Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cardiovasc Toxicol ; 24(5): 435-471, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38555547

RESUMEN

Cigarette smoking is positively and robustly associated with cardiovascular disease (CVD), including hypertension, atherosclerosis, cardiac arrhythmias, stroke, thromboembolism, myocardial infarctions, and heart failure. However, after more than a decade of ENDS presence in the U.S. marketplace, uncertainty persists regarding the long-term health consequences of ENDS use for CVD. New approach methods (NAMs) in the field of toxicology are being developed to enhance rapid prediction of human health hazards. Recent technical advances can now consider impact of biological factors such as sex and race/ethnicity, permitting application of NAMs findings to health equity and environmental justice issues. This has been the case for hazard assessments of drugs and environmental chemicals in areas such as cardiovascular, respiratory, and developmental toxicity. Despite these advances, a shortage of widely accepted methodologies to predict the impact of ENDS use on human health slows the application of regulatory oversight and the protection of public health. Minimizing the time between the emergence of risk (e.g., ENDS use) and the administration of well-founded regulatory policy requires thoughtful consideration of the currently available sources of data, their applicability to the prediction of health outcomes, and whether these available data streams are enough to support an actionable decision. This challenge forms the basis of this white paper on how best to reveal potential toxicities of ENDS use in the human cardiovascular system-a primary target of conventional tobacco smoking. We identify current approaches used to evaluate the impacts of tobacco on cardiovascular health, in particular emerging techniques that replace, reduce, and refine slower and more costly animal models with NAMs platforms that can be applied to tobacco regulatory science. The limitations of these emerging platforms are addressed, and systems biology approaches to close the knowledge gap between traditional models and NAMs are proposed. It is hoped that these suggestions and their adoption within the greater scientific community will result in fresh data streams that will support and enhance the scientific evaluation and subsequent decision-making of tobacco regulatory agencies worldwide.


Asunto(s)
Enfermedades Cardiovasculares , Sistemas Electrónicos de Liberación de Nicotina , Vapeo , Humanos , Medición de Riesgo , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/prevención & control , Animales , Vapeo/efectos adversos , Vapeo/tendencias , Factores de Riesgo , Nicotina/efectos adversos , Nicotina/administración & dosificación , Agonistas Nicotínicos/efectos adversos , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/toxicidad , Seguridad de Productos para el Consumidor , Sistema Cardiovascular/efectos de los fármacos , Cardiotoxicidad , Factores de Riesgo de Enfermedad Cardiaca , Cigarrillo Electrónico a Vapor/efectos adversos
2.
Annu Rev Med ; 75: 353-367, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-37582489

RESUMEN

Cannabis, the most commonly used recreational drug, is illicit in many areas of the world. With increasing decriminalization and legalization, cannabis use is increasing in the United States and other countries. The adverse effects of cannabis are unclear because its status as a Schedule 1 drug in the United States restricts research. Despite a paucity of data, cannabis is commonly perceived as a benign or even beneficial drug. However, recent studies show that cannabis has adverse cardiovascular and pulmonary effects and is linked with malignancy. Moreover, case reports have shown an association between cannabis use and neuropsychiatric disorders. With growing availability, cannabis misuse by minors has led to increasing incidences of overdose and toxicity. Though difficult to detect, cannabis intoxication may be linked to impaired driving and motor vehicle accidents. Overall, cannabis use is on the rise, and adverse effects are becoming apparent in clinical data sets.


Asunto(s)
Cannabis , Sobredosis de Droga , Humanos , Cannabis/efectos adversos
4.
Cell ; 185(10): 1676-1693.e23, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35489334

RESUMEN

Epidemiological studies reveal that marijuana increases the risk of cardiovascular disease (CVD); however, little is known about the mechanism. Δ9-tetrahydrocannabinol (Δ9-THC), the psychoactive component of marijuana, binds to cannabinoid receptor 1 (CB1/CNR1) in the vasculature and is implicated in CVD. A UK Biobank analysis found that cannabis was an risk factor for CVD. We found that marijuana smoking activated inflammatory cytokines implicated in CVD. In silico virtual screening identified genistein, a soybean isoflavone, as a putative CB1 antagonist. Human-induced pluripotent stem cell-derived endothelial cells were used to model Δ9-THC-induced inflammation and oxidative stress via NF-κB signaling. Knockdown of the CB1 receptor with siRNA, CRISPR interference, and genistein attenuated the effects of Δ9-THC. In mice, genistein blocked Δ9-THC-induced endothelial dysfunction in wire myograph, reduced atherosclerotic plaque, and had minimal penetration of the central nervous system. Genistein is a CB1 antagonist that attenuates Δ9-THC-induced atherosclerosis.


Asunto(s)
Cannabis , Enfermedades Cardiovasculares , Alucinógenos , Analgésicos , Animales , Agonistas de Receptores de Cannabinoides/farmacología , Dronabinol/farmacología , Células Endoteliales , Genisteína/farmacología , Genisteína/uso terapéutico , Inflamación/tratamiento farmacológico , Ratones , Receptor Cannabinoide CB1 , Receptores de Cannabinoides
5.
JCI Insight ; 6(7)2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33830086

RESUMEN

Human pluripotent stem cells (PSCs), which are composed of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), provide an opportunity to advance cardiac cell therapy-based clinical trials. However, an important hurdle that must be overcome is the risk of teratoma formation after cell transplantation due to the proliferative capacity of residual undifferentiated PSCs in differentiation batches. To tackle this problem, we propose the use of a minimal noncardiotoxic doxorubicin dose as a purifying agent to selectively target rapidly proliferating stem cells for cell death, which will provide a purer population of terminally differentiated cardiomyocytes before cell transplantation. In this study, we determined an appropriate in vitro doxorubicin dose that (a) eliminates residual undifferentiated stem cells before cell injection to prevent teratoma formation after cell transplantation and (b) does not cause cardiotoxicity in ESC-derived cardiomyocytes (CMs) as demonstrated through contractility analysis, electrophysiology, topoisomerase activity assay, and quantification of reactive oxygen species generation. This study establishes a potentially novel method for tumorigenic-free cell therapy studies aimed at clinical applications of cardiac cell transplantation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Doxorrubicina/administración & dosificación , Células Madre Embrionarias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Células Madre Pluripotentes/citología , Animales , Apoptosis/efectos de los fármacos , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Células Madre Embrionarias/trasplante , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Ratones SCID , Especies Reactivas de Oxígeno/metabolismo , Teratoma/prevención & control
6.
J Vasc Med Surg ; 9(7)2021.
Artículo en Inglés | MEDLINE | ID: mdl-36276915

RESUMEN

Cigarette smoking is undoubtedly the single most important risk factor and trigger for vasospastic angina, a condition also known as Prinzmetal angina secondary to coronary artery vasospasm. Even decades before vasospastic angina was first described by Dr. Myron Prinzmetal and his colleagues in 1959, there had been suspected connections between smoking and coronary artery vasospasm in what was alluded to then as "tobacco angina." The intimate relationship between smoking and vasospastic angina has since been extensively researched and validated through decades of epidemiological and clinical studies. The fact that smoking would aggravate vasospastic angina comes with very little surprise, as it has been shown to adversely impact many of the disease processes thought to underlie vasospastic angina, including autonomic dysfunction, endothelial dysfunction, smooth muscle hyperactivity, and genetic susceptibility. While avoidance of smoking is the first logical step in managing smokers with vasospastic angina, there have been reported cases of vasospastic angina paradoxically triggered by smoking cessation or relieved with smoking resumption or nicotine replacement therapy. Thus, there appears to be patient-specific factors that could significantly alter the close connection between smoking and vasospastic angina, warranting further mechanistic investigations. In this review, we will examine this complicated relationship between smoking and vasospastic angina from multiple perspectives (historical, mechanistic, and clinical) and call attention to the "smoking paradox," which, with further elucidation, may provide additional insight into the complex mechanisms of VSA and potentially new strategies to treat medically refractory VSA, at least in selected individuals.

7.
Pharmacol Rev ; 72(1): 320-342, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31871214

RESUMEN

Human induced pluripotent stem cells (iPSCs) have emerged as an effective platform for regenerative therapy, disease modeling, and drug discovery. iPSCs allow for the production of limitless supply of patient-specific somatic cells that enable advancement in cardiovascular precision medicine. Over the past decade, researchers have developed protocols to differentiate iPSCs to multiple cardiovascular lineages, as well as to enhance the maturity and functionality of these cells. Despite significant advances, drug therapy and discovery for cardiovascular disease have lagged behind other fields such as oncology. We speculate that this paucity of drug discovery is due to a previous lack of efficient, reproducible, and translational model systems. Notably, existing drug discovery and testing platforms rely on animal studies and clinical trials, but investigations in animal models have inherent limitations due to interspecies differences. Moreover, clinical trials are inherently flawed by assuming that all individuals with a disease will respond identically to a therapy, ignoring the genetic and epigenomic variations that define our individuality. With ever-improving differentiation and phenotyping methods, patient-specific iPSC-derived cardiovascular cells allow unprecedented opportunities to discover new drug targets and screen compounds for cardiovascular disease. Imbued with the genetic information of an individual, iPSCs will vastly improve our ability to test drugs efficiently, as well as tailor and titrate drug therapy for each patient.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/terapia , Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes Inducidas/citología , Medicina de Precisión/métodos , Animales , Fármacos Cardiovasculares/uso terapéutico , Linaje de la Célula , Desarrollo de Medicamentos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/trasplante , Ensayos Clínicos Controlados Aleatorios como Asunto
8.
PLoS One ; 13(8): e0202778, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30169548

RESUMEN

BACKGROUND: MicroRNAs (miR) are small non-coding RNAs that regulate diverse biological functions. The bicistronic gene miR-143/145 determines cell fate and phenotype of vascular smooth muscle cells (VSMC), in part, by destabilizing Elk-1 mRNA. The transcription factor c-Myb also regulates differentiation and proliferation of VSMC, and here we test whether these effects may be mediated by miR-143/145. METHODS & RESULTS: Flow cytometry of cardiovascular-directed d3.75 embryoid bodies (EBs) isolated smooth muscle progenitors with specific cell surface markers. In c-myb knockout (c-myb -/-) EB, these progenitors manifest low levels of miR-143 (19%; p<0.05) and miR-145 (6%; p<0.01) expression as compared to wild-type (wt) EB. Primary VSMC isolated from transgenic mice with diminished expression (c-myblx/lx) or reduced activity (c-mybh/h) of c-Myb also manifest low levels of miR-143 (c-myblx/lx: 50%; c-mybh/h: 41%), and miR-145 (c-myblx/lx: 49%; c-mybh/h: 56%), as compared to wt (P<0.05). Sequence alignment identified four putative c-Myb binding sites (MBS1-4) in the proximal promoter (PP) of the miR-143/145 gene. PP-reporter constructs revealed that point mutations in MBS1 and MBS4 abrogated c-Myb-dependent transcription from the miR-143/145 PP (P<0.01). Chromatin immunoprecipitation (ChIP) revealed preferential c-Myb binding at MBS4 (p<0.001). By conjugating Elk-1 3'-untranslated region (UTR) to a reporter and co-transducing wt VSMC with this plus a miR-143-antagomir, and co-transducing c-myblx/lx VSMC with this plus a miR-143-mimic, we demonstrate that c-Myb's ability to repress Elk-1 is mediated by miR-143. CONCLUSION: c-Myb regulates VSMC gene expression by transcriptional activation of miR-143/145.


Asunto(s)
MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Regiones no Traducidas 3' , Animales , Antagomirs/metabolismo , Sitios de Unión , Células Cultivadas , Inmunoprecipitación de Cromatina , Ratones , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Mutación Puntual , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myb/genética , Activación Transcripcional/genética , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo
9.
Clin Sci (Lond) ; 131(15): 1801-1814, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28424290

RESUMEN

Objective: Few methods enable molecular and cellular studies of vascular aging or Type 2 diabetes (T2D). Here, we report a new approach to studying human vascular smooth muscle cell (VSMC) pathophysiology by examining VSMCs differentiated from progenitors found in skin. Approach and results: Skin-derived precursors (SKPs) were cultured from biopsies (N=164, ∼1 cm2) taken from the edges of surgical incisions of older adults (N=158; males 72%; mean age 62.7 ± 13 years) undergoing cardiothoracic surgery, and differentiated into VSMCs at high efficiency (>80% yield). The number of SKPs isolated from subjects with T2D was ∼50% lower than those without T2D (cells/g: 0.18 ± 0.03, N=58 versus 0.40 ± 0.05, N=100, P<0.05). Importantly, SKP-derived VSMCs from subjects with T2D had higher Fluo-5F-determined baseline cytosolic Ca2+ concentrations (AU: 1,968 ± 160, N=7 versus 1,386 ± 170, N=13, P<0.05), and a trend toward greater Ca2+ cycling responses to norepinephrine (NE) (AUC: 177,207 ± 24,669, N=7 versus 101,537 ± 15,881, N=20, P<0.08) despite a reduced frequency of Ca2+ cycling (events s-1 cell-1: 0.011 ± 0.004, N=8 versus 0.021 ± 0.003, N=19, P<0.05) than those without T2D. SKP-derived VSMCs from subjects with T2D also manifest enhanced sensitivity to phenylephrine (PE) in an impedance-based assay (EC50 nM: 72.3 ± 63.6, N=5 versus 3,684 ± 3,122, N=9, P<0.05), and impaired wound closure in vitro (% closure: 21.9 ± 3.6, N=4 versus 67.0 ± 10.3, N=4, P<0.05). Compared with aortic- and saphenous vein-derived primary VSMCs, SKP-derived VSMCs are functionally distinct, but mirror defects of T2D also exhibited by primary VSMCs. CONCLUSION: Skin biopsies from older adults yield sufficient SKPs to differentiate VSMCs, which reveal abnormal phenotypes of T2D that survive differentiation and persist even after long-term normoglycemic culture.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/fisiología , Piel/patología , Células Madre/patología , Anciano , Biopsia , Calcio/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/efectos de los fármacos , Norepinefrina/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
10.
Arterioscler Thromb Vasc Biol ; 36(7): 1367-76, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27174098

RESUMEN

OBJECTIVE: Vascular smooth muscle cells (VSMCs) are believed to dedifferentiate and proliferate in response to vessel injury. Recently, adventitial progenitor cells were implicated as a source of VSMCs involved in vessel remodeling. c-Myb is a transcription factor known to regulate VSMC proliferation in vivo and differentiation of VSMCs from mouse embryonic stem cell-derived progenitors in vitro. However, the role of c-Myb in regulating specific adult vascular progenitor cell populations was not known. Our objective was to examine the role of c-Myb in the proliferation and differentiation of Sca1(+) adventitial VSMC progenitor cells. APPROACH AND RESULTS: Using mice with wild-type or hypomorphic c-myb (c-myb(h/h)), BrdU (bromodeoxyuridine) uptake and flow cytometry revealed defective proliferation of Sca1(+) adventitial VSMC progenitor cells at 8, 14, and 28 days post carotid artery denudation injury in c-myb(h/h) arteries. c-myb(h/h) cKit(+)CD34(-)Flk1(-)Sca1(+)CD45(-)Lin(-) cells failed to proliferate, suggesting that c-myb regulates the activation of specific Sca1(+) progenitor cells in vivo and in vitro. Although expression levels of transforming growth factor-ß1 did not vary between wild-type and c-myb(h/h) carotid arteries, in vitro differentiation of c-myb(h/h) Sca1(+) cells manifested defective transforming growth factor-ß1-induced VSMC differentiation. This is mediated by reduced transcriptional activation of myocardin because chromatin immunoprecipitation revealed c-Myb binding to the myocardin promoter only during differentiation of Sca1(+) cells, myocardin promoter mutagenesis identified 2 specific c-Myb-responsive binding sites, and adenovirus-mediated expression of myocardin rescued the phenotype of c-myb(h/h) progenitors. CONCLUSIONS: These data support a role for c-Myb in the regulation of VSMC progenitor cells and provide novel insight into how c-myb regulates VSMC differentiation through myocardin.


Asunto(s)
Adventicia/metabolismo , Ataxina-1/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Diferenciación Celular , Proliferación Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Células Madre/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Adventicia/efectos de los fármacos , Adventicia/lesiones , Adventicia/patología , Animales , Sitios de Unión , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Genotipo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Proteínas Nucleares/genética , Fenotipo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myb/genética , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/patología , Factores de Tiempo , Transactivadores/genética , Transcripción Genética , Transfección , Factor de Crecimiento Transformador beta1/farmacología , Remodelación Vascular
11.
EMBO J ; 26(3): 730-40, 2007 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-17235287

RESUMEN

ATP-dependent chromatin remodeling complexes play a critical role in chromatin dynamics. A large number of in vitro studies have pointed towards nucleosome sliding as the principal remodeling outcome of SWI/SNF action, whereas few have described histone octamer transfer as the principal outcome. In contrast, recent in vivo studies have linked the activity of SWI/SNF to histone eviction in trans from gene promoters. In this study, we have found that the chimeric transcription factor Gal4-VP16 can enhance SWI/SNF histone octamer transfer activity, resulting in targeted histone eviction from a nucleosome probe. This effect is dependent on the presence of the activation domain. We observed that under conditions mimicking the in vivo relative abundance of SWI/SNF with respect to the total number of nucleosomes in a cell nucleus, the accessibility of the transcription factor binding site is the first determinant in the sequence of events leading to nucleosome remodeling. We propose a model mechanism for this transcription factor-mediated enhancement of SWI/SNF octamer transfer activity.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Proteínas de Unión al ADN/metabolismo , Modelos Biológicos , Nucleosomas/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Factores de Transcripción/metabolismo , Adenosina Trifosfatasas , Secuencia de Bases , Ensamble y Desensamble de Cromatina/genética , Sondas de ADN/genética , Datos de Secuencia Molecular , Nucleosomas/genética , Estructura Terciaria de Proteína , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética
12.
Mol Cell Biol ; 24(16): 7043-58, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15282305

RESUMEN

We have programmed human cells to express physiological levels of recombinant RNA polymerase II (RNAPII) subunits carrying tandem affinity purification (TAP) tags. Double-affinity chromatography allowed for the simple and efficient isolation of a complex containing all 12 RNAPII subunits, the general transcription factors TFIIB and TFIIF, the RNAPII phosphatase Fcp1, and a novel 153-kDa polypeptide of unknown function that we named RNAPII-associated protein 1 (RPAP1). The TAP-tagged RNAPII complex is functionally active both in vitro and in vivo. A role for RPAP1 in RNAPII transcription was established by shutting off the synthesis of Ydr527wp, a Saccharomyces cerevisiae protein homologous to RPAP1, and demonstrating that changes in global gene expression were similar to those caused by the loss of the yeast RNAPII subunit Rpb11. We also used TAP-tagged Rpb2 with mutations in fork loop 1 and switch 3, two structural elements located strategically within the active center, to start addressing the roles of these elements in the interaction of the enzyme with the template DNA during the transcription reaction.


Asunto(s)
Proteínas Portadoras/aislamiento & purificación , Proteínas Portadoras/metabolismo , Mutación , Subunidades de Proteína/aislamiento & purificación , Subunidades de Proteína/metabolismo , ARN Polimerasa II/aislamiento & purificación , ARN Polimerasa II/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Proteínas Portadoras/genética , ADN/metabolismo , Etiquetas de Secuencia Expresada , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multienzimáticos , Fosfoproteínas Fosfatasas/aislamiento & purificación , Fosfoproteínas Fosfatasas/metabolismo , Regiones Promotoras Genéticas , Conformación Proteica , Subunidades de Proteína/genética , ARN Polimerasa II/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Homología de Secuencia de Ácido Nucleico , Factor de Transcripción TFIIB/genética , Factor de Transcripción TFIIB/aislamiento & purificación , Factor de Transcripción TFIIB/metabolismo , Factores de Transcripción TFII/genética , Factores de Transcripción TFII/aislamiento & purificación , Factores de Transcripción TFII/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA