Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 295(21): 7492-7500, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32299910

RESUMEN

Severe congenital neutropenia (SCN) is characterized by a near absence of neutrophils, rendering individuals with this disorder vulnerable to recurrent life-threatening infections. The majority of SCN cases arise because of germline mutations in the gene elastase, neutrophil-expressed (ELANE) encoding the neutrophil granule serine protease neutrophil elastase. Treatment with a high dose of granulocyte colony-stimulating factor increases neutrophil production and reduces infection risk. How ELANE mutations produce SCN remains unknown. The currently proposed mechanism is that ELANE mutations promote protein misfolding, resulting in endoplasmic reticulum stress and activation of the unfolded protein response (UPR), triggering death of neutrophil precursors and resulting in neutropenia. Here we studied the ELANE mutation p.G185R, often associated with greater clinical severity (e.g. decreased responsiveness to granulocyte colony-stimulating factor and increased leukemogenesis). Using an inducible expression system, we observed that this ELANE mutation diminishes enzymatic activity and granulocytic differentiation without significantly affecting cell proliferation, cell death, or UPR induction in murine myeloblast 32D and human promyelocytic NB4 cells. Impaired differentiation was associated with decreased expression of genes encoding critical hematopoietic transcription factors (Gfi1, Cebpd, Cebpe, and Spi1), cell surface proteins (Csf3r and Gr1), and neutrophil granule proteins (Mpo and Elane). Together, these findings challenge the currently prevailing model that SCN results from mutant ELANE, which triggers endoplasmic reticulum stress, UPR, and apoptosis.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea , Regulación Enzimológica de la Expresión Génica , Granulocitos/enzimología , Elastasa de Leucocito , Mutación Missense , Neutropenia/congénito , Respuesta de Proteína Desplegada , Sustitución de Aminoácidos , Animales , Apoptosis , Línea Celular Tumoral , Síndromes Congénitos de Insuficiencia de la Médula Ósea/enzimología , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Estrés del Retículo Endoplásmico , Humanos , Elastasa de Leucocito/biosíntesis , Elastasa de Leucocito/genética , Ratones , Neutropenia/enzimología , Neutropenia/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
PLoS Genet ; 14(9): e1007642, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30216339

RESUMEN

PAX5, one of nine members of the mammalian paired box (PAX) family of transcription factors, plays an important role in B cell development. Approximately one-third of individuals with pre-B acute lymphoblastic leukemia (ALL) acquire heterozygous inactivating mutations of PAX5 in malignant cells, and heterozygous germline loss-of-function PAX5 mutations cause autosomal dominant predisposition to ALL. At least in mice, Pax5 is required for pre-B cell maturation, and leukemic remission occurs when Pax5 expression is restored in a Pax5-deficient mouse model of ALL. Together, these observations indicate that PAX5 deficiency reversibly drives leukemogenesis. PAX5 and its two most closely related paralogs, PAX2 and PAX8, which are not mutated in ALL, exhibit overlapping expression and function redundantly during embryonic development. However, PAX5 alone is expressed in lymphocytes, while PAX2 and PAX8 are predominantly specific to kidney and thyroid, respectively. We show that forced expression of PAX2 or PAX8 complements PAX5 loss-of-function mutation in ALL cells as determined by modulation of PAX5 target genes, restoration of immunophenotypic and morphological differentiation, and, ultimately, reduction of replicative potential. Activation of PAX5 paralogs, PAX2 or PAX8, ordinarily silenced in lymphocytes, may therefore represent a novel approach for treating PAX5-deficient ALL. In pursuit of this strategy, we took advantage of the fact that, in kidney, PAX2 is upregulated by extracellular hyperosmolarity. We found that hyperosmolarity, at potentially clinically achievable levels, transcriptionally activates endogenous PAX2 in ALL cells via a mechanism dependent on NFAT5, a transcription factor coordinating response to hyperosmolarity. We also found that hyperosmolarity upregulates residual wild type PAX5 expression in ALL cells and modulates gene expression, including in PAX5-mutant primary ALL cells. These findings specifically demonstrate that osmosensing pathways may represent a new therapeutic target for ALL and more broadly point toward the possibility of using gene paralogs to rescue mutations driving cancer and other diseases.


Asunto(s)
Riñón/metabolismo , Osmorregulación , Factor de Transcripción PAX2/metabolismo , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Animales , Linfocitos B/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Células HEK293 , Humanos , Soluciones Hipertónicas/farmacología , Riñón/efectos de los fármacos , Masculino , Ratones , Mutación , Osmorregulación/efectos de los fármacos , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX5/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Blood ; 129(15): 2103-2110, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28179280

RESUMEN

GATA family proteins play essential roles in development of many cell types, including hematopoietic, cardiac, and endodermal lineages. The first three factors, GATAs 1, 2, and 3, are essential for normal hematopoiesis, and their mutations are responsible for a variety of blood disorders. Acquired and inherited GATA1 mutations contribute to Diamond-Blackfan anemia, acute megakaryoblastic leukemia, transient myeloproliferative disorder, and a group of related congenital dyserythropoietic anemias with thrombocytopenia. Conversely, germ line mutations in GATA2 are associated with GATA2 deficiency syndrome, whereas acquired mutations are seen in myelodysplastic syndrome, acute myeloid leukemia, and in blast crisis transformation of chronic myeloid leukemia. The fact that mutations in these genes are commonly seen in blood disorders underscores their critical roles and highlights the need to develop targeted therapies for transcription factors. This review focuses on hematopoietic disorders that are associated with mutations in two prominent GATA family members, GATA1 and GATA2.


Asunto(s)
Factor de Transcripción GATA1 , Factor de Transcripción GATA2 , Enfermedades Hematológicas , Hematopoyesis , Mutación , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Animales , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Humanos
4.
EMBO J ; 31(9): 2103-16, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22446391

RESUMEN

The function of metabolic state in stemness is poorly understood. Mouse embryonic stem cells (ESC) and epiblast stem cells (EpiSC) are at distinct pluripotent states representing the inner cell mass (ICM) and epiblast embryos. Human embryonic stem cells (hESC) are similar to EpiSC stage. We now show a dramatic metabolic difference between these two stages. EpiSC/hESC are highly glycolytic, while ESC are bivalent in their energy production, dynamically switching from glycolysis to mitochondrial respiration on demand. Despite having a more developed and expanding mitochondrial content, EpiSC/hESC have low mitochondrial respiratory capacity due to low cytochrome c oxidase (COX) expression. Similarly, in vivo epiblasts suppress COX levels. These data reveal EpiSC/hESC functional similarity to the glycolytic phenotype in cancer (Warburg effect). We further show that hypoxia-inducible factor 1α (HIF1α) is sufficient to drive ESC to a glycolytic Activin/Nodal-dependent EpiSC-like stage. This metabolic switch during early stem-cell development may be deterministic.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Activinas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , ADN Mitocondrial/análisis , Femenino , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Prostaglandina-Endoperóxido Sintasas/metabolismo
5.
Blood ; 124(12): 1926-30, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25114263

RESUMEN

Familial platelet disorder with predisposition to acute myeloid leukemia (FPD/AML) is an autosomal dominant disease of the hematopoietic system that is caused by heterozygous mutations in RUNX1. FPD/AML patients have a bleeding disorder characterized by thrombocytopenia with reduced platelet numbers and functions, and a tendency to develop AML. No suitable animal models exist for FPD/AML, as Runx11/2 mice and zebra fish do not develop bleeding disorders or leukemia. Here we derived induced pluripotent stem cells (iPSCs) from 2 patients in a family with FPD/AML, and found that the FPD iPSCs display defects in megakaryocytic differentiation in vitro. We corrected the RUNX1 mutation in 1 FPD iPSC line through gene targeting, which led to normalization of megakaryopoiesis of the iPSCs in culture. Our results demonstrate successful in vitro modeling of FPD with patient-specific iPSCs and confirm that RUNX1 mutations are responsible for megakaryopoietic defects in FPD patients.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/genética , Trastornos de la Coagulación Sanguínea Heredados/terapia , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/terapia , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Mutación Missense , Reparación del Gen Blanco/métodos , Animales , Trastornos de la Coagulación Sanguínea Heredados/patología , Trastornos de las Plaquetas Sanguíneas/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/trasplante , Leucemia Mieloide Aguda/patología , Ratones , Trombopoyesis/genética
6.
Blood ; 123(4): 562-9, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24184683

RESUMEN

Hereditary neutropenia is usually caused by heterozygous germline mutations in the ELANE gene encoding neutrophil elastase (NE). How mutations cause disease remains uncertain, but two hypotheses have been proposed. In one, ELANE mutations lead to mislocalization of NE. In the other, ELANE mutations disturb protein folding, inducing an unfolded protein response in the endoplasmic reticulum (ER). In this study, we describe new types of mutations that disrupt the translational start site. At first glance, they should block translation and are incompatible with either the mislocalization or misfolding hypotheses, which require mutant protein for pathogenicity. We find that start-site mutations, instead, force translation from downstream in-frame initiation codons, yielding amino-terminally truncated isoforms lacking ER-localizing (pre) and zymogen-maintaining (pro) sequences, yet retain essential catalytic residues. Patient-derived induced pluripotent stem cells recapitulate hematopoietic and molecular phenotypes. Expression of the amino-terminally deleted isoforms in vitro reduces myeloid cell clonogenic capacity. We define an internal ribosome entry site (IRES) within ELANE and demonstrate that adjacent mutations modulate IRES activity, independently of protein-coding sequence alterations. Some ELANE mutations, therefore, appear to cause neutropenia via the production of amino-terminally deleted NE isoforms rather than by altering the coding sequence of the full-length protein.


Asunto(s)
Elastasa de Leucocito/genética , Elastasa de Leucocito/metabolismo , Mutación , Neutropenia/metabolismo , Biosíntesis de Proteínas , Apoptosis , Codón , Análisis Mutacional de ADN , Retículo Endoplásmico/metabolismo , Células HL-60 , Humanos , Células Madre Pluripotentes Inducidas/citología , Neutrófilos/citología , Fenotipo , Desnaturalización Proteica , Pliegue de Proteína , Isoformas de Proteínas/metabolismo , Células U937
7.
Br J Haematol ; 171(1): 13-28, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26018193

RESUMEN

Lymphocytes are unique among cells in that they undergo programmed DNA breaks and translocations, but that special property predisposes them to chromosomal instability (CIN), a cardinal feature of neoplastic lymphoid cells that manifests as whole chromosome- or translocation-based aneuploidy. In several lymphoid malignancies translocations may be the defining or diagnostic markers of the diseases. CIN is a cornerstone of the mutational architecture supporting lymphoid neoplasia, though it is perhaps one of the least understood components of malignant transformation in terms of its molecular mechanisms. CIN is associated with prognosis and response to treatment, making it a key area for impacting treatment outcomes and predicting prognoses. Here we will review the types and mechanisms of CIN found in Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma and the lymphoid leukaemias, with emphasis placed on pathogenic mutations affecting DNA recombination, replication and repair; telomere function; and mitotic regulation of spindle attachment, centrosome function, and chromosomal segregation. We will discuss the means by which chromosome-level genetic aberrations may give rise to multiple pathogenic mutations required for carcinogenesis and conclude with a discussion of the clinical applications of CIN and aneuploidy to diagnosis, prognosis and therapy.


Asunto(s)
Inestabilidad Cromosómica , Roturas del ADN , Replicación del ADN , ADN de Neoplasias , Neoplasias Hematológicas , Recombinación Genética , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/terapia , Humanos
8.
Nat Methods ; 9(1): 78-80, 2011 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-22120468

RESUMEN

Because mutations are inevitable, the genome of each cell in a multicellular organism becomes unique and therefore encodes a record of its ancestry. Here we coupled arbitrary single primer PCR with next-generation DNA sequencing to catalog mutations and deconvolve the phylogeny of cultured mouse cells. This study helps pave the way toward construction of retrospective cell-fate maps based on mutations accumulating in genomes of somatic cells.


Asunto(s)
Linaje de la Célula/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mutación , Análisis de Secuencia de ADN/métodos , Animales , Simulación por Computador , Genoma , Ratones , Filogenia , Reacción en Cadena de la Polimerasa/métodos , Reproducibilidad de los Resultados
9.
Blood ; 119(5): 1283-91, 2012 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-22147895

RESUMEN

Recent work has established that heterozygous germline GATA2 mutations predispose carriers to familial myelodysplastic syndrome (MDS)/acute myeloid leukemia (AML), "MonoMAC" syndrome, and DCML deficiency. Here, we describe a previously unreported MDS family carrying a missense GATA2 mutation (p.Thr354Met), one patient with MDS/AML carrying a frameshift GATA2 mutation (p.Leu332Thrfs*53), another with MDS harboring a GATA2 splice site mutation, and 3 patients exhibiting MDS or MDS/AML who have large deletions encompassing the GATA2 locus. Intriguingly, 2 MDS/AML or "MonoMAC" syndrome patients with GATA2 deletions and one with a frameshift mutation also have primary lymphedema. Primary lymphedema occurs as a result of aberrations in the development and/or function of lymphatic vessels, spurring us to investigate whether GATA2 plays a role in the lymphatic vasculature. We demonstrate here that GATA2 protein is present at high levels in lymphatic vessel valves and that GATA2 controls the expression of genes important for programming lymphatic valve development. Our data expand the phenotypes associated with germline GATA2 mutations to include predisposition to primary lymphedema and suggest that complete haploinsufficiency or loss of function of GATA2, rather than missense mutations, is the key predisposing factor for lymphedema onset. Moreover, we reveal a crucial role for GATA2 in lymphatic vascular development.


Asunto(s)
Factor de Transcripción GATA2/genética , Mutación de Línea Germinal , Leucemia Mieloide Aguda/genética , Vasos Linfáticos/metabolismo , Linfedema/congénito , Síndromes Mielodisplásicos/genética , Adolescente , Adulto , Animales , Células Cultivadas , Niño , Femenino , Factor de Transcripción GATA2/metabolismo , Factor de Transcripción GATA2/fisiología , Mutación de Línea Germinal/fisiología , Humanos , Recién Nacido , Linfangiogénesis/genética , Linfedema/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/patología , Síndrome , Adulto Joven
10.
J Biol Chem ; 287(46): 39083-93, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22988245

RESUMEN

The malignant cell in classical Hodgkin lymphoma (HL) is the binucleated giant Reed-Sternberg cell. Chromosomal instability and mitotic errors may contribute to HL pathogenesis; one potential mitotic regulator is the kelch protein KLHDC8B, which localizes to the midbody, is expressed during mitosis, and is mutated in a subset of familial and sporadic HL. We report that disrupting KLHDC8B function in HeLa cells, B lymphoblasts, and fibroblasts leads to significant increases in multinucleation, multipolar mitoses, failed abscission, asymmetric segregation of daughter nuclei, formation of anucleated daughter cells, centrosomal amplification, and aneuploidy. We recapitulated the major pathologic features of the Reed-Sternberg cell and concluded that KLHDC8B is essential for mitotic integrity and maintenance of chromosomal stability. The significant impact of KLHDC8B implicates the central roles of mitotic regulation and chromosomal segregation in the pathogenesis of HL and provides a novel molecular mechanism for chromosomal instability in HL.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Centrosoma/ultraestructura , Mitosis , Aneuploidia , Linfocitos B/citología , Proteínas de Ciclo Celular/química , Núcleo Celular/metabolismo , Inestabilidad Cromosómica , Citocinesis , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Enfermedad de Hodgkin/metabolismo , Humanos , Cariotipificación , Fenotipo , Células de Reed-Sternberg/metabolismo
11.
BMC Genomics ; 14: 39, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23327737

RESUMEN

BACKGROUND: The C. elegans cell fate map, in which the lineage of its approximately 1000 cells is visibly charted beginning from the zygote, represents a developmental biology milestone. Nematode development is invariant from one specimen to the next, whereas in mammals, aspects of development are probabilistic, and development exhibits variation between even genetically identical individuals. Consequently, a single defined cell fate map applicable to all individuals cannot exist. RESULTS: To determine the extent to which patterns of cell lineage are conserved between different mice, we have employed the recently developed method of "phylogenetic fate mapping" to compare cell fate maps in siblings. In this approach, somatic mutations arising in individual cells are used to retrospectively deduce lineage relationships through phylogenetic and-as newly investigated here-related analytical approaches based on genetic distance. We have cataloged genomic mutations at an average of 110 mutation-prone polyguanine (polyG) tracts for about 100 cells clonally isolated from various corresponding tissues of each of two littermates of a hypermutable mouse strain. CONCLUSIONS: We find that during mouse development, muscle and fat arise from a mixed progenitor cell pool in the germ layer, but, contrastingly, vascular endothelium in brain derives from a smaller source of progenitor cells. Additionally, formation of tissue primordia is marked by establishment of left and right lateral compartments, with restricted cell migration between divisions. We quantitatively demonstrate that development represents a combination of stochastic and deterministic events, offering insight into how chance influences normal development and may give rise to birth defects.


Asunto(s)
Ratones/crecimiento & desarrollo , Ratones/genética , Mutación , Animales , Proliferación Celular , Reparación del ADN/genética , Evolución Molecular , Femenino , Mitosis/genética , Filogenia , Análisis de la Célula Individual
12.
Pharmacol Rev ; 62(4): 726-59, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21079042

RESUMEN

Polymorphonuclear neutrophils are the first cells recruited to inflammatory sites and form the earliest line of defense against invading microorganisms. Neutrophil elastase, proteinase 3, and cathepsin G are three hematopoietic serine proteases stored in large quantities in neutrophil cytoplasmic azurophilic granules. They act in combination with reactive oxygen species to help degrade engulfed microorganisms inside phagolysosomes. These proteases are also externalized in an active form during neutrophil activation at inflammatory sites, thus contributing to the regulation of inflammatory and immune responses. As multifunctional proteases, they also play a regulatory role in noninfectious inflammatory diseases. Mutations in the ELA2/ELANE gene, encoding neutrophil elastase, are the cause of human congenital neutropenia. Neutrophil membrane-bound proteinase 3 serves as an autoantigen in Wegener granulomatosis, a systemic autoimmune vasculitis. All three proteases are affected by mutations of the gene (CTSC) encoding dipeptidyl peptidase I, a protease required for activation of their proform before storage in cytoplasmic granules. Mutations of CTSC cause Papillon-Lefèvre syndrome. Because of their roles in host defense and disease, elastase, proteinase 3, and cathepsin G are of interest as potential therapeutic targets. In this review, we describe the physicochemical functions of these proteases, toward a goal of better delineating their role in human diseases and identifying new therapeutic strategies based on the modulation of their bioavailability and activity. We also describe how nonhuman primate experimental models could assist with testing the efficacy of proposed therapeutic strategies.


Asunto(s)
Catepsina G/química , Catepsina G/fisiología , Elastasa de Leucocito/fisiología , Terapia Molecular Dirigida , Mieloblastina/fisiología , Animales , Dominio Catalítico , Catepsina G/antagonistas & inhibidores , Humanos , Elastasa de Leucocito/antagonistas & inhibidores , Elastasa de Leucocito/química , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/enzimología , Mieloblastina/antagonistas & inhibidores , Mieloblastina/química , Neutropenia/tratamiento farmacológico , Neutropenia/enzimología , Enfermedad de Papillon-Lefevre/tratamiento farmacológico , Enfermedad de Papillon-Lefevre/enzimología
13.
Nat Genet ; 34(3): 308-12, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12778173

RESUMEN

Mice lacking the transcriptional repressor oncoprotein Gfi1 are unexpectedly neutropenic. We therefore screened GFI1 as a candidate for association with neutropenia in affected individuals without mutations in ELA2 (encoding neutrophil elastase), the most common cause of severe congenital neutropenia (SCN; ref. 3). We found dominant negative zinc finger mutations that disable transcriptional repressor activity. The phenotype also includes immunodeficient lymphocytes and production of a circulating population of myeloid cells that appear immature. We show by chromatin immunoprecipitation, gel shift, reporter assays and elevated expression of ELA2 in vivo in neutropenic individuals that GFI1 represses ELA2, linking these two genes in a common pathway involved in myeloid differentiation.


Asunto(s)
Proteínas de Unión al ADN/genética , Elastasa de Leucocito/genética , Mutación Missense , Neutropenia/genética , Factores de Transcripción , Adulto , Anciano , Preescolar , Cromosomas/inmunología , Ensayo de Unidades Formadoras de Colonias , Femenino , Humanos , Lactante , Luciferasas/metabolismo , Masculino , Neutropenia/sangre , Neutropenia/etiología , Neutrófilos/enzimología , Linaje , Pruebas de Precipitina , Regiones Promotoras Genéticas , Proto-Oncogenes Mas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Dedos de Zinc
14.
Nat Genet ; 35(1): 90-6, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12897784

RESUMEN

Cyclic hematopoiesis is a stem cell disease in which the number of neutrophils and other blood cells oscillates in weekly phases. Autosomal dominant mutations of ELA2, encoding the protease neutrophil elastase, found in lysosome-like granules, cause cyclic hematopoiesis and most cases of the pre-leukemic disorder severe congenital neutropenia (SCN; ref. 3) in humans. Over 20 different mutations of neutrophil elastase have been identified, but their consequences are elusive, because they confer no consistent effects on enzymatic activity. The similar autosomal recessive disease of dogs, canine cyclic hematopoiesis, is not caused by mutations in ELA2 (data not shown). Here we show that homozygous mutation of the gene encoding the dog adaptor protein complex 3 (AP3) beta-subunit, directing trans-Golgi export of transmembrane cargo proteins to lysosomes, causes canine cyclic hematopoiesis. C-terminal processing of neutrophil elastase exposes an AP3 interaction signal responsible for redirecting neutrophil elastase trafficking from membranes to granules. Disruption of either neutrophil elastase or AP3 perturbs the intracellular trafficking of neutrophil elastase. Most mutations in ELA2 that cause human cyclic hematopoiesis prevent membrane localization of neutrophil elastase, whereas most mutations in ELA2 that cause SCN lead to exclusive membrane localization.


Asunto(s)
Complejo 3 de Proteína Adaptadora/genética , Enfermedades de los Perros/genética , Elastasa de Leucocito/genética , Mutación , Neutropenia/genética , Neutrófilos/enzimología , Complejo 3 de Proteína Adaptadora/metabolismo , Animales , Membrana Celular/enzimología , Enfermedades de los Perros/enzimología , Perros , Hematopoyesis , Humanos , Datos de Secuencia Molecular , Neutropenia/enzimología , Linaje , Señales de Clasificación de Proteína , Transporte de Proteínas
15.
Stem Cell Rev Rep ; 19(8): 2980-2990, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37702917

RESUMEN

Embryonic development is a continuum in vivo. Transcriptional analysis can separate established human embryonic stem cells (hESC) into at least four distinct developmental pluripotent stages, two naïve and two primed, early and late relative to the intact epiblast. In this study we primarily show that exposure of frozen human blastocysts to an inhibitor of checkpoint kinase 1 (CHK1) upon thaw greatly enhances establishment of karyotypically normal late naïve hESC cultures. These late naïve cells are plastic and can be toggled back to early naïve and forward to early primed pluripotent stages. The early primed cells are transcriptionally equivalent to the post inner cell mass intermediate (PICMI) stage seen one day following transfer of human blastocysts into in vitro culture and are stable at an earlier stage than conventional primed hESC.


Asunto(s)
Técnicas de Cultivo de Célula , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Células Madre Embrionarias Humanas , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Blastocisto/citología , Células Madre Pluripotentes/citología
16.
Mol Carcinog ; 51(11): 881-94, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22012859

RESUMEN

Elastase is the only currently identified target protein for indole-3-carbinol (I3C), a naturally occurring hydrolysis product of glucobrassicin in cruciferous vegetables such as broccoli, cabbage, and Brussels sprouts that induces a cell cycle arrest and apoptosis of human breast cancer cells. In vitro elastase enzymatic assays demonstrated that I3C and at lower concentrations its more potent derivative 1-benzyl-indole-3-carbinol (1-benzyl-I3C) act as non-competitive allosteric inhibitors of elastase activity. Consistent with these results, in silico computational simulations have revealed the first predicted interactions of I3C and 1-benzyl-I3C with the crystal structure of human neutrophil elastase, and identified a potential binding cluster on an external surface of the protease outside of the catalytic site that implicates elastase as a target protein for both indolecarbinol compounds. The Δ205 carboxyterminal truncation of elastase, which disrupts the predicted indolecarbinol binding site, is enzymatically active and generates a novel I3C resistant enzyme. Expression of the wild type and Δ205 elastase in MDA-MB-231 human breast cancer cells demonstrated that the carboxyterminal domain of elastase is required for the I3C and 1-benzyl-I3C inhibition of enzymatic activity, accumulation of the unprocessed form of the CD40 elastase substrate (a tumor necrosis factor receptor family member), disruption of NFκB nuclear localization and transcriptional activity, and induction of a G1 cell cycle arrest. Surprisingly, expression of the Δ205 elastase molecule failed to reverse indolecarbinol stimulated apoptosis, establishing an elastase-dependent bifurcation point in anti-proliferative signaling that uncouples the cell cycle and apoptotic responses in human breast cancer cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/enzimología , Ciclo Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Elastasa de Leucocito/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/química , Femenino , Humanos , Indoles/química , Elastasa de Leucocito/antagonistas & inhibidores , Elastasa de Leucocito/química , Elastasa de Leucocito/genética , Modelos Moleculares , Mutación , FN-kappa B/análisis , FN-kappa B/metabolismo , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Verduras/química
17.
Proc Natl Acad Sci U S A ; 106(46): 19450-4, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19887639

RESUMEN

To mimic events and molecules involved in type 1 insulin-dependent diabetes mellitus (T1D), we previously designed a transgenic (tg) mouse model where the viral nucleoprotein (NP) gene of lymphocytic choriomeningitis virus (LCMV) was expressed in the thymus to delete high affinity antiself (virus) T cells and in insulin-producing beta cells of the islets of Langerhans. Such tg mice, termed RIP-LCMV, fail to spontaneously develop diabetes. In contrast, when these mice are challenged with LCMV, they develop diabetes as they display hyperglycemia, low to absent levels of pancreatic insulin, and abundant mononuclear cell infiltrates in the islets. However, expressing the adenovirus early region (E3) gene in beta cells along with the LCMV transgene aborted the T1D. The present study utilizes this combined tg model (RIP LCMV x RIP E3) to define the requirement(s) of either pro-apoptotic TNF and Fas pathways or MHC class I up-regulation on beta cells for virus-induced T1D. Inhibitors to either pathway (TNF/Fas or MHC class I) are encoded in the E3 gene complex. To accomplish this task either the E3 region encoding the inhibitors of TNF and Fas pathways or the region encoding gp-19, a protein that inhibits transport of MHC class I molecules out of the endoplasmic reticulum were deleted in the RIP LCMV x RIP E3 model. Thus only the gp-19 is required to abort the virus-induced T1D. In contrast, removal of TNF- and Fas-pathway inhibitory genes had no effect on E3-mediated prevention of T1D.


Asunto(s)
Proteínas E3 de Adenovirus/inmunología , Apoptosis/inmunología , Diabetes Mellitus Experimental/virología , Diabetes Mellitus Tipo 1/virología , Células Secretoras de Insulina/virología , Proteínas E3 de Adenovirus/genética , Animales , Apoptosis/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Transgénicos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Receptor fas/genética , Receptor fas/inmunología
18.
Proc Natl Acad Sci U S A ; 106(49): 20871-6, 2009 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-19926851

RESUMEN

Chronic inflammation predisposes to a variety of human cancers. Affected tissues slowly accumulate mutations, some of which affect growth regulation and drive successive waves of clonal evolution, whereas a far greater number are functionally neutral and serve only to passively mark expanding clones. Ulcerative colitis (UC) is an inflammatory bowel disease, in which up to 10% of patients eventually develop colon cancer. Here we have mapped mutations in hypermutable intergenic and intronic polyguanine tracts in patients with UC to delineate the extent of clonal expansions associated with carcinogenesis. We genotyped colon biopsies for length altering mutations at 28 different polyguanine markers. In eight patients without neoplasia, we detected only two mutations in a single individual from among 37 total biopsies. In contrast, for 11 UC patients with neoplasia elsewhere in the colon, we identified 63 mutations in 51 nondysplastic biopsies, and every patient possessed at least one mutant clone. A subset of clones were large and extended over many square centimeters of colon. Of these, some occurred as isolated populations in nondysplastic tissue, considerably distant from neoplastic lesions. Other large clones included regions of cancer, suggesting that the tumor arose within a preexisting clonal field. Our results demonstrate that neutral mutations in polyguanine tracts serve as a unique tool for identifying fields of clonal expansions, which may prove clinically useful for distinguishing a subset of UC patients who are at risk for developing cancer.


Asunto(s)
Colitis Ulcerosa/patología , Neoplasias del Colon/diagnóstico , Proliferación Celular , Células Clonales , Neoplasias del Colon/patología , Electroforesis en Gel de Agar , Genotipo , Guanina/metabolismo , Humanos , Modelos Biológicos , Mutación/genética
19.
Proc Natl Acad Sci U S A ; 106(35): 14920-5, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19706467

RESUMEN

Classical Hodgkin lymphoma (cHL) is a malignancy of B-cell origin in which the neoplastic cells, known as "Reed-Sternberg" (RS) cells, are characteristically binucleated. Here we describe a family where multiple individuals developing cHL have inherited a reciprocal translocation between chromosomes 2 and 3. The translocation disrupts KLHDC8B, an uncharacterized gene from a region (3p21.31) previously implicated in lymphoma and related malignancies, resulting in its loss of expression. We tested KLHDC8B as a candidate gene for cHL and found that a 5'-UTR polymorphism responsible for decreasing its translational expression is associated with cHL in probands from other families with cHL and segregates with disease in those pedigrees. In one of three informative sporadic cases of cHL, we detected loss of heterozygosity (LOH) for KLHDC8B in RS cells, but not reactive T lymphocytes, purified from a malignant lymph node. KLHDC8B encodes a protein predicted to contain seven kelch repeat domains. KLHDC8B is expressed during mitosis, where it localizes to the midbody structure connecting cells about to separate during cytokinesis, and it is degraded after cell division. Depletion of KLHDC8B through RNA interference leads to an increase in binucleated cells, implicating its reduced expression in the formation of cHL's signature RS cell.


Asunto(s)
Antígenos de Neoplasias/genética , Núcleo Celular/genética , Cromosomas Humanos Par 2 , Cromosomas Humanos Par 3 , Enfermedad de Hodgkin/genética , Mutación , Regiones no Traducidas 5' , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Secuencia de Bases , Femenino , Predisposición Genética a la Enfermedad , Enfermedad de Hodgkin/patología , Humanos , Masculino , Persona de Mediana Edad , Linaje , Células de Reed-Sternberg/metabolismo , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Adulto Joven
20.
Semin Cancer Biol ; 20(5): 294-303, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20951806

RESUMEN

Cancer arises as the result of a natural selection process among cells of the body, favoring lineages bearing somatic mutations that bestow them with a proliferative advantage. Of the thousands of mutations within a tumor, only a small fraction functionally drive its growth; the vast majority are mere passengers of minimal biological consequence. Yet the presence of any mutation, independent of its role in facilitating proliferation, tags a cell's clonal descendants in a manner that allows them to be distinguished from unrelated cells. Such markers of cell lineage can be used to identify the abnormal proliferative signature of neoplastic clonal evolution, even at a stage which predates morphologically recognizable dysplasia. This article focuses on molecular techniques for assessing cellular clonality in humans with an emphasis on how they may be used for early detection of tumorigenic processes. We discuss historical as well as contemporary approaches and consider ways in which powerful new genomic technologies might be harnessed to develop a future generation of early cancer diagnostics.


Asunto(s)
Detección Precoz del Cáncer , Mutación , Neoplasias/diagnóstico , Neoplasias/genética , Linaje de la Célula , Células Clonales , Metilación de ADN , Reparación de la Incompatibilidad de ADN , Variación Genética , Genoma Mitocondrial , Humanos , Inestabilidad de Microsatélites , Neoplasias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA