Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38740370

RESUMO

The placenta is a temporary organ that provides communication between the mother and fetus. Maternal diabetes and abnormal placental angiogenesis may be linked. We investigated the angiogenesis mechanism resulting from VEGF and glucose stimulation in PECs obtained from human term placenta. Immunohistochemistry was performed to characterize PECs obtained from human term placenta. D-glucose was added to the medium containing PECs to establish normoglycemic and hyperglycemic conditions. The expression levels of VEGF, VEGFR-1 and VEGFR-2 genes and proteins in PECs from the control and experimental groups were analyzed by RT-PCR and Western blotting, respectively. With 48-hours incubation, gene expressions increased due to hyperglycemia, while protein levels increased due to the combined effect of VEGF and hyperglycemia. While VEGFR-2 gene expression and protein amounts increased in 24-hours due to the combined effect of VEGF and hyperglycemia, the effect of VEGF stimulation and glucose level on VEGFR-2 decreased in 48-hour incubation with time. VEGF, VEGFR-1 and VEGFR-2 genes and proteins were affected by hyperglycemic conditions in PECs. Hyperglycemia occurring in various conditions such as gestational diabetes mellitus and diabetes mellitus may affect VEGF, VEGFR-1 and VEGFR-2 genes and proteins of PECs derived from human term placenta.

2.
J Neurosci Res ; 102(1): e25247, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37800665

RESUMO

This study aimed to assess the focal cerebral ischemia-induced changes in learning and memory together with glutamatergic pathway in rats and the effects of treatment of the animals with transcranial Direct Current Stimulation (tDCS). One hundred male rats were divided into five groups as sham, tDCS, Ischemia/Reperfusion (IR), IR + tDCS, and IR + E-tDCS groups. Learning, memory, and locomotor activity functions were evaluated by behavioral experiments in rats. Glutamate and glutamine levels, alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionate receptor (AMPAR1), N-Methyl-D-Aspartate receptors (NMDAR1 and NMDAR2A), vesicular glutamate transporter-1 (VGLUT-1), and excitatory amino acid transporters (EAAT1-3) mRNA expressions in hippocampus tissues were measured. Ischemic areas were analyzed by TTC staining. The increase was observed in IR + tDCS, and IR + E-tDCS groups compared to the IR group while a significant decrease was observed in IR group compared to the sham in the locomotor activity, learning, and memory tests. While glutamate and glutamine levels, AMPAR1, NMDAR1, NMDAR2A, VGLUT1, and EAAT1 mRNA expressions were significantly higher in IR group compared to the sham group, it was found to be significantly lower in IR + tDCS and IR + E-tDCS groups compared to the IR group. EAAT2 and EAAT3 mRNA expressions were significantly higher in IR + tDCS and IR + E-tDCS groups compared to the IR group. Ischemic areas were significantly decreased in IR + tDCS and IR + E-tDCS groups compared to the IR group. Current results suggest that tDCS application after ischemia improves learning and memory disorders and these effects of tDCS may be provided through transporters that regulate glutamate levels.


Assuntos
Isquemia Encefálica , Estimulação Transcraniana por Corrente Contínua , Ratos , Masculino , Animais , Ratos Sprague-Dawley , Glutamina/metabolismo , Hipocampo/metabolismo , Isquemia Encefálica/terapia , Isquemia Encefálica/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/farmacologia , Isquemia/metabolismo , Glutamatos , RNA Mensageiro/metabolismo
3.
Int J Neurosci ; 132(3): 258-268, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32772609

RESUMO

INTRODUCTION: Reduction of blood flow below a threshold value in brain regions locally or globally is called cerebral ischemia and proper treatment requires either the restoration of normal blood flow and/or the administration of neuroprotective therapies. Human trophoblast progenitor cells (hTPCs) give rise to the placenta and are responsible for the invasion and vascular remodeling of the maternal vessels within the uterus. Here, we tested whether hTPCs promoted to differentiate along neural lineages may exhibit therapeutic properties in the setting of cerebral ischemia in vivo. MATERIALS AND METHODS: Cerebral ischemia was generated in rats via middle cerebral artery occlusion and, after 24 h, hTPCs were injected systemically via tail vein. Animals were sacrified at Day 3 or 11. RESULTS: TTC staining indicated that infarct volumes were smaller in hTPC treated animals. Visible myelin recovery was observed in the hTPC injected group with Luxol Fast Blue staining. On Day 11 after hTPC transplantation, DLX5 and VEGF expression, as well as 2 and 10 d after hTPC transplantation, NKX2.2 were significantly increased; while LHX6, Olig1, PDGFRα, VEGFR1 and VEGFR2 showed trends toward improved expression in brain tissue via immunoblot analysis. Neuron-like differentiated cells were positive for both NeuN and Cresyl Violet staining. CONCLUSION: Here, we demonstrate for the first time that hTPCs enhance the expression of angiogenic and neurogenic factors in rat brain after stroke. Transplantation of hTPCs could form the basis of novel therapeutic approaches for the treatment of stroke in the clinical setting.


Assuntos
Isquemia Encefálica , Acidente Vascular Cerebral , Animais , Isquemia Encefálica/tratamento farmacológico , Feminino , Humanos , Infarto da Artéria Cerebral Média , Neurogênese , Placenta/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Células-Tronco , Acidente Vascular Cerebral/terapia , Trofoblastos/metabolismo
4.
Acta Histochem ; 123(3): 151694, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33571695

RESUMO

Our research aimed to compare the epigenetic alterations between placentae of in vitro fertilization (IVF) patients and spontaneous pregnancies. Additionally, the expression levels of proliferation markers (PCNA, Ki67) and glucose transporter proteins (GLUT1, GLUT3) were assessed in control and IVF placentae to examine the possible consequences of epigenetic alterations on placental development. Control group placentae were obtained from spontaneous pregnancies of healthy women (n = 16). IVF placentae were obtained from fresh (n = 16) and frozen (n = 16) embryo transfer pregnancies. A group of maternal and paternal imprint genes H19, IGF2, IGF2, IGF2R, PHLDA2, PLAGL1, MASH2, GRB10, PEG1, PEG3, and PEG10 were detected by Real-Time PCR. Additionally, PCNA, Ki67, GLUT1, and GLUT3 protein levels were assessed by immunohistochemistry and western blot. In the fresh embryo transfer placenta group (fETP), gene expression of paternal PEG1 and PEG10 was upregulated compared with the control group. Increased gene expression in paternal PEG1 and maternal IGFR2 genes was detected in the frozen embryo transfer placenta group (FET) compared with the control group. Conversely, expression levels of H19 and IGF2 genes were downregulated in the FET group. On the other hand, GLUT3 and PCNA expression was increased in FET group placentae. IVF techniques affect placental imprinted gene expressions which are important for proper placental development. Imprinted genes are differently expressed in fresh ET placentae and frozen ET placentae. In conclusion, these data indicate that altered imprinted gene expression may affect glucose transport and cell proliferation, therefore play an important role in placental development.


Assuntos
Transferência Embrionária , Expressão Gênica/fisiologia , Fator de Crescimento Insulin-Like II/metabolismo , Placenta/metabolismo , Adulto , Transferência Embrionária/métodos , Feminino , Fertilização in vitro/métodos , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Masculino , Gravidez , Receptor IGF Tipo 2/metabolismo
5.
Biotech Histochem ; 96(8): 594-607, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33522283

RESUMO

Chronic kidney disease (CKD) is characterized by disruption of the glomerulus, tubule and vascular structures by renal fibrosis. Mesenchymal stem cells (MSC) ameliorate CKD. We investigated the effects of human amnion derived MSC (hAMSC) on fibrosis using expression of transforming growth factor beta (TGF-ß), collagen type I (COL-1) and bone morphogenetic protein (BMP-7). We also investigated levels of urinary creatinine and nitrogen in CKD. We used a 5/6 nephrectomy (5/6 Nx) induced CKD model. We used 36 rats in six groups of six animals: sham group, 5/6 Nx group, 15 days after 5/6 Nx (5/6 Nx + 15) group, 30 days after 5/6 Nx (5/6 Nx + 30) group, transfer of hAMSC 15 days after 5/6 Nx (5/6 Nx + hAMSC + 15) group and transfer of hAMSC 30 days after 5/6 Nx (5/6 Nx + hAMSC + 30) group. We isolated 106 hAMSC from the amnion and transplanted them via the rat tail vein into the 5/6 Nx + hAMSC + 15 and 5/6 Nx + hAMSC + 30 groups. We measured the expression of BMP-7, COL-1 and TGF-ß using western blot and immunohistochemistry, and their gene expressions were analyzed by quantitative real time PCR. TGF-ß and COL-1 protein, and gene expressions were increased in the 5/6 Nx +30 group compared to the 5/6 Nx + hAMSC + 30 group. Conversely, both protein and gene expression of BMP-7 was increased in 5/6 Nx + hAMSC + 30 group compared to the 5/6 Nx groups. Increased TGF-ß together with decreased BMP-7 expression may cause fibrosis by epithelial-mesenchymal transition due to chronic renal injury. Increased COL-1 levels cause accumulation of extracellular matrix in CKD. Levels of urea, creatinine and nitrogen were increased significantly in 5/6 Nx + 15 and 5/6 Nx + 30 groups compared to the hAMSC groups. We found that hAMSC ameliorate CKD.


Assuntos
Células-Tronco Mesenquimais , Insuficiência Renal Crônica , Âmnio , Animais , Fibrose , Rim/patologia , Nefrectomia , Ratos , Insuficiência Renal Crônica/patologia
6.
Int J Stem Cells ; 12(1): 151-161, 2019 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-30595007

RESUMO

BACKGROUND AND OBJECTIVES: The feature of chronic kidney failure (CKF) is loss of kidney functions due to erosion of healthy tissue and fibrosis. Recent studies showed that Mesenchymal stem cells (MSCs) differentiated into tubular epithelial cells thus renal function and structures renewed.Furthermore, MSCs protect renal function in CKF. Therefore, we aimed to investigate whether human amnion-derived mesenchymal stem cells (hAMSCs) can repair fibrosis and determine the effects on proliferation and apoptosis mechanisms in chronic kidney failure. METHODS AND RESULTS: In this study, rat model of CKF was constituted by applying Aristolochic acid (AA). hAMSCs were isolated from term placenta amnion membrane and transplanted into tail vein of rats. At the end of 30 days and 60 days of recovery period, we examined expressions of PCNA, p57 and Parp-1 by western blotting. Immunoreactivity of PCNA, Ki67, IL-6 and Collagen type I were detected by immunohistochemistry. Besides, apoptosis was detected by TUNEL. Serum creatinine and urea were measured. Expressions of PCNA and Ki67 increased in hAMSC groups compared with AA group. Furthermore, expressions of PARP-1 apoptosis marker and p57 cell cycle inhibitory protein increased in AA group significantly according to control, hAMSC groups and sham groups. IL-6 proinflammatory cytokine increased in AA group significantly according to control, hAMSCs groups and sham groups. Expressions of Collagen type I protein reduced in hAMSCs groups compared to AA group. After hAMSC treatment, serum creatinine and urea levels significantly decreased compared to AA group. After injection of hAMSC to rats, Masson’s Trichrome and Sirius Red staining showed fibrosis reduction in kidney. CONCLUSIONS: According to our results hAMSCs can be ameliorate renal failure.

7.
J Gynecol Obstet Hum Reprod ; 48(3): 193-199, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30553049

RESUMO

OBJECTIVE: The mammalian target of rapamycin (mTOR) signaling pathway has newly been recommended to be a nutrient sensor in the placenta. It is speculated that mTORC1 may be activated in diabetes, associated with increased placental nutrient availability. Thus, we aimed to investigate the mTOR signaling pathway both in diabetic and non-diabetic placenta and searched for the alterations of angiogenic factors VEGF, VEGFR1 and VEGFR2. METHODS: Streptozotocin (STZ) was administered by intravenous injection in doses of 60 mg/kg body weight and STZ injected rats were exposed to Everolimus (Rapamycin analog) and sacrificed at gestational days 14 and 20. mTORC1 and mTORC2 target proteins and angiogenic factors were analyzed at protein and mRNA levels in the placenta. Soluble VEGF A and Insulin protein levels were determined in blood serum. RESULTS: Placenta and embryo weights were altered after STZ and/or Rapamycin administration. mTOR pathway inhibition was confirmed by decreased p70S6K (Thr389) phosphorylation levels. We found that maternal diabetic environment led to an increase in Akt phosphorylation at 14th and decrease at 20th gestational days. Serum levels of Insulin in 14 th and 20 th days of gestation were decreased in Rapamycin and diabetic groups. On the other side serum levels of Soluble VEGF were increased in 14 th and decreased in 20 th days of pregnancy. CONCLUSION: According to our results, it might be suggested that angiogenesis related proteins will be related with placental growth regulation and mTOR may be a candidate pathway mediating the process in normal and diabetic pregnancy.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Imunossupressores/farmacologia , Neovascularização Fisiológica , Placenta/metabolismo , Complicações na Gravidez/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Fator A de Crescimento do Endotélio Vascular/sangue , Animais , Modelos Animais de Doenças , Feminino , Neovascularização Fisiológica/efeitos dos fármacos , Placenta/efeitos dos fármacos , Gravidez , Ratos , Ratos Wistar , Serina-Treonina Quinases TOR/efeitos dos fármacos
8.
Int J Mol Cell Med ; 7(4): 203-211, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31516879

RESUMO

Trophoblast stem cells develop from polar trophectoderm and give rise to the cells that generate the placenta. Trophoblast cells are responsible for the uterine invasion and vascular remodeling during the implantation of the embryo. However this knowledge is not yet to be confirmed for trophoblast progenitor cells (TPCs). In this study, we aimed to demonstrate that human TPCs (hTPCs) express and release angiogenic factors for the first time. TPCs were isolated from the term placenta. Then immunophenotyping was performed by FACS method by analyzing caudal type homeobox 2 (CDX2) and eomesodermin (EOMES). Immunofluorescence staining of CDX2 and EOMES was performed on these cells. Lastly, angiogenesis-related proteins were detected by western blot and ELISA assays. The isolated cells were positive for trophoblast stem cell markers CDX2 and EOMES in 92.5% and 92.7% of cells, respectively showing the characteristics of TPCs. The investigation of vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor 1 (VEGFR1), and vascular endothelial growth factor receptor 2 (VEGFR2) at protein and mRNA level in comparison with human umbilical vein endothelial cells (HUVECs), revealed that human TPCs (hTPCs) have higher levels of VEGF and VEGFR1 transcripts. Additionally soluble forms of VEGF and VEGFR1 were detected in supernatants of hTPCs. With this information, TPCs seem to be promising for regenerative cell therapies by increasing angiogenesis.

9.
Acta Histochem ; 117(2): 137-47, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25596037

RESUMO

Intrauterine growth restriction (IUGR) is a major clinical problem, which causes perinatal morbidity and mortality. One of the reasons for IUGR is abnormal placentation. In rats, fetal-placental exposure to maternally administered glucocorticoids decreases birth weight and placental weight. Proper placental development depends on the proliferation and differentiation of trophoblasts. Our knowledge about the mitotic regulators that play key roles in synchronizing these events is limited. Also the mechanisms underlying the placental growth inhibitory effects of glucocorticoids have not been elucidated. The aim of this study was to investigate the immunolocalization, mRNA and protein levels of proliferating cell nuclear antigen (PCNA), cyclin D3, p27 and p57 in normal and dexamethasone-induced IUGR Wistar rat placentas by reverse transcriptase polymerase chain reaction (RT-PCR), immunohistochemistry and Western blot. We also compared apoptotic cell numbers at the light microscopic level via terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) and transmission electron microscopy. Glucocorticoid levels were higher in IUGR rats than in control rats after 60 and 120min of injection. We showed reduced gene and protein expressions of PCNA and cyclin D3 and increased expressions of p27 and p57 in IUGR placentas compared to control placentas. Apoptotic cell number was higher in the placentas of the IUGR group. In brief we found that maternal dexamethasone treatment led to a shift from cell proliferation to apoptosis in IUGR placentas. Dexamethasone induced placental and embryonal abnormalities which could be associated with reduced expressions of PCNA and cyclin D3, increased expressions of p27 and p57 and increased rate of apoptosis in IUGR placentas.


Assuntos
Anti-Inflamatórios/efeitos adversos , Apoptose/efeitos dos fármacos , Ciclina D3/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Dexametasona/efeitos adversos , Retardo do Crescimento Fetal , Placenta , Antígeno Nuclear de Célula em Proliferação/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Dexametasona/farmacologia , Feminino , Retardo do Crescimento Fetal/induzido quimicamente , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Placenta/metabolismo , Placenta/patologia , Gravidez , Ratos , Ratos Wistar
10.
Ann Anat ; 198: 34-40, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25479925

RESUMO

During pregnancy, glucocorticoids (GCs) are used for fetal lung maturation in women at risk of preterm labor. Exogenous GCs do not have exclusively beneficial effects and repeated use of GCs remains controversial. It has been observed that GC exposed rats have smaller placentas and intrauterine growth retarded fetuses. In this study, we questioned whether or not glucocorticoids effect placental angiogenesis mechanisms. One of the most important signaling pathways among several downstream of VEGFR-2 is PI3K/Akt which subsequently activates the mammalian target of rapamycin. Therefore, we hypothesized that overexposure to GCs may adversely affect placental angiogenesis mechanisms by regulating pro-angiogenic factors and their receptors via Akt/mTOR pathway. According to our results Dexamethasone, a synthetic glucocorticoid, administration led to a decrease in VEGF, PIGF expression during pregnancy. VEGFR2 expression was first decreased at gestational day 14 and afterwards increased at gestational days 16, 18 and 20 in rat placentas. These results are in accordance with the reduced phosphorylation of Akt, 4EBP1 and p70S6K. Dexamethasone injection also resulted in a reduction of VEGF, VEGFR1, and VEGFR2 mRNA expression at gestational days 14 and 20, but PIGF mRNA expression was not altered. Growth retarded fetuses seen in Dexamethasone treated pregnancies, may be a result of altered angiogenic factor expression of the placenta mediated via altered mTOR pathway signaling.


Assuntos
Proteínas Angiogênicas/metabolismo , Dexametasona/farmacologia , Neovascularização Fisiológica/fisiologia , Placenta/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Glucocorticoides/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Placenta/efeitos dos fármacos , Gravidez , Prenhez , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
11.
Histol Histopathol ; 29(6): 743-56, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24346807

RESUMO

Diabetic pregnancy is associated with complications such as early and late embryonic death, fetal growth disorders, placental abnormalities, and embryonal-placental metabolic disorders. Excessive apoptosis and/or changes of proliferation mechanisms are seen as a major event in the pathogenesis of diabetes-induced embryonic death, placental weight and structural anomalies. Akt and ERK1/2 proteins are important for placental and fetal development associated with cellular proliferation and differentiation mechanisms. The mechanism underlying the placental growth regulatory effects of hyperglycemia have not been elucidated. Moreover, it is still not determined how Akt and ERK1/2 proteins related proliferation and apoptosis mechanisms are influenced by Streptozotocin (STZ) induced diabetic rat placental development. The aim of this study was to investigate the expression levels and spatio-temporal immunolocalizations of Akt, p-Akt, ERK1/2 and p-ERK1/2 proteins in normal and STZ-treated diabetic rat placental development. In order to compose the diabetic group, pregnant females were injected with a single dose of 40 mg/kg STZ intraperitonally seven days before their sacrifice at 12th, 14th, 16th, 18th and 20th day of their gestation. We found that maternal diabetic environment led to a decrease in ERK1/2 and Akt phosphorylation during rat placental development. It could be said that MAPK-ERK1/2 and PI3K/Akt cell signaling pathways are affected from hyperglycemic conditions in rat placentas. In conclusion, hyperglycemia-induced placental and embryonal developmental abnormalities could be associated with reduction of Akt and ERK1/2 phosphorylation.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Placenta/metabolismo , Gravidez em Diabéticas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose/fisiologia , Proliferação de Células/fisiologia , Feminino , Fosforilação , Placentação/fisiologia , Gravidez , Gravidez em Diabéticas/induzido quimicamente , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Estreptozocina
12.
Food Chem Toxicol ; 52: 129-36, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23168241

RESUMO

We aimed at investigating the effects of sulfite-induced lipid peroxidation and apoptosis mediated by secretory phospholipase A2 (sPLA2) on somatosensory evoked potentials (SEP) alterations in rats. Thirty male albino Wistar rats were randomized into three experimental groups as follows; control (C), sodium metabisulfite treated (S), sodium metabisulfite+quinacrine treated (SQ). Sodium metabisulfite (100 mg/kg/day) was given by gastric gavage for 5 weeks and 10 mg/kg/day quinacrine was applied as a single dose of intraperitoneal injection for the same period. The latencies of SEP components were significantly prolonged in the S group and returned to control levels following quinacrine administration. Plasma-S-sulfonate level was increased in S and SQ groups. TBARS levels in the S group were significantly higher than those detected in controls. Quinacrine significantly decreased brain TBARS levels in the SQ group compared with the S group. Quinacrine treatment did not have an effect on the increased sPLA2 level of the sulfite administered group. Immunohistochemistry showed that sulfite caused an increase in caspase-3 and TUNEL positive cells, restored to control levels via quinacrine administration. This study showed that sPLA2 might play a role in ingested sulfite-induced SEP alterations, oxidative stress, apoptotic cell death and DNA damage in the brain.


Assuntos
Encéfalo/efeitos dos fármacos , Fosfolipases A2 Secretórias/metabolismo , Quinacrina/farmacologia , Sulfitos/toxicidade , Animais , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Caspase 3/metabolismo , Potenciais Evocados/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Sulfitos/administração & dosagem , Ácidos Sulfônicos/sangue , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
13.
Cell Biochem Funct ; 30(1): 47-53, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21987433

RESUMO

The placenta is a glucocorticoid target organ, and glucocorticoids (GCs) are essential for the development and maturation of fetal organs. They are widely used for treatment of a variety of diseases during pregnancy. In various tissues, GCs have regulated by glucose transport systems; however, their effects on glucose transporters in the human placental endothelial cells (HPECs) are unknown. In the present study, HPECs were cultured 24 h in the presence or absence of 0.5, 5 and 50 µmol · l(-1) of synthetic GC triamcinolone (TA). The glucose carrier proteins GLUT 1, GLUT 3 and GC receptor (GR) were detected in the HPECs. We showed increased expression of GLUT 1 and GLUT 3 proteins and messenger RNA (mRNA) levels (p < 0.05) after 24-h cell culture in the presence of 0.5, 5 and 50 µmol · l(-1) of TA. In contrast, GR protein and mRNA expressions were down-regulated (p < 0.05) with 0.5, 5 and 50 µmol · l(-1) of TA 24-h cell culture. The results demonstrate that GCs are potent regulators of placental GLUT 1 and GLUT 3 expression through GR. Excessive exposure to GCs causes maternal and fetal hypoglycemia and diminished fetal growth. We speculate that to compensate for fetal hypoglycemia and diminished fetal growth, the expression of placental endothelial glucose transporters might be increased.


Assuntos
Células Endoteliais/efeitos dos fármacos , Glucocorticoides/farmacologia , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 3/metabolismo , Placenta/efeitos dos fármacos , Triancinolona/farmacologia , Células Endoteliais/metabolismo , Feminino , Humanos , Placenta/citologia , Placenta/metabolismo , Gravidez , Receptores de Glucocorticoides/metabolismo
14.
Acta Histochem ; 114(1): 31-40, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21371741

RESUMO

Intrauterine growth restriction (IUGR) is a major clinical problem which causes perinatal morbidity and mortality. Although fetuses with IUGR form a heterogeneous group, a major etiological factor is abnormal placentation. Despite the fact that placental development requires the coordinated action of trophoblast proliferation and differentiation, there are few studies on cell cycle regulators, which play the main roles in the coordination of these events. Moreover it is still not determined how mechanisms of coordination of proliferation and differentiation are influenced by dexamethasone-induced IUGR in the placenta. The aim of the study was to investigate the spatial and temporal immunolocalization of proliferating cell nuclear antigen (PCNA), Ki67, p27 and p57 in normal and IUGR placental development in pregnant Wistar rats. The study demonstrated altered expressions of distinct cell cycle proteins and cyclin dependent kinase inhibitors (CKIs) in IUGR placental development compared to control placental development. We found reduced immunostaining of PCNA and Ki67 and increased immunostaining of p27 and p57 in the dexamethasone-induced IUGR placental development compared to control placental development. In conclusion, our data show that the cell populations in the placenta stain for a number of cell cycle related proteins and that these staining patterns change as a function of both gestational age and abnormal placentation.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/análise , Inibidor de Quinase Dependente de Ciclina p57/análise , Dexametasona/farmacologia , Retardo do Crescimento Fetal/induzido quimicamente , Antígeno Ki-67/análise , Placenta/efeitos dos fármacos , Prenhez/fisiologia , Antígeno Nuclear de Célula em Proliferação/análise , Animais , Feminino , Saúde , Imuno-Histoquímica , Gravidez , Ratos , Ratos Wistar
15.
Folia Histochem Cytobiol ; 49(2): 325-34, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21744335

RESUMO

In various tissues, glucocorticoids (GCs) are known to downregulate glucose transport systems; however, their effects on glucose transporters (GLUTs) in the placenta of a diabetic rat are unknown. Glucocorticoid hormone action within the cell is regulated by the glucocorticoid receptor (GR). Thus, this study was designed to investigate the relationship between GR and glucose transporter expression in the placenta of the diabetic rat. Our immunohistochemical results indicated that GR and glucose transporter protein 1 (GLUT 1) are expressed ubiquitously in the trophoblast and endothelial cells of the labyrinthine zone, where maternal fetal transport takes place in the rat placenta. Expression of GR in the junctional zone of the rat placenta was detected in giant cells, and in some spongiotrophoblast cells, but not in the glycogen cells. GLUT 1 was present, especially in glycogen cells during early pregnancy, and in the spongiotrophoblast cells of the junctional zone during late pregnancy. Amounts of GR and GLUT 1 protein were increased towards the end of gestation both in the control and the diabetic placenta. However, at days 17 and 19 of gestation, only the placental GR protein was significantly increased in the streptozotocin-induced diabetic rats compared to control rats. Diabetes led to a significant decrease in placental weight at gestation day 15. In contrast, at gestational days 17 and 21, the weights of the diabetic placenta were significantly increased as compared with the controls. Moreover, diabetes induced fetus intrauterine growth retardation at gestational days 13, 17 and 21. In conclusion, the localization pattern of GR and GLUT 1 proteins in the same cell types led us to believe that there might be a relationship between GR and GLUT 1 expressions at the cellular level. GLUT 1 does not play a pivotal role in diabetic pregnancies. However, placental growth abnormalities during diabetic pregnancy may be related to the amount of GR.


Assuntos
Diabetes Mellitus Experimental/embriologia , Diabetes Mellitus Experimental/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Placenta/embriologia , Placenta/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/complicações , Feminino , Feto/metabolismo , Hiperglicemia/sangue , Hiperglicemia/complicações , Hiperglicemia/embriologia , Immunoblotting , Imuno-Histoquímica , Placenta/patologia , Gravidez , Transporte Proteico , Ratos , Estreptozocina
16.
Histochem Cell Biol ; 136(3): 267-78, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21789682

RESUMO

As cell cycle regulation is fundamental to the normal growth and development of the placenta, the aim of the present study was to determine the immunolocalizations of cell cycle related proteins, which have key roles in proliferation, differentiation and apoptosis during the development of the rat placenta. Here immunohistochemistry has been used to localize G1 cyclins (D1, D3, E), which are major determinants of proliferation, CIP/KIP inhibitors (p21, p27, p57), p53 as a master regulator and proliferating cell nuclear antigen in all cell types of the rat term placenta. The proportion of each cell type immunolabeled was counted. Cyclin D1 and cyclin D3 were present mostly in cells of the fetal aspect of the placenta, whereas the G1/S cyclin E was present only in the spongio- and labyrinthine trophoblast populations. Among the CIP/KIP inhibitors, p21 was present only in cells of the fetal aspect whereas p27 and p57 were found in all cell types studied. p53 was only found in a small proportion of cells with no co-localization of p53 and p21. The data suggest that the cells of the fetal side of the rat placenta still have some proliferation potential which is kept in check by expression of the CIP/KIP cell cycle inhibitors, whereas cells of the maternal aspect have lost this potential. Apoptosis is only marginal in the term rat placenta. In conclusion, proliferation and apoptosis in rat placental cells appears controlled mostly by the CIP/KIP inhibitors in late pregnancy.


Assuntos
Ciclina D1/metabolismo , Ciclina D3/metabolismo , Ciclina E/metabolismo , Ciclina G1/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Placenta/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Feminino , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Ratos Sprague-Dawley , Trofoblastos/metabolismo
17.
J Mol Histol ; 42(3): 237-49, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21512721

RESUMO

The placenta is a complicated tissue that lies between maternal and fetal compartments. Although the architecture of the human and rodent placentas differ a little in their details, their overall structures and the molecular mechanisms of placental developments are thought to be very similar. In rats, fetal-placental exposure to maternally administered glucocorticoids decreases birth weight and placental weight. The mechanism underlying the placental growth inhibitory effects of glucocorticoids have not been elucidated. Moreover it is still not determined that how Akt and ERK1/2 proteins related proliferation and apoptosis mechanisms are influenced by dexamethasone-induced IUGR (Intrauterine Growth Restriction) placentas. The aim of this study was to investigate the expression levels and spatio-temporal immunolocalizations of Akt, p-Akt, ERK1/2 and p-ERK1/2 proteins in normal and dexamethasone treated placental development in pregnant Wistar rats. Pregnant rats were subcutaneously injected with 100 µg/kg dexamethasone 21-acetate in 0.1 ml 10% ethanol on day 10 and 12 of gestation. Afterwards injection was continued as 200 µg/kg until they were killed on day 12 (injection started on day 10), 14, 16, 18 and 20 (injections started on day 12) of pregnancy. Placental and embryonal tissues were collected for immunohistochemistry and Western blot analysis. We found that maternal dexamethasone treatment led to a decrease in ERK1/2 and Akt activation during rat placental development. The decrease in Akt and ERK1/2 activations may result with cell survival inhibition or apoptosis stimulation. Hence, dexamethasone induced placental and embryonal developmental abnormalities could be associated with reduction of Akt and ERK1/2 activation.


Assuntos
Dexametasona/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Placenta/efeitos dos fármacos , Placentação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Embrião de Mamíferos/anatomia & histologia , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/genética , Feminino , Placenta/anatomia & histologia , Gravidez , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Ratos Wistar
18.
Biophys Chem ; 119(3): 234-9, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16233946

RESUMO

Heparin binding to human low density lipoproteins (LDL) and the effect of heparin on the ability of LDL to bind to the LDL receptor has been investigated. Emphasis has been made on the physiological conditions of temperature, pH and the ionic strength. Intrinsic fluorescence spectroscopy of LDL has been applied to follow heparin binding. Fluorescence anisotropy has been measured to describe the changes in apoB and dansyl-heparin dynamics upon binding. Eu3+-labeled LDL binding to the intact LDL receptor has been monitored by time-resolved fluorescence spectroscopy technique. We have found that heparin binds to LDL under the physiological conditions, probably by Van der Waals interactions and hydrogen bonding. Temperature seems to be the most important factor influencing the interaction. Furthermore, the presence of heparin inhibits LDL binding to the intact LDL receptor that might have consequences on the cholesterol metabolism in vivo.


Assuntos
Fibrinolíticos/farmacologia , Heparina/farmacologia , Lipoproteínas LDL/metabolismo , Apolipoproteínas B/metabolismo , Células Cultivadas/efeitos dos fármacos , Európio/metabolismo , Polarização de Fluorescência , Heparina/análogos & derivados , Heparina/metabolismo , Humanos , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Rim/metabolismo , Lipoproteínas LDL/química , Receptores de LDL/metabolismo , Espectrometria de Fluorescência , Temperatura
19.
J Cardiovasc Pharmacol ; 45(5): 418-30, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15821437

RESUMO

The GPI-anchored protein T-cadherin was found to be an atypical LDL binding site that is expressed in various types of cells, including endothelial cells, smooth muscle cells, and neurons. Notably, the expression of T-cadherin was reduced in numerous types of cancers, although it was up-regulated in tumor-penetrating blood vessels, atherosclerotic lesions, and during neointima formation. Despite these intriguing findings, our knowledge of the physiological role and the signal transduction pathways associated with this protein is limited. Therefore, T-cadherin was overexpressed in the human umbilical vein-derived endothelial cell line EA.hy926, the human embryonic kidney cell line HEK293, and LDL-initiated signal transduction, and its consequences were elucidated. Our data revealed that T-cadherin serves as a receptor specifically for LDL. Following LDL binding to T-cadherin, mitogenic signal transduction was initiated that involved activation of PLC and IP3 formation, which subsequently yielded intracellular Ca2+ mobilization. Downstream to these early phenomena, activation of tyrosine kinase(s) Erk 1/2 kinase, and the translocation of NF kappa B toward the nucleus were found. Finally, overexpression of T-cadherin in HEK293 cells resulted in accelerated cell proliferation in an LDL-dependent manner, although cell viability was not influenced. Because LDL uptake was not facilitated by T-cadherin, our data suggest that T-cadherin serves as a signaling receptor for LDL that facilitates an LDL-dependent mitogenic signal in the vasculature.


Assuntos
Caderinas/metabolismo , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases/metabolismo , Western Blotting , Caderinas/genética , Linhagem Celular , Endotélio Vascular/citologia , Fluorometria , Humanos , Imuno-Histoquímica , Proteína Quinase 1 Ativada por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Veias Umbilicais/citologia
20.
Br J Pharmacol ; 140(8): 1351-62, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14645143

RESUMO

The endocannabinoid anandamide has been reported to affect neuronal cells, immune cells and smooth muscle cells via either CB1 or CB2 receptors. In endothelial cells, the receptors involved in activating signal transduction are still unclear, despite the fact that anandamide is produced in this cell type. The present study was designed to explore in detail the effect of this endocannabinoid on Ca2+ signaling in single cells of a calf pulmonary endothelial cell line. Anandamide initiated a transient Ca2+ elevation that was prevented by the CB2 receptor antagonist SR144528, but not by the CB1 antagonist SR141716A. These data were confirmed by molecular identification of the bovine CB2 receptor in these endothelial cells by partial sequencing. The phospholipase C inhibitor 1-[6-[[(17beta)-3-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl]-1H-pyrrole-2,5dione and the inositol 1,4,5-trisphosphate receptor antagonist 2-aminoethoxydiphenylborate prevented Ca2+ signaling in response to anandamide. Using an improved cameleon probe targeted to the endoplasmic reticulum (ER), fura-2 and ratiometric-pericam, which is targeted to the mitochondria, anandamide was found to induce Ca2+ depletion of the ER accompanied by the activation of capacitative Ca2+ entry (CCE) and a transient elevation of mitochondrial Ca2+. These data demonstrate that anandamide stimulates the endothelial cells used in this study via CB2 receptor-mediated activation of phospholipase C, formation of inositol 1,4,5-trisphosphate, Ca2+ release from the ER and subsequent activation of CCE. Moreover, the cytosolic Ca2+ elevation was accompanied by a transient Ca2+ increase in the mitochondria. Thus, in addition to its actions on smooth muscle cells, anandamide also acts as a powerful stimulus for endothelial cells.


Assuntos
Ácidos Araquidônicos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio , Moduladores de Receptores de Canabinoides/farmacologia , Endotélio Vascular/metabolismo , Receptor CB2 de Canabinoide/fisiologia , Fosfolipases Tipo C/metabolismo , Animais , Ácidos Araquidônicos/metabolismo , Sequência de Bases , Cálcio/metabolismo , Moduladores de Receptores de Canabinoides/metabolismo , Bovinos , Linhagem Celular , Endocanabinoides , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Dados de Sequência Molecular , Alcamidas Poli-Insaturadas , Artéria Pulmonar/citologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fosfolipases Tipo C/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...