Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 30(5): 1097-1154, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37100955

RESUMEN

Apoptosis is a form of regulated cell death (RCD) that involves proteases of the caspase family. Pharmacological and genetic strategies that experimentally inhibit or delay apoptosis in mammalian systems have elucidated the key contribution of this process not only to (post-)embryonic development and adult tissue homeostasis, but also to the etiology of multiple human disorders. Consistent with this notion, while defects in the molecular machinery for apoptotic cell death impair organismal development and promote oncogenesis, the unwarranted activation of apoptosis promotes cell loss and tissue damage in the context of various neurological, cardiovascular, renal, hepatic, infectious, neoplastic and inflammatory conditions. Here, the Nomenclature Committee on Cell Death (NCCD) gathered to critically summarize an abundant pre-clinical literature mechanistically linking the core apoptotic apparatus to organismal homeostasis in the context of disease.


Asunto(s)
Apoptosis , Caspasas , Animales , Humanos , Apoptosis/genética , Muerte Celular , Caspasas/genética , Caspasas/metabolismo , Carcinogénesis , Mamíferos/metabolismo
2.
bioRxiv ; 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36945615

RESUMEN

Interspecies chimera formation with human pluripotent stem cells (PSCs) holds great promise to generate humanized animal models and provide donor organs for transplant. However, the approach is currently limited by low levels of human cells ultimately represented in chimeric embryos. Different strategies have been developed to improve chimerism by genetically editing donor human PSCs. To date, however, it remains unexplored if human chimerism can be enhanced in animals through modifying the host embryos. Leveraging the interspecies PSC competition model, here we discovered retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) signaling, an RNA sensor, in "winner" cells plays an important role in the competitive interactions between co-cultured mouse and human PSCs. We found that genetic inactivation of Ddx58/Ifih1-Mavs-Irf7 axis compromised the "winner" status of mouse PSCs and their ability to outcompete PSCs from evolutionarily distant species during co-culture. Furthermore, by using Mavs-deficient mouse embryos we substantially improved unmodified donor human cell survival. Comparative transcriptome analyses based on species-specific sequences suggest contact-dependent human-to-mouse transfer of RNAs likely plays a part in mediating the cross-species interactions. Taken together, these findings establish a previously unrecognized role of RNA sensing and innate immunity in "winner" cells during cell competition and provides a proof-of-concept for modifying host embryos, rather than donor PSCs, to enhance interspecies chimerism.

3.
Mob DNA ; 13(1): 30, 2022 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-36461093

RESUMEN

BACKGROUND: LINE-1s, Alus and SVAs are the only retrotransposition competent elements in humans. Their mobilization followed by insertional mutagenesis is often linked to disease. Apart from these rare integration events, accumulation of retrotransposition intermediates in the cytoplasm is potentially pathogenic due to induction of inflammatory response pathways. Although the retrotransposition of LINE-1 and Alu retroelements has been studied in considerable detail, there are mixed observations about the localization of their RNAs. RESULTS: We undertook a comprehensive and unbiased approach to analyze retroelement RNA localization using common cell lines and publicly available datasets containing RNA-sequencing data from subcellular fractions. Using our customized analytic pipeline, we compared localization patterns of RNAs transcribed from retroelements and single-copy protein coding genes. Our results demonstrate a generalized characteristic pattern of retroelement RNA nuclear localization that is conserved across retroelement classes as well as evolutionarily young and ancient elements. Preferential nuclear enrichment of retroelement transcripts was consistently observed in cell lines, in vivo and across species. Moreover, retroelement RNA localization patterns were dynamic and subject to change during development, as seen in zebrafish embryos. CONCLUSION: The pronounced nuclear localization of transcripts arising from ancient as well as de novo transcribed retroelements suggests that these transcripts are retained in the nucleus as opposed to being re-imported to the nucleus or degraded in the cytoplasm. This raises the possibility that there is adaptive value associated with this localization pattern to the host, the retroelements or possibly both.

4.
Dev Cell ; 57(15): 1833-1846.e6, 2022 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-35820415

RESUMEN

p53 genes are conserved transcriptional activators that respond to stress. These proteins can also downregulate genes, but the mechanisms are not understood and are generally assumed to be indirect. Here, we investigate synthetic and native cis-regulatory elements in Drosophila to examine opposing features of p53-mediated transcriptional control in vivo. We show that transcriptional repression by p53 operates continuously through canonical DNA binding sites that confer p53-dependent transactivation at earlier developmental stages. p53 transrepression is correlated with local H3K9me3 chromatin marks and occurs without the need for stress or Chk2. In sufficiency tests, two p53 isoforms qualify as transrepressors and a third qualifies as a transcriptional activator. Targeted isoform-specific knockouts dissociate these opposing transcriptional activities, highlighting features that are dispensable for transactivation but critical for repression and for proper germ cell formation. Together, these results demonstrate that certain p53 isoforms function as constitutive tissue-specific repressors, raising important implications for tumor suppression by the human counterpart.


Asunto(s)
Cromatina , Proteína p53 Supresora de Tumor , Animales , Sitios de Unión , Cromatina/genética , Drosophila/genética , Drosophila/metabolismo , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo
5.
Nat Genet ; 53(5): 672-682, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33833453

RESUMEN

Transposable elements or transposons are major players in genetic variability and genome evolution. Aberrant activation of long interspersed element-1 (LINE-1 or L1) retrotransposons is common in human cancers, yet their tumor-type-specific functions are poorly characterized. We identified MPHOSPH8/MPP8, a component of the human silencing hub (HUSH) complex, as an acute myeloid leukemia (AML)-selective dependency by epigenetic regulator-focused CRISPR screening. Although MPP8 is dispensable for steady-state hematopoiesis, MPP8 loss inhibits AML development by reactivating L1s to induce the DNA damage response and cell cycle exit. Activation of endogenous or ectopic L1s mimics the phenotype of MPP8 loss, whereas blocking retrotransposition abrogates MPP8-deficiency-induced phenotypes. Expression of AML oncogenic mutations promotes L1 suppression, and enhanced L1 silencing is associated with poor prognosis in human AML. Hence, while retrotransposons are commonly recognized for their cancer-promoting functions, we describe a tumor-suppressive role for L1 retrotransposons in myeloid leukemia.


Asunto(s)
Silenciador del Gen , Leucemia Mieloide/genética , Elementos de Nucleótido Esparcido Largo/genética , Animales , Sistemas CRISPR-Cas/genética , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Genoma Humano , Inestabilidad Genómica , Hematopoyesis/genética , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/genética
6.
Genes Dev ; 34(21-22): 1439-1451, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33060137

RESUMEN

p53 is a potent tumor suppressor and commonly mutated in human cancers. Recently, we demonstrated that p53 genes act to restrict retrotransposons in germline tissues of flies and fish but whether this activity is conserved in somatic human cells is not known. Here we show that p53 constitutively restrains human LINE1s by cooperatively engaging sites in the 5'UTR and stimulating local deposition of repressive histone marks at these transposons. Consistent with this, the elimination of p53 or the removal of corresponding binding sites in LINE1s, prompted these retroelements to become hyperactive. Concurrently, p53 loss instigated chromosomal rearrangements linked to LINE sequences and also provoked inflammatory programs that were dependent on reverse transcriptase produced from LINE1s. Taken together, our observations establish that p53 continuously operates at the LINE1 promoter to restrict autonomous copies of these mobile elements in human cells. Our results further suggest that constitutive restriction of these retroelements may help to explain tumor suppression encoded by p53, since erupting LINE1s produced acute oncogenic threats when p53 was absent.


Asunto(s)
Regulación de la Expresión Génica/genética , Elementos de Nucleótido Esparcido Largo/genética , Retroelementos/genética , Proteína p53 Supresora de Tumor/metabolismo , Sitios de Unión , Línea Celular , Eliminación de Gen , Reordenamiento Génico/genética , Código de Histonas/genética , Humanos , Inmunidad/genética , Elementos de Nucleótido Esparcido Largo/inmunología , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteína p53 Supresora de Tumor/genética
7.
Fly (Austin) ; 14(1-4): 3-9, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32615907

RESUMEN

Glutamine: fructose-6-phosphate amidotransferase (GFAT) enzymes catalyse the first committed step of the hexosamine biosynthesis pathway (HBP) using glutamine and fructose-6-phosphate to form glucosamine-6-phosphate (GlcN6P). Numerous species (e.g. mouse, rat, zebrafish, chicken) including humans and Drosophila encode two broadly expressed copies of this enzyme but whether these perform redundant, partially overlapping or distinct functions is not known. To address this question, we produced single gene null mutations in the fly counterparts of gfat1 and gfat2. Deletions for either enzyme were fully lethal and homozygotes lacking either GFAT1 or GFAT2 died at or prior to the first instar larval stage. Therefore, when genetically eliminated, neither isoform was able to compensate for the other. Importantly, dietary supplementation with D-glucosamine-6-phosphate rescued GFAT2 deficiency and restored viability to gfat2-/- mutants. In contrast, glucosamine-6-phosphate did not rescue gfat1-/- animals.


Asunto(s)
Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Sistemas CRISPR-Cas , Suplementos Dietéticos , Proteínas de Drosophila/genética , Regulación Enzimológica de la Expresión Génica , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/genética , Larva , Mutación , Sobrevida
8.
Mol Biol Cell ; 30(11): 1339-1351, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30892991

RESUMEN

TP53 is the most frequently mutated gene in human cancers, and despite intensive research efforts, genome-scale studies of p53 function in whole animal models are rare. The need for such in vivo studies is underscored by recent challenges to established paradigms, indicating that unappreciated p53 functions contribute to cancer prevention. Here we leveraged the Drosophila system to interrogate p53 function in a postmitotic context. In the developing embryo, p53 robustly activates important apoptotic genes in response to radiation-induced DNA damage. We recently showed that a p53 enhancer (p53RErpr) near the cell death gene reaper forms chromatin contacts and enables p53 target activation across long genomic distances. Interestingly, we found that this canonical p53 apoptotic program fails to activate in adult heads. Moreover, this failure to exhibit apoptotic responses was not associated with altered chromatin contacts. Instead, we determined that p53 does not occupy the p53RErpr enhancer in this postmitotic tissue as it does in embryos. Through comparative RNA-seq and chromatin immunoprecipitation-seq studies of developing and postmitotic tissues, we further determined that p53 regulates distinct transcriptional programs in adult heads, including DNA repair, metabolism, and proteolysis genes. Strikingly, in the postmitotic context, p53-binding landscapes were poorly correlated with nearby transcriptional effects, raising the possibility that p53 enhancers could be generally acting through long distances.


Asunto(s)
Reparación del ADN , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Inmunoprecipitación de Cromatina , ADN/metabolismo , ADN/efectos de la radiación , Daño del ADN , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Radiación Ionizante , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN , Proteína p53 Supresora de Tumor/genética
9.
Mob DNA ; 9: 21, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30211913

RESUMEN

The Mobile Genetic Elements and Genome Plasticity conference was hosted by Keystone Symposia in Santa Fe, NM USA, February 11-15, 2018. The organizers were Marlene Belfort, Evan Eichler, Henry Levin and Lynn Maquat. The goal of this conference was to bring together scientists from around the world to discuss the function of transposable elements and their impact on host species. Central themes of the meeting included recent innovations in genome analysis and the role of mobile DNA in disease and evolution. The conference included 200 scientists who participated in poster presentations, short talks selected from abstracts, and invited talks. A total of 58 talks were organized into eight sessions and two workshops. The topics varied from mechanisms of mobilization, to the structure of genomes and their defense strategies to protect against transposable elements.

10.
Trends Genet ; 34(11): 846-855, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30195581

RESUMEN

p53, the most commonly mutated tumor suppressor, is a transcription factor known to regulate proliferation, senescence, and apoptosis. Compelling studies have found that p53 may prevent oncogenesis through effectors that are unrelated to these canonical processes and recent findings have uncovered ancient roles for p53 in the containment of mobile elements. Together, these developments raise the possibility that some p53-driven cancers could result from unrestrained transposons. Here, we explore evidence linking conserved features of p53 biology to the control of transposons. We also show how p53-deficient cells can be exploited to probe the behavior of transposons and illustrate how unrestrained transposons incited by p53 loss might contribute to human malignancies.


Asunto(s)
Elementos Transponibles de ADN/genética , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proliferación Celular/genética , Senescencia Celular/genética , Inestabilidad Genómica/genética , Humanos
11.
Cell Death Differ ; 25(3): 486-541, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29362479

RESUMEN

Over the past decade, the Nomenclature Committee on Cell Death (NCCD) has formulated guidelines for the definition and interpretation of cell death from morphological, biochemical, and functional perspectives. Since the field continues to expand and novel mechanisms that orchestrate multiple cell death pathways are unveiled, we propose an updated classification of cell death subroutines focusing on mechanistic and essential (as opposed to correlative and dispensable) aspects of the process. As we provide molecularly oriented definitions of terms including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence, and mitotic catastrophe, we discuss the utility of neologisms that refer to highly specialized instances of these processes. The mission of the NCCD is to provide a widely accepted nomenclature on cell death in support of the continued development of the field.


Asunto(s)
Muerte Celular , Animales , Humanos , Lisosomas/metabolismo , Lisosomas/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Necrosis/metabolismo , Necrosis/patología
12.
Curr Biol ; 27(19): 3010-3016.e3, 2017 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-28966088

RESUMEN

Retrotransposons are a pervasive class of mobile elements present in the genomes of virtually all forms of life [1, 2]. In metazoans, these are preferentially active in the germline, which, in turn, mounts defenses that restrain their activity [3, 4]. Here we report that certain classes of retrotransposons ensure transgenerational inheritance by invading presumptive germ cells before they are formed. Using sensitized Drosophila and zebrafish models, we found that diverse classes of retrotransposons migrate to the germ plasm, a specialized region of the oocyte that prefigures germ cells and specifies the germline of descendants in the fertilized egg. In Drosophila, we found evidence for a "stowaway" model, whereby Tahre retroelements traffic to the germ plasm by mimicking oskar RNAs and engaging the Staufen-dependent active transport machinery. Consistent with this, germ plasm determinants attracted retroelement RNAs even when these components were ectopically positioned in bipolar oocytes. Likewise, vertebrate retrotransposons similarly migrated to the germ plasm in zebrafish oocytes. Together, these results suggest that germ plasm targeting represents a fitness strategy adopted by some retrotransposons to ensure transgenerational propagation.


Asunto(s)
Drosophila melanogaster/genética , Oocitos/metabolismo , Retroelementos/genética , Pez Cebra/genética , Animales , Herencia/genética , Oocitos/crecimiento & desarrollo , ARN Mensajero/metabolismo
13.
Bioessays ; 38(11): 1111-1116, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27644006

RESUMEN

Throughout the animal kingdom, p53 genes function to restrain mobile elements and recent observations indicate that transposons become derepressed in human cancers. Together, these emerging lines of evidence suggest that cancers driven by p53 mutations could represent "transpospoathies," i.e. disease states linked to eruptions of mobile elements. The transposopathy hypothesis predicts that p53 acts through conserved mechanisms to contain transposon movement, and in this way, prevents tumor formation. How transposon eruptions provoke neoplasias is not well understood but, from a broader perspective, this hypothesis also provides an attractive framework to explore unrestrained mobile elements as inciters of late-onset idiopathic disease. Also see the video abstract here.


Asunto(s)
Elementos Transponibles de ADN , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética , Animales , Humanos , Mutación , Neoplasias/metabolismo
14.
Genes Dev ; 30(1): 64-77, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26701264

RESUMEN

Throughout the animal kingdom, p53 genes govern stress response networks by specifying adaptive transcriptional responses. The human member of this gene family is mutated in most cancers, but precisely how p53 functions to mediate tumor suppression is not well understood. Using Drosophila and zebrafish models, we show that p53 restricts retrotransposon activity and genetically interacts with components of the piRNA (piwi-interacting RNA) pathway. Furthermore, transposon eruptions occurring in the p53(-) germline were incited by meiotic recombination, and transcripts produced from these mobile elements accumulated in the germ plasm. In gene complementation studies, normal human p53 alleles suppressed transposons, but mutant p53 alleles from cancer patients could not. Consistent with these observations, we also found patterns of unrestrained retrotransposons in p53-driven mouse and human cancers. Furthermore, p53 status correlated with repressive chromatin marks in the 5' sequence of a synthetic LINE-1 element. Together, these observations indicate that ancestral functions of p53 operate through conserved mechanisms to contain retrotransposons. Since human p53 mutants are disabled for this activity, our findings raise the possibility that p53 mitigates oncogenic disease in part by restricting transposon mobility.


Asunto(s)
Genes p53/genética , Retroelementos/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Drosophila/genética , Femenino , Variación Genética , Humanos , Masculino , Ratones , Mutación/genética , Neoplasias/genética , Retroelementos/genética , Pez Cebra/genética
15.
Mech Dev ; 138 Pt 3: 349-55, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26226435

RESUMEN

Elimination of cells and tissues by apoptosis is a highly conserved and tightly regulated process. In Drosophila, the entire wing epithelium is completely removed shortly after eclosion. The cells that make up this epithelium are collectively eliminated through a highly synchronized form of apoptotic cell death, involving canonical apoptosome genes. Here we present evidence that collective cell death does not require cell-cell contact and show that transcription of the IAP antagonist, head involution defective, is acutely induced in wing epithelial cells prior to this process. hid mRNAs accumulate to levels that exceed a component of the ribosome and likewise, Hid protein becomes highly abundant in these same cells. hid function is required for collective cell death, since loss of function mutants shows persisting wing epithelial cells and, furthermore, silencing of the hormone bursicon in the CNS produced collective cell death defective phenotypes manifested in the wing epithelium. Taken together, our observations suggest that acute induction of Hid primes wing epithelial cells for collective cell death and that Bursicon is a strong candidate to trigger this process, possibly by activating the abundant pool of Hid protein already present.


Asunto(s)
Apoptosis/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Neuropéptidos/fisiología , Alas de Animales/citología , Alas de Animales/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Apoptosis/genética , Adhesión Celular , Comunicación Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Genes de Insecto , Proteínas Inhibidoras de la Apoptosis/metabolismo , Hormonas de Invertebrados/antagonistas & inhibidores , Hormonas de Invertebrados/genética , Hormonas de Invertebrados/fisiología , Neuropéptidos/genética , Alas de Animales/metabolismo
16.
Cell Rep ; 10(7): 1096-109, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25704813

RESUMEN

The p53 tumor suppressor plays a key role in maintaining cellular integrity. In response to diverse stress signals, p53 can trigger apoptosis to eliminate damaged cells or cell-cycle arrest to enable cells to cope with stress and survive. However, the transcriptional networks underlying p53 pro-survival function are incompletely understood. Here, we show that in oncogenic-Ras-expressing cells, p53 promotes oxidative phosphorylation (OXPHOS) and cell survival upon glucose starvation. Analysis of p53 transcriptional activation domain mutants reveals that these responses depend on p53 transactivation function. Using gene expression profiling and ChIP-seq analysis, we identify several p53-inducible fatty acid metabolism-related genes. One such gene, Acad11, encoding a protein involved in fatty acid oxidation, is required for efficient OXPHOS and cell survival upon glucose starvation. This study provides new mechanistic insight into the pro-survival function of p53 and suggests that targeting this pathway may provide a strategy for therapeutic intervention based on metabolic perturbation.


Asunto(s)
Acil-CoA Deshidrogenasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acil-CoA Deshidrogenasa/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Redes Reguladoras de Genes , Glucosa/farmacología , Humanos , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación Oxidativa/efectos de los fármacos , Estructura Terciaria de Proteína , Interferencia de ARN , Alineación de Secuencia , Estrés Fisiológico , Activación Transcripcional , Trasplante Heterólogo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética
17.
PLoS One ; 9(8): e104858, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25121966

RESUMEN

BACKGROUND: Ionizing radiation is genotoxic to cells. Healthy tissue toxicity in patients and radiation resistance in tumors present common clinical challenges in delivering effective radiation therapies. Radiation response is a complex, polygenic trait with unknown genetic determinants. The Drosophila Genetic Reference Panel (DGRP) provides a model to investigate the genetics of natural variation for sensitivity to radiation. METHODS AND FINDINGS: Radiation response was quantified in 154 inbred DGRP lines, among which 92 radiosensitive lines and 62 radioresistant lines were classified as controls and cases, respectively. A case-control genome-wide association screen for radioresistance was performed. There are 32 single nucleotide polymorphisms (SNPs) associated with radio resistance at a nominal p<10(-5); all had modest effect sizes and were common variants with the minor allele frequency >5%. All the genes implicated by those SNP hits were novel, many without a known role in radiation resistance and some with unknown function. Variants in known DNA damage and repair genes associated with radiation response were below the significance threshold of p<10(-5) and were not present among the significant hits. No SNP met the genome-wide significance threshold (p = 1.49 × 10(-7)), indicating a necessity for a larger sample size. CONCLUSIONS: Several genes not previously associated with variation in radiation resistance were identified. These genes, especially the ones with human homologs, form the basis for exploring new pathways involved in radiation resistance in novel functional studies. An improved DGRP model with a sample size of at least 265 lines and ideally up to 793 lines is recommended for future studies of complex traits.


Asunto(s)
Drosophila melanogaster/efectos de la radiación , Estudio de Asociación del Genoma Completo , Tolerancia a Radiación/genética , Animales , Drosophila melanogaster/genética , Polimorfismo de Nucleótido Simple
19.
Elife ; 3: e01530, 2014 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-24618896

RESUMEN

Oncogenic stress provokes tumor suppression by p53 but the extent to which this regulatory axis is conserved remains unknown. Using a biosensor to visualize p53 action, we find that Drosophila p53 is selectively active in gonadal stem cells after exposure to stressors that destabilize the genome. Similar p53 activity occurred in hyperplastic growths that were triggered either by the Ras(V12) oncoprotein or by failed differentiation programs. In a model of transient sterility, p53 was required for the recovery of fertility after stress, and entry into the cell cycle was delayed in p53(-) stem cells. Together, these observations establish that the stem cell compartment of the Drosophila germline is selectively licensed for stress-induced activation of the p53 regulatory network. Furthermore, the findings uncover ancestral links between p53 and aberrant proliferation that are independent of DNA breaks and predate evolution of the ARF/Mdm2 axis. DOI: http://dx.doi.org/10.7554/eLife.01530.001.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ovario/metabolismo , Nicho de Células Madre , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Técnicas Biosensibles , Puntos de Control del Ciclo Celular , Proliferación Celular , Daño del ADN , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Fertilidad , Regulación de la Expresión Génica , Inestabilidad Genómica , Infertilidad/genética , Infertilidad/metabolismo , Infertilidad/fisiopatología , Ovario/patología , Ovario/fisiopatología , Transducción de Señal , Células Madre/patología , Estrés Fisiológico , Factores de Tiempo , Proteína p53 Supresora de Tumor/genética
20.
Genes Dev ; 27(22): 2433-8, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24240233

RESUMEN

We examined how a p53 enhancer transmits regulatory information in vivo. Using genetic ablation together with digital chromosome conformation capture and fluorescent in situ hybridization, we found that a Drosophila p53 enhancer region (referred to as the p53 response element [p53RE]) physically contacts targets in cis and across the centromere to control stress-responsive transcription at these sites. Furthermore, when placed at ectopic genomic positions, fragments spanning this element re-established chromatin contacts and partially restored target gene regulation to mutants lacking the native p53RE. Therefore, a defined p53 enhancer region is sufficient for long-range chromatin interactions that enable multigenic regulation.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Drosophila/genética , Drosophila/metabolismo , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Proteína p53 Supresora de Tumor/genética , Animales , Centrómero/metabolismo , Unión Proteica , Estrés Fisiológico/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...