are possible only through extensive remodelling of the coronary tree. Adult hearts that were once anaemic in utero are more resistant to hypoxic stress as adults but it is not known whether such an adaptation would be deleterious in later life. These studies indicate the need for investigation into the basic mechanisms of coronary tree remodelling in the immature myocardium. New information on these mechanisms is likely to lead to better prevention of and therapies for adult-onset coronary disease.


Subject(s)
Anemia, Neonatal/embryology , Anemia, Neonatal/physiopathology , Coronary Circulation , Fetal Heart/embryology , Fetal Heart/growth & development , Fetal Hypoxia/embryology , Fetal Hypoxia/physiopathology , Animals , Animals, Newborn , Female , Humans , Infant, Newborn , Pregnancy , Sheep
16.
Pediatr Neurol ; 30(4): 247-53, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15087102

ABSTRACT

Recirculation after transient in utero ischemia has previously been found to be accompanied by delayed deterioration of cellular bioenergetic state and of mitochondrial function in the fetal rat brain. Our objective was to assess whether the delayed deterioration is a result of the activation of mitochondrial permeability transition which is observed ultrastructurally as mitochondrial swelling. The respiratory activities and ultrastructure of isolated mitochondria and the cellular bioenergetic state in fetal rat brain were examined at the end of 30 minutes of in utero ischemia and after 1, 2, 3 and 4 hours of recirculation. Cyclosporin A, a potent and virtually specific mitochondrial permeability transition blocker, or vehicle was administered 1 hour after recirculation. In the vehicle-treated animals, the transient ischemia was associated with a delayed deterioration of cellular bioenergetic state and mitochondrial activities at 4 hours of recirculation. The number of swollen mitochondria increased markedly after 4 hours of recirculation. The deterioration and the swelling were prevented by cyclosporin A. The present study indicates that cyclosporin A treatment improves recovery of fetal brain energy metabolism and inhibits the mitochondrial swelling after transient in utero ischemia. The results suggest that mitochondria and mitochondrial permeability transition may be involved in the development of ischemic brain damage in the immature rat.


Subject(s)
Brain Damage, Chronic/embryology , Brain Ischemia/embryology , Fetal Hypoxia/embryology , Ion Channels/physiology , Mitochondrial Swelling/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain Damage, Chronic/pathology , Brain Ischemia/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Cyclosporine/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Fetal Hypoxia/pathology , Ion Channels/drug effects , Ion Channels/ultrastructure , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Pregnancy , Rats , Rats, Wistar , Reperfusion Injury/embryology , Reperfusion Injury/pathology
17.
Pediatr Neurol ; 26(2): 116-22, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11897475

ABSTRACT

Because accumulation of oxidative modification products seems to relate to aging and has not been fully studied in fetal brains, an immunohistochemical examination was performed on nine brains ranging from 22-40 weeks of gestation. These brains did not demonstrate lesions except hypoxic-ischemic changes. Advanced glycation end products and 4-hydroxynonenal are generally reported to be negative in neurons of normal young brains, but, in the present study, distinct positive immunoreaction was observed in neurons of fetal brains. Positive immunoreaction appeared earlier in the medulla oblongata than in the cerebrum, and 4-hydroxynonenal began to accumulate earlier than advanced glycation end products. As for glial cells, advanced glycation end products and 4-hydroxynonenal were positive in reactive astrocytes in mid- to late gestation. Because hypoxic-ischemic changes were observed in most of the patients, it is possible that oxidative stress caused by hypoxic-ischemic may be involved in the accumulation of these products in the fetal brain. 8-Hydroxy-2'-deoxyguanosine was negative even in patients demonstrating positive reaction for advanced glycation end products and 4-hydroxynonenal. In the fetal brain, DNA might be strongly protected from oxidative damage. 4-Hydroxynonenal is generally positive in the cytoplasm but was positive in the nucleus of immature neurons and glial cells in the present study, suggesting a unique metabolism of the fetal brain.


Subject(s)
Brain/embryology , Deoxyguanosine/analogs & derivatives , Fetal Hypoxia/embryology , Oxidative Stress/physiology , 8-Hydroxy-2'-Deoxyguanosine , Brain/pathology , DNA Damage , Deoxyguanosine/metabolism , Female , Fetal Hypoxia/pathology , Frontal Lobe/embryology , Frontal Lobe/pathology , Gestational Age , Glycation End Products, Advanced/metabolism , Humans , Infant, Newborn , Medulla Oblongata/embryology , Medulla Oblongata/pathology , Pregnancy , Superoxide Dismutase/metabolism , Temporal Lobe/embryology , Temporal Lobe/pathology
18.
Biol Neonate ; 71(6): 385-94, 1997.
Article in English | MEDLINE | ID: mdl-9197341

ABSTRACT

Our aim was to determine the effects of 12 h of hypoxaemia on cerebral blood flow (CBF) and cerebral O2 delivery in ovine fetuses at 0.6 gestation. During fetal hypoxaemia, induced by reduced uterine blood flow, fetal SaO2 and PaO2 were reduced (p < 0.01) from control values of 77.0 +/- 1.6% and 27.3 +/- 1.0 mm Hg, respectively, to 28.4 +/- 3.4% and 15.6 +/- 0.6 mm Hg; fetal pHa decreased from control values of 7.37 +/- 0.01 to 7.20 +/- 0.02 at 3 h, but returned to control values before 12 h. CBF (ml/min/100 g) was 2.0- to 2.6-fold higher (p < 0.01) than control values during hypoxaemia, but only 1.7-fold higher (p < 0.01) at 3 h when pHa was lowest. Cerebral O2 delivery (ml/min/100 g) was lower (p < 0.01) than control values of 3.15 +/- 0.29 at 1.5h (2.09 +/- 0.36) and 3h (1.84 +/- 0.22) of hypoxaemia and higher 1 h after hypoxaemia had ceased (3.81 +/- 0.22, p < 0.01). We conclude that the ovine fetus at 0.6 gestation is unable to sustain increased CBF and hence maintain cerebral O2 delivery during the first 6 h of hypoxaemia, a time which coincides with acidaemia; in contrast, at 6 and 12 h of hypoxaemia, when pHa was normal, cerebral O2 delivery was similar to control values. Reduced cerebral O2 delivery during the early, acidaemic, stages of hypoxaemia may lead to impaired neural development.


Subject(s)
Acidosis/physiopathology , Cerebrovascular Circulation/physiology , Fetal Diseases/physiopathology , Hypoxia, Brain/physiopathology , Hypoxia/physiopathology , Oxygen/metabolism , Acidosis/complications , Acidosis/embryology , Animals , Brain/blood supply , Female , Fetal Diseases/embryology , Fetal Hypoxia/complications , Fetal Hypoxia/embryology , Fetal Hypoxia/physiopathology , Hydrogen-Ion Concentration , Hypoxia/complications , Hypoxia/embryology , Hypoxia, Brain/complications , Hypoxia, Brain/embryology , Oxygen/blood , Pregnancy , Regional Blood Flow , Sheep , Time Factors , Uterus/blood supply , Vascular Resistance
19.
Morfologiia ; 107(7-12): 22-9, 1994.
Article in Russian | MEDLINE | ID: mdl-8680569

ABSTRACT

Kidneys of the foetuses in cases of normal pregnancy (37) and in gestosis (31) were studied in following gestative terms; 24-28, 30-36 and 39-40 weeks. Dynamics of specific volume of structural elements of cortical substance (CS) and of external and internal zones of medullary substance (MS) of kidney was established in normal pregnancy and in gestosis. In normal pregnancy specific volume of the CS vessels and tubules increases in proportion with gestative term. Differences in dynamics of specific volume of vessels and tubules were found in external and internal zones of MS. Specific volume of nephrogenic blastema and tubules of cortex and medulla reduces in gestosis, placental insufficiency and hypoxia of the foetus. Specific volume of the CS stroma and vessels of both zones also increases in combined gestosis, while in the one without combination with other diseases specific volume of the cortical and medullary stroma increases. Correlative analysis revealed that the growth of these structures of cortex and medulla is dependent on the increase of the number of the connections in proportion with the gestative term. Number, direction and significance of correlative connections, reflecting adaptive possibilities of the foetal kidney structures in conditions of prolonged hypoxia were found to alter in gestosis.


Subject(s)
Kidney/embryology , Pre-Eclampsia/embryology , Abortion, Criminal , Abortion, Therapeutic , Female , Fetal Hypoxia/embryology , Gestational Age , Humans , Organ Size , Pregnancy , Reference Values
20.
Behav Neural Biol ; 57(2): 93-102, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1586357

ABSTRACT

Norway rat fetuses (Rattus norvegicus) exhibit a stereotypic behavioral response when the umbilical cord is experimentally compressed with a vascular clamp. In this study, the development of the fetal behavioral response to cord compression was compared in altricial and precocial rodents, which differ markedly in neural and motor maturity at the time of birth. Both altricial and precocial species showed some form of behavioral response to umbilical cord compression. Fetuses of two altricial species, Norway rats and Mongolian gerbils (Meriones unguiculatus), expressed hyperactivity in response to cord compression throughout the last third of gestation. In contrast, precocial cotton rats (Sigmodon hispidus) and spiny mice (Acomys cahirinus) did not respond to cord compression until relatively late in gestation. Thus, altricial and precocial species do not express the cord compression response during comparable periods of neural development: precocial species are much more mature at the earliest expression of this behavior than altricial species. These findings are consistent with the interpretation that the cord compression response is a behavioral adaptation that can promote survival of the fetus in utero.


Subject(s)
Fetal Hypoxia/veterinary , Fetal Movement/physiology , Muridae/embryology , Animals , Fetal Hypoxia/embryology , Gestational Age , Motor Activity/physiology , Nervous System/embryology , Rats , Species Specificity , Stereotyped Behavior/physiology
SELECTION OF CITATIONS
SEARCH DETAIL