Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
NPJ Breast Cancer ; 7(1): 58, 2021 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-34031428

RESUMO

ID proteins are helix-loop-helix (HLH) transcriptional regulators frequently overexpressed in cancer. ID proteins inhibit basic-HLH transcription factors often blocking differentiation and sustaining proliferation. A small-molecule, AGX51, targets ID proteins for degradation and impairs ocular neovascularization in mouse models. Here we show that AGX51 treatment of cancer cell lines impairs cell growth and viability that results from an increase in reactive oxygen species (ROS) production upon ID degradation. In mouse models, AGX51 treatment suppresses breast cancer colonization in the lung, regresses the growth of paclitaxel-resistant breast tumors when combined with paclitaxel and reduces tumor burden in sporadic colorectal neoplasia. Furthermore, in cells and mice, we fail to observe acquired resistance to AGX51 likely the result of the inability to mutate the binding pocket without loss of ID function and efficient degradation of the ID proteins. Thus, AGX51 is a first-in-class compound that antagonizes ID proteins, shows strong anti-tumor effects and may be further developed for the management of multiple cancers.

3.
Cell Rep ; 29(1): 62-75.e7, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31577956

RESUMO

Id helix-loop-helix (HLH) proteins (Id1-4) bind E protein bHLH transcription factors, preventing them from forming active transcription complexes that drive changes in cell states. Id proteins are primarily expressed during development to inhibit differentiation, but they become re-expressed in adult tissues in diseases of the vasculature and cancer. We show that the genetic loss of Id1/Id3 reduces ocular neovascularization in mouse models of wet age-related macular degeneration (AMD) and retinopathy of prematurity (ROP). An in silico screen identifies AGX51, a small-molecule Id antagonist. AGX51 inhibits the Id1-E47 interaction, leading to ubiquitin-mediated degradation of Ids, cell growth arrest, and reduced viability. AGX51 is well-tolerated in mice and phenocopies the genetic loss of Id expression in AMD and ROP models by inhibiting retinal neovascularization. Thus, AGX51 is a first-in-class compound that antagonizes an interaction formerly considered undruggable and that may have utility in the management of multiple diseases.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neovascularização Patológica/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Células HCT116 , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteína 1 Inibidora de Diferenciação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Neovascularização Patológica/metabolismo
4.
EMBO J ; 37(2): 201-218, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29196303

RESUMO

Whole chromosome gains or losses (aneuploidy) are a hallmark of ~70% of human tumors. Modeling the consequences of aneuploidy has relied on perturbing spindle assembly checkpoint (SAC) components, but interpretations of these experiments are clouded by the multiple functions of these proteins. Here, we used a Cre recombinase-mediated chromosome loss strategy to individually delete mouse chromosomes 9, 10, 12, or 14 in tetraploid immortalized murine embryonic fibroblasts. This methodology also involves the generation of a dicentric chromosome intermediate, which subsequently undergoes a series of breakage-fusion-bridge (BFB) cycles. While the aneuploid cells generally display a growth disadvantage in vitro, they grow significantly better in low adherence sphere-forming conditions and three of the four lines are transformed in vivo, forming large and invasive tumors in immunocompromised mice. The aneuploid cells display increased chromosomal instability and DNA damage, a mutator phenotype associated with tumorigenesis in vivo Thus, these studies demonstrate a causative role for whole chromosome loss and the associated BFB-mediated instability in tumorigenesis and may shed light on the early consequences of aneuploidy in mammalian cells.


Assuntos
Deleção Cromossômica , Cromossomos de Mamíferos , Fibroblastos/metabolismo , Neoplasias Experimentais , Tetraploidia , Animais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Cromossomos de Mamíferos/genética , Cromossomos de Mamíferos/metabolismo , Fibroblastos/patologia , Camundongos , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia
5.
Cell Rep ; 19(9): 1832-1845, 2017 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-28564602

RESUMO

The mitotic checkpoint ensures proper segregation of chromosomes by delaying anaphase until all kinetochores are bound to microtubules. This inhibitory signal is composed of a complex containing Mad2, which inhibits anaphase progression. The complex can be disassembled by p31comet and TRIP13; however, TRIP13 knockdown has been shown to cause only a mild mitotic delay. Overexpression of checkpoint genes, as well as TRIP13, is correlated with chromosomal instability (CIN) in cancer, but the initial effects of Mad2 overexpression are prolonged mitosis and decreased proliferation. Here, we show that TRIP13 overexpression significantly reduced, and TRIP13 reduction significantly exacerbated, the mitotic delay associated with Mad2 overexpression, but not that induced by microtubule depolymerization. The combination of Mad2 overexpression and TRIP13 loss reduced the ability of checkpoint complexes to disassemble and significantly inhibited the proliferation of cells in culture and tumor xenografts. These results identify an unexpected dependency on TRIP13 in cells overexpressing Mad2.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Mad2/metabolismo , Mitose , Animais , Sequência de Bases , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Humanos , Camundongos , Mitose/efeitos dos fármacos , Morfolinas/farmacologia , Neoplasias/metabolismo , Neoplasias/patologia , Nocodazol/farmacologia , Fenótipo , Purinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Am J Pathol ; 185(11): 2983-93, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26348574

RESUMO

Inhibitor of DNA binding (ID)-1 is important for angiogenesis during embryogenesis and tumor development. Whether ID1 expression in endothelial cells of the colon is required for normal response to injury is unknown. We demonstrate that Id1 is up-regulated in colonic endothelial cells in an experimental model of colitis and in the inflamed mucosa of patients with inflammatory bowel disease. Because prostaglandin E2 and tumor necrosis factor-α are also elevated in colitis, we determined whether these factors could induce ID1 transcription in cultured endothelial cells. Tumor necrosis factor-α stimulated ID1 transcription via early growth response 1 protein (Egr-1). By contrast, the induction of ID1 by prostaglandin E2 was mediated by cAMP response element-binding protein (CREB). To determine whether the increased ID1 levels in the endothelial cells of inflamed mucosa were an adaptive response that modulated the severity of tissue injury, Id1 was conditionally depleted in the endothelium of mice, which sensitized the mice to more severe chemical colitis, including more severe diarrhea, bleeding, and histological injury, and shorter colon compared with control mice. Moreover, depletion of Id1 in the vasculature was associated with increased CD31(+) aggregates and increased vascular permeability in inflamed mucosa compared with those in Id1 wild-type control mice. These results suggest that endothelial ID1 up-regulation in inflamed colonic mucosa is an adaptive response that modulates the severity of tissue injury.


Assuntos
Colite/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Proteína 1 Inibidora de Diferenciação/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Cultivadas , Colite/induzido quimicamente , Colite/patologia , Colo/metabolismo , Colo/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/genética , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio/metabolismo , Endotélio/patologia , Humanos , Doenças Inflamatórias Intestinais/patologia , Proteína 1 Inibidora de Diferenciação/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Knockout , Fator de Necrose Tumoral alfa/genética , Regulação para Cima
7.
Cancer Prev Res (Phila) ; 8(4): 303-11, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25623217

RESUMO

Different mechanisms contribute to the development of sporadic, hereditary and colitis-associated colorectal cancer. Inhibitor of DNA binding/differentiation (Id) proteins act as dominant-negative antagonists of basic helix-loop-helix transcription factors. Id1 is a promising target for cancer therapy, but little is known about its role in the development of colon cancer. We used immunohistochemistry to demonstrate that Id1 is overexpressed in human colorectal adenomas and carcinomas, whether sporadic or syndromic. Furthermore, elevated Id1 levels were found in dysplasia and colon cancer arising in patients with inflammatory bowel disease. Because levels of PGE2 are also elevated in both colitis and colorectal neoplasia, we determined whether PGE2 could induce Id1. PGE2 via EP4 stimulated protein kinase A activity resulting in enhanced pCREB-mediated Id1 transcription in human colonocytes. To determine the role of Id1 in carcinogenesis, two mouse models were used. Consistent with the findings in humans, Id1 was overexpressed in tumors arising in both Apc(Min) (/+) mice, a model of familial adenomatous polyposis, and in experimental colitis-associated colorectal neoplasia. Id1 deficiency led to significant decrease in the number of intestinal tumors in Apc(Min) (/+) mice and prolonged survival. In contrast, Id1 deficiency did not affect the number or size of tumors in the model of colitis-associated colorectal neoplasia, likely due to exacerbation of colitis associated with Id1 loss. Collectively, these results suggest that Id1 plays a role in gastrointestinal carcinogenesis. Our findings also highlight the need for different strategies to reduce the risk of colitis-associated colorectal cancer compared with sporadic or hereditary colorectal cancer.


Assuntos
Adenoma/patologia , Colite/complicações , Neoplasias do Colo/patologia , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Genes APC/fisiologia , Doenças Inflamatórias Intestinais/patologia , Proteína 1 Inibidora de Diferenciação/fisiologia , Adenoma/metabolismo , Animais , Western Blotting , Carcinogênese , Células Cultivadas , Imunoprecipitação da Cromatina , Colite/induzido quimicamente , Colite/patologia , Neoplasias do Colo/etiologia , Neoplasias do Colo/prevenção & controle , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Técnicas Imunoenzimáticas , Doenças Inflamatórias Intestinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Bladder Cancer ; 1(2): 159-170, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-27376116

RESUMO

Background: Bladder cancer is one of the most common malignant genitourinary diseases worldwide. Despite advances in surgical technique, medical oncology and radiation therapy, cure of invasive tumors remains elusive for patients with late stage disease. Therefore, new therapeutic strategies are needed to improve the response rates with regard to recurrence, invasion and metastasis. Objective: Inhibitor of DNA binding (Id) proteins have been proposed as therapeutic targets due to the key regulatory role they exert in multiple steps of cancer. We aimed to explore the role of Id proteins in bladder cancer development and the pattern of expression of Id proteins in bladder carcinomas. Methods: We used a well-established chemically induced model of bladder carcinogenesis. Wild type and Id-deficient mice were given N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN) in the drinking water and urinary bladder lesions were analyzed histopathologically and stained for Id1. We assessed the effects of Id1 inactivation in cultured bladder cancer cells and in a model of metastatic lung colonization. We also performed Id1 staining of human urothelial carcinoma samples and matched lymph node metastases. Results: Id1 protein was overexpressed in the BBN-induced model of bladder cancer. Id1 deficiency resulted in the development of urinary bladder tumors with areas of extensive hemorrhage and decreased invasiveness when compared to wild type mice. Id1 inactivation led to decreased cell growth in vitro and lung colonization in vivo of human bladder cancer cells. Immunohistochemistry performed on human urothelial carcinoma samples showed Id1 positive staining in both primary tumors and lymph node metastases. Conclusions: In summary, our studies reveal the physiological relevance of Id1 in bladder cancer progression and suggest that targeting Id1 may be important in the development of novel therapies for the treatment of bladder cancer.

9.
Stem Cell Reports ; 3(5): 716-24, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25418719

RESUMO

LGR5 and BMI1 mark intestinal stem cells in crypt base columnar cells and +4 position cells, respectively, but characterization of functional markers in these cell populations is limited. ID1 maintains the stem cell potential of embryonic, neural, and long-term repopulating hematopoietic stem cells. Here, we show in both human and mouse intestine that ID1 is expressed in cycling columnar cells, +4 position cells, and transit-amplifying cells in the crypt. Lineage tracing revealed ID1+ cells to be self-renewing, multipotent stem/progenitor cells that are responsible for the long-term renewal of the intestinal epithelium. Single ID1+ cells can generate long-lived organoids resembling mature intestinal epithelium. Complete knockout of Id1 or selective deletion of Id1 in intestinal epithelium or in LGR5+ stem cells sensitizes mice to chemical-induced colon injury. These experiments identify ID1 as a marker for intestinal stem/progenitor cells and demonstrate a role for ID1 in maintaining the potential for repair in response to colonic injury.


Assuntos
Biomarcadores/metabolismo , Proteína 1 Inibidora de Diferenciação/metabolismo , Mucosa Intestinal/metabolismo , Células-Tronco Multipotentes/metabolismo , Animais , Proliferação de Células , Colite/induzido quimicamente , Colite/genética , Colite/metabolismo , Colo/lesões , Colo/metabolismo , Sulfato de Dextrana , Expressão Gênica , Humanos , Imuno-Histoquímica , Proteína 1 Inibidora de Diferenciação/genética , Mucosa Intestinal/citologia , Intestinos/citologia , Antígeno Ki-67/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Organoides/citologia , Organoides/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Cell Rep ; 5(5): 1228-42, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24332369

RESUMO

ID genes are required for breast cancer colonization of the lungs, but the mechanism remains poorly understood. Here, we show that Id1 expression induces a stem-like phenotype in breast cancer cells while retaining epithelial properties, contrary to the notion that cancer stem-like properties are inextricably linked to the mesenchymal state. During metastatic colonization, Id1 induces a mesenchymal-to-epithelial transition (MET), specifically in cells whose mesenchymal state is dependent on the Id1 target protein Twist1, but not at the primary site, where this state is controlled by the zinc finger protein Snail1. Knockdown of Id expression in metastasizing cells prevents MET and dramatically reduces lung colonization. Furthermore, Id1 is induced by transforming growth factor (TGF)-ß only in cells that have first undergone epithelial-to-mesenchymal transition (EMT), demonstrating that EMT is a prerequisite for subsequent Id1-induced MET during lung colonization. Collectively, these studies underscore the importance of Id-mediated phenotypic switching during distinct stages of breast cancer metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Transição Epitelial-Mesenquimal , Proteína 1 Inibidora de Diferenciação/metabolismo , Neoplasias Pulmonares/secundário , Proteínas Nucleares/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Neoplasias da Mama/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Células MCF-7 , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Transdução de Sinais , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo
11.
Stem Cells Dev ; 21(3): 384-93, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22013995

RESUMO

Understanding the mechanism by which embryonic stem (ES) cells self-renew is crucial for the realization of their therapeutic potential. Earlier, overexpression of Id proteins was shown to be sufficient to maintain mouse ES cells in a self-renewing state even in the absence of serum. Here, we use ES cells derived from Id deficient mice to investigate the requirement for Id proteins in maintaining ES cell self-renewal. We find that Id1(-/-) ES cells have a defect in self-renewal and a propensity to differentiate. We observe that chronic or acute loss of Id1 leads to a down-regulation of Nanog, a critical regulator of self-renewal. In addition, in the absence of Id1, ES cells express elevated levels of Brachyury, a marker of mesendoderm differentiation. We find that loss of both Nanog and Id1 is required for the up-regulation of Brachyury, and ectopic Nanog expression in Id1(-/-) ES cells rescues the self-renewal defect, indicating that Nanog is the major downstream target of Id1. These results identify Id1 as a critical factor in the maintenance of ES cell self-renewal and suggest a plausible mechanism for its control of lineage commitment.


Assuntos
Células-Tronco Embrionárias/citologia , Proteínas Fetais/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteína 1 Inibidora de Diferenciação/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Meios de Cultura/metabolismo , Técnicas de Cultura Embrionária/métodos , Células-Tronco Embrionárias/metabolismo , Feminino , Proteínas Fetais/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Camundongos , Camundongos Knockout , Proteína Homeobox Nanog , Plasmídeos/genética , Plasmídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas com Domínio T/genética , Regulação para Cima
12.
Nat Biotechnol ; 26(1): 91-100, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18176556

RESUMO

Transcription factors are important targets for the treatment of a variety of malignancies but are extremely difficult to inhibit, as they are located in the cell's nucleus and act mainly by protein-DNA and protein-protein interactions. The transcriptional regulators Id1 and Id3 are attractive targets for cancer therapy as they are required for tumor invasiveness, metastasis and angiogenesis. We report here the development of an antitumor agent that downregulates Id1 effectively in tumor endothelial cells in vivo. Efficient delivery and substantial reduction of Id1 protein levels in the tumor endothelium were effected by fusing an antisense molecule to a peptide known to home specifically to tumor neovessels. In two different tumor models, systemic delivery of this drug led to enhanced hemorrhage, hypoxia and inhibition of primary tumor growth and metastasis, similar to what is observed in Id1 knockout mice. Combination with the Hsp90 inhibitor 17-(allylamino)-17-demethoxygeldanamycin yielded virtually complete growth suppression of aggressive breast tumors.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Marcação de Genes/métodos , Proteínas de Neoplasias/genética , Oligonucleotídeos Antissenso/genética , Peptídeos/genética , Fatores de Transcrição/genética , Animais , Linhagem Celular Tumoral , Camundongos , Oligonucleotídeos Antissenso/administração & dosagem , Peptídeos/administração & dosagem
13.
Cancer Res ; 66(22): 10870-7, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17108123

RESUMO

Id proteins are a class of dominant-negative antagonists of helix-loop-helix transcription factors and have been shown to control differentiation of a variety of cell types in diverse organisms. Although the importance of Id1 in tumor endothelial cells is well established, the expression and role of the Id1 protein in human cancer cells is controversial. To explore this issue, we developed and characterized a highly specific rabbit monoclonal antibody against Id1 to assess its expression in human breast, prostate, and bladder malignancies. Our results show that in usual types of human mammary carcinomas, the Id1 protein is expressed exclusively in the endothelium. Interestingly, we detected nuclear expression of the Id1 protein in the tumor cells in 10 of 45 cases of poorly differentiated and highly aggressive carcinoma with metaplastic morphology. Similarly, only 1 of 30 prostate cancer samples showed Id1-positive tumor cells, whereas in almost all, endothelial cells showed high Id1 expression. Intriguingly, whereas normal prostate glands do not show any Id1 protein expression, basal layer cells of benign prostate glands in proximity to tumors expressed high levels of the Id1 protein. In contrast to the lack of Id1 expression in the usual types of mammary and prostate cancers, the majority of transitional cell bladder tumors showed Id1 protein expression in both tumor and endothelial cells. These results suggest that further refinement of Id1 expression patterns in a variety of tumor types will be necessary to identify and study the functional roles played by Id1 in human neoplastic processes.


Assuntos
Proteína 1 Inibidora de Diferenciação/biossíntese , Neoplasias/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Células Endoteliais/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , Imuno-Histoquímica , Proteína 1 Inibidora de Diferenciação/imunologia , Masculino , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Neoplasias/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Coelhos , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia
14.
Gene ; 283(1-2): 117-24, 2002 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-11867218

RESUMO

T-box (Tbx) genes represent a phylogenetically conserved family of transcription factors that play important roles during embryonic development. Tbx family members have been shown to either activate or inhibit gene expression. However, little is known about the domains within Tbx proteins responsible for mediating gene transcription. While Tbx2 is known to repress gene expression, the domain(s) within Tbx2 remains poorly defined. Deletion of the carboxy-terminus of Tbx2, which contains a domain that is highly conserved with Tbx3 and ET, which has been demonstrated to contain a repression domain, only minimally diminishes the ability of Tbx2 to repress gene expression. However, in combination with the carboxy-terminal truncation, deletion of the amino acids located amino-terminal to the T-box abolished the ability of Tbx2 to repress gene expression. Both of these domains were capable of repressing gene expression when linked to the GAL4 DNA binding domain. In contrast to these two repression domains, the T-box was capable of weakly activating gene expression depending on the promoter context. Deletion analysis of the T-box suggests that this activation domain is located in the amino-terminal end of the T-box. These results reveal a novel transcription repression domain, confirm the presence of a previously implicated domain, and suggest a novel role for the T-box. Taken together, these results provide the basis for understanding the molecular mechanism whereby Tbx2 regulates gene expression and subsequently controls embryonic development.


Assuntos
Proteínas com Domínio T/fisiologia , Transcrição Gênica/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Linhagem Celular , Regulação da Expressão Gênica , Genes Reporter/genética , Vetores Genéticos/genética , Genótipo , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Proteínas com Domínio T/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...