Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Immunol ; 206(10): 2271-2276, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33941655

RESUMEN

T cell development is predicated on the successful rearrangement of the TCR gene loci, which encode for Ag-specific receptors. Recombination-activating gene (RAG) 2 is required for TCR gene rearrangements, which occur during specific stages of T cell development. In this study, we differentiated human pluripotent stem cells with a CRISPR/Cas9-directed deletion of the RAG2 gene (RAG2-KO) to elucidate the requirement for the TCR ß-chain in mediating ß-selection during human T cell development. In stark contrast to mice, human RAG2-KO T lineage progenitors progressed to the CD4+CD8+ double-positive (DP) stage in the absence of TCRß rearrangements. Nonetheless, RAG2-KO DPs retrovirally transduced to express a rearranged TCR ß-chain showed increased survival and proliferation as compared with control-transduced RAG2-KO DPs. Furthermore, transcriptomic analysis showed that TCRß- and control-transduced RAG2-KO DPs differed in gene pathways related to survival and proliferation. Our results provide important insights as to the distinct requirement for the TCR ß-chain during human T cell development.


Asunto(s)
Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Diferenciación Celular/genética , Células Madre Embrionarias Humanas/citología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Técnicas de Inactivación de Genes , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T/genética , Hematopoyesis/genética , Humanos , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Transducción Genética
2.
Trends Immunol ; 37(12): 889-901, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27789110

RESUMEN

T cells, as orchestrators of the adaptive immune response, serve important physiological and potentially therapeutic roles, for example in cancer immunotherapy. T cells are readily isolated from patients; however, the yield of antigen-specific T cells is limited, thus making their clinical use challenging. Therefore, the generation of T lymphocytes from hematopoietic stem/progenitor cells (HSPCs) and human pluripotent stem cells (PSCs) in vitro provides an attractive method for the large-scale production and genetic manipulation of T cells. In this review, we discuss recent strategies for the generation of T cells from human HSPCs and PSCs in vitro. Continued advancement in the generation of human T cells in vitro will expand their benefits and therapeutic potential in the clinic.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Madre Hematopoyéticas/fisiología , Inmunoterapia Adoptiva/métodos , Linfopoyesis , Neoplasias/terapia , Células Madre Pluripotentes/fisiología , Linfocitos T/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Ingeniería Genética , Humanos , Técnicas In Vitro , Neoplasias/inmunología , Técnicas de Cultivo de Órganos , Linfocitos T/trasplante
3.
Blood ; 128(7): 934-47, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27297795

RESUMEN

Novel agents such as the Bcl-2 inhibitor venetoclax (ABT-199) are changing treatment paradigms for chronic lymphocytic leukemia (CLL) but important problems remain. Although some patients exhibit deep and durable responses to venetoclax as a single agent, other patients harbor subpopulations of resistant leukemia cells that mediate disease recurrence. One hypothesis for the origin of resistance to venetoclax is by kinase-mediated survival signals encountered in proliferation centers that may be unique for individual patients. An in vitro microenvironment model was developed with primary CLL cells that could be incorporated into an automated high-content microscopy-based screen of kinase inhibitors (KIs) to identify agents that may improve venetoclax therapy in a personalized manner. Marked interpatient variability was noted for which KIs were effective; nevertheless, sunitinib was identified as the most common clinically available KI effective in overcoming venetoclax resistance. Examination of the underlying mechanisms indicated that venetoclax resistance may be induced by microenvironmental signals that upregulate antiapoptotic Bcl-xl, Mcl-1, and A1, which can be counteracted more efficiently by sunitinib than by ibrutinib or idelalisib. Although patient-specific drug responses are common, for many patients, combination therapy with sunitinib may significantly improve the therapeutic efficacy of venetoclax.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Inhibidores de Proteínas Quinasas/análisis , Inhibidores de Proteínas Quinasas/uso terapéutico , Sulfonamidas/uso terapéutico , Adenina/análogos & derivados , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Microambiente Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Imagenología Tridimensional , Indoles/farmacología , Mutación/genética , Piperidinas , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Quinazolinonas/farmacología , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Sulfonamidas/farmacología , Sunitinib , Regulación hacia Arriba/efectos de los fármacos , Proteína bcl-X/metabolismo
4.
Blood ; 128(6): 783-93, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27301863

RESUMEN

Primary immunodeficiency diseases comprise a group of heterogeneous genetic defects that affect immune system development and/or function. Here we use in vitro differentiation of human induced pluripotent stem cells (iPSCs) generated from patients with different recombination-activating gene 1 (RAG1) mutations to assess T-cell development and T-cell receptor (TCR) V(D)J recombination. RAG1-mutants from severe combined immunodeficient (SCID) patient cells showed a failure to sustain progression beyond the CD3(--)CD4(-)CD8(-)CD7(+)CD5(+)CD38(-)CD31(-/lo)CD45RA(+) stage of T-cell development to reach the CD3(-/+)CD4(+)CD8(+)CD7(+)CD5(+)CD38(+)CD31(+)CD45RA(-) stage. Despite residual mutant RAG1 recombination activity from an Omenn syndrome (OS) patient, similar impaired T-cell differentiation was observed, due to increased single-strand DNA breaks that likely occur due to heterodimers consisting of both an N-terminal truncated and a catalytically dead RAG1. Furthermore, deep-sequencing analysis of TCR-ß (TRB) and TCR-α (TRA) rearrangements of CD3(-)CD4(+)CD8(-) immature single-positive and CD3(+)CD4(+)CD8(+) double-positive cells showed severe restriction of repertoire diversity with preferential usage of few Variable, Diversity, and Joining genes, and skewed length distribution of the TRB and TRA complementary determining region 3 sequences from SCID and OS iPSC-derived cells, whereas control iPSCs yielded T-cell progenitors with a broadly diversified repertoire. Finally, no TRA/δ excision circles (TRECs), a marker of TRA/δ locus rearrangements, were detected in SCID and OS-derived T-lineage cells, consistent with a pre-TCR block in T-cell development. This study compares human T-cell development of SCID vs OS patients, and elucidates important differences that help to explain the wide range of immunologic phenotypes that result from different mutations within the same gene of various patients.


Asunto(s)
Proteínas de Homeodominio/genética , Células Madre Pluripotentes Inducidas/patología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/patología , Linfocitos T/patología , Células Cultivadas , Roturas del ADN , Genes RAG-1 , Humanos , Lactante , Mutación , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Recombinación V(D)J
5.
Methods Mol Biol ; 2580: 249-260, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36374462

RESUMEN

For nearly a generation now, OP9-DL1 and OP9-DL4 cells have provided an efficient and reliable cell system to generate T cells from mouse and human hematopoietic stem cells (HSCs) and pluripotent stem cells. OP9-DL1 and OP9-DL4 were originally derived from the OP9 mouse bone marrow stromal cell line, which was transduced to ectopically express Delta-like 1 or 4 proteins, respectively. OP9-DL cells mimic the thymic microenvironment in that when cocultured with mouse or human (h) HSCs, they interact with and activate Notch receptors present on HSCs, required for T cell differentiation. The HSC/OP9-DL cocultures require additional cytokines that are necessary for survival and proliferation of hematopoietic cells. For hHSCs, these factors are interleukin-7 (IL-7), stem cell factor (SCF), and FMS-like tyrosine kinase 3 ligand (FLT3L) that are normally exogenously added to the cocultures. In this chapter, we describe methods for establishing a novel and improved version of OP9-DL4 cells, called OP9-DL4-7FS cells that circumvent the addition of these costly cytokines, by transducing OP9-DL4 cell line to express human IL-7, FLT3L, and SCF (7FS). Herein, we describe the protocol for the generation of OP9-DL4-7FS cells and the conditions for OP9-DL4-7FS/hHSC coculture to support T cell lineage initiation and expansion while comparing it to the now "classic" OP9-DL4 coculture. The use of OP9-DL4-7FS cell system will provide an improved and cost-effective method to the commonly used OP9-DL/HSC coculture for studying both mouse and human T cell development.


Asunto(s)
Citocinas , Interleucina-7 , Humanos , Ratones , Animales , Interleucina-7/metabolismo , Citocinas/metabolismo , Diferenciación Celular , Células Madre Hematopoyéticas , Técnicas de Cocultivo , Linfocitos T , Células del Estroma/metabolismo
6.
J Cell Sci ; 123(Pt 2): 236-45, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20026641

RESUMEN

Disruption of the gene encoding protein tyrosine kinase 6 (PTK6) leads to increased growth, impaired enterocyte differentiation and higher levels of nuclear beta-catenin in the mouse small intestine. Here, we demonstrate that PTK6 associates with nuclear and cytoplasmic beta-catenin and inhibits beta-catenin- and T-cell factor (TCF)-mediated transcription. PTK6 directly phosphorylates beta-catenin on Tyr64, Tyr142, Tyr331 and/or Tyr333, with the predominant site being Tyr64. However, mutation of these sites does not abrogate the ability of PTK6 to inhibit beta-catenin transcriptional activity. Outcomes of PTK6-mediated regulation appear to be dependent on its intracellular localization. In the SW620 colorectal adenocarcinoma cell line, nuclear-targeted PTK6 negatively regulates endogenous beta-catenin/TCF transcriptional activity, whereas membrane-targeted PTK6 enhances beta-catenin/TCF regulated transcription. Levels of TCF4 and the transcriptional co-repressor TLE/Groucho increase in SW620 cells expressing nuclear-targeted PTK6. Knockdown of PTK6 in SW620 cells leads to increased beta-catenin/TCF transcriptional activity and increased expression of beta-catenin/TCF target genes Myc and Survivin. Ptk6-null BAT-GAL mice, containing a beta-catenin-activated LacZ reporter transgene, have increased levels of beta-galactosidase expression in the gastrointestinal tract. The ability of PTK6 to negatively regulate beta-catenin/TCF transcription by modulating levels of TCF4 and TLE/Groucho could contribute to its growth-inhibitory activities in vivo.


Asunto(s)
Espacio Intracelular/enzimología , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , beta Catenina/metabolismo , Familia-src Quinasas/metabolismo , Animales , Línea Celular Tumoral , Regulación hacia Abajo/genética , Activación Enzimática , Genes Reporteros , Humanos , Intestinos/enzimología , Intestinos/patología , Ratones , Ratones Transgénicos , Fosforilación , Fosfotirosina/metabolismo , Unión Proteica , Especificidad por Sustrato , Factores de Transcripción TCF/metabolismo , Transcripción Genética , beta Catenina/genética
7.
Biochim Biophys Acta ; 1806(1): 66-73, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20193745

RESUMEN

Protein tyrosine kinase 6 (PTK6), also referred to as breast tumor kinase BRK, is a member of a distinct family of kinases that is evolutionarily related to the SRC family of tyrosine kinases. While not expressed in the normal mammary gland, PTK6 expression is detected in a large proportion of human mammary gland tumors. In breast tumor cells, PTK6 promotes growth factor signaling and cell migration. PTK6 expression is also increased in a number of other epithelial tumors, including ovarian and colon cancer. In contrast, PTK6 is expressed in diverse normal epithelia, including the linings of the gastrointestinal tract, skin and prostate, where its expression correlates with cell cycle exit and differentiation. Disruption of the mouse Ptk6 gene leads to increased growth and impaired differentiation in the small intestine that is accompanied by increased AKT and Wnt signaling. Following total body irradiation, PTK6 expression is induced in proliferating progenitor cells of the intestine, where it plays an essential role in DNA-damage induced apoptosis. A distinguishing feature of PTK6 is its flexibility in intracellular localization, due to a lack of amino-terminal myristoylation/palmitoylation. Recently a number of substrates of PTK6 have been identified, including nuclear RNA-binding proteins and transcription factors. We discuss PTK6 signaling, its apparent conflicting roles in cancer and normal epithelia, and its potential as a therapeutic target in epithelial cancers.


Asunto(s)
Proteínas de Neoplasias/fisiología , Neoplasias/etiología , Proteínas Tirosina Quinasas/fisiología , Animales , Humanos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neoplasias/tratamiento farmacológico , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Transducción de Señal
8.
EMBO Rep ; 10(9): 1036-42, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19644500

RESUMEN

The tumour suppressor ARF (alternative reading frame), which is mutated or silenced in various tumours, has a crucial role in tumour surveillance to suppress unwarranted cell growth and proliferation. ARF has also been linked to the DNA-damage-induced response of p53 because of its ability to inhibit murine double minute 2 (MDM2). Here, however, we provide genetic evidence for a role of ARF in nucleotide excision repair (NER) that is independent of p53. Cells lacking ARF are deficient in NER. Expression of ARF restores the repair activity, which coincides with increased expression of the damaged-DNA recognition protein xeroderma pigmentosum, complementation group C (XPC). We provide evidence that, by disrupting the interaction between E2F transcription factor 4 (E2F4) and DRTF polypeptide 1 (DP1), ARF reduces the interaction of the E2F4-p130 repressor complex with the promoter of XPC to ensure high-level expression of XPC. Together, our results point to an important 'care-taker'-type tumour-suppression function for ARF in NER through the increased expression of XPC.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Factor de Transcripción E2F4/metabolismo , Animales , Secuencia de Bases , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/deficiencia , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Dímeros de Pirimidina/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo
9.
Nat Commun ; 12(1): 5023, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408144

RESUMEN

T cells are pivotal effectors of the immune system and can be harnessed as therapeutics for regenerative medicine and cancer immunotherapy. An unmet challenge in the field is the development of a clinically relevant system that is readily scalable to generate large numbers of T-lineage cells from hematopoietic stem/progenitor cells (HSPCs). Here, we report a stromal cell-free, microbead-based approach that supports the efficient in vitro development of both human progenitor T (proT) cells and T-lineage cells from CD34+cells sourced from cord blood, GCSF-mobilized peripheral blood, and pluripotent stem cells (PSCs). DL4-µbeads, along with lymphopoietic cytokines, induce an ordered sequence of differentiation from CD34+ cells to CD34+CD7+CD5+ proT cells to CD3+αß T cells. Single-cell RNA sequencing of human PSC-derived proT cells reveals a transcriptional profile similar to the earliest thymocytes found in the embryonic and fetal thymus. Furthermore, the adoptive transfer of CD34+CD7+ proT cells into immunodeficient mice demonstrates efficient thymic engraftment and functional maturation of peripheral T cells. DL4-µbeads provide a simple and robust platform to both study human T cell development and facilitate the development of engineered T cell therapies from renewable sources.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas de Unión al Calcio/inmunología , Células Madre Hematopoyéticas/citología , Linfopoyesis , Enfermedades de Inmunodeficiencia Primaria/terapia , Linfocitos T/citología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD34/genética , Antígenos CD34/inmunología , Proteínas de Unión al Calcio/genética , Linaje de la Célula , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/inmunología , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/inmunología , Enfermedades de Inmunodeficiencia Primaria/fisiopatología , Linfocitos T/inmunología , Linfocitos T/trasplante
10.
Stem Cell Reports ; 13(6): 1126-1141, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31813827

RESUMEN

Induced pluripotent stem cells (iPSC) derived from healthy individuals are important controls for disease-modeling studies. Here we apply precision health to create a high-quality resource of control iPSCs. Footprint-free lines were reprogrammed from four volunteers of the Personal Genome Project Canada (PGPC). Multilineage-directed differentiation efficiently produced functional cortical neurons, cardiomyocytes and hepatocytes. Pilot users demonstrated versatility by generating kidney organoids, T lymphocytes, and sensory neurons. A frameshift knockout was introduced into MYBPC3 and these cardiomyocytes exhibited the expected hypertrophic phenotype. Whole-genome sequencing-based annotation of PGPC lines revealed on average 20 coding variants. Importantly, nearly all annotated PGPC and HipSci lines harbored at least one pre-existing or acquired variant with cardiac, neurological, or other disease associations. Overall, PGPC lines were efficiently differentiated by multiple users into cells from six tissues for disease modeling, and variant-preferred healthy control lines were identified for specific disease settings.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Sistemas CRISPR-Cas , Autorrenovación de las Células , Separación Celular , Ectodermo/citología , Ectodermo/metabolismo , Edición Génica , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Organoides , Fenotipo , Linfocitos T/metabolismo , Secuenciación Completa del Genoma
11.
Sci Signal ; 11(533)2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29871911

RESUMEN

The transient receptor potential (TRP) family is a large family of widely expressed ion channels that regulate the intracellular concentration of ions and metals and respond to various chemical and physical stimuli. TRP subfamily M member 7 (TRPM7) is unusual in that it contains both an ion channel and a kinase domain. TRPM7 is a divalent cation channel with preference for Ca2+ and Mg2+ It is required for the survival of DT40 cells, a B cell line; however, deletion of TRPM7 in T cells does not impair their development. We found that expression of TRPM7 was required for B cell development in mice. Mice that lacked TRPM7 in B cells failed to generate peripheral B cells because of a developmental block at the pro-B cell stage. The loss of TRPM7 kinase activity alone did not affect the proportion of peripheral mature B cells or the development of B cells in the bone marrow. However, supplementation with a high concentration of extracellular Mg2+ partially rescued the development of TRPM7-deficient B cells in vitro. Thus, our findings identify a critical role for TRPM7 ion channel activity in B cell development.


Asunto(s)
Linfocitos B/citología , Linfocitos B/fisiología , Linfopoyesis , Magnesio/metabolismo , Células Mieloides/fisiología , Canales Catiónicos TRPM/fisiología , Animales , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología
12.
Nat Commun ; 8: 15380, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28541275

RESUMEN

Efforts to recapitulate haematopoiesis, a process guided by spatial and temporal inductive signals, to generate haematopoietic progenitors from human pluripotent stem cells (hPSCs) have focused primarily on exogenous signalling pathway activation or inhibition. Here we show haemogenic niches can be engineered using microfabrication strategies by micropatterning hPSC-derived haemogenic endothelial (HE) cells into spatially-organized, size-controlled colonies. CD34+VECAD+ HE cells were generated with multi-lineage potential in serum-free conditions and cultured as size-specific haemogenic niches that displayed enhanced blood cell induction over non-micropatterned cultures. Intra-colony analysis revealed radial organization of CD34 and VECAD expression levels, with CD45+ blood cells emerging primarily from the colony centroid area. We identify the induced interferon gamma protein (IP-10)/p-38 MAPK signalling pathway as the mechanism for haematopoietic inhibition in our culture system. Our results highlight the role of spatial organization in hPSC-derived blood generation, and provide a quantitative platform for interrogating molecular pathways that regulate human haematopoiesis.


Asunto(s)
Hemangioblastos/citología , Hemangioblastos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Cadherinas/metabolismo , Diferenciación Celular , Ingeniería Celular/métodos , Línea Celular , Linaje de la Célula , Quimiocina CXCL10/metabolismo , Medio de Cultivo Libre de Suero , Femenino , Hemangioblastos/trasplante , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Antígenos Comunes de Leucocito/metabolismo , Ratones , Células Madre Pluripotentes/trasplante , Transducción de Señal , Nicho de Células Madre
13.
Stem Cell Reports ; 9(3): 779-795, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28803914

RESUMEN

Hematopoietic stem cells arise from mesoderm-derived hemogenic endothelium (HE) during embryogenesis in a process termed endothelial-hematopoietic transition (EHT). To better understand the gene networks that control this process, we investigated the role of the transcription factor HEB (TCF12) by disrupting the TCF12 gene locus in human embryonic stem cells (hESCs) and inducing them to differentiate toward hematopoietic outcomes. HEB-deficient hESCs retained key features of pluripotency, including expression of SOX2 and SSEA-4 and teratoma formation, while NANOG expression was reduced. Differentiation of HEB-/- hESCs toward hematopoietic fates revealed a severe defect in mesodermal development accompanied by decreased expression of regulators of mesoendodermal fate choices. We also identified independent defects in HE formation at the molecular and cellular levels, as well as a failure of T cell development. All defects were largely rescued by re-expression of HEB. Taken together, our results identify HEB as a critical regulator of human mesodermal and hematopoietic specification.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Marcación de Gen , Hematopoyesis , Mesodermo/citología , Antígenos CD/metabolismo , Tipificación del Cuerpo , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Cuerpos Embrioides/citología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Endodermo/citología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Sitios Genéticos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Mieloides/citología , Células Mieloides/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Análisis de Secuencia de ARN , Linfocitos T/citología , Linfocitos T/metabolismo
14.
J Exp Med ; 214(3): 623-637, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28148688

RESUMEN

We studied three patients with severe skeletal dysplasia, T cell immunodeficiency, and developmental delay. Whole-exome sequencing revealed homozygous missense mutations affecting exostosin-like 3 (EXTL3), a glycosyltransferase involved in heparan sulfate (HS) biosynthesis. Patient-derived fibroblasts showed abnormal HS composition and altered fibroblast growth factor 2 signaling, which was rescued by overexpression of wild-type EXTL3 cDNA. Interleukin-2-mediated STAT5 phosphorylation in patients' lymphocytes was markedly reduced. Interbreeding of the extl3-mutant zebrafish (box) with Tg(rag2:green fluorescent protein) transgenic zebrafish revealed defective thymopoiesis, which was rescued by injection of wild-type human EXTL3 RNA. Targeted differentiation of patient-derived induced pluripotent stem cells showed a reduced expansion of lymphohematopoietic progenitor cells and defects of thymic epithelial progenitor cell differentiation. These data identify EXTL3 mutations as a novel cause of severe immune deficiency with skeletal dysplasia and developmental delay and underline a crucial role of HS in thymopoiesis and skeletal and brain development.


Asunto(s)
Enfermedades del Desarrollo Óseo/etiología , Discapacidades del Desarrollo/etiología , Síndromes de Inmunodeficiencia/etiología , Mutación , N-Acetilglucosaminiltransferasas/genética , Animales , Preescolar , Femenino , Heparitina Sulfato/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Lactante , Linfocitos/fisiología , Pez Cebra
15.
Cell Stem Cell ; 19(2): 205-216, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27184401

RESUMEN

The E3 ubiquitin ligase Mule is often overexpressed in human colorectal cancers, but its role in gut tumorigenesis is unknown. Here, we show in vivo that Mule controls murine intestinal stem and progenitor cell proliferation by modulating Wnt signaling via c-Myc. Mule also regulates protein levels of the receptor tyrosine kinase EphB3 by targeting it for proteasomal and lysosomal degradation. In the intestine, EphB/ephrinB interactions position cells along the crypt-villus axis and compartmentalize incipient colorectal tumors. Our study thus unveils an important new avenue by which Mule acts as an intestinal tumor suppressor by regulation of the intestinal stem cell niche.


Asunto(s)
Efrina-B3/metabolismo , Intestinos/citología , Lisosomas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Nicho de Células Madre , Ubiquitina-Proteína Ligasas/metabolismo , Vía de Señalización Wnt , Poliposis Adenomatosa del Colon/patología , Alelos , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Endocitosis , Células HEK293 , Humanos , Ratones Noqueados , Modelos Biológicos , Mutación/genética , Células de Paneth/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Supresoras de Tumor , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia
16.
Cancer Res ; 73(17): 5426-37, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23856248

RESUMEN

The intracellular tyrosine kinase protein tyrosine kinase 6 (PTK6) lacks a membrane-targeting SH4 domain and localizes to the nuclei of normal prostate epithelial cells. However, PTK6 translocates from the nucleus to the cytoplasm in human prostate tumor cells. Here, we show that while PTK6 is located primarily within the cytoplasm, the pool of active PTK6 in prostate cancer cells localizes to membranes. Ectopic expression of membrane-targeted active PTK6 promoted epithelial-mesenchymal transition in part by enhancing activation of AKT, thereby stimulating cancer cell migration and metastases in xenograft models of prostate cancer. Conversely, siRNA-mediated silencing of endogenous PTK6 promoted an epithelial phenotype and impaired tumor xenograft growth. In mice, PTEN deficiency caused endogenous active PTK6 to localize at membranes in association with decreased E-cadherin expression. Active PTK6 was detected at membranes in some high-grade human prostate tumors, and PTK6 and E-cadherin expression levels were inversely correlated in human prostate cancers. In addition, high levels of PTK6 expression predicted poor prognosis in patients with prostate cancer. Our findings reveal novel functions for PTK6 in the pathophysiology of prostate cancer, and they define this kinase as a candidate therapeutic target. Cancer Res; 73(17); 5426-37. ©2013 AACR.


Asunto(s)
Membrana Celular/metabolismo , Transición Epitelial-Mesenquimal , Proteínas de Neoplasias/metabolismo , Fosfohidrolasa PTEN/fisiología , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Citoplasma/metabolismo , Humanos , Inmunoprecipitación , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Noqueados , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/secundario , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
17.
Curr Opin Immunol ; 24(5): 617-24, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22841347

RESUMEN

The advent of reprogramming technology has greatly advanced the field of stem cell biology and nurtured our hope to create patient specific renewable stem cell sources. While the number of reports of disease specific induced pluripotent stem cells is continuously rising, the field becomes increasingly more aware that induced pluripotent stem cells are not as similar to embryonic stem cells as initially assumed. Our state of the art understanding of human induced pluripotent stem cells, their capacity, their limitations and their promise as it pertains to the study and treatment of primary immunodeficiencies, is the content of this review.


Asunto(s)
Diferenciación Celular/inmunología , Síndromes de Inmunodeficiencia/patología , Síndromes de Inmunodeficiencia/terapia , Células Madre Pluripotentes/fisiología , Células Madre Pluripotentes/trasplante , Medicina Regenerativa/métodos , Animales , Trasplante de Células/métodos , Trasplante de Células/tendencias , Modelos Animales de Enfermedad , Humanos , Síndromes de Inmunodeficiencia/fisiopatología , Células Madre Pluripotentes/patología , Medicina Regenerativa/tendencias
18.
PLoS One ; 6(3): e14789, 2011 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-21479203

RESUMEN

Protein tyrosine kinase 6 (PTK6), also called breast tumor kinase (BRK), is expressed in epithelial cells of various tissues including the prostate. Previously it was shown that PTK6 is localized to epithelial cell nuclei in normal prostate, but becomes cytoplasmic in human prostate tumors. PTK6 is also primarily cytoplasmic in the PC3 prostate adenocarcinoma cell line. Sequencing revealed expression of wild type full-length PTK6 transcripts in addition to an alternative transcript lacking exon 2 in PC3 cells. The alternative transcript encodes a 134 amino acid protein, referred to here as ALT-PTK6, which shares the first 77 amino acid residues including the SH3 domain with full length PTK6. RT-PCR was used to show that ALT-PTK6 is coexpressed with full length PTK6 in established human prostate and colon cell lines, as well as in primary cell lines derived from human prostate tissue and tumors. Although interaction between full-length PTK6 and ALT-PTK6 was not detected, ALT-PTK6 associates with the known PTK6 substrates Sam68 and ß-catenin in GST pull-down assays. Coexpression of PTK6 and ALT-PTK6 led to suppression of PTK6 activity and reduced association of PTK6 with tyrosine phosphorylated proteins. While ALT-PTK6 alone did not influence ß-catenin/TCF transcriptional activity in a luciferase reporter assay, it enhanced PTK6-mediated inhibition of ß-catenin/TCF transcription by promoting PTK6 nuclear functions. Ectopic expression of ALT-PTK6 led to reduced expression of the ß-catenin/TCF targets Cyclin D1 and c-Myc in PC3 cells. Expression of tetracycline-inducible ALT-PTK6 blocked the proliferation and colony formation of PC3 cells. Our findings suggest that ALT-PTK6 is able to negatively regulate growth and modulate PTK6 activity, protein-protein associations and/or subcellular localization. Fully understanding functions of ALT-PTK6 and its impact on PTK6 signaling will be critical for development of therapeutic strategies that target PTK6 in cancer.


Asunto(s)
Empalme Alternativo/genética , Proteínas de Neoplasias/genética , Proteínas Tirosina Quinasas/genética , beta Catenina/antagonistas & inhibidores , Línea Celular Tumoral , Núcleo Celular/enzimología , Proliferación Celular , Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Proteínas de Neoplasias/metabolismo , Fosforilación , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Transcripción Genética , Ensayo de Tumor de Célula Madre , beta Catenina/metabolismo
19.
Cell Cycle ; 9(20): 4190-9, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20953141

RESUMEN

Protein tyrosine kinase 6 (PTK6) is an intracellular tyrosine kinase that is nuclear in epithelial cells of the normal prostate, but cytoplasmic in prostate tumors and in the PC3 prostate tumor cell line. The impact of altered PTK6 intracellular localization in prostate tumor cells has not been extensively explored. Knockdown of endogenous cytoplasmic PTK6 resulted in decreased PC3 cell proliferation and colony formation, suggesting that cytoplasmic PTK6 stimulates oncogenic pathways. In contrast, reintroduction of PTK6 into nuclei of PC3 cells had a negative effect on growth. Enhanced tyrosine phosphorylation of the PTK6 substrate Sam68 was detected in cells expressing nuclear-targeted PTK6. We found that mechanisms regulating nuclear localization of PTK6 are intact in PC3 cells. Transiently overexpressed PTK6 readily enters the nucleus. Ectopic expression of ALT-PTK6, a catalytically inactive splice variant of PTK6, did not affect localization of endogenous PTK6 in PC3 cells. Using leptomycin B, we confirmed that cytoplasmic localization of endogenous PTK6 is not due to Crm-1/exportin-1 mediated nuclear export. In addition, overexpression of the PTK6 nuclear substrate Sam68 is not sufficient to bring PTK6 into the nucleus. While exogenous PTK6 was readily detected in the nucleus when transiently expressed at high levels, low-level expression of inducible wild type PTK6 in stable cell lines resulted in its cytoplasmic retention. Our results suggest that retention of PTK6 in the cytoplasm of prostate cancer cells disrupts its ability to regulate nuclear substrates and leads to aberrant growth. In prostate cancer, restoring PTK6 nuclear localization may have therapeutic advantages.


Asunto(s)
Citoplasma/enzimología , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Carioferinas/metabolismo , Masculino , Proteínas de Neoplasias/genética , Señales de Localización Nuclear , Proteínas Tirosina Quinasas/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología , Proteína Exportina 1
20.
Cell Cycle ; 9(1): 86-9, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20016279

RESUMEN

ARF is a vital tumor suppressor and its loss contributes significantly to cancer. The frequency in which ARF is mutated, deleted or silenced is second to the loss of p53. The most documented and widely accepted activity of ARF is mediated through its activation of the p53 transcriptional program by inhibiting MDM2 function. However, several lines of evidence have surfaced demonstrating that ARF possesses p53-independent functions. One of these p53-independent functions is ARF's regulation of the E2F family. The E2F/DP transcription factor is critical for cell cycle progression. The balance between activator and repressor E2Fs regulates the expression of E2F target genes and thus cell proliferation as well as other cellular functions such as checkpoint, chromosome assembly and repair. Through its ability to bind directly to DP1, ARF can cause dissociation of both activator and repressor E2Fs. While the regulation of the activator E2Fs is related to cell cycle arrest, there is evidence that the regulation of the repressors, E2F4 and E2F5, is significant in maintaining genomic stability.


Asunto(s)
Neoplasias/metabolismo , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Factores de Transcripción E2F/metabolismo , Humanos , Modelos Biológicos , Neoplasias/patología , Unión Proteica/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA