Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(2): e29236, 2012.
Article in English | MEDLINE | ID: mdl-22363401

ABSTRACT

BACKGROUND: Inadequate placental development is associated with a high incidence of early embryonic lethality and serious pregnancy disorders in both humans and mice. However, the lack of well-defined trophoblast-specific gene regulatory elements has hampered investigations regarding the role of specific genes in placental development and fetal growth. PRINCIPAL FINDINGS: By random assembly of placental enhancers from two previously characterized genes, trophoblast specific protein α (Tpbpa) and adenosine deaminase (Ada), we identified a chimeric Tpbpa/Ada enhancer that when combined with the basal Ada promoter provided the highest luciferase activity in cultured human trophoblast cells, in comparison with non-trophoblast cell lines. We used this chimeric enhancer arrangement to drive the expression of a Cre recombinase transgene in the placentas of transgenic mice. Cre transgene expression occurred throughout the placenta but not in maternal organs examined or in the fetus. SIGNIFICANCE: In conclusion, we have provided both in vitro and in vivo evidence for a novel genetic system to achieve placental transgene expression by the use of a chimeric Tpbpa/Ada enhancer driven transgene. The availability of this expression vector provides transgenic opportunities to direct the production of desired proteins to the placenta.


Subject(s)
DNA/genetics , Gene Targeting , Integrases/metabolism , Placenta/metabolism , Recombination, Genetic/genetics , Adenosine Deaminase/genetics , Animals , Cell Line , Cells, Cultured , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation , Genes, Reporter/genetics , Humans , Luciferases/genetics , Male , Mice , Mice, Transgenic , Organ Specificity/genetics , Pregnancy , Pregnancy Proteins/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Transgenes/genetics , Trophoblasts/metabolism
2.
J Immunol ; 186(10): 6024-34, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21482739

ABSTRACT

Preeclampsia (PE) is a life-threatening hypertensive disorder of pregnancy. Elevated circulating endothelin-1 (ET-1) is associated with the disease. However the molecular basis of increased ET-1 production and its role in PE are unknown. This study aimed to investigate the causative factors, pathological role of elevated ET-1 production in PE, and the underlying mechanisms. In this study, we found that IgG from women with PE, in contrast to IgG from normotensive pregnant women, induced preproET-1 mRNA expression via angiotensin II type 1 receptor activation in kidneys and placentas in pregnant mice. The ET-A receptor-specific antagonist BQ123 significantly attenuated autoantibody-induced hypertension, proteinuria, and renal damage in pregnant mice, demonstrating that autoantibody-induced ET-1 production contributes to pathophysiology. Mechanistically, we discovered that IL-6 functioned downstream of TNF-α signaling, contributing to increased ET-1 production in pregnant mice. IL-6 blockade inhibited preeclamptic features in autoantibody-injected pregnant mice. Extending the data to human studies, we found that IL-6 was a key cytokine underlying ET-1 induction mediated by IgG from women with PE in human placental villous explants and that endothelial cells are a key source of ET-1. Overall, we provide human and mouse studies showing that angiotensin II type I receptor-agonistic autoantibody is a novel causative factor responsible for elevated ET-1 production and that increased TNF-α/IL-6 signaling is a key mechanism underlying increased ET-1 production and subsequent maternal features. Significantly, our findings revealed novel factors and signaling cascades involved in ET-1 production, subsequent disease symptom development, and possible therapeutic intervention in the management of PE.


Subject(s)
Autoantibodies/immunology , Endothelin-1/metabolism , Interleukin-6/metabolism , Pre-Eclampsia/immunology , Pre-Eclampsia/metabolism , Receptor, Angiotensin, Type 1/immunology , Receptor, Angiotensin, Type 1/metabolism , Animals , Disease Models, Animal , Endothelin-1/biosynthesis , Endothelin-1/blood , Endothelin-1/genetics , Female , Humans , Immunoglobulin G/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Kidney/metabolism , Mice , Placenta/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
Hypertension ; 55(5): 1246-53, 2010 May.
Article in English | MEDLINE | ID: mdl-20351341

ABSTRACT

Preeclampsia is a prevalent life-threatening hypertensive disorder of pregnancy for which the pathophysiology remains largely undefined. Recently, a circulating maternal autoantibody, the angiotensin II type I (AT(1)) receptor agonistic autoantibody (AA), has emerged as a contributor to disease features. Increased circulating maternal tumor necrosis factor alpha (TNF-alpha) is also associated with the disease; however, it is unknown whether this factor directly contributes to preeclamptic symptoms. Here we report that this autoantibody increases the proinflammatory cytokine TNF-alpha in the circulation of AT(1)-AA-injected pregnant mice but not in nonpregnant mice. Coinjection of AT(1)-AA with a TNF-alpha neutralizing antibody reduced cytokine availability in AT(1)-AA-injected pregnant mice. Moreover, TNF-alpha blockade in AT(1)-AA-injected pregnant mice significantly attenuated the key features of preeclampsia. Autoantibody-induced hypertension was reduced from 131+/-4 to 110+/-4 mm Hg, and proteinuria was reduced from 212+/-25 to 155+/-23 microg of albumin per milligram of creatinine (both P<0.05). Injection of AT(1)-AA increased the serum levels of circulating soluble fms-like tyrosine kinase 1 and soluble endoglin (34.1+/-5.1, 2.4+/-0.3 ng/mL, respectively) and coinjection with the TNF-alpha blocker significantly reduced their levels (21.7+/-3.4 and 1.2+/-0.4 ng/mL, respectively). Renal damage and placental abnormalities were also decreased by TNF-alpha blockade. Lastly, the elevated circulating TNF-alpha in preeclamptic patients is significantly correlated with the AT(1)-AA bioactivity in our patient cohort. Similarly, the autoantibody, through AT(1) receptor-mediated TNF-alpha induction, contributed to increased soluble fms-like tyrosine kinase 1, soluble endoglin secretion, and increased apoptosis in cultured human villous explants. Overall, AT(1)-AA is a novel candidate that induces TNF-alpha, a cytokine that may play an important pathogenic role in preeclampsia.


Subject(s)
Autoantibodies/physiology , Pre-Eclampsia/physiopathology , Receptor, Angiotensin, Type 1/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cesarean Section , Disease Models, Animal , Female , Humans , Mice , Pre-Eclampsia/immunology , Pre-Eclampsia/prevention & control , Pregnancy , Receptor, Angiotensin, Type 1/immunology , Reference Values , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood
4.
Circulation ; 121(3): 436-44, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20065159

ABSTRACT

BACKGROUND: Preeclampsia is a prevalent life-threatening hypertensive disorder of pregnancy. The circulating antiangiogenic factor, soluble endoglin (sEng), is elevated in the blood circulation of women with preeclampsia and contributes to disease pathology; however, the underlying mechanisms responsible for its induction in preeclampsia are unknown. METHODS AND RESULTS: Here, we discovered that a circulating autoantibody, the angiotensin receptor agonistic autoantibody (AT(1)-AA), stimulates sEng production via AT(1) angiotensin receptor activation in pregnant mice but not in nonpregnant mice. We subsequently demonstrated that the placenta is a major source contributing to sEng induction in vivo and that AT(1)-AA-injected pregnant mice display impaired placental angiogenesis. Using drug screening, we identified tumor necrosis factor-alpha as a circulating factor increased in the serum of autoantibody-injected pregnant mice contributing to AT(1)-AA-mediated sEng induction in human umbilical vascular endothelial cells. Subsequently, among all the drugs screened, we found that hemin, an inducer of heme oxygenase, functions as a break to control AT(1)-AA-mediated sEng induction by suppressing tumor necrosis factor-alpha signaling in human umbilical vascular endothelial cells. Finally, we demonstrated that the AT(1)-AA-mediated decreased angiogenesis seen in human placenta villous explants was attenuated by tumor necrosis factor-alpha-neutralizing antibodies, soluble tumor necrosis factor-alpha receptors, and hemin by abolishing both sEng and soluble fms-like tyrosine kinase-1 induction. CONCLUSIONS: Our findings demonstrate that AT(1)-AA-mediated tumor necrosis factor-alpha induction, by overcoming its negative regulator, heme oxygenase-1, is a key underlying mechanism responsible for impaired placental angiogenesis by inducing both sEng and soluble fms-like tyrosine kinase-1 secretion from human villous explants. Our results provide important new targets for diagnosis and therapeutic intervention in the management of preeclampsia.


Subject(s)
Autoantibodies/blood , Intracellular Signaling Peptides and Proteins/blood , Pre-Eclampsia/blood , Pre-Eclampsia/immunology , Receptor, Angiotensin, Type 1/immunology , Tumor Necrosis Factor-alpha/blood , Animals , Autoantibodies/pharmacology , Chorionic Villi/blood supply , Chorionic Villi/immunology , Chorionic Villi/metabolism , Endoglin , Endothelial Cells/metabolism , Female , Heme Oxygenase-1/metabolism , Humans , Immunoglobulin G/blood , Immunoglobulin G/pharmacology , Mice , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/immunology , Placenta/blood supply , Placenta/immunology , Placenta/metabolism , Pregnancy , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , Solubility
5.
J Exp Med ; 206(12): 2809-22, 2009 Nov 23.
Article in English | MEDLINE | ID: mdl-19887397

ABSTRACT

Growth-restricted fetuses are at risk for a variety of lifelong medical conditions. Preeclampsia, a life-threatening hypertensive disorder of pregnancy, is associated with fetuses who suffer from intrauterine growth restriction (IUGR). Recently, emerging evidence indicates that preeclamptic women harbor AT(1) receptor agonistic autoantibodies (AT(1)-AAs) that contribute to the disease features. However, the exact role of AT(1)-AAs in IUGR and the underlying mechanisms have not been identified. We report that these autoantibodies are present in the cord blood of women with preeclampsia and retain the ability to activate AT(1) receptors. Using an autoantibody-induced animal model of preeclampsia, we show that AT(1)-AAs cross the mouse placenta, enter fetal circulation, and lead to small fetuses with organ growth retardation. AT(1)-AAs also induce apoptosis in the placentas of pregnant mice, human villous explants, and human trophoblast cells. Finally, autoantibody-induced IUGR and placental apoptosis are diminished by either losartan or an autoantibody-neutralizing peptide. Thus, these studies identify AT(1)-AA as a novel causative factor of preeclampsia-associated IUGR and offer two possible underlying mechanisms: a direct detrimental effect on fetal development by crossing the placenta and entering fetal circulation, and indirectly through AT(1)-AA-induced placental damage. Our findings highlight AT(1)-AAs as important therapeutic targets.


Subject(s)
Autoantibodies/immunology , Fetal Growth Retardation/immunology , Maternal-Fetal Exchange/immunology , Pre-Eclampsia/immunology , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/immunology , Adult , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Autoantibodies/blood , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Female , Fetal Growth Retardation/blood , Fetal Growth Retardation/drug therapy , Humans , Losartan/pharmacology , Mice , Peptides/pharmacology , Pre-Eclampsia/blood , Pre-Eclampsia/drug therapy , Pregnancy , Trophoblasts/immunology , Trophoblasts/metabolism
6.
Hypertension ; 51(4): 1010-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18259044

ABSTRACT

Preeclampsia is a pregnancy-specific hypertensive syndrome that causes substantial maternal and fetal morbidity and mortality. Recent evidence indicates that maternal endothelial dysfunction in preeclampsia results from increased soluble Fms-like tyrosine kinase-1 (sFlt-1), a circulating antiangiogenic protein. Factors responsible for excessive production of sFlt-1 in preeclampsia have not been identified. We tested the hypothesis that angiotensin II type 1 (AT(1)) receptor activating autoantibodies, which occur in women with preeclampsia, contribute to increased production of sFlt-1. IgG from women with preeclampsia stimulates the synthesis and secretion of sFlt-1 via AT(1) receptor activation in pregnant mice, human placental villous explants, and human trophoblast cells. Using FK506 or short-interfering RNA targeted to the calcineurin catalytic subunit mRNA, we determined that calcineurin/nuclear factor of activated T-cells signaling functions downstream of the AT(1) receptor to induce sFlt-1 synthesis and secretion by AT(1)-receptor activating autoantibodies. AT(1)-receptor activating autoantibody-induced sFlt-1 secretion resulted in inhibition of endothelial cell migration and capillary tube formation in vitro. Overall, our studies demonstrate that an autoantibody from women with preeclampsia induces sFlt-1 production via angiotensin receptor activation and downstream calcineurin/nuclear factor of activated T-cells signaling. These autoantibodies represent potentially important targets for diagnosis and therapeutic intervention.


Subject(s)
Autoantibodies/pharmacology , Calcineurin/metabolism , Pre-Eclampsia/immunology , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/immunology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Autoantibodies/blood , Cells, Cultured , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/pharmacology , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Solubility , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trophoblasts/cytology , Trophoblasts/immunology
7.
J Immunol ; 179(6): 3391-5, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785770

ABSTRACT

Preeclampsia is a pregnancy-induced hypertensive disorder that causes substantial maternal and fetal morbidity and mortality. Despite being a leading cause of maternal death and a major contributor to maternal and perinatal morbidity, the mechanisms responsible for the pathogenesis of preeclampsia are poorly understood. Recent studies indicate that women with preeclampsia have autoantibodies that activate the angiotensin receptor, AT1, and that autoantibody-mediated receptor activation contributes to pathophysiology associated with preeclampsia. The research reviewed here raises the intriguing possibility that preeclampsia may be a pregnancy-induced autoimmune disease.


Subject(s)
Autoimmune Diseases/metabolism , Autoimmune Diseases/physiopathology , Pre-Eclampsia/immunology , Pre-Eclampsia/metabolism , Receptor, Angiotensin, Type 1/immunology , Animals , Autoantibodies/metabolism , Autoantibodies/physiology , Autoimmune Diseases/immunology , Female , Humans , Pre-Eclampsia/pathology , Pregnancy , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/physiology
8.
Circ Res ; 100(1): 88-95, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17158338

ABSTRACT

Maternal endothelial dysfunction in preeclampsia is associated with increased soluble fms-like tyrosine kinase-1 (sFlt-1), a circulating antagonist of vascular endothelial growth factor and placental growth factor. Angiotensin II (Ang II) is a potent vasoconstrictor that increases concomitant with sFlt-1 during pregnancy. Therefore, we speculated that Ang II may promote the expression of sFlt-1 in pregnancy. Here we report that infusion of Ang II significantly increases circulating levels of sFlt-1 in pregnant mice, thereby demonstrating that Ang II is a regulator of sFlt-1 secretion in vivo. Furthermore, Ang II stimulated sFlt-1 production in a dose- and time-dependent manner from human villous explants and cultured trophoblasts but not from endothelial cells, suggesting that trophoblasts are the primary source of sFlt-1 during pregnancy. As expected, Ang II-induced sFlt-1 secretion resulted in the inhibition of endothelial cell migration and in vitro tube formation. In vitro and in vivo studies with losartan, small interfering RNA specific for calcineurin and FK506 demonstrated that Ang II-mediated sFlt-1 release was via Ang II type 1 receptor activation and calcineurin signaling, respectively. These findings reveal a previously unrecognized regulatory role for Ang II on sFlt-1 expression in murine and human pregnancy and suggest that elevated sFlt-1 levels in preeclampsia may be caused by a dysregulation of the local renin/angiotensin system.


Subject(s)
Angiotensin II/physiology , Calcineurin/metabolism , Pregnancy/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vasoconstrictor Agents/metabolism , Angiotensin II/pharmacology , Animals , Cell Line, Transformed , Cell Movement/physiology , Chorionic Villi/metabolism , Endothelial Cells/physiology , Female , Humans , Mice , Neovascularization, Physiologic/physiology , Placenta/metabolism , Receptor, Angiotensin, Type 1/metabolism , Trophoblasts/metabolism , Vascular Endothelial Growth Factor Receptor-1/blood , Vascular Endothelial Growth Factor Receptor-1/physiology , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...