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












Base de datos
Intervalo de año de publicación
1.
Metabolism ; 137: 155331, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36228741

RESUMEN

BACKGROUND: The triglyceride (TG) transfer activity of microsomal triglyceride transfer protein (MTP) is essential for lipoprotein assembly in the liver and intestine; however, its function in adipose tissue, which does not assemble lipoproteins, is unknown. Here we have elucidated the function of MTP in adipocytes. APPROACH AND RESULTS: We demonstrated that MTP is present on lipid droplets in human adipocytes. Adipose-specific MTP deficient (A-Mttp-/-) male and female mice fed an obesogenic diet gained less weight than Mttpf/f mice, had less fat mass, smaller adipocytes and were insulin sensitive. A-Mttp-/- mice showed higher energy expenditure than Mttpf/f mice. During a cold challenge, A-Mttp-/- mice maintained higher body temperature by mobilizing more fatty acids. Biochemical studies indicated that MTP deficiency de-repressed adipose triglyceride lipase (ATGL) activity and increased TG lipolysis. Both wild type MTP and mutant MTP deficient in TG transfer activity interacted with and inhibited ATGL activity. Thus, the TG transfer activity of MTP is not required for ATGL inhibition. C-terminally truncated ATGL that retains its lipase activity interacted less efficiently than full-length ATGL. CONCLUSION: Our findings demonstrate that adipose-specific MTP deficiency increases ATGL-mediated TG lipolysis and enhances energy expenditure, thereby resisting diet-induced obesity. We speculate that the regulatory function of MTP involving protein-protein interactions might have evolved before the acquisition of TG transfer activity in vertebrates. Adipose-specific inhibition of MTP-ATGL interactions may ameliorate obesity while avoiding the adverse effects associated with inhibition of the lipid transfer activity of MTP.


Asunto(s)
Lipasa , Lipólisis , Animales , Femenino , Humanos , Masculino , Ratones , Adipocitos/metabolismo , Lipasa/metabolismo , Lípidos/farmacología , Obesidad/metabolismo
2.
Int J STEM Educ ; 8(1): 3, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33532174

RESUMEN

BACKGROUND: Transforming the culture of STEM higher education to be more inclusive and help more students reach STEM careers is challenging. Herein, we describe a new model for STEM higher education transformation, the Sustainable, Transformative Engagement across a Multi-Institution/Multidisciplinary STEM, (STEM)2, "STEM-squared", Network. The Network embraces a pathways model, as opposed to a pipeline model, to STEM career entry. It is founded upon three strong theoretical frameworks: Communities of Transformation, systems design for organizational change, and emergent outcomes for the diffusion of innovations in STEM education. Currently composed of five institutions-three private 4-year universities and two public community colleges-the Network capitalizes on the close geographic proximity and shared student demographics to effect change across the classroom, disciplinary, institutional, and inter-institutional levels. RESULTS: The (STEM)2 Network has increased the extent to which participants feel empowered to be change agents for STEM higher education reform and has increased collaboration across disciplines and institutions. Participants were motivated to join the Network to improve STEM education, to improve the transfer student experience, to collaborate with colleagues across disciplines and institutions, and because they respected the leadership team. Participants continue to engage in the Network because of the collaborations created, opportunities for professional growth, opportunities to improve STEM education, and a sense that the Network is functioning as intended. CONCLUSION: The goal to increase the number and diversity of people entering STEM careers is predicated on transforming the STEM higher education system to embrace a pathways model to a STEM career. The (STEM)2 Network is achieving this by empowering faculty to transform the system from the inside. While the systemic transformation of STEM higher education is challenging, the (STEM)2 Network directly addresses those challenges by bridging disciplinary and institutional silos and leveraging the reward structure of the current system to support faculty as they work to transform this very system. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s40594-020-00262-z.

3.
G3 (Bethesda) ; 10(5): 1599-1612, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32152006

RESUMEN

The endosymbiotic theory proposes that eukaryotes evolved from the symbiotic relationship between anaerobic (host) and aerobic prokaryotes. Through iterative genetic transfers, the mitochondrial and nuclear genomes coevolved, establishing the mitochondria as the hub of oxidative metabolism. To study this coevolution, we disrupt mitochondrial-nuclear epistatic interactions by using strains that have mitochondrial DNA (mtDNA) and nuclear DNA (nDNA) from evolutionarily divergent species. We undertake a multifaceted approach generating introgressed Drosophila strains containing D. simulans mtDNA and D. melanogaster nDNA with Sirtuin 4 (Sirt4)-knockouts. Sirt4 is a nuclear-encoded enzyme that functions, exclusively within the mitochondria, as a master regulator of oxidative metabolism. We exposed flies to the drug rapamycin in order to eliminate TOR signaling, thereby compromising the cytoplasmic crosstalk between the mitochondria and nucleus. Our results indicate that D. simulans and D. melanogaster mtDNA haplotypes display opposite Sirt4-mediated phenotypes in the regulation of whole-fly oxygen consumption. Moreover, our data reflect that the deletion of Sirt4 rescued the metabolic response to rapamycin among the introgressed strains. We propose that Sirt4 is a suitable candidate for studying the properties of mitochondrial-nuclear epistasis in modulating mitochondrial metabolism.


Asunto(s)
ADN Mitocondrial , Drosophila , Estrés Oxidativo , Sirolimus/farmacología , Sirtuinas/genética , Animales , ADN Mitocondrial/genética , Drosophila/efectos de los fármacos , Drosophila/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Haplotipos , Mitocondrias/genética , Fenotipo
4.
Heliyon ; 5(2): e01217, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30805566

RESUMEN

Drosophila melanogaster is a powerful model to study mitochondrial respiratory chain defects, particularly succinate dehydrogenase (SDH) deficiency. Mutations in sdh genes cause degenerative disorders and often lead to death. Therapies for such pathologies are based on a combination of vitamins and dietary supplements, and are rarely effective. In Drosophila, mutations in several of the genes encoding SDH resemble the pathology of SDH deficiency in humans, enabling the Drosophila model to be used in finding treatments for this condition. Here we show that exposure to the drug rapamycin improves the survival of sdh mutant strains, the activity of SDH and the impaired climbing associated with sdh mutations. However, the production of reactive oxygen species, the oxygen consumption of isolated mitochondria and the resistance to hyperoxia were minimally affected. Our results contribute to the current research seeking a treatment for mitochondrial disease.

5.
J Vis Exp ; (103)2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26485391

RESUMEN

Since mitochondria play roles in amino acid metabolism, carbohydrate metabolism and fatty acid oxidation, defects in mitochondrial function often compromise the lives of those who suffer from these complex diseases. Detecting mitochondrial metabolic changes is vital to the understanding of mitochondrial disorders and mitochondrial responses to pharmacological agents. Although mitochondrial metabolism is at the core of metabolic regulation, the detection of subtle changes in mitochondrial metabolism may be hindered by the overrepresentation of other cytosolic metabolites obtained using whole organism or whole tissue extractions. Here we describe an isolation method that detected pronounced mitochondrial metabolic changes in Drosophila that were distinct between whole-fly and mitochondrial enriched preparations. To illustrate the sensitivity of this method, we used a set of Drosophila harboring genetically diverse mitochondrial DNAs (mtDNA) and exposed them to the drug rapamycin. Using this method we showed that rapamycin modifies mitochondrial metabolism in a mitochondrial-genotype-dependent manner. However, these changes are much more distinct in metabolomics studies when metabolites were extracted from mitochondrial enriched fractions. In contrast, whole tissue extracts only detected metabolic changes mediated by the drug rapamycin independently of mtDNAs.


Asunto(s)
Drosophila melanogaster/ultraestructura , Mitocondrias/metabolismo , Animales , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Mitocondrias/genética , Oxidación-Reducción
6.
Dis Model Mech ; 8(8): 843-54, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26035388

RESUMEN

Communication between the mitochondrial and nuclear genomes is vital for cellular function. The assembly of mitochondrial enzyme complexes, which produce the majority of cellular energy, requires the coordinated expression and translation of both mitochondrially and nuclear-encoded proteins. The joint genetic architecture of this system complicates the basis of mitochondrial diseases, and mutations both in mitochondrial DNA (mtDNA)- and nuclear-encoded genes have been implicated in mitochondrial dysfunction. Previously, in a set of mitochondrial-nuclear introgression strains, we characterized a dual genome epistasis in which a naturally occurring mutation in the Drosophila simulans simw(501) mtDNA-encoded transfer RNA (tRNA) for tyrosine (tRNA(Tyr)) interacts with a mutation in the nuclear-encoded mitochondrially localized tyrosyl-tRNA synthetase from Drosophila melanogaster. Here, we show that the incompatible mitochondrial-nuclear combination results in locomotor defects, reduced mitochondrial respiratory capacity, decreased oxidative phosphorylation (OXPHOS) enzyme activity and severe alterations in mitochondrial morphology. Transgenic rescue strains containing nuclear variants of the tyrosyl-tRNA synthetase are sufficient to rescue many of the deleterious phenotypes identified when paired with the simw(501) mtDNA. However, the severity of this defective mito-nuclear interaction varies across traits and genetic backgrounds, suggesting that the impact of mitochondrial dysfunction might be tissue specific. Because mutations in mitochondrial tRNA(Tyr) are associated with exercise intolerance in humans, this mitochondrial-nuclear introgression model in Drosophila provides a means to dissect the molecular basis of these, and other, mitochondrial diseases that are a consequence of the joint genetic architecture of mitochondrial function.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Núcleo Celular/metabolismo , Drosophila melanogaster/metabolismo , Enfermedades Mitocondriales/metabolismo , ARN de Transferencia/metabolismo , Estructuras Animales/anatomía & histología , Animales , Animales Modificados Genéticamente , Respiración de la Célula , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Epistasis Genética , Vuelo Animal , Genotipo , Mitocondrias Musculares/ultraestructura , Actividad Motora , Músculos/ultraestructura , Fosforilación Oxidativa , Iniciación de la Cadena Peptídica Traduccional , Tirosina/metabolismo
7.
J Cell Sci ; 127(Pt 10): 2282-90, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24610944

RESUMEN

Downregulation of the mammalian target of rapamycin (mTOR) pathway by its inhibitor rapamycin is emerging as a potential pharmacological intervention that mimics the beneficial effects of dietary restriction. Modulation of mTOR has diverse effects on mitochondrial metabolism and biogenesis, but the role of the mitochondrial genotype in mediating these effects remains unknown. Here, we use novel mitochondrial genome replacement strains in Drosophila to test the hypothesis that genes encoded in mitochondrial DNA (mtDNA) influence the mTOR pathway. We show that rapamycin increases mitochondrial respiration and succinate dehydrogenase activity, decreases H2O2 production and generates distinct shifts in the metabolite profiles of isolated mitochondria versus whole Drosophila. These effects are disabled when divergent mitochondrial genomes from D. simulans are placed into a common nuclear background, demonstrating that the benefits of rapamycin to mitochondrial metabolism depend on genes encoded in the mtDNA. Rapamycin is able to enhance mitochondrial respiration when succinate dehydrogenase activity is blocked, suggesting that the beneficial effects of rapamycin on these two processes are independent. Overall, this study provides the first evidence for a link between mitochondrial genotype and the effects of rapamycin on mitochondrial metabolic pathways.


Asunto(s)
ADN Mitocondrial/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Sirolimus/farmacología , Animales , ADN Mitocondrial/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Oxidación-Reducción , Serina-Treonina Quinasas TOR/metabolismo
8.
IOSR J Pharm ; 4(8): 43-51, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28593179

RESUMEN

Rapamycin treatment is considered a pharmacological intervention with the potential to mimic the longevity benefits of dietary manipulations. However, how rapamycin interacts with nutrition is not fully understood. Here we studied the effect of rapamycin on the longevity of Drosophila under a range of dietary conditions. In diets low in nutrients, rapamycin reduced longevity in a dosage-dependent manner. This dosage effect requires some nutrients as rapamycin has no impact on survival under starvation conditions. Under a balanced diet of yeast and sugar, rapamycin had no repeatable beneficial effect on organismal longevity. These results show that the effect of rapamycin on longevity is sensitive to the nutritional environment and it can reduce lifespan when nutrients are limited.

9.
PLoS One ; 6(12): e29513, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22242130

RESUMEN

Resveratrol is a plant-derived polyphenol that extends lifespan and healthspan in model organism. Despite extensive investigation, the biological processes mediating resveratrol's effects have yet to be elucidated. Because repression of translation shares many of resveratrol's beneficial effects, we hypothesized that resveratrol was a modulator of protein synthesis. We studied the effect of the drug on the H4-II-E rat hepatoma cell line. Initial studies showed that resveratrol inhibited global protein synthesis. Given the role of the mammalian Target of Rapamycin (mTOR) in regulating protein synthesis, we examined the effect of resveratrol on mTOR signaling. Resveratrol inhibited mTOR self-phosphorylation and the phosphorylation of mTOR targets S6K1 and eIF4E-BP1. It attenuated the formation of the translation initiation complex eIF4F and increased the phosphorylation of eIF2α. The latter event, also a mechanism for translation inhibition, was not recapitulated by mTOR inhibitors. The effects on mTOR signaling were independent of effects on AMP-activated kinase or AKT. We conclude that resveratrol is an inhibitor of global protein synthesis, and that this effect is mediated through modulation of mTOR-dependent and independent signaling.


Asunto(s)
Hígado/efectos de los fármacos , Hígado/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Estilbenos/farmacología , Proteínas Quinasas Activadas por AMP , Animales , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 4F Eucariótico de Iniciación/metabolismo , Hígado/enzimología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Estabilidad Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Resveratrol , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo
10.
PLoS One ; 5(7): e11521, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20634900

RESUMEN

One way animals may cope with nutrient deprivation is to broadly repress translation by inhibiting 5'-cap initiation. However, under these conditions specific proteins remain essential to survival during fasting. Such peptides may be translated through initiation at 5'UTR Internal Ribosome Entry Sites (IRES). Here we show that the Drosophila melanogaster Forkhead box type O (dFoxO) transcription factor is required for adult survival during fasting, and that the 5'UTR of dfoxO has the ability to initiate IRES-mediated translation in cell culture. Previous work has shown that insulin negatively regulates dFoxO through AKT-mediated phosphorylation while dFoxO itself induces transcription of the insulin receptor dInR, which also harbors IRES. Here we report that IRES-mediated translation of both dFoxO and dInR is activated in fasted Drosophila S2 cells at a time when cap-dependent translation is reduced. IRES mediated translation of dFoxO and dInR may be essential to ensure function and sensitivity of the insulin signaling pathway during fasting.


Asunto(s)
Regiones no Traducidas 5'/fisiología , Proteínas de Drosophila/metabolismo , Ayuno/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regiones no Traducidas 5'/genética , Animales , Northern Blotting , Western Blotting , Línea Celular , Drosophila , Proteínas de Drosophila/genética , Factores de Transcripción Forkhead/genética , Reacción en Cadena de la Polimerasa , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
11.
Proc Natl Acad Sci U S A ; 105(17): 6368-73, 2008 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-18434551

RESUMEN

Ablation of germ-line precursor cells in Caenorhabditis elegans extends lifespan by activating DAF-16, a forkhead transcription factor (FOXO) repressed by insulin/insulin-like growth factor (IGF) signaling (IIS). Signals from the gonad might thus regulate whole-organism aging by modulating IIS. To date, the details of this systemic regulation of aging by the reproductive system are not understood, and it is unknown whether such effects are evolutionarily conserved. Here we report that eliminating germ cells (GCs) in Drosophila melanogaster increases lifespan and modulates insulin signaling. Long-lived germ-line-less flies show increased production of Drosophila insulin-like peptides (dilps) and hypoglycemia but simultaneously exhibit several characteristics of IIS impedance, as indicated by up-regulation of the Drosophila FOXO (dFOXO) target genes 4E-BP and l (2)efl and the insulin/IGF-binding protein IMP-L2. These results suggest that signals from the gonad regulate lifespan and modulate insulin sensitivity in the fly and that the gonadal regulation of aging is evolutionarily conserved.


Asunto(s)
Drosophila melanogaster/metabolismo , Células Germinativas/metabolismo , Insulina/metabolismo , Longevidad , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Femenino , Regulación de la Expresión Génica , Genes de Insecto , Células Germinativas/citología , Masculino , Ovario/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Testículo/citología
12.
BMC Dev Biol ; 7: 106, 2007 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-17880703

RESUMEN

BACKGROUND: The organization of the different tissues of an animal requires mechanisms that regulate cell-cell adhesion to promote and maintain the physical separation of adjacent cell populations. In the Drosophila imaginal wing disc the iroquois homeobox genes are expressed in the notum anlage and contribute to the specification of notum identity. These genes are not expressed in the adjacent wing hinge territory. These territories are separated by an approximately straight boundary that in the mature disc is associated with an epithelial fold. The mechanism by which these two cell populations are kept separate is unclear. RESULTS: Here we show that the Iro-C genes participate in keeping the notum and wing cell populations separate. Indeed, within the notum anlage, cells not expressing Iro-C tend to join together and sort out from their Iro-C expressing neighbours. We also show that apposition of Iro-C expressing and non-expressing cells induces invagination and apico-basal shortening of the Iro-C- cells. This effect probably underlies formation of the fold that separates the notum and wing hinge territories. In addition, cells overexpressing a member of the Iro-C contact one another and become organized in a network of thin strings that surrounds and isolates large groups of non-overexpressing cells. The strings appear to exert a pulling force along their longitudinal axis. CONCLUSION: Apposition of cells expressing and non-expressing the Iro-C, as it occurs in the notum-wing hinge border of the Drosophila wing disc, influences cell behaviour. It leads to cell sorting, and cellular invagination and apical-basal shortening. These effects probably account for keeping the prospective notum and wing hinge cell populations separate and underlie epithelial fold formation. Cells that overexpress a member of the Iro-C and that confront non-expressing cells establish contacts between themselves and become organized in a network of thin strings. This is a complex and unique phenotype that might be important for the generation of a straight notum-wing hinge border.


Asunto(s)
Movimiento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo , Animales , Forma de la Célula , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Proteínas de Homeodominio/genética , Fenotipo , Alas de Animales/citología
13.
Development ; 132(18): 4087-96, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16093324

RESUMEN

During development, the imaginal wing disc of Drosophila is subdivided into territories separated by developmental boundaries. The best characterized boundaries delimit compartments defined by cell-lineage restrictions. Here, we analyze the formation of a boundary that does not rely on such restrictions, namely, that which separates the notum (body wall) and the wing hinge (appendage). It is known that the homeobox genes of the Iroquois complex (Iro-C) define the notum territory and that the distal limit of the Iro-C expression domain demarks the boundary between the notum and the wing hinge. However, it is unclear how this boundary is established and maintained. We now find that msh, a homeobox gene of the Msx family, is strongly expressed in the territory of the hinge contiguous to the Iro-C domain. Loss- and gain-of-function analyses show that msh maintains Iro-C repressed in the hinge, while Iro-C prevents high level expression of msh in the notum. Thus, a mutual repression between msh and Iro-C is essential to set the limit between the contiguous domains of expression of these genes and therefore to establish and/or maintain the boundary between body wall and wing. In addition, we find that msh is necessary for proper growth of the hinge territory and the differentiation of hinge structures. msh also participates in the patterning of the notum, where it is expressed at low levels.


Asunto(s)
Tipificación del Cuerpo/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/embriología , Epistasis Genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Alas de Animales/embriología , Animales , Clonación Molecular , Oligonucleótidos , beta-Galactosidasa
14.
Genetics ; 163(4): 1403-12, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12702684

RESUMEN

The Tufted(1) (Tft(1)) dominant mutation promotes the generation of ectopic bristles (macrochaetae) in the dorsal mesothorax of Drosophila. Here we show that Tft(1) corresponds to a gain-of-function allele of the proneural gene amos that is associated with a chromosomal aberration at 36F-37A. This causes ectopic expression of amos in large domains of the lateral-dorsal embryonic ectoderm, which results in supernumerary neurons of the PNS, and in the notum region of the third instar imaginal wing, which gives rise to the mesothoracic extra bristles. Revertants of Tft(1), which lack ectopic neurons and bristles, do not show ectopic expression of amos. One revertant is a loss-of-function allele of amos and has a recessive phenotype in the embryonic PNS. Our results suggest that both normal and ectopic Tft(1) bristles are generated following similar rules, and both are subjected to Notch-mediated lateral inhibition. The ability of Tft(1) bristles to appear close together may be due to amos having a stronger proneural capacity than that of other proneural genes like asense and scute. This ability might be related to the wild-type function of amos in promoting development of large clusters of closely spaced olfactory sensilla.


Asunto(s)
Drosophila/genética , Factores de Crecimiento Nervioso/genética , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila/anatomía & histología , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...