Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
medRxiv ; 2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37986997

RESUMEN

PURPOSE: Gamma delta T-cell receptor-positive acute lymphoblastic leukemia (γδ T-ALL) is a high-risk but poorly characterized disease. METHODS: We studied clinical features of 200 pediatric γδ T-ALL, and compared the prognosis of 93 cases to 1,067 protocol-matched non-γδ T-ALL. Genomic features were defined by transcriptome and genome sequencing. Experimental modeling was used to examine the mechanistic impacts of genomic alterations. Therapeutic vulnerabilities were identified by high throughput drug screening of cell lines and xenografts. RESULTS: γδ T-ALL in children under three was extremely high-risk with 5-year event-free survival (33% v. 70% [age 3-<10] and 73% [age ≥10], P =9.5 x 10 -5 ) and 5-year overall survival (49% v. 78% [age 3-<10] and 81% [age ≥10], P =0.002), differences not observed in non-γδ T-ALL. γδ T-ALL in this age group was enriched for genomic alterations activating LMO2 activation and inactivating STAG2 inactivation ( STAG2/LMO2 ). Mechanistically, we show that inactivation of STAG2 profoundly perturbs chromatin organization by altering enhancer-promoter looping resulting in deregulation of gene expression associated with T-cell differentiation. Drug screening showed resistance to prednisolone, consistent with clinical slow treatment response, but identified a vulnerability in DNA repair pathways arising from STAG2 inactivation, which was efficaciously targeted by Poly(ADP-ribose) polymerase (PARP) inhibition, with synergism with HDAC inhibitors. Ex-vivo drug screening on PDX cells validated the efficacy of PARP inhibitors as well as other potential targets including nelarabine. CONCLUSION: γδ T-ALL in children under the age of three is extremely high-risk and enriched for STAG2/LMO2 ALL. STAG2 loss perturbs chromatin conformation and differentiation, and STAG2/LMO2 ALL is sensitive to PARP inhibition. These data provide a diagnostic and therapeutic framework for pediatric γδ T-ALL. SUPPORT: The authors are supported by the American and Lebanese Syrian Associated Charities of St Jude Children's Research Hospital, NCI grants R35 CA197695, P50 CA021765 (C.G.M.), the Henry Schueler 41&9 Foundation (C.G.M.), and a St. Baldrick's Foundation Robert J. Arceci Innovation Award (C.G.M.), Gabriella Miller Kids First X01HD100702 (D.T.T and C.G.M.) and R03CA256550 (D.T.T. and C.G.M.), F32 5F32CA254140 (L.M.), and a Garwood Postdoctoral Fellowship of the Hematological Malignancies Program of the St Jude Children's Research Hospital Comprehensive Cancer Center (S.K.). This project was supported by the National Cancer Institute of the National Institutes of Health under the following award numbers: U10CA180820, UG1CA189859, U24CA114766, U10CA180899, U10CA180866 and U24CA196173. DISCLAIMER: The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The funding agencies were not directly involved in the design of the study, gathering, analysis and interpretation of the data, writing of the manuscript, or decision to submit the manuscript for publication.

2.
Cell Rep ; 42(7): 112804, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37453060

RESUMEN

The bone marrow microenvironment (BME) drives drug resistance in acute lymphoblastic leukemia (ALL) through leukemic cell interactions with bone marrow (BM) niches, but the underlying mechanisms remain unclear. Here, we show that the interaction between ALL and mesenchymal stem cells (MSCs) through integrin ß1 induces an epithelial-mesenchymal transition (EMT)-like program in MSC-adherent ALL cells, resulting in drug resistance and enhanced survival. Moreover, single-cell RNA sequencing analysis of ALL-MSC co-culture identifies a hybrid cluster of MSC-adherent ALL cells expressing both B-ALL and MSC signature genes, orchestrated by a WNT/ß-catenin-mediated EMT-like program. Blockade of interaction between ß-catenin and CREB binding protein impairs the survival and drug resistance of MSC-adherent ALL cells in vitro and results in a reduction in leukemic burden in vivo. Targeting of this WNT/ß-catenin-mediated EMT-like program is a potential therapeutic approach to overcome cell extrinsically acquired drug resistance in ALL.


Asunto(s)
Transición Epitelial-Mesenquimal , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , beta Catenina , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Técnicas de Cocultivo , Resistencia a Medicamentos , Proliferación Celular , Microambiente Tumoral
3.
JCI Insight ; 7(23)2022 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-36477361

RESUMEN

NK cell deficiencies (NKD) are a type of primary immune deficiency in which the major immunologic abnormality affects NK cell number, maturity, or function. Since NK cells contribute to immune defense against virally infected cells, patients with NKD experience higher susceptibility to chronic, recurrent, and fatal viral infections. An individual with recurrent viral infections and mild hypogammaglobulinemia was identified to have an X-linked damaging variant in the transcription factor gene ELF4. The variant does not decrease expression but disrupts ELF4 protein interactions and DNA binding, reducing transcriptional activation of target genes and selectively impairing ELF4 function. Corroborating previous murine models of ELF4 deficiency (Elf4-/-) and using a knockdown human NK cell line, we determined that ELF4 is necessary for normal NK cell development, terminal maturation, and function. Through characterization of the NK cells of the proband, expression of the proband's variant in Elf4-/- mouse hematopoietic precursor cells, and a human in vitro NK cell maturation model, we established this ELF4 variant as a potentially novel cause of NKD.


Asunto(s)
Factores de Transcripción , Animales , Humanos , Ratones , Proteínas de Unión al ADN/genética , Células Asesinas Naturales , Factores de Transcripción/genética
4.
Exp Mol Med ; 52(10): 1663-1672, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33067577

RESUMEN

Chronic myeloid leukemia is a hematological cancer driven by the oncoprotein BCR-ABL1, and lifelong treatment with tyrosine kinase inhibitors extends patient survival to nearly the life expectancy of the general population. Despite advances in the development of more potent tyrosine kinase inhibitors to induce a durable deep molecular response, more than half of patients relapse upon treatment discontinuation. This clinical finding supports the paradigm that leukemia stem cells feed the neoplasm, resist tyrosine kinase inhibition, and reactivate upon drug withdrawal depending on the fitness of the patient's immune surveillance. This concept lends support to the idea that treatment-free remission is not achieved solely with tyrosine kinase inhibitors and that new molecular targets independent of BCR-ABL1 signaling are needed in order to develop adjuvant therapy to more efficiently eradicate the leukemia stem cell population responsible for chemoresistance and relapse. Future efforts must focus on the identification of new targets to support the discovery of potent and safe small molecules able to specifically eradicate the leukemic stem cell population. In this review, we briefly discuss molecular maintenance in leukemia stem cells in chronic myeloid leukemia and provide a more in-depth discussion of the dual-specificity kinase DYRK2, which has been identified as a novel actionable checkpoint in a critical leukemic network. DYRK2 controls the activation of p53 and proteasomal degradation of c-MYC, leading to impaired survival and self-renewal of leukemia stem cells; thus, pharmacological activation of DYRK2 as an adjuvant to standard therapy has the potential to induce treatment-free remission.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Animales , Proteínas Portadoras/metabolismo , Autorrenovación de las Células/genética , Susceptibilidad a Enfermedades , Metabolismo Energético , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Quinasas DyrK
5.
Blood ; 134(22): 1960-1972, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31515251

RESUMEN

Leukemia stem cells are a rare population with a primitive progenitor phenotype that can initiate, sustain, and recapitulate leukemia through a poorly understood mechanism of self-renewal. Here, we report that Krüppel-like factor 4 (KLF4) promotes disease progression in a murine model of chronic myeloid leukemia (CML)-like myeloproliferative neoplasia by repressing an inhibitory mechanism of preservation in leukemia stem/progenitor cells with leukemia-initiating capacity. Deletion of the Klf4 gene severely abrogated the maintenance of BCR-ABL1(p210)-induced CML by impairing survival and self-renewal in BCR-ABL1+ CD150+ lineage-negative Sca-1+ c-Kit+ leukemic cells. Mechanistically, KLF4 repressed the Dyrk2 gene in leukemic stem/progenitor cells; thus, loss of KLF4 resulted in elevated levels of dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 2 (DYRK2), which were associated with inhibition of survival and self-renewal via depletion of c-Myc protein and p53 activation. In addition to transcriptional regulation, stabilization of DYRK2 protein by inhibiting ubiquitin E3 ligase SIAH2 with vitamin K3 promoted apoptosis and abrogated self-renewal in murine and human CML stem/progenitor cells. Altogether, our results suggest that DYRK2 is a molecular checkpoint controlling p53- and c-Myc-mediated regulation of survival and self-renewal in CML cells with leukemic-initiating capacity that can be targeted with small molecules.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Eliminación de Gen , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Noqueados , Células Madre Neoplásicas/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Vitamina K 3/farmacología , Quinasas DyrK
6.
Methods Mol Biol ; 1686: 173-182, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29030821

RESUMEN

Hematopoietic stem cells (HSCs) represent an important target cell population in bone marrow transplantation, cell and gene therapy applications, and the development of leukemia models for research. Because the hematopoietic progeny carries the genetic information of HSCs and replenishes the blood and immune system, corrective gene transfer into HSCs provides an ideal therapeutic approach for many monogenic hematological diseases and a useful tool for studies of HSC function and blood formation in normal and malignant hematopoiesis. However, the efficiency of gene transfer into HSCs has been limited by several features of viral vectors, viral titer, methods of viral transduction, and the property of stem cell quiescence. In this chapter, we describe the production of retrovirus using murine stem cell virus (MSCV)-based retroviral vectors and purification and transduction of murine HSCs.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Fase de Descanso del Ciclo Celular , Retroviridae/genética , Transducción Genética , Animales , Antígenos CD34/metabolismo , Células Cultivadas , Citometría de Flujo , Vectores Genéticos , Ratones
7.
Sci Rep ; 7: 41389, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-28128342

RESUMEN

Zika virus (ZIKV) is an emerging mosquito-borne (Aedes genus) arbovirus of the Flaviviridae family. Although ZIKV has been predominately associated with a mild or asymptomatic dengue-like disease, its appearance in the Americas has been accompanied by a multi-fold increase in reported incidence of fetal microcephaly and brain malformations. The source and mode of vertical transmission from mother to fetus is presumptively transplacental, although a causal link explaining the interval delay between maternal symptoms and observed fetal malformations following infection has been missing. In this study, we show that primary human placental trophoblasts from non-exposed donors (n = 20) can be infected by primary passage ZIKV-FLR isolate, and uniquely allowed for ZIKV viral RNA replication when compared to dengue virus (DENV). Consistent with their being permissive for ZIKV infection, primary trophoblasts expressed multiple putative ZIKV cell entry receptors, and cellular function and differentiation were preserved. These findings suggest that ZIKV-FLR strain can replicate in human placental trophoblasts without host cell destruction, thereby serving as a likely permissive reservoir and portal of fetal transmission with risk of latent microcephaly and malformations.


Asunto(s)
Placenta/patología , Trofoblastos/virología , Replicación Viral/fisiología , Virus Zika/fisiología , Adulto , Células Cultivadas , Dengue/patología , Dengue/virología , Virus del Dengue/fisiología , Femenino , Células Gigantes/metabolismo , Células Gigantes/patología , Humanos , Ligandos , MicroARNs/genética , MicroARNs/metabolismo , Filogenia , Embarazo , ARN Viral/metabolismo , Receptores Virales/metabolismo , Receptores Toll-Like/metabolismo , Infección por el Virus Zika/patología , Infección por el Virus Zika/virología
8.
J Leukoc Biol ; 99(5): 673-85, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26908828

RESUMEN

Krüppel-like factor 4 is a zinc finger protein with dual functions that can act as a transcriptional activator and repressor of genes involved in cell proliferation, differentiation, and apoptosis. Although most studies have focused on terminally differentiated epithelial cells, evidence suggests that Krüppel-like factor 4 regulates the development and function of the myeloid and lymphoid blood lineages. The ability of Krüppel-like factor 4 to dedifferentiate from somatic cells into pluripotent stem cells in cooperation with other reprogramming factors suggests its potential function in the preservation of tissue-specific stem cells. Additionally, emerging interest in the redifferentiation of induced pluripotent stem cells into blood cells to correct hematologic deficiencies and malignancies warrants further studies on the role of Krüppel-like factor 4 in steady-state blood formation.


Asunto(s)
Reprogramación Celular , Hematopoyesis , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Reprogramación Celular/genética , Cromatina/metabolismo , Regulación de la Expresión Génica , Hematopoyesis/genética , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Modelos Biológicos
9.
Immunol Cell Biol ; 93(7): 605-15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25666096

RESUMEN

Since its discovery, diverse functions have been attributed to the G0/G1 switch gene 2 (G0S2), from lipid metabolism to control of cell proliferation. Our group showed for the first time that G0S2 promotes quiescence in hematopoietic stem cells by interacting with and retaining nucleolin around the nucleus. Herein, we report the role of G0S2 in the differentiation and function of CD8(+) T cells examined in mice with an embryonic deletion of the G0s2 gene. G0S2 expression in naïve CD8(+) T cells decreased immediately after T-cell receptor activation downstream of the mitogen-activated protein kinase, calcium/calmodulin, phosphatidylinositol 3'-kinase and mammalian target of rapamycin pathways. Surprisingly, G0S2-null naïve CD8(+) T cells displayed increased basal and spare respiratory capacity that was not associated with increased mitochondrial biogenesis but with increased phosphorylation of AMP-activated protein kinase α. Naïve CD8(+) T cells showed increased proliferation in response to in vitro activation and in vivo lymphopenia; however, naïve CD8(+) T cells expressing the OT-1 transgene exhibited normal differentiation of naïve cells to effector and memory CD8(+) T cells upon infection with Listeria monocytogenes in a wild-type or a G0s2-null environment, with increased circulating levels of free fatty acids. Collectively, our results suggest that G0S2 inhibits energy production by oxidative phosphorylation to fine-tune proliferation in homeostatic conditions.


Asunto(s)
Linfocitos T CD8-positivos/citología , Proteínas de Ciclo Celular/fisiología , Mitocondrias/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , División Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ácidos Grasos no Esterificados/sangre , Femenino , Genes Reporteros , Homeostasis , Listeria monocytogenes , Listeriosis/inmunología , Listeriosis/metabolismo , Activación de Linfocitos , Linfopenia/inmunología , Linfopenia/metabolismo , Linfopoyesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación Oxidativa , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal , Organismos Libres de Patógenos Específicos
10.
Leuk Res ; 38(2): 210-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24183236

RESUMEN

G0/G1 switch gene 2 (G0S2) is a basic protein with ill-defined function that inhibits the proliferation of hematopoietic stem cells. Herein, we show that treatment of K562 cells with 5-azacytidine (5-Aza) resulted in a 24-fold increase in G0S2 expression and a reduction in cell growth. Conversely, gene demethylation in the presence of G0S2-specific shRNA restored proliferation, further supporting an inhibitory role for G0S2 in cell proliferation. Elevated levels of G0S2 inhibited the division of K562 cells by sequestering the nucleolar phosphoprotein nucleolin in the cytosol. G0S2 inhibited the proliferation of leukemia cells in vivo in xenograft models. Collectively, our data identify a new mechanism that controls proliferation in K562 cells, suggesting a possible tumor suppressor function in leukemia cells.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proliferación Celular , Citosol/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Genes Supresores de Tumor/fisiología , Células HL-60 , Humanos , Células K562 , Ratones , Ratones Desnudos , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Nucleolina
11.
Immunol Lett ; 156(1-2): 94-101, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24075846

RESUMEN

The transcription factor Krüppel-like factor 4 (KLF4) can activate or repress gene expression in a cell-context dependent manner. We have previously shown that KLF4 inhibits the proliferation of naïve CD8(+) T cells in vitro downstream of the transcription factor ELF4. In this work, we describe a novel role of KLF4 in the differentiation of CD8(+) T cells upon infection. Loss of KLF4 had minimal effect on thymic T cell development and distribution of mature T cells in the spleen, blood, and lymph nodes. KLF4-deficient naïve CD8(+) T cells also displayed normal homeostatic proliferation upon adoptive transfer into lymphopenic hosts. However, activation of KLF4-deficient naïve CD8(+) T cells by in vitro TCR crosslink and co-stimulation resulted in increased proliferation. Furthermore, naïve KLF4-deficient OT-I CD8(+) T cells generated increased numbers of functional memory CD8(+) T cells compared to wild type OT-I CD8(+) T cells co-injected in the same recipient in both primary and recall responses to Listeria monocytogenes-OVA. Collectively, our data demonstrate that KLF4 regulates differentiation of functional memory CD8(+) T cells while sparing development and homeostasis of naïve CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Factores de Transcripción de Tipo Kruppel/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Citometría de Flujo , Homeostasis/inmunología , Memoria Inmunológica/inmunología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/inmunología , Listeriosis/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología
12.
Cell Cycle ; 12(15): 2376-83, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23839041

RESUMEN

Cellular quiescence is a reversible cell cycle arrest that is poised to re-enter the cell cycle in response to a combination of cell-intrinsic factors and environmental cues. In hematopoietic stem cells, a coordinated balance between quiescence and differentiating proliferation ensures longevity and prevents both genetic damage and stem cell exhaustion. However, little is known about how all these processes are integrated at the molecular level. We will briefly review the environmental and intrinsic control of stem cell quiescence and discuss a new model that involves a protein-to-protein interaction between G0S2 and the phospho-nucleoprotein nucleolin in the cytosol.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/fisiología , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/fisiología , Proliferación Celular , Humanos , Datos de Secuencia Molecular , Fosfoproteínas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/metabolismo , Nucleolina
13.
PLoS One ; 7(12): e51251, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251470

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common hematological cancer in children. Although risk-adaptive therapy, CNS-directed chemotherapy, and supportive care have improved the survival of ALL patients, disease relapse is still the leading cause of cancer-related death in children. Therefore, new drugs are needed as frontline treatments in high-risk disease and as salvage agents in relapsed ALL. In this study, we report that purified sulforaphane, a natural isothiocyanate found in cruciferous vegetables, has anti-leukemic properties in a broad range of ALL cell lines and primary lymphoblasts from pediatric T-ALL and pre-B ALL patients. The treatment of ALL leukemic cells with sulforaphane resulted in dose-dependent apoptosis and G2/M cell cycle arrest, which was associated with the activation of caspases (3, 8, and 9), inactivation of PARP, p53-independent upregulation of p21(CIP1/WAF1), and inhibition of the Cdc2/Cyclin B1 complex. Interestingly, sulforaphane also inhibited the AKT and mTOR survival pathways in most of the tested cell lines by lowering the levels of both total and phosphorylated proteins. Finally, the administration of sulforaphane to the ALL xenograft models resulted in a reduction of tumor burden, particularly following oral administration, suggesting a potential role as an adjunctive agent to improve the therapeutic response in high-risk ALL patients with activated AKT signaling.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Tiocianatos/farmacología , Línea Celular Tumoral , Humanos , Isotiocianatos , Sulfóxidos
14.
PLoS One ; 7(5): e38280, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22693613

RESUMEN

Bone marrow hematopoietic stem cells (HSCs) balance proliferation and differentiation by integrating complex transcriptional and post-translational mechanisms regulated by cell intrinsic and extrinsic factors. We found that transcripts of G(0)/G(1) switch gene 2 (G0S2) are enriched in lineage(-) Sca-1(+) c-kit(+) (LSK) CD150(+) CD48(-) CD41(-) cells, a population highly enriched for quiescent HSCs, whereas G0S2 expression is suppressed in dividing LSK CD150(+) CD48(-) cells. Gain-of-function analyses using retroviral expression vectors in bone marrow cells showed that G0S2 localizes to the mitochondria, endoplasmic reticulum, and early endosomes in hematopoietic cells. Co-transplantation of bone marrow cells transduced with the control or G0S2 retrovirus led to increased chimerism of G0S2-overexpressing cells in femurs, although their contribution to the blood was reduced. This finding was correlated with increased quiescence in G0S2-overexpressing HSCs (LSK CD150(+) CD48(-)) and progenitor cells (LS(-)K). Conversely, silencing of endogenous G0S2 expression in bone marrow cells increased blood chimerism upon transplantation and promoted HSC cell division, supporting an inhibitory role for G0S2 in HSC proliferation. A proteomic study revealed that the hydrophobic domain of G0S2 interacts with a domain of nucleolin that is rich in arginine-glycine-glycine repeats, which results in the retention of nucleolin in the cytosol. We showed that this cytosolic retention of nucleolin occurs in resting, but not proliferating, wild-type LSK CD150(+) CD48(-) cells. Collectively, we propose a novel model of HSC quiescence in which elevated G0S2 expression can sequester nucleolin in the cytosol, precluding its pro-proliferation functions in the nucleolus.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Citosol/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Células Sanguíneas/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Células 3T3 NIH , Fosfoproteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Nucleolina
15.
J Leukoc Biol ; 91(5): 739-50, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22345706

RESUMEN

The development and survival of NK cells rely on a complex, spatiotemporal gene expression pattern regulated by specific transcription factors in NK cells and tissue-specific microenvironments supported by hematopoietic cells. Here, we show that somatic deletion of the KLF4 gene, using inducible and lineage-specific cre-transgenic mice, leads to a significant reduction of NK cells (NK1.1(+) TCR-ß(-)) in the blood and spleen but not in the BM, liver, or LNs. Functional and immunophenotypic analyses revealed increased apoptosis of CD27(+/-) CD11b(+) NK cells in the spleen of KLF4-deficient mice, although remaining NK cells were able to lyse tumor target cells and produce IFN-γ. A normal recovery of adoptively transferred KLF4-deficient NK cells in WT hosts suggested that the survival defect was not intrinsic of NK cells. However, BM chimeras using KLF4-deficient mice as donors indicated that reduced survival of NK cells depended on BM-derived hematopoietic cells in the spleen. The number of CD11c(hi) DCs, which are known to support NK cell survival, was reduced significantly in the spleen of KLF4-deficient mice, likely a result of a lower number of precDC progenitor cells in this tissue. Taken together, our data suggest that the pluripotency-associated gene KLF4 is required for the maintenance of DCs in the spleen and consequently, survival of differentiated NK cells in this tissue.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Factores de Transcripción de Tipo Kruppel/fisiología , Bazo/citología , Animales , Apoptosis , Western Blotting , Trasplante de Médula Ósea , Diferenciación Celular , Células Dendríticas/inmunología , Femenino , Citometría de Flujo , Inmunofenotipificación , Integrasas/metabolismo , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Factor 4 Similar a Kruppel , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Bazo/inmunología , Bazo/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 31(5): 1185-91, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21350194

RESUMEN

OBJECTIVE: The regeneration of the hematopoietic system in bone marrow after chemotherapy depends on a balance between the quiescence and proliferation of lineage-specific progenitor cells. Even though the vascular network in bone is damaged by cytoablation, the transcriptional control of quiescence in endothelial cells is not well known. In this study, we investigated the role of the transcription factor E74-like factor (ELF4) in the proliferation of endothelial cells in bone marrow. METHODS AND RESULTS: Loss-of-function models were used to study the role of ELF4 in human and murine endothelial cells. ELF4 promotes cell cycle entry by activating cyclin-dependent kinase-4 in human umbilical vein endothelial cells. Elf4-null mice exhibited enhanced recovery of bone marrow CD45- CD31+ endothelial cells and sinusoidal blood vessels following administration of 5-fluorouracil. CONCLUSIONS: Loss of ELF4 leads to increased quiescence in bone marrow endothelial cells by the deregulation of cyclin-dependent kinase-4 expression and to enhanced regeneration of sinusoidal blood vessels.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Resistencia a Medicamentos , Células Endoteliales/efectos de los fármacos , Fluorouracilo/farmacología , Agonistas Mieloablativos/farmacología , Factores de Transcripción/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Células COS , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Neovascularización Fisiológica/efectos de los fármacos , Regiones Promotoras Genéticas , Interferencia de ARN , Factores de Tiempo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transfección
17.
Nat Immunol ; 10(6): 618-26, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19412182

RESUMEN

Transcription factors that regulate the quiescence, proliferation and homing of lymphocytes are critical for effective immune system function. Here we demonstrate that the transcription factor ELF4 directly activated the tumor suppressor KLF4 'downstream' of T cell antigen receptor signaling to induce cell cycle arrest in naive CD8(+) T cells. Elf4- and Klf4-deficient mice accumulated CD8(+)CD44(hi) T cells during steady-state conditions and generated more memory T cells after immunization. The homeostatic population expansion of CD8(+)CD44(hi) T cells in Elf4-null mice resulted in a redistribution of cells to nonlymphoid tissue because of lower expression of the transcription factor KLF2 and the surface proteins CCR7 and CD62L. Our work describes the combinatorial effect of lymphocyte-intrinsic factors on the homeostasis, activation and homing of T cells.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción/metabolismo , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/citología , Ciclo Celular , Movimiento Celular , Regulación de la Expresión Génica , Memoria Inmunológica , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
18.
Cell Biochem Funct ; 26(1): 24-32, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17154359

RESUMEN

Embryonic stem (ES) cells have the potential to develop into various cell lineages including hemangioblasts (Flk1+), a common progenitor for hematopoietic and vascular endothelial cells. Previous studies indicate that Flk1+ cells, a marker for hemangioblast, can be derived from ES cell and that Flk1+ can be differentiated into hematopoietic or endothelial cells depending on culture conditions. We developed an improved in vitro system to generate Flk1+-enriched cultures from mouse ES cells and used this in vitro system to study the role of Wnt signalling in early endothelial progenitor cells. We determined the expression of the Wnt and Frizzled genes in Flk1+ cells derived from mouse ES cells. RT-PCR analyses identified significantly higher expression of non-canonical Wnt5a and Wnt11 genes in Flk1+ cells compared to Flk1- cells. In contrast, expression of canonical Wnt3a gene was reduced in Flk1+ cells. In addition, Frizzled2, Frizzled5 and Frizzled7 genes were also expressed at a higher level in Flk1+ cells. The differential expression of Wnt and Frizzled genes in Flk1+ cells provides a novel insight into the role of non-canonical Wnt signalling in vascular endothelial fate determination.


Asunto(s)
Células Madre Embrionarias/metabolismo , Receptores Frizzled/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Receptores Acoplados a Proteínas G/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Proteínas Wnt/genética , Secuencia de Aminoácidos , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Células Madre Embrionarias/citología , Receptores Frizzled/biosíntesis , Receptores Frizzled/fisiología , Ratones , Datos de Secuencia Molecular , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/genética , Proteínas Wnt/biosíntesis , Proteínas Wnt/fisiología , Proteína Wnt-5a
19.
IUBMB Life ; 59(2): 90-8, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17454300

RESUMEN

This study examined whether cell cycle regulatory proteins, such as cyclin-dependent kinases (CDKs), cyclins, and CDK inhibitors, regulate type II collagen expression and mediate interlukin-1 (IL-1beta)-induced suppression of type II collagen expression in articular chondrocytes. IL-1beta inhibited type II collagen expression, but activated CDK6. Ectopic expression of CDK2 did not alter type II collagen expression. However, overexpression of CDK6 inhibited type II collagen expression, whereas inhibition of CDK6 activity blocked IL-1beta-induced suppression of type II collagen expression. IL-1beta upregulated the expression of cyclin D1, which is known to activate CDK6. In turn, overexpression of cyclin D1 suppressed type II collagen expression. In contrast to cyclin D1, IL-1beta triggered down-regulation of the CDK inhibitor, p21. Overexpression of p21 blocked IL-1beta- or CDK6-induced suppression of type II collagen expression. Our results collectively indicate that CDK6/cyclin D1/p21 complex regulates type II collagen expression in articular chondrocytes.


Asunto(s)
Condrocitos/metabolismo , Colágeno Tipo II/biosíntesis , Ciclina D1/fisiología , Quinasa 6 Dependiente de la Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Animales , Western Blotting , Condrocitos/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/fisiología , Regulación de la Expresión Génica , Interleucina-1beta/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/biosíntesis , Conejos
20.
Eur J Cell Biol ; 86(2): 111-23, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17208333

RESUMEN

Beta-catenin is implicated in quite different cellular processes, which require a fine-tuned regulation of its function. Here we demonstrate that cyclin-dependent kinase 6 (CDK6), in association with cyclin D1 (CCND1), directly binds to beta-catenin. We showed that CCND1-CDK6 phosphorylates beta-catenin on serine 45 (S45). This phosphorylation creates a priming site for glycogen synthase kinase 3beta (GSK3beta) and is both necessary and sufficient to initiate the beta-catenin phosphorylation-degradation cascade. Moreover, co-immunoprecipitation assays using Wnt3a-conditioned medium reveals that while Wnt stimulation leads to the dissociation of beta-catenin from axin and casein kinase Ialpha (CKIalpha), Wnt treatment promotes an increase in CCND1 level and the association of beta-catenin with CCND1-CDK6. Furthermore, Wnt3a-stimulated cytosolic beta-catenin levels were higher in CDK6 knockout mouse embryonic fibroblasts (CDK6-/- MEFs) compared to wild-type MEFs. Thus, the CCND1-CDK6 complex is like to negatively regulate Wnt signaling by mediating beta-catenin phosphorylation and its subsequent degradation in Wnt-stimulated cells.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/metabolismo , Proteínas Quinasas/metabolismo , Factores de Transcripción TCF/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Bovinos , Células Cultivadas , Ciclina D1/metabolismo , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Noqueados , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica/genética , Proteínas Quinasas/genética , Serina/metabolismo , Transactivadores/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...