Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Wound Repair Regen ; 21(1): 103-12, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23126606

RESUMO

While cutaneous wounds of late-gestational fetuses and on through adulthood result in scar formation, wounds incurred early in gestation have been shown to heal scarlessly. Unique properties of fetal fibroblasts are believed to mediate this scarless healing process. In this study, microarray analysis was used to identify differences in the gene expression profiles of cultured fibroblasts from embryonic day 15 (E15; midgestation) and embryonic day 18 (E18; late-gestation) skin. Sixty-two genes were differentially expressed and 12 of those genes are associated with inflammation, a process that correlates with scar formation in fetal wounds. One of the differentially expressed inflammatory genes was cyclooxygenase-1 (COX-1). COX-1 was more highly expressed in E18 fibroblasts than in E15 fibroblasts, and these differences were confirmed at the gene and protein level. Differences in COX-1 protein expression were also observed in fetal skin by immunohistochemical and immunofluorescence staining. The baseline differences in gene expression found in mid- and late-gestational fetal fibroblasts suggest that developmental alterations in fibroblasts could be involved in the transition from scarless to fibrotic fetal wound healing. Furthermore, baseline differences in the expression of inflammatory genes by fibroblasts in E15 and E18 skin may contribute to inflammation and scar formation late in gestation.


Assuntos
Cicatriz/patologia , Feto/metabolismo , Fibroblastos/metabolismo , Idade Gestacional , Pele/patologia , Cicatrização , Análise de Variância , Animais , Células Cultivadas , Cicatriz/embriologia , Ciclo-Oxigenase 1/metabolismo , Feminino , Feto/citologia , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Inflamação , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Substâncias Luminescentes , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Gravidez , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Pele/embriologia , Pele/lesões , Fator de Crescimento Transformador beta/metabolismo
2.
Resuscitation ; 84(1): 114-20, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22982155

RESUMO

BACKGROUND: The effect of hyperoxygenation at reperfusion, particularly in the setting of cardiac arrest, remains unclear. This issue was studied in a prolonged cardiac arrest model consisting of 25 min cardiac arrest in a rat resuscitated with cardiopulmonary bypass (CPB). The objective of this study was to determine the effect of hyperoxygenation following prolonged cardiac arrest resuscitation on mitochondrial and cardiac function. METHODS: Male Sprague-Dawley rats (400-450 g) were anesthetized with ketamine and xylazine and instrumented for closed chest cardiopulmonary bypass (CPB). Following a 25-min KCl-induced cardiac arrest, the animals were resuscitated by CPB with 100% oxygen. Three minutes after successful return of spontaneous circulation (ROSC), the animals received either normoxemic reperfusion (CPB with 40-50% oxygen) or hyperoxemic reperfusion (CPB with 100% oxygen) for 1 h. Post-resuscitation hemodynamics, cardiac function, mitochondrial function and immunostaining of 3-nitrotyrosine were compared between the two different treatment groups. RESULTS: At 1 h after ROSC, the hyperoxemic reperfusion group had a significant higher mean arterial pressure, less metabolic acidosis and better diastolic function than the normoxemic reperfusion group. Cardiac mitochondria from the hyperoxemic reperfusion group had a higher respiratory control ratio (RCR) and cardiac tissue showed less nitroxidative stress compared to the normoxemic reperfusion group. CONCLUSIONS: One hour of hyperoxemic reperfusion after 25 min of cardiac arrest in an in vivo CPB model resulted in significant short-term improvement in myocardial and mitochondrial function compared with 1h of normoxemic reperfusion. This myocardial response may differ from previously reported post-arrest hyperoxia mediated effects following shorter arrest times.


Assuntos
Ponte Cardiopulmonar/métodos , Parada Cardíaca/terapia , Hiperóxia , Oxigenoterapia/métodos , Análise de Variância , Animais , Gasometria , Parada Cardíaca/fisiopatologia , Hemodinâmica , Imuno-Histoquímica , Masculino , Mitocôndrias Cardíacas/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Tirosina/análogos & derivados , Tirosina/análise
3.
J Invest Dermatol ; 132(2): 458-65, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21993557

RESUMO

Scar formation is a potentially detrimental process of tissue restoration in adults, affecting organ form and function. During fetal development, cutaneous wounds heal without inflammation or scarring at early stages of development; however, they begin to heal with significant inflammation and scarring as the skin becomes more mature. One possible cell type that could regulate the change from scarless to fibrotic healing is the mast cell. We show here that dermal mast cells in scarless wounds generated at embryonic day 15 (E15) are fewer in number, less mature, and do not degranulate in response to wounding as effectively as mast cells of fibrotic wounds made at embryonic day 18 (E18). Differences were also observed between cultured mast cells from E15 and E18 skin, with regard to degranulation and preformed cytokine levels. Injection of mast cell lysates into E15 wounds disrupted scarless healing, suggesting that mast cells interfere with scarless repair. Finally, wounds produced at E18, which normally heal with a scar, healed with significantly smaller scars in mast cell-deficient Kit(W/W-v) mice compared with Kit(+/+) littermates. Together, these data suggest that mast cells enhance scar formation, and that these cells may mediate the transition from scarless to fibrotic healing during fetal development.


Assuntos
Cicatriz/etiologia , Feto/fisiologia , Mastócitos/fisiologia , Cicatrização , Fatores Etários , Animais , Células Cultivadas , Feminino , Liberação de Histamina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
5.
J Dermatol Sci ; 56(1): 19-26, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19643582

RESUMO

BACKGROUND: Keratinocytes at wound margins undergo partial epithelial to mesenchymal transition (EMT). Based on previous in vitro and ex vivo findings, Slug (Snai2), a transcriptional regulator of EMT in development, may play an important role in this process. OBJECTIVES: This study was designed to validate an in vivo role for Slug in wound healing. METHODS: Excisional wounds in Slug null and wild type mice were examined histologically at 6, 24, 48, and 72h after wounding; reepithelialization was measured and immunohistochemistry for keratins 8, 10, 14, and 6 and E-cadherin was performed. In 20 Slug null and 20 wild type mice exposed three times weekly to two minimal erythemal doses of UVR, the development of non-healing cutaneous ulcers was documented. Ulcers were examined histologically and by immunohistochemistry. RESULTS: The reepithelialization component of excisional wound healing was reduced 1.7-fold and expression of the Slug target genes keratin 8 and E-cadherin was increased at wound margins in Slug null compared to wild type mice. In contrast, no differences in expression of keratins 10 or 14 or in markers of proliferation K6 and Ki-67 were observed. Forty per cent of Slug null mice but no wild type mice developed non-healing cutaneous ulcers in response to chronic UVR. Keratinocytes at ulcer margins expressed high levels of keratin 8 and retained E-cadherin expression, thus resembling excisional wounds. CONCLUSION: Slug is an important modulator of successful wound repair in adult tissue and may be critical for maintaining epidermal integrity in response to chronic injury.


Assuntos
Caderinas/metabolismo , Queratinócitos/fisiologia , Queratinas/metabolismo , Pele/metabolismo , Fatores de Transcrição/metabolismo , Cicatrização/genética , Animais , Doença Crônica , Feminino , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Lesões Experimentais por Radiação/metabolismo , Pele/patologia , Úlcera Cutânea/metabolismo , Úlcera Cutânea/patologia , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética
6.
Lab Invest ; 88(8): 831-41, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18458671

RESUMO

We previously reported ultraviolet radiation (UVR) induction of Slug, a Snail family zinc-finger transcription factor, in the epidermis of mice; we now report that Slug-knockout mice are, unexpectedly, more resistant to sunburn than wild-type mice. There was a marked difference between the cutaneous inflammatory response in the skin of Slug-knockout and wild-type mice from 12 h to 1 week following a single exposure to 3 minimal erythemal doses of UVR. Slug-knockout mice showed a much reduced immediate increase in skin thickness and neutrophil infiltration compared to wild-type mice. However, there were as many or more intraepidermal T cells, dermal mast cells, and dermal blood vessels in the UVR-exposed skin of Slug-knockout mice as in the skin of wild-type mice. Differences in cytokine and chemokine expression following UVR appeared to account for at least some differences between the genotypes in cutaneous inflammatory response. Despite the reported antiapoptotic and antiproliferative role for Slug in some cell types, we observed little difference between the genotypes in UVR-induced keratinocyte apoptosis or proliferation. Our findings indicate an unexpected but important role for Slug in the acute cutaneous inflammatory response to UVR.


Assuntos
Fatores Imunológicos/metabolismo , Lesões Experimentais por Radiação/metabolismo , Pele/efeitos da radiação , Fatores de Transcrição/metabolismo , Raios Ultravioleta , Animais , Apoptose/efeitos da radiação , Dano ao DNA , Dermatite/imunologia , Dermatite/metabolismo , Dermatite/patologia , Feminino , Expressão Gênica , Fatores Imunológicos/genética , Masculino , Camundongos , Camundongos Knockout , Lesões Experimentais por Radiação/imunologia , Lesões Experimentais por Radiação/patologia , Pele/imunologia , Pele/metabolismo , Pele/patologia , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética
7.
Am J Pathol ; 171(5): 1629-39, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17916597

RESUMO

Snai2, encoded by the SNAI2 gene, has been shown to modulate epithelial-mesenchymal transformation (EMT), the conversion of sessile epithelial cells attached to adjacent cells and to the basement membrane into dissociated and motile fibroblastic cells. EMT occurs during development, wound healing, and carcinoma progression. Using Snai2-null mice (Snai2(lacZ)), we evaluated the role of Snai2 in UV radiation (UVR)-induced skin carcinogenesis. In chronically UVR-exposed nontumor-bearing skin from Snai2-null mice, inflammation and epidermal proliferation were decreased compared with wild-type (+/+) skin. Snai2-null mice had a consistently lower tumor burden than +/+ mice. In addition, null mice developed fewer aggressive spindle cell tumors, believed to arise from squamous cell carcinomas that have undergone EMT, than +/+ mice; however, the difference in tumor type distribution between the two genotypes was not statistically significant. No metastases were observed in either the +/+ or Snai2-null mice. Using quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry, we showed that the spindle cell tumors in the Snai2-null mice demonstrated impaired EMT, as shown by decreased vimentin and increased cadherin 1 expression. This study confirms a role for Snai2 in EMT, but demonstrates that Snai2 expression is not required for the development or progression of UVR-induced skin tumors.


Assuntos
Transformação Celular Neoplásica , Neoplasias Induzidas por Radiação/patologia , Neoplasias Cutâneas/patologia , Fatores de Transcrição/biossíntese , Raios Ultravioleta/efeitos adversos , Animais , Caderinas/metabolismo , Diferenciação Celular/fisiologia , Epiderme/metabolismo , Epiderme/patologia , Inflamação/patologia , Camundongos , Camundongos Knockout , Neoplasias Induzidas por Radiação/imunologia , Neoplasias Induzidas por Radiação/metabolismo , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética
8.
Toxicol Pathol ; 35(6): 819-26, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17943656

RESUMO

Ultraviolet radiation (UVR) is a risk factor for the development of ocular disease in humans, including acute photokeratitis, chronic corneal spheroidal degeneration, and cataract formation. This report describes the ocular lesions seen in 21 mice chronically exposed to UVR as part of a skin carcinogenicity study. All globes were affected to varying degrees. The primary lesion, not previously reported in UVR-exposed mice, was marked loss of keratocytes relative to age-matched controls. Secondary lesions included corneal stromal thinning, keratoconus, corneal vascularization and fibrosis, keratitis, globe rupture, and phthisis bulbi. In addition, more than 90% of UVR-exposed and unexposed lenses had evidence of cataract formation; this is the first report of the occurrence of spontaneous cataracts in 129 mice. In a subsequent study, apoptotic cells were identified histologically and by cleaved caspase 3 immunoreactivity in the corneal epithelium and, less commonly, in the corneal stroma after acute UVR exposure. Based on this finding, we propose that the loss of keratocytes observed in the chronic study was due to UVR-induced apoptosis.


Assuntos
Córnea/efeitos da radiação , Raios Ultravioleta , Aldeído Desidrogenase/fisiologia , Animais , Catarata/etiologia , Córnea/patologia , Feminino , Masculino , Metaloproteinases da Matriz/fisiologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo
9.
J Histochem Cytochem ; 52(7): 959-65, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15208362

RESUMO

The Slug transcription factor plays an important role in epithelial-mesenchymal transformation during embryogenesis and is expressed in adult tissues during carcinogenesis. By detecting expression of a Slug-beta-galactosidase fusion protein, we have now demonstrated that Slug is also re-expressed in a variety of normal tissues in the adult mouse. Slug is expressed at relatively high levels in patchy stretches of basal cells in stratified and pseudostratified epithelium, including skin, oral mucosa, esophagus, stomach, rectum, cervix, and trachea. Slug is also found at variable levels in fibroblasts and stromal smooth muscle cells in many tissues. Sites of more intense Slug expression in mesenchymal tissues include cartilage, kidney glomeruli, lung, ovary, and uterus. Therefore, Slug expression is not restricted to the period of embryonic development or to pathological processes. The pattern of localization to basal cells in various epithelia suggests that Slug may play a role in the cell migration that occurs during continual renewal of these tissues.


Assuntos
Fatores de Transcrição/biossíntese , Animais , Epitélio/metabolismo , Imuno-Histoquímica , Camundongos , Especificidade de Órgãos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Dedos de Zinco , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...