Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.428
Filtrar
1.
J Agric Food Chem ; 72(19): 11221-11229, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38703356

RESUMEN

Liposcelis bostrychophila, commonly known as booklouse, is an important stored-product pest worldwide. Studies have demonstrated that booklices have developed resistance to several insecticides. In this study, an integument esterase gene, LbEST-inte4, with upregulated expression, was characterized in L. bostrychophila. Knockdown of LbEST-inte4 resulted in a substantial increase in the booklice susceptibility to malathion. Overexpression of LbEST-inte4 in Drosophila melanogaster significantly enhanced its malathion tolerance. Molecular modeling and docking analysis suggested potential interactions between LbEST-inte4 and malathion. When overexpressed LbEST-inte4 in Sf9 cells, a notable elevation in esterase activity and malathion tolerance was observed. HPLC analysis indicated that the LbEST-inte4 enzyme could effectively degrade malathion. Taken together, the upregulated LbEST-inte4 appears to contribute to malathion tolerance in L. bostrychophila by facilitating the depletion of malathion. This study elucidates the molecular mechanism underlying malathion detoxification and provides the foundations for the development of effective prevention and control measures against psocids.


Asunto(s)
Esterasas , Proteínas de Insectos , Insectos , Insecticidas , Malatión , Animales , Malatión/metabolismo , Malatión/química , Malatión/toxicidad , Malatión/farmacología , Insecticidas/metabolismo , Insecticidas/química , Insecticidas/farmacología , Esterasas/metabolismo , Esterasas/genética , Esterasas/química , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Proteínas de Insectos/química , Insectos/efectos de los fármacos , Resistencia a los Insecticidas/genética , Inactivación Metabólica , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/metabolismo
2.
J Biol Chem ; 300(1): 105491, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37995940

RESUMEN

l-2-hydroxyglutarate dehydrogenase (L2HGDH) is a mitochondrial membrane-associated metabolic enzyme, which catalyzes the oxidation of l-2-hydroxyglutarate (l-2-HG) to 2-oxoglutarate (2-OG). Mutations in human L2HGDH lead to abnormal accumulation of l-2-HG, which causes a neurometabolic disorder named l-2-hydroxyglutaric aciduria (l-2-HGA). Here, we report the crystal structures of Drosophila melanogaster L2HGDH (dmL2HGDH) in FAD-bound form and in complex with FAD and 2-OG and show that dmL2HGDH exhibits high activity and substrate specificity for l-2-HG. dmL2HGDH consists of an FAD-binding domain and a substrate-binding domain, and the active site is located at the interface of the two domains with 2-OG binding to the re-face of the isoalloxazine moiety of FAD. Mutagenesis and activity assay confirmed the functional roles of key residues involved in the substrate binding and catalytic reaction and showed that most of the mutations of dmL2HGDH equivalent to l-2-HGA-associated mutations of human L2HGDH led to complete loss of the activity. The structural and biochemical data together reveal the molecular basis for the substrate specificity and catalytic mechanism of L2HGDH and provide insights into the functional roles of human L2HGDH mutations in the pathogeneses of l-2-HGA.


Asunto(s)
Oxidorreductasas de Alcohol , Encefalopatías Metabólicas Innatas , Drosophila melanogaster , Modelos Moleculares , Animales , Humanos , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/metabolismo , Encefalopatías Metabólicas Innatas/enzimología , Encefalopatías Metabólicas Innatas/genética , Encefalopatías Metabólicas Innatas/fisiopatología , Drosophila melanogaster/enzimología , Glutaratos/metabolismo , Mutación , Dominio Catalítico/genética , Especificidad por Sustrato/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
Nature ; 623(7985): 122-131, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37722602

RESUMEN

A fundamental and unresolved question in regenerative biology is how tissues return to homeostasis after injury. Answering this question is essential for understanding the aetiology of chronic disorders such as inflammatory bowel diseases and cancer1. We used the Drosophila midgut2 to investigate this and discovered that during regeneration a subpopulation of cholinergic3 neurons triggers Ca2+ currents among intestinal epithelial cells, the enterocytes, to promote return to homeostasis. We found that downregulation of the conserved cholinergic enzyme acetylcholinesterase4 in the gut epithelium enables acetylcholine from specific Egr5 (TNF in mammals)-sensing cholinergic neurons to activate nicotinic receptors in innervated enterocytes. This activation triggers high Ca2+, which spreads in the epithelium through Innexin2-Innexin7 gap junctions6, promoting enterocyte maturation followed by reduction of proliferation and inflammation. Disrupting this process causes chronic injury consisting of ion imbalance, Yki (YAP in humans) activation7, cell death and increase of inflammatory cytokines reminiscent of inflammatory bowel diseases8. Altogether, the conserved cholinergic pathway facilitates epithelial Ca2+ currents that heal the intestinal epithelium. Our findings demonstrate nerve- and bioelectric9-dependent intestinal regeneration and advance our current understanding of how a tissue returns to homeostasis after injury.


Asunto(s)
Señalización del Calcio , Calcio , Neuronas Colinérgicas , Drosophila melanogaster , Enterocitos , Intestinos , Animales , Humanos , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Calcio/metabolismo , Neuronas Colinérgicas/metabolismo , Drosophila melanogaster/enzimología , Drosophila melanogaster/metabolismo , Enterocitos/metabolismo , Homeostasis , Inflamación/enzimología , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Intestinos/citología , Intestinos/metabolismo , Receptores Nicotínicos/metabolismo , Modelos Animales de Enfermedad
4.
J Biol Chem ; 299(8): 104961, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37380077

RESUMEN

Myosin-1D (myo1D) is important for Drosophila left-right asymmetry, and its effects are modulated by myosin-1C (myo1C). De novo expression of these myosins in nonchiral Drosophila tissues promotes cell and tissue chirality, with handedness depending on the paralog expressed. Remarkably, the identity of the motor domain determines the direction of organ chirality, rather than the regulatory or tail domains. Myo1D, but not myo1C, propels actin filaments in leftward circles in in vitro experiments, but it is not known if this property contributes to establishing cell and organ chirality. To further explore if there are differences in the mechanochemistry of these motors, we determined the ATPase mechanisms of myo1C and myo1D. We found that myo1D has a 12.5-fold higher actin-activated steady-state ATPase rate, and transient kinetic experiments revealed myo1D has an 8-fold higher MgADP release rate compared to myo1C. Actin-activated phosphate release is rate limiting for myo1C, whereas MgADP release is the rate-limiting step for myo1D. Notably, both myosins have among the tightest MgADP affinities measured for any myosin. Consistent with ATPase kinetics, myo1D propels actin filaments at higher speeds compared to myo1C in in vitro gliding assays. Finally, we tested the ability of both paralogs to transport 50 nm unilamellar vesicles along immobilized actin filaments and found robust transport by myo1D and actin binding but no transport by myo1C. Our findings support a model where myo1C is a slow transporter with long-lived actin attachments, whereas myo1D has kinetic properties associated with a transport motor.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Lateralidad Funcional , Miosina Tipo I , Animales , Actinas/metabolismo , Cinética , Miosina Tipo I/química , Miosina Tipo I/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Dominios Proteicos , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/enzimología
5.
J Cell Physiol ; 238(3): 647-658, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36745702

RESUMEN

Cardiomyopathy is a common disease of cardiac muscle that negatively affects cardiac function. HDAC3 commonly functions as corepressor by removing acetyl moieties from histone tails. However, a deacetylase-independent role of HDAC3 has also been described. Cardiac deletion of HDAC3 causes reduced cardiac contractility accompanied by lipid accumulation, but the molecular function of HDAC3 in cardiomyopathy remains unknown. We have used powerful genetic tools in Drosophila to investigate the enzymatic and nonenzymatic roles of HDAC3 in cardiomyopathy. Using the Drosophila heart model, we showed that cardiac-specific HDAC3 knockdown (KD) leads to prolonged systoles and reduced cardiac contractility. Immunohistochemistry revealed structural abnormalities characterized by myofiber disruption in HDAC3 KD hearts. Cardiac-specific HDAC3 KD showed increased levels of whole-body triglycerides and increased fibrosis. The introduction of deacetylase-dead HDAC3 mutant in HDAC3 KD background showed comparable results with wild-type HDAC3 in aspects of contractility and Pericardin deposition. However, deacetylase-dead HDAC3 mutants failed to improve triglyceride accumulation. Our data indicate that HDAC3 plays a deacetylase-independent role in maintaining cardiac contractility and preventing Pericardin deposition as well as a deacetylase-dependent role to maintain triglyceride homeostasis.


Asunto(s)
Cardiomiopatías , Modelos Animales de Enfermedad , Proteínas de Drosophila , Drosophila melanogaster , Histona Desacetilasas , Animales , Cardiomiopatías/enzimología , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Técnicas de Silenciamiento del Gen , Corazón/fisiología , Histona Desacetilasas/deficiencia , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/química , Histonas/metabolismo , Miocardio/metabolismo , Triglicéridos/metabolismo , Homeostasis
6.
Genetics ; 223(2)2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36482767

RESUMEN

Transvection, a type of trans-regulation of gene expression in which regulatory elements on one chromosome influence elements on a paired homologous chromosome, is itself a complex biological phenotype subject to modification by genetic background effects. However, relatively few studies have explored how transvection is affected by distal genetic variation, perhaps because it is strongly influenced by local regulatory elements and chromosomal architecture. With the emergence of the "hub" model of transvection and a series of studies showing variation in transvection effects, it is becoming clear that genetic background plays an important role in how transvection influences gene transcription. We explored the effects of genetic background on transvection by performing two independent genome wide association studies (GWASs) using the Drosophila genetic reference panel (DGRP) and a suite of Malic enzyme (Men) excision alleles. We found substantial variation in the amount of transvection in the 149 DGRP lines used, with broad-sense heritability of 0.89 and 0.84, depending on the excision allele used. The specific genetic variation identified was dependent on the excision allele used, highlighting the complex genetic interactions influencing transvection. We focussed primarily on genes identified as significant using a relaxed P-value cutoff in both GWASs. The most strongly associated genetic variant mapped to an intergenic single nucleotide polymorphism (SNP), located upstream of Tiggrin (Tig), a gene that codes for an extracellular matrix protein. Variants in other genes, such transcription factors (CG7368 and Sima), RNA binding proteins (CG10418, Rbp6, and Rig), enzymes (AdamTS-A, CG9743, and Pgant8), proteins influencing cell cycle progression (Dally and Eip63E) and signaling proteins (Atg-1, Axo, Egfr, and Path) also associated with transvection in Men. Although not intuitively obvious how many of these genes may influence transvection, some have been previously identified as promoting or antagonizing somatic homolog pairing. These results identify several candidate genes to further explore in the understanding of transvection in Men and in other genes regulated by transvection. Overall, these findings highlight the complexity of the interactions involved in gene regulation, even in phenotypes, such as transvection, that were traditionally considered to be primarily influenced by local genetic variation.


Asunto(s)
Estudio de Asociación del Genoma Completo , Malato Deshidrogenasa , Animales , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Malato Deshidrogenasa/metabolismo
7.
Elife ; 112022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35037620

RESUMEN

Neuronal health depends on quality control functions of autophagy, but mechanisms regulating neuronal autophagy are poorly understood. Previously, we showed that in Drosophila starvation-independent quality control autophagy is regulated by acinus (acn) and the Cdk5-dependent phosphorylation of its serine437 (Nandi et al., 2017). Here, we identify the phosphatase that counterbalances this activity and provides for the dynamic nature of acinus-serine437 (acn-S437) phosphorylation. A genetic screen identified six phosphatases that genetically interacted with an acn gain-of-function model. Among these, loss of function of only one, the PPM-type phosphatase Nil (CG6036), enhanced pS437-acn levels. Cdk5-dependent phosphorylation of acn-S437 in nil1 animals elevates neuronal autophagy and reduces the accumulation of polyQ proteins in a Drosophila Huntington's disease model. Consistent with previous findings that Cd2+ inhibits PPM-type phosphatases, Cd2+ exposure elevated acn-S437 phosphorylation which was necessary for increased neuronal autophagy and protection against Cd2+-induced cytotoxicity. Together, our data establish the acn-S437 phosphoswitch as critical integrator of multiple stress signals regulating neuronal autophagy.


Asunto(s)
Autofagia/genética , Cadmio/metabolismo , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/fisiología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Serina/genética , Estrés Fisiológico/efectos de los fármacos , Animales , Autofagia/efectos de los fármacos , Autofagia/fisiología , Cadmio/toxicidad , Intoxicación por Cadmio , Drosophila melanogaster/enzimología , Femenino , Masculino , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Serina/metabolismo
8.
Pigment Cell Melanoma Res ; 35(1): 26-37, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34388859

RESUMEN

Melanin is a widely distributed phenolic pigment that is biosynthesized from tyrosine and its hydroxylated product, dopa, in all animals. However, recent studies reveal a significant deviation from this paradigm, as insects appear to use dopamine rather than dopa as the major precursor of melanin. This observation calls for a reconsideration of the insect melanogenic pathway. While phenoloxidases and laccases can oxidize dopamine for dopaminechrome production, the fate of dopaminechrome remains undetermined. Dopachrome decarboxylase/tautomerase, encoded by yellow-f/f2 of Drosophila melanogaster, can convert dopaminechrome into 5,6-dihydroxyindole, but the same enzyme from other organisms does not act on dopaminechrome, suggesting the existence of a specific dopaminechrome tautomerase (DPT). We now report the identification of this novel enzyme that biosynthesizes 5,6-dihydroxyindole from dopaminechrome in Drosophila. Dopaminechrome tautomerase acted on both dopaminechrome and N-methyl dopaminechrome but not on dopachrome or other aminochromes tested. Our biochemical and molecular studies reveal that this enzyme is encoded by the yellow-h gene, a member of the yellow gene family, and advance our understanding of the physiological functions of this gene family. Identification and characterization of DPT clarifies the precursor for melanin biosynthetic pathways and proves the existence of an independent melanogenic pathway in insects that utilizes dopamine as the primary precursor.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Oxidorreductasas Intramoleculares , Melaninas , Animales , Animales Modificados Genéticamente , Línea Celular , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Indoles/metabolismo , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Melaninas/biosíntesis , Mutación
9.
Nucleic Acids Res ; 49(22): 13108-13121, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34878141

RESUMEN

Mutations in genes encoding mitochondrial aminoacyl-tRNA synthetases are linked to diverse diseases. However, the precise mechanisms by which these mutations affect mitochondrial function and disease development are not fully understood. Here, we develop a Drosophila model to study the function of dFARS2, the Drosophila homologue of the mitochondrial phenylalanyl-tRNA synthetase, and further characterize human disease-associated FARS2 variants. Inactivation of dFARS2 in Drosophila leads to developmental delay and seizure. Biochemical studies reveal that dFARS2 is required for mitochondrial tRNA aminoacylation, mitochondrial protein stability, and assembly and enzyme activities of OXPHOS complexes. Interestingly, by modeling FARS2 mutations associated with human disease in Drosophila, we provide evidence that expression of two human FARS2 variants, p.G309S and p.D142Y, induces seizure behaviors and locomotion defects, respectively. Together, our results not only show the relationship between dysfunction of mitochondrial aminoacylation system and pathologies, but also illustrate the application of Drosophila model for functional analysis of human disease-causing variants.


Asunto(s)
Discapacidades del Desarrollo/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Mitocondriales/genética , Mutación , Fenilalanina-ARNt Ligasa/genética , ARN de Transferencia/genética , Convulsiones/genética , Animales , Línea Celular , Discapacidades del Desarrollo/enzimología , Modelos Animales de Enfermedad , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/enzimología , Técnicas de Silenciamiento del Gen , Humanos , Microscopía Electrónica de Transmisión , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/deficiencia , Fosforilación Oxidativa , Fenilalanina-ARNt Ligasa/deficiencia , ARN de Transferencia/metabolismo , Convulsiones/enzimología , Aminoacilación de ARN de Transferencia
10.
Development ; 148(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34918741

RESUMEN

Genetic and genomic analysis in Drosophila suggests that hematopoietic progenitors likely transition into terminal fates via intermediate progenitors (IPs) with some characteristics of either, but perhaps maintaining IP-specific markers. In the past, IPs have not been directly visualized and investigated owing to lack of appropriate genetic tools. Here, we report a Split GAL4 construct, CHIZ-GAL4, that identifies IPs as cells physically juxtaposed between true progenitors and differentiating hemocytes. IPs are a distinct cell type with a unique cell-cycle profile and they remain multipotent for all blood cell fates. In addition, through their dynamic control of the Notch ligand Serrate, IPs specify the fate of direct neighbors. The Ras pathway controls the number of IP cells and promotes their transition into differentiating cells. This study suggests that it would be useful to characterize such intermediate populations of cells in mammalian hematopoietic systems.


Asunto(s)
Proteínas de Drosophila/genética , Hematopoyesis/genética , Proteína Jagged-1/genética , Receptores Notch/genética , Factores de Transcripción/genética , Animales , Células Sanguíneas/citología , Células Sanguíneas/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hemocitos , Lectinas/genética , Receptores de Interleucina/genética , Transducción de Señal/genética , Células Madre/citología , Células Madre/metabolismo
11.
Elife ; 102021 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-34605405

RESUMEN

PERK is an endoplasmic reticulum (ER) transmembrane sensor that phosphorylates eIF2α to initiate the Unfolded Protein Response (UPR). eIF2α phosphorylation promotes stress-responsive gene expression most notably through the transcription factor ATF4 that contains a regulatory 5' leader. Possible PERK effectors other than ATF4 remain poorly understood. Here, we report that the bZIP transcription factor Xrp1 is required for ATF4-independent PERK signaling. Cell-type-specific gene expression profiling in Drosophila indicated that delta-family glutathione-S-transferases (gstD) are prominently induced by the UPR-activating transgene Rh1G69D. Perk was necessary and sufficient for such gstD induction, but ATF4 was not required. Instead, Perk and other regulators of eIF2α phosphorylation regulated Xrp1 protein levels to induce gstDs. The Xrp1 5' leader has a conserved upstream Open Reading Frame (uORF) analogous to those that regulate ATF4 translation. The gstD-GFP reporter induction required putative Xrp1 binding sites. These results indicate that antioxidant genes are highly induced by a previously unrecognized UPR signaling axis consisting of PERK and Xrp1.


Asunto(s)
Antioxidantes/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Glutatión Transferasa/metabolismo , Discos Imaginales/enzimología , eIF-2 Quinasa/metabolismo , Animales , Animales Modificados Genéticamente , Sitios de Unión , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Estrés del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glutatión Transferasa/genética , Discos Imaginales/embriología , Sistemas de Lectura Abierta , Fosforilación , Rodopsina/genética , Rodopsina/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada , eIF-2 Quinasa/genética
12.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638976

RESUMEN

Lysosomal degradation, the common destination of autophagy and endocytosis, is one of the most important elements of eukaryotic metabolism. The small GTPases Rab39A and B are potential new effectors of this pathway, as their malfunction is implicated in severe human diseases like cancer and neurodegeneration. In this study, the lysosomal regulatory role of the single Drosophila Rab39 ortholog was characterized, providing valuable insight into the potential cell biological mechanisms mediated by these proteins. Using a de novo CRISPR-generated rab39 mutant, we found no failure in the early steps of endocytosis and autophagy. On the contrary, we found that Rab39 mutant nephrocytes internalize and degrade endocytic cargo at a higher rate compared to control cells. In addition, Rab39 mutant fat body cells contain small yet functional autolysosomes without lysosomal fusion defect. Our data identify Drosophila Rab39 as a negative regulator of lysosomal clearance during both endocytosis and autophagy.


Asunto(s)
Autofagia/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Endocitosis/genética , Lisosomas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas , Proteínas de Drosophila/genética , Larva/enzimología , Larva/genética , Fenotipo , Proteínas de Unión al GTP rab/genética
13.
Cell Rep ; 37(3): 109834, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686333

RESUMEN

WNTs play key roles in development and disease, signaling through Frizzled (FZD) seven-pass transmembrane receptors and numerous co-receptors including ROR and RYK family receptor tyrosine kinases (RTKs). We describe crystal structures and WNT-binding characteristics of extracellular regions from the Drosophila ROR and RYK orthologs Nrk (neurospecific receptor tyrosine kinase) and Derailed-2 (Drl-2), which bind WNTs though a FZD-related cysteine-rich domain (CRD) and WNT-inhibitory factor (WIF) domain respectively. Our crystal structures suggest that neither Nrk nor Drl-2 can accommodate the acyl chain typically attached to WNTs. The Nrk CRD contains a deeply buried bound fatty acid, unlikely to be exchangeable. The Drl-2 WIF domain lacks the lipid-binding site seen in WIF-1. We also find that recombinant DWnt-5 can bind Drosophila ROR and RYK orthologs despite lacking an acyl chain. Alongside analyses of WNT/receptor interaction sites, our structures provide further insight into how WNTs may recruit RTK co-receptors into signaling complexes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Modelos Moleculares , Proteínas del Tejido Nervioso/genética , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Células Sf9 , Relación Estructura-Actividad , Proteínas Wnt/genética
14.
Cell Rep ; 37(3): 109874, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686334

RESUMEN

Embryos repair wounds rapidly, with no inflammation or scarring, in a process that involves polarization of the actomyosin cytoskeleton. Actomyosin polarization results in the assembly of a contractile cable around the wound that drives wound closure. Here, we demonstrate that a contractile actomyosin cable is not sufficient for rapid wound repair in Drosophila embryos. We show that wounding causes activation of the serine/threonine kinase p38 mitogen-activated protein kinase (MAPK) in the cells adjacent to the wound. p38 activation reduces the levels of wound-induced reactive oxygen species in the cells around the wound, limiting wound size. In addition, p38 promotes an increase in volume in the cells around the wound, thus facilitating the collective cell movements that drive rapid wound healing. Our data indicate that p38 regulates cell volumes through the sodium-potassium-chloride cotransporter NKCC1. Our work reveals cell growth and cell survival as cell behaviors critical for embryonic wound repair.


Asunto(s)
Proliferación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Cicatrización de Heridas , Heridas y Lesiones/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Animales Modificados Genéticamente , Tamaño de la Célula , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Miosina Tipo II/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Factores de Tiempo , Heridas y Lesiones/genética , Heridas y Lesiones/patología , Proteínas Quinasas p38 Activadas por Mitógenos/genética
15.
Elife ; 102021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34515029

RESUMEN

The preinitiation complex (PIC) for transcriptional initiation by RNA polymerase (Pol) II is composed of general transcription factors that are highly conserved. However, analysis of ChIP-seq datasets reveals kinetic and compositional differences in the transcriptional initiation process among eukaryotic species. In yeast, Mediator associates strongly with activator proteins bound to enhancers, but it transiently associates with promoters in a form that lacks the kinase module. In contrast, in human, mouse, and fly cells, Mediator with its kinase module stably associates with promoters, but not with activator-binding sites. This suggests that yeast and metazoans differ in the nature of the dynamic bridge of Mediator between activators and Pol II and the composition of a stable inactive PIC-like entity. As in yeast, occupancies of TATA-binding protein (TBP) and TBP-associated factors (Tafs) at mammalian promoters are not strictly correlated. This suggests that within PICs, TFIID is not a monolithic entity, and multiple forms of TBP affect initiation at different classes of genes. TFIID in flies, but not yeast and mammals, interacts strongly at regions downstream of the initiation site, consistent with the importance of downstream promoter elements in that species. Lastly, Taf7 and the mammalian-specific Med26 subunit of Mediator also interact near the Pol II pause region downstream of the PIC, but only in subsets of genes and often not together. Species-specific differences in PIC structure and function are likely to affect how activators and repressors affect transcriptional activity.


Asunto(s)
Complejo Mediador/metabolismo , ARN Polimerasa II/metabolismo , Factores Generales de Transcripción/metabolismo , Iniciación de la Transcripción Genética , Animales , Línea Celular , Bases de Datos Genéticas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Regulación Fúngica de la Expresión Génica , Humanos , Complejo Mediador/química , Complejo Mediador/genética , Ratones , Regiones Promotoras Genéticas , Conformación Proteica , ARN Polimerasa II/química , ARN Polimerasa II/genética , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidad de la Especie , Relación Estructura-Actividad , Factores Asociados con la Proteína de Unión a TATA/genética , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Factores Generales de Transcripción/química , Factores Generales de Transcripción/genética , Sitio de Iniciación de la Transcripción
16.
Int J Mol Sci ; 22(17)2021 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-34502551

RESUMEN

The evolutionarily conserved c-Jun N-terminal kinase (JNK) signaling pathway is a critical genetic determinant in the control of longevity. In response to extrinsic and intrinsic stresses, JNK signaling is activated to protect cells from stress damage and promote survival. In Drosophila, global JNK upregulation can delay aging and extend lifespan, whereas tissue/organ-specific manipulation of JNK signaling impacts lifespan in a context-dependent manner. In this review, focusing on several tissues/organs that are highly associated with age-related diseases-including metabolic organs (intestine and fat body), neurons, and muscles-we summarize the distinct effects of tissue/organ-specific JNK signaling on aging and lifespan. We also highlight recent progress in elucidating the molecular mechanisms underlying the tissue-specific effects of JNK activity. Together, these studies highlight an important and comprehensive role for JNK signaling in the regulation of longevity in Drosophila.


Asunto(s)
Envejecimiento/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Longevidad/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Animales , Encéfalo/citología , Encéfalo/enzimología , Drosophila melanogaster/enzimología , Cuerpo Adiposo/enzimología , Modelos Biológicos , Neuronas/enzimología
17.
Chem Commun (Camb) ; 57(83): 10879-10882, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34590626

RESUMEN

Dicer-2 cleaves double-stranded RNA into siRNAs in a terminus-dependent manner as part of D. melanogaster's RNA interference pathway. Using ultrafast fluorescence, we probe the local environment of chromophores at the dsRNA terminus upon binding by Dicer-2 and interrogate the effects of Loquacious-PD, an accessory protein. We find substrate-selective modes of molecular recognition that distinguish between blunt and 3'overhang termini, but whose differences are greatly reduced by Loquacious-PD. These results connect the molecular recognition properties of Dicer-2 to its selective processing of dsRNAs with different termini and to its need for Loquacious-PD to efficiently produce endogenous siRNAs.


Asunto(s)
Proteínas de Drosophila/metabolismo , ARN Helicasas/metabolismo , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/metabolismo , Animales , Carbocianinas/química , Drosophila melanogaster/enzimología , Colorantes Fluorescentes/química , ARN Bicatenario/química
18.
Cell Rep ; 36(9): 109644, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34469735

RESUMEN

In holometabolous insects, metamorphic timing and body size are controlled by a neuroendocrine axis composed of the ecdysone-producing prothoracic gland (PG) and its presynaptic neurons (PGNs) producing PTTH. Although PTTH/Torso signaling is considered the primary mediator of metamorphic timing, recent studies indicate that other unidentified PGN-derived factors also affect timing. Here, we demonstrate that the receptor tyrosine kinases anaplastic lymphoma kinase (Alk) and PDGF and VEGF receptor-related (Pvr), function in coordination with PTTH/Torso signaling to regulate pupariation timing and body size. Both Alk and Pvr trigger Ras/Erk signaling in the PG to upregulate expression of ecdysone biosynthetic enzymes, while Alk also suppresses autophagy by activating phosphatidylinositol 3-kinase (PI3K)/Akt. The Alk ligand Jelly belly (Jeb) is produced by the PGNs and serves as a second PGN-derived tropic factor, while Pvr activation mainly relies on autocrine signaling by PG-derived Pvf2 and Pvf3. These findings illustrate that a combination of juxtacrine and autocrine signaling regulates metamorphic timing, the defining event of holometabolous development.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Glándulas Endocrinas/enzimología , Metamorfosis Biológica , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Animales Modificados Genéticamente , Comunicación Autocrina , Tamaño Corporal , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Ecdisona/metabolismo , Glándulas Endocrinas/embriología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , IMP Deshidrogenasa/genética , IMP Deshidrogenasa/metabolismo , Quinasas Janus/genética , Quinasas Janus/metabolismo , Mutación , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Péptidos de Invertebrados/genética , Receptores de Péptidos de Invertebrados/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Crecimiento Endotelial Vascular/genética , Factores de Crecimiento Endotelial Vascular/metabolismo
19.
Viruses ; 13(8)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34452330

RESUMEN

Eukaryotic nucleic acid methyltransferase (MTase) proteins are essential mediators of epigenetic and epitranscriptomic regulation. DNMT2 belongs to a large, conserved family of DNA MTases found in many organisms, including holometabolous insects such as fruit flies and mosquitoes, where it is the lone MTase. Interestingly, despite its nomenclature, DNMT2 is not a DNA MTase, but instead targets and methylates RNA species. A growing body of literature suggests that DNMT2 mediates the host immune response against a wide range of pathogens, including RNA viruses. Curiously, although DNMT2 is antiviral in Drosophila, its expression promotes virus replication in mosquito species. We, therefore, sought to understand the divergent regulation, function, and evolution of these orthologs. We describe the role of the Drosophila-specific host protein IPOD in regulating the expression and function of fruit fly DNMT2. Heterologous expression of these orthologs suggests that DNMT2's role as an antiviral is host-dependent, indicating a requirement for additional host-specific factors. Finally, we identify and describe potential evidence of positive selection at different times throughout DNMT2 evolution within dipteran insects. We identify specific codons within each ortholog that are under positive selection and find that they are restricted to four distinct protein domains, which likely influence substrate binding, target recognition, and adaptation of unique intermolecular interactions. Collectively, our findings highlight the evolution of DNMT2 in Dipteran insects and point to structural, regulatory, and functional differences between mosquito and fruit fly homologs.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Dípteros/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimología , Drosophila melanogaster/microbiología , Interacciones Huésped-Patógeno , Wolbachia/fisiología , Adaptación Biológica , Aedes/enzimología , Aedes/genética , Aedes/inmunología , Aedes/microbiología , Secuencia de Aminoácidos , Animales , ADN (Citosina-5-)-Metiltransferasas/química , ADN (Citosina-5-)-Metiltransferasas/inmunología , Dípteros/clasificación , Dípteros/enzimología , Dípteros/inmunología , Proteínas de Drosophila/química , Proteínas de Drosophila/inmunología , Drosophila melanogaster/genética , Drosophila melanogaster/inmunología , Evolución Molecular , Filogenia , Conformación Proteica , Alineación de Secuencia , Wolbachia/genética
20.
J Immunol ; 207(6): 1616-1626, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34452932

RESUMEN

The evolutionarily conserved immune deficiency (IMD) signaling pathway shields Drosophila against bacterial infections. It regulates the expression of antimicrobial peptides encoding genes through the activation of the NF-κB transcription factor Relish. Tight regulation of the signaling cascade ensures a balanced immune response, which is otherwise highly harmful. Several phosphorylation events mediate intracellular progression of the IMD pathway. However, signal termination by dephosphorylation remains largely elusive. Here, we identify the highly conserved protein phosphatase 4 (PP4) complex as a bona fide negative regulator of the IMD pathway. RNA interference-mediated gene silencing of PP4-19c, PP4R2, and Falafel, which encode the catalytic and regulatory subunits of the phosphatase complex, respectively, caused a marked upregulation of bacterial-induced antimicrobial peptide gene expression in both Drosophila melanogaster S2 cells and adult flies. Deregulated IMD signaling is associated with reduced lifespan of PP4-deficient flies in the absence of any infection. In contrast, flies overexpressing this phosphatase are highly sensitive to bacterial infections. Altogether, our results highlight an evolutionarily conserved function of PP4c in the regulation of NF-κB signaling from Drosophila to mammals.


Asunto(s)
Proteínas de Drosophila/deficiencia , Drosophila melanogaster/enzimología , Drosophila melanogaster/inmunología , Inmunidad Innata , FN-kappa B/metabolismo , Fosfoproteínas Fosfatasas/deficiencia , Transducción de Señal/inmunología , Animales , Péptidos Catiónicos Antimicrobianos/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Expresión Génica , Silenciador del Gen , Longevidad/genética , Longevidad/inmunología , Fosfoproteínas Fosfatasas/genética , Interferencia de ARN , Transducción de Señal/genética , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...