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










Base de datos
Intervalo de año de publicación
1.
medRxiv ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38260438

RESUMEN

Phospholipase C isozymes (PLCs) hydrolyze phosphatidylinositol 4,5-bisphosphate into inositol 1,4,5-trisphosphate and diacylglycerol, important signaling molecules involved in many cellular processes. PLCG1 encodes the PLCγ1 isozyme that is broadly expressed. Hyperactive somatic mutations of PLCG1 are observed in multiple cancers, but only one germline variant has been reported. Here we describe three unrelated individuals with de novo heterozygous missense variants in PLCG1 (p.Asp1019Gly, p.His380Arg, and p.Asp1165Gly) who exhibit variable phenotypes including hearing loss, ocular pathology and cardiac septal defects. To model these variants in vivo, we generated the analogous variants in the Drosophila ortholog, small wing (sl). We created a null allele slT2A and assessed the expression pattern. sl is broadly expressed, including in wing discs, eye discs, and a subset of neurons and glia. Loss of sl causes wing size reductions, ectopic wing veins and supernumerary photoreceptors. We document that mutant flies exhibit a reduced lifespan and age-dependent locomotor defects. Expressing wild-type sl in slT2A mutant rescues the loss-of-function phenotypes whereas expressing the variants causes lethality. Ubiquitous overexpression of the variants also reduces viability, suggesting that the variants are toxic. Ectopic expression of an established hyperactive PLCG1 variant (p.Asp1165His) in the wing pouch causes severe wing phenotypes, resembling those observed with overexpression of the p.Asp1019Gly or p.Asp1165Gly variants, further arguing that these two are gain-of-function variants. However, the wing phenotypes associated with p.His380Arg overexpression are mild. Our data suggest that the PLCG1 de novo heterozygous missense variants are pathogenic and contribute to the features observed in the probands.

2.
Nat Commun ; 13(1): 3156, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672419

RESUMEN

Many vital processes in the eye are under circadian regulation, and circadian dysfunction has emerged as a potential driver of eye aging. Dietary restriction is one of the most robust lifespan-extending therapies and amplifies circadian rhythms with age. Herein, we demonstrate that dietary restriction extends lifespan in Drosophila melanogaster by promoting circadian homeostatic processes that protect the visual system from age- and light-associated damage. Altering the positive limb core molecular clock transcription factor, CLOCK, or CLOCK-output genes, accelerates visual senescence, induces a systemic immune response, and shortens lifespan. Flies subjected to dietary restriction are protected from the lifespan-shortening effects of photoreceptor activation. Inversely, photoreceptor inactivation, achieved via mutating rhodopsin or housing flies in constant darkness, primarily extends the lifespan of flies reared on a high-nutrient diet. Our findings establish the eye as a diet-sensitive modulator of lifespan and indicates that vision is an antagonistically pleiotropic process that contributes to organismal aging.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Ojo , Animales , Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Longevidad/genética , Factores de Transcripción/genética
3.
Methods Mol Biol ; 2472: 235-276, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35674905

RESUMEN

Rare variants in the many genes related to Notch signaling cause diverse Mendelian diseases that affect myriad organ systems. In addition, genome- and exome-wide association studies have linked common and rare variants in Notch-related genes to common diseases and phenotypic traits. Moreover, somatic mutations in these genes have been observed in many types of cancer, some of which are classified as oncogenic and others as tumor suppressive. While functional characterization of some of these variants has been performed through experimental studies, the number of "variants of unknown significance" identified in patients with diverse conditions keeps increasing as high-throughput sequencing technologies become more commonly used in the clinic. Furthermore, as disease gene discovery efforts identify rare variants in human genes that have yet to be linked to a disease, the demand for functional characterization of variants in these "genes of unknown significance" continues to increase. In this chapter, we describe a workflow to functionally characterize a rare variant in a Notch signaling related gene that was found to be associated with late-onset Alzheimer's disease. This pipeline involves informatic analysis of the variant of interest using diverse human and model organism databases, followed by in vivo experiments in the fruit fly Drosophila melanogaster. The protocol described here can be used to study variants that affect amino acids that are not conserved between human and fly. By "humanizing" the almondex gene in Drosophila with mutant alleles and heterologous genomic rescue constructs, a missense variant in TM2D3 (TM2 Domain Containing 3) was shown to be functionally damaging. This, and similar approaches, greatly facilitate functional interpretations of genetic variants in the human genome and propel personalized medicine.


Asunto(s)
Drosophila melanogaster , Drosophila , Animales , Drosophila/genética , Drosophila melanogaster/genética , Exoma , Predisposición Genética a la Enfermedad , Genómica/métodos , Humanos , Fenotipo
4.
PLoS Genet ; 17(12): e1009962, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34905536

RESUMEN

TM2 domain containing (TM2D) proteins are conserved in metazoans and encoded by three separate genes in each model organism species that has been sequenced. Rare variants in TM2D3 are associated with Alzheimer's disease (AD) and its fly ortholog almondex is required for embryonic Notch signaling. However, the functions of this gene family remain elusive. We knocked-out all three TM2D genes (almondex, CG11103/amaretto, CG10795/biscotti) in Drosophila and found that they share the same maternal-effect neurogenic defect. Triple null animals are not phenotypically worse than single nulls, suggesting these genes function together. Overexpression of the most conserved region of the TM2D proteins acts as a potent inhibitor of Notch signaling at the γ-secretase cleavage step. Lastly, Almondex is detected in the brain and its loss causes shortened lifespan accompanied by progressive motor and electrophysiological defects. The functional links between all three TM2D genes are likely to be evolutionarily conserved, suggesting that this entire gene family may be involved in AD.


Asunto(s)
Proteínas de Drosophila , Proteínas de la Membrana , Neurogénesis , Receptores Notch , Animales , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Técnicas de Inactivación de Genes , Proteínas de la Membrana/genética , Mutación/genética , Neurogénesis/genética , Neuronas/metabolismo , Receptores Notch/genética , Transducción de Señal/genética
5.
Elife ; 92020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286230

RESUMEN

Retromer, including Vps35, Vps26, and Vps29, is a protein complex responsible for recycling proteins within the endolysosomal pathway. Although implicated in both Parkinson's and Alzheimer's disease, our understanding of retromer function in the adult brain remains limited, in part because Vps35 and Vps26 are essential for development. In Drosophila, we find that Vps29 is dispensable for embryogenesis but required for retromer function in aging adults, including for synaptic transmission, survival, and locomotion. Unexpectedly, in Vps29 mutants, Vps35 and Vps26 proteins are normally expressed and associated, but retromer is mislocalized from neuropil to soma with the Rab7 GTPase. Further, Vps29 phenotypes are suppressed by reducing Rab7 or overexpressing the GTPase activating protein, TBC1D5. With aging, retromer insufficiency triggers progressive endolysosomal dysfunction, with ultrastructural evidence of impaired substrate clearance and lysosomal stress. Our results reveal the role of Vps29 in retromer localization and function, highlighting requirements for brain homeostasis in aging.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Transmisión Sináptica/fisiología , Proteínas de Transporte Vesicular/metabolismo , Animales , Drosophila , Proteínas de Drosophila/metabolismo
6.
Neuron ; 106(4): 589-606.e6, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32169171

RESUMEN

ACOX1 (acyl-CoA oxidase 1) encodes the first and rate-limiting enzyme of the very-long-chain fatty acid (VLCFA) ß-oxidation pathway in peroxisomes and leads to H2O2 production. Unexpectedly, Drosophila (d) ACOX1 is mostly expressed and required in glia, and loss of ACOX1 leads to developmental delay, pupal death, reduced lifespan, impaired synaptic transmission, and glial and axonal loss. Patients who carry a previously unidentified, de novo, dominant variant in ACOX1 (p.N237S) also exhibit glial loss. However, this mutation causes increased levels of ACOX1 protein and function resulting in elevated levels of reactive oxygen species in glia in flies and murine Schwann cells. ACOX1 (p.N237S) patients exhibit a severe loss of Schwann cells and neurons. However, treatment of flies and primary Schwann cells with an antioxidant suppressed the p.N237S-induced neurodegeneration. In summary, both loss and gain of ACOX1 lead to glial and neuronal loss, but different mechanisms are at play and require different treatments.


Asunto(s)
Acil-CoA Oxidasa/genética , Axones/enzimología , Degeneración Nerviosa/genética , Neuroglía/enzimología , Animales , Axones/patología , Drosophila , Humanos , Ratones , Mutación , Degeneración Nerviosa/enzimología , Neuroglía/patología , Ratas
7.
Dev Growth Differ ; 62(1): 80-93, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31782145

RESUMEN

Notch signaling plays crucial roles in the control of cell fate and physiology through local cell-cell interactions. The core processes of Notch signal transduction are well established, but the mechanisms that fine-tune the pathway in various developmental and post-developmental contexts are less clear. Drosophila almondex, which encodes an evolutionarily conserved double-pass transmembrane protein, was identified in the 1970s as a maternal-effect gene that regulates Notch signaling in certain contexts, but its mechanistic function remains obscure. In this study, we examined the role of almondex in Notch signaling during early Drosophila embryogenesis. We found that in addition to being required for lateral inhibition in the neuroectoderm, almondex is also partially required for Notch signaling-dependent single-minded expression in the mesectoderm. Furthermore, we found that almondex is required for proper subcellular Notch receptor distribution in the neuroectoderm, specifically during mid-stage 5 development. The absence of maternal almondex during this critical window of time caused Notch to accumulate abnormally in cells in a mesh-like pattern. This phenotype did not include any obvious change in subcellular Delta ligand distribution, suggesting that it does not result from a general vesicular-trafficking defect. Considering that dynamic Notch trafficking regulates signal output to fit the specific context, we speculate that almondex may facilitate Notch activation by regulating intracellular Notch receptor distribution during early embryogenesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Neurogénesis , Receptores Notch/metabolismo , Transducción de Señal , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Receptores Notch/genética
8.
Am J Hum Genet ; 105(6): 1237-1253, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31785787

RESUMEN

We report an early-onset autosomal-recessive neurological disease with cerebellar atrophy and lysosomal dysfunction. We identified bi-allelic loss-of-function (LoF) variants in Oxidative Resistance 1 (OXR1) in five individuals from three families; these individuals presented with a history of severe global developmental delay, current intellectual disability, language delay, cerebellar atrophy, and seizures. While OXR1 is known to play a role in oxidative stress resistance, its molecular functions are not well established. OXR1 contains three conserved domains: LysM, GRAM, and TLDc. The gene encodes at least six transcripts, including some that only consist of the C-terminal TLDc domain. We utilized Drosophila to assess the phenotypes associated with loss of mustard (mtd), the fly homolog of OXR1. Strong LoF mutants exhibit late pupal lethality or pupal eclosion defects. Interestingly, although mtd encodes 26 transcripts, severe LoF and null mutations can be rescued by a single short human OXR1 cDNA that only contains the TLDc domain. Similar rescue is observed with the TLDc domain of NCOA7, another human homolog of mtd. Loss of mtd in neurons leads to massive cell loss, early death, and an accumulation of aberrant lysosomal structures, similar to what we observe in fibroblasts of affected individuals. Our data indicate that mtd and OXR1 are required for proper lysosomal function; this is consistent with observations that NCOA7 is required for lysosomal acidification.


Asunto(s)
Atrofia/patología , Enfermedades Cerebelosas/patología , Lisosomas/patología , Proteínas Mitocondriales/metabolismo , Enfermedades del Sistema Nervioso/patología , Estrés Oxidativo , Adolescente , Adulto , Animales , Atrofia/genética , Atrofia/metabolismo , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/metabolismo , Niño , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Lisosomas/metabolismo , Masculino , Proteínas Mitocondriales/genética , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Linaje , Fenotipo , Adulto Joven
9.
Glycobiology ; 28(11): 849-859, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30169771

RESUMEN

In multicellular organisms, glycosylation regulates various developmental signaling pathways including the Notch pathway. One of the O-linked glycans added to epidermal growth factor-like (EGF) repeats in animal proteins including the Notch receptors is the xylose-xylose-glucose-O oligosaccharide. Drosophila glucoside xylosyltransferase (Gxylt) Shams negatively regulates Notch signaling in specific contexts. Since Shams adds the first xylose residue to O-glucose, its loss-of-function phenotype could be due to the loss of the first xylose, the second xylose or both. To examine the contribution of the second xylose residues to Drosophila Notch signaling, we have performed biochemical and genetic analysis on CG11388, which is the Drosophila homolog of human xyloside xylosyltransferase 1 (XXYLT1). Experiments in S2 cells indicated that similar to human XXYLT1, CG11388 can add the second xylose to xylose-glucose-O glycans. Flies lacking both copies of CG11388 (Xxylt) are viable and fertile and do not show gross phenotypes indicative of altered Notch signaling. However, genetic interaction experiments show that in sensitized genetic backgrounds with decreased or increased Notch pathway components, loss of Xxylt promotes Delta-mediated activation of Notch. Unexpectedly, we find that in such sensitized backgrounds, even loss of one copy of the fly Gxylt shams enhances Delta-mediated Notch activation. Taken together, these data indicate that while the first xylose plays a key role in tuning the Delta-mediated Notch signaling in Drosophila, the second xylose has a fine-tuning role only revealed in sensitized genetic backgrounds.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Factor de Crecimiento Epidérmico/química , Antecedentes Genéticos , Pentosiltransferasa/química , Pentosiltransferasa/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Proteínas de Drosophila/genética , Humanos , Pentosiltransferasa/genética , Receptores Notch/genética , Transducción de Señal/genética , UDP Xilosa Proteína Xilosiltransferasa
10.
Elife ; 72018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30091705

RESUMEN

We generated two new genetic tools to efficiently tag genes in Drosophila. The first, Double Header (DH) utilizes intronic MiMIC/CRIMIC insertions to generate artificial exons for GFP mediated protein trapping or T2A-GAL4 gene trapping in vivo based on Cre recombinase to avoid embryo injections. DH significantly increases integration efficiency compared to previous strategies and faithfully reports the expression pattern of genes and proteins. The second technique targets genes lacking coding introns using a two-step cassette exchange. First, we replace the endogenous gene with an excisable compact dominant marker using CRISPR making a null allele. Second, the insertion is replaced with a protein::tag cassette. This sequential manipulation allows the generation of numerous tagged alleles or insertion of other DNA fragments that facilitates multiple downstream applications. Both techniques allow precise gene manipulation and facilitate detection of gene expression, protein localization and assessment of protein function, as well as numerous other applications.


Asunto(s)
Drosophila/genética , Entomología/métodos , Marcación de Gen/métodos , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Genes Reporteros , Integrasas/metabolismo , Mutagénesis Insercional
11.
Elife ; 72018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29565247

RESUMEN

We generated a library of ~1000 Drosophila stocks in which we inserted a construct in the intron of genes allowing expression of GAL4 under control of endogenous promoters while arresting transcription with a polyadenylation signal 3' of the GAL4. This allows numerous applications. First, ~90% of insertions in essential genes cause a severe loss-of-function phenotype, an effective way to mutagenize genes. Interestingly, 12/14 chromosomes engineered through CRISPR do not carry second-site lethal mutations. Second, 26/36 (70%) of lethal insertions tested are rescued with a single UAS-cDNA construct. Third, loss-of-function phenotypes associated with many GAL4 insertions can be reverted by excision with UAS-flippase. Fourth, GAL4 driven UAS-GFP/RFP reports tissue and cell-type specificity of gene expression with high sensitivity. We report the expression of hundreds of genes not previously reported. Finally, inserted cassettes can be replaced with GFP or any DNA. These stocks comprise a powerful resource for assessing gene function.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Biblioteca de Genes , Factores de Transcripción/genética , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Perfilación de la Expresión Génica , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mutagénesis Insercional , Especificidad de Órganos/genética , Factores de Transcripción/metabolismo
12.
PLoS Genet ; 13(7): e1006910, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28715417

RESUMEN

Hox transcription factors specify distinct cell types along the anterior-posterior axis of metazoans by regulating target genes that modulate signaling pathways. A well-established example is the induction of Epidermal Growth Factor (EGF) signaling by an Abdominal-A (Abd-A) Hox complex during the specification of Drosophila hepatocyte-like cells (oenocytes). Previous studies revealed that Abd-A is non-cell autonomously required to promote oenocyte fate by directly activating a gene (rhomboid) that triggers EGF secretion from sensory organ precursor (SOP) cells. Neighboring cells that receive the EGF signal initiate a largely unknown pathway to promote oenocyte fate. Here, we show that Abd-A also plays a cell autonomous role in inducing oenocyte fate by activating the expression of the Pointed-P1 (PntP1) ETS transcription factor downstream of EGF signaling. Genetic studies demonstrate that both PntP1 and PntP2 are required for oenocyte specification. Moreover, we found that PntP1 contains a conserved enhancer (PntP1OE) that is activated in oenocyte precursor cells by EGF signaling via direct regulation by the Pnt transcription factors as well as a transcription factor complex consisting of Abd-A, Extradenticle, and Homothorax. Our findings demonstrate that the same Abd-A Hox complex required for sending the EGF signal from SOP cells, enhances the competency of receiving cells to select oenocyte cell fate by up-regulating PntP1. Since PntP1 is a downstream effector of EGF signaling, these findings provide insight into how a Hox factor can both trigger and potentiate the EGF signal to promote an essential cell fate along the body plan.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Factor de Crecimiento Epidérmico/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Proteínas de Unión al ADN/genética , Drosophila/enzimología , Proteínas de Drosophila/genética , Elementos de Facilitación Genéticos , Factor de Crecimiento Epidérmico/genética , Regulación del Desarrollo de la Expresión Génica , Hepatocitos/citología , Hepatocitos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas/genética , Órganos de los Sentidos/crecimiento & desarrollo , Factores de Transcripción/genética
13.
Cell Host Microbe ; 21(6): 719-730.e6, 2017 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-28579255

RESUMEN

Clearance of bacteria by macrophages involves internalization of the microorganisms into phagosomes, which are then delivered to endolysosomes for enzymatic degradation. These spatiotemporally segregated processes are not known to be functionally coupled. Here, we show that lysosomal degradation of bacteria sustains phagocytic uptake. In Drosophila and mammalian macrophages, lysosomal dysfunction due to loss of the endolysosomal Cl- transporter ClC-b/CLCN7 delayed degradation of internalized bacteria. Unexpectedly, defective lysosomal degradation of bacteria also attenuated further phagocytosis, resulting in elevated bacterial load. Exogenous application of bacterial peptidoglycans restored phagocytic uptake in the lysosomal degradation-defective mutants via a pathway requiring cytosolic pattern recognition receptors and NF-κB. Mammalian macrophages that are unable to degrade internalized bacteria also exhibit compromised NF-κB activation. Our findings reveal a role for phagolysosomal degradation in activating an evolutionarily conserved signaling cascade, which ensures that continuous uptake of bacteria is preceded by lysosomal degradation of microbes.


Asunto(s)
Bacterias/inmunología , Inmunidad Innata/inmunología , Lisosomas/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Fagocitosis/fisiología , Animales , Citocinas/metabolismo , Drosophila/inmunología , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Mutación , FN-kappa B/metabolismo , Fagosomas/metabolismo , Células RAW 264.7 , Transducción de Señal/fisiología
14.
PLoS Genet ; 12(10): e1006327, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27764101

RESUMEN

We performed an exome-wide association analysis in 1393 late-onset Alzheimer's disease (LOAD) cases and 8141 controls from the CHARGE consortium. We found that a rare variant (P155L) in TM2D3 was enriched in Icelanders (~0.5% versus <0.05% in other European populations). In 433 LOAD cases and 3903 controls from the Icelandic AGES sub-study, P155L was associated with increased risk and earlier onset of LOAD [odds ratio (95% CI) = 7.5 (3.5-15.9), p = 6.6x10-9]. Mutation in the Drosophila TM2D3 homolog, almondex, causes a phenotype similar to loss of Notch/Presenilin signaling. Human TM2D3 is capable of rescuing these phenotypes, but this activity is abolished by P155L, establishing it as a functionally damaging allele. Our results establish a rare TM2D3 variant in association with LOAD susceptibility, and together with prior work suggests possible links to the ß-amyloid cascade.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas de Drosophila/genética , Proteínas de la Membrana/genética , Receptores Notch/genética , Tropomiosina/genética , Edad de Inicio , Anciano , Alelos , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Apolipoproteínas E/genética , Drosophila melanogaster/genética , Exoma/genética , Femenino , Estudio de Asociación del Genoma Completo , Genómica , Humanos , Islandia , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Mutación , Fenotipo , Población Blanca
15.
PLoS Biol ; 14(6): e1002472, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27254664

RESUMEN

Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) is neuroprotective in numerous preclinical models of neurodegeneration. Here, we show that brain nmnat2 mRNA levels correlate positively with global cognitive function and negatively with AD pathology. In AD brains, NMNAT2 mRNA and protein levels are reduced. NMNAT2 shifts its solubility and colocalizes with aggregated Tau in AD brains, similar to chaperones, which aid in the clearance or refolding of misfolded proteins. Investigating the mechanism of this observation, we discover a novel chaperone function of NMNAT2, independent from its enzymatic activity. NMNAT2 complexes with heat shock protein 90 (HSP90) to refold aggregated protein substrates. NMNAT2's refoldase activity requires a unique C-terminal ATP site, activated in the presence of HSP90. Furthermore, deleting NMNAT2 function increases the vulnerability of cortical neurons to proteotoxic stress and excitotoxicity. Interestingly, NMNAT2 acts as a chaperone to reduce proteotoxic stress, while its enzymatic activity protects neurons from excitotoxicity. Taken together, our data indicate that NMNAT2 exerts its chaperone or enzymatic function in a context-dependent manner to maintain neuronal health.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Western Blotting , Encéfalo/patología , Encéfalo/fisiopatología , Células COS , Células Cultivadas , Chlorocebus aethiops , Cognición/fisiología , Femenino , Proteínas HSP90 de Choque Térmico/genética , Humanos , Masculino , Ratones Transgénicos , Microscopía Fluorescente , Persona de Mediana Edad , Chaperonas Moleculares/genética , Mutación , Neuronas/citología , Neuronas/metabolismo , Nicotinamida-Nucleótido Adenililtransferasa/genética , Unión Proteica , Pliegue de Proteína , Estabilidad Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Trends Neurosci ; 36(11): 632-40, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23968695

RESUMEN

Proper brain function requires neuronal homeostasis over a range of environmental challenges. Neuronal activity, injury, and aging stress the nervous system, and lead to neuronal dysfunction and degeneration. Nevertheless, most organisms maintain healthy neurons throughout life, implying the existence of active maintenance mechanisms. Recent studies have revealed a key neuronal maintenance and protective function for nicotinamide mononucleotide adenylyl transferases (NMNATs). We review evidence that NMNATs protect neurons through multiple mechanisms in different contexts, and highlight functions that either require or are independent of NMNAT catalytic activity. We then summarize data supporting a role for NMNATs in neuronal maintenance and raise intriguing questions on how NMNATs preserve neuronal integrity and facilitate proper neural function throughout life.


Asunto(s)
Evolución Molecular , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Mononucleótido de Nicotinamida/metabolismo , Nucleotidiltransferasas/metabolismo , Animales , Biocatálisis , Humanos , Degeneración Nerviosa/patología , Neuronas/patología
17.
Development ; 139(9): 1611-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22438572

RESUMEN

Cis-regulatory modules (CRMs) ensure specific developmental outcomes by mediating both proper spatiotemporal gene expression patterns and appropriate transcriptional levels. In Drosophila, the precise transcriptional control of the serine protease rhomboid regulates EGF signaling to specify distinct cell types. Recently, we identified a CRM that activates rhomboid expression and thereby EGF secretion from a subset of abdominal sensory organ precursor cells (SOPs) to induce an appropriate number of lipid-processing cells called oenocytes. Here, we use scanning mutagenesis coupled with reporter assays, biochemistry and genetics to dissect the transcriptional mechanisms regulating SOP-specific rhomboid activation. Our results show that proper spatial activity of the rhomboid CRM is dependent upon direct integration of the abdomen-specific Hox factor Abdominal-A and the SOP-restricted Pax2 factor. In addition, we show that the Extradenticle and Homothorax Hox co-factors are differentially integrated on the rhomboid CRM by abdominal versus thoracic Hox proteins in the presence of Pax2. Last, we show that Abdominal-A uses both Pax2-dependent and Pax2-independent mechanisms to stimulate rhomboid CRM activity to induce proper oenocyte numbers. Thus, these data demonstrate how a CRM integrates Hox and neural transcriptional inputs to regulate the appropriate spatial pattern and levels of EGF secretion to specify an essential cell fate.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/embriología , Factor de Crecimiento Epidérmico/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal/fisiología , Animales , Animales Modificados Genéticamente , Ensayo de Cambio de Movilidad Electroforética , Inmunoprecipitación , Luciferasas , Mutagénesis , Células-Madre Neurales/metabolismo , Factor de Transcripción PAX2/metabolismo , Factores de Transcripción/metabolismo
18.
Neural Dev ; 6: 20, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21539742

RESUMEN

BACKGROUND: The concept of an equivalence group, a cluster of cells with equal potential to adopt the same specific fate, has served as a useful paradigm to understand neural cell type specification. In the Drosophila eye, a set of five cells, called the 'R7 equivalence group', generates a single photoreceptor neuron and four lens-secreting epithelial cells. This choice between neuronal versus non-neuronal cell fates rests on differential requirements for, and cross-talk between, Notch/Delta- and Ras/mitogen-activated protein kinase (MAPK)-dependent signaling pathways. However, many questions remain unanswered related to how downstream events of these two signaling pathways mediate distinct cell fate decisions. RESULTS: Here, we demonstrate that two direct downstream targets of Ras and Notch signaling, the transcription factors Prospero and dPax2, are essential regulators of neuronal versus non-neuronal cell fate decisions in the R7 equivalence group. Prospero controls high activated MAPK levels required for neuronal fate, whereas dPax2 represses Delta expression to prevent neuronal fate. Importantly, activity from both factors is required for proper cell fate decisions to occur. CONCLUSIONS: These data demonstrate that Ras and Notch signaling are integrated during cell fate decisions within the R7 equivalence group through the combinatorial and opposing activities of Pros and dPax2. Our study provides one of the first examples of how the differential expression and synergistic roles of two independent transcription factors determine cell fate within an equivalence group. Since the integration of Ras and Notch signaling is associated with many developmental and cancer models, these findings should provide new insights into how cell specificity is achieved by ubiquitously used signaling pathways in diverse biological contexts.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Proteínas Nucleares/fisiología , Factor de Transcripción PAX2/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/fisiología , Proteínas ras/metabolismo , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Drosophila , Proteínas de Drosophila/genética , Ojo/citología , Ojo/embriología , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas del Tejido Nervioso/genética , Neuronas/clasificación , Proteínas Nucleares/genética , Factor de Transcripción PAX2/genética , Células Fotorreceptoras , Pupa , Receptores Notch/genética , Retina/citología , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/fisiología , Factores de Transcripción/genética , Proteínas ras/genética
19.
Dev Biol ; 344(2): 1060-70, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20478292

RESUMEN

The atonal (ato) proneural gene specifies different numbers of sensory organ precursor (SOP) cells within distinct regions of the Drosophila embryo in an epidermal growth factor-dependent manner through the activation of the rhomboid (rho) protease. How ato activates rho, and why it does so in only a limited number of sensory cells remains unclear. We previously identified a rho enhancer (RhoBAD) that is active within a subset of abdominal SOP cells to induce larval oenocytes and showed that RhoBAD is regulated by an Abdominal-A (Abd-A) Hox complex and the Senseless (Sens) transcription factor. Here, we show that ato is also required for proper RhoBAD activity and oenocyte formation. Transgenic reporter assays reveal RhoBAD contains two conserved regions that drive SOP gene expression: RhoD mediates low levels of expression in both thoracic and abdominal SOP cells, whereas RhoA drives strong expression within abdominal SOP cells. Ato indirectly stimulates both elements and enhances RhoA reporter activity by interfering with the ability of the Sens repressor to bind DNA. As RhoA is also directly regulated by Abd-A, we propose a model for how the Ato and Sens proneural factors are integrated with an abdominal Hox factor to regulate region-specific SOP gene expression.


Asunto(s)
Drosophila/genética , Factores de Transcripción/metabolismo , Animales , Drosophila/citología , Drosophila/metabolismo , Embrión no Mamífero , Unión Proteica/genética , Órganos de los Sentidos/metabolismo , Factores de Transcripción/genética
20.
Dev Cell ; 15(2): 298-308, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18694568

RESUMEN

Hox factors are key regulators of distinct cells, tissues, and organs along the body plan. However, little is known about how Hox factors regulate cell-specific gene expression to pattern diverse tissues. Here, we show an unexpected Hox transcriptional mechanism: the permissive regulation of EGF secretion, and thereby cell specification, by antagonizing the Senseless transcription factor in the peripheral nervous system. rhomboid expression in a subset of sensory cells stimulates EGF secretion to induce hepatocyte-like cell development. We identified a rhomboid enhancer that is active in these cells and show that an abdominal Hox complex directly competes with Senseless for enhancer binding, with the transcriptional outcome dependent upon their relative binding activities. Thus, Hox-Senseless antagonism forms a molecular switch that integrates neural and anterior-posterior positional information. As the vertebrate senseless homolog is essential for neural development as well as hematopoiesis, we propose Hox-Senseless antagonism will broadly control cell fate decisions.


Asunto(s)
Proteínas de Drosophila/antagonistas & inhibidores , Drosophila melanogaster/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Sistema Nervioso Periférico/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Abdomen/embriología , Animales , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Unión Proteica , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/genética , Transcripción Genética , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...