Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 28(20): 4466-4478, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-35653119

RESUMO

PURPOSE: Propagation of Ewing sarcoma requires precise regulation of EWS::FLI1 transcriptional activity. Determining the mechanisms of fusion regulation will advance our understanding of tumor progression. Here we investigated whether HOXD13, a developmental transcription factor that promotes Ewing sarcoma metastatic phenotypes, influences EWS::FLI1 transcriptional activity. EXPERIMENTAL DESIGN: Existing tumor and cell line datasets were used to define EWS::FLI1 binding sites and transcriptional targets. Chromatin immunoprecipitation and CRISPR interference were employed to identify enhancers. CUT&RUN and RNA sequencing defined binding sites and transcriptional targets of HOXD13. Transcriptional states were investigated using bulk and single-cell transcriptomic data from cell lines, patient-derived xenografts, and patient tumors. Mesenchymal phenotypes were assessed by gene set enrichment, flow cytometry, and migration assays. RESULTS: We found that EWS::FLI1 creates a de novo GGAA microsatellite enhancer in a developmentally conserved regulatory region of the HOXD locus. Knockdown of HOXD13 led to widespread changes in expression of developmental gene programs and EWS::FLI1 targets. HOXD13 binding was enriched at established EWS::FLI1 binding sites where it influenced expression of EWS::FLI1-activated genes. More strikingly, HOXD13 bound and activated EWS::FLI1-repressed genes, leading to adoption of mesenchymal and migratory cell states that are normally suppressed by the fusion. Single-cell analysis confirmed that direct transcriptional antagonism between HOXD13-mediated gene activation and EWS::FLI1-dependent gene repression defines the state of Ewing sarcoma cells along a mesenchymal axis. CONCLUSIONS: Ewing sarcoma tumors are comprised of tumor cells that exist along a mesenchymal transcriptional continuum. The identity of cells along this continuum is, in large part, determined by the competing activities of EWS::FLI1 and HOXD13. See related commentary by Weiss and Bailey, p. 4360.


Assuntos
Sarcoma de Ewing , Linhagem Celular Tumoral , Plasticidade Celular , Imunoprecipitação da Cromatina , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteína Proto-Oncogênica c-fli-1/genética , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/genética , Proteína EWS de Ligação a RNA/metabolismo , Sarcoma de Ewing/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Stem Cell Reports ; 16(3): 626-640, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33606989

RESUMO

Heterotopic ossification (HO) is a form of pathological cell-fate change of mesenchymal stem/precursor cells (MSCs) that occurs following traumatic injury, limiting range of motion in extremities and causing pain. MSCs have been shown to differentiate to form bone; however, their lineage and aberrant processes after trauma are not well understood. Utilizing a well-established mouse HO model and inducible lineage-tracing mouse (Hoxa11-CreERT2;ROSA26-LSL-TdTomato), we found that Hoxa11-lineage cells represent HO progenitors specifically in the zeugopod. Bioinformatic single-cell transcriptomic and epigenomic analyses showed Hoxa11-lineage cells are regionally restricted mesenchymal cells that, after injury, gain the potential to undergo differentiation toward chondrocytes, osteoblasts, and adipocytes. This study identifies Hoxa11-lineage cells as zeugopod-specific ectopic bone progenitors and elucidates the fate specification and multipotency that mesenchymal cells acquire after injury. Furthermore, this highlights homeobox patterning genes as useful tools to trace region-specific progenitors and enable location-specific gene deletion.


Assuntos
Osso e Ossos/metabolismo , Diferenciação Celular , Linhagem da Célula , Células-Tronco Mesenquimais/metabolismo , Ossificação Heterotópica/genética , Ossificação Heterotópica/metabolismo , Osteogênese , Adipócitos/metabolismo , Animais , Condrócitos/metabolismo , Modelos Animais de Doenças , Expressão Ectópica do Gene , Epigenômica , Feminino , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Ossificação Heterotópica/patologia , Osteoblastos/metabolismo , Análise de Célula Única , Tendões/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(13): 7296-7304, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32170021

RESUMO

Hox genes are indispensable for the proper patterning of the skeletal morphology of the axial and appendicular skeleton during embryonic development. Recently, it has been demonstrated that Hox expression continues from embryonic stages through postnatal and adult stages exclusively in a skeletal stem cell population. However, whether Hox genes continue to function after development has not been rigorously investigated. We generated a Hoxd11 conditional allele and induced genetic deletion at adult stages to show that Hox11 genes play critical roles in skeletal homeostasis of the forelimb zeugopod (radius and ulna). Conditional loss of Hox11 function at adult stages leads to replacement of normal lamellar bone with an abnormal woven bone-like matrix of highly disorganized collagen fibers. Examining the lineage from the Hox-expressing mutant cells demonstrates no loss of stem cell population. Differentiation in the osteoblast lineage initiates with Runx2 expression, which is observed similarly in mutants and controls. With loss of Hox11 function, however, osteoblasts fail to mature, with no progression to osteopontin or osteocalcin expression. Osteocyte-like cells become embedded within the abnormal bony matrix, but they completely lack dendrites, as well as the characteristic lacuno-canalicular network, and do not express SOST. Together, our studies show that Hox11 genes continuously function in the adult skeleton in a region-specific manner by regulating differentiation of Hox-expressing skeletal stem cells into the osteolineage.


Assuntos
Osso e Ossos/embriologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Animais , Osso e Ossos/metabolismo , Diferenciação Celular , Condrócitos/metabolismo , Feminino , Membro Anterior/embriologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Homeobox/genética , Genes Homeobox/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Esqueleto/embriologia , Fatores de Transcrição/metabolismo
4.
Nat Commun ; 10(1): 3168, 2019 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-31320650

RESUMO

Multipotent mesenchymal stromal cells (MSCs) are required for skeletal formation, maintenance, and repair throughout life; however, current models posit that postnatally arising long-lived adult MSCs replace transient embryonic progenitor populations. We previously reported exclusive expression and function of the embryonic patterning transcription factor, Hoxa11, in adult skeletal progenitor-enriched MSCs. Here, using a newly generated Hoxa11-CreERT2 lineage-tracing system, we show Hoxa11-lineage marked cells give rise to all skeletal lineages throughout the life of the animal and persist as MSCs. Hoxa11 lineage-positive cells give rise to previously described progenitor-enriched MSC populations marked by LepR-Cre and Osx-CreER, placing them upstream of these populations. Our studies establish that Hox-expressing cells are skeletal stem cells that arise from the earliest stages of skeletal development and self-renew throughout the life of the animal.


Assuntos
Adipócitos/citologia , Condrócitos/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Animais , Regeneração Óssea/genética , Osso e Ossos/citologia , Osso e Ossos/embriologia , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Receptores para Leptina/genética , Fator de Transcrição Sp7/genética
5.
Cancers (Basel) ; 11(3)2019 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-30845695

RESUMO

Ewing sarcomas predominantly arise in pelvic and stylopod bones (i.e., femur and humerus), likely as a consequence of EWS-FLI1 oncogene-induced transformation of mesenchymal stem/progenitor cells (MSCs). MSCs located in the embryonic superficial zone cells (eSZ) of limbs express anatomically distinct posterior Hox genes. Significantly, high expression of posterior HOXD genes, especially HOXD13, is a hallmark of Ewing sarcoma. These data drove our hypothesis that Hox genes in posterior skeleton MSCs contribute to Ewing sarcoma tumorigenesis. We isolated eSZ cells from stylopod and zeugopod (i.e., tibia/fibula, radius/ulna) bones, from wild-type and Hoxd13 mutant embryos, and tested the impact of EWS-FLI1 transduction on cell proliferation, gene expression, and tumorigenicity. Our data demonstrate that both stylopod and zeugopod eSZ cells tolerate EWS-FLI1 but that stylopod eSZ cells are relatively more susceptible, demonstrating changes in proliferation and gene expression consistent with initiation of malignant transformation. Significantly, loss of Hoxd13 had no impact, showing that it is dispensable for the initiation of EWS-FLI1-induced transformation in mouse MSCs. These findings show that MSCs from anatomically distinct sites are differentially susceptible to EWS-FLI1-induced transformation, supporting the premise that the dominant presentation of Ewing sarcoma in pelvic and stylopod bones is attributable to anatomically-defined differences in MSCs.

6.
Curr Top Dev Biol ; 132: 451-486, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30797517

RESUMO

The limb musculoskeletal system provides a primary means for locomotion, manipulation of objects and protection for most vertebrate organisms. Intricate integration of the bone, tendon and muscle tissues are required for function. These three tissues arise largely independent of one another, but the connections formed during later development are maintained throughout life and are re-established following injury. Each of these tissues also have mesenchymal stem/progenitor cells that function in maintenance and repair. Here in, we will review the major events in the development of limb skeleton, tendon, and muscle tissues, their response to injury, and discuss current knowledge regarding resident progenitor/stem cells within each tissue that participate in development, repair, and regeneration in vivo.


Assuntos
Extremidades/embriologia , Extremidades/fisiologia , Desenvolvimento Musculoesquelético/fisiologia , Regeneração/fisiologia , Animais , Osso e Ossos/citologia , Osso e Ossos/embriologia , Osso e Ossos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Músculos/citologia , Músculos/embriologia , Músculos/fisiologia , Desenvolvimento Musculoesquelético/genética , Regeneração/genética , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Tendões/citologia , Tendões/embriologia , Tendões/fisiologia
7.
J Bone Miner Res ; 32(8): 1750-1760, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28470721

RESUMO

The processes that govern fracture repair rely on many mechanisms that recapitulate embryonic skeletal development. Hox genes are transcription factors that perform critical patterning functions in regional domains along the axial and limb skeleton during development. Much less is known about roles for these genes in the adult skeleton. We recently reported that Hox11 genes, which function in zeugopod development (radius/ulna and tibia/fibula), are also expressed in the adult zeugopod skeleton exclusively in PDGFRα+/CD51+/LepR+ mesenchymal stem/stromal cells (MSCs). In this study, we use a Hoxa11eGFP reporter allele and loss-of-function Hox11 alleles, and we show that Hox11 expression expands after zeugopod fracture injury, and that loss of Hox11 function results in defects in endochondral ossification and in the bone remodeling phase of repair. In Hox11 compound mutant fractures, early chondrocytes are specified but show defects in differentiation, leading to an overall deficit in the cartilage production. In the later stages of the repair process, the hard callus remains incompletely remodeled in mutants due, at least in part, to abnormal bone matrix organization. Overall, our data supports multiple roles for Hox11 genes following fracture injury in the adult skeleton. © 2017 American Society for Bone and Mineral Research.


Assuntos
Alelos , Remodelação Óssea/genética , Condrócitos/metabolismo , Consolidação da Fratura , Fraturas Ósseas , Proteínas de Homeodomínio , Animais , Condrócitos/patologia , Feminino , Fraturas Ósseas/genética , Fraturas Ósseas/metabolismo , Fraturas Ósseas/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Camundongos Mutantes
8.
Dev Cell ; 39(6): 653-666, 2016 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-27939685

RESUMO

Posterior Hox genes (Hox9-13) are critical for patterning the limb skeleton along the proximodistal axis during embryonic development. Here we show that Hox11 paralogous genes, which developmentally pattern the zeugopod (radius/ulna and tibia/fibula), remain regionally expressed in the adult skeleton. Using Hoxa11EGFP reporter mice, we demonstrate expression exclusively in multipotent mesenchymal stromal cells (MSCs) in the bone marrow of the adult zeugopod. Hox-positive cells express PDGFRα and CD51, are marked by LepR-Cre, and exhibit colony-forming unit fibroblast activity and tri-lineage differentiation in vitro. Loss of Hox11 function leads to fracture repair defects, including reduced cartilage formation and delayed ossification. Hox mutant cells are defective in osteoblastic and chondrogenic differentiation in tri-lineage differentiation experiments, and these defects are zeugopod specific. In the stylopod (humerus and femur) and sternum, bone marrow MSCs express other regionally restricted Hox genes, and femur fractures heal normally in Hox11 mutants. Together, our data support regional Hox expression and function in skeletal MSCs.


Assuntos
Envelhecimento/metabolismo , Células da Medula Óssea/metabolismo , Proteínas de Homeodomínio/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Consolidação da Fratura , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Mesenquimais , Camundongos Endogâmicos C57BL
9.
PLoS One ; 10(12): e0145225, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26710299

RESUMO

The Hedgehog (Hh) signaling pathway directs a multitude of cellular responses during embryogenesis and adult tissue homeostasis. Stimulation of the pathway results in activation of Hh target genes by the transcription factor Ci/Gli, which binds to specific motifs in genomic enhancers. In Drosophila, only a few enhancers (patched, decapentaplegic, wingless, stripe, knot, hairy, orthodenticle) have been shown by in vivo functional assays to depend on direct Ci/Gli regulation. All but one (orthodenticle) contain more than one Ci/Gli site, prompting us to directly test whether homotypic clustering of Ci/Gli binding sites is sufficient to define a Hh-regulated enhancer. We therefore developed a computational algorithm to identify Ci/Gli clusters that are enriched over random expectation, within a given region of the genome. Candidate genomic regions containing Ci/Gli clusters were functionally tested in chicken neural tube electroporation assays and in transgenic flies. Of the 22 Ci/Gli clusters tested, seven novel enhancers (and the previously known patched enhancer) were identified as Hh-responsive and Ci/Gli-dependent in one or both of these assays, including: Cuticular protein 100A (Cpr100A); invected (inv), which encodes an engrailed-related transcription factor expressed at the anterior/posterior wing disc boundary; roadkill (rdx), the fly homolog of vertebrate Spop; the segment polarity gene gooseberry (gsb); and two previously untested regions of the Hh receptor-encoding patched (ptc) gene. We conclude that homotypic Ci/Gli clustering is not sufficient information to ensure Hh-responsiveness; however, it can provide a clue for enhancer recognition within putative Hedgehog target gene loci.


Assuntos
Biologia Computacional/métodos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Proteínas Hedgehog/genética , Proteínas Oncogênicas/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Embrião de Galinha , Drosophila melanogaster/genética , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas Hedgehog/metabolismo , Tubo Neural/metabolismo , Sequências Reguladoras de Ácido Nucleico , Análise de Sequência de DNA , Transdução de Sinais/genética , Asas de Animais/embriologia , Proteína GLI1 em Dedos de Zinco
10.
Dev Biol ; 402(2): 239-52, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25848697

RESUMO

Proper levels of Hedgehog (HH) signaling are essential during embryonic development and adult tissue homeostasis. A central mechanism to control HH pathway activity is through the regulation of secreted HH ligands at the plasma membrane. Recent studies have revealed a collective requirement for the cell surface co-receptors GAS1, CDON and BOC in HH signal transduction. Despite their requirement in HH pathway function, the mechanisms by which these proteins act to promote HH signaling remain poorly understood. Here we focus on the function of the two structurally related co-receptors, CDON and BOC. We utilized an in vivo gain-of-function approach in the developing chicken spinal cord to dissect the structural requirements for CDON and BOC function in HH signal transduction. Notably, we find that although CDON and BOC display functional redundancy during HH-dependent ventral neural patterning, these molecules utilize distinct molecular mechanisms to execute their HH-promoting effects. Specifically, we define distinct membrane attachment requirements for CDON and BOC function in HH signal transduction. Further, we identify novel and separate extracellular motifs in CDON and BOC that are required to promote HH signaling. Together, these data suggest that HH co-receptors employ distinct mechanisms to mediate HH pathway activity.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteínas Hedgehog/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Medula Espinal/embriologia , Animais , Western Blotting , Células COS , Moléculas de Adesão Celular/genética , Membrana Celular/metabolismo , Embrião de Galinha , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Eletroporação , Imunofluorescência , Processamento de Imagem Assistida por Computador , Imunoprecipitação , Modelos Moleculares , Estrutura Terciária de Proteína , Receptores de Superfície Celular/genética , Medula Espinal/metabolismo
11.
Cell Rep ; 9(2): 484-94, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25310976

RESUMO

Pancreatic cancer, a hypovascular and highly desmoplastic cancer, is characterized by tumor expression of Hedgehog (HH) ligands that signal to fibroblasts in the surrounding stroma that in turn promote tumor survival and growth. However, the mechanisms and consequences of stromal HH pathway activation are not well understood. Here, we show that the HH coreceptors GAS1, BOC, and CDON are expressed in cancer-associated fibroblasts. Deletion of two coreceptors (Gas1 and Boc) in fibroblasts reduces HH responsiveness. Strikingly, these fibroblasts promote greater tumor growth in vivo that correlates with increased tumor-associated vascularity. In contrast, deletion of all three coreceptors (Gas1, Boc, and Cdon) results in the near complete abrogation of HH signaling and a corresponding failure to promote tumorigenesis and angiogenesis. Collectively, these data identify a role for HH dosage in pancreatic cancer promotion and may explain the clinical failure of HH pathway blockade as a therapeutic approach in pancreatic cancer.


Assuntos
Dosagem de Genes , Proteínas Hedgehog/metabolismo , Neovascularização Patológica/genética , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Deleção de Genes , Proteínas Hedgehog/genética , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Camundongos , Neovascularização Patológica/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
12.
Development ; 140(16): 3423-34, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23900540

RESUMO

Hedgehog (HH) signaling is essential for vertebrate and invertebrate embryogenesis. In Drosophila, feedback upregulation of the HH receptor Patched (PTC; PTCH in vertebrates), is required to restrict HH signaling during development. By contrast, PTCH1 upregulation is dispensable for early HH-dependent patterning in mice. Unique to vertebrates are two additional HH-binding antagonists that are induced by HH signaling, HHIP1 and the PTCH1 homologue PTCH2. Although HHIP1 functions semi-redundantly with PTCH1 to restrict HH signaling in the developing nervous system, a role for PTCH2 remains unresolved. Data presented here define a novel role for PTCH2 as a ciliary localized HH pathway antagonist. While PTCH2 is dispensable for normal ventral neural patterning, combined removal of PTCH2- and PTCH1-feedback antagonism produces a significant expansion of HH-dependent ventral neural progenitors. Strikingly, complete loss of PTCH2-, HHIP1- and PTCH1-feedback inhibition results in ectopic specification of ventral cell fates throughout the neural tube, reflecting constitutive HH pathway activation. Overall, these data reveal an essential role for ligand-dependent feedback inhibition of vertebrate HH signaling governed collectively by PTCH1, PTCH2 and HHIP1.


Assuntos
Padronização Corporal , Proteínas de Transporte/metabolismo , Glicoproteínas de Membrana/metabolismo , Tubo Neural/metabolismo , Receptores de Superfície Celular/metabolismo , Células 3T3 , Animais , Proteínas de Transporte/genética , Embrião de Galinha , Embrião de Mamíferos/metabolismo , Retroalimentação Fisiológica , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Ligantes , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Patched , Receptor Patched-1 , Receptor Patched-2 , Ligação Proteica , Receptores de Superfície Celular/genética , Transdução de Sinais , Transcrição Gênica
13.
Pediatr Crit Care Med ; 13(5): 549-53, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22805159

RESUMO

OBJECTIVE: Among infants with single-ventricle heart disease who require surgical palliation, central venous access is routinely obtained via the umbilical or femoral veins. Both routes are associated with potential complications, including thrombosis. We sought to analyze the clinical outcomes of patients with umbilical venous catheter vs. femoral central venous catheter placement at the time of initial central venous access in this high-risk patient population. DESIGN: This was a retrospective study, with data collected including demographics, catheter type, duration, complications, and clinical outcomes. Patients were designated as group 1 (initial umbilical venous catheter placed, n = 70) or group 2 (initial femoral central venous catheter placed, n = 19). SETTING: The study was conducted at a single tertiary care referral institution. PATIENTS: We included all 89 patients who underwent single-ventricle palliation at this institution in 2007 and 2008. MEASUREMENTS AND MAIN RESULTS: The overall rates of survival to hospital discharge, thrombosis, and iliofemoral vein occlusion were 82%, 18%, and 21%, respectively. The proportion of thrombosis was 11% in group 1, compared with 42% in group 2 (p < .01). The proportion of iliofemoral vein occlusion was 16% in group 1, compared with 42% in group 2 (p = .02). The proportions of catheter-associated bloodstream infection, need for transhepatic access, and ultrasound-documented thrombus at the inferior vena caval-right atrial junction did not differ significantly between the groups. Patients with non-tunneled femoral central venous catheters for ≥14 days had a higher prevalence of thrombosis (52%) than those with femoral central venous catheters for <14 days (13%) but no difference in the prevalence of iliofemoral vein occlusion. CONCLUSIONS: In this population, initial placement of an umbilical venous catheter rather than a femoral venous catheter resulted in significantly lower risks of catheter thrombosis and iliofemoral vein occlusion. For femoral venous catheters, the prevalence of thrombosis, but not of iliofemoral vein occlusion, is proportional to the duration of catheterization.


Assuntos
Cateterismo Venoso Central/efeitos adversos , Cateterismo Venoso Central/métodos , Sepse/etiologia , Tromboembolia Venosa/etiologia , Trombose Venosa/etiologia , Cateteres Venosos Centrais/efeitos adversos , Intervalos de Confiança , Feminino , Veia Femoral , Técnica de Fontan , Cardiopatias Congênitas/cirurgia , Humanos , Recém-Nascido , Tempo de Internação , Modelos Logísticos , Masculino , Razão de Chances , Estudos Retrospectivos , Estatísticas não Paramétricas , Fatores de Tempo , Veias Umbilicais
14.
Dev Cell ; 20(6): 775-87, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21664576

RESUMO

Secreted Hedgehog (HH) ligands signal through the canonical receptor Patched (PTCH1). However, recent studies implicate three additional HH-binding, cell-surface proteins, GAS1, CDO, and BOC, as putative coreceptors for HH ligands. A central question is to what degree these coreceptors function similarly and what their collective requirement in HH signal transduction is. Here we provide evidence that GAS1, CDO, and BOC play overlapping and essential roles during HH-mediated ventral neural patterning of the mammalian neural tube. Specifically, we demonstrate two important roles for these molecules: an early role in cell fate specification of multiple neural progenitors and a later role in motor neuron progenitor maintenance. Most strikingly, genetic loss-of-function experiments indicate an obligatory requirement for GAS1, CDO, and BOC in HH pathway activity in multiple tissues.


Assuntos
Moléculas de Adesão Celular/fisiologia , Proteínas de Ciclo Celular/fisiologia , Proteínas Hedgehog/fisiologia , Imunoglobulina G/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Neurônios/metabolismo , Receptores de Superfície Celular/fisiologia , Animais , Padronização Corporal , Embrião de Galinha , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Extremidades/embriologia , Imunofluorescência , Proteínas Ligadas por GPI/fisiologia , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Sistema Nervoso/citologia , Sistema Nervoso/metabolismo , Neurônios/citologia , Transdução de Sinais , Proteína Gli2 com Dedos de Zinco
15.
J Child Adolesc Psychopharmacol ; 20(4): 309-15, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20807069

RESUMO

The antiglutamatergic drug riluzole (Rilutek) is presently being used off label in the treatment of psychiatric conditions in adult patients and, increasingly, in children. This article briefly reviews the pharmacology of this drug and its current investigative and clinical uses and adverse effects. It also reports on our experience to date in the study of the drug in children, with emphasis on adverse effects noted so far in these younger patients.


Assuntos
Transtornos Mentais/tratamento farmacológico , Transtorno Obsessivo-Compulsivo/tratamento farmacológico , Riluzol/uso terapêutico , Adulto , Fatores Etários , Animais , Criança , Ensaios Clínicos como Assunto , Antagonistas de Aminoácidos Excitatórios/efeitos adversos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Humanos , Transtornos Mentais/fisiopatologia , Transtorno Obsessivo-Compulsivo/fisiopatologia , Riluzol/efeitos adversos , Riluzol/farmacologia
16.
Pharmacol Biochem Behav ; 95(2): 235-41, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20122953

RESUMO

Neurotensin receptor type 1 (NTS1) is known to mediate a variety of biological functions of neurotensin (NT) in the central nervous system. In this study, we found that NTS1 null mice displayed decreased sensitivity to the ataxic effect of ethanol on the rotarod and increased ethanol consumption when given a free choice between ethanol and tap water containing bottles. Interestingly, the administration of NT69L, a brain-permeable NT analog, increased ethanol sensitivity in wild-type littermates but had no such effect in NTS1 null mice, suggesting that NTS1 contributes to NT-mediated ethanol intoxication. Furthermore, the daily treatment of NT69L, for 4 consecutive days, significantly reduced alcohol preference and consumption in wild-type littermates but had no such effects in NTS1 null mice in a two-bottle drinking experiment. Our study provides evidence for possible pharmacological roles of NT69L in which it increases sensitivity to the ataxic effect, and decreases voluntary consumption, of ethanol. Our study also demonstrates NTS1-mediated behavioral effects of NT69L. Therefore, our findings will be useful for understanding some aspects of alcoholism as well as to develop novel pharmacological therapeutic options for humans.


Assuntos
Etanol/toxicidade , Receptores de Neurotensina/fisiologia , Animais , Sequência de Bases , Primers do DNA , Etanol/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Neurotensina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...