Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
BMJ ; 379: e068950, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-36442873
2.
Endocrinol Metab Clin North Am ; 51(4): 817-836, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36244695

RESUMEN

Some women experience bothersome symptoms around the time of menopause that may have a negative impact on their quality of life and prompt them to seek treatments. Menopausal hormone therapy was historically the treatment of choice. However, medical contraindications and personal preference for nonhormonal therapy have prompted the evaluation of a range of nonhormonal pharmacologic and non-pharmacologic therapies. This review provides an update focusing on the latest evidence-based approach for the management of bothersome symptoms of menopause.


Asunto(s)
Sofocos , Calidad de Vida , Terapia de Reemplazo de Estrógeno , Femenino , Terapia de Reemplazo de Hormonas , Sofocos/tratamiento farmacológico , Humanos , Menopausia
3.
Front Endocrinol (Lausanne) ; 13: 1067648, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36589798

RESUMEN

Introduction: Embryo implantation failure leads to infertility. As an important approach to regulate implantation, endometrial epithelial cells produce and secrete factors apically into the uterine cavity in the receptive phase to prepare the initial blastocyst adhesion and implantation. Organoids were recently developed from human endometrial epithelium with similar apical-basal polarity compared to endometrial gland making it an ideal model to study endometrial epithelial secretions. Methods: Endometrial organoids were established using endometrial biopsies from women with primary infertility and normal fertility. Fertile and infertile organoids were treated with hormones to model receptive phase of the endometrial epithelium and intra-organoid fluid (IOF) was collected to compare the apical protein secretion profile and function on trophoblast cell adhesion. Results: Our data show that infertile organoids were dysregulated in their response to estrogen and progesterone treatment. Proteomic analysis of organoid apical secretions identified 150 dysregulated proteins between fertile and infertile groups (>1.5-fold change). Trophoblast progenitor spheroids (blastocyst surrogates) treated with infertile organoid apical secretions significantly compromised their adhesion to organoid epithelial cell monolayers compared to fertile group (P < 0.0001). Discussion: This study revealed that endometrial organoid apical secretions alter trophoblast cell adhesiveness relative to fertility status of women. It paves the way to determine the molecular mechanisms by which endometrial epithelial apical released factors regulate blastocyst initial attachment and implantation.


Asunto(s)
Infertilidad Femenina , Trofoblastos , Humanos , Femenino , Trofoblastos/metabolismo , Proteómica , Endometrio/metabolismo , Útero/metabolismo , Infertilidad Femenina/metabolismo , Proteínas/metabolismo
4.
Pregnancy Hypertens ; 14: 125-130, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30527099

RESUMEN

Preeclampsia is associated with intermittent placental hypoxia, inflammation and the release of antiangiogenic factors, namely sFLT-1 and sEng. These factors cause maternal endothelial dysfunction and the manifestation of clinical disease. Disulfiram is a dehydrogenase inhibitor used to treat alcoholism and has been suggested as a proteasome inhibitor. Inhibiting the proteasome has been previously shown to reduce FLT-1 gene expression. Thus, we aim to investigate whether disulfiram alters the secretion of sFLT-1 and sEng and reduces endothelial dysfunction. METHODS AND RESULTS: We assessed the effects of disulfiram on primary cytotrophoblast and human umbilical vein endothelial cells (HUVECs). Disulfiram significantly reduced mRNA expression of membrane bound FLT-1 and sFLT-1 variants in primary cytotrophoblasts, which translated into a significant reduction in the protein secretion of sFLT-1. Additionally, sFLT-1 was reduced in primary HUVECs treated with disulfiram, whilst sEng was only reduced in primary cytotrophoblasts. Next, we investigated the effect of disulfiram on endothelial dysfunction using primary HUVECs treated with 5% preeclamptic serum ±â€¯disulfiram. Serum from preeclamptic women induced endothelial dysfunction evidenced by increased mRNA expression of vascular cell adhesion molecule-1 (VCAM-1) and adhesion of peripheral blood mononuclear cells (PBMCs) to HUVECs. The addition of disulfiram reduced VCAM-1 mRNA expression, however did not affect the adhesion of PBMCs to endothelial cells. Lastly, we assessed the effects of disulfiram on the 20S subunit of the proteasome and found disulfiram did not inhibit this subunit in either primary cytotrophoblast or HUVECs. CONCLUSIONS: Disulfiram quenches sFLT-1 and sEng via mechanisms independent of the 20S subunit of the proteasome. Understanding of the mechanisms by which disulfiram inhibits antiangiogenic secretion may reveal insights into the pathogenesis and potential therapeutic targets for preeclampsia.


Asunto(s)
Inhibidores del Acetaldehído Deshidrogenasa/farmacología , Disulfiram/farmacología , Endoglina/efectos de los fármacos , Placenta/efectos de los fármacos , Preeclampsia/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Endoglina/genética , Endoglina/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Placenta/metabolismo , Embarazo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
J Clin Neurosci ; 48: 114-117, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29239824

RESUMEN

Köhlmeier-Degos disease is rare idiopathic vasculopathy, the exact pathogenesis of which remains unclear. Here, we review pertinent literatutre and present a case of a Köhlmeier-Degos disease with central nervous system involvement followed-up over 11 years with various neuroimaging modalities. Evolution of neurovascular and neuropathological changes over an extended time period has not been previously described.


Asunto(s)
Circulación Cerebrovascular , Trastornos Cerebrovasculares/fisiopatología , Papulosis Atrófica Maligna/fisiopatología , Adulto , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Trastornos Cerebrovasculares/diagnóstico por imagen , Trastornos Cerebrovasculares/etiología , Progresión de la Enfermedad , Femenino , Humanos , Hidrocefalia/diagnóstico por imagen , Hidrocefalia/etiología , Angiografía por Resonancia Magnética , Imagen por Resonancia Magnética , Papulosis Atrófica Maligna/complicaciones , Papulosis Atrófica Maligna/diagnóstico por imagen , Neuroimagen , Siderosis/diagnóstico por imagen , Siderosis/etiología
6.
Plast Reconstr Surg Glob Open ; 5(4): e1317, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28507874

RESUMEN

The free vascularized fibula flap has been widely used for clavicle reconstruction. Limited evidence exists for the long-term outcome of clavicle reconstruction using the free vascularized fibula flap in adults. We report the functional and aesthetic outcome in a 52-year old man a decade after clavicle reconstruction using a free vascularized fibula flap in combination with a modified Richardson Hook Plate. At the 10-year follow-up, panoramic shoulder X-ray showed the modified Richardson Hook Plate had remained firmly in place with the fibula and the hook positioned beneath the acromion. Functionally, the patient presented with a constant shoulder score of 77, with a pain-free symmetrical full range of motion. In conclusion, reconstruction of lateral clavicle defect using free vascularized fibula flap in conjunction with modified Richardson Hook plate may provide patients with excellent long-term functional and aesthetic outcomes.

7.
Placenta ; 48: 110-118, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27871461

RESUMEN

INTRODUCTION: Preeclampsia is a serious complication affecting 5-8% of pregnancies. Central to its pathogenesis is placental hypoxia and inflammation which leads to secretion of soluble fms-like tyrosine kinase 1 (sFlt-1). sFlt-1 causes widespread endothelial dysfunction. The molecular mechanisms regulating sFlt-1 production remain poorly understood. Recently, a binding site for the nuclear factor activated T cells (NFAT) transcription factor has been found on fms-like tyrosine kinase 1 (FLT-1) promoter. METHODS: We assessed whether inhibiting NFAT impacts FLT-1, sFlt-1 and cytokine expression, as well as sFlt-1 secretion in primary cytotrophoblasts, placental explants and human umbilical vein endothelial cells (HUVECs). We investigated whether NFAT is regulated by hypoxia in primary cytotrophoblasts. We characterised the expression of NFAT1-4 in preterm preeclamptic compared to gestationally matched placentas. RESULTS: Inhibiting NFAT reduced FLT-1 and sFlt-1 splice variant e15a transcription, concordant with reduced total sFlt-1 and sFlt-1 e15a secretion from primary human cytotrophoblasts. This effect appeared tissue specific as inhibiting NFAT did not change sFlt-1 secretion from endothelial cells. Inhibiting NFAT also reduced transcription of inflammatory cytokines IL-1ß and IL-10 in primary cytotrophoblasts. NFAT1 and NFAT3 mRNA expression were significantly increased under hypoxia (1% O2). Inhibiting NFAT under hypoxia significantly reduced FLT-1 and sFlt-1 e15a transcription, but did not reduce sFlt-1 secretion. NFAT mRNA and protein localisation was not different in preeclamptic compared to gestationally matched placenta. DISCUSSION: NFAT positively regulates placental FLT-1 and sFlt-1 e15a, secretion of sFlt-1 and inflammatory cytokine expression. It may be involved in the pathophysiology of preeclampsia.


Asunto(s)
Hipoxia de la Célula/fisiología , Factores de Transcripción NFATC/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , Placenta/efectos de los fármacos , Embarazo , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
8.
Placenta ; 48: 72-79, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27871476

RESUMEN

INTRODUCTION: Preeclampsia is a serious complication of pregnancy affecting 5% of pregnancies. Our team identified 137 genes highly expressed in placenta relative to other human tissues. Here, we have explored a role for steroid sulfatase (STS) in preeclampsia by characterising STS expression and the functional effects of STS on primary placental trophoblasts. METHODS: Characterisation of STS was performed on preterm preeclamptic and gestation-matched normotensive preterm controls who delivered at <34 weeks gestation. We characterised placental and maternal whole blood STS mRNA and placental protein expression via qRT-PCR, immunohistochemistry and Western Blot. To assess whether STS is involved in sFlt1 secretion and syncytialisation, we administered siRNA to silence STS in primary trophoblasts before measuring sFlt1 and hCG secretion and E-Cadherin expression. RESULTS: A custom array containing 45 placental specific genes identified 10 genes significantly altered in the placentas of preeclamptic patients relative to normotensive gestation-matched controls. Of these genes, qRT-PCR and western blot on a larger cohort confirmed that the expression of STS was significantly elevated in preeclamptic placentas (n = 44) relative to gestation matched controls (n = 26). Given placental RNA leaks in to the maternal circulation, we also assessed STS mRNA expression in the whole blood of patients with preeclampsia and found it was significantly increased relative to normotensive controls. siRNA knockdown of STS in primary trophoblast resulted in a modest but significant reduction in sFlt1 secretion, but had no affect on hCG secretion or E-Cadherin protein expression. DISCUSSION: STS is increased in preeclamptic placentas and maternal whole blood. Our data suggests that STS may affect sFlt1 secretion by regulating sFlt1-i13 transcription, and not via alterations in syncytialisation.


Asunto(s)
Placenta/enzimología , Preeclampsia/metabolismo , Esteril-Sulfatasa/metabolismo , Trofoblastos/enzimología , Adolescente , Adulto , Cadherinas/metabolismo , Femenino , Humanos , Preeclampsia/sangre , Embarazo , Esteril-Sulfatasa/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
9.
Biol Reprod ; 94(3): 59, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26819475

RESUMEN

The anti-angiogenic protein, soluble fms-like tyrosine kinase-1 (sFLT-1), plays a central role in preeclamptic pathophysiology. A splice variant of FLT-1 (VEGF receptor 1), sFLT-1 is released in excessive amounts from the preeclamptic placenta into the maternal circulation, where it causes endothelial dysfunction manifesting as end-organ disease. However, the mechanisms regulating its production within the placenta remain poorly understood. Recently it was shown in endothelial cells that Jumonji domain containing protein 6 (JMJD6) hydroxylates U2 small nuclear ribonucleoprotein auxiliary factor 65-kDa subunit (U2AF65, a component of the splicesome). The hydroxylation by JMJD6 is oxygen dependent. Under hypoxia, JMJD6 is less able to hydroxylate U2AF65, and this unhydroxylated form of U2AF65 biases splicing of FLT-1 to sFLT-1. We assessed whether oxygen-sensing JMJD6 is differentially expressed in preeclamptic placenta and regulates sFLT-1 splicing in placenta via U2AF65. JMJD6 protein expression was significantly reduced in preterm preeclamptic placenta (P < 0.0001; n = 21) relative to preterm controls (n = 10). Exposing both placental and endothelial cells to hypoxia significantly reduced JMJD6 mRNA and increased sFLT-1 mRNA and protein expression. Silencing JMJD6 in primary endothelial and trophoblast cells significantly increased sFLT-1 secretion. Next, we examined whether these molecules may be directly interacting. We demonstrated that placental U2AF65 colocalized with JMJD6. In turn, we found JMJD6 directly interacts with U2AF65, which in turn produces sFLT-1 mRNA transcripts. Taken together, our findings provide evidence that JMJD6 may play a role in regulating the production of sFLT-1 in the preeclamptic placenta. Decreased placental JMJD6 expression may be an important component to the pathophysiology of preeclampsia.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Isoformas de Proteínas/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Regulación hacia Abajo , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Oxígeno/metabolismo , Embarazo , Isoformas de Proteínas/genética , Factor de Empalme U2AF/genética , Factor de Empalme U2AF/metabolismo , Regulación hacia Arriba , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
10.
Hypertension ; 66(6): 1251-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416849

RESUMEN

In preeclampsia, the antiangiogenic factor soluble fms-like tyrosine kinase-1 (sFLT-1) is released from placenta into the maternal circulation, causing endothelial dysfunction and organ injury. A recently described splice variant, sFLT-1 e15a, is primate specific and the most abundant placentally derived sFLT-1. Therefore, it may be the major sFLT-1 isoform contributing to the pathophysiology of preeclampsia. sFLT-1 e15a protein remains poorly characterized: its bioactivity has not been comprehensively examined, and serum levels in normal and preeclamptic pregnancy have not been reported. We generated and validated an sFLT-1 e15a-specific ELISA to further characterize serum levels during pregnancy, and in the presence of preeclampsia. Furthermore, we performed assays to examine the bioactivity and antiangiogenic properties of sFLT-1 e15a protein. sFLT-1 e15a was expressed in the syncytiotrophoblast, and serum levels rose across pregnancy. Strikingly, serum levels were increased 10-fold in preterm preeclampsia compared with normotensive controls. We confirmed sFLT-1 e15a is bioactive and is able to inhibit vascular endothelial growth factor signaling of vascular endothelial growth factor receptor 2 and block downstream Akt phosphorylation. Furthermore, sFLT-1 e15a has antiangiogenic properties. sFLT-1 e15a decreased endothelial cell migration, invasion, and inhibited endothelial cell tube formation. Administering sFLT-1 e15a blocked vascular endothelial growth factor induced sprouts from mouse aortic rings ex vivo. We have demonstrated that sFLT-1 e15a is increased in preeclampsia, antagonizes vascular endothelial growth factor signaling, and has antiangiogenic activity. Future development of diagnostics and therapeutics for preeclampsia should consider targeting placentally derived sFLT-1 e15a.


Asunto(s)
Placenta/metabolismo , Preeclampsia/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Western Blotting , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Recién Nacido , Ratones Endogámicos C57BL , Microscopía Fluorescente , Preeclampsia/sangre , Preeclampsia/genética , Embarazo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Trofoblastos/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/farmacología
11.
Reprod Fertil Dev ; 26(2): 351-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23484525

RESUMEN

Severe early onset pre-eclampsia is a serious pregnancy complication, believed to arise as a result of persistent placental hypoxia due to impaired placentation. Pregnancy-associated plasma protein A2 (PAPPA2) is very highly expressed in the placenta relative to all other tissues. There is some evidence that PAPPA2 mRNA and protein are increased in association with pre-eclampsia. The aim of the present study was to characterise the mRNA and protein expression, as well as localisation, of PAPPA2 in an independent cohort of severe early onset pre-eclamptic placentas. We also examined whether exposing placental explants to hypoxia (1% oxygen) changed the expression of PAPPA2. Expression of PAPPA2 mRNA and protein was upregulated in severe early onset pre-eclamptic placentas compared with preterm controls and localised to the syncytiotrophoblast. Interestingly, protein localisation was markedly reduced in term placenta. Syncytialisation of BeWo cells did not change PAPPA2 expression. However, hypoxia upregulated PAPPA2 mRNA and protein expression in primary placental explants. Together, our data suggest that PAPPA2 may be upregulated in severe pre-eclampsia and, functionally, this may be mediated via increased placental hypoxia known to occur with this pregnancy disorder.


Asunto(s)
Placenta/metabolismo , Preeclampsia/metabolismo , Proteína Plasmática A Asociada al Embarazo/metabolismo , Estudios de Casos y Controles , Hipoxia de la Célula , Línea Celular , Femenino , Edad Gestacional , Humanos , Preeclampsia/diagnóstico , Preeclampsia/genética , Embarazo , Proteína Plasmática A Asociada al Embarazo/genética , Índice de Severidad de la Enfermedad , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Trofoblastos/metabolismo , Regulación hacia Arriba , Victoria
12.
Hum Reprod ; 28(5): 1172-80, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23434834

RESUMEN

STUDY QUESTION: What is the nature of cellular Corin expression in human gestational tissues? SUMMARY ANSWER: CORIN is expressed in non-pregnant late secretory phase endometrium, first trimester human implantation sites and is up-regulated with decidualization ex vivo. WHAT IS KNOWN ALREADY: Adequate trophoblast invasion and spiral artery remodeling/transformation is critical for successful implantation. CORIN, best known for its role in activating atrial natruietic peptide (ANP) to regulate blood pressure, has recently been proposed to be centrally involved in trophoblast invasion and spiral artery remodeling. It is postulated that ANP, activated by CORIN, promotes trophoblast invasion and that a deficiency causes pre-eclampsia. Mice deficient in either Corin or ANP displayed poor trophoblast invasion, impaired spiral artery remodeling and phenocopied human pre-eclampsia. However, the precise cellular localization of CORIN within human gestational tissues has not been well characterized. STUDY DESIGN, SIZE, DURATION: We measured CORIN protein localization in a number of human gestational tissues relevant to early embryo/placental implantation: non-pregnant (NP) endometrial biopsies (n = 5 per phase of the menstrual cycle), first trimester placental bed biopsies (n = 12) and pre-term control (n = 10) and severe early onset preeclamptic placentas (n = 15). Endometrial stromal cells were isolated from human endometrial biopsies (n = 5) and induced to decidualize ex vivo. Finally, CORIN concentrations were measured in serum obtained from pregnant women during the first trimester of whom, 56 subsequently ended up with a healthy term delivery (controls), 18 developed fetal growth restriction (FGR) and 21 had a miscarriage. PARTICIPANTS/MATERIALS, SETTING, METHODS: We performed immunohistochemistry to assess CORIN localization. Changes in Corin mRNA expression in human endometrial stromal cells decidualized ex vivo were measured by quantitative RT-PCR, and levels of CORIN within human sera were measured by ELISA. MAIN RESULTS AND THE ROLE OF CHANCE: CORIN was expressed in both NP late secretory phase endometrium and first trimester decidua within placental bed biopsies. Importantly, decidualization of primary human endometrial cells ex vivo significantly increased Corin expression (P < 0.05). CORIN was also detected within the villous cytotrophoblast, but there was no change in mRNA levels in placentas complicated by severe preterm pre-eclampsia when compared with pre-term controls. Although CORIN was detected in first trimester serum, levels did not change across gestation, nor could they predict miscarriage or FGR (other disorders of impaired placental invasion). LIMITATIONS, REASONS FOR CAUTION: Owing to the fact that we utilized early pregnancy human specimens, this is mainly a descriptive study with a limited amount of functional experiments. WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to thoroughly characterize Corin mRNA and protein expression in human gestational tissue. Our findings support recent data from murine studies collectively suggesting that CORIN plays a critical role in trophoblast migration and spiral artery remodeling during early pregnancy in humans. Therefore, further studies of CORIN biology in early pregnancy may identify new therapeutic targets to improve implantation quality in early pregnancy and potentially reduce the rates of pregnancy complications caused by inadequate implantation (pre-eclampsia, FGR and miscarriage). STUDY FUNDING/COMPETING INTEREST(S): This study was supported by The National Health and Medical Research Council of Australia (Salary support #490970, #490995). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. The authors declare that no competing interests exist.


Asunto(s)
Decidua/metabolismo , Endometrio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Serina Endopeptidasas/metabolismo , Aborto Espontáneo/genética , Adulto , Arterias/patología , Biopsia , Decidua/patología , Endometrio/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Retardo del Crecimiento Fetal/genética , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Placenta/metabolismo , Placenta/patología , Preeclampsia/metabolismo , Embarazo , Resultado del Embarazo , Primer Trimestre del Embarazo , Trofoblastos/citología , Adulto Joven
13.
Endocrinology ; 154(2): 911-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23288908

RESUMEN

Abnormal trophoblast growth can cause life-threatening disorders such as ectopic pregnancy, choriocarcinoma, and placenta accreta. EnGeneIC Delivery Vehicles (EDVs) are nanocells that can promote tissue-specific delivery of drugs and may be useful to medically treat such disorders. The objective of this study was to determine whether EDVs loaded with the chemotherapeutic doxorubicin and targeting the epidermal growth factor receptor (EGFR, very highly expressed on the placental surface) can regress placental cells in vitro, ex vivo, and in vivo. In female SCID mice, EGFR-targeted EDVs induced greater inhibition of JEG-3 (choriocarcinoma cells) tumor xenografts, compared with EDVs targeting an irrelevant antigen (nontargeted EDVs) or naked doxorubicin. EGFR-targeted EDVs were more readily taken up by human placental explants ex vivo and induced increased apoptosis (M30 antibody) compared with nontargeted EDVs. In vitro, EGFR-targeted EDVs administered to JEG-3 cells resulted in a dose-dependent inhibition of cell viability, proliferation, and increased apoptosis, a finding confirmed by continuous monitoring by xCELLigence. In conclusion, EGFR-targeted EDVs loaded with doxorubicin significantly inhibited trophoblastic tumor cell growth in vivo and in vitro and induced significant cell death ex vivo, potentially mediated by increasing apoptosis and decreasing proliferation. EDVs may be a novel nanoparticle treatment for ectopic pregnancy and other disorders of trophoblast growth.


Asunto(s)
Doxorrubicina/administración & dosificación , Receptores ErbB/metabolismo , Nanopartículas/administración & dosificación , Embarazo Ectópico/tratamiento farmacológico , Neoplasias Trofoblásticas/tratamiento farmacológico , Animales , Femenino , Ratones , Ratones SCID , Embarazo , Células Tumorales Cultivadas , Neoplasias Uterinas/tratamiento farmacológico
14.
J Clin Endocrinol Metab ; 98(3): E429-36, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23337725

RESUMEN

CONTEXT: Fetal growth restriction (FGR) is a leading cause of perinatal mortality, yet no reliable screening test exists. Placental specific mRNA in the maternal circulation may reflect changes in the placental transcriptome in FGR and could be a novel biomarker for FGR. OBJECTIVE: The aim of the study was to identify placental specific RNA detectable in the maternal circulation and examine whether they are differentially expressed in severe preterm FGR. DESIGN: In silico screening was used to identify placental specific RNAs. Their expression in cases of severe FGR vs controls was examined in both maternal blood and placenta by microarray, RT-PCR, and in situ hybridization. RESULTS: Via in silico analysis, we identified 137 genes very highly expressed in the placenta relative to other tissues. Using microarray, we found that they were detectable in the maternal blood and were globally dysregulated with preterm FGR; 75 genes (55%) had a ≥1.5-fold differential expression compared to controls. Eight genes (ERVWE-1, PSG1, PLAC4, TAC3, PLAC3, CRH, CSH1, and KISS1) were validated by RT-PCR to be significantly increased in both maternal blood and placenta in a larger cohort of severe FGR compared to controls. In situ hybridization confirmed PAPPA2 and ERVWE-1 localized to the syncytiotrophoblast. CONCLUSION: There is global differential expression of placental specific mRNA in the maternal blood in pregnancies complicated by severe preterm FGR. Placental specific mRNA in maternal blood may represent a new class of biomarkers for preterm FGR.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Placenta/fisiología , Proteínas Gestacionales/genética , ARN Mensajero/genética , Transcriptoma , Adulto , Hormona Liberadora de Corticotropina/genética , Femenino , Retardo del Crecimiento Fetal/metabolismo , Productos del Gen env/genética , Humanos , Recién Nacido , Recien Nacido Prematuro/sangre , Kisspeptinas/genética , Lactógeno Placentario/genética , Embarazo , Proteína Plasmática A Asociada al Embarazo/genética , Glicoproteínas beta 1 Específicas del Embarazo/genética , ARN Mensajero/sangre , Receptores de Taquicininas/genética
15.
Rev Endocr Metab Disord ; 13(4): 235-51, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22847235

RESUMEN

The functional layer of the human endometrium is a highly regenerative tissue undergoing monthly cycles of growth, differentiation and shedding during a woman's reproductive years. Fluctuating levels of circulating estrogen and progesterone orchestrate this dramatic remodeling of human endometrium. The thin inactive endometrium of postmenopausal women which resembles the permanent basal layer of cycling endometrium retains the capacity to respond to exogenous sex steroid hormones to regenerate into a thick functional endometrium capable of supporting pregnancy. Endometrial regeneration also follows parturition and endometrial resection. In non menstruating rodents, endometrial epithelium undergoes rounds of proliferation and apoptosis during estrus cycles. The recent identification of adult stem cells in both human and mouse endometrium suggests that epithelial progenitor cells and the mesenchymal stem/stromal cells have key roles in the cyclical regeneration of endometrial epithelium and stroma. This review will summarize the evidence for endometrial stem/progenitor cells, examine their role in mouse models of endometrial epithelial repair and estrogen-induced endometrial regeneration, and also describe the generation of endometrial-like epithelium from human embryonic stem cells. With markers now available for identifying endometrial mesenchymal stem/stromal cells, their possible role in gynecological diseases associated with abnormal endometrial proliferation and their potential application in cell-based therapies to regenerate reproductive and other tissues will be discussed.


Asunto(s)
Endometrio/fisiología , Regeneración , Células Madre/citología , Animales , Femenino , Humanos , Trasplante de Células Madre , Enfermedades Uterinas/terapia
16.
PLoS One ; 7(6): e39864, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768148

RESUMEN

Preeclampsia is a major pregnancy complication, characterized by severe endothelial dysfunction, hypertension and maternal end-organ damage. Soluble endoglin is an anti-angiogenic protein released from placenta and thought to play a central role in causing the endothelial dysfunction and maternal organ injury seen in severe preeclampsia. We recently reported MMP-14 was the protease producing placentally-derived soluble endoglin by cleaving full-length endoglin present on the syncytiotrophoblast surface. This find identifies a specific drug target for severe preeclampsia; interfering with MMP-14 mediated cleavage of endoglin could decrease soluble endoglin production, ameliorating clinical disease. However, experimental MMP-14 inhibition alone only partially repressed soluble endoglin production, implying other proteases might have a role in producing soluble endoglin. Here we investigated whether MMP-15--phylogenetically the closest MMP relative to MMP-14 with 66% sequence similarity--also cleaves endoglin to produce soluble endoglin. MMP-15 was localized to the syncytiotrophoblast layer of the placenta, the same site where endoglin was localized. Interestingly, it was significantly (p = 0.03) up-regulated in placentas from severe early-onset preeclamptic pregnancies (n = 8) compared to gestationally matched preterm controls (n = 8). However, siRNA knockdown of MMP-15 yielded no significant decrease of soluble endoglin production from either HUVECs or syncytialised BeWo cells in vitro. Importantly, concurrent siRNA knockdown of both MMP-14 and MMP-15 in HUVECS did not yield further decrease in soluble endoglin production compared to MMP-14 siRNA alone. We conclude MMP-15 is up-regulated in preeclampsia, but does not cleave endoglin to produce soluble endoglin.


Asunto(s)
Antígenos CD/metabolismo , Metaloproteinasa 15 de la Matriz/metabolismo , Receptores de Superficie Celular/metabolismo , Regulación hacia Arriba , Adulto , Estudios de Cohortes , Endoglina , Femenino , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Metaloproteinasa 15 de la Matriz/deficiencia , Metaloproteinasa 15 de la Matriz/genética , Preeclampsia/enzimología , Preeclampsia/genética , Preeclampsia/patología , Embarazo , Transporte de Proteínas , Solubilidad , Trofoblastos/enzimología , Trofoblastos/patología
17.
Fertil Steril ; 98(1): 11-20, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22657248

RESUMEN

Adult stem cells have been identified in the highly regenerative human endometrium on the basis of their functional attributes. They can reconstruct endometrial tissue in vivo suggesting their possible use in treating disorders associated with inadequate endometrium. The identification of specific markers for endometrial mesenchymal stem cells and candidate markers for epithelial progenitor cells enables the potential use of endometrial stem/progenitor cells in reconstructing endometrial tissue in Asherman syndrome and intrauterine adhesions.


Asunto(s)
Endometrio/fisiología , Regeneración/fisiología , Trasplante de Células Madre , Células Madre/fisiología , Adulto , Células Madre Adultas/fisiología , Endometrio/citología , Femenino , Humanos , Modelos Biológicos , Trasplante de Células Madre/métodos , Células Madre/citología , Cicatrización de Heridas/fisiología
18.
Biol Reprod ; 86(6): 184, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22402967

RESUMEN

The human endometrium is incredibly dynamic, undergoing monthly cycles of growth and regression during a woman's reproductive life. Endometrial repair at the cessation of menstruation is critical for reestablishment of a functional endometrium receptive for embryo implantation; however, little is understood about the mechanisms behind this rapid and highly efficient process. This study utilized a functional mouse model of endometrial breakdown and repair to assess changes in endometrial vasculature that accompany these dynamic processes. Given that adult endometrial stem/progenitor cells identified in human and mouse endometrium are likely contributors to the remarkable regenerative capacity of endometrium, we also assessed label-retaining cells (LRC) as candidate stromal stem/progenitor cells and examined their relationship with endometrial vasculature. Newborn mouse pups were pulse-labeled with bromodeoxyuridine (BrdU) and chased for 5 wk before decidualization, endometrial breakdown, and repair were induced by hormonal manipulation. Mean vessel density did not change significantly throughout breakdown and repair; however, significantly elevated endothelial cell proliferation was observed in decidual tissue. Stromal LRC were identified throughout breakdown and repair, with significantly fewer observed during endometrial repair than before decidualization. A significantly higher percentage of LRC were associated with vasculature during repair than before decidualization, and a proportion were undergoing proliferation, indicative of their functional capacity. This study is the first to examine the endometrial vasculature and candidate stromal stem/progenitor cells in a functional mouse model of endometrial breakdown and repair and provides functional evidence suggesting that perivascular LRC may contribute to endometrial stromal expansion during the extensive remodeling associated with this process.


Asunto(s)
Endometrio/fisiología , Células Madre/citología , Actinas/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Endometrio/irrigación sanguínea , Endometrio/citología , Células Endoteliales/fisiología , Estrógenos , Ciclo Estral , Femenino , Ratones , Ratones Endogámicos C57BL , Microvasos/anatomía & histología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Progesterona , Células Madre/metabolismo
19.
Histochem Cell Biol ; 137(4): 527-36, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22231913

RESUMEN

Lim1 encodes a homeodomain transcription factor required for head, kidney and female reproductive tract development in the murine embryo. Recently, Lim1 expression was documented in several adult murine and human organs. In the developing female reproductive tract, Lim1 expression was first detected in the Müllerian ducts. Using immunofluorescence, we detected LIM1 expression in a developmental model of human female reproductive tract which was established by recombination of neonatal uterine mesenchyme with human embryonic stem cells. In addition, we report a dynamic expression of Lim1/LIM1 in neonatal and adult mouse, and adult human endometrial epithelium and stroma as revealed by immunofluorescence and quantitative real-time polymerase chain reaction. LIM1 expression was also observed in several endometrial epithelial cancer cell lines (ECC-1, Ishikawa, and HEC1A). Furthermore, we found that Activin A significantly upregulated LIM1 mRNA expression in ECC-1 cells. These studies demonstrate previously unreported Lim1/LIM1 expression in neonatal, adult mouse and human endometrium suggesting Lim1/LIM1 may have a role in endometrial development and remodelling.


Asunto(s)
Endometrio/embriología , Endometrio/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/metabolismo , Adulto , Animales , Endometrio/citología , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Técnicas de Cultivo de Tejidos , Factores de Transcripción/genética
20.
PLoS One ; 6(6): e21136, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21698266

RESUMEN

BACKGROUND: Recent studies have identified stem/progenitor cells in human and mouse uterine epithelium, which are postulated to be responsible for tissue regeneration and proliferative disorders of human endometrium. These progenitor cells are thought to be derived from Müllerian duct (MD), the primordial female reproductive tract (FRT). METHODOLOGY/PRINCIPAL FINDINGS: We have developed a model of human reproductive tract development in which inductive neonatal mouse uterine mesenchyme (nMUM) is recombined with green fluorescent protein (GFP)-tagged human embryonic stem cells (hESCs); GFP-hESC (ENVY). We demonstrate for the first time that hESCs can be differentiated into cells with a human FRT epithelial cell phenotype. hESC derived FRT epithelial cells emerged from cultures containing MIXL1(+) mesendodermal precursors, paralleling events occurring during normal organogenesis. Following transplantation, nMUM treated embryoid bodies (EBs) generated epithelial structures with a typical MD phenotype that expressed the MD markers PAX2, HOXA10. Functionally, the hESCs derived FRT epithelium responded to exogenous estrogen by proliferating and secreting uterine-specific glycodelin A (GdA). CONCLUSIONS/SIGNIFICANCE: These data show nMUM can induce differentiation of hESC to form the FRT epithelium. This may provide a model to study early developmental events of the human FRT.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Epiteliales/citología , Genitales Femeninos/crecimiento & desarrollo , Animales , Femenino , Genitales Femeninos/citología , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...