Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Dyn ; 246(11): 779-801, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28481428

RESUMO

BACKGROUND: Austrofundulus limnaeus is an annual killifish from the Maracaibo basin of Venezuela. Annual killifishes are unique among vertebrates in their ability to enter into a state of dormancy at up to three distinct developmental stages termed diapause I, II, and III. These embryos are tolerant of a wide variety of environmental stresses and develop relatively slowly compared with nonannual fishes. RESULTS: These traits make them an excellent model for research on interactions between the genome and the environment during development, and an excellent choice for developmental biology laboratories. Furthermore, A. limnaeus is relatively easy to maintain in a laboratory setting and has a high fecundity, making it an excellent candidate as an emerging model for studies of development, and for defining the limits of developmental buffering in vertebrates. CONCLUSIONS: This study reports for the first time on the detailed development of A. limnaeus and provides a photographic and illustrated atlas of embryos on the two developmental trajectories possible in this species. Developmental Dynamics 246:779-801, 2017. © 2017 The Authors Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Assuntos
Biologia do Desenvolvimento/métodos , Fundulidae/embriologia , Interação Gene-Ambiente , Animais , Embrião não Mamífero , Fundulidae/crescimento & desenvolvimento , Peixes Listrados/embriologia , Peixes Listrados/crescimento & desenvolvimento , Modelos Animais
2.
Oncogene ; 35(5): 577-586, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25893305

RESUMO

Non-small-cell lung cancer (NSCLC) is a common malignancy with a poor prognosis. Despite progress targeting oncogenic drivers, there are no therapies targeting tumor-suppressor loss. Smad4 is an established tumor suppressor in pancreatic and colon cancer; however, the consequences of Smad4 loss in lung cancer are largely unknown. We evaluated Smad4 expression in human NSCLC samples and examined Smad4 alterations in large NSCLC data sets and found that reduced Smad4 expression is common in human NSCLC and occurs through a variety of mechanisms, including mutation, homozygous deletion and heterozygous loss. We modeled Smad4 loss in lung cancer by deleting Smad4 in airway epithelial cells and found that Smad4 deletion both initiates and promotes lung tumor development. Interestingly, both Smad4(-/-) mouse tumors and human NSCLC samples with reduced Smad4 expression demonstrated increased DNA damage, whereas Smad4 knockdown in lung cancer cells reduced DNA repair and increased apoptosis after DNA damage. In addition, Smad4-deficient NSCLC cells demonstrated increased sensitivity to both chemotherapeutics that inhibit DNA topoisomerase and drugs that block double-strand DNA break repair by non-homologous end joining. In sum, these studies establish Smad4 as a lung tumor suppressor and suggest that the defective DNA repair phenotype of Smad4-deficient tumors can be exploited by specific therapeutic strategies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Proteína Smad4/deficiência , Inibidores da Topoisomerase/farmacologia , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Reparo do DNA , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Proteína Smad4/genética , Proteína Smad4/metabolismo
3.
Mol Carcinog ; 53(10): 841-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23625632

RESUMO

Lung adenocarcinoma (AdC) and lung squamous cell carcinoma (SCC) are the most common non-small cell lung cancer (NSCLC) subtypes, however, most genetic mouse models of lung cancer produce predominantly, if not exclusively, AdC. Whether this is secondary to targeting mutations to the distal airway cells or to the use of activating Kras mutations that drive AdC formation is unknown. We previously showed that targeting Kras(G12D) activation and transforming growth factor ß receptor type II (TGFßRII) deletion to airway basal cells via a keratin promoter induced formation of both lung AdC and SCC. In this study we assessed if targeting phosphatase and tensin homologue (PTEN) deletion to airway basal cells could initiate lung tumor formation or increase lung SCC formation. We found that PTEN deletion is capable of initiating both lung AdC and SCC formation when targeted to basal cells and although PTEN deletion is a weaker tumor initiator than Kras(G12D) with low tumor multiplicity and long latency, tumors initiated by PTEN deletion were larger and displayed more malignant conversion than Kras(G12D) initiated tumors. That PTEN deletion did not increase lung SCC formation compared to Kras(G12D) activation, suggests that the initiating genetic event does not dictate tumor histology when genetic alterations are targeted to a specific cell. These studies also confirm that basal cells of the conducting airway are capable of giving rise to multiple NSCLC tumor types.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasia de Células Basais/metabolismo , PTEN Fosfo-Hidrolase/genética , Animais , Carcinoma de Células Escamosas/genética , Deleção de Genes , Humanos , Neoplasias Pulmonares/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação de Sentido Incorreto , Neoplasia de Células Basais/genética , PTEN Fosfo-Hidrolase/deficiência , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
4.
Clin Cancer Res ; 18(8): 2173-83, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22399565

RESUMO

PURPOSE: Lung adenocarcinoma and lung squamous cell carcinoma (SCC) are the most common non-small cell lung cancer (NSCLC) subtypes. This study was designed to determine whether reduced expression of TGFß type II receptor (TGFßRII) promotes lung adenocarcinoma and SCC carcinogenesis. EXPERIMENTAL DESIGN: We examined TGFßRII expression at the protein and mRNA levels in human NSCLC samples and assessed the relationship between TGFßRII expression and clinicopathologic parameters. To determine whether sporadic TGFßRII deletion in airway epithelial cells induces NSCLC formation, we targeted TGFßRII deletion alone and in combination with oncogenic Kras(G12D) to murine airways using a keratin 5 (K5) promoter and inducible Cre recombinase. RESULTS: Reduced TGFßRII expression in human NSCLC is associated with male gender, smoking, SCC histology, reduced differentiation, increased tumor stage, increased nodal metastasis, and reduced survival. Homozygous or heterozygous TGFßRII deletion in mouse airway epithelia increases the size and number of Kras(G12D)-initiated adenocarcinoma and SCC. TGFßRII deletion increases proliferation, local inflammation, and TGFß ligand elaboration; TGFßRII knockdown in airway epithelial cells increases migration and invasion. CONCLUSIONS: Reduced TGFßRII expression in human NSCLC is associated with more aggressive tumor behavior and inflammation that is, at least partially, mediated by increased TGFß1 expression. TGFßRII deletion in mouse airway epithelial cells promotes adenocarcinoma and SCC formation, indicating that TGFßRII loss plays a causal role in lung carcinogenesis. That TGFßRII shows haploid insufficiency suggests that a 50% TGFßRII protein reduction would negatively impact lung cancer prognosis.


Assuntos
Adenocarcinoma/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/patologia , Neoplasias Pulmonares/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Adenocarcinoma de Pulmão , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/mortalidade , Linhagem Celular Tumoral , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Masculino , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética
5.
Cancer Prev Res (Phila) ; 5(2): 283-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22086679

RESUMO

Squamous cell carcinoma (SCC) and premalignant endobronchial lesions have been difficult to study in murine models. In this study, we evaluate the topical N-nitroso-tris-chloroethylurea (NTCU) murine SCC model, determine the extent to which resulting premalignant airway dysplasia develops, discuss clinicopathologic grading criteria in lesion progression, and confirm that immunohistochemical (IHC) staining patterns are consistent with those observed in human endobronchial dysplasia and SCC. Male and female FVB mice were treated biweekly with topical NTCU (4, 8, or 40 mmol/L) or vehicle for 32 weeks. Following sacrifice, squamous cell lesions were enumerated and categorized into the following groups: flat atypia, low-grade dysplasia, high-grade dysplasia, and invasive SCC. The 40 mmol/L NTCU concentration produced the entire spectrum of premalignant dysplasias and squamous cell carcinomas, but was associated with poor survival. Concentrations of 4 and 8 mmol/L NTCU were better tolerated and produced only significant levels of flat atypia. Squamous origin of the range of observed lesions was confirmed with IHC staining for cytokeratin 5/6, p63, thyroid transcription factor-1 (TTF-1), and Napsin-A. This study shows that topical application of high-dose NTCU produces endobronchial premalignant lesions with classic squamous characteristics and should allow for improved preclinical evaluation of potential chemopreventive agents.


Assuntos
Neoplasias Brônquicas/induzido quimicamente , Carcinógenos/toxicidade , Carcinoma de Células Escamosas/induzido quimicamente , Carmustina/análogos & derivados , Modelos Animais de Doenças , Lesões Pré-Cancerosas/induzido quimicamente , Administração Tópica , Animais , Neoplasias Brônquicas/mortalidade , Neoplasias Brônquicas/patologia , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Carmustina/toxicidade , Feminino , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Lesões Pré-Cancerosas/mortalidade , Lesões Pré-Cancerosas/patologia , Taxa de Sobrevida
6.
Stem Cells ; 29(4): 725-35, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21312316

RESUMO

Tissue-resident mesenchymal stem cells (MSCs) are important regulators of tissue repair or regeneration, fibrosis, inflammation, angiogenesis, and tumor formation. Here, we define a population of resident lung MSCs (luMSCs) that function to regulate the severity of bleomycin injury via modulation of the T-cell response. Bleomycin-induced loss of these endogenous luMSCs and elicited fibrosis (pulmonary fibrosis), inflammation, and pulmonary arterial hypertension (PAH). Replacement of resident stem cells by administration of isolated luMSCs attenuated the bleomycin-associated pathology and mitigated the development of PAH. In addition, luMSC modulated a decrease in numbers of lymphocytes and granulocytes in bronchoalveolar fluid and demonstrated an inhibition of effector T-cell proliferation in vitro. Global gene expression analysis indicated that the luMSCs are a unique stromal population differing from lung fibroblasts in terms of proinflammatory mediators and profibrotic pathways. Our results demonstrate that luMSCs function to protect lung integrity after injury; however, when endogenous MSCs are lost, this function is compromised illustrating the importance of this novel population during lung injury. The definition of this population in vivo in both murine and human pulmonary tissue facilitates the development of a therapeutic strategy directed at the rescue of endogenous cells to facilitate lung repair during injury.


Assuntos
Bleomicina/efeitos adversos , Proliferação de Células , Células-Tronco Mesenquimais/efeitos dos fármacos , Fibrose Pulmonar/induzido quimicamente , Linfócitos T/citologia , Animais , Bleomicina/farmacologia , Hipertensão Pulmonar Primária Familiar , Hipertensão Pulmonar/patologia , Lesão Pulmonar , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Fibrose Pulmonar/patologia
7.
Int J Biol Sci ; 6(1): 68-79, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20140084

RESUMO

Systems capable of targeting genetic manipulations to keratin-positive airway basal cells are more poorly developed than systems targeting other airway epithelial cell populations and this has likely hindered development of animal models of diseases such as lung squamous cell carcinoma. Although keratin promoter driven-Cre recombinase constructs are potentially useful for targeting these cells, these constructs have substantially higher activity in the skin and oral epithelium than in the airways. We developed a method for delivering RU486, the conditional activator of Cre recombinase progesterone receptor (CrePR) fusion proteins to the lung and then examined the activity of three keratin-driven CrePR constructs in the conducting airways. We also developed a technique for survival bronchioalveolar lavage on non-ventilated animals to examine the effects of the acetone/oil vehicle required to deliver RU486 to the lung. K5CrePR1 and K14CrePR1 constructs differ only in the keratin promoter used to target CrePR1 expression while K5Cre*PR contains a truncated progesterone receptor designed to reduce RU486-independent Cre activity. While all three constructs demonstrate RU486-inducible Cre activity in the conducting airways, both construct activity and tightness of regulation vary considerably. K5Cre*PR is the most tightly regulated Cre driver making it ideal for targeting somatic mutations to the airway epithelia while K5CrePR1 and K14CrePR1 may be better suited to studying diseases of the conducting airways where gene targeting of keratin expressing cells and their derivatives is desired.


Assuntos
Técnicas Genéticas , Queratinas/genética , Regiões Promotoras Genéticas , Mucosa Respiratória/metabolismo , Acetona , Animais , Lavagem Broncoalveolar/métodos , Hormônios/administração & dosagem , Hormônios/farmacologia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Integrases/genética , Integrases/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mifepristona/administração & dosagem , Mifepristona/farmacologia , Óleos , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Traqueia/efeitos dos fármacos , Traqueia/metabolismo
8.
J Clin Invest ; 119(11): 3408-19, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19841536

RESUMO

Smad4 is a central mediator of TGF-beta signaling, and its expression is downregulated or lost at the malignant stage in several cancer types. In this study, we found that Smad4 was frequently downregulated not only in human head and neck squamous cell carcinoma (HNSCC) malignant lesions, but also in grossly normal adjacent buccal mucosa. To gain insight into the importance of this observation, we generated mice in which Smad4 was deleted in head and neck epithelia (referred to herein as HN-Smad4-/- mice) and found that they developed spontaneous HNSCC. Interestingly, both normal head and neck tissue and HNSCC from HN-Smad4-/- mice exhibited increased genomic instability, which correlated with downregulated expression and function of genes encoding proteins in the Fanconi anemia/Brca (Fanc/Brca) DNA repair pathway linked to HNSCC susceptibility in humans. Consistent with this, further analysis revealed a correlation between downregulation of Smad4 protein and downregulation of the Brca1 and Rad51 proteins in human HNSCC. In addition to the above changes in tumor epithelia, both normal head and neck tissue and HNSCC from HN-Smad4-/- mice exhibited severe inflammation, which was associated with increased expression of TGF-beta1 and activated Smad3. We present what we believe to be the first single gene-knockout model for HNSCC, in which both HNSCC formation and invasion occurred as a result of Smad4 deletion. Our results reveal an intriguing connection between Smad4 and the Fanc/Brca pathway and highlight the impact of epithelial Smad4 loss on inflammation.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/fisiopatologia , Instabilidade Genômica/genética , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/fisiopatologia , Proteína Smad4/genética , Proteína Smad4/metabolismo , Animais , Carcinoma de Células Escamosas/patologia , Reparo do DNA/genética , Regulação para Baixo , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Deleção de Genes , Genes BRCA1 , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Inflamação/genética , Inflamação/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rad51 Recombinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...