Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Annu Rev Cell Dev Biol ; 34: 59-84, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30074816

RESUMEN

In recent years, thin membrane protrusions such as cytonemes and tunneling nanotubes have emerged as a novel mechanism of intercellular communication. Protrusion-based cellular interactions allow for specific communication between participating cells and have a distinct spectrum of advantages compared to secretion- and diffusion-based intercellular communication. Identification of protrusion-based signaling in diverse systems suggests that this mechanism is a ubiquitous and prevailing means of communication employed by many cell types. Moreover, accumulating evidence indicates that protrusion-based intercellular communication is often involved in pathogenesis, including cancers and infections. Here we review our current understanding of protrusion-based intercellular communication.


Asunto(s)
Comunicación Celular/genética , Linaje de la Célula/genética , Extensiones de la Superficie Celular/genética , Endocitosis/genética , Humanos , Nanotubos/química , Transducción de Señal/genética
2.
Semin Cell Dev Biol ; 129: 126-134, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35260295

RESUMEN

Cell-cell communications are central to a variety of physiological and pathological processes in multicellular organisms. Cells often rely on cellular protrusions to communicate with one another, which enable highly selective and efficient signaling within complex tissues. Owing to significant improvements in imaging techniques, identification of signaling protrusions has increased in recent years. These protrusions are structurally specialized for signaling and facilitate interactions between cells. Therefore, physical regulation of these structures must be key for the appropriate strength and pattern of signaling outcomes. However, the typical approaches for understanding signaling regulation tend to focus solely on changes in signaling molecules, such as gene expression, protein-protein interaction, and degradation. In this short review, we summarize the studies proposing the removal of different types of signaling protrusions-including cilia, neurites, MT (microtubule based)-nanotubes and microvilli-and discuss their mechanisms and significance in signaling regulation.


Asunto(s)
Comunicación Celular , Extensiones de la Superficie Celular , Extensiones de la Superficie Celular/metabolismo , Microtúbulos/metabolismo , Neuritas , Transducción de Señal
3.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33753475

RESUMEN

Stem cells divide asymmetrically to generate a stem cell and a differentiating daughter cell. Yet, it remains poorly understood how a stem cell and a differentiating daughter cell can receive distinct levels of niche signal and thus acquire different cell fates (self-renewal versus differentiation), despite being adjacent to each other and thus seemingly exposed to similar levels of niche signaling. In the Drosophila ovary, germline stem cells (GSCs) are maintained by short range bone morphogenetic protein (BMP) signaling; the BMP ligands activate a receptor that phosphorylates the downstream molecule mothers against decapentaplegic (Mad). Phosphorylated Mad (pMad) accumulates in the GSC nucleus and activates the stem cell transcription program. Here, we demonstrate that pMad is highly concentrated in the nucleus of the GSC, while it quickly decreases in the nucleus of the differentiating daughter cell, the precystoblast (preCB), before the completion of cytokinesis. We show that a known Mad phosphatase, Dullard (Dd), is required for the asymmetric partitioning of pMad. Our mathematical modeling recapitulates the high sensitivity of the ratio of pMad levels to the Mad phosphatase activity and explains how the asymmetry arises in a shared cytoplasm. Together, these studies reveal a mechanism for breaking the symmetry of daughter cells during asymmetric stem cell division.


Asunto(s)
División Celular Asimétrica/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Poro Nuclear/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Células Madre/fisiología , Factores de Transcripción/metabolismo , Animales , Animales Modificados Genéticamente , Núcleo Celular , Drosophila melanogaster , Femenino , Oocitos , Fosforilación/genética , Activación Transcripcional
4.
PLoS Biol ; 18(12): e3001003, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33315855

RESUMEN

Stem-cell niche signaling is short-range in nature, such that only stem cells but not their differentiating progeny receive self-renewing signals. At the apical tip of the Drosophila testis, 8 to 10 germline stem cells (GSCs) surround the hub, a cluster of somatic cells that organize the stem-cell niche. We have previously shown that GSCs form microtubule-based nanotubes (MT-nanotubes) that project into the hub cells, serving as the platform for niche signal reception; this spatial arrangement ensures the reception of the niche signal specifically by stem cells but not by differentiating cells. The receptor Thickveins (Tkv) is expressed by GSCs and localizes to the surface of MT-nanotubes, where it receives the hub-derived ligand Decapentaplegic (Dpp). The fate of Tkv receptor after engaging in signaling on the MT-nanotubes has been unclear. Here we demonstrate that the Tkv receptor is internalized into hub cells from the MT-nanotube surface and subsequently degraded in the hub cell lysosomes. Perturbation of MT-nanotube formation and Tkv internalization from MT-nanotubes into hub cells both resulted in an overabundance of Tkv protein in GSCs and hyperactivation of a downstream signal, suggesting that the MT-nanotubes also serve a second purpose to dampen the niche signaling. Together, our results demonstrate that MT-nanotubes play dual roles to ensure the short-range nature of niche signaling by (1) providing an exclusive interface for the niche ligand-receptor interaction; and (2) limiting the amount of stem cell receptors available for niche signal reception.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/fisiología , Nicho de Células Madre/fisiología , Células Madre/metabolismo , Células Madre Germinales Adultas/metabolismo , Células Madre Germinales Adultas/fisiología , Animales , Diferenciación Celular/fisiología , Drosophila melanogaster/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Ligandos , Masculino , Microtúbulos/metabolismo , Microtúbulos/fisiología , Transducción de Señal/fisiología , Células Madre/citología , Testículo/metabolismo
5.
Nature ; 523(7560): 329-32, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26131929

RESUMEN

Stem cell niches provide resident stem cells with signals that specify their identity. Niche signals act over a short range such that only stem cells but not their differentiating progeny receive the self-renewing signals. However, the cellular mechanisms that limit niche signalling to stem cells remain poorly understood. Here we show that the Drosophila male germline stem cells form previously unrecognized structures, microtubule-based nanotubes, which extend into the hub, a major niche component. Microtubule-based nanotubes are observed specifically within germline stem cell populations, and require intraflagellar transport proteins for their formation. The bone morphogenetic protein (BMP) receptor Tkv localizes to microtubule-based nanotubes. Perturbation of microtubule-based nanotubes compromises activation of Dpp signalling within germline stem cells, leading to germline stem cell loss. Moreover, Dpp ligand and Tkv receptor interaction is necessary and sufficient for microtubule-based nanotube formation. We propose that microtubule-based nanotubes provide a novel mechanism for selective receptor-ligand interaction, contributing to the short-range nature of niche-stem-cell signalling.


Asunto(s)
Drosophila melanogaster/citología , Microtúbulos/química , Microtúbulos/metabolismo , Nanotubos , Transducción de Señal , Nicho de Células Madre/fisiología , Testículo/citología , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ligandos , Masculino , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo
6.
Semin Cell Dev Biol ; 129: 61-62, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35927120
7.
Mol Reprod Dev ; 83(8): 675-83, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27434704

RESUMEN

Adult stem cells reside in specialized microenvironments, called niches, that maintain stem cells in an undifferentiated and self-renewing state. Defining and understanding the mechanisms that restrict niche signaling exclusively to stem cells is crucial to determine how stem cells undergo self-renewal while their progeny, often located just one cell diameter away from the niche, differentiate. Despite extensive studies on the signaling pathways that operate within stem cells and their niches, how this segregation occurs remains elusive. Here we review recent progress on the characterization of niche-stem cell interactions, with a focus on emerging mechanisms that spatially restrict niche signaling. Mol. Reprod. Dev. 83: 675-683, 2016 © 2016 Wiley Periodicals, Inc.


Asunto(s)
Genitales/fisiología , Transducción de Señal/fisiología , Nicho de Células Madre/fisiología , Células Madre/fisiología , Animales , Genitales/citología , Humanos , Células Madre/citología
8.
Development ; 138(23): 5079-86, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22031544

RESUMEN

Tight regulation of stem cell proliferation is fundamental to tissue homeostasis, aging and tumor suppression. Although stem cells are characterized by their high potential to proliferate throughout the life of the organism, the mechanisms that regulate the cell cycle of stem cells remain poorly understood. Here, we show that the Cdc25 homolog String (Stg) is a crucial regulator of germline stem cells (GSCs) and cyst stem cells (CySCs) in Drosophila testis. Through knockdown and overexpression experiments, we show that Stg is required for stem cell maintenance and that a decline in its expression during aging is a critical determinant of age-associated decline in stem cell function. Furthermore, we show that restoration of Stg expression reverses the age-associated decline in stem cell function but leads to late-onset tumors. We propose that Stg/Cdc25 is a crucial regulator of stem cell function during tissue homeostasis and aging.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Senescencia Celular/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Homeostasis/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Células Madre/fisiología , Testículo/citología , Animales , Drosophila melanogaster/citología , Masculino , Microscopía Fluorescente
9.
bioRxiv ; 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38798517

RESUMEN

Within a given tissue, the stem cell niche provides the microenvironment for stem cells suitable for their self-renewal. Conceptually, the niche space constrains the size of a stem-cell pool, as the cells sharing the niche compete for its space. It has been suggested that either neutral- or non-neutral-competition of stem cells changes the clone dynamics of stem cells. Theoretically, if the rate of asymmetric division is high, the stem cell competition is limited, thus suppressing clonal expansion. However, the effects of asymmetric division on clone dynamics have never been experimentally tested. Here, using the Drosophila germline stem cell (GSC) system, as a simple model of the in-vivo niche, we examine the effect of division modes (asymmetric or symmetric) on clonal dynamics by combining experimental approaches with mathematical modeling. Our experimental data and computational model both suggest that the rate of asymmetric division is proportional to the time a stem cell clone takes to expand. Taken together, our data suggests that asymmetric division is essential for maintaining the genetic variation of stem cells and thus serves as a critical mechanism for safeguarding fertility over the animal age or preventing multiple disorders caused by the clonal expansion of stem cells.

10.
Nat Commun ; 15(1): 1166, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326318

RESUMEN

Drosophila male germline stem cells (GSCs) reside at the tip of the testis and surround a cluster of niche cells. Decapentaplegic (Dpp) is one of the well-established ligands and has a major role in maintaining stem cells located in close proximity. However, the existence and the role of the diffusible fraction of Dpp outside of the niche have been unclear. Here, using genetically-encoded nanobodies called Morphotraps, we physically block Dpp diffusion without interfering with niche-stem cell signaling and find that a diffusible fraction of Dpp is required to ensure differentiation of GSC daughter cells, opposite of its role in maintenance of GSC in the niche. Our work provides an example in which a soluble niche ligand induces opposed cellular responses in stem cells versus in differentiating descendants to ensure spatial control of the niche. This may be a common mechanism to regulate tissue homeostasis.


Asunto(s)
Proteínas de Drosophila , Animales , Masculino , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ligandos , Diferenciación Celular/fisiología , Drosophila/metabolismo , Transducción de Señal/fisiología , Nicho de Células Madre/fisiología , Células Germinativas/metabolismo , Drosophila melanogaster/metabolismo
11.
Methods Mol Biol ; 2677: 127-138, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37464239

RESUMEN

The Drosophila male germline provides a strong model system to understand numerous developmental and cell-biological processes, owing to a well-defined anatomy and cell type markers in combination with various genetic tools available for the Drosophila system. A major weakness of this system has been the difficulty of approaches for obtaining material for biochemical assays, proteomics, and genomic or transcriptomic profiling due to small-size and complex tissues. However, the recent development of techniques has started allowing us the usage of a low amount of material for these analyses and now we can strategize many new experiments. The method for enrichment or isolation of rare populations of cells is still challenging and should meaningfully influence the reliability of the results. Here, we provide our semi-optimized protocol of enrichment of undifferentiated germ cells and somatic cells from non-tumorous Drosophila testis, which we have successfully improved after multiple trials.


Asunto(s)
Proteínas de Drosophila , Testículo , Masculino , Animales , Testículo/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Reproducibilidad de los Resultados , Diferenciación Celular/genética , Células Germinativas/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo
12.
bioRxiv ; 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37131641

RESUMEN

In the Drosophila germline stem cell system, maintenance of the stem cell pool requires "dedifferentiation", in which differentiating cells reattach to the niche and reacquire stem cell properties. However, the mechanism of dedifferentiation remains poorly understood. Here, using long-term live imaging, we show that dedifferentiated cells immediately re-enter mitosis with correct spindle orientation after reattachment to the niche. Analysis of cell cycle markers revealed that these dedifferentiating cells are all in G2 phase. In addition, we found that the observed G2 block during dedifferentiation likely corresponds to a centrosome orientation checkpoint (COC), a previously reported polarity checkpoint. We show that re-activation of a COC is likely required for the dedifferentiation thus ensuring asymmetric division even in dedifferentiated stem cells. Taken together, our study demonstrates the remarkable ability of dedifferentiating cells to reacquire the ability to divide asymmetrically.

13.
PLoS One ; 17(11): e0276704, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36342916

RESUMEN

Microtubule acetylation is found in populations of stable, long-lived microtubules, occurring on the conserved lysine 40 (K40) residue of α-tubulin by α-tubulin acetyltransferases (αTATs). α-tubulin K40 acetylation has been shown to stabilize microtubules via enhancing microtubule resilience against mechanical stress. Here we show that a previously uncharacterized αTAT, Drosophila CG17003/leaky (lky), is required for α-tubulin K40 acetylation in early germ cells in Drosophila ovary. We found that loss of lky resulted in a progressive egg chamber fusion phenotype accompanied with mislocalization of germline-specific Vasa protein in somatic follicle cells. The same phenotype was observed upon replacement of endogenous α-tubulin84B with non-acetylatable α-tubulin84BK40A, suggesting α-tubulin K40 acetylation is responsible for the phenotype. Chemical disturbance of microtubules by Colcemid treatment resulted in a mislocalization of Vasa in follicle cells within a short period of time (~30 min), suggesting that the observed mislocalization is likely caused by direct leakage of cellular contents between germline and follicle cells. Taken together, this study provides a new function of α-tubulin acetylation in maintaining the cellular identity possibly by preventing the leakage of tissue-specific gene products between juxtaposing distinct cell types.


Asunto(s)
Drosophila , Tubulina (Proteína) , Animales , Femenino , Acetilación , Tubulina (Proteína)/metabolismo , Drosophila/metabolismo , Ovario/metabolismo , Microtúbulos/metabolismo , Células Germinativas/metabolismo
14.
Nat Commun ; 13(1): 3981, 2022 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-35810185

RESUMEN

Pairing of homologous chromosomes in somatic cells provides the opportunity of interchromosomal interaction between homologous gene regions. In the Drosophila male germline, the Stat92E gene is highly expressed in a germline stem cell (GSC) and gradually downregulated during the differentiation. Here we show that the pairing of Stat92E is always tight in GSCs and immediately loosened in differentiating daughter cells, gonialblasts (GBs). Disturbance of Stat92E pairing by relocation of one locus to another chromosome or by knockdown of global pairing/anti-pairing factors both result in a failure of Stat92E downregulation, suggesting that the pairing is required for the decline in transcription. Furthermore, the Stat92E enhancer, but not its transcription, is required for the change in pairing state, indicating that pairing is not a consequence of transcriptional changes. Finally, we show that the change in Stat92E pairing is dependent on asymmetric histone inheritance during the asymmetric division of GSCs. Taken together, we propose that the changes in Stat92E pairing status is an intrinsically programmed mechanism for enabling prompt cell fate switch during the differentiation of stem cells.


Asunto(s)
Proteínas de Drosophila , Alelos , Animales , Diferenciación Celular/genética , Drosophila/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Células Germinativas
15.
Methods Mol Biol ; 2346: 79-90, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33460026

RESUMEN

The Drosophila ovary is an exceptional model for studying cell-cell interactions in vivo. Cells communicate with each other in a highly coordinated manner. Accurate spatiotemporal regulation of cell-cell interaction is critical for the development of eggs. Ultrastructural analysis using electron microscopy (EM) permits the visualization of both cells and subcellular signaling structures with high resolution. Here we describe a method for the processing of intact fly ovaries by scanning electron microscopy (SEM).


Asunto(s)
Ovario/ultraestructura , Animales , Comunicación Celular , Drosophila , Femenino , Ovario/citología
16.
Cells ; 9(2)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31979180

RESUMEN

The Drosophila ovary offers a suitable model system to study the mechanisms that orchestrate diverse cellular processes. Oogenesis starts from asymmetric stem cell division, proper differentiation and the production of fully patterned oocytes equipped with all the maternal information required for embryogenesis. Spatial and temporal regulation of cell-cell interaction is particularly important to fulfill accurate biological outcomes at each step of oocyte development. Progress has been made in understanding diverse cell physiological regulation of signaling. Here we review the roles of specialized cellular machinery in cell-cell communication in different stages of oogenesis.


Asunto(s)
Comunicación Celular , Drosophila/citología , Oocitos/citología , Animales , Exocitosis , Transducción de Señal , Nicho de Células Madre
17.
Methods Mol Biol ; 1463: 49-62, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27734346

RESUMEN

Asymmetric cell division (ACD) is utilized in many stem cell systems to produce two daughter cells with different cell fates. Despite the fundamental importance of ACD during development and tissue homeostasis, the nature of ACD is far from being fully understood. Step-by-step observation of events during ACD allows us to understand processes that lead to ACD. Here we describe examples of how we evaluate ACD in vivo using the Drosophila male germline stem cell system.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Espermatozoides/citología , Animales , División Celular Asimétrica , Regulación del Desarrollo de la Expresión Génica , Homeostasis , Masculino , Huso Acromático/metabolismo
18.
Sci Rep ; 7(1): 2502, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28566755

RESUMEN

Although the mechanisms that balance self-renewal and differentiation of a stem cell lineage have been extensively studied, it remains poorly understood how tissues that contain multiple stem cell lineages maintain balanced proliferation among distinct lineages: when stem cells of a particular lineage proliferate, how do the other lineages respond to maintain the correct ratio of cells among linages? Here, we show that Merlin (Mer), a homolog of the human tumor suppressor neurofibromatosis 2, is required to coordinate proliferation of germline stem cells (GSCs) and somatic cyst stem cells (CySCs) in the Drosophila testis. Mer mutant CySCs fail to coordinate their proliferation with that of GSCs in multiple settings, and can be triggered to undergo tumorous overproliferation. Mer executes its function by stabilizing adherens junctions. Given the known role of Mer in contact-dependent inhibition of proliferation, we propose that the proliferation of CySCs are regulated by crowdedness, or confluency, of cells in their lineage with respect to that of germline, thereby coordinating the proliferation of two lineages.


Asunto(s)
Proliferación Celular/genética , Neurofibromina 2/genética , Células Madre/citología , Testículo/crecimiento & desarrollo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Masculino , Transducción de Señal/genética , Nicho de Células Madre/genética , Testículo/metabolismo
19.
Trends Cell Biol ; 26(7): 526-534, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27032616

RESUMEN

Information exchange between different cells makes multicellular life possible. Signaling between cells can occur over long distances, as in the case of hormone signaling, or it can take place over short distances between immediately juxtaposed neighbors, as in the case of stem cell-niche signaling. The ability of signal-sending and -receiving cells to communicate with one another in a specific manner is of paramount importance in the proper development and function of tissues. Growing evidence indicates that different cellular protrusions help to achieve specificity in signaling that occurs between distinct cell types. Here, we focus on new roles for cellular protrusions in cell-to-cell communication, drawing special attention to how stem cells use specialized extensions to promote reception of self-renewing signals emanating from the niche.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Transducción de Señal , Animales , Cilios/metabolismo , Humanos , Microtúbulos/metabolismo , Nanotubos/química , Células Madre/metabolismo
20.
G3 (Bethesda) ; 6(12): 3849-3857, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27678520

RESUMEN

Conserved from yeast to humans, the Paf1 complex participates in a number of diverse processes including transcriptional initiation and polyadenylation. This complex typically includes five proteins: Paf1, Rtf1, Cdc73, Leo1, and Ctr9. Previous efforts identified clear Drosophila homologs of Paf1, Rtf1, and Cdc73 based on sequence similarity. Further work showed that these proteins help to regulate gene expression and are required for viability. To date, a Drosophila homolog of Ctr9 has remained uncharacterized. Here, we show that the gene CG2469 encodes a functional Drosophila Ctr9 homolog. Both human and Drosophila Ctr9 localize to the nuclei of Drosophila cells and appear enriched in histone locus bodies. RNAi knockdown of Drosophila Ctr9 results in a germline stem cell loss phenotype marked by defects in the morphology of germ cell nuclei. A molecular null mutation of Drosophila Ctr9 results in lethality and a human cDNA CTR9 transgene rescues this phenotype. Clonal analysis in the ovary using this null allele reveals that loss of Drosophila Ctr9 results in a reduction of global levels of histone H3 trimethylation of lysine 4 (H3K4me3), but does not compromise the maintenance of stem cells in ovaries. Given the differences between the null mutant and RNAi knockdown phenotypes, the germ cell defects caused by RNAi likely result from the combined loss of Drosophila Ctr9 and other unidentified genes. These data provide further evidence that the function of this Paf1 complex component is conserved across species.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/embriología , Drosophila/genética , Genes Esenciales , Proteínas Nucleares/genética , Fosfoproteínas/genética , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Evolución Molecular , Femenino , Técnicas de Inactivación de Genes , Estudios de Asociación Genética , Células Germinativas/metabolismo , Humanos , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fenotipo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Unión Proteica , Interferencia de ARN , Células Madre/metabolismo , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA