Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Hum Genet ; 107(5): 911-931, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33098770

RESUMEN

Human fetuses with trisomy 21 (T21) have atypical brain development that is apparent sonographically in the second trimester. We hypothesize that by analyzing and integrating dysregulated gene expression and pathways common to humans with Down syndrome (DS) and mouse models we can discover novel targets for prenatal therapy. Here, we tested the safety and efficacy of apigenin, identified with this approach, in both human amniocytes from fetuses with T21 and in the Ts1Cje mouse model. In vitro, T21 cells cultured with apigenin had significantly reduced oxidative stress and improved antioxidant defense response. In vivo, apigenin treatment mixed with chow was administered prenatally to the dams and fed to the pups over their lifetimes. There was no significant increase in birth defects or pup deaths resulting from prenatal apigenin treatment. Apigenin significantly improved several developmental milestones and spatial olfactory memory in Ts1Cje neonates. In addition, we noted sex-specific effects on exploratory behavior and long-term hippocampal memory in adult mice, and males showed significantly more improvement than females. We demonstrated that the therapeutic effects of apigenin are pleiotropic, resulting in decreased oxidative stress, activation of pro-proliferative and pro-neurogenic genes (KI67, Nestin, Sox2, and PAX6), reduction of the pro-inflammatory cytokines INFG, IL1A, and IL12P70 through the inhibition of NFκB signaling, increase of the anti-inflammatory cytokines IL10 and IL12P40, and increased expression of the angiogenic and neurotrophic factors VEGFA and IL7. These studies provide proof of principle that apigenin has multiple therapeutic targets in preclinical models of DS.


Asunto(s)
Apigenina/farmacología , Síndrome de Down/tratamiento farmacológico , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Células Madre/efectos de los fármacos , Líquido Amniótico/citología , Líquido Amniótico/metabolismo , Animales , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Síndrome de Down/genética , Síndrome de Down/inmunología , Síndrome de Down/patología , Conducta Exploratoria/efectos de los fármacos , Femenino , Feto , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Interleucina-7/genética , Interleucina-7/inmunología , Antígeno Ki-67/genética , Antígeno Ki-67/inmunología , Masculino , Ratones , Nestina/genética , Nestina/inmunología , Neurogénesis/genética , Estrés Oxidativo/efectos de los fármacos , Factor de Transcripción PAX6/genética , Factor de Transcripción PAX6/inmunología , Embarazo , Cultivo Primario de Células , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/inmunología , Factores Sexuales , Células Madre/metabolismo , Células Madre/patología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/inmunología
2.
Pediatr Dev Pathol ; 22(1): 70-74, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29652239

RESUMEN

Simpson-Golabi-Behmel syndrome type I (SGBS, OMIM312870), caused by defects of the GPC3 and GPC4 genes on chromosome Xq26, is an X-linked recessive macrosomia/multiple congenital anomaly disorder characterized by somatic overgrowth, coarse facial features, variable congenital anomalies, increased tumor risk, and mild-to-moderate neurodevelopmental anomalies. We report the postmortem findings in 3 second-trimester male siblings with SGBS who displayed ambiguous genitalia (in all 3) and gonadal dysgenesis (ovotestis) (in 1), thus expanding the SGBS spectrum to include these disorders of sex development.


Asunto(s)
Anomalías Múltiples/diagnóstico , Arritmias Cardíacas/diagnóstico , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico , Gigantismo/diagnóstico , Cardiopatías Congénitas/diagnóstico , Discapacidad Intelectual/diagnóstico , Trastornos Ovotesticulares del Desarrollo Sexual/diagnóstico , Anomalías Múltiples/patología , Arritmias Cardíacas/patología , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Gigantismo/patología , Cardiopatías Congénitas/patología , Humanos , Discapacidad Intelectual/patología , Masculino , Trastornos Ovotesticulares del Desarrollo Sexual/patología , Mortinato
3.
Breast Cancer Res Treat ; 157(3): 405-11, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27180259

RESUMEN

For dual probe HER2 FISH assay, the 2013 CAP/ASCO guideline recommendations lowered the HER2/CEP17 ratio cut off for HER2 amplification to ≥2.0 and introduced an average HER2 copy number criterion for HER2 amplification (≥6.0/cell) and HER2 equivocal categories (≥4 and <6/cell). The HER2/CEP17 equivocal category is eliminated. The aim of this study is to assess the impact of 2013 HER2 FISH testing guideline recommendations update on the assignment of HER2 status with dual probe HER2 FISH assay. Dual probe HER2 FISH assay results on breast cancers from 09/2009 to 07/2015 that underwent reflex HER2 FISH testing after equivocal HER2 (2+) immunohistochemistry (IHC) were reviewed. HER2 copy number, CEP17 signals, and HER2/CEP ratios were noted. HER2 status was assigned as HER2 negative (HER2-), HER2 equivocal (HER2e), and HER2 amplified (HER2+) by applying both 2007 and 2013 CAP/ASCO HER2 FISH guideline recommendations and results were compared. New guidelines reclassified HER2 FISH status in a significant proportion of cases (8.3 %, 69/836; p = .021). There were 22 (2.6 %) more HER2+, 17 (2.1 %) more HER2e, and 39 (4.1 %) fewer HER2- tumors. Change of HER2 status correlated significantly with ≥3 CEP17 signals (38 vs. 2 %; p < .001). The 2013 CAP/ASCO guideline recommendations for HER2 FISH testing by dual probe assay increased the HER2 amplified and HER2 equivocal tumors. Increase in HER2 equivocal tumors would potentially increase the frequency of repeat HER2 testing. Tumors with ≥3 CEP17 signals, so-called chromosome 17 polysomy, are more likely to be impacted and classified as HER2 equivocal.


Asunto(s)
Neoplasias de la Mama/clasificación , Hibridación Fluorescente in Situ/métodos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Cromosomas Humanos Par 17/genética , Análisis Citogenético/métodos , Femenino , Amplificación de Genes , Humanos , Proteínas Asociadas a Microtúbulos , Fosfoproteínas/genética , Guías de Práctica Clínica como Asunto , Estudios Retrospectivos
4.
Exp Cell Res ; 335(2): 224-37, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25999147

RESUMEN

Protein phosphatase 6 (PP6) is a ubiquitous Ser/Thr phosphatase involved in an array of cellular processes. To assess the potential of PP6 as a therapeutic target in liver disorders, we attenuated expression of the PP6 catalytic subunit in HepG2 cells using lentiviral-transduced shRNA. Two PP6 knock-down (PP6KD) cell lines (90% reduction of PP6-C protein content) were studied in depth. Both proliferated at a rate similar to control cells. However, flow cytometry indicated G2/M cell cycle arrest that was accounted for by a shift of the cells from a diploid to tetraploid state. PP6KD cells did not show an increase in apoptosis, nor did they exhibit reduced viability in the presence of bleomycin or taxol. Gene expression analysis by microarray showed attenuated anti-inflammatory signaling. Genes associated with DNA replication were downregulated. Mass spectrometry-based phosphoproteomic analysis yielded 80 phosphopeptides representing 56 proteins that were significantly affected by a stable reduction in PP6-C. Proteins involved in DNA replication, DNA damage repair and pre-mRNA splicing were overrepresented among these. PP6KD cells showed intact mTOR signaling. Our studies demonstrated involvement of PP6 in a diverse set of biological pathways and an adaptive response that may limit the effectiveness of targeting PP6 in liver disorders.


Asunto(s)
Fosfoproteínas Fosfatasas/fisiología , Adaptación Fisiológica , Dominio Catalítico , Proliferación Celular , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Fenotipo , Fosfoproteínas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Subunidades de Proteína/fisiología , Proteoma/metabolismo , ARN Interferente Pequeño/genética , Transcriptoma
5.
Ann Neurol ; 76(4): 581-93, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25044251

RESUMEN

OBJECTIVE: Recently, Christianson syndrome (CS) has been determined to be caused by mutations in the X-linked Na(+) /H(+) exchanger 6 (NHE6). We aimed to determine the diagnostic criteria and mutational spectrum for CS. METHODS: Twelve independent pedigrees (14 boys, age = 4-19 years) with mutations in NHE6 were administered standardized research assessments, and mutations were characterized. RESULTS: The mutational spectrum was composed of 9 single nucleotide variants, 2 indels, and 1 copy number variation deletion. All mutations were protein-truncating or splicing mutations. We identified 2 recurrent mutations (c.1498 c>t, p.R500X; and c.1710 g>a, p.W570X). Otherwise, all mutations were unique. In our study, 7 of 12 mutations (58%) were de novo, in contrast to prior literature wherein mutations were largely inherited. We also report prominent neurological, medical, and behavioral symptoms. All CS participants were nonverbal and had intellectual disability, epilepsy, and ataxia. Many had prior diagnoses of autism and/or Angelman syndrome. Other neurologic symptoms included eye movement abnormalities (79%), postnatal microcephaly (92%), and magnetic resonance imaging evidence of cerebellar atrophy (33%). Regression was noted in 50%, with recurrent presentations involving loss of words and/or the ability to walk. Medical symptoms, particularly gastrointestinal symptoms, were common. Height and body mass index measures were below normal ranges in most participants. Behavioral symptoms included hyperkinetic behavior (100%), and a majority exhibited high pain threshold. INTERPRETATION: This is the largest cohort of independent CS pedigrees reported. We propose diagnostic criteria for CS. CS represents a novel neurogenetic disorder with general relevance to autism, intellectual disability, Angelman syndrome, epilepsy, and regression.


Asunto(s)
Ataxia/complicaciones , Ataxia/genética , Discapacidades del Desarrollo/genética , Epilepsia/complicaciones , Epilepsia/genética , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/genética , Microcefalia/complicaciones , Microcefalia/genética , Mutación/genética , Trastornos de la Motilidad Ocular/complicaciones , Trastornos de la Motilidad Ocular/genética , Intercambiadores de Sodio-Hidrógeno/genética , Adolescente , Ataxia/patología , Trastorno Autístico/etiología , Trastorno Autístico/genética , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Niño , Preescolar , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/patología , Progresión de la Enfermedad , Electroencefalografía , Epilepsia/etiología , Epilepsia/patología , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Genotipo , Humanos , Discapacidad Intelectual/patología , Imagen por Resonancia Magnética , Masculino , Microcefalia/patología , Trastornos de la Motilidad Ocular/patología , Fenotipo , Análisis de Regresión , Adulto Joven
6.
Am J Med Genet A ; 164A(5): 1227-33, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24458767

RESUMEN

Monochorionic-diamniotic twins are usually monozygotic twins and known to be associated with adverse obstetric and perinatal outcomes. Cases of discordant karyotype of monozygotic twins are rare and most involves sex chromosomes. We present the first case of monochorionic twins with discordant karyotype manifested as mosaic trisomy 14 in one twin (B) and a normal karyotype in the other (A). We describe the postmortem pathological and imaging findings of the trisomic twin and for the first time neuropathological findings of this entity. Metaphase chromosome analysis of twin B using fetal tissue showed a 47,XX, +14 karyotype. Chromosomal microarray analysis (CMA) using fetal tissue revealed 38% mosaicism. CMA with placental tissue from both sides demonstrated normal karyotype and confirmed monozygosity, highlighting the value of array based testing on diagnosing mosaicism and zygosity.


Asunto(s)
Trisomía/diagnóstico , Trisomía/genética , Gemelos Monocigóticos , Alelos , Autopsia , Cromosomas Humanos Par 14/genética , Hibridación Genómica Comparativa , Resultado Fatal , Femenino , Genotipo , Humanos , Cariotipificación , Imagen por Resonancia Magnética , Mosaicismo , Polimorfismo de Nucleótido Simple , Ultrasonografía Prenatal
7.
Am J Med Genet A ; 161A(4): 787-91, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23495067

RESUMEN

We report on a pedigree with a pair of brothers each with minor anomalies, developmental delay, and autistic-symptoms who share an unbalanced translocation (not detectable by karyotype). The unbalanced translocation involves a 7.1 Mb loss of the terminal portion of 10q, and a 4.2 Mb gain of 11q. One of the brothers also developed a cerebellar juvenile pilocytic astrocytoma. The father was found to be a balanced carrier and the couple had a previous miscarriage. We demonstrate that the breakpoint for the triplicated region from chromosome 11 is adjacent to two IgLON genes, namely Neurotrimin (NTM) and Opioid Binding Protein/Cell Adhesion Molecule-like (OPCML). These genes are highly similar neural cell adhesion molecules that have been implicated in synaptogenesis and oncogenesis, respectively. The children also have a 10q deletion and are compared to other children with the 10q deletion syndrome which generally does not involve autism spectrum disorders (ASDs) or cancer. Together these data support a role for NTM and OPCML in developmental delay and potentially in cancer susceptibility.


Asunto(s)
Astrocitoma/genética , Neoplasias Cerebelosas/genética , Trastornos Generalizados del Desarrollo Infantil/genética , Deleción Cromosómica , Translocación Genética , Trisomía , Astrocitoma/diagnóstico , Neoplasias Cerebelosas/diagnóstico , Niño , Trastornos Generalizados del Desarrollo Infantil/diagnóstico , Preescolar , Cromosomas Humanos Par 10 , Cromosomas Humanos Par 11 , Hibridación Genómica Comparativa , Proteínas del Citoesqueleto , Humanos , Hibridación Fluorescente in Situ , Cariotipo , Masculino , Linaje , Proteínas/genética
8.
Proc Natl Acad Sci U S A ; 106(23): 9425-9, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19474297

RESUMEN

To characterize the differences between second trimester Down syndrome (DS) and euploid fetuses, we used Affymetrix microarrays to compare gene expression in uncultured amniotic fluid supernatant samples. Functional pathway analysis highlighted the importance of oxidative stress, ion transport, and G protein signaling in the DS fetuses. Further evidence supporting these results was derived by correlating the observed gene expression patterns to those of small molecule drugs via the Connectivity Map. Our results suggest that there are secondary adverse consequences of DS evident in the second trimester, leading to testable hypotheses about possible antenatal therapy for DS.


Asunto(s)
Líquido Amniótico/metabolismo , Síndrome de Down/genética , Síndrome de Down/metabolismo , Feto/metabolismo , Perfilación de la Expresión Génica , Estrés Oxidativo , Femenino , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo
9.
Hum Genet ; 129(3): 295-305, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21152935

RESUMEN

Trisomy 18 is a common human aneuploidy that is associated with significant perinatal mortality. Unlike the well-characterized "critical region" in trisomy 21 (21q22), there is no corresponding region on chromosome 18 associated with its pathogenesis. The high morbidity and mortality of affected individuals has limited extensive investigations. In order to better understand the molecular mechanisms underlying the congenital anomalies observed in this condition, we investigated the in utero gene expression profile of second trimester fetuses affected with trisomy 18. Total RNA was extracted from cell-free amniotic fluid supernatant from aneuploid fetuses and euploid controls matched for gestational age and hybridized to Affymetrix U133 Plus 2.0 arrays. Individual differentially expressed transcripts were obtained by two-tailed t tests. Over-represented functional pathways among these genes were identified with DAVID and Ingenuity(®) Pathways Analysis. Results show that three hundred and fifty-two probe sets representing 251 annotated genes were statistically significantly differentially expressed between trisomy 18 and controls. Only 7 genes (2.8% of the annotated total) were located on chromosome 18, including ROCK1, an up-regulated gene involved in valvuloseptal and endocardial cushion formation. Pathway analysis indicated disrupted function in ion transport, MHCII/T cell mediated immunity, DNA repair, G-protein mediated signaling, kinases, and glycosylation. Significant down-regulation of genes involved in adrenal development was identified, which may explain both the abnormal maternal serum estriols and the pre and postnatal growth restriction in trisomy 18. Comparison of this gene set to one previously generated for trisomy 21 fetuses revealed only six overlapping differentially regulated genes. This study contributes novel information regarding functional developmental gene expression differences in fetuses with trisomy 18.


Asunto(s)
Trisomía , Glándulas Suprarrenales/crecimiento & desarrollo , Glándulas Suprarrenales/metabolismo , Algoritmos , Cromosomas Humanos Par 18/genética , Reparación del ADN/genética , Regulación hacia Abajo , Cojinetes Endocárdicos , Estriol/sangre , Femenino , Proteínas de Unión al GTP/genética , Perfilación de la Expresión Génica , Glicosilación , Humanos , Inmunidad Celular/genética , Transporte Iónico/genética , Anotación de Secuencia Molecular , Embarazo , Segundo Trimestre del Embarazo , Trisomía/genética , Regulación hacia Arriba , Quinasas Asociadas a rho/genética
10.
Exp Mol Pathol ; 89(3): 248-59, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20655306

RESUMEN

Cholangiocarcinoma, a severe form of biliary cancer, has a high mortality rate resulting partially from the advanced stage of disease at earliest diagnosis. A better understanding of the progressive molecular and cellular changes occurring during spontaneous cholangiocarcinogenesis is needed to identify potential biomarkers for diagnosis/prognosis or targets for novel therapeutics. Here, we show that with continued passage (p) in vitro, rat bile duct epithelial cells (BDEC) accumulated neoplastic characteristics that by mid-passage (p31-85) included alterations in morphology, increased growth rate, growth factor independence, decreased cell adhesion, loss of cholangiocyte markers expressed at low passage (p<30), and onset of aneuploidy. At high passage (p>85), BDEC cultures showed increasing numbers of cells expressing activated, tyrosine phosphorylated ErbB-2/Neu, a receptor tyrosine kinase previously reported to be at elevated levels in cholangiocarcinomas. Enrichment for high passage ErbB-2/Neu-positive cells yielded several anchorage-independent sub-lines with elevated levels of activated ErbB-2/Neu and increased expression of cyclooxygenase-2 (COX-2). When injected into immunodeficient beige/nude/xid mice, these sub-lines formed poorly differentiated cystic tumors strongly positive for rat cholangiocyte markers, a finding consistent with a previous report showing the susceptibility of high passage, non-tumorigenic BDEC to transformation by activated ErbB-2/Neu. Mid passage BDEC, in contrast, were resistant to the transforming activity of activated ErbB-2/Neu and remained anchorage dependent in vitro and non-tumorigenic in vivo following stable transfection. Based on these findings, we concluded that during progression to high passage, cultured BDEC undergo preneoplastic changes that enhance their susceptibility to transformation by ErbB-2/Neu. The ability to generate cells at different points in the process of spontaneous neoplastic transformation offers a valuable model system for identifying molecular features that determine whether over-expression of activated ErbB-2/Neu is necessary and sufficient to induce neoplastic conversion.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Transformación Celular Neoplásica/genética , Células Epiteliales/patología , Receptor ErbB-2/genética , Animales , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Separación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Células Epiteliales/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Immunoblotting , Ratones , Ratas , Receptor ErbB-2/metabolismo , Transfección
11.
Gynecol Oncol Rep ; 32: 100561, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32258332

RESUMEN

The human epidermal growth factor receptor 2 (Her2) is tested in many human cancers, including breast, bladder, pancreatic, ovarian and stomach. The American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) have issued Clinical Practice Guidelines for reporting Her2 results for breast carcinomas (Wolf et al., 2018). For the last 1-2 years Her2/neu is tested in endometrial serous carcinoma, especially in recurrent tumors or non-responsive tumors as an option for additional treatment. College of American Pathologists (CAP) offers a template for prognostic marker reporting results for specimens with endometrial carcinomas (Fitzgibbons et al., 2019). Her2/neu testing by immunohistochemistry (IHC) mandates rigorous fixation time control, e.g., fixation time should fall within 6-72 h (Recommendations for Her2 Testing in Breast Cancer, 2013). For that reason, in breast cancers, Her2/neu testing is done on initial core biopsy specimens. The test is however, repeated on excision specimen in high grade tumors where Her2/neu expression was initially negative on core biopsies. For endometrial serous carcinoma no guidelines have been set or proposed as of yet. The Gynecologic Oncologists request this test because of proven benefit of adding Trastuzumab (Fader et al., 2018) and that is why it is important to documenting the findings in this report in the literature so that an informed request can be made by the treating oncologist when multiple tissue samples from the same patient are available for testing. Similarly pathologists also can decide which would be the best sample to test when no instruction is received. We report here three separate scenarios of uterine serous carcinomas in which the Her2/neu expressions were unique enough to justify documentation and therefore have implications for determining which specimen is ideal for the Her2 overexpression testing and likely to have highest possibility in identifying the Her2/neu overexpressed clone in the tumor which would expand the therapeutic options for the patients.

12.
Am J Pathol ; 173(1): 42-56, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18535181

RESUMEN

Premature infants are at risk for bronchopulmonary dysplasia, a complex condition characterized by impaired alveolar development and increased alveolar epithelial apoptosis. The functional involvement of pulmonary apoptosis in bronchopulmonary dysplasia- associated alveolar disruption remains undetermined. The aims of this study were to generate conditional lung-specific Fas-ligand (FasL) transgenic mice and to determine the effects of FasL-induced respiratory epithelial apoptosis on alveolar remodeling in postcanalicular lungs. Transgenic (TetOp)(7)-FasL responder mice, generated by pronuclear microinjection, were bred with Clara cell secretory protein (CCSP)-rtTA activator mice. Doxycycline (Dox) was administered from embryonal day 14 to postnatal day 7, and lungs were studied between embryonal day 19 and postnatal day 21. Dox administration induced marked respiratory epithelium-specific FasL mRNA and protein up-regulation in double-transgenic CCSP-rtTA(+)/(TetOp)(7)-FasL(+) mice compared with single-transgenic CCSP-rtTA(+) littermates. The Dox-induced FasL up-regulation was associated with dramatically increased apoptosis of alveolar type II cells and Clara cells, disrupted alveolar development, decreased vascular density, and increased postnatal lethality. These data demonstrate that FasL-induced alveolar epithelial apoptosis during postcanalicular lung remodeling is sufficient to disrupt alveolar development after birth. The availability of inducible lung-specific FasL transgenic mice will facilitate studies of the role of apoptosis in normal and disrupted alveologenesis and may lead to novel therapeutic approaches for perinatal and adult pulmonary diseases characterized by dysregulated apoptosis.


Asunto(s)
Apoptosis/fisiología , Células Epiteliales/patología , Proteína Ligando Fas/metabolismo , Enfermedades Pulmonares/patología , Alveolos Pulmonares/crecimiento & desarrollo , Animales , Secuencia de Bases , Western Blotting , Modelos Animales de Enfermedad , Proteína Ligando Fas/genética , Femenino , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Enfermedades Pulmonares/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transgenes
13.
Int J Radiat Oncol Biol Phys ; 67(2): 405-9, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17097832

RESUMEN

PURPOSE: To determine the overall survival for patients with locally advanced, HER2 overexpressing, esophageal adenocarcinoma receiving trastuzumab, paclitaxel, cisplatin, and radiation on a Phase I-II study. METHODS AND MATERIALS: Patients with adenocarcinoma of the esophagus without distant organ metastases and 2+/3+ HER2 overexpression by immunohistochemistry (IHC) were eligible. All patients received cisplatin 25 mg/m2 and paclitaxel 50 mg/m2 weekly for 6 weeks with radiation therapy (RT) 50.4 Gy. Patients received trastuzumab at dose levels of 1, 1.5, or 2 mg/kg weekly for 5 weeks after an initial bolus of 2, 3, or 4 mg/kg. RESULTS: Nineteen patients were entered: 7 (37%) had celiac adenopathy, and 7 (37%) had retroperitoneal, portal adenopathy, or scalene adenopathy. Fourteen of 19 patients (74%) had either 3+ HER2 expression by immunohistochemistry, or an increase in HER2 gene copy number by HER2 gene amplification or high polysomy by fluorescence in situ hybridization. The median survival of all patients was 24 months and the 2-year survival was 50%. CONCLUSIONS: Assessment of the effect of trastuzumab in the treatment of patients with esophageal adenocarcinoma overexpressing HER2 is limited by the small number of patients in this study. Overall survival, however, was similar to prior studies without an increase in toxicity. Evaluation of HER2 status should be performed in future trials for patients with adenocarcinoma of the esophagus that investigate therapies targeting the HER family.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/radioterapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/radioterapia , Adenocarcinoma/metabolismo , Adenocarcinoma/secundario , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Cisplatino/administración & dosificación , Terapia Combinada/métodos , Esquema de Medicación , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Femenino , Amplificación de Genes , Genes erbB-2/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Paclitaxel/administración & dosificación , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Análisis de Supervivencia , Trastuzumab
15.
Sci Rep ; 6: 32353, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27586445

RESUMEN

Anatomical and functional brain abnormalities begin during fetal life in Down syndrome (DS). We hypothesize that novel prenatal treatments can be identified by targeting signaling pathways that are consistently perturbed in cell types/tissues obtained from human fetuses with DS and mouse embryos. We analyzed transcriptome data from fetuses with trisomy 21, age and sex-matched euploid controls, and embryonic day 15.5 forebrains from Ts1Cje, Ts65Dn, and Dp16 mice. The new datasets were compared to other publicly available datasets from humans with DS. We used the human Connectivity Map (CMap) database and created a murine adaptation to identify FDA-approved drugs that can rescue affected pathways. USP16 and TTC3 were dysregulated in all affected human cells and two mouse models. DS-associated pathway abnormalities were either the result of gene dosage specific effects or the consequence of a global cell stress response with activation of compensatory mechanisms. CMap analyses identified 56 molecules with high predictive scores to rescue abnormal gene expression in both species. Our novel integrated human/murine systems biology approach identified commonly dysregulated genes and pathways. This can help to prioritize therapeutic molecules on which to further test safety and efficacy. Additional studies in human cells are ongoing prior to pre-clinical prenatal treatment in mice.


Asunto(s)
Síndrome de Down/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Redes y Vías Metabólicas/genética , Ubiquitina Tiolesterasa/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/metabolismo , Síndrome de Down/patología , Embrión de Mamíferos , Femenino , Feto , Dosificación de Gen , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Ratones Transgénicos , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/farmacología , Biología de Sistemas/métodos , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
16.
JAMA ; 293(7): 836-42, 2005 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-15713773

RESUMEN

CONTEXT: No molecular biological tests are available to monitor the ongoing development of human fetuses in vivo. OBJECTIVE: To determine whether cell-free fetal messenger RNA (mRNA) in amniotic fluid can be detected using oligonucleotide microarrays to study large-scale gene expression in living human fetuses, with analysis of sex, gestational age, and fetal pathology as variables. DESIGN, SETTING, AND PATIENTS: Four samples of cell-free amniotic fluid were analyzed from pregnant women between 20 and 32 weeks' gestation and undergoing amnioreduction for polyhydramnios associated with twin-twin transfusion syndrome or hydrops fetalis (cases). The control consisted of 6 pooled amniotic fluid samples from women at 17 weeks' gestation and undergoing genetic amniocentesis. After extraction from the normally discarded fraction of amniotic fluid, RNA was amplified twice, labeled, and analyzed using gene expression microarrays. MAIN OUTCOME MEASURE: Relative mRNA expression in cell-free samples of amniotic fluid from fetuses with polyhydramnios at different gestational ages vs cell-free amniotic fluid from a pooled control. RESULTS: Thirty-six percent of 22,283 probe sets represented on the arrays were present in the cell-free amniotic fluid, and a median of 20% of all probe sets differed between cases and the pooled control. Only male samples expressed 1 Y chromosome transcript. The expression of some developmental transcripts, such as surfactant proteins, mucins, and keratins, changed with gestational age by up to 64-fold. A water transporter gene transcript was increased up to 18-fold in both twin-twin transfusion samples. Placental gene transcripts were not present in any samples. CONCLUSIONS: This pilot study demonstrates that cell-free fetal mRNA can be extracted from amniotic fluid and successfully hybridized to gene expression microarrays. Preliminary analysis suggests that gene expression changes can be detected in fetuses of different sexes, gestational age, and disease status. Cell-free mRNA in amniotic fluid appears to originate from the fetus and not the placenta.


Asunto(s)
Amniocentesis , Desarrollo Fetal/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/análisis , Femenino , Edad Gestacional , Humanos , Masculino , Proyectos Piloto
17.
Clin Lab Med ; 23(2): 481-502, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12848455

RESUMEN

In the past few decades, enormous progress has been made in the field of prenatal molecular genetic testing. Based on the inheritance patterns of the disease and type of mutation, prenatal diagnosis is possible using direct or indirect methods of detection. Although direct mutation analysis is highly accurate, accuracy of indirect mutation analysis depends on the distance of the DNA marker to the disease locus. In the past decade, the discovery of new concepts--such as atypical inheritance patterns due to UPD and imprinting and triplet repeat disorders--have helped to increase understanding of the molecular basis of these unusual genetic disorders. Prenatal diagnosis using a single cell from a blastomere is rapidly becoming routine in clinical practice. Noninvasive procedures to obtain fetal DNA for molecular testing also are progressing very rapidly. With the completion of the genome project, resources now are available for developing new technologies, such as microarrays (DNA chips), for accurate, simultaneous, mutation detection. The next few decades hold the promise of many more advances in genetic testing, drug discovery, and therapy.


Asunto(s)
Enfermedades Fetales/diagnóstico , Enfermedades Genéticas Congénitas/diagnóstico , Biología Molecular/métodos , Diagnóstico Prenatal , Adulto , Femenino , Enfermedades Fetales/genética , Enfermedades Genéticas Congénitas/genética , Humanos , Edad Materna , Embarazo , Embarazo de Alto Riesgo
18.
Pediatr Dev Pathol ; 17(5): 374-81, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24971487

RESUMEN

We report a case of a 31-week-gestation male newborn who died soon after birth from intractable respiratory failure and persistent pulmonary hypertension. The pregnancy had been complicated by intermittent bleeding between 13 and 20 weeks' gestation, attributed to peripheral placental separation, as well as bilateral fetal adrenal hemorrhage, first detected at 17 weeks' gestation. Postmortem examination revealed small, calcified adrenal glands as well as several remote cerebral and cerebellar infarcts. The lungs were hypoplastic (lung weight/body weight ratio: 1.64%; 10th percentile for 28-36 weeks' gestation: 2.27%) and distorted by exaggerated lobulation. Microscopically, the lungs exhibited several developmental anomalies, including focal acinar dysgenesis suggestive of arrested development in the pseudoglandular stage of development (8-16 weeks' gestation) (mainly in the upper lobes), and features of bronchial obstruction, including focal lobular hyperplasia and microcystic maldevelopment (mainly in the lower lobes). The adrenal and cerebral findings were consistent with a severe early-gestation hypoxic-ischemic insult, likely related to peripheral placental separation and chronic abruption. The co-occurrence and timing of these well-recognized hypoxic lesions provide further evidence that certain developmental lung anomalies, such as focal acinar dysplasia, focal lobular hyperplasia, and microcystic maldevelopment, may, at least in some cases, have a hypoxic/ischemic etiology.


Asunto(s)
Desprendimiento Prematuro de la Placenta/patología , Infarto Cerebral/patología , Enfermedades Pulmonares/patología , Pulmón/patología , Hemorragia Uterina , Corteza Suprarrenal/patología , Adulto , Infarto Cerebral/complicaciones , Infarto Cerebral/diagnóstico , Femenino , Feto/patología , Humanos , Recién Nacido , Pulmón/crecimiento & desarrollo , Enfermedades Pulmonares/complicaciones , Enfermedades Pulmonares/diagnóstico , Embarazo
19.
J Mol Diagn ; 13(5): 565-70, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21827969

RESUMEN

Microarray analysis of cell-free RNA in amniotic fluid (AF) supernatant has revealed differential fetal gene expression as a function of gestational age and karyotype. Once informative genes are identified, research moves to a more focused platform such as quantitative reverse transcriptase-PCR. Standardized NanoArray PCR (SNAP) is a recently developed gene profiling technology that enables the measurement of transcripts from samples containing reduced quantities or degraded nucleic acids. We used a previously developed SNAP gene panel as proof of concept to determine whether fetal functional gene expression could be ascertained from AF supernatant. RNA was extracted and converted to cDNA from 19 AF supernatant samples of euploid fetuses between 15 to 20 weeks of gestation, and transcript abundance of 21 genes was measured. Statistically significant differences in expression, as a function of advancing gestational age, were observed for 5 of 21 genes. ANXA5, GUSB, and PPIA showed decreasing gene expression over time, whereas CASC3 and ZNF264 showed increasing gene expression over time. Statistically significantly increased expression of MTOR and STAT2 was seen in female compared with male fetuses. This study demonstrates the feasibility of focused fetal gene expression analysis using SNAP technology. In the future, this technique could be optimized to examine specific genes instrumental in fetal organ system function, which could be a useful addition to prenatal care.


Asunto(s)
Líquido Amniótico/metabolismo , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Análisis por Micromatrices/métodos , Reacción en Cadena de la Polimerasa/métodos , Femenino , Humanos , Pulmón/metabolismo , Masculino , Análisis de Regresión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA