Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 307(10): L765-74, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25217661

RESUMO

Emerging epidemiological evidence reveals a link between lung disease and exposure to indoor pollutants such as perfluorinated compounds (PFCs). PFC exposure during critical developmental stages may increase asthma susceptibility. Thus, in a murine model, we tested the hypothesis that early life and continued exposure to two ubiquitous household PFCs, perfluorooctanoic acid (PFOA) and perflurooctanesulfonic acid (PFOS), can induce lung dysfunction that exacerbates allergen-induced airway hyperresponsiveness (AHR) and inflammation. Balb/c mice were exposed to PFOA or PFOS (4 mg/kg chow) from gestation day 2 to 12 wk of age by feeding pregnant and nursing dams, and weaned pups. Some pups were also sensitized and challenged with ovalbumin (OVA). We assessed lung function and inflammatory cell and cytokine expression in the lung and examined bronchial goblet cell number. PFOA, but not PFOS, without the OVA sensitization/challenge induced AHR concomitant with a 25-fold increase of lung macrophages. PFOA exposure did not affect OVA-induced lung inflammatory cell number. In contrast, PFOS exposure inhibited OVA-induced lung inflammation, decreasing total cell number in lung lavage by 68.7%. Interferon-γ mRNA in the lung was elevated in all PFC-exposed groups. Despite these effects, neither PFOA nor PFOS affected OVA-induced AHR. Our data do not reveal PFOA or PFOS exposure as a risk factor for more severe allergic asthma-like symptoms, but PFOA alone can induce airway inflammation and alter airway function.


Assuntos
Poluentes Atmosféricos/toxicidade , Ácidos Alcanossulfônicos/toxicidade , Asma/imunologia , Caprilatos/toxicidade , Fluorocarbonos/toxicidade , Células Caliciformes/imunologia , Pulmão/imunologia , Exposição Materna/efeitos adversos , Animais , Asma/induzido quimicamente , Asma/patologia , Feminino , Células Caliciformes/patologia , Interferon gama/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Gravidez
2.
Cardiovasc Res ; 87(1): 102-10, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20097674

RESUMO

AIMS: The fibroblast growth factor (FGF) family plays an important role in cardiac growth and development. However, only FGF-16 RNA levels are reported to increase during the perinatal period and to be expressed preferentially in the myocardium, suggesting control at the transcriptional level and a role for FGF-16 in the postnatal heart. Beyond the identification of two TATA-like elements (TATA1 and TATA2) in the mouse FGF-16 promoter region and the preferential cardiac activity of TATA2, there is no report of Fgf-16 gene regulation. Assessment of promoter sequences, however, reveals putative nuclear factor-kappaB (NF-kappaB) elements, suggesting that Fgf-16 is regulated via NF-kappaB activation and thereby implicated in a number of cardiac events. Thus, the Fgf-16 gene was investigated as a target for NF-kappaB activation in cardiac cells. METHODS AND RESULTS: Assessments of Fgf-16 promoter activity were made using truncated and transfected hybrid genes with NF-kappaB inhibitors and/or beta-adrenergic stimulation via isoproterenol (IsP) treatment (a known NF-kappaB activator) in culture, and on endogenous mouse and human Fgf-16 genes in situ. The mouse Fgf-16 promoter region was stimulated in response to IsP treatment, but this response was lost with NF-kappaB inhibitor pretreatment. Deletion analysis revealed IsP responsiveness linked to sequences between TATA2 and TATA1 and, more specifically, a NF-kappaB element upstream and adjacent to TATA1 that associates with NF-kappaB p50/p65 subunits in chromatin. Finally, TATA1 and the proximal NF-kappaB element are conserved in the human genome and responsive to IsP. CONCLUSION: The mouse and human Fgf-16 gene is a target for NF-kappaB activation in the postnatal heart.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Fatores de Crescimento de Fibroblastos/metabolismo , Isoproterenol/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , NF-kappa B/metabolismo , Adulto , Idoso , Animais , Animais Recém-Nascidos , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Sequência Conservada , Feminino , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Miócitos Cardíacos/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Fatores de Tempo , Transfecção , Adulto Jovem
3.
Biochem Biophys Res Commun ; 373(2): 270-4, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18565327

RESUMO

Fibroblast growth factor 16 (FGF-16) expression has previously been detected in mouse heart at mid-gestation in the endocardium and epicardium, suggesting a role in embryonic heart development. More specifically, exogenously applied FGF-16 has been shown to stimulate growth of embryonic myocardial cells in tissue explants. We have generated mice lacking FGF-16 by targeting the Fgf16 locus on the X chromosome. Elimination of Fgf16 expression resulted in embryonic death as early as day 11.5 (E11.5). External abnormalities, including hemorrhage in the heart and ventral body region as well as facial defects, began to appear in null embryos from E11.5. Morphological analysis of FGF-16 null hearts revealed cardiac defects including chamber dilation, thinning of the atrial and ventricular walls, and poor trabeculation, which were visible at E10.5 and more pronounced at E11.5. These findings indicate FGF-16 is required for embryonic heart development in mid-gestation through its positive effect on myocardial growth.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Cardiopatias Congênitas/genética , Coração/embriologia , Animais , Anormalidades Craniofaciais/genética , Perda do Embrião/genética , Feminino , Marcação de Genes , Cardiopatias Congênitas/patologia , Camundongos , Camundongos Knockout , Gravidez , Segundo Trimestre da Gravidez , Cromossomo X/genética
4.
Am J Physiol Cell Physiol ; 294(5): C1242-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18337564

RESUMO

FGF-16 has been reported to be preferentially expressed in the adult rat heart. We have investigated the expression of FGF-16 in the perinatal and postnatal heart and its functional significance in neonatal rat cardiac myocytes. FGF-16 mRNA accumulation was observed by quantitative RT-PCR between neonatal days 1 and 7, with this increased expression persisting into adulthood. FGF-2 has been shown to increase neonatal rat cardiac myocyte proliferative potential via PKC activation. Gene array analysis revealed that FGF-16 inhibited the upregulation by FGF-2 of cell cycle promoting genes including cyclin F and Ki67. Furthermore, the CDK4/6 inhibitor gene Arf/INK4A was upregulated with the combination of FGF-16 and FGF-2 but not with either factor on its own. The effect on Ki67 was validated by protein immunodetection, which also showed that FGF-16 significantly decreased FGF-2-induced Ki67 labeling of cardiac myocytes, although it alone had no effect on Ki67 labeling. Inhibition of p38 MAPK potentiated cardiac myocyte proliferation induced by FGF-2 but did not alter the inhibitory action of FGF-16. Receptor binding assay showed that FGF-16 can compete with FGF-2 for binding sites including FGF receptor 1. FGF-16 had no effect on activated p38, ERK1/2, or JNK/SAPK after FGF-2 treatment. However, FGF-16 inhibited PKC-alpha and PKC-epsilon activation induced by FGF-2 and, importantly, IGF-1. Collectively, these data suggest that expression and release of FGF-16 in the neonatal myocardium interfere with cardiac myocyte proliferative potential by altering the local signaling environment via modulation of PKC activation and cell cycle-related gene expression.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Técnicas de Cultura de Células , Ciclo Celular/fisiologia , Divisão Celular , Ativação Enzimática , Fatores de Crescimento de Fibroblastos/genética , Gliceraldeído-3-Fosfato Desidrogenases/genética , Cinética , Proteína Quinase C/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
5.
Neuroendocrinology ; 83(3-4): 145-53, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17047377

RESUMO

The somatic cells of a multicellular organism contain an identical complement of genes that need to be expressed specifically and appropriately to allow the normal development and functions associated with an organism. In the eukaryotic cell nucleus, genes are packaged with nucleoprotein histones into chromatin. The human growth hormone (GH)/chorionic somatomammotropin (CS) gene family offers an excellent model to study the relationship between chromatin structure and transcription factor binding in terms of tissue-specific gene expression. The GH/CS gene family consists of five genes (GH-N, GH-V, CS-A, CS-B and CS-L), contained in a single locus on chromosome 17. Although they share approximately 94% sequence similarity, GH-N expression is restricted to pituitary somatotropes while the four placental GH/CS genes are expressed in the villus syncytiotrophoblast. Appropriate expression in vivo is dependent on remote sequences found 14-32 kb upstream of GH-N in the loci of adjacent genes, and these sequences are characterized by five (I-V) nuclease-hypersensitive sites (HS). Pituitary-specific factor Pit-1 binds at HS I/II and plays an essential role in chromatin remodeling and GH-N expression; however, the processes that lead to HS I/II accessibility are unknown. We discuss the possibility that Pit-1-driven remodeling at HS III may precede that at HS I/II in the pituitary. Also, in pituitary chromatin, all five GH/CS genes share similar nuclease sensitivity, suggesting that the conformation of the placental genes is not inhibitory to transcription. Given that the promoters of both GH-N and the placental GH/CS genes contain Pit-1-binding sites, possible mechanisms to restrict placenta GH/CS promoter activity in the pituitary are discussed, including active repression via P sequences located upstream of each of the placental GH/CS genes. Positively or negatively influencing those components known to be important for pituitary transcription may link epigenetic events to key transcription factors in the overall picture of tissue-specific control of gene expression.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , Regulação da Expressão Gênica/fisiologia , Hormônio do Crescimento Humano/metabolismo , Hipófise/metabolismo , Fator de Transcrição Pit-1/metabolismo , Montagem e Desmontagem da Cromatina/genética , Cromossomos Humanos Par 17/genética , Cromossomos Humanos Par 17/metabolismo , Hormônio do Crescimento Humano/genética , Humanos , Especificidade de Órgãos/genética , Hipófise/citologia
6.
Mol Endocrinol ; 20(3): 598-607, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16239259

RESUMO

The human GH family consists of five genes, including the placental chorionic somatomammotropins (CS), within a single locus on chromosome 17. Based on nuclease sensitivity, the entire GH/CS locus is accessible in pituitary chromatin, yet only GH-N is expressed. Previously, we reported a P sequence element (263P) capable of repressing placental CS-A promoter activity in transfected pituitary (GC) cells, and our data indicated a possible role for nuclear factor-1 (NF-1) and regulatory factor X1 in this repression. In this study we show the formation of two independent pituitary complexes in vitro: a repressor complex containing NF-1 and a nonfunctional complex containing regulatory factor X1. In vitro repressor function is stabilized by the presence of P sequence element C (PSE-C), downstream of the previously characterized PSE-A and PSE-B. Repressor function is also dependent on an intact Pit-1 binding site in the CS-A promoter. EMSAs with PSE-C reveal binding of the hepatocyte nuclear factor-3/forkhead (HNF-3/fkh) family of transcription factors in rat pituitary GC cells. This observation is extended to human pituitary tissue, where HNF-3alpha's association with P sequences is confirmed by chromatin immunoprecipitation. Furthermore, protein-protein interactions between HNF-3alpha and NF-1 family members are demonstrated. These results identify HNF-3alpha as an additional member of the pituitary P sequence regulatory complex, implicating it in tissue-specific expression of the human GH/CS family.


Assuntos
Fator 3-alfa Nuclear de Hepatócito/metabolismo , Hormônio do Crescimento Humano/metabolismo , Hipófise/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fator 3-alfa Nuclear de Hepatócito/genética , Hormônio do Crescimento Humano/genética , Humanos , Dados de Sequência Molecular , Complexos Multiproteicos , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo , Hipófise/citologia , Lactogênio Placentário/genética , Lactogênio Placentário/metabolismo , Regiões Promotoras Genéticas , Ratos , Fatores de Transcrição de Fator Regulador X , Sequências Reguladoras de Ácido Nucleico , Fator de Transcrição Pit-1/genética , Fator de Transcrição Pit-1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Cardiovasc Res ; 62(3): 548-57, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15158147

RESUMO

OBJECTIVE: Fibroblast growth factor-2 (FGF-2) exerts its cardioprotective effect through cell surface receptor signaling and may play a role in the normal maintenance of a healthy myocardium. One mechanism of FGF-2 release from contracting cardiomyocytes is through transient sarcolemmal disruption, with accumulation in the extracellular matrix. Continuous FGF-2 release would require a link to synthesis and, thus, we examined regulation of FGF-2 promoter activity in cardiomyocytes as a potential target for optimizing cardioprotection. METHODS AND RESULTS: To investigate autoregulation, neonatal rat cardiomyocytes, (NRCM), were transfected with approximately 1 or 0.1 kb of rat FGF-2 promoter sequences linked to luciferase, -1058FGF-2p.luc and -110FGF-2p.luc, and treated with or without FGF-2. FGF-2 promoter activity was significantly increased approximately 2.5-fold with both genes. The proximal promoter region of rat FGF-2 contains putative binding sites for the early growth response-1 (Egr-1) and stimulating protein 1 (Sp1) transcription factors. Overexpression of Egr-1 and Sp1 increased -1058FGF-2p.luc expression by 4.4- and 8.7-fold, respectively. Mutation of Egr-1 and overlapping Sp1 sites did not blunt the response of -110FGF-2p.luc to FGF-2 treatment but did significantly reduce basal promoter activity. Transgenic mice expressing -1058FGF-2p.luc were treated with isoproterenol (IsP) to increase heart rate and endogenous FGF-2 release. FGF-2 promoter activity was stimulated significantly at 6 h, and increases in both FGF-2 and its receptor mRNA levels were also detected. In contrast, no effect of IsP was seen on -1058FGF-2p.luc or -110FGF-2p.luc in transfected NRCMs. CONCLUSIONS: FGF-2 released from cardiomyocytes may act to regulate its own synthesis at the transcriptional level. The mechanism does not appear to require an intact Egr-1 site in the proximal promoter region. This may, however, reflect redundancy in the control of FGF-2 promoter activity as our data support a stimulatory role for Egr-1 and Sp1.


Assuntos
Fator 2 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Miócitos Cardíacos/metabolismo , Transcrição Gênica , Agonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Isoproterenol/farmacologia , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Regiões Promotoras Genéticas , Ratos , Receptores Proteína Tirosina Quinases/análise , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/análise
8.
Can J Physiol Pharmacol ; 82(12): 1044-52, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15644945

RESUMO

In the field of cardiovascular research, a number of independent approaches have been explored to protect the heart from acute and chronic ischemic damage. Fibroblast growth factor-2 (FGF-2) recently has received considerable attention with respect to its angiogenic potential. While therapeutic angiogenesis may serve to salvage chronically ischemic myocardium, more acute treatments are in demand to increase cardiac resistance to injury (preconditioning) and to guard against secondary injury after an acute ischemic insult. Here, we look beyond the angiogenic potential of FGF-2 and examine its acute cardioprotective activity as demonstrated under experimental conditions, both as an agent of a preconditioning-like response and for secondary injury prevention at the time of reperfusion. Factors to consider in moving to the clinical setting will be discussed, including issues of dosage, treatment duration, and routes of administration. Finally, issues of safety and clinical trial design will be considered. The prospect of such a multipotent growth factor having clinical usefulness opens the door to effective treatment of both acute and chronic ischemic heart disease, something well worth the attention of the cardiovascular community.


Assuntos
Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Cardiopatias/prevenção & controle , Neovascularização Patológica/prevenção & controle , Animais , Humanos , Precondicionamento Isquêmico Miocárdico
9.
Cardiovasc Res ; 57(1): 8-19, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12504809

RESUMO

Fibroblast growth factor-2 (FGF-2) is a potent regulator of many cellular functions and phenomena, including cell proliferation, differentiation, survival, adhesion, migration, motility and apoptosis, and processes such as limb formation, wound healing, tumorigenesis, angiogenesis, vasculogenesis and blood vessel remodeling. In the adult myocardium, FGF-2 is expressed by various cell types, including cardiomyocytes, fibroblasts and smooth muscle cells. The biological effects of FGF-2 in the myocardium are mediated by the high-affinity tyrosine kinase receptor FGFR-1, the major FGF receptor in the heart. Here, we give an overview of current insights into the multiple roles of FGF-2 in the myocardium, as they pertain to two basic phenomena: ischemia-reperfusion injury and cardiac hypertrophy. The first category includes roles for FGF-2 in cardioprotection, the inflammatory response, angiogenesis and vascular remodeling, while the second includes myocyte hypertrophy, fibrosis, and gap junction functioning (conduction). Given the strong evidence for FGF-2 as both a cardioprotective and angiogenic agent, the therapeutic potential of FGF-2 in the ischemic myocardium is discussed.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Miocárdio/metabolismo , Neovascularização Fisiológica/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia , Adulto , Animais , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Proteínas Contráteis/metabolismo , Fibroblastos/patologia , Humanos , Células Musculares/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA