Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 132(3): 463-73, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18267076

RESUMEN

Primordial germ cell (PGC) migration in zebrafish is directed by the chemokine SDF-1a that activates its receptor CXCR4b. Little is known about the molecular mechanisms controlling the distribution of this chemoattractant in vivo. We demonstrate that the activity of a second SDF-1/CXCL12 receptor, CXCR7, is crucial for proper migration of PGCs toward their targets. We show that CXCR7 functions primarily in the somatic environment rather than within the migrating cells. In CXCR7 knocked-down embryos, the PGCs exhibit a phenotype that signifies defects in SDF-1a gradient formation as the cells fail to polarize effectively and to migrate toward their targets. Indeed, somatic cells expressing CXCR7 show enhanced internalization of the chemokine suggesting that CXCR7 acts as a sink for SDF-1a, thus allowing the dynamic changes in the transcription of sdf-1a to be mirrored by similar dynamics at the protein level.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/metabolismo , Células Germinativas/citología , Receptores CXCR/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Polaridad Celular , Embrión no Mamífero/citología , Regulación del Desarrollo de la Expresión Génica , Receptores CXCR/genética , Proteínas de Pez Cebra/genética
2.
N Engl J Med ; 388(24): 2303, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37314724
3.
Mol Ther ; 27(6): 1126-1138, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31005597

RESUMEN

Clinical success of autologous CD19-directed chimeric antigen receptor T cells (CAR Ts) in acute lymphoblastic leukemia and non-Hodgkin lymphoma suggests that CAR Ts may be a promising therapy for hematological malignancies, including multiple myeloma. However, autologous CAR T therapies have limitations that may impact clinical use, including lengthy vein-to-vein time and manufacturing constraints. Allogeneic CAR T (AlloCAR T) therapies may overcome these innate limitations of autologous CAR T therapies. Unlike autologous cell therapies, AlloCAR T therapies employ healthy donor T cells that are isolated in a manufacturing facility, engineered to express CARs with specificity for a tumor-associated antigen, and modified using gene-editing technology to limit T cell receptor (TCR)-mediated immune responses. Here, transcription activator-like effector nuclease (TALEN) gene editing of B cell maturation antigen (BCMA) CAR Ts was used to confer lymphodepletion resistance and reduced graft-versus-host disease (GvHD) potential. The safety profile of allogeneic BCMA CAR Ts was further enhanced by incorporating a CD20 mimotope-based intra-CAR off switch enabling effective CAR T elimination in the presence of rituximab. Allogeneic BCMA CAR Ts induced sustained antitumor responses in mice supplemented with human cytokines, and, most importantly, maintained their phenotype and potency after scale-up manufacturing. This novel off-the-shelf allogeneic BCMA CAR T product is a promising candidate for clinical evaluation.


Asunto(s)
Antígeno de Maduración de Linfocitos B/inmunología , Trasplante de Células/métodos , Inmunoterapia Adoptiva/métodos , Mieloma Múltiple/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/trasplante , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno de Maduración de Linfocitos B/genética , Donantes de Sangre , Línea Celular Tumoral , Trasplante de Células/efectos adversos , Citotoxicidad Inmunológica/genética , Edición Génica , Vectores Genéticos , Enfermedad Injerto contra Huésped/terapia , Humanos , Inmunoterapia Adoptiva/efectos adversos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/patología , Supervivencia sin Progresión , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Rituximab/uso terapéutico , Linfocitos T/metabolismo , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Transducción Genética , Trasplante Homólogo/métodos
4.
Development ; 138(14): 2909-14, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21693511

RESUMEN

The active migration of primordial germ cells (PGCs) from their site of specification towards their target is a valuable model for investigating directed cell migration within the complex environment of the developing embryo. In several vertebrates, PGC migration is guided by Cxcl12, a member of the chemokine superfamily. Interestingly, two distinct Cxcl12 paralogs are expressed in zebrafish embryos and contribute to the chemotattractive landscape. Although this offers versatility in the use of chemokine signals, it also requires a mechanism through which migrating cells prioritize the relevant cues that they encounter. Here, we show that PGCs respond preferentially to one of the paralogs and define the molecular basis for this biased behavior. We find that a single amino acid exchange switches the relative affinity of the Cxcl12 ligands for one of the duplicated Cxcr4 receptors, thereby determining the functional specialization of each chemokine that elicits a distinct function in a distinct process. This scenario represents an example of protein subfunctionalization--the specialization of two gene copies to perform complementary functions following gene duplication--which in this case is based on receptor-ligand interaction. Such specialization increases the complexity and flexibility of chemokine signaling in controlling concurrent developmental processes.


Asunto(s)
Movimiento Celular/fisiología , Quimiocina CXCL12/metabolismo , Evolución Molecular , Células Germinativas/fisiología , Receptores CXCR4/metabolismo , Pez Cebra/embriología , Sustitución de Aminoácidos , Animales , Línea Celular , Quimiocina CXCL12/genética , Técnicas de Silenciamiento del Gen , Humanos , Hibridación in Situ , Microscopía Confocal , Espectrometría de Fluorescencia , Pez Cebra/metabolismo
5.
Cancer Immunol Res ; 10(9): 1069-1083, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35881865

RESUMEN

Although cytokine support can enhance CAR T-cell function, coadministering cytokines or engineering CAR T cells to secrete cytokines can result in toxicities. To mitigate these safety risks, we engineered iTurboCAR T cells that coexpress a novel inducible Turbo (iTurbo) cytokine signaling domain. iTurbo domains consist of modular components that are customizable to a variety of activating inputs, as well as cytokine signaling outputs multiplexable for combinatorial signaling outcomes. Unlike most canonical cytokine receptors that are heterodimeric, iTurbo domains leverage a compact, homodimeric design that minimizes viral vector cargo. Using an iTurbo domain activated by the clinically validated dimerizer, AP1903, homodimeric iTurbo domains instigated signaling that mimicked the endogenous heterodimeric cytokine receptor. Different iTurbo domains programmed iTurboCAR T cells toward divergent phenotypes and resulted in improved antitumor efficacy. iTurbo domains, therefore, offer the flexibility for user-programmable signaling outputs, permitting control over cellular phenotype and function while minimizing viral cargo footprint.


Asunto(s)
Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T , Citocinas , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal , Linfocitos T
6.
Cancer Res Commun ; 2(3): 158-171, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-36874402

RESUMEN

Multiple myeloma remains an incurable plasma cell malignancy despite the rapidly evolving treatment landscape. Chimeric antigen receptor T cells targeted against BCMA have recently shown great promise in relapsed refractory multiple myeloma; however, all patients ultimately still progress from their disease. Lack of CAR T-cell persistence, impaired T-cell fitness in autologous CAR T-cell products and the presence of an immunosuppressive bone marrow (BM) microenvironment are contributory factors to treatment failure. We generated anti-BCMA CAR T cells from healthy donors (HD) and patients with multiple myeloma at different stages of disease to compare their T-cell profile, fitness, and cytotoxic activity in preclinical studies. We also used an ex vivo assay with multiple myeloma BM biopsies from distinct genomic subgroups to test the efficacy of HD-derived CAR T cells in a clinically relevant model. HD volunteers showed increased T-cell counts, higher CD4/CD8 ratio, and expanded naïve T-cell population compared with patients with multiple myeloma. After anti-BCMA CAR T-cell production, patients with relapsed multiple myeloma had lower frequencies of CAR+ T cells, decreased central memory phenotype, and increased checkpoint inhibitory markers compared with HD-derived products, which compromised their expansion and cytotoxicity against multiple myeloma cells in vitro. Importantly, HD-derived CAR T cells efficiently killed primary multiple myeloma cells within the BM microenvironment of different multiple myeloma genomic subgroups and their cytotoxic activity could be boosted with gamma secretase inhibitors. In conclusion, allogeneic anti-BCMA CAR T cells are a potential therapeutic strategy for patients with relapsed multiple myeloma and should be further developed in the clinic. Significance: Multiple myeloma is an incurable cancer of the plasma cells. A new therapy with anti-BCMA CAR T cells - the patient's own T cells genetically engineered to find and kill myeloma cancer cells - has shown encouraging results. Unfortunately, patients still relapse. In this study, we propose to use T cells from HD volunteers, which have a stronger T-cell fitness, higher cancer killing capacity, and are ready to be administered when needed.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple , Receptores Quiméricos de Antígenos , Humanos , Mieloma Múltiple/terapia , Receptores Quiméricos de Antígenos/genética , Inhibidores y Moduladores de Gamma Secretasa , Recurrencia Local de Neoplasia , Linfocitos T , Microambiente Tumoral
7.
Nat Commun ; 12(1): 710, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514714

RESUMEN

Antibody-based therapeutics have experienced a rapid growth in recent years and are now utilized in various modalities spanning from conventional antibodies, antibody-drug conjugates, bispecific antibodies to chimeric antigen receptor (CAR) T cells. Many next generation antibody therapeutics achieve enhanced potency but often increase the risk of adverse events. Antibody scaffolds capable of exhibiting inducible affinities could reduce the risk of adverse events by enabling a transient suspension of antibody activity. To demonstrate this, we develop conditionally activated, single-module CARs, in which tumor antigen recognition is directly modulated by an FDA-approved small molecule drug. The resulting CAR T cells demonstrate specific cytotoxicity of tumor cells comparable to that of traditional CARs, but the cytotoxicity is reversibly attenuated by the addition of the small molecule. The exogenous control of conditional CAR T cell activity allows continual modulation of therapeutic activity to improve the safety profile of CAR T cells across all disease indications.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia Adoptiva/métodos , Metotrexato/administración & dosificación , Neoplasias/terapia , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/efectos de los fármacos , Animales , Línea Celular Tumoral , Terapia Combinada/métodos , Femenino , Células HEK293 , Humanos , Inmunoterapia Adoptiva/efectos adversos , Ratones , Neoplasias/inmunología , Cultivo Primario de Células , Receptores Quiméricos de Antígenos/inmunología , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T/efectos de los fármacos , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Sci STKE ; 2007(383): pe16, 2007 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-17456805

RESUMEN

Cell differentiation, cell proliferation, cell death, and cell migration are tightly controlled during animal development and adult homeostasis. Failure to regulate these processes can result in tumor formation and metastasis. Aberrant cells are therefore often cleared by induction of cell death. Recent work has elucidated the mechanism of elimination of mouse primordial germ cells that fail to migrate properly and highlights the similarity of this mechanism to those governing the same phenomenon in Drosophila. In addition, these studies underscore the different functions a single signaling pathway can have in controlling cell survival, cell proliferation, and cell migration during different phases of development.


Asunto(s)
Movimiento Celular/fisiología , Drosophila melanogaster/citología , Células Germinativas/citología , Animales , Apoptosis , Diferenciación Celular , Quimiotaxis , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Embrión no Mamífero/ultraestructura , Femenino , Gónadas/citología , Gónadas/embriología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Modelos Biológicos , Fosfatidato Fosfatasa/genética , Fosfatidato Fosfatasa/fisiología , Fosfolípidos/fisiología , Proteínas Proto-Oncogénicas c-kit/fisiología , Factor de Células Madre/fisiología
9.
Sci Rep ; 8(1): 8972, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29895885

RESUMEN

CAR T-cell therapies hold great promise for treating a range of malignancies but are however challenged by the complexity of their production and by the adverse events related to their activity. Here we report the development of the CubiCAR, a tri-functional CAR architecture that enables CAR T-cell detection, purification and on-demand depletion by the FDA-approved antibody Rituximab. This novel architecture has the potential to streamline the manufacturing of CAR T-cells, allow their tracking and improve their overall safety.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/cirugía , Receptores Quiméricos de Antígenos/inmunología , Rituximab/farmacología , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/patología
10.
JCI Insight ; 1(20): e89289, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27942588

RESUMEN

T cells that enter tumors are largely tolerized, but how that process is choreographed and how the ensuing "dysfunctional" tumor-infiltrating lymphocytes (TILs) are maintained are poorly understood and are difficult to assess in spontaneous disease. We exploited an autochthonous model of breast cancer for high-resolution imaging of the early and later stages of tumor residence to understand the relationships between cellular behaviors and cellular phenotypes. "Dysfunctional" differentiation began within the first days of tumor residence with an initial phase in which T cells arrest, largely on tumor-associated macrophages. Within 10 days, cellular motility increased and resembled a random walk, suggesting a relative absence of TCR signaling. We then studied the concurrent and apparently contradictory phenomenon that many of these cells express molecular markers of activation and were visualized undergoing active cell division. We found that whereas proliferation did not require ongoing TCR/ZAP70 signaling, instead this is driven in part by intratumoral IL-15 cytokine. Thus, TILs undergo sequential reprogramming by the tumor microenvironment and are actively retained, even while being antigen insensitive. We conclude that this program effectively fills the niche with ineffective yet cytokine-dependent TILs, and we propose that these might compete with new clones, when they arise.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Interleucina-15/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T/inmunología , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína Tirosina Quinasa ZAP-70/metabolismo
11.
Dev Cell ; 34(5): 493-504, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26321127

RESUMEN

Postnatal organogenesis occurs in an immune competent environment and is tightly controlled by interplay between positive and negative regulators. Innate immune cells have beneficial roles in postnatal tissue remodeling, but roles for the adaptive immune system are currently unexplored. Here we show that adaptive immune responses participate in the normal postnatal development of a non-lymphoid epithelial tissue. Since the mammary gland (MG) is the only organ developing predominantly after birth, we utilized it as a powerful system to study adaptive immune regulation of organogenesis. We found that antigen-mediated interactions between mammary antigen-presenting cells and interferon-γ (IFNγ)-producing CD4+ T helper 1 cells participate in MG postnatal organogenesis as negative regulators, locally orchestrating epithelial rearrangement. IFNγ then affects luminal lineage differentiation. This function of adaptive immune responses, regulating normal development, changes the paradigm for studying players of postnatal organogenesis and provides insights into immune surveillance and cancer transformation.


Asunto(s)
Inmunidad Adaptativa/inmunología , Células Presentadoras de Antígenos/inmunología , Mama/inmunología , Células Epiteliales/citología , Epitelio/metabolismo , Organogénesis/inmunología , Animales , Células Presentadoras de Antígenos/citología , Mama/crecimiento & desarrollo , Mama/metabolismo , Células Epiteliales/inmunología , Epitelio/inmunología , Femenino , Humanos , Inmunidad Innata/inmunología , Interferón gamma/metabolismo , Ratones
12.
Cancer Cell ; 26(5): 638-52, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-25446897

RESUMEN

It is well understood that antigen-presenting cells (APCs) within tumors typically do not maintain cytotoxic T cell (CTL) function, despite engaging them. Across multiple mouse tumor models and human tumor biopsies, we have delineated the intratumoral dendritic cell (DC) populations as distinct from macrophage populations. Within these, CD103(+) DCs are extremely sparse and yet remarkably capable CTL stimulators. These are uniquely dependent on IRF8, Zbtb46, and Batf3 transcription factors and are generated by GM-CSF and FLT3L cytokines. Regressing tumors have higher proportions of these cells, T-cell-dependent immune clearance relies on them, and abundance of their transcripts in human tumors correlates with clinical outcome. This cell type presents opportunities for prognostic and therapeutic approaches across multiple cancer types.


Asunto(s)
Células Dendríticas/inmunología , Neoplasias Mamarias Experimentales/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD/metabolismo , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Femenino , Humanos , Inmunoterapia Adoptiva , Macrófagos/metabolismo , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Ratones Endogámicos C57BL , Células Mieloides/inmunología , Células Mieloides/metabolismo , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral
13.
Cancer Cell ; 21(3): 402-17, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22439936

RESUMEN

The nature and site of tumor-antigen presentation to immune T cells by bone-marrow-derived cells within the tumor microenvironment remains unresolved. We generated a fluorescent mouse model of spontaneous immunoevasive breast cancer and identified a subset of myeloid cells with significant similarity to dendritic cells and macrophages that constitutively ingest tumor-derived proteins and present processed tumor antigens to reactive T cells. Using intravital live imaging, we determined that infiltrating tumor-specific T cells engage in long-lived interactions with these cells, proximal to the tumor. In vitro, these cells capture cytotoxic T cells in signaling-competent conjugates but do not support full activation or sustain cytolysis. The spatiotemporal dynamics revealed here implicate nonproductive interactions between T cells and antigen-presenting cells on the tumor margin.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Reactividad Cruzada , Células Dendríticas/inmunología , Linfocitos T/inmunología , Microambiente Tumoral , Animales , Presentación de Antígeno , Neoplasias de la Mama/patología , Femenino , Humanos , Activación de Linfocitos , Ratones , Células Mieloides/inmunología
14.
Nat Cell Biol ; 12(1): 47-53; sup pp 1-11, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20010816

RESUMEN

Cell migration is central to embryonic development, homeostasis and disease, processes in which cells move as part of a group or individually. Whereas the mechanisms controlling single-cell migration in vitro are relatively well understood, less is known about the mechanisms promoting the motility of individual cells in vivo. In particular, it is not clear how cells that form blebs in their migration use those protrusions to bring about movement in the context of the three-dimensional cellular environment. Here we show that the motility of chemokine-guided germ cells within the zebrafish embryo requires the function of the small Rho GTPases Rac1 and RhoA, as well as E-cadherin-mediated cell-cell adhesion. Using fluorescence resonance energy transfer we demonstrate that Rac1 and RhoA are activated in the cell front. At this location, Rac1 is responsible for the formation of actin-rich structures, and RhoA promotes retrograde actin flow. We propose that these actin-rich structures undergoing retrograde flow are essential for the generation of E-cadherin-mediated traction forces between the germ cells and the surrounding tissue and are therefore crucial for cell motility in vivo.


Asunto(s)
Cadherinas/metabolismo , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Germinativas/metabolismo , Proteína de Unión al GTP rac1/fisiología , Proteína de Unión al GTP rhoA/fisiología , Animales , Cadherinas/genética , Polaridad Celular , Células Cultivadas , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Pez Cebra
16.
Cell Adh Migr ; 2(2): 69-70, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19262101

RESUMEN

Primordial Germ Cell (PGC) migration in zebrafish is guided by SDF-1a. Binding of this chemokine to its receptor CXCR4b activates downstream signalling cascades leading to cell polarization and directed migration towards the attractant source. Despite the detailed information available concerning the role of SDF-1 in guiding the PGCs to their targets, little was known regarding the molecular mechanisms controlling the distribution of SDF-1a within the tissue. We have recently shown that the activity of a second SDF-1/CXCL12 receptor, CXCR7 is crucial for proper migration of PGCs. Although CXCR4 and CXCR7 are structurally related and serve as receptors for the same ligand, they appear to serve very different functions during PGC migration. Here we discuss a model according to which CXCR4b translates the polarized distribution of SDF-1 into directed PGC migration, while CXCR7 acts as a high-affinity decoy receptor and facilitates the migration of PGCs by shaping the distribution of the chemokine in the environment.


Asunto(s)
Movimiento Celular , Células Germinativas/citología , Células Germinativas/metabolismo , Receptores CXCR/metabolismo , Pez Cebra/metabolismo , Animales , Quimiocina CXCL12/metabolismo , Transducción de Señal
17.
Cell ; 131(7): 1273-86, 2007 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-18155131

RESUMEN

MicroRNAs (miRNAs) are inhibitors of gene expression capable of controlling processes in normal development and cancer. In mammals, miRNAs use a seed sequence of 6-8 nucleotides (nt) to associate with 3' untranslated regions (3'UTRs) of mRNAs and inhibit their expression. Intriguingly, occasionally not only the miRNA-targeting site but also sequences in its vicinity are highly conserved throughout evolution. We therefore hypothesized that conserved regions in mRNAs may serve as docking platforms for modulators of miRNA activity. Here we demonstrate that the expression of dead end 1 (Dnd1), an evolutionary conserved RNA-binding protein (RBP), counteracts the function of several miRNAs in human cells and in primordial germ cells of zebrafish by binding mRNAs and prohibiting miRNAs from associating with their target sites. These effects of Dnd1 are mediated through uridine-rich regions present in the miRNA-targeted mRNAs. Thus, our data unravel a novel role of Dnd1 in protecting certain mRNAs from miRNA-mediated repression.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Células Germinativas/metabolismo , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Proteínas de Pez Cebra/metabolismo , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Conexina 43/genética , Conexina 43/metabolismo , Secuencia Conservada , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/genética , Secuencias Reguladoras de Ácido Ribonucleico , Transfección , Pez Cebra , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA