Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Cell Mol Neurobiol ; 32(6): 1031-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22410672

ABSTRACT

Compelling evidence shows that the offspring subjected to uncontrolled hyperlycemia during gestation display behavioral, neurochemical, and cellular abnormalities during adulthood. However, the molecular mechanisms underlying these defects remain elusive. Previous studies have shown an increased rate of apoptosis and a decreased index of neuronal proliferation associated with diabetic embryopathy. The aim of the present study was to determine whether impairments in apoptotic related proteins also occur in the developing central nervous system from non-malformed embryos exposed to uncontrolled gestational hyperglycemia. Pregnant rats injected with either streptozotocin or vehicle were killed on gestational day 19. Offspring brains were quickly removed to evaluate protein expression by Western blotting. Embryonic brains from diabetic rats exhibited a decrease in the cell survival p-Akt expression (52.83 ± 24.35%) and in the pro-apoptotic protein Bax (56.16 ± 6.47%). Moreover, the anti-apoptotic protein Bcl-2 showed a non-significant increase while there were no changes in Procaspase 3 or cleaved Caspase 3 proteins. The cytoskeleton proteins NF-200 and GFAP were also examined. Neither NF-200 nor GFAP showed differences in embryonic brains from diabetic rats compared to controls. Altogether, these results indicate that both proliferation and apoptotic pathways are decreased in the brain from the developing offspring of diabetic rats. Since selective neuronal apoptosis, as well as selective cell proliferation, are specifically involved in brain organogenesis, it is possible that simultaneous impairments during the perinatal period contribute to the long lasting alterations observed in the adult brain.


Subject(s)
Apoptosis , Brain/embryology , Brain/pathology , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/pathology , Down-Regulation , Signal Transduction , Animals , Blood Glucose/metabolism , Brain/enzymology , Caspase 3/metabolism , Cell Proliferation , Cytoskeleton/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Female , Fetus/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hyperglycemia/complications , Hyperglycemia/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
2.
Birth Defects Res A Clin Mol Teratol ; 94(10): 770-81, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22786762

ABSTRACT

Maternal diabetes and obesity are independent risk factors for neural tube defects, although it is unclear whether the effects are mediated by common pathogenic mechanisms. In this manuscript, we report a genome-wide survey of histone acetylation in neurulation stage embryos from mouse pregnancies with different metabolic conditions: maternal diabetes, and maternal consumption of a high fat content diet. We find that maternal diabetes, and independently, exposure to high-fat diet, are associated with increases and decreases of H3 and H4 histone acetylation in the embryo. Intriguingly, changes of H3K27 acetylation marks are significantly enriched near genes known to cause neural tube defects in mouse mutants. These data suggest that epigenetic changes in response to diet and metabolic condition may contribute to increased risk for neural tube defects in diabetic and obese pregnancies. Importantly, the responses to high-fat diet and maternal diabetes were distinct, suggesting that perturbed embryonic development under these conditions is mediated by different molecular pathways. This conclusion is supported by morphometric analyses that reveal a trend for maternal diabetes to delay embryonic development in the C57BL/6 strain, while high-fat diet appears to be associated with accelerated development. Taken together, our results link changes in histone acetylation to metabolic conditions during pregnancy, and implicate distinct epigenetic mechanisms in susceptibility to neural tube defects under conditions of maternal diabetes and obesity.


Subject(s)
Embryonic Development/genetics , Epigenesis, Genetic/physiology , Pregnancy in Diabetics/genetics , Algorithms , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/etiology , Diabetes, Gestational/genetics , Diet, High-Fat , Dietary Fats , Embryo, Mammalian , Embryonic Development/drug effects , Epigenomics , Female , Male , Mice , Mice, Inbred C57BL , Pregnancy , Pregnancy in Diabetics/pathology , Prenatal Exposure Delayed Effects/genetics , Streptozocin
3.
Rev Invest Clin ; 61(1): 41-52, 2009.
Article in Spanish | MEDLINE | ID: mdl-19507474

ABSTRACT

Human epidemiological and experimental animal studies have shown that suboptimal environments in the womb and during early neonatal life alter growth and may program offspring susceptibility to lifelong health problems. One of the most interesting and significant feature of developmental programming is the evidence that adverse consequences of altered intrauterine environments can be passed from first generation to second generation offspring. To obtain the transgenerational phenotype, a negative environment is required during fetal or early neonatal life, the physiologic phenotype or disease can be transmitted through the germ line and the subsequent generations are not directly exposed to the environmental factor. The hypothesis has become well accepted by compelling animal studies that define the outcome of specific challenges such as: 1) nutrient restriction or overfeeding during pregnancy and lactation; 2) uterine blood flow restriction; 3) fetal exposure to inappropriately high levels of glucocorticoids, and 4) experimental maternal diabetes. Maternal protein restriction in the rat adversely affects glucose metabolism of male and female second generation offspring in a gender and developmental time window-specific manner. Other studies have proved transgenerational passage of effects resulting from treatment of pregnant rats with dexamethasone by either maternal or paternal lines. First generation female diabetic offspring of F0 rats treated with streptozotocin during pregnancy had F2 offspring with altered glucose and carbohydrate metabolism. The studies suggest that the mechanisms involved in developmental programming are likely epigenetic rather than due to DNA sequence mutations. Many individuals all over the world experience undernutrition, stress, hyperglycemia and other negative environmental factors during pregnancy and/or lactation. Insult during this critical period of development may induce malprogramming and adversely alter not only the F1 generation but also future generations. Preventing or treating these conditions will help to minimize the risk of transmission of metabolic diseases to future generations.


Subject(s)
Metabolic Diseases/etiology , Prenatal Exposure Delayed Effects , Animals , Birth Weight , Diabetes Mellitus, Experimental/embryology , Diabetes, Gestational/metabolism , Diet , Disease Susceptibility , Epigenesis, Genetic , Female , Fetal Growth Retardation/etiology , Fetal Macrosomia/etiology , Glucocorticoids/adverse effects , Humans , Hyperinsulinism/embryology , Hyperinsulinism/metabolism , Infant, Newborn , Lactation , Male , Malnutrition/complications , Malnutrition/embryology , Metabolic Diseases/embryology , Metabolic Diseases/genetics , Metabolic Syndrome/embryology , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Models, Biological , Oxazoles/toxicity , Pregnancy , Pregnancy Complications/metabolism , Rats , Rats, Wistar
4.
Birth Defects Res A Clin Mol Teratol ; 82(10): 710-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18937363

ABSTRACT

BACKGROUND: Congenital defects are a major complication of diabetic pregnancy, and the leading cause of infant death in the first year of life. Caudal dysgenesis, occurring up to 200-fold more frequently in children born to diabetic mothers, is a hallmark of diabetic pregnancy. Given that there is also an at least threefold higher risk for heart defects and NTDs, it is important to identify the underlying molecular mechanisms for aberrant embryonic development. METHODS: We have investigated gene expression in a transgenic mouse model of caudal dysgenesis, and in a pharmacological model using situ hybridization and quantitative real-time PCR. RESULTS: We identified altered expression of several molecules that control developmental processes and embryonic growth. CONCLUSIONS: The results from our models point towards major implication of altered Wnt signaling in the pathogenesis of developmental anomalies associated with embryonic exposure to maternal diabetes.


Subject(s)
Diabetes Mellitus, Experimental/embryology , Fetal Diseases/metabolism , Neural Tube Defects/etiology , Pregnancy in Diabetics , Signal Transduction , Wnt Proteins/metabolism , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Embryo, Mammalian/metabolism , Female , Fetal Diseases/pathology , Mice , Mice, Transgenic , Neural Tube Defects/metabolism , Pregnancy , Wnt Proteins/genetics
5.
Toxicol Pathol ; 36(4): 529-51, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18467681

ABSTRACT

Type 2 diabetes mellitus (TTDM) is characterized by progressive loss of glucose control through multifactorial mechanisms. The search for an understanding of TTDM has relied on animal models since the realization of the importance of the pancreas in controlling plasma glucose concentration. Rodent models of TTDM are developed to express hyperglycemia and not islet degeneration per se. Degeneration of the islets of Langerhans with beta-cell loss is secondary to insulin resistance and is regarded as the more important lesion. Despite this, differences between models are seen in the development and progression of islet degeneration. Assessing the differences between the models is important to appreciate the various aspects of TTDM and understand their advantages as well as their deficiencies. Relevant animal models of TTDM provide opportunities to investigate important physiological and cell biological processes that may ultimately lead to development of targeted therapies. This article reviews the importance, advantages, and limitations of rodent models of TTDM in relation to the histopathological changes that characterize islet degeneration. Pathophysiological mechanisms that contribute to islet degeneration are also discussed and are placed into the context of changes in islet histological appearances.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Islets of Langerhans/pathology , Rodentia , Animals , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression , Humans , Insulin Resistance , Islets of Langerhans/embryology , Streptozocin , Transgenes
6.
J Clin Invest ; 87(3): 821-30, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1847938

ABSTRACT

Fetuses of streptozotocin-induced diabetic rats exhibited delayed lung maturation and a 40% reduction in the steady-state level of lung Na+,K(+)-ATPase alpha 1 subunit mRNA and Na+,K(+)-ATPase activity at 21 d of gestation. In in situ hybridization experiments the signal specific for Na(+)-pump alpha 1 subunit message was strongest above columnar epithelial cells of air-conducting structures. Strong labeling was also present above cuboidal cells lining the forming alveoli, but not above mesenchymal cells. Immunocytochemical localization of the protein paralleled the distribution of the mRNA. Mesenchymal cells were more abundant in fetal lungs of diabetic mothers, and thus the decreased overall levels of Na+,K(+)-ATPase may result from the observed morphological pulmonary immaturity. One day after birth there was no apparent difference in lung morphology at the light microscopic level, in the localization or the steady-state level of Na+,K(+)-ATPase alpha 1 isoform mRNA, or in enzyme activity. Na+,K(+)-ATPase has a likely role in the active phase of fluid absorption in the airways of newborns before the onset of breathing. Decreased fluid clearance and lack of thinning of the lung's connective tissue may contribute to the increased risk for respiratory distress in infants of diabetic mothers.


Subject(s)
Diabetes Mellitus, Experimental/embryology , Lung/embryology , Pregnancy in Diabetics/embryology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blotting, Northern , Bronchi/enzymology , Diabetes Mellitus, Experimental/metabolism , Epithelium/enzymology , Female , Gene Expression , Immunoenzyme Techniques , Lung/enzymology , Nucleic Acid Hybridization , Pregnancy , Pulmonary Alveoli/enzymology , Rats , Sodium-Potassium-Exchanging ATPase/genetics
7.
Reprod Toxicol ; 23(4): 486-98, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17482424

ABSTRACT

We aimed to investigate the extent to which maternal diabetes with or without folic acid (FA) supplementation affects mRNA levels and protein distribution of ROS scavenging enzymes, vascular endothelial growth factor-A (Vegf-A), folate binding protein-1 (Folbp-1), and apoptosis-associated proteins in the yolk sacs of rat embryos on gestational days 10 and 11. Commencing at conception and throughout pregnancy, half of the streptozotocin-diabetic and half of the control rats received daily FA injections. Maternal diabetes impaired vascular morphology and decreased CuZnSOD and GPX-1 gene expression in yolk sacs. Maternal diabetes also increased the levels of CuZnSOD protein, increased the Bax/Bcl-2 protein ratio and decreased Vegf-A protein distribution. FA treatment normalized vascular morphology, decreased mRNA levels of all three SOD isoforms and increased Vegf-A mRNA levels, rectified CuZnSOD protein distribution and Bax/Bcl-2 ratio. A teratogenic diabetic environment produces a state of vasculopathy, oxidative stress, and mild apoptosis in the yolk sac. FA administration normalizes vascular morphology, diminishes apoptotic rate, and increases Vegf-A gene expression and protein distribution in the yolk sac of diabetic rats.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Diabetes Mellitus, Experimental/drug therapy , Folic Acid/pharmacology , Oxidoreductases/metabolism , Pregnancy in Diabetics/drug therapy , Vascular Endothelial Growth Factor A/metabolism , Yolk Sac/drug effects , Animals , Antioxidants/therapeutic use , Caspase 3/metabolism , Catalase/metabolism , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Female , Folic Acid/therapeutic use , Gene Expression Regulation, Developmental/drug effects , Gestational Age , Glutathione Peroxidase/metabolism , Glycine N-Methyltransferase/metabolism , Oxidoreductases/genetics , Pregnancy , Pregnancy in Diabetics/genetics , Pregnancy in Diabetics/metabolism , Pregnancy in Diabetics/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Vascular Endothelial Growth Factor A/genetics , Yolk Sac/enzymology , Yolk Sac/pathology , bcl-2-Associated X Protein/metabolism
8.
Sci Rep ; 6: 37172, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27849017

ABSTRACT

Engulfment and cell motility 1 (ELMO1) functions as a guanine exchange factor for Rac1 and was recently found to protect endothelial cells from apoptosis. Genome wide association studies suggest that polymorphisms within human elmo1 act as a potential contributing factor for the development of diabetic nephropathy. Yet, the function of ELMO1 with respect to the glomerulus and how this protein contributes to renal pathology was unknown. Thus, this study aimed to identify the role played by ELMO1 in renal development in zebrafish, under hyperglycaemic conditions, and in diabetic nephropathy patients. In zebrafish, hyperglycaemia did not alter renal ELMO1 expression. However, hyperglycaemia leads to pathophysiological and functional alterations within the pronephros, which could be rescued via ELMO1 overexpression. Zebrafish ELMO1 crispants exhibited a renal pathophysiology due to increased apoptosis which could be rescued by the inhibition of apoptosis. In human samples, immunohistochemical staining of ELMO1 in nondiabetic, diabetic and polycystic kidneys localized ELMO1 in glomerular podocytes and in the tubules. However, ELMO1 was not specifically or distinctly regulated under either one of the disease conditions. Collectively, these results highlight ELMO1 as an important factor for glomerular protection and renal cell survival via decreasing apoptosis, especially under diabetic conditions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Diabetes Mellitus, Experimental/embryology , Kidney/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Humans , Kidney/pathology , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
Diabetes ; 65(1): 149-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26450995

ABSTRACT

High glucose in vivo and in vitro induces neural tube defects (NTDs). CITED2 (CBP/p300-interacting transactivator with ED-rich tail 2) is essential for neural tube closure. We explored the regulatory mechanism underlying CITED2 expression and its relationship with miRNA and endoplasmic reticulum (ER) stress. miR-200b levels were increased by maternal diabetes or high glucose in vitro, and this increase was abrogated by transgenic overexpression of superoxide dismutase 1 (SOD1) or an SOD1 mimetic. CITED2 was the target of miR-200b and was downregulated by high glucose. Two miR-200b binding sites in the 3'-untranslated region of the CITED2 mRNA were required for inhibiting CITED2 expression. The miR-200b mimic and a CITED2 knockdown mimicked the stimulative effect of high glucose on unfolded protein response (UPR) and ER stress, whereas the miR-200b inhibitor and CITED2 overexpression abolished high glucose-induced UPR signaling, ER stress, and apoptosis. The ER stress inhibitor, 4-phenylbutyrate, blocked CITED2 knockdown-induced apoptosis. Furthermore, the miR-200b inhibitor reversed high glucose-induced CITED2 downregulation, ER stress, and NTDs in cultured embryos. Thus, we showed a novel function of miR-200b and CITED2 in high glucose-induced UPR and ER stress, suggesting that miR-200b and CITED2 are critical for ER homeostasis and NTD formation in the developing embryo.


Subject(s)
Diabetes Mellitus, Experimental/embryology , Endoplasmic Reticulum Stress/genetics , MicroRNAs/genetics , Neural Tube Defects/embryology , RNA, Messenger/metabolism , Repressor Proteins/genetics , Trans-Activators/genetics , Unfolded Protein Response/genetics , 3' Untranslated Regions , Animals , Apoptosis/genetics , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Embryo, Mammalian , Female , Gene Knockdown Techniques , Immunoblotting , In Situ Nick-End Labeling , In Vitro Techniques , Mice , Mice, Transgenic , Neural Stem Cells , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Phenylbutyrates/pharmacology , Pregnancy , Repressor Proteins/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Trans-Activators/metabolism
10.
Diabetes ; 33(3): 281-4, 1984 Mar.
Article in English | MEDLINE | ID: mdl-6698318

ABSTRACT

Embryos from rats rendered diabetic with streptozotocin for 1 wk or more before conception were examined on day 11.5 of gestation (i.e., at the 26-29-somite stage of normal rat embryonic development). The studies were designed to assess whether poorly regulated maternal diabetes is associated with demonstrable abnormalities even during this early phase of embryogenesis. We found that manifest retardations in growth and development were invariably present as judged by significant reductions in crown-rump length and somite number, respectively. Total protein and DNA content of the embryos were also reduced, although not symmetrically, so that protein/DNA ratios were increased. Gross dysmorphogenic lesions in neural tissue disproportional to the overall growth retardation at 11.5 days could not be demonstrated. The findings suggest that maternal diabetes can compromise intra-uterine growth and development during the period preceding and coinciding with the establishment of circulation in the allantoic placenta. The possible multifactorial determinants remain to be elucidated. It also remains to be established whether the early embryotoxicity provides a setting conducive to the increased dysmorphogenesis that is traditionally recognized during the later stages of pregnancy complicated by diabetes.


Subject(s)
Diabetes Mellitus, Experimental/embryology , Fetal Growth Retardation/etiology , Morphogenesis , Animals , Blood Glucose/analysis , Body Weight , DNA/analysis , Diabetes Mellitus, Experimental/complications , Embryo, Mammalian/analysis , Female , Fetal Viability , Gestational Age , Pregnancy , Pregnancy in Diabetics/complications , Rats
11.
Diabetes ; 35(12): 1356-63, 1986 Dec.
Article in English | MEDLINE | ID: mdl-3770312

ABSTRACT

The effects of maternal diabetes on somatic development and activity of the polyol pathway were investigated during early and late gestation in a rat model for diabetic pregnancy. We studied embryo-fetal growth, mortality, and malformation rate in the offspring of nondiabetic rats and in the offspring of diabetic rats either treated with an aldose reductase inhibitor during gestation or left untreated. The numbers of embryo-fetal resorptions and malformations were significantly increased in the diabetic groups compared with the controls despite maternal treatment with the aldose reductase inhibitor. The sorbitol content of embryos and membranes from the diabetic rats in early gestation was increased 3-5 times over the control values. Similarly, elevated sorbitol levels were observed in the fetal livers and placentas of the diabetic rats in late gestation. Administration of the aldose reductase inhibitor to the pregnant diabetic rats normalized the sorbitol levels in the embryos and their membranes, whereas the sorbitol contents of the fetal livers and placentas were significantly lowered but not completely corrected. Furthermore, in the diabetic groups, no differences in sorbitol levels could be demonstrated between malformed and nonmalformed offspring. The results of this study suggest that enhanced polyol metabolism leading to increased sorbitol accumulation is present in the embryos of diabetic mothers as early as organogenesis. This accumulation is apparently not a major factor in the early developmental disturbances (e.g., growth perturbations and congenital malformations) of diabetic pregnancy.


Subject(s)
Diabetes Mellitus, Experimental/embryology , Pregnancy in Diabetics/metabolism , Sorbitol/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Embryo, Mammalian/analysis , Embryo, Mammalian/metabolism , Female , Fetal Growth Retardation/etiology , Fetus/analysis , Fetus/metabolism , Gestational Age , Humans , Liver/embryology , Pregnancy , Rats , Rats, Inbred Strains , Sorbitol/analysis
12.
Diabetes ; 49(1): 101-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10615956

ABSTRACT

Previous studies have suggested that reactive oxygen species (ROS) are mediators in the teratogenic process of diabetic pregnancy. In an animal model for diabetic pregnancy, offspring of the H rat strain show minor dysmorphogenesis when the mother is diabetic, whereas the offspring of diabetic rats of a sister strain, U, display major morphologic malformations. Earlier studies have shown that embryonic catalase activity is higher in the H than in the U strain, and maternal diabetes increases this difference in activity. The aim of this study was to characterize the influence of genetic predisposition on diabetic embryopathy by comparing the mRNA levels of ROS-metabolizing enzymes in the two strains. We determined the mRNA levels of catalase, glutathione peroxidase, gamma-glutamylcystein-synthetase, glutathione reductase, and superoxide dismutase (CuZn-SOD and Mn-SOD) in day 11 embryos of normal and diabetic H and U rats using semiquantitative reverse transcription-polymerase chain reaction. The mRNA levels of catalase and Mn-SOD were increased in H embryos as a response to maternal diabetes, and no differences were found for the other genes. Sequence analysis of the catalase promoter indicated that the difference in mRNA levels may result from different regulation of transcription. Sequence analysis of the catalase cDNA revealed no differences between the two strains in the translated region, suggesting that the previously observed difference in the electrophoretic mobility in zymograms is due to posttranslational modifications. An impaired expression of scavenging enzymes in response to ROS excess can thus be an integral part of a genetic predisposition to embryonic dysmorphogenesis.


Subject(s)
Catalase/genetics , Congenital Abnormalities/genetics , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/enzymology , RNA, Messenger/metabolism , Superoxide Dismutase/genetics , Animals , Diabetes Mellitus, Experimental/metabolism , Embryo, Mammalian/metabolism , Genetic Predisposition to Disease , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley
13.
Diabetes ; 51(9): 2811-6, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12196475

ABSTRACT

Maternal diabetes increases the risk of congenital malformations in the offspring of affected pregnancies. This increase arises from the teratogenic effect of the maternal diabetic milieu on the developing embryo, although the mechanism of this action is poorly understood. In the present study, we examined whether the vitamin A metabolite retinoic acid (RA), a common drug with well-known teratogenic properties, may interact with maternal diabetes to alter the incidence of congenital malformations in mice. Our results show that when treated with RA, embryos of diabetic mice are significantly more prone than embryos of nondiabetic mice to develop caudal regression, a defect that is highly associated with diabetic pregnancy in humans. By studying the vestigial tail (Wnt-3a(vt)) mutant, we provide evidence that Wnt-3a, a gene that controls the development of the caudal region, is directly involved in the pathogenic pathway of RA-induced caudal regression. We further show that the molecular basis of the increased susceptibility of embryos of diabetic mice to RA involves enhanced downregulation of Wnt-3a expression. This positive interaction between RA and maternal diabetes may have implications for humans in suggesting increased susceptibility to environmental teratogens during diabetic pregnancy.


Subject(s)
Abnormalities, Drug-Induced/embryology , Abnormalities, Multiple/etiology , Pregnancy in Diabetics/complications , Pregnancy in Diabetics/embryology , Teratogens , Tretinoin/adverse effects , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/embryology , Down-Regulation , Female , Genetic Predisposition to Disease/genetics , Mice , Mice, Inbred ICR , Mutation , Pregnancy , Proteins/genetics , Wnt Proteins , Wnt3 Protein , Wnt3A Protein
14.
Diabetes ; 47(12): 1960-6, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9836531

ABSTRACT

The increased rate of embryonic dysmorphogenesis in diabetic pregnancy is correlated with the severity and duration of the concurrent hyperglycemia during early gestation. Whole embryo culture was used to investigate a possible association of hyperglycemia-induced disturbances of embryo development with tissue levels of the three alpha-oxoaldehydes: glyoxal, methylglyoxal, and 3-deoxyglucosone (3-DG). Rat embryos exposed to high glucose levels in vitro showed severe dysmorphogenesis and a 17-fold increased concentration of 3-DG compared with control embryos cultured in a low glucose concentration. Exogenous 3-DG (100 micromol/l) added to the medium of control cultures yielded an increased embryonic malformation rate and a 3-DG concentration similar to that of embryos cultured in high glucose. Addition of superoxide dismutase (SOD) to the culture medium decreased the malformation rates of embryos exposed to either high glucose or high 3-DG levels, but it did not decrease the high embryonic 3-DG concentrations caused by either agent. Our results implicate the potent glycating agent 3-DG as a teratogenic factor in diabetic embryopathy. In addition, the anti-teratogenic effect of SOD administration appears to occur downstream of 3-DG formation, suggesting that 3-DG accumulation leads to superoxide-mediated embryopathy.


Subject(s)
Deoxyglucose/analogs & derivatives , Diabetes Mellitus, Experimental/metabolism , Embryo, Mammalian/drug effects , Teratogens/toxicity , Animals , Deoxyglucose/metabolism , Deoxyglucose/toxicity , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/embryology , Dose-Response Relationship, Drug , Embryo, Mammalian/abnormalities , Female , Glucose/administration & dosage , Glucose/pharmacology , Glyoxal/metabolism , Hyperglycemia/physiopathology , Male , Pregnancy , Pyruvaldehyde/metabolism , Rats
15.
Diabetes ; 46(7): 1189-97, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9200655

ABSTRACT

Neural tube defects are among the most common of the malformations associated with diabetic embryopathy. To study the molecular mechanisms by which neural tube defects occur during diabetic pregnancy, we have developed a new experimental system using pregnant diabetic mice. In this system, the rate of neural tube defects is about three times higher in embryos of diabetic mice than in embryos of nondiabetic mice. Most of the defects affected presumptive midbrain and hindbrain structures and included open defects (i.e., exencephaly) and gross maldevelopment. By semiquantitative reverse transcription-polymerase chain reaction and in situ hybridization, we found that expression of Pax-3, a gene required for neural tube closure in the area of the midbrain and hindbrain, is significantly reduced in the embryos of diabetic mice. The same regions of the neural tube where Pax-3 had been underexpressed were found subsequently to contain high concentrations of cells undergoing apoptosis. Reduced expression of Pax-3 appears to be responsible for this apoptosis because apoptotic cells were also found at sites of neural tube defects in embryos carrying null mutation of the Pax-3 gene. Finally, mouse strains that carry null mutations in Pax-3 develop neural tube defects that resemble the malformations that occur in embryos of diabetic mice. These results suggest that Pax-3 is an important developmental control gene, expression of which is disturbed in embryos of diabetic mice, and that as a consequence, apoptosis of the neural tube occurs. This pathway may be responsible for many of the neural tube defects resulting from diabetic pregnancy.


Subject(s)
Apoptosis/physiology , DNA-Binding Proteins/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Embryo, Mammalian/abnormalities , Neural Tube Defects/etiology , Pregnancy in Diabetics/complications , Transcription Factors , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/embryology , Embryo, Mammalian/physiology , Female , Gene Expression/genetics , In Situ Hybridization , Litter Size , Mice , Mice, Inbred ICR , Neural Tube Defects/diagnosis , Neural Tube Defects/genetics , PAX3 Transcription Factor , Paired Box Transcription Factors , Polymerase Chain Reaction , Pregnancy , Pregnancy in Diabetics/chemically induced , Pregnancy in Diabetics/embryology , RNA, Messenger/analysis , RNA, Messenger/genetics
16.
Diabetes ; 46(6): 1054-61, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9166679

ABSTRACT

An association between excess oxygen radical activity and disturbed embryogenesis in diabetic pregnancy has been suggested. In the present study, the protective capacity of vitamin E with different treatment regimens was investigated in early and late pregnancy of streptozotocin-induced diabetic rats. Daily gavaging of 0.2 g/kg or 0.8 g/kg of vitamin E exerted moderate protective effects. In contrast, treatment with a diet enriched with 2% (wt/wt) of vitamin E, yielding an approximate daily dosage of 2 g/kg of vitamin E, clearly restored both embryonic and fetal morphology. High-performance liquid chromatography measurement showed that maternal diabetes decreased embryonic content of vitamin E. When pregnant diabetic animals were supplemented with vitamin E, increased concentrations of the vitamin were found in maternal, embryonic, and fetal tissues. Thus, despite marked accumulation of vitamin E in maternal tissues, the compound apparently reached the conceptus. Thiobarbituric acid reactive substances (TBARS) were estimated as a measure of lipid peroxidation, and no changes were observed in maternal tissue, embryonic tissue, placenta, and fetal brain in the untreated diabetic group. In contrast, a fivefold increase of TBARS was found in fetal liver, a rise that was reduced with vitamin E treatment of the diabetic pregnant rats and completely normalized with 2% vitamin E in the diet. Congenital malformations caused by experimental diabetes can be prevented by antioxidants in vivo. These findings further corroborate the notion that an imbalance in the metabolism of free oxygen radicals is involved in the embryonic maldevelopment of diabetic pregnancy, and suggest a direction for prophylactic treatment in the future.


Subject(s)
Diabetes Mellitus, Experimental/embryology , Embryo, Mammalian/abnormalities , Embryonic and Fetal Development/drug effects , Pregnancy in Diabetics/embryology , Vitamin E/pharmacology , Animals , Blood Glucose/analysis , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Embryo, Mammalian/drug effects , Embryo, Mammalian/physiology , Embryonic and Fetal Development/physiology , Female , Gestational Age , Lipid Peroxidation/drug effects , Maternal-Fetal Exchange/drug effects , Organ Size/drug effects , Placenta/drug effects , Placenta/physiology , Pregnancy , Pregnancy Outcome , Pregnancy in Diabetics/drug therapy , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/analysis , Thiobarbituric Acid Reactive Substances/metabolism , Vitamin E/blood , Vitamin E/therapeutic use
17.
Congenit Anom (Kyoto) ; 45(3): 73-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16131363

ABSTRACT

The mechanism of diabetic embryopathy was investigated using in vitro experiments in a rat embryo culture system and in streptozotocin-induced diabetic pregnant rats. The energy metabolism in embryos during early organogenesis was characterized by a high rate of glucose utilization and lactic acid production (anaerobic glycolysis). Embryos uninterruptedly underwent glycolysis. When embryos were cultured with hypoglycemic serum, such embryos showed malformations in association with a significant reduction in glycolysis. In a diabetic environment, hyperglycemia caused an increased glucose flux into embryonic cells without a down-regulation of GLUT1 and an increased metabolic overload on mitochondria, leading to an increased formation of reactive oxygen species (ROS). Activation of the hexamine pathway, subsequently occurring with increased protein carbonylation and increased lipid peroxidation, also contributed to the increased generation of ROS. Hyperglycemia also caused a myo-inositol deficiency with a competitive inhibition of ambient glucose, which might have been associated with a diminished phosphoinositide signal transduction. In the presence of low activity of the mitochondrial oxidative glucose metabolism, the ROS scavenging system in the embryo was not sufficiently developed. Diabetes further weakened the antioxidant system, especially, the enzyme for GSH synthesis, gamma-GCS, thereby reducing the GSH concentration. GSH depletion also disturbed prostaglandin biosynthesis. An increased formation of ROS in a diminished GSH-dependent antioxidant system may, therefore, play an important role in the development of embryonic malformations in diabetes.


Subject(s)
Congenital Abnormalities/etiology , Diabetes Mellitus, Experimental/embryology , Embryo Culture Techniques/methods , Models, Animal , Pregnancy in Diabetics , Pregnancy, Animal , Animals , Apoptosis/drug effects , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Dinoprostone/metabolism , Dinoprostone/pharmacology , Female , Glucose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Models, Biological , Pregnancy , Rats , Reactive Oxygen Species
18.
J Matern Fetal Neonatal Med ; 28(4): 403-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24766077

ABSTRACT

OBJECTIVE: To evaluate the effects of folic acid (FA)-induced renal failure in young offspring of diabetic mothers. METHODS: The offspring of streptozotocin-induced diabetic dams were divided into four groups: CC (controls receiving vehicle); DC (diabetics receiving vehicle); CA (controls receiving FA solution, 250 mg/kg) and DA (diabetics receiving FA solution, 250 mg/kg). Renal function tests and morphometry results were analyzed. RESULTS: An increase in creatinine and urea levels was observed in CA and DA groups at two and five months. FA administration caused a significant reduction in the number of glomeruli in the offspring of diabetic dams. The diabetes group treated with FA had fewer glomeruli compared to controls at two and five months. FA caused an increase in the area of the urinary space both in controls and offspring of diabetic dams at two and five months. The number of glomeruli and area of the urinary space at two months were negatively correlated. CONCLUSIONS: Fetal programing promotes remarkable changes in kidney morphology and function in offspring. We suggest that the morphological changes in the kidneys are more pronounced when fetal programing is associated with newly acquired diseases, e.g. renal failure induced by FA.


Subject(s)
Acute Kidney Injury/embryology , Acute Kidney Injury/pathology , Diabetes Mellitus, Experimental/pathology , Fetal Development , Pregnancy in Diabetics/pathology , Prenatal Exposure Delayed Effects/pathology , Acute Kidney Injury/physiopathology , Animals , Blood Pressure , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/physiopathology , Female , Fetal Development/drug effects , Heart Rate , Kidney/physiopathology , Kidney Function Tests , Pregnancy , Pregnancy in Diabetics/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Wistar , Streptozocin
19.
Endocrinology ; 144(5): 1703-11, 2003 May.
Article in English | MEDLINE | ID: mdl-12697674

ABSTRACT

GLUT8 is a novel glucose transporter protein that is widely distributed in tissues including liver, a central organ of regulation of glucose homeostasis. The purpose of the current study was to investigate expression and regulation of hepatic GLUT8 mRNA and protein. Therefore, Northern and immunoblot analysis, semiquantitative RT-PCR, and immunofluorescence microscopy were performed using mouse livers at different stages of embryonic and postnatal development and in type 1 (streprozotocin treated) and type 2 (GLUT4 heterozygous) diabetes. GLUT8 mRNA and protein expression in embryonic liver was differentially regulated depending on the prenatal and postnatal developmental stage of the mice. Immunofluorescence microscopy of liver from wild-type mice demonstrated the highest levels of GLUT8 protein in perivenous hepatocytes pointing to its role in regulation of glycolytic flux. In diabetic scenarios, GLUT8 mRNA levels were correlated with circulating insulin; specifically, GLUT8 mRNA decreased in a type 1 diabetes model and increased in a type 2 diabetes model, suggesting a regulatory role for insulin in GLUT8 mRNA expression. While up-regulation of GLUT8 protein occurred in both models of diabetes, only in streptozotocin diabetic livers was GLUT8 zonation altered. These data demonstrate that GLUT8 mRNA and protein are differentially regulated in liver in response to physiologic and pathologic (diabetes) milieu and suggests that GLUT8 is intimately linked to glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Liver/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Animals , Animals, Newborn/metabolism , Diabetes Mellitus, Experimental/embryology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/embryology , Diabetes Mellitus, Type 2/embryology , Diabetes Mellitus, Type 2/genetics , Female , Fetus/metabolism , Gene Deletion , Glucose Transport Proteins, Facilitative , Glucose Transporter Type 4 , Heterozygote , Immunologic Techniques , Mice , Mice, Inbred CBA , Monosaccharide Transport Proteins/genetics , RNA, Messenger/metabolism , Reference Values
20.
J Endocrinol ; 139(1): 97-105, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8254299

ABSTRACT

It was observed in the course of other studies that rat fetal lung extracts inhibited proliferation of fetal lung cells in culture. The purpose of the present study was to isolate and characterize this cytostatic factor. It was found that fetal lungs contained a 16 kDa cytostatic factor and its concentration was twofold greater in fetal lungs of diabetic rats compared with control rats. This fetal lung cytostatic protein (FLCP) was purified by reversed-phase, heparin-affinity and gel filtration high-performance liquid chromatography and SDS-PAGE. The purified protein was electroblotted onto polyvinylidene difluoride membrane and subjected to sequence analysis. The amino-terminal sequence of this fetal lung cytostatic protein was P E P A K S A P A P X K G I G K Q X X K A X X K A ... and showed significant homology with histone H2B; however, the amino acid composition of FLCP suggested that it may be structurally distinct from histone H2B. Ion-spray mass spectrometry suggested that FLCP was made up of at least two species of the protein with molecular weights of 13,776.1 and 14,007.3 and was different from the molecular weight of rat histone H2B predicted by its cDNA sequence. The concentration of FLCP, based on amino acid compositions, was 0.32 nmol/g and 0.83 nmol/g wet fetal lung from non-diabetic and diabetic rats respectively. These findings suggest that the fetal rat lung produces a regulatory factor bearing considerable homology with but possibly different from histone H2B and that fetal lung immaturity during diabetic pregnancy might be contributed to by an increase in this factor.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Fetal Proteins/isolation & purification , Lung/embryology , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Diabetes Mellitus, Experimental/embryology , Electrophoresis, Polyacrylamide Gel , Fetal Proteins/genetics , Histones/genetics , Lung/chemistry , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL