Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Ultrasound Obstet Gynecol ; 62(2): 248-254, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36971026

RESUMEN

OBJECTIVE: To examine the relationship between umbilical cord insertion site, placental pathology and adverse pregnancy outcome in a cohort of normal and complicated pregnancies. METHODS: Sonographic measurement of the cord insertion and detailed placental pathology were performed in 309 participants. Associations between cord insertion site, placental pathology and adverse pregnancy outcome (pre-eclampsia, preterm birth, small-for-gestational age) were examined. RESULTS: A total of 93 (30%) participants were identified by pathological examination to have a peripheral cord insertion site. Only 41 of the 93 (44%) peripheral cords were detected by prenatal ultrasound. Peripherally inserted cords were associated significantly (P < 0.0001) with diagnostic placental pathology (most commonly with maternal vascular malperfusion (MVM)); of which 85% had an adverse pregnancy outcome. In cases of isolated peripheral cords, without placental pathology, the incidence of adverse outcome was not statistically different when compared to those with central cord insertion and no placental pathology (31% vs 18%; P = 0.3). A peripheral cord with an abnormal umbilical artery (UA) pulsatility index (PI) corresponded to an adverse outcome in 96% of cases compared to 29% when the UA-PI was normal. CONCLUSIONS: This study demonstrates that peripheral cord insertion is often part of the spectrum of findings of MVM disease and is associated with adverse pregnancy outcome. However, adverse outcome was uncommon when there was an isolated peripheral cord insertion and no placental pathology. Therefore, additional sonographic and biochemical features of MVM should be sought when a peripheral cord is observed. © 2023 International Society of Ultrasound in Obstetrics and Gynecology.


Asunto(s)
Placenta , Resultado del Embarazo , Cordón Umbilical , Femenino , Humanos , Recién Nacido , Embarazo , Placenta/patología , Nacimiento Prematuro , Arterias Umbilicales/diagnóstico por imagen , Cordón Umbilical/diagnóstico por imagen , Cordón Umbilical/patología
2.
BJOG ; 125(8): 1009-1017, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29193660

RESUMEN

OBJECTIVE: Preterm birth (PTB) is associated with excess maternal cardiovascular disease risk. We considered that women with PTB and placental evidence of maternal malperfusion would be particularly affected. DESIGN: Pregnancy cohort study. SETTING: Pittsburgh, PA, USA. POPULATION: Women with PTB (n = 115) and term births (n = 210) evaluated 4-12 years after pregnancy. METHODS: Cardiometabolic risk markers were compared in women with prior PTB versus term births; pre-eclampsia and growth restriction cases were excluded. Placental evidence of maternal vascular malperfusion (vasculopathy, infarct, advanced villous maturation, perivillous fibrin, intervillous fibrin deposition), acute infection/inflammation (chorioamnionitis, funisitis, deciduitus) and villitis of unknown aetiology (chronic inflammation) was used to classify PTBs. MAIN OUTCOME MEASURES: Carotid artery intima-media thickness (IMT), fasting lipids, blood pressure (BP) and inflammatory markers measured after delivery. RESULTS: Women with PTB and malperfusion lesions had higher total cholesterol (+13.5 mg/dl) and systolic BP (+4.0 mmHg) at follow up compared with women with term births, accounting for age, race, pre-pregnancy BMI, and smoking (P < 0.05). Women with PTB and malperfusion accompanied by inflammatory lesions had the most atherogenic profile after pregnancy (cholesterol +18.7, apolipoprotein B + 12.7 mg/dl; all P < 0.05), adjusted for pre-pregnancy features. Carotid IMT was higher in this group (+0.037 cm, P = 0.031) accounting for pre-pregnancy factors; differences were attenuated after adjusting for BP and atherogenic lipids at follow up (+0.027, P = 0.095). CONCLUSION: PTBs with placental malperfusion were associated with an excess maternal cardiometabolic risk burden in the decade after pregnancy. The placenta may offer insight into subtypes of PTB related to maternal cardiovascular disease. TWEETABLE ABSTRACT: Preterm births with placental malperfusion may mark women at higher cardiovascular disease risk.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Placenta/irrigación sanguínea , Nacimiento Prematuro/fisiopatología , Daño por Reperfusión/complicaciones , Adulto , Presión Sanguínea , Grosor Intima-Media Carotídeo , Femenino , Humanos , Periodo Posparto , Embarazo , Nacimiento Prematuro/etiología , Estudios Prospectivos , Factores de Riesgo
3.
J Med Virol ; 83(6): 1031-3, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21503916

RESUMEN

Polyomavirus BK (BKV) is a widely latent pathogen in man. Although viral reactivation during pregnancy has been demonstrated, and polyomaviruses have been linked to chromosomal abnormalities, a pathogenic role for BKV in fetoplacental disease has not been explored. We performed in situ hybridization studies with BKV probes on cases of villitis of unknown etiology (102), diffuse villitis (25), and spontaneous abortion (22). We found no evidence that BKV plays a role in the pathogenesis of these common fetoplacental disorders.


Asunto(s)
Aborto Espontáneo/virología , Virus BK/patogenicidad , Enfermedades Placentarias/virología , Infecciones por Polyomavirus/virología , Infecciones Tumorales por Virus/virología , Aborto Espontáneo/patología , Adolescente , Adulto , Virus BK/genética , Vellosidades Coriónicas/patología , ADN Viral/genética , Femenino , Humanos , Hibridación in Situ/métodos , Inflamación/virología , Masculino , Enfermedades Placentarias/patología , Infecciones por Polyomavirus/patología , Embarazo , Complicaciones del Embarazo , Complicaciones Infecciosas del Embarazo/patología , Complicaciones Infecciosas del Embarazo/virología , Estudios Retrospectivos , Infecciones Tumorales por Virus/patología
4.
J Clin Invest ; 105(8): 1057-65, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10772650

RESUMEN

Recent observations suggest that immune response is involved in the development of pancreatitis. However, the exact pathogenesis underlying this immune-mediated response is still under debate. TGF-beta has been known to be an important regulating factor in maintaining immune homeostasis. To determine the role of TGF-beta in the initiation or progression of pancreatitis, TGF-beta signaling was inactivated in mouse pancreata by overexpressing a dominant-negative mutant form of TGF-beta type II receptor in the pancreas, under control of the pS2 mouse trefoil peptide promoter. Transgenic mice showed marked increases in MHC class II molecules and matrix metalloproteinase expression in pancreatic acinar cells. These mice also showed increased susceptibility to cerulein-induced pancreatitis. This pancreatitis was characterized by severe pancreatic edema, inflammatory cell infiltration, T- and B-cell hyperactivation, IgG-type autoantibodies against pancreatic acinar cells, and IgM-type autoantibodies against pancreatic ductal epithelial cells. Therefore, TGF-beta signaling seems to be essential either in maintaining the normal immune homeostasis and suppressing autoimmunity or in preserving the integrity of pancreatic acinar cells.


Asunto(s)
Pancreatitis/etiología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inducido químicamente , Linfocitos B/inmunología , Ceruletida , Citocinas/genética , Femenino , Expresión Génica , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Masculino , Ratones , Ratones Transgénicos , Mutagénesis , Pancreatitis/inmunología , Pancreatitis/metabolismo , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/genética
5.
Can Respir J ; 2017: 9064046, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28316463

RESUMEN

Introduction. Failure of the vascular pulmonary remodeling at birth often manifests as pulmonary hypertension (PHT) and is associated with a variety of neonatal lung disorders including a uniformly fatal developmental disorder known as alveolar capillary dysplasia with misalignment of pulmonary veins (ACD/MPV). Serum serotonin regulation has been linked to pulmonary vascular function and disease, and serotonin transporter (SERT) is thought to be one of the key regulators in these processes. We sought to find evidence of a role that SERT plays in the neonatal respiratory adaptation process and in the pathomechanism of ACD/MPV. Methods. We used histology and immunohistochemistry to determine the timetable of SERT protein expression in normal human fetal and postnatal lungs and in cases of newborn and childhood PHT of varied etiology. In addition, we tested for a SERT gene promoter defect in ACD/MPV patients. Results. We found that SERT protein expression begins at 30 weeks of gestation, increases to term, and stays high postnatally. ACD/MPV patients had diminished SERT expression without SERT promoter alteration. Conclusion. We concluded that SERT/serotonin pathway is crucial in the process of pulmonary vascular remodeling/adaptation at birth and plays a key role in the pathobiology of ACD/MPV.


Asunto(s)
Pulmón/metabolismo , Síndrome de Circulación Fetal Persistente/etiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Adaptación Fisiológica , Femenino , Humanos , Lactante , Recién Nacido , Pulmón/embriología , Masculino , Síndrome de Circulación Fetal Persistente/metabolismo , Regiones Promotoras Genéticas , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
6.
Cancer Res ; 59(19): 4834-42, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10519393

RESUMEN

Transforming growth factor (TGF)-betas are multifunctional growth factors, the properties of which include the potent inhibition of epithelial cell growth. Expression patterns of TGF-betas and TGF-beta receptors in the normal prostate indicate that these growth regulators play key roles in prostatic development and proliferative homeostasis. Importantly, TGF-beta receptor levels are frequently diminished in malignant human prostate tissue. To test the hypothesis that loss of TGF-beta responsiveness is causally involved in the tumorigenic process, we have used retroviral transduction to introduce a dominant-negative mutant type II TGF-beta receptor (DNR) into the premalignant rat prostatic epithelial cell line, NRP-152. High-level expression of the DNR abolished the ability of TGF-beta to inhibit cell growth, to promote cell differentiation, and to induce apoptosis, and it partially blocked the induction of extracellular matrix gene expression. When injected into nude mice, NRP-152-DNR cells formed carcinomas at 13 of 34 sites, compared with 0 of 30 sites for parental and control cells (P = 0.0001). We conclude that the type II TGF-beta receptor is an important tumor suppressor in the prostate, and furthermore, that loss of TGF-beta responsiveness can contribute early in the tumorigenic process by causing the malignant transformation of preneoplastic cells.


Asunto(s)
Transformación Celular Neoplásica , Proteínas de la Matriz Extracelular/genética , Regulación de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata/patología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Factor de Crecimiento Transformador beta/farmacología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Células Epiteliales , Humanos , Masculino , Ratones , Ratones Desnudos , Próstata , Neoplasias de la Próstata/genética , Proteínas Serina-Treonina Quinasas , Ratas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteínas Recombinantes/metabolismo , Transfección , Trasplante Heterólogo
7.
Oncogene ; 19(15): 1868-74, 2000 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-10773876

RESUMEN

The tumor suppressor SMAD4, also known as DPC4, deleted in pancreatic cancer, is a central mediator of TGF-beta signaling. It was previously shown that mice homozygous for a null mutation of Smad4 (Smad4-/-) died prior to gastrulation displaying impaired extraembryonic membrane formation and endoderm differentiation. Here we show that Smad4+/- mice began to develop polyposis in the fundus and antrum when they were over 6 - 12 months old, and in the duodenum and cecum in older animals at a lower frequency. With increasing age, polyps in the antrum show sequential changes from hyperplasia, to dysplasia, in-situ carcinoma, and finally invasion. These alterations are initiated by a dramatic expansion of the gastric epithelium where Smad4 is expressed. However, loss of the remaining Smad4 wild-type allele was detected only in later stages of tumor progression, suggesting that haploinsufficiency of Smad4 is sufficient for tumor initiation. Our data also showed that overexpression of TGF-beta1 and Cyclin D1 was associated with increased proliferation of gastric polyps and tumors. These studies demonstrate that Smad4 functions as a tumor suppressor in the gastrointestinal tract and also provide a valuable model for screening factors that promote or prevent gastric tumorigenesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes Supresores de Tumor , Pólipos/genética , Neoplasias Gástricas/genética , Transactivadores/genética , Factores de Edad , Animales , Ciclina D1/metabolismo , Haploidia , Pérdida de Heterocigocidad , Ratones , Pólipos/patología , Proteína Smad4 , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/metabolismo
8.
Rejuvenation Res ; 8(1): 18-28, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15798371

RESUMEN

Point and deletion mutations and a general depletion of mammalian mitochondrial DNA (mtDNA) give rise to a wide variety of medical syndromes that are refractory to treatment, possibly including aging itself. While gene therapy directed at correcting such deficits in the mitochondrial genome may offer some therapeutic benefits, there are inherent problems associated with a direct approach. These problems are primarily due to the high mitochondrial genome copy number in each cell and the mitochondrial genome being "protected" inside the double-membrane mitochondrial organelle. In an alternative approach there is evidence that genes normally present in the mitochondrial genome can be incorporated into the nuclear genome. To extend such studies, we modified the Chinese Hamster Ovary (CHO) mtDNA-located ATPase6 gene (possessing a mutation which confers oligomycin resistance- oli(r)) by altering the mtDNA code to the universal code (U-code) to permit the correct translation of its mRNA in the cytoplasm. The U-code construct was inserted into the nuclear genome (nucDNA) of a wild type CHO cell. The expressed transgene products enabled the transformed CHO cell lines to grow in up to 1000 ng mL(-1) oligomycin, while untransformed sensitive CHO cells were eliminated in 1 ng mL(-1) oligomycin. This approach, termed allotopic expression, provides a model that may make possible the transfer of all 13 mtDNA mammalian protein-encoding genes to the nucDNA, for treatments of mtDNA disorders. The CHO mtATPase6 protein is 85% identical to both the mouse and human mtATPase6 protein; these proteins are highly conserved in the region of the oligomycin resistance mutation. They are also well conserved in the regions of the oligomycin resistance mutation of the mouse, and in the region of a mutation found in Leigh's syndrome (T8993G), also called NARP (neurogenic weakness, ataxia, retinitis pigmentosum). It is likely that the CHO oli(r) mtATPase6 Ucode construct could impart oligomycin-resistance in human and mouse cells, as well as function in place of the mutant ATPase subunit in a NARP cell line. Preliminary experiments on human cybrids homoplasmic for the NARP mutation (kindly supplied by D.C. Wallace), transformed with our construct, display an increased oligomycin resistance that supports these suppositions.


Asunto(s)
ADN Mitocondrial/genética , Técnicas de Transferencia de Gen , ATPasas de Translocación de Protón Mitocondriales/genética , Animales , Células CHO , Cricetinae , Terapia Genética , Enfermedad de Leigh/genética , Oligomicinas , Plásmidos , Biosíntesis de Proteínas/fisiología , Transgenes
9.
Placenta ; 36(6): 687-92, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25798785

RESUMEN

INTRODUCTION: Normal placental function is critical to optimize fetal growth and development, but few perinatal studies incorporate placental measures. Our objectives were to link clinical placental pathology records to birth records, and validate an automated abstraction strategy. METHODS: Of the 47,329 deliveries at our hospital from 2008 to 2012, we retrieved electronic copies of pathology reports (n = 21,585, 45.4%). Pathology data were extracted with Extensible Markup Language (XML) script using Java and structured query language (SQL) transformed the text information into variables that were linked to delivery data. A subgroup of records was selected for a validation study that compared automated to manual abstraction (n = 144). RESULTS: Linked birth-placental records included 93% of all preterm (<37 weeks, n = 5108) and 37.1% of term births (n = 14,019). Over 90% of deliveries complicated by preeclampsia, chronic hypertension, or gestational diabetes included pathology data. The validation study indicated excellent agreement, sensitivity and specificity between the two abstraction strategies. DISCUSSION: We demonstrate a reliable approach to electronically integrate placental pathology and delivery data. These linked data provide a platform to identify risk factors and sequelae associated with placental lesions.


Asunto(s)
Diabetes Gestacional/patología , Retardo del Crecimiento Fetal/patología , Enfermedades Placentarias/patología , Placenta/patología , Preeclampsia/patología , Bases de Datos Factuales , Femenino , Humanos , Embarazo , Estudios Retrospectivos
10.
Pediatr Pulmonol ; 49(10): 985-90, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24574430

RESUMEN

INTRODUCTION: The mammalian lung possesses the highest level of angiotensin converting enzyme (ACE) amongst all the organs. ACE is known to generate angiotensin (AT)-II from AT-I and to regulate serum bradykinin level, thereby controlling blood pressure. Recent data, however, indicate a role for ACE derived AT-II in angiogenesis, pulmonary hypertension, and neonatal lung disease. The ontogeny of ACE in humans has not been investigated. We studied pulmonary ACE expression during human lung development and in human bronchopulmonary dysplasia (BPD). MATERIAL AND METHODS: Human fetal autopsy lung tissue representing all three trimesters (12, 13, 16, 18, 24, 34, 39, and 40 weeks of gestational age (WGA)), as well as from 1 to 10 years of age with no significant lung pathology were used. In addition lung sections of patients with BPD (n = 5) were selected. The slides were immunostained using an anti-ACE monoclonal antibody. The temporal and spatial pattern of ACE expression was contrasted to that of the pan-endothelial marker CD31. Staining intensity was graded. RESULTS: Mildly diffuse and strong microvascular endothelial immunreactivity for ACE was seen in the human fetus as early as 12 WGA. ACE expression peaked at mid gestation and remained high throughout gestation and postnatally. In BPD lungs ACE endothelial staining was largely absent, and when focal staining was observed the intensity was weak. CONCLUSION: We established that ACE expression is present in the human fetal lung as early as 12 WGA, remains active pre- and postnatally, and ACE expression was downregulated in BPD lungs. We speculate that ACE may be involved in the process of lung development.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Pulmón/enzimología , Peptidil-Dipeptidasa A/metabolismo , Animales , Niño , Preescolar , Regulación hacia Abajo , Endotelio/metabolismo , Femenino , Feto , Humanos , Lactante , Recién Nacido , Pulmón/embriología , Pulmón/patología , Masculino , Coloración y Etiquetado
11.
Placenta ; 34(3): 248-55, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23332415

RESUMEN

Leukocyte-associated immunoglobulin-like receptor 2 (LAIR2) was identified on a global gene expression microarray analysis of surplus chorionic villus sampling (CVS) tissues as down-regulated in the first trimester of preeclampsia pregnancies. LAIR2 is the soluble receptor counterpart to LAIR1, an inhibitory receptor found on multiple immune cell subsets. In situ and immunohistochemical studies have previously shown that placental expression of LAIR2 expression is highly restricted, confined to the more distal portions of extravillous trophoblast (EVT) cell columns. This study examines LAIR2 expression in deeper layers of trophoblasts in the placental implantation site, maternal decidua and maternal spiral arterioles. Immunohistochemical staining detected LAIR2 expression on a subset of EVT within the implantation site. This trophoblast included the invasive EVT infiltrating the maternal decidual vessels and the EVT forming the endovascular trophoblastic plugs. More specifically, LAIR2-positive EVT showed a striking predilection for maternal decidual arterioles and the immediately surrounding decidua. Moreover, the appearance of EVT expressing LAIR2 in these areas was contemporaneous with the process of spiral arteriole remodeling. Based on these findings, we suggest that LAIR2-expressing EVT may play an important role in the remodeling of maternal spiral arterioles.


Asunto(s)
Arteriolas/fisiología , Decidua/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Receptores Inmunológicos/metabolismo , Trofoblastos/metabolismo , Adulto , Arteriolas/citología , Diferenciación Celular , Decidua/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Placenta/citología , Embarazo , Receptores Inmunológicos/genética , Regeneración
12.
Placenta ; 31(10): 880-5, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20692035

RESUMEN

PURPOSE: A global gene expression microarray analysis of surplus chorionic villus sampling (CVS) tissues identified leukocyte-associated immunoglobulin-like receptor 2 (LAIR2) as down-regulated in the first trimester of pregnancies destined for preeclampsia. Neither the localization nor the function of LAIR2 has been examined in the placenta. Localization studies were conducted in placental tissues to determine the precise sites of LAIR2 mRNA production and protein binding. RESULTS: Quantitative real time polymerase chain reaction (qRT-PCR) indicated LAIR2 expression in CVS, but none in breast, lymph node, kidney, skin, uterus, or third trimester placentas. In situ hybridization (ISH) revealed a highly restricted LAIR2 localization. LAIR2 mRNA was found only in the more distal portions of trophoblast anchoring cell columns, adjacent to the invading extravillous trophoblast (EVT). Immunohistochemistry (IHC) detected intracellular LAIR2 staining in these same cells. Extracellular staining of this soluble receptor was found in the acellular material between invasive EVT cells distal to the anchoring cell columns. CONCLUSIONS: ISH and IHC staining for LAIR2 detected specific, highly localized expression at the leading edge of EVT anchoring cell columns in first trimester placentas. This staining likely identifies the site of production for this soluble receptor. Following secretion, the receptor appears to bind extracellular material among the invasive EVT. The precise restriction of this protein only to the sites of EVT invasion strongly suggests that it functions to regulate this invasion. The decreased LAIR2 expression noted in first trimester placentas that ultimately developed preeclampsia further suggests that alterations in LAIR2 may play an etiologic role in preeclampsia.


Asunto(s)
Vellosidades Coriónicas/metabolismo , Preeclampsia/metabolismo , Primer Trimestre del Embarazo/metabolismo , Receptores Inmunológicos/metabolismo , Trofoblastos/metabolismo , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Preeclampsia/etiología , Embarazo , ARN/química , ARN/genética , Receptores Inmunológicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Placenta ; 31(9): 781-4, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20667590

RESUMEN

MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression at the post-transcriptional level. While mostly intracellular, a portion of cellular miRNAs is released to the circulation and their level in the plasma is altered in certain pathological conditions such as cancer, and also during pregnancy. We examined the circulating levels of a set of trophoblastic miRNAs, which we recently found to be regulated by hypoxia, in the plasma of pregnant women with fetal growth restriction (FGR). Pregnancy was associated with increased plasma levels of several placenta-specific miRNAs, compared to non-pregnant controls. Among pregnant women, the overall levels of miRNA species that we analyzed were increased by 1.84-fold (p < or = 0.01) in plasma of women with pregnancies complicated by FGR, but decreased in FGR placentas by 24% (p < or = 0.01) compared to values from uncomplicated pregnancies. Together, our results show that plasma concentration of miRNAs is regulated in pregnancy, and that FGR is associated with increased circulating miRNA levels, highlighting the need to explore plasma miRNAs as potential biomarkers for placental diseases.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Hipoxia/metabolismo , MicroARNs/metabolismo , Placenta/metabolismo , Adulto , Femenino , Humanos , Embarazo
14.
J Biol Chem ; 276(28): 26542-9, 2001 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-11356828

RESUMEN

X-linked inhibitor of apoptosis protein (XIAP) is a potent suppressor of apoptotic cell death, which functions by directly inhibiting caspases, the principal effectors of apoptosis. Here we report that XIAP can also function as a cofactor in the regulation of gene expression by transforming growth factor-beta (TGF-beta). XIAP, but not the related proteins c-IAP1 or c-IAP2, associated with several members of the type I class of the TGF-beta receptor superfamily and potentiated TGF-beta-induced signaling. Although XIAP-mediated activation of c-Jun N-terminal kinase and nuclear factor kappa B was found to require the TGF-beta signaling intermediate Smad4, the ability of XIAP to suppress apoptosis was found to be Smad4-independent. These data implicate a role for XIAP in TGF-beta-mediated signaling that is distinct from its anti-apoptotic functions.


Asunto(s)
Proteínas/fisiología , Factor de Crecimiento Transformador beta/fisiología , Apoptosis/fisiología , Línea Celular , Regulación de la Expresión Génica/fisiología , Humanos , Transducción de Señal , Proteína Inhibidora de la Apoptosis Ligada a X
15.
Wound Repair Regen ; 8(3): 179-91, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10886809

RESUMEN

Expression of endogenous transforming growth factor-beta1 is reduced in many animal models of impaired wound healing, and addition of exogenous transforming growth factor-beta has been shown to improve healing. To test the hypothesis that endogenous transforming growth factor-beta1 is essential for normal wound repair, we have studied wound healing in mice in which the transforming growth factor-beta1 gene has been deleted by homologous recombination. No perceptible differences were observed in wounds made in 3-10-day-old neonatal transforming growth factor-beta1 null mice compared to wild-type littermates. To preclude interference from maternally transferred transforming growth factor-beta1, cutaneous wounds were also made on the backs of 30-day-old transforming growth factor-beta1 null and littermate control mice treated with rapamycin, which extends their lifetime and suppresses the inflammatory response characteristic of the transforming growth factor-beta1 null mice. Again, no impairment in healing was seen in transforming growth factor-beta1 null mice. Instead these wounds showed an overall reduction in the amount of granulation tissue and an increased rate of epithelialization compared to littermate controls. Our data suggest that release of transforming growth factor-beta1 from degranulating platelets or secretion by infiltrating macrophages and fibroblasts is not critical to initiation or progression of tissue repair and that endogenous transforming growth factor-beta1 may actually function to increase inflammation and retard wound closure.


Asunto(s)
Factor de Crecimiento Transformador beta/fisiología , Cicatrización de Heridas/fisiología , Factores de Edad , Animales , Modelos Animales de Enfermedad , Tejido de Granulación/patología , Inmunosupresores/uso terapéutico , Inflamación/fisiopatología , Ratones , Sirolimus/uso terapéutico , Heridas y Lesiones/patología
16.
Genes Dev ; 12(11): 1587-92, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9620846

RESUMEN

SMAD proteins mediate signals from receptor serine-threonine kinases (RSKs) of the TGF-beta superfamily. We demonstrate here that HGF and EGF, which signal through RTKs, can also mediate SMAD-dependent reporter gene activation and induce rapid phosphorylation of endogenous SMAD proteins by kinase(s) downstream of MEK1. HGF induces phosphorylation and nuclear translocation of epitope-tagged Smad2 and a mutation that blocks TGF-beta signaling also blocks HGF signal transduction. Smad2 may thus act as a common positive effector of TGF-beta- and HGF-induced signals and serve to modulate cross talk between RTK and RSK signaling pathways.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal , Transactivadores , Animales , Línea Celular , Factor de Crecimiento Epidérmico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor , Factor de Crecimiento de Hepatocito/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Tirosina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/agonistas , Proteína Smad2 , Activación Transcripcional , Transfección
17.
Nature ; 401(6755): 804-8, 1999 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-10548107

RESUMEN

Specialized epithelia known as M cells overlying the lymphoid follicles of Peyer's patches are important in the mucosal immune system, but also provide a portal of entry for pathogens such as Salmonella typhimurium, Mycobacterium bovis, Shigella flexneri, Yersinia enterocolitica and reoviruses. Penetration of intestinal M cells and epithelial cells by Salmonella typhimurium requires the invasion genes of Salmonella Pathogenicity Island 1 (SPI1). SPI1-deficient S. typhimurium strains gain access to the spleen following oral administration and cause lethal infection in mice without invading M cells or localizing in Peyer's patches, which indicates that Salmonella uses an alternative strategy to disseminate from the gastrointestinal tract. Here we report that Salmonella is transported from the gastrointestinal tract to the bloodstream by CD18-expressing phagocytes, and that CD18-deficient mice are resistant to dissemination of Salmonella to the liver and spleen after oral administration. This CD18-dependent pathway of extraintestinal dissemination may be important for the development of systemic immunity to gastrointestinal pathogens, because oral challenge with SPI1-deficient S. typhimurium elicits a specific systemic IgG humoral immune response, despite an inability to stimulate production of specific mucosal IgA.


Asunto(s)
Antígenos CD18/metabolismo , Intestinos/microbiología , Fagocitos/microbiología , Salmonelosis Animal/microbiología , Salmonella typhimurium/fisiología , 3-Fosfoshikimato 1-Carboxiviniltransferasa , Transferasas Alquil y Aril/genética , Animales , Bacteriemia/microbiología , Proteínas Bacterianas/genética , Transporte Biológico , Antígenos CD18/genética , Fimbrias Bacterianas/genética , Genes Bacterianos , Inmunidad Mucosa , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Fagocitos/metabolismo , Salmonelosis Animal/inmunología , Salmonella typhimurium/genética , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad
18.
J Biol Chem ; 275(3): 2115-22, 2000 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-10636916

RESUMEN

Transforming growth factor-beta (TGF-beta) family members signal through a unique set of intracellular proteins called Smads. Smad4, previously identified as the tumor suppressor DPC4, is functionally distinct among the Smad family, and is required for the assembly and transcriptional activation of diverse, Smad-DNA complexes. We previously identified a 48-amino acid proline-rich regulatory element within the middle linker domain of this molecule, the Smad4 activation domain (SAD), which is essential for mediating these signaling activities. We now characterize the functional activity of the SAD. Mutants lacking the SAD are still able to form complexes with other Smad family members and associated transcription factors, but cannot activate transcription in these complexes. Furthermore, the SAD itself is able to activate transcription in heterologous reporter assays, identifying it as a proline-rich transcriptional activation domain, and indicating that the SAD is both necessary and sufficient to activate Smad-dependent transcriptional responses. We show that transcriptional activation by the SAD is p300-dependent, and demonstrate that this activity is associated with a physical interaction of the SAD with the amino terminus of p300. These data identify a novel function of the middle linker region of Smad4, and define the role of the SAD as an important locus determining the transcriptional activation of the Smad complex.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas Nucleares/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Activación Transcripcional , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular , Citoplasma/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína p300 Asociada a E1A , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Datos de Secuencia Molecular , Pruebas de Precipitina , Prolina/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteína Smad2 , Proteína Smad4 , Transactivadores/genética , Transcripción Genética
19.
Genes Dev ; 15(4): 455-66, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11230153

RESUMEN

Hepatitis B, one of the most common infectious diseases in the world, is closely associated with acute and chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Many clinical investigations have revealed that hepatic fibrosis is an important component of these liver diseases caused by chronic hepatitis B. TGF-beta signaling plays an important role in the pathogenesis of fibrosis in chronic hepatitis and cirrhosis. As these diseases are associated with hepatitis B virus (HBV) infection, we examined the possibility that the HBV-encoded pX oncoprotein regulates TGF-beta signaling. We show that pX enhances transcriptional activity in response to TGF-beta, BMP-2, and activin by stabilizing the complex of Smad4 with components of the basic transcriptional machinery. Additionally, confocal microscopic studies suggest that pX facilitates and potentiates the nuclear translocation of Smads, further enhancing TGF-beta signaling. Our studies suggest a new paradigm for amplification of Smad-mediated signaling by an oncoprotein and suggest that enhanced Smad-mediated signaling may contribute to HBV-associated liver fibrosis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Virus de la Hepatitis B/genética , Cirrosis Hepática/virología , Proteínas Oncogénicas Virales/fisiología , Transducción de Señal/fisiología , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células 3T3 , Animales , Núcleo Celular/metabolismo , Virus de la Hepatitis B/patogenicidad , Ratones , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Unión Proteica , Proteína Smad4 , Activación Transcripcional
20.
J Biol Chem ; 276(22): 19332-9, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11279102

RESUMEN

Sorting nexins (SNX) comprise a family of proteins with homology to several yeast proteins, including Vps5p and Mvp1p, that are required for the sorting of proteins to the yeast vacuole. Human SNX1, -2, and -4 have been proposed to play a role in receptor trafficking and have been shown to bind to several receptor tyrosine kinases, including receptors for epidermal growth factor, platelet-derived growth factor, and insulin as well as the long form of the leptin receptor, a glycoprotein 130-associated receptor. We now describe a novel member of this family, SNX6, which interacts with members of the transforming growth factor-beta family of receptor serine-threonine kinases. These receptors belong to two classes: type II receptors that bind ligand, and type I receptors that are subsequently recruited to transduce the signal. Of the type II receptors, SNX6 was found to interact strongly with ActRIIB and more moderately with wild type and kinase-defective mutants of TbetaRII. Of the type I receptors, SNX6 was found to interact only with inactivated TbetaRI. SNXs 1-4 also interacted with the transforming growth factor-beta receptor family, showing different receptor preferences. Conversely, SNX6 behaved similarly to the other SNX proteins in its interactions with receptor tyrosine kinases. Strong heteromeric interactions were also seen among SNX1, -2, -4, and -6, suggesting the formation in vivo of oligomeric complexes. These findings are the first evidence for the association of the SNX family of molecules with receptor serine-threonine kinases.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Factor de Crecimiento Transformador beta/química , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Línea Celular , Clonación Molecular , Epítopos , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Ligandos , Luciferasas/metabolismo , Datos de Secuencia Molecular , Pruebas de Precipitina , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Nexinas de Clasificación , Distribución Tisular , Transfección , Técnicas del Sistema de Dos Híbridos , Proteínas de Transporte Vesicular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA