Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
STAR Protoc ; 2(1): 100230, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33364620

RESUMO

We developed an adeno-associated virus (AAV) vector-based technique to label mouse neostriatal neurons comprising direct and indirect pathways with different fluorescent proteins and analyze their axonal projections. The AAV vector expresses GFP or RFP in the presence or absence of Cre recombinase and should be useful for labeling two cell populations exclusively dependent on its expression. Here, we describe the AAV vector design, stereotaxic injection of the AAV vector, and a highly sensitive immunoperoxidase method for axon visualization. For complete details on the use and execution of this protocol, please refer to Okamoto et al. (2020).


Assuntos
Dependovirus , Vetores Genéticos , Neostriado/metabolismo , Vias Neurais/metabolismo , Neurônios/metabolismo , Transdução Genética , Animais , Integrases/biossíntese , Integrases/genética , Camundongos , Neostriado/citologia , Vias Neurais/citologia , Neurônios/citologia
2.
Exp Eye Res ; 202: 108370, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33264655

RESUMO

Cre-mediated modulation of gene function in the murine retinal pigment epithelium (RPE) has been widely used, but current postnatal RPE-selective Cre driver lines suffer from limited recombination efficiency and/or ectopic or mosaic expression. We sought to generate a transgenic mouse line with consistently efficient RPE-selective Cre activity that could be temporally regulated. We used ϕC31 integrase to insert a DNA construct encoding a human BEST1 promoter fragment driving a Cre recombinase estrogen receptor fusion (BEST1-CreERT2) at the Rosa26 locus of C57BL/6J mice. Rosa26BEST1-CreERT2 mice were bred with a tdTomato reporter line and to mice with a Cre-conditional allele of Tfam. 4-hydroxytamoxifen or vehicle was delivered by four consecutive daily intraperitoneal injections. TdTomato was robustly expressed in the RPE of mice of both sexes for inductions beginning at P14 (males 90.7 ± 4.5%, females 84.7 ± 3.2%) and at 7 weeks (males 84.3 ± 7.0%, females 82 ± 3.6%). <0.6% of Muller glia also expressed tdTomato, but no tdTomato fluorescence was observed in other ocular cells or in multiple non-ocular tissues, with the exception of sparse foci in the testis. No evidence of retinal toxicity was observed in mice homozygous for the transgene induced beginning at P14 and assessed at 7-10 months. RPE-selective ablation of Tfam beginning at P14 led to reduced retinal thickness at 8 months of age and diminished retinal electrical responses at 12 months, as expected. These findings demonstrate that we have generated a mouse line with consistently efficient, tamoxifen-mediated postnatal induction of Cre recombination in the RPE and a small fraction of Muller glia. This line should be useful for temporally regulated modulation of gene function in the murine RPE.


Assuntos
Regulação da Expressão Gênica , Integrases/genética , Degeneração Macular/genética , Epitélio Pigmentado da Retina/metabolismo , Animais , Modelos Animais de Doenças , Eletrorretinografia , Integrases/biossíntese , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA/genética , RNA/metabolismo , Epitélio Pigmentado da Retina/patologia , Tomografia de Coerência Óptica
3.
J Neuroinflammation ; 17(1): 304, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-33059703

RESUMO

BACKGROUND: Interleukin-6 (IL-6) is a pleiotropic cytokine that controls numerous physiological processes both in basal and neuroinflammatory conditions, including the inflammatory response to experimental autoimmune encephalomyelitis (EAE). IL-6 is produced by multiple peripheral and central cells, and until now, the putative roles of IL-6 from different cell types have been evaluated through conditional cell-specific IL-6 knockout mice. Nevertheless, these mice probably undergo compensatory responses of IL-6 from other cells, which makes it difficult to assess the role of each source of IL-6. METHODS: To give some insight into this problem, we have produced a novel mouse model: a conditional reversible IL-6 KO mouse (IL6-DIO-KO). By using double-inverted, open-reading-frame (DIO) technology, we created a mouse line with the loss of Il6 expression in all cells that can be restored by the action of Cre recombinase. Since microglia are one of the most important sources and targets of IL-6 into the central nervous system, we have recovered microglial Il6 expression in IL6-DIO-KO mice through breeding to Cx3cr1-CreER mice and subsequent injection of tamoxifen (TAM) when mice were 10-16 weeks old. Then, they were immunized with myelin oligodendrocyte glycoprotein 35-55 peptide (MOG35-55) 7 weeks after TAM treatment to induce EAE. Clinical symptoms and demyelination, CD3 infiltration, and gliosis in the spinal cord were evaluated. RESULTS: IL6-DIO-KO mice were resistant to EAE, validating the new model. Restoration of microglial Il6 was sufficient to develop a mild version of EAE-related clinical symptoms and neuropathology. CONCLUSIONS: IL6-DIO-KO mouse is an excellent model to understand in detail the role of specific cellular sources of IL-6 within a recovery-of-function paradigm in EAE.


Assuntos
Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/metabolismo , Integrases/biossíntese , Interleucina-6/biossíntese , Microglia/metabolismo , Sequência de Aminoácidos , Animais , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/patologia , Expressão Gênica , Integrases/genética , Interleucina-6/deficiência , Interleucina-6/genética , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia
4.
Am J Respir Cell Mol Biol ; 62(6): 692-698, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32208105

RESUMO

Type II alveolar epithelial cells (AEC2s) play an essential role in the function and maintenance of the pulmonary epithelium. Several transgenic mice have been developed to study the function of these cells in vivo by using the human SFTPC promoter to drive expression of Cre recombinase. The precise activity of each of these transgenic alleles has not been studied, and previous reports suggest that their activity can depend on breeding strategies. We bred mice with a conditional allele of the essential telomere capping protein TRF2 with two different SFTPC-Cre-transgenic strains and observed opposite phenotypes (100% lethality vs. 100% viability). We characterized the Cre recombinase activity in these two transgenic lines and found that the contrasting phenotypes were driven by difference in embryonic expression of the two transgenes, likely due to position effects or differences in the transgenic constructs. We also tested if SFTPC-Cre activity was dependent on maternal or paternal inheritance. When paternally inherited, both SFTPC-Cre alleles produced offspring with constitutive reporter activity independent of the inheritance of the Cre allele, suggesting that Cre recombinase was expressed in the male germline before meiosis. Immunohistochemical analysis of the testis showed reporter activity during spermatogenesis. Analysis of single-cell RNA sequencing data from murine and human testis demonstrated SFTPC expression uniquely during human spermatogenesis, suggesting that use of the human promoter in these constructs is responsible for male germline activity. Our data highlight the importance of careful analysis of transgenic allele activity and identify an SFTPC-Cre allele that is useful for panepithelial targeting in the mouse.


Assuntos
Integrases/genética , Regiões Promotoras Genéticas/genética , Proteína C Associada a Surfactante Pulmonar/genética , Transgenes , Alelos , Células Epiteliais Alveolares/metabolismo , Animais , Linhagem da Célula , Senescência Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Letais , Genes Reporter , Estudos de Associação Genética , Humanos , Integrases/biossíntese , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes/metabolismo , Análise de Célula Única , Espermatogênese , Homeostase do Telômero/genética , Proteína 2 de Ligação a Repetições Teloméricas/biossíntese , Proteína 2 de Ligação a Repetições Teloméricas/genética , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
5.
eNeuro ; 6(5)2019.
Artigo em Inglês | MEDLINE | ID: mdl-31451604

RESUMO

Genetically modified mice have become standard tools in neuroscience research. Our understanding of the basal ganglia in particular has been greatly assisted by BAC mutants with selective transgene expression in striatal neurons forming the direct or indirect pathways. However, for more sophisticated behavioral tasks and larger intracranial implants, rat models are preferred. Furthermore, BAC lines can show variable expression patterns depending upon genomic insertion site. We therefore used CRISPR/Cas9 to generate two novel knock-in rat lines specifically encoding Cre recombinase immediately after the dopamine D1 receptor (Drd1a) or adenosine 2a receptor (Adora2a) loci. Here, we validate these lines using in situ hybridization and viral vector mediated transfection to demonstrate selective, functional Cre expression in the striatal direct and indirect pathways, respectively. We used whole-genome sequencing to confirm the lack of off-target effects and established that both rat lines have normal locomotor activity and learning in simple instrumental and Pavlovian tasks. We expect these new D1-Cre and A2a-Cre rat lines will be widely used to study both normal brain functions and neurological and psychiatric pathophysiology.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Integrases/genética , Receptor A2A de Adenosina/genética , Receptores de Dopamina D1/genética , Animais , Feminino , Técnicas de Introdução de Genes/métodos , Integrases/biossíntese , Masculino , Ratos , Ratos Long-Evans , Ratos Transgênicos , Receptor A2A de Adenosina/biossíntese , Receptores de Dopamina D1/biossíntese
6.
J Biol Chem ; 292(21): 8594-8604, 2017 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-28377509

RESUMO

The liver possesses a remarkable capacity to regenerate after damage. There is a heated debate on the origin of new hepatocytes after injuries in adult liver. Hepatic stem/progenitor cells have been proposed to produce functional hepatocytes after injury. Recent studies have argued against this model and suggested that pre-existing hepatocytes, rather than stem cells, contribute new hepatocytes. This hepatocyte-to-hepatocyte model is mainly based on labeling of hepatocytes with Cre-recombinase delivered by the adeno-associated virus. However, the impact of virus infection on cell fate determination, consistency of infection efficiency, and duration of Cre-virus in hepatocytes remain confounding factors that interfere with the data interpretation. Here, we generated a new genetic tool Alb-DreER to label almost all hepatocytes (>99.5%) and track their contribution to different cell lineages in the liver. By "pulse-and-chase" strategy, we found that pre-existing hepatocytes labeled by Alb-DreER contribute to almost all hepatocytes during normal homeostasis and after liver injury. Virtually all hepatocytes in the injured liver are descendants of pre-existing hepatocytes through self-expansion. We concluded that stem cell differentiation is unlikely to be responsible for the generation of a substantial number of new hepatocytes in adult liver. Our study also provides a new mouse tool for more precise in vivo genetic study of hepatocytes in the field.


Assuntos
Diferenciação Celular , Hepatócitos , Regeneração Hepática , Fígado , Células-Tronco , Animais , Rastreamento de Células/métodos , Hepatócitos/metabolismo , Hepatócitos/patologia , Integrases/biossíntese , Integrases/genética , Fígado/lesões , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Transgênicos , Células-Tronco/metabolismo , Células-Tronco/patologia
7.
Oncotarget ; 8(11): 18303-18311, 2017 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-28407687

RESUMO

Cytokeratin 19 (K19) is expressed in various differentiated cells, including gastric, intestinal and bronchial epithelial cells, and liver duct cells. Here, we generated a transgenic mouse line, K19-Cre, in which the expression of Cre recombinase was controlled by the promoter of K19. To test the tissue distribution and excision activity of Cre recombinase, K19-Cre transgenic mice were bred with Rosa26 reporter strain and a mouse strain that carries PTEN conditional alleles (PTENLoxp/Loxp). At mRNA level, Cre was strongly expressed in the stomach, lung and intestine, while in stomach, lung, and liver at protein level. The immunoreactivity to Cre was strongly observed the cytoplasm of gastric, bronchial and intestinal epithelial cells. Cre activity was detectable in gastric, bronchial and intestinal epithelial cells, according to LacZ staining. In K19-Cre/PTEN Loxp/Loxp mice, PTEN was abrogated in stomach, intestine, lung, liver and breast, the former two of which were verified by in situ PCR. There appeared breast cancer with PTEN loss. These data suggest that K19 promoter may be a useful tool to study the pathophysiological functions of cytokeratin 19-positive cells, especially gastrointestinal epithelial cells. Cell specificity of neoplasia is not completely attributable to the cell-specific expression of oncogenes and cell-specific loss of tumor suppressor genes.


Assuntos
Integrases/biossíntese , Queratina-19/genética , Neoplasias Gástricas/metabolismo , Animais , Células Epiteliais/metabolismo , Humanos , Queratina-19/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Regiões Promotoras Genéticas , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/genética
8.
Genesis ; 54(12): 626-635, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27731922

RESUMO

Temporal and spatial regulation of genes mediated by tissue-specific promoters and conditional gene expression systems provide a powerful tool to study gene function in health, disease, and during development. Although transgenic mice expressing the Cre recombinase in the gastric epithelium have been reported, there is a lack of models that allow inducible and reversible gene modification in the stomach. Here, we exploited the gastrointestinal epithelium-specific expression pattern of the three trefoil factor (Tff) genes and bacterial artificial chromosome transgenesis to generate a novel mouse strain that expresses the CreERT2 recombinase and the reverse tetracycline transactivator (rtTA). The Tg(Tff1-CreERT2;Tff2-rtTA;Tff3-Luc) strain confers tamoxifen-inducible irreversible somatic recombination and allows simultaneous doxycycline-dependent reversible gene activation in the gastric epithelium of developing and adult mice. This strain also confers luciferase activity to the intestinal epithelium to enable in vivo bioluminescence imaging. Using fluorescent reporters as conditional alleles, we show Tff1-CreERT2 and Tff2-rtTA transgene activity in a partially overlapping subset of long-term regenerating gastric stem/progenitor cells. Therefore, the Tg(Tff1-CreERT2;Tff2-rtTA;Tff3-Luc) strain can confer intermittent transgene expression to gastric epithelial cells that have undergone previous gene modification, and may be suitable to genetically model therapeutic intervention during development, tumorigenesis, and other genetically tractable diseases. Birth Defects Research (Part A) 106:626-635, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Mucosa Intestinal/metabolismo , Fator Trefoil-1/biossíntese , Fator Trefoil-2/biossíntese , Fator Trefoil-3/biossíntese , Animais , Regulação da Expressão Gênica no Desenvolvimento , Integrases/biossíntese , Integrases/genética , Mucosa Intestinal/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Recombinases/genética , Transgenes/genética , Fator Trefoil-1/genética , Fator Trefoil-2/genética , Fator Trefoil-3/genética
9.
Genesis ; 53(11): 709-17, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26454009

RESUMO

We have established a novel Cre mouse line, using genomic elements encompassing the Nrp2 locus, present within a bacterial artificial chromosome clone. By crossing this Cre driver line to R26R LacZ reporter mice, we have documented the temporal expression and lineage traced tissues in which Cre is expressed. Nrp2-Cre drives expression in primitive blood cells arising from the yolk sac, venous and lymphatic endothelial cells, peripheral sensory ganglia, and the lung bud. This mouse line will provide a new tool to researchers wishing to study the development of various tissues and organs in which this Cre driver is expressed, as well as allow tissue-specific knockout of genes of interest to study protein function. This work also presents the first evidence for expression of Nrp2 protein in a mesodermal progenitor with restricted hematopoietic potential, which will significantly advance the study of primitive erythropoiesis. genesis 53:709-717, 2015. © 2015 Wiley Periodicals, Inc.


Assuntos
Linhagem da Célula , Integrases/biossíntese , Camundongos Transgênicos , Neuropilina-2/genética , Células-Tronco/metabolismo , Animais , Desenvolvimento Embrionário/genética , Endotélio Vascular/citologia , Eritrócitos/metabolismo , Gânglios/citologia , Células-Tronco Hematopoéticas/citologia , Modelos Biológicos , Crista Neural/citologia , Células-Tronco Neurais/citologia
10.
J Virol ; 89(17): 9044-60, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26085166

RESUMO

UNLABELLED: To induce and trigger innate and adaptive immune responses, antigen-presenting cells (APCs) take up and process antigens. Retroviral particles are capable of transferring not only genetic information but also foreign cargo proteins when they are genetically fused to viral structural proteins. Here, we demonstrate the capacity of lentiviral protein transfer vectors (PTVs) for targeted antigen transfer directly into APCs and thereby induction of cytotoxic T cell responses. Targeting of lentiviral PTVs to APCs can be achieved analogously to gene transfer vectors by pseudotyping the particles with truncated wild-type measles virus (MV) glycoproteins (GPs), which use human SLAM (signaling lymphocyte activation molecule) as a main entry receptor. SLAM is expressed on stimulated lymphocytes and APCs, including dendritic cells. SLAM-targeted PTVs transferred the reporter protein green fluorescent protein (GFP) or Cre recombinase with strict receptor specificity into SLAM-expressing CHO and B cell lines, in contrast to broadly transducing vesicular stomatitis virus G protein (VSV-G) pseudotyped PTVs. Primary myeloid dendritic cells (mDCs) incubated with targeted or nontargeted ovalbumin (Ova)-transferring PTVs stimulated Ova-specific T lymphocytes, especially CD8(+) T cells. Administration of Ova-PTVs into SLAM-transgenic and control mice confirmed the observed predominant induction of antigen-specific CD8(+) T cells and demonstrated the capacity of protein transfer vectors as suitable vaccines for the induction of antigen-specific immune responses. IMPORTANCE: This study demonstrates the specificity and efficacy of antigen transfer by SLAM-targeted and nontargeted lentiviral protein transfer vectors into antigen-presenting cells to trigger antigen-specific immune responses in vitro and in vivo. The observed predominant activation of antigen-specific CD8(+) T cells indicates the suitability of SLAM-targeted and also nontargeted PTVs as a vaccine for the induction of cytotoxic immune responses. Since cytotoxic CD8(+) T lymphocytes are a mainstay of antitumoral immune responses, PTVs could be engineered for the transfer of specific tumor antigens provoking tailored antitumoral immunity. Therefore, PTVs can be used as safe and efficient alternatives to gene transfer vectors or live attenuated replicating vector platforms, avoiding genotoxicity or general toxicity in highly immunocompromised patients, respectively. Thereby, the potential for easy envelope exchange allows the circumventing of neutralizing antibodies, e.g., during repeated boost immunizations.


Assuntos
Antígenos CD/imunologia , Vetores Genéticos/genética , Ovalbumina/imunologia , Receptores de Superfície Celular/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Antígenos CD/biossíntese , Células CHO , Cricetulus , Células Dendríticas/imunologia , Proteínas de Fluorescência Verde/biossíntese , Células HEK293 , Humanos , Integrases/biossíntese , Integrases/genética , Lentivirus/genética , Ativação Linfocitária/imunologia , Vírus do Sarampo/genética , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Transporte Proteico , Receptores de Superfície Celular/biossíntese , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Transfecção , Vacinas de Subunidades Antigênicas/imunologia , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética
11.
Cardiovasc Res ; 104(1): 15-23, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25082846

RESUMO

AIMS: Inducible gene targeting in mice using the Cre/LoxP system has become a valuable tool to analyse the roles of specific genes in the adult heart. However, the commonly used Myh6-MerCreMer system requires time-consuming breeding schedules and is potentially associated with cardiac side effects, which may result in transient cardiac dysfunction. The aim of our study was to establish a rapid and simple system for cardiac gene inactivation in conditional knockout mice by gene transfer of a Cre recombinase gene using adeno-associated viral vectors of serotype 9 (AAV9). METHODS AND RESULTS: AAV9 vectors expressing Cre under the control of a human cardiac troponin T promoter (AAV-TnT-Cre) enabled a highly efficient Cre/LoxP switching in cardiomyocytes 2 weeks after injection into 5- to 6-week-old ROSA26-LacZ reporter mice. Recombination efficiency was at least as high as observed with the Myh6-MerCreMer system. No adverse side effects were detected upon application of AAV-TnT-Cre. As proof of principle, we studied AAV-TnT-Cre in a conditional knockout model (Srf-flex1 mice) to deplete the myocardium of the transcription factor serum response factor (SRF). Four weeks after AAV-TnT-Cre injection, a strong decrease in the cardiac expression of SRF mRNA and protein was observed. Furthermore, mice developed a severe cardiac dysfunction with increased interstitial fibrosis in accordance with the central role of SRF for the expression of contractile and calcium trafficking proteins in the heart. CONCLUSIONS: AAV9-mediated expression of Cre is a promising approach for rapid and efficient conditional cardiac gene knockout in adult mice.


Assuntos
Dependovirus/genética , Técnicas de Silenciamento de Genes , Integrases/genética , Miócitos Cardíacos/metabolismo , Animais , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Dependovirus/enzimologia , Regulação para Baixo , Fibrose , Genótipo , Integrases/biossíntese , Óperon Lac , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/patologia , Fenótipo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , RNA não Traduzido/genética , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Fatores de Tempo , Troponina T/genética , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda
12.
Breast Cancer Res ; 16(1): R11, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24457046

RESUMO

INTRODUCTION: Effective in vivo models of breast cancer are crucial for studying the development and progression of the disease in humans. We sought to engineer a novel mouse model of polyomavirus middle T antigen (PyV mT)-mediated mammary tumourigenesis in which inducible expression of this well-characterized viral oncoprotein is coupled to Cre recombinase (TetO-PyV mT-IRES-Cre recombinase or MIC). METHODS: MIC mice were crossed to the mouse mammary tumour virus (MMTV)-reverse tetracycline transactivator (rtTA) strain to generate cohorts of virgin females carrying one or both transgenes. Experimental (rtTA/MIC) and control (rtTA or MIC) animals were administered 2 mg/mL doxycycline beginning as early as eight weeks of age and monitored for mammary tumour formation, in parallel with un-induced controls of the same genotypes. RESULTS: Of the rtTA/MIC virgin females studied, 90% developed mammary tumour with complete penetrance to all glands in response to doxycycline and a T50 of seven days post-induction, while induced or un-induced controls remained tumour-free after one year of induction. Histological analyses of rtTA/MIC mammary glands and tumour revealed that lesions followed the canonical stepwise progression of PyV mT tumourigenesis, from hyperplasia to mammary intraepithelial neoplasia/adenoma, carcinoma, and invasive carcinoma that metastasizes to the lung; at each of these stages expression of PyV mT and Cre recombinase transgenes was confirmed. Withdrawal of doxycycline from rtTA/MIC mice with end-stage mammary tumours led to rapid regression, yet animals eventually developed PyV mT-expressing and -non-expressing recurrent masses with varied tumour histopathologies. CONCLUSIONS: We have successfully created a temporally regulated mouse model of PyV mT-mediated mammary tumourigenesis that can be used to study Cre recombinase-mediated genetic changes simultaneously. While maintaining all of the hallmark features of the well-established constitutive MMTV-PyV mT model, the utility of this strain derives from the linking of PyV mT and Cre recombinase transgenes; mammary epithelial cells are thereby forced to couple PyV mT expression with conditional ablation of a given gene. This transgenic mouse model will be an important research tool for identifying synthetic viable genetic events that enable PyV mT tumours to evolve in the absence of a key signaling pathway.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Transformação Celular Neoplásica/genética , Integrases/genética , Neoplasias Mamárias Experimentais/genética , Vírus do Tumor Mamário do Camundongo/genética , Animais , Antígenos Transformantes de Poliomavirus/biossíntese , Sequência de Bases , Carcinoma in Situ/genética , Carcinoma in Situ/patologia , Modelos Animais de Doenças , Progressão da Doença , Doxiciclina/farmacologia , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Hiperplasia/genética , Hiperplasia/patologia , Integrases/biossíntese , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Análise de Sequência de DNA
13.
Lab Invest ; 93(8): 868-79, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23732814

RESUMO

The protective effect of heme oxygenase-1 (HO-1) expression in cardiovascular disease has been previously demonstrated using transgenic animal models in which HO-1 is constitutively overexpressed in the heart. However, the temporal requirements for protection by HO-1 induction relative to injury have not been investigated, but are essential to employ HO-1 as a therapeutic strategy in human cardiovascular disease states. Therefore, we generated mice with cardiac-specific, tamoxifen (TAM)-inducible overexpression of a human HO-1 (hHO-1) transgene (myosin heavy chain (MHC)-HO-1 mice) by breeding mice with cardiac-specific expression of a TAM-inducible Cre recombinase (MHC-Cre mice), with mice containing an hHO-1 transgene preceded by a floxed-stop signal. MHC-HO-1 mice overexpress HO-1 mRNA and the enzymatically active protein following TAM administration (40 mg/kg body weight on 2 consecutive days). In MHC-Cre controls, TAM administration leads to severe, acute cardiac toxicity, cardiomyocyte necrosis, and 80% mortality by day 3. This cardiac toxicity is accompanied by a significant increase in inflammatory cells in the heart that are predominantly neutrophils. In MHC-HO-1 mice, HO-1 overexpression ameliorates the depression of cardiac function and high mortality rate observed in MHC-Cre mice following TAM administration and attenuates cardiomyocyte necrosis and neutrophil infiltration. These results highlight that HO-1 induction is sufficient to prevent the depression of cardiac function observed in mice with TAM-inducible Cre recombinase expression by protecting the heart from necrosis and neutrophil infiltration. These findings are important because MHC-Cre mice are widely used in cardiovascular research despite the limitations imposed by Cre-induced cardiac toxicity, and also because inflammation is an important pathological component of many human cardiovascular diseases.


Assuntos
Cardiopatias/prevenção & controle , Heme Oxigenase-1/metabolismo , Integrases/biossíntese , Miocárdio/enzimologia , Doença Aguda , Animais , Antineoplásicos Hormonais/farmacologia , Modelos Animais de Doenças , Indução Enzimática , Feminino , Coração/efeitos dos fármacos , Coração/fisiopatologia , Cardiopatias/enzimologia , Cardiopatias/mortalidade , Cardiopatias/patologia , Heme Oxigenase-1/genética , Humanos , Integrases/genética , Longevidade/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Necrose/induzido quimicamente , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Taxa de Sobrevida , Tamoxifeno/farmacologia
14.
J Vis Exp ; (72): e50156, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23407503

RESUMO

Genomic, proteomic, transcriptomic, and epigenomic analyses of human tumors indicate that there are thousands of anomalies within each cancer genome compared to matched normal tissue. Based on these analyses it is evident that there are many undiscovered genetic drivers of cancer(1). Unfortunately these drivers are hidden within a much larger number of passenger anomalies in the genome that do not directly contribute to tumor formation. Another aspect of the cancer genome is that there is considerable genetic heterogeneity within similar tumor types. Each tumor can harbor different mutations that provide a selective advantage for tumor formation(2). Performing an unbiased forward genetic screen in mice provides the tools to generate tumors and analyze their genetic composition, while reducing the background of passenger mutations. The Sleeping Beauty (SB) transposon system is one such method(3). The SB system utilizes mobile vectors (transposons) that can be inserted throughout the genome by the transposase enzyme. Mutations are limited to a specific cell type through the use of a conditional transposase allele that is activated by Cre Recombinase. Many mouse lines exist that express Cre Recombinase in specific tissues. By crossing one of these lines to the conditional transposase allele (e.g. Lox-stop-Lox-SB11), the SB system is activated only in cells that express Cre Recombinase. The Cre Recombinase will excise a stop cassette that blocks expression of the transposase allele, thereby activating transposon mutagenesis within the designated cell type. An SB screen is initiated by breeding three strains of transgenic mice so that the experimental mice carry a conditional transposase allele, a concatamer of transposons, and a tissue-specific Cre Recombinase allele. These mice are allowed to age until tumors form and they become moribund. The mice are then necropsied and genomic DNA is isolated from the tumors. Next, the genomic DNA is subjected to linker-mediated-PCR (LM-PCR) that results in amplification of genomic loci containing an SB transposon. LM-PCR performed on a single tumor will result in hundreds of distinct amplicons representing the hundreds of genomic loci containing transposon insertions in a single tumor(4). The transposon insertions in all tumors are analyzed and common insertion sites (CISs) are identified using an appropriate statistical method(5). Genes within the CIS are highly likely to be oncogenes or tumor suppressor genes, and are considered candidate cancer genes. The advantages of using the SB system to identify candidate cancer genes are: 1) the transposon can easily be located in the genome because its sequence is known, 2) transposition can be directed to almost any cell type and 3) the transposon is capable of introducing both gain- and loss-of-function mutations(6). The following protocol describes how to devise and execute a forward genetic screen using the SB transposon system to identify candidate cancer genes (Figure 1).


Assuntos
Elementos de DNA Transponíveis/genética , Genes Neoplásicos , Neoplasias/genética , Reação em Cadeia da Polimerase/métodos , Transposases/genética , Animais , Humanos , Integrases/biossíntese , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos
15.
J Vis Exp ; (59): e3581, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22230968

RESUMO

The ability of two or more cells of the same type to fuse has been utilized in metazoans throughout evolution to form many complex organs, including skeletal muscle, bone and placenta. Contemporary studies demonstrate fusion of cells of the same type confers enhanced function. For example, when the trophoblast cells of the placenta fuse to form the syncytiotrophoblast, the syncytiotrophoblast is better able to transport nutrients and hormones across the maternal-fetal barrier than unfused trophoblasts(1-4). More recent studies demonstrate fusion of cells of different types can direct cell fate. The "reversion" or modification of cell fate by fusion was once thought to be limited to cell culture systems. But the advent of stem cell transplantation led to the discovery by us and others that stem cells can fuse with somatic cells in vivo and that fusion facilitates stem cell differentiation(5-7). Thus, cell fusion is a regulated process capable of promoting cell survival and differentiation and thus could be of central importance for development, repair of tissues and even the pathogenesis of disease. Limiting the study of cell fusion, is lack of appropriate technology to 1) accurately identify fusion products and to 2) track fusion products over time. Here we present a novel approach to address both limitations via induction of bioluminescence upon fusion (Figure 1); bioluminescence can be detected with high sensitivity in vivo(8-15). We utilize a construct encoding the firefly luciferase (Photinus pyralis) gene placed adjacent to a stop codon flanked by LoxP sequences. When cells expressing this gene fuse with cells expressing the Cre recombinase protein, the LoxP sites are cleaved and the stop signal is excised allowing transcription of luciferase. Because the signal is inducible, the incidence of false-positive signals is very low. Unlike existing methods which utilize the Cre/LoxP system(16, 17), we have incorporated a "living" detection signal and thereby afford for the first time the opportunity to track the kinetics of cell fusion in vivo. To demonstrate the approach, mice ubiquitously expressing Cre recombinase served as recipients of stem cells transfected with a construct to express luciferase downstream of a floxed stop codon. Stem cells were transplanted via intramyocardial injection and after transplantation intravital image analysis was conducted to track the presence of fusion products in the heart and surrounding tissues over time. This approach could be adapted to analyze cell fusion in any tissue type at any stage of development, disease or adult tissue repair.


Assuntos
Fusão Celular/métodos , Integrases/genética , Recombinação Genética , Animais , Integrases/biossíntese , Luciferases de Vaga-Lume/genética , Medições Luminescentes/métodos , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Transfecção/métodos
16.
J Vis Exp ; (54)2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21841766

RESUMO

Normal brain function relies not only on embryonic development when major neuronal pathways are established, but also on postnatal development when neural circuits are matured and refined. Misregulation at this stage may lead to neurological and psychiatric disorders such as autism and schizophrenia. Many genes have been studied in the prenatal brain and found crucial to many developmental processes. However, their function in the postnatal brain is largely unknown, partly because their deletion in mice often leads to lethality during neonatal development, and partly because their requirement in early development hampers the postnatal analysis. To overcome these obstacles, floxed alleles of these genes are currently being generated in mice. When combined with transgenic alleles that express Cre recombinase in specific cell types, conditional deletion can be achieved to study gene function in the postnatal brain. However, this method requires additional alleles and extra time (3-6 months) to generate the mice with appropriate genotypes, thereby limiting the expansion of the genetic analysis to a large scale in the mouse brain. Here we demonstrate a complementary approach that uses virally-expressed Cre to study these floxed alleles rapidly and systematically in postnatal brain development. By injecting recombinant adeno-associated viruses (rAAVs) encoding Cre into the neonatal brain, we are able to delete the gene of interest in different regions of the brain. By controlling the viral titer and coexpressing a fluorescent protein marker, we can simultaneously achieve mosaic gene inactivation and sparse neuronal labeling. This method bypasses the requirement of many genes in early development, and allows us to study their cell autonomous function in many critical processes in postnatal brain development, including axonal and dendritic growth, branching, and tiling, as well as synapse formation and refinement. This method has been used successfully in our own lab (unpublished results) and others, and can be extended to other viruses, such as lentivirus, as well as to the expression of shRNA or dominant active proteins. Furthermore, by combining this technique with electrophysiology as well as recently-developed optical imaging tools, this method provides a new strategy to study how genetic pathways influence neural circuit development and function in mice and rats.


Assuntos
Alelos , Encéfalo/fisiologia , Dependovirus/genética , Integrases/genética , Recombinação Genética , Animais , Dependovirus/enzimologia , Integrases/biossíntese , Camundongos , Ratos
17.
Cancer Res ; 71(13): 4686-95, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21551250

RESUMO

Stem cells are under strict regulation by both intrinsic factors and the microenvironment. There is increasing evidence that many cancers initiate through acquisition of genetic mutations (loss of intrinsic control) in stem cells or their progenitors, followed by alterations of the surrounding microenvironment (loss of extrinsic control). In neurofibromatosis type 1 (NF1), deregulation of Ras signaling results in development of multiple neurofibromas, complex tumors of the peripheral nerves. Neurofibromas arise from the Schwann cell lineage following loss of function at the NF1 locus, which initiates a cascade of interactions with other cell types in the microenvironment and additional cell autonomous modifications. In this study, we sought to identify whether a temporal "window of opportunity" exists during which cells of the Schwann cell lineage can give rise to neurofibromas following loss of NF1. We showed that acute loss of NF1 in both embryonic and adult Schwann cells can lead to neurofibroma formation. However, the embryonic period when Schwann cell precursors and immature Schwann cells are most abundant coincides with enhanced susceptibility to plexiform neurofibroma tumorigenesis. This model has important implications for understanding early cellular events that dictate neurofibroma development, as well as for the development of novel therapies targeting these tumors.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Genes da Neurofibromatose 1 , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/patologia , Células de Schwann/fisiologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hiperplasia , Integrases/biossíntese , Integrases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/biossíntese , Proteína Proteolipídica de Mielina/genética , Nervos Periféricos/patologia , Gravidez , Recombinação Genética , Células de Schwann/patologia , Tamoxifeno/farmacologia , Transgenes
18.
Cancer Res ; 71(13): 4675-85, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21551249

RESUMO

Plexiform neurofibromas are peripheral nerve sheath tumors initiated by biallelic mutation of the NF1 tumor suppressor gene in the Schwann cell lineage. To understand whether neurofibroma formation is possible after birth, we induced Nf1 loss of function with an inducible proteolipid protein Cre allele. Perinatal loss of Nf1 resulted in the development of small plexiform neurofibromas late in life, whereas loss in adulthood caused large plexiform neurofibromas and morbidity beginning 4 months after onset of Nf1 loss. A conditional EGFP reporter allele identified cells showing recombination, including peripheral ganglia satellite cells, peripheral nerve S100ß+ myelinating Schwann cells, and peripheral nerve p75+ cells. Neurofibromas contained cells with Remak bundle disruption but no recombination within GFAP+ nonmyelinating Schwann cells. Extramedullary lympho-hematopoietic expansion was also observed in PlpCre;Nf1fl/fl mice. These tumors contained EGFP+/Sca-1+ stromal cells among EGFP-negative lympho-hematopoietic cells indicating a noncell autonomous effect and unveiling a role of Nf1-deleted microenvironment on lympho-hematopoietic proliferation in vivo. Together these findings define a tumor suppressor role for Nf1 in the adult and narrow the range of potential neurofibroma-initiating cell populations.


Assuntos
Inativação Gênica , Genes da Neurofibromatose 1 , Integrases/biossíntese , Proteína Proteolipídica de Mielina/biossíntese , Neurofibromatose 1/genética , Tamoxifeno/farmacologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Integrases/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética
19.
Genesis ; 48(10): 585-95, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20715174

RESUMO

When compared with the many tamoxifen-activated Cre mouse lines available for gene manipulation studies, relatively few RU486-inducible Cre mice are in use, due to leakiness issues. Here, we report the generation of an RU486-inducible triple fusion gene (GCrePR1e), consisting of green fluorescent protein, Cre, and the progesterone receptor ligand-binding domain (F642-L901). We sought to improve the GCrePR1e by selecting a truncated human lactoferrin (Lf) promoter to drive its expression, based on the promoter's low basal activity and innate sensitivity to RU486. The resulting vector displayed decreased leakiness and increased Cre induction by RU486 through transcriptional and posttranslational regulation in in vitro transfection assays. Inducible GCrePR1e expression was found in most organs of Lf-GCrePR1e transgenic mice and highly activated in the salivary gland, spleen, and lymph nodes. In the bigenic mouse generated by crossing the Lf-GCrePR1e mouse and the Cre reporter mouse (R26R-LacZ), we found that RU486-induced LacZ expression only in the mucous acini and striated ducts of the salivary gland and had very low background recombination in the untreated mice. Our results demonstrated that the Lf-CrePR1e vector was suitable for in vitro recombination in culture models, and Lf-CrePR1e transgenic mice could mediate spatially restricted and RU486-induced gene manipulation in the salivary gland.


Assuntos
Proteínas de Fluorescência Verde/genética , Integrases/genética , Lactoferrina/genética , Mifepristona/farmacologia , Proteínas Recombinantes de Fusão/genética , Glândulas Salivares/metabolismo , Animais , Estradiol/farmacologia , Genes Reporter , Vetores Genéticos/genética , Humanos , Integrases/biossíntese , Integrases/metabolismo , Óperon Lac , Luciferases de Vaga-Lume/metabolismo , Camundongos , Camundongos Transgênicos , Progesterona/farmacologia , Regiões Promotoras Genéticas/genética , Receptores de Progesterona/genética , Recombinação Genética , Tamoxifeno/farmacologia , Transfecção
20.
J Immunol ; 184(6): 2793-804, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20142358

RESUMO

Mechanisms that regulate the lifespan of CD4(+)CD8(+) double-positive (DP) thymocytes help shape the peripheral T cell repertoire. However, the molecular mechanisms controlling DP thymocyte survival remain poorly understood. The Myb proto-oncogene encodes a transcription factor required during multiple stages of T cell development. We demonstrate that Myb mRNA expression is upregulated as thymocytes differentiate from the double-negative into the metabolically quiescent, small, preselection DP stage during T cell development. Using a conditional deletion mouse model, we demonstrate that Myb-deficient DP thymocytes undergo premature apoptosis, resulting in a limited Tcralpha repertoire biased toward 5' Jalpha segment usage. Premature apoptosis occurs specifically in the small preselection DP compartment in an alphabetaTCR-independent manner and is a consequence of decreased Bcl-xL expression. Forced Bcl-xL expression is able to rescue survival, and reintroduction of c-Myb restores both Bcl-xL expression and the small preselection DP compartment. We further demonstrate that c-Myb promotes transcription at the Bcl2l1 locus via a genetic pathway that is independent of the expression of T cell-specific factor-1 or RORgammat, two transcription factors that induce Bcl-xL expression in T cell development. Thus, Bcl-xL is a novel mediator of c-Myb activity during normal T cell development.


Assuntos
Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Proteínas Proto-Oncogênicas c-myb/fisiologia , Timo/imunologia , Timo/metabolismo , Regulação para Cima/imunologia , Proteína bcl-X/biossíntese , Animais , Apoptose/genética , Apoptose/imunologia , Antígenos CD4/genética , Antígenos CD8/genética , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Células Clonais , Técnicas de Cocultura , Integrases/biossíntese , Integrases/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myb/deficiência , Proteínas Proto-Oncogênicas c-myb/genética , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/citologia , Regulação para Cima/genética , Proteína bcl-X/genética , Proteína bcl-X/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA