Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Mol Sci ; 23(4)2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35216266

RESUMO

BRIL (bone restricted ifitm-like; also known as IFITM5) is a transmembrane protein expressed in osteoblasts. Although its role in skeletal development and homeostasis is unknown, mutations in BRIL result in rare dominant forms of osteogenesis imperfecta. The pathogenic mechanism has been proposed to be a gain-of or neomorphic function. To understand the function of BRIL and its OI type V mutant (MALEP BRIL) and whether they could activate signaling pathways in osteoblasts, we performed a luciferase reporter assay screen based on the activity of 26 transcription factors. When overexpressed in MC3T3-E1 and MLO-A5 cells, the MALEP BRIL activated the reporters dependent on MEF2, NFATc, and NR4A significantly more. Additional co-transfection experiments with MEF2C and NFATc1 and a number of their modulators (HDAC4, calcineurin, RCAN, FK506) confirmed the additive or synergistic activation of the pathways by MALEP, and suggested a coordinated regulation involving calcineurin. Endogenous levels of Nr4a members, as well as Ptgs2, were upregulated by MALEP BRIL. Y2H and co-immunoprecipitation indicated that BRIL interacted with CAML, but its contribution as the most upstream stimulator of the Ca2+-calcineurin-MEF2/NFATc cascade was not confirmed convincingly. Altogether the data presented provide the first ever readout to monitor for BRIL activity and suggest a potential gain-of-function causative effect for MALEP BRIL in OI type V, leading to perturbed signaling events and gene expression.


Assuntos
Proteínas de Membrana/genética , Mutação/genética , Fatores de Transcrição NFATC/genética , Osteoblastos/metabolismo , Osteogênese Imperfeita/genética , Ativação Transcricional/genética , Células 3T3 , Animais , Calcineurina/genética , Cálcio/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Camundongos , Osteogênese Imperfeita/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética
2.
J Biol Chem ; 288(19): 13278-94, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23530031

RESUMO

BACKGROUND: BRIL is a bone-specific membrane protein that is involved in osteogenesis imperfecta type V. RESULTS: Bril transcription is activated by Sp1, Sp3, OSX, and GLI2 and by CpG demethylation. CONCLUSION: Regulation of Bril involves trans-acting factors integrating at conserved promoter elements and epigenetic modifications. SIGNIFICANCE: Identification of the mechanisms governing Bril transcription is important to understand its role in skeletal biology. Bril encodes a small membrane protein present in osteoblasts. In humans, a single recurrent mutation in the 5'-UTR of BRIL causes osteogenesis imperfecta type V. The exact function of BRIL and the mechanism by which it contributes to disease are still unknown. The goal of the current study was to characterize the mechanisms governing Bril transcription in humans, rats, and mice. In the three species, as detected by luciferase reporter assays in UMR106 cells, we found that most of the base-line regulatory activity was localized within ∼250 bp upstream of the coding ATG. Co-transfection experiments indicated that Sp1 and Sp3 were potent inducers of the promoter activity, through the binding of several GC-rich boxes. Osterix was a weak activator but acted cooperatively with Sp1 and GLI2 to synergistically induce the BRIL promoter. GLI2, a mediator of hedgehog signaling pathway, was also a potent activator of BRIL through a single GLI binding site. Correspondingly, agonists of the hedgehog pathway (purmorphamine and Indian hedgehog) in MC3T3 osteoblasts led to increased BRIL levels. The BRIL promoter activity was also found to be negatively modulated through two different mechanisms. First, the ZFP354C zinc finger protein repressed basal and Sp1-induced activity. Second, CpG methylation of the promoter region correlated with an inactive state and prevented Sp1 activation. The data provide the very first analyses of the cis- and trans-acting factors regulating Bril transcription. They revealed key roles for the Sp members and GLI2 that possibly cooperate to activate Bril when the promoter becomes demethylated.


Assuntos
Metilação de DNA , Inativação Gênica , Proteínas de Membrana/genética , Transcrição Gênica , Ativação Transcricional , Células 3T3 , Animais , Sequência de Bases , Osso e Ossos/metabolismo , Diferenciação Celular , Sequência Conservada , Ilhas de CpG , Feminino , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Anotação de Sequência Molecular , Dados de Sequência Molecular , Proteínas Nucleares/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica , Ratos , Proteínas Repressoras/metabolismo , Transdução de Sinais , Fator de Transcrição Sp1/fisiologia , TATA Box , Proteína Gli2 com Dedos de Zinco
3.
Bone ; 154: 116218, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34571201

RESUMO

Physical forces are critical for successful function of many organs including bone. Interestingly, the timing of exercise during the day alters physiology and gene expression in many organs due to circadian rhythms. Circadian clocks in tissues, such as bone, express circadian clock genes that target tissue-specific genes, resulting in tissue-specific rhythmic gene expression (clock-controlled genes). We hypothesized that the adaptive response of bone to mechanical loading is regulated by circadian rhythms. First, mice were sham loaded and sacrificed 8 h later, which amounted to tissues being collected at zeitgeber time (ZT)2, 6, 10, 14, 18, and 22. Cortical bone of the tibiae collected from these mice displayed diurnal expression of core clock genes and key osteocyte and osteoblast-related genes, such as the Wnt-signaling inhibitors Sost and Dkk1, indicating these are clock-controlled genes. Serum bone turnover markers did not display rhythmicity. Second, mice underwent a single bout of in vivo loading at either ZT2 or ZT14 and were sacrificed 1, 8, or 24 h after loading. Loading at ZT2 resulted in Sost upregulation, while loading at ZT14 led to Sost and Dkk1 downregulation. Third, mice underwent daily in vivo tibial loading over 2 weeks administered either in the morning, (ZT2, resting phase) or evening (ZT14, active phase). In vivo microCT was performed at days 0, 5, 10, and 15 and conventional histomorphometry was performed at day 15. All outcome measures indicated a robust response to loading, but only microCT-based time-lapse morphometry showed that loading at ZT14 resulted in a greater endocortical bone formation response compared to mice loaded at ZT2. The decreased Sost and Dkk1 expression coincident with the modest, but significant time-of-day specific increase in adaptive bone formation, suggests that circadian clocks influence bone mechanoresponse.


Assuntos
Relógios Circadianos , Ritmo Circadiano , Animais , Osso e Ossos , Relógios Circadianos/genética , Ritmo Circadiano/fisiologia , Osso Cortical , Camundongos , Osteócitos , Osteogênese/fisiologia
4.
Sci Rep ; 10(1): 7790, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32385306

RESUMO

Snorc (Small NOvel Rich in Cartilage) has been identified as a chondrocyte-specific gene in the mouse. Yet little is known about the SNORC protein biochemical properties, and mechanistically how the gene is regulated transcriptionally in a tissue-specific manner. The goals of the present study were to shed light on those important aspects. The chondrocyte nature of Snorc expression was confirmed in mouse and rat tissues, in differentiated (day 7) ATDC5, and in RCS cells where it was constitutive. Topological mapping and biochemical analysis brought experimental evidences that SNORC is a type I protein carrying a chondroitin sulfate (CS) attached to serine 44. The anomalous migration of SNORC on SDS-PAGE was due to its primary polypeptide features, suggesting no additional post-translational modifications apart from the CS glycosaminoglycan. A highly conserved SOX9-binding enhancer located in intron 1 was necessary to drive transcription of Snorc in the mouse, rat, and human. The enhancer was active independently of orientation and whether located in a heterologous promoter or intron. Crispr-mediated inactivation of the enhancer in RCS cells caused reduction of Snorc. Transgenic mice carrying the intronic multimerized enhancer drove high expression of a ßGeo reporter in chondrocytes, but not in the hypertrophic zone. Altogether these data confirmed the chondrocyte-specific nature of Snorc and revealed dependency on the intronic enhancer binding of SOX9 for transcription.


Assuntos
Condrócitos/metabolismo , Regulação da Expressão Gênica , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteoglicanas/genética , Proteoglicanas/metabolismo , Fatores de Transcrição SOX9/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Células Cultivadas , Sulfatos de Condroitina/metabolismo , Humanos , Proteínas de Membrana/química , Camundongos , Camundongos Transgênicos , Fases de Leitura Aberta , Ligação Proteica , Domínios Proteicos , Proteoglicanas/química , Ratos , Sítio de Iniciação de Transcrição , Transcrição Gênica
5.
Bone ; 107: 131-142, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29174564

RESUMO

Osteogenesis imperfecta (OI) type V is caused by an autosomal dominant mutation in the IFITM5 gene, also known as BRIL. The c.-14C>T mutation in the 5'UTR of BRIL creates a novel translational start site adding 5 residues (MALEP) in frame with the natural coding of BRIL. A neomorphic function has been proposed for the MALEP-BRIL but the mechanisms at play are still unknown. In order to further understand the effects of MALEP-BRIL in vivo, we generated a knockin (KI) mouse model having the exact genetic -14C>T replica of patients with OI type V. Live KI descendants were never obtained from 2 male mosaic founders. Skeletal staining with alizarin red/alcian blue and µCT imaging of KI embryos revealed striking skeletal anomalies such as hypomineralized skull, short and bent long bones, and frail and wavy ribs. Histology and histochemical labeling revealed that midshaft of long bones was filled with hypertrophic chondrocytes, lacked a defined primary ossification center with the absence of defined cortices. Gene expression monitoring at E15.5 and E17.5 showed no change in Osx but decreased Bril itself as well as other differentiated osteoblast markers (Ibsp, Bglap, Sost). However, upregulation of Ptgs2 and Nr4a3 suggested that a pro-inflammatory reaction was activated. Primary osteoblasts from KI calvaria showed delayed differentiation and mineralization, with decreased abundance of BRIL. However, the upregulation AdipoQ and Fabp4 in young cultures indicated a possible switch in fate towards adipogenesis. Altogether our data suggest that the low level expression of MALEP-BRIL in Osx+ mesenchymal progenitors blunted their further differentiation into mature osteoblasts, which may have resulted in part from an inflammatory response.


Assuntos
Modelos Animais de Doenças , Proteínas de Membrana/genética , Osteoblastos/patologia , Osteogênese Imperfeita/genética , Osteogênese Imperfeita/patologia , Animais , Animais Geneticamente Modificados , Sistemas CRISPR-Cas , Diferenciação Celular/genética , Edição de Genes/métodos , Técnicas de Introdução de Genes , Inflamação/genética , Inflamação/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Mutação , Osteoblastos/metabolismo , Osteogênese/genética
6.
PLoS One ; 12(9): e0184568, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28880886

RESUMO

BRIL (bone-restricted IFITM-like), is a short transmembrane protein expressed almost exclusively in osteoblasts. Although much is known about its bone-restricted gene expression pattern and protein biochemical and topological features, little information is available for BRIL physiological function. Two autosomal dominant forms of osteogenesis imperfecta (OI) are caused by distinct, but recurrent mutations in the BRIL gene. Yet, the underlying mechanisms by which those mutations lead to OI are still poorly understood. A previous report indicated that BRIL knockout (KO) mice had bone deformities, shortened long bones, and reproductive problems. Here we generated and systematically analyzed the skeletal phenotype of a new global Bril KO/LacZ knockin mouse model. KO mice reproduced and thrived normally up to 12 month of age. The skeletal phenotype of KO and WT littermates was assessed at embryonic (E13.5 to E18.5) and postnatal (2 days, 3 weeks, 3 months and 8 months) time-points. Embryos from E13.5 through to E18.5 showed significant X-Gal staining in all skeletal elements without any apparent patterning anomalies. Although bone deformities were never observed at any postnatal ages, minor and transient differences were noted in terms of bone length and static uCT parameters, but not systematically across all ages and genders. These changes, however, were not accompanied by significant alteration in bone material properties as assessed by a 3-point bending test. In addition, no changes were detected in circulating serum markers of bone turnover (P1NP, CTX-I, and osteocalcin). Gene expression monitoring also revealed no major impact of the loss of BRIL. Further, when mice were challenged with a surgically-induced fracture in tibia, bones repaired equally well in the KO mice as compared to WT. Finally, we showed that BRIL C-terminus is not a bona fide binding site for calcium. In conclusion, our in depth analysis suggest that skeletal patterning, bone mass accrual and remodeling in mice proceeded independent of BRIL.


Assuntos
Proteínas de Membrana/metabolismo , Osteoblastos/metabolismo , Animais , Desenvolvimento Ósseo/genética , Desenvolvimento Ósseo/fisiologia , Cálcio/metabolismo , Feminino , Homeostase/genética , Homeostase/fisiologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Osteogênese/genética , Osteogênese/fisiologia , Osteogênese Imperfeita/genética , Osteogênese Imperfeita/metabolismo , Gravidez , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Nucleic Acids Res ; 30(19): 4285-94, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12364608

RESUMO

We have developed a functional genomics tool to identify the subset of cDNAs encoding secreted and membrane-bound proteins within a library (the 'secretome'). A Sindbis virus replicon was engineered such that the envelope protein precursor no longer enters the secretory pathway. cDNA fragments were fused to the mutant precursor and expression screened for their ability to restore membrane localization of envelope proteins. In this way, recombinant replicons were released within infectious viral particles only if the cDNA fragment they contain encodes a secretory signal. By using engineered viral replicons to selectively export cDNAs of interest in the culture medium, the methodology reported here efficiently filters genetic information in mammalian cells without the need to select individual clones. This adaptation of the 'signal trap' strategy is highly sensitive (1/200 000) and efficient. Indeed, of the 2546 inserts that were retrieved after screening various libraries, more than 97% contained a putative signal peptide. These 2473 clones encoded 419 unique cDNAs, of which 77% were previously annotated. Of the 94 cDNAs encoding proteins of unknown function, 24% either had no match in databases or contained a secretory signal that could not be predicted from electronic data.


Assuntos
Proteínas de Membrana/genética , Sindbis virus/genética , Animais , Linhagem Celular , DNA Complementar/genética , Biblioteca Gênica , Engenharia Genética , Genoma Viral , Proteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Sinais Direcionadores de Proteínas/genética , Replicon/genética , Sindbis virus/crescimento & desenvolvimento , Transfecção
8.
Gene ; 334: 123-31, 2004 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-15256262

RESUMO

Terminal differentiation of keratinocytes results in the formation of a cornified layer composed of cross-linked intracellular and extracellular material. Using a signal trap expression screening strategy, we have identified four cDNAs encoding secreted proteins potentially involved in this process. One of the cDNAs is identical to the short isoform of suprabasin, a recently described epidermis-specific protein, which is shown here to contain a functional secretory signal. The second cDNA, sk89, encodes a protein of 493 amino acids, rich in glycine and serine residues. The third cDNA encodes a C-terminal fragment of SK89 (amino acids 410-493). It comprises exons 13 to 18 of the sk89 locus but transcription starts at an isoform-specific exon encoding a distinct secretory signal. The fourth cDNA encodes keratinocyte differentiation-associated protein (KDAP), a precursor protein of 102 amino acids. Subcellular localization by immunofluorescence and detection of the tagged proteins by Western blotting confirmed that the four proteins are secreted. Northern analysis and in situ hybridization revealed that expression of the corresponding genes was restricted to the suprabasal keratinocytes of the epidermis. These genes encoding epidermis-specific secreted products are found in a conserved cluster on human chromosome 19q13.12 and on mouse chromosome 7A3.


Assuntos
Antígenos de Diferenciação/genética , Ácido Aspártico Endopeptidases/genética , Família Multigênica/genética , Proteínas/genética , Pele/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Diferenciação/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Sequência de Bases , Western Blotting , Linhagem Celular , Mapeamento Cromossômico/métodos , Cromossomos Humanos Par 19/genética , Cromossomos de Mamíferos/genética , Clonagem Molecular , Sequência Conservada/genética , DNA Complementar/química , DNA Complementar/genética , Éxons , Perfilação da Expressão Gênica , Ordem dos Genes , Genes/genética , Proteínas de Fluorescência Verde , Humanos , Íntrons , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Análise de Sequência de DNA , Transfecção
9.
J Bone Miner Res ; 29(9): 2004-16, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24715519

RESUMO

BRIL/IFITM5 is a membrane protein present almost exclusively in osteoblasts, which is believed to adopt a type III (N-out/C-out) topology. Mutations in IFITM5 cause OI type V, but the characteristics of the mutant protein and the mechanism involved are still unknown. The purpose of the current study was to re-assess the topology, localization, and biochemical properties of BRIL and compare it to the OI type V mutant in MC3T3 osteoblasts. Immunofluorescence labeling was performed with antibodies directed against BRIL N- or C-terminus. In intact cells, BRIL labeling was conspicuously detected at the plasma membrane only with the anti-C antibody. Detection of BRIL N-terminus was only possible after cell permeabilization, revealing both plasma membrane and Golgi labeling. Trypsinization of live cells expressing BRIL only cleaved off the C-terminus, confirming that it is a type II protein and that its N-terminus is intracellular. A truncated form of BRIL lacking the last 18 residues did not appear to affect localization, whereas mutation of a single leucine to arginine within the transmembrane segment abolished plasma membrane targeting. BRIL is first targeted to the endoplasmic reticulum as the entry point to the secretory pathway and rapidly traffics to the Golgi via a COPII-dependent pathway. BRIL was found to be palmitoylated and two conserved cysteine residues (C52 and C53) were critical for targeting to the plasma membrane. The OI type V mutant BRIL, having a five residue extension (MALEP) at its N-terminus, presented with exactly the same topological and biochemical characteristics as wild type BRIL. In contrast, the S42 > L mutant BRIL was trapped intracellularly in the Golgi. BRIL proteins and transcripts were equally detected in bone from a patient with OI type V, suggesting that the cause of the disease is a gain of function mediated by a faulty intracellular activity of the mutant BRIL.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas Mutantes/metabolismo , Osteogênese Imperfeita/genética , Sequência de Aminoácidos , Animais , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Membrana Celular/metabolismo , Cisteína/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Lipoilação , Proteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/química , Mutação/genética , Estabilidade Proteica , Transporte Proteico , Relação Estrutura-Atividade
10.
J Bone Miner Res ; 23(9): 1497-508, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18442316

RESUMO

In the course of attempting to define the bone "secretome" using a signal-trap screening approach, we identified a gene encoding a small membrane protein novel to osteoblasts. Although previously identified in silico as ifitm5, no localization or functional studies had been undertaken on this gene. We characterized the expression patterns and localization of this gene in vitro and in vivo and assessed its role in matrix mineralization in vitro. The bone specificity and shown role in mineralization led us to rename the gene bone restricted ifitm-like protein (Bril). Bril encodes a 14.8-kDa 134 amino acid protein with two transmembrane domains. Northern blot analysis showed bone-specific expression with no expression in other embryonic or adult tissues. In situ hybridization and immunohistochemistry in mouse embryos showed expression localized on the developing bone. Screening of cell lines showed Bril expression to be highest in osteoblasts, associated with the onset of matrix maturation/mineralization, suggesting a role in bone formation. Functional evidence of a role in mineralization was shown by adenovirus-mediated Bril overexpression and lentivirus-mediated Bril shRNA knockdown in vitro. Elevated Bril resulted in dose-dependent increases in mineralization in UMR106 and rat primary osteoblasts. Conversely, knockdown of Bril in MC3T3 osteoblasts resulted in reduced mineralization. Thus, we identified Bril as a novel osteoblast protein and showed a role in mineralization, possibly identifying a new regulatory pathway in bone formation.


Assuntos
Osso e Ossos/metabolismo , Calcificação Fisiológica , Proteínas de Membrana/metabolismo , Proteínas/metabolismo , Adenoviridae , Sequência de Aminoácidos , Animais , Biomarcadores/metabolismo , Western Blotting , Linhagem Celular , Clonagem Molecular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Imunofluorescência , Perfilação da Expressão Gênica , Humanos , Hibridização In Situ , Lentivirus , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Osteoblastos/citologia , Osteoblastos/metabolismo , Proteínas/química , Proteínas/genética , Ratos , Alinhamento de Sequência
11.
J Biol Chem ; 278(50): 50563-71, 2003 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-14523025

RESUMO

Although a number of secreted factors have been demonstrated to be bone regulators, none of these are unique to bone. Using a viral-based signal-trap strategy we have identified a novel gene we have termed "osteocrin." A 1280-bp mRNA encodes osteocrin producing a mature protein of 103 amino acids with a molecular mass of 11.4 kDa. Osteocrin shows no homology with any known gene except for two conserved sequence motifs reminiscent of dibasic cleavage sites found in peptide hormone precursors. Immunofluorescence and Western blot analysis confirmed the secretory nature of osteocrin. Two protein species were identified in the medium of cells overexpressing osteocrin, a full-length 11.4 kDa species and a processed approximately 5 kDa species. Mutation of the 76KKKR79 dibasic cleavage site abolished the appearance of this smaller osteocrin fragment. By in situ hybridization in mouse embryos, osteocrin was expressed specifically in Cbfa-1-positive, osteocalcin-negative osteoblasts. Immunohistochemistry on adult mouse bone showed osteocrin localization in osteoblasts and young osteocytes. By Northern blot analysis, osteocrin expression was only detected in bone, expression peaking just after birth and decreasing markedly with age. In primary osteoblastic cell cultures osteocrin expression coincided with matrix formation then decreased in very mature cultures. Treatment of cultures with 1,25-dihydroxyvitamin D3 resulted in a rapid dose-dependent down-regulation of osteocrin expression, suggesting direct regulation. Chronic treatment of primary cultures with osteocrin-conditioned media inhibited mineralization and reduced osteocalcin and alkaline phosphatase expression. These results suggest that osteocrin represents a novel, unique vitamin D-regulated bone-specific protein that appears to act as a soluble osteoblast regulator.


Assuntos
Osteoblastos/metabolismo , Proteínas/química , Proteínas/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Northern Blotting , Western Blotting , Calcitriol/farmacologia , Clonagem Molecular , Meios de Cultivo Condicionados/farmacologia , Primers do DNA/farmacologia , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Microscopia de Fluorescência , Modelos Genéticos , Dados de Sequência Molecular , Proteínas Musculares , Mutação , Fenótipo , Proteínas/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Fatores de Transcrição , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA