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.
Cell Tissue Res ; 389(2): 309-326, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35661920

RESUMEN

The heart's limited regenerative capacity raises the need for novel stem cell-based therapeutic approaches for cardiac regeneration. However, the use of stem cells is restrictive due to poor determination of their properties and the factors that regulate them. Here, we investigated the role of desmin, the major muscle-specific intermediate filament protein, in the characteristics and differentiation capacity of cardiac side population (CSP) and Sca1+ stem cells of adult mice. We found that desmin deficiency affects the microenvironment of the cells and leads to increased numbers of CSP but not Sca1+ cells; CSP subpopulation composition is altered, the expression of the senescence marker p16INK4a in Sca1+ cells is increased, and early cardiomyogenic commitment is impaired. Specifically, we found that mRNA levels of the cardiac transcription factors Mef2c and Nkx2.5 were significantly reduced in des-/- CSP and Sca1+ cells, while differentiation of CSP and Sca1+ cells demonstrated that in the absence of desmin, the levels of Nkx2.5, Mef2c, Tnnt2, Hey2, and Myh6 mRNA are differentially affected. Thus, desmin deficiency restricts the regenerative potential of CSP and Sca1+ cells, both directly and indirectly through their microenvironment.


Asunto(s)
Miocitos Cardíacos , Células Madre , Animales , Diferenciación Celular/genética , Desmina/genética , Desmina/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo
2.
Cell Tissue Res ; 385(3): 675-696, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34037836

RESUMEN

The desmin-associated protein myospryn, encoded by the cardiomyopathy-associated gene 5 (CMYA5), is a TRIM-like protein associated to the BLOC-1 (Biogenesis of Lysosomes Related Organelles Complex 1) protein dysbindin. Human myospryn mutations are linked to both cardiomyopathy and schizophrenia; however, there is no evidence of a direct causative link of myospryn to these diseases. Therefore, we sought to unveil the role of myospryn in heart and brain. We have genetically inactivated the myospryn gene by homologous recombination and demonstrated that myospryn null hearts have dilated phenotype and compromised cardiac function. Ultrastructural analyses revealed that the sarcomere organization is not obviously affected; however, intercalated disk (ID) integrity is impaired, along with mislocalization of ID and sarcoplasmic reticulum (SR) protein components. Importantly, cardiac and skeletal muscles of myospryn null mice have severe mitochondrial defects with abnormal internal vacuoles and extensive cristolysis. In addition, swollen SR and T-tubules often accompany the mitochondrial defects, strongly implying a potential link of myospryn together with desmin to SR- mitochondrial physical and functional cross-talk. Furthermore, given the reported link of human myospryn mutations to schizophrenia, we performed behavioral studies, which demonstrated that myospryn-deficient male mice display disrupted startle reactivity and prepulse inhibition, asocial behavior, decreased exploratory behavior, and anhedonia. Brain neurochemical and ultrastructural analyses revealed prefrontal-striatal monoaminergic neurotransmitter defects and ultrastructural degenerative aberrations in cerebellar cytoarchitecture, respectively, in myospryn-deficient mice. In conclusion, myospryn is essential for both cardiac and brain structure and function and its deficiency leads to cardiomyopathy and schizophrenia-associated symptoms.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas Musculares/deficiencia , Miocardio/patología , Esquizofrenia/genética , Animales , Femenino , Humanos , Masculino , Ratones
3.
Front Cardiovasc Med ; 6: 32, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31001541

RESUMEN

Following an insult by both intrinsic and extrinsic pathways, complex cellular, and molecular interactions determine a successful recovery or inadequate repair of damaged tissue. The efficiency of this process is particularly important in the heart, an organ characterized by very limited regenerative and repair capacity in higher adult vertebrates. Cardiac insult is characteristically associated with fibrosis and heart failure, as a result of cardiomyocyte death, myocardial degeneration, and adverse remodeling. Recent evidence implies that resident non-cardiomyocytes, fibroblasts but also macrophages -pillars of the innate immunity- form part of the inflammatory response and decisively affect the repair process following a cardiac insult. Multiple studies in model organisms (mouse, zebrafish) of various developmental stages (adult and neonatal) combined with genetically engineered cell plasticity and differentiation intervention protocols -mainly targeting cardiac fibroblasts or progenitor cells-reveal particular roles of resident and recruited innate immune cells and their secretome in the coordination of cardiac repair. The interplay of innate immune cells with cardiac fibroblasts and cardiomyocytes is emerging as a crucial platform to help our understanding and, importantly, to allow the development of effective interventions sufficient to minimize cardiac damage and dysfunction after injury.

4.
Nat Commun ; 10(1): 1405, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926812

RESUMEN

Lung adenocarcinoma (LUAD)-derived Wnts increase cancer cell proliferative/stemness potential, but whether they impact the immune microenvironment is unknown. Here we show that LUAD cells use paracrine Wnt1 signaling to induce immune resistance. In TCGA, Wnt1 correlates strongly with tolerogenic genes. In another LUAD cohort, Wnt1 inversely associates with T cell abundance. Altering Wnt1 expression profoundly affects growth of murine lung adenocarcinomas and this is dependent on conventional dendritic cells (cDCs) and T cells. Mechanistically, Wnt1 leads to transcriptional silencing of CC/CXC chemokines in cDCs, T cell exclusion and cross-tolerance. Wnt-target genes are up-regulated in human intratumoral cDCs and decrease upon silencing Wnt1, accompanied by enhanced T cell cytotoxicity. siWnt1-nanoparticles given as single therapy or part of combinatorial immunotherapies act at both arms of the cancer-immune ecosystem to halt tumor growth. Collectively, our studies show that Wnt1 induces immunologically cold tumors through cDCs and highlight its immunotherapeutic targeting.


Asunto(s)
Inmunidad Adaptativa , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/inmunología , Quimiocinas/genética , Células Dendríticas/metabolismo , Silenciador del Gen , Proteína Wnt1/metabolismo , Traslado Adoptivo , Animales , Línea Celular Tumoral , Proliferación Celular , Quimiocinas/metabolismo , Humanos , Evasión Inmune , Ratones Endogámicos C57BL , Interferencia de ARN , Transducción de Señal , Linfocitos T/metabolismo , Regulación hacia Arriba
5.
Biophys Rev ; 10(4): 1007-1031, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30027462

RESUMEN

Intermediate filament (IF) proteins are critical regulators in health and disease. The discovery of hundreds of mutations in IF genes and posttranslational modifications has been linked to a plethora of human diseases, including, among others, cardiomyopathies, muscular dystrophies, progeria, blistering diseases of the epidermis, and neurodegenerative diseases. The major IF proteins that have been linked to cardiomyopathies and heart failure are the muscle-specific cytoskeletal IF protein desmin and the nuclear IF protein lamin, as a subgroup of the known desminopathies and laminopathies, respectively. The studies so far, both with healthy and diseased heart, have demonstrated the importance of these IF protein networks in intracellular and intercellular integration of structure and function, mechanotransduction and gene activation, cardiomyocyte differentiation and survival, mitochondrial homeostasis, and regulation of metabolism. The high coordination of all these processes is obviously of great importance for the maintenance of proper, life-lasting, and continuous contraction of this highly organized cardiac striated muscle and consequently a healthy heart. In this review, we will cover most known information on the role of IFs in the above processes and how their deficiency or disruption leads to cardiomyopathy and heart failure.

6.
J Biol Chem ; 292(18): 7435-7451, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28292929

RESUMEN

The activity of the E3 ligase, SMURF2, is antagonized by an intramolecular, autoinhibitory interaction between its C2 and Hect domains. Relief of SMURF2 autoinhibition is induced by TGFß and is mediated by the inhibitory SMAD, SMAD7. In a proteomic screen for endomembrane interactants of the RING-domain E3 ligase, RNF11, we identified SMURF2, among a cohort of Hect E3 ligases previously implicated in TGFß signaling. Reconstitution of the SMURF2·RNF11 complex in vitro unexpectedly revealed robust SMURF2 E3 ligase activity, with biochemical properties previously restricted to the SMURF2·SMAD7 complex. Using in vitro binding assays, we find that RNF11 can directly compete with SMAD7 for SMURF2 and that binding is mutually exclusive and dependent on a proline-rich domain. Moreover, we found that co-expression of RNF11 and SMURF2 dramatically reduced SMURF2 ubiquitylation in the cell. This effect is strictly dependent on complex formation and sorting determinants that regulate the association of RNF11 with membranes. RNF11 is overexpressed in certain tumors, and, importantly, we found that depletion of this protein down-regulated gene expression of several TGFß-responsive genes, dampened cell proliferation, and dramatically reduced cell migration in response to TGFß. Our data suggest for the first time that the choice of binding partners for SMURF2 can sustain or repress TGFß signaling, and RNF11 may promote TGFß-induced cell migration.


Asunto(s)
Proteínas Portadoras/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal/fisiología , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células 3T3-L1 , Animales , Proteínas Portadoras/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Proteínas de Unión al ADN , Células HeLa , Humanos , Ratones , Complejos Multiproteicos/genética , Proteína smad7/genética , Factor de Crecimiento Transformador beta/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/fisiología
7.
J Cell Sci ; 129(20): 3705-3720, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27566162

RESUMEN

The association of desmin with the α-crystallin Β-chain (αΒ-crystallin; encoded by CRYAB), and the fact that mutations in either one of them leads to heart failure in humans and mice, suggests a potential compensatory interplay between the two in cardioprotection. To address this hypothesis, we investigated the consequences of αΒ-crystallin overexpression in the desmin-deficient (Des-/-) mouse model, which possesses a combination of the pathologies found in most cardiomyopathies, with mitochondrial defects as a hallmark. We demonstrated that cardiac-specific αΒ-crystallin overexpression ameliorates all these defects and improves cardiac function to almost wild-type levels. Protection by αΒ-crystallin overexpression is linked to maintenance of proper mitochondrial protein levels, inhibition of abnormal mitochondrial permeability transition pore activation and maintenance of mitochondrial membrane potential (Δψm). Furthermore, we found that both desmin and αΒ-crystallin are localized at sarcoplasmic reticulum (SR)-mitochondria-associated membranes (MAMs), where they interact with VDAC, Mic60 - the core component of mitochondrial contact site and cristae organizing system (MICOS) complex - and ATP synthase, suggesting that these associations could be crucial in mitoprotection at different levels.


Asunto(s)
Desmina/metabolismo , Homeostasis , Mitocondrias/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Cadena B de alfa-Cristalina/metabolismo , Animales , Antioxidantes/farmacología , Supervivencia Celular/efectos de los fármacos , Desmina/deficiencia , Pruebas de Función Cardíaca , Homeostasis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Proteínas Mitocondriales/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/ultraestructura , Condicionamiento Físico Animal , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Estrés Fisiológico/efectos de los fármacos , Natación , Canales Aniónicos Dependientes del Voltaje/metabolismo
8.
Methods Enzymol ; 568: 427-59, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26795479

RESUMEN

Intermediate filament (IF) cytoskeleton comprises the fine-tuning cellular machinery regulating critical homeostatic mechanisms. In skeletal and cardiac muscle, deficiency or disturbance of the IF network leads to severe pathology, particularly in the latter. The three-dimensional scaffold of the muscle-specific IF protein desmin interconnects key features of the cardiac muscle cells, including the Z-disks, intercalated disks, plasma membrane, nucleus, mitochondria, lysosomes, and potentially sarcoplasmic reticulum. This is crucial for the highly organized striated muscle, in which effective energy production and transmission as well as mechanochemical signaling are tightly coordinated among the organelles and the contractile apparatus. The role of desmin and desmin-associated proteins in the biogenesis, trafficking, and organelle function, as well as the development, differentiation, and survival of the cardiac muscle begins to be enlightened, but the precise mechanisms remain elusive. We propose a set of experimental tools that can be used, in vivo and in vitro, to unravel crucial new pathways by which the IF cytoskeleton facilitates proper organelle function, homeostasis, and cytoprotection and further understand how its disturbance and deficiency lead to disease.


Asunto(s)
Desmina/metabolismo , Miocardio/metabolismo , Animales , Células Cultivadas , Humanos , Miocitos Cardíacos/metabolismo
9.
Curr Opin Cell Biol ; 32: 113-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25680090

RESUMEN

Maintenance of the highly organized striated muscle tissue requires a cell-wide dynamic network that through interactions with all vital cell structures, provides an effective mechanochemical integrator of morphology and function, absolutely necessary for intra-cellular and intercellular coordination of all muscle functions. A good candidate for such a system is the desmin intermediate filament cytoskeletal network. Human desmin mutations and post-translational modifications cause disturbance of this network, thus leading to loss of function of both desmin and its binding partners, as well as potential toxic effects of the formed aggregates. Both loss of normal function and gain of toxic function are linked to mitochondrial defects, cardiomyocyte death, muscle degeneration and development of skeletal myopathy and cardiomyopathy.


Asunto(s)
Muerte Celular , Desmina/metabolismo , Músculo Estriado/citología , Enfermedades Musculares/patología , Animales , Membrana Celular/metabolismo , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Desmina/química , Desmina/genética , Humanos , Filamentos Intermedios/metabolismo , Mitocondrias/metabolismo , Músculo Estriado/metabolismo , Mutación , Procesamiento Proteico-Postraduccional
10.
EMBO Rep ; 11(12): 969-76, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20948544

RESUMEN

We have identified the E3 ligase Traf7 as a direct MyoD1 target and show that cell cycle exit-an early event in muscle differentiation-is linked to decreased Traf7 expression. Depletion of Traf7 accelerates myogenesis, in part through downregulation of nuclear factor-κB (NF-κB) activity. We used a proteomic screen to identify NEMO, the NF-κB essential modulator, as a Traf7-interacting protein. Finally, we show that ubiquitylation of NF-κB essential modulator is regulated exclusively by Traf7 activity in myoblasts. Our results suggest a new mechanism by which MyoD1 function is coupled to NF-κB activity through Traf7, regulating the balance between cell cycle progression and differentiation during myogenesis.


Asunto(s)
Desarrollo de Músculos/genética , Proteína MioD/metabolismo , FN-kappa B/metabolismo , Transcripción Genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Ciclina D1/metabolismo , Regulación de la Expresión Génica , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Mioblastos/citología , Mioblastos/metabolismo , Fosforilación , Unión Proteica , Proteína de Retinoblastoma/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/deficiencia , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Ubiquitina/metabolismo , Ubiquitinación
11.
Genes Dev ; 23(1): 37-53, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19136625

RESUMEN

Current models posit that E2F transcription factors can be divided into members that either activate or repress transcription, in part through collaboration with the retinoblastoma (pRb) tumor suppressor family. The E2f3 locus encodes E2f3a and E2f3b proteins, and available data suggest that they regulate cell cycle-dependent gene expression through opposing transcriptional activating and repressing activities in growing and quiescent cells, respectively. However, the role, if any, of E2F proteins, and in particular E2f3, in myogenic differentiation is not well understood. Here, we dissect the contributions of E2f3 isoforms and other activating and repressing E2Fs to cell cycle exit and differentiation by performing genome-wide identification of isoform-specific targets. We show that E2f3a and E2f3b target genes are involved in cell growth, lipid metabolism, and differentiation in an isoform-specific manner. Remarkably, using gene silencing, we show that E2f3b, but not E2f3a or other E2F family members, is required for myogenic differentiation, and that this requirement for E2f3b does not depend on pRb. Our functional studies indicate that E2f3b specifically attenuates expression of genes required to promote differentiation. These data suggest how diverse E2F isoforms encoded by a single locus can play opposing roles in cell cycle exit and differentiation.


Asunto(s)
Diferenciación Celular , Factor de Transcripción E2F3/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mioblastos/citología , Mioblastos/metabolismo , Animales , Línea Celular , Factores de Transcripción E2F/metabolismo , Factor de Transcripción E2F3/genética , Ratones , Unión Proteica , Isoformas de Proteínas , Proteínas Represoras/metabolismo
12.
Mol Cell ; 27(1): 53-66, 2007 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-17612490

RESUMEN

Using genome-wide approaches, we have elucidated the regulatory circuitry governed by the XBP1 transcription factor, a key effector of the mammalian unfolded protein response (UPR), in skeletal muscle and secretory cells. We identified a core group of genes involved in constitutive maintenance of ER function in all cell types and tissue- and condition-specific targets. In addition, we identified a cadre of unexpected targets that link XBP1 to neurodegenerative and myodegenerative diseases, as well as to DNA damage and repair pathways. Remarkably, we found that XBP1 regulates functionally distinct targets through different sequence motifs. Further, we identified Mist1, a critical regulator of differentiation, as an important target of XBP1, providing an explanation for developmental defects associated with XBP1 loss of function. Our results provide a detailed picture of the regulatory roadmap governed by XBP1 in distinct cell types as well as insight into unexplored functions of XBP1.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Redes Reguladoras de Genes/genética , Proteínas Nucleares/metabolismo , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión , Inmunoprecipitación de Cromatina , Biología Computacional , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Regulación de la Expresión Génica , Genoma/genética , Ratones , Datos de Secuencia Molecular , Desarrollo de Músculos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Enfermedades Neurodegenerativas/genética , Proteínas Nucleares/genética , Unión Proteica , Pliegue de Proteína , Ratas , Factores de Transcripción del Factor Regulador X , Reproducibilidad de los Resultados , Especificidad por Sustrato , Factores de Transcripción , Proteína 1 de Unión a la X-Box
13.
Proc Natl Acad Sci U S A ; 102(34): 12129-34, 2005 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-16099835

RESUMEN

Rhabdoid tumors are aggressive pediatric malignancies for which, currently, there are no effective or standard treatment strategies. Rhabdoid tumors arise because of the loss of the tumor suppressor gene INI1. We have previously demonstrated that INI1 represses Cyclin D1 transcription in rhabdoid cells by directly recruiting histone deacetylase 1 complex to its promoter, leading to G(0)-G(1) arrest. Expression of Cyclin D1 overcomes cell cycle arrest mediated by INI1 and Cyclin D1 overexpression in human rhabdoid tumors is a common phenomenon. However, it is not clear whether Cyclin D1 is a critical downstream target of INI1 in vivo and whether the derepression of this gene is essential for rhabdoid tumorigenesis. To determine the requirement of Cyclin D1 for genesis of rhabdoid tumors in vivo, we developed Ini1 heterozygous mice by targeted disruption. We found that the tumors developed in these Ini1+/- mice are rhabdoid, defective for Ini1 protein, and like the human tumors, express Cyclin D1. We crossed Ini1+/- mice to Cyclin D1-/- mice and found that Ini1+/- mice with Cyclin D1 deficiency did not develop any spontaneous tumors, in contrast to the parental Ini1+/- mice. These results strongly support the hypothesis that Cyclin D1 is a key mediator in the genesis of rhabdoid tumors. Our results provide an in vivo proof of concept that drugs that target Cyclin D1 expression or activity could be potentially effective as novel therapeutic agents for rhabdoid tumors.


Asunto(s)
Ciclo Celular/fisiología , Ciclina D1/genética , Proteínas de Unión al ADN/genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica/genética , Tumor Rabdoide/genética , Factores de Transcripción/genética , Animales , Western Blotting , Ciclo Celular/genética , Proteínas Cromosómicas no Histona , Cruzamientos Genéticos , Cartilla de ADN , Proteínas de Unión al ADN/metabolismo , Marcación de Gen , Inmunohistoquímica , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína SMARCB1 , Factores de Transcripción/metabolismo
14.
Genes Dev ; 19(5): 553-69, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15706034

RESUMEN

We have combined genome-wide transcription factor binding and expression profiling to assemble a regulatory network controlling the myogenic differentiation program in mammalian cells. We identified a cadre of overlapping and distinct targets of the key myogenic regulatory factors (MRFs)--MyoD and myogenin--and Myocyte Enhancer Factor 2 (MEF2). We discovered that MRFs and MEF2 regulate a remarkably extensive array of transcription factor genes that propagate and amplify the signals initiated by MRFs. We found that MRFs play an unexpectedly wide-ranging role in directing the assembly and usage of the neuromuscular junction. Interestingly, these factors also prepare myoblasts to respond to diverse types of stress. Computational analyses identified novel combinations of factors that, depending on the differentiation state, might collaborate with MRFs. Our studies suggest unanticipated biological insights into muscle development and highlight new directions for further studies of genes involved in muscle repair and responses to stress and damage.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Desarrollo de Músculos/genética , Proteína MioD/genética , Mioblastos , Miogenina/genética , Diferenciación Celular/fisiología , Línea Celular , Proteínas de Unión al ADN , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Transcripción MEF2 , Desarrollo de Músculos/fisiología , Proteína MioD/metabolismo , Mioblastos/fisiología , Factores Reguladores Miogénicos , Miogenina/metabolismo , Unión Neuromuscular/genética , Unión Neuromuscular/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Transcripción , Transcripción Genética/genética , Transcripción Genética/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...