Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 781
Filter
1.
Int J Mol Sci ; 22(22)2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34830088

ABSTRACT

For developmental processes, we know most of the gene networks controlling specific cell responses. We still have to determine how these networks cooperate and how signals become integrated. The JNK pathway is one of the key elements modulating cellular responses during development. Yet, we still know little about how the core components of the pathway interact with additional regulators or how this network modulates cellular responses in the whole organism in homeostasis or during tissue morphogenesis. We have performed a promoter analysis, searching for potential regulatory sequences of puckered (puc) and identified different specific enhancers directing gene expression in different tissues and at different developmental times. Remarkably, some of these domains respond to the JNK activity, but not all. Altogether, these analyses show that puc expression regulation is very complex and that JNK activities participate in non-previously known processes during the development of Drosophila.


Subject(s)
Drosophila Proteins , Gene Expression Regulation, Enzymologic , Morphogenesis/genetics , Phosphoprotein Phosphatases , Response Elements , Signal Transduction/genetics , Animals , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/genetics
2.
Elife ; 102021 10 12.
Article in English | MEDLINE | ID: mdl-34636720

ABSTRACT

Associative learning allows animals to use past experience to predict future events. The circuits underlying memory formation support immediate and sustained changes in function, often in response to a single example. Larval Drosophila is a genetic model for memory formation that can be accessed at molecular, synaptic, cellular, and circuit levels, often simultaneously, but existing behavioral assays for larval learning and memory do not address individual animals, and it has been difficult to form long-lasting memories, especially those requiring synaptic reorganization. We demonstrate a new assay for learning and memory capable of tracking the changing preferences of individual larvae. We use this assay to explore how activation of a pair of reward neurons changes the response to the innately aversive gas carbon dioxide (CO2). We confirm that when coupled to CO2 presentation in appropriate temporal sequence, optogenetic reward reduces avoidance of CO2. We find that learning is switch-like: all-or-none and quantized in two states. Memories can be extinguished by repeated unrewarded exposure to CO2 but are stabilized against extinction by repeated training or overnight consolidation. Finally, we demonstrate long-lasting protein synthesis dependent and independent memory formation.


Brains learn from experience. They take events from the past, link them together, and use them to predict the future. This is true for fruit flies, Drosophila melanogaster, as well as for humans. One of the main questions in the field of neuroscience is, how does this kind of associative learning happen? Fruit fly larvae can learn to associate a certain smell with a sugar reward. When a group of larvae learn to associate a smell with sugar, most but not all of them will approach that smell in the future. This shows associative learning in action, but it raises a big question. Did the larvae that failed to approach the smell fail to learn, or did they just happen to make a mistake finding the smell? Given another chance, would exactly the same larvae approach the smell as the first time? In other words, did all the larvae learn a little, or did some larvae learn completely and others learn nothing? To find out, Lesar et al. built a computer-controlled maze to test whether individual fruit fly larvae liked or avoided a smell. Whenever a larva reached the middle of the Y-shaped maze, it could choose to go down one of two remaining corridors. One corridor contained air and the other carbon dioxide, a gas they would naturally avoid. Lesar et al. taught each larva to like carbon dioxide by activating reward neurons in its brain while filling the maze with carbon dioxide gas. Studying each larva as it navigated the maze revealed that they learn in a single jump, a 'lightbulb moment'. When Lesar et al. activated the reward neurons, the larva either 'got it' and stopped avoiding carbon dioxide altogether, or it did not. In the second case, it behaved as if it had received no training at all. Classic and modern experiments on people suggest that humans might also learn in jumps, but research on our own brains is challenging. Fruit flies are an excellent model organism to study memory formation because they are easy to breed, and it is easy to manipulate their genetic code. Work in flies has already revealed many of the genes and cells responsible for learning and memory. But, to find the specific brain changes that explain learning, researchers need to know whether the animals they are examining have actually learned something. This new maze could help researchers to identify those individuals, making it easier to find out exactly how associative learning works.


Subject(s)
Association Learning , Avoidance Learning , Behavior, Animal , Drosophila melanogaster/physiology , Memory , Animals , Animals, Genetically Modified , Carbon Dioxide , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Extinction, Psychological , Larva/genetics , Larva/metabolism , Larva/physiology , Odorants , Olfactory Perception , Optogenetics , Reward , Smell , Time Factors
3.
Cell Rep ; 37(4): 109896, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706247

ABSTRACT

Highly specific expression patterns can be caused by the overlapping activities of activator and repressor sequences in enhancers. However, few studies illuminate how these sequences evolve in the origin of new enhancers. Here, we show that expression of the bond gene in the semicircular wall epithelium (swe) of the Drosophila melanogaster male ejaculatory bulb (EB) is controlled by an enhancer consisting of an activator region that requires Abdominal-B driving expression in the entire EB and a repressor region that restricts this expression to the EB swe. Although this expression pattern is independently gained in the distantly related Scaptodrosophila lebanonensis and does not require Abdominal-B, we show that functionally similar repressor sequences are present in Scaptodrosophila and also in species that do not express bond in the EB. We suggest that during enhancer evolution, repressor sequences can precede the evolution of activator sequences and may lead to similar but independently evolved expression patterns.


Subject(s)
Acetyltransferases , Drosophila Proteins , Enhancer Elements, Genetic , Evolution, Molecular , Gene Expression Regulation , Acetyltransferases/biosynthesis , Acetyltransferases/genetics , Animals , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , Male , Species Specificity
4.
J Neurosci ; 41(40): 8338-8350, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34429376

ABSTRACT

Rhythmic rest-activity cycles are controlled by an endogenous clock. In Drosophila, this clock resides in ∼150 neurons organized in clusters whose hierarchy changes in response to environmental conditions. The concerted activity of the circadian network is necessary for the adaptive responses to synchronizing environmental stimuli. Thus far, work was devoted to unravel the logic of the coordination of different clusters focusing on neurotransmitters and neuropeptides. We further explored communication in the adult male brain through ligands belonging to the bone morphogenetic protein (BMP) pathway. Herein we show that the lateral ventral neurons (LNvs) express the small morphogen decapentaplegic (DPP). DPP expression in the large LNvs triggered a period lengthening phenotype, the downregulation of which caused reduced rhythmicity and affected anticipation at dawn and dusk, underscoring DPP per se conveys time-of-day relevant information. Surprisingly, DPP expression in the large LNvs impaired circadian remodeling of the small LNv axonal terminals, likely through local modulation of the guanine nucleotide exchange factor Trio. These findings open the provocative possibility that the BMP pathway is recruited to strengthen/reduce the connectivity among specific clusters along the day and thus modulate the contribution of the clusters to the circadian network.SIGNIFICANCE STATEMENT The circadian clock relies on the communication between groups of so-called clock neurons to coordinate physiology and behavior to the optimal times across the day, predicting and adapting to a changing environment. The circadian network relies on neurotransmitters and neuropeptides to fine-tune connectivity among clock neurons and thus give rise to a coherent output. Herein we show that decapentaplegic, a ligand belonging to the BMP retrograde signaling pathway required for coordinated growth during development, is recruited by a group of circadian neurons in the adult brain to trigger structural remodeling of terminals on a daily basis.


Subject(s)
Central Pattern Generators/physiology , Circadian Rhythm/physiology , Drosophila Proteins/biosynthesis , Nerve Net/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila melanogaster , Male
5.
Neuron ; 109(18): 2864-2883.e8, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34384519

ABSTRACT

The molecular and cellular mechanisms underlying complex axon morphogenesis are still poorly understood. We report a novel, evolutionary conserved function for the Drosophila Wnk kinase (dWnk) and its mammalian orthologs, WNK1 and 2, in axon branching. We uncover that dWnk, together with the neuroprotective factor Nmnat, antagonizes the axon-destabilizing factors D-Sarm and Axundead (Axed) during axon branch growth, revealing a developmental function for these proteins. Overexpression of D-Sarm or Axed results in axon branching defects, which can be blocked by overexpression of dWnk or Nmnat. Surprisingly, Wnk kinases are also required for axon maintenance of adult Drosophila and mouse cortical pyramidal neurons. Requirement of Wnk for axon maintenance is independent of its developmental function. Inactivation of dWnk or mouse Wnk1/2 in mature neurons leads to axon degeneration in the adult brain. Therefore, Wnk kinases are novel signaling components that provide a safeguard function in both developing and adult axons.


Subject(s)
Armadillo Domain Proteins/biosynthesis , Axons/metabolism , Cytoskeletal Proteins/biosynthesis , Drosophila Proteins/biosynthesis , Evolution, Molecular , Morphogenesis/physiology , Protein Serine-Threonine Kinases/biosynthesis , Animals , Armadillo Domain Proteins/antagonists & inhibitors , Armadillo Domain Proteins/genetics , Cell Line, Tumor , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster , Female , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Pregnancy , Protein Serine-Threonine Kinases/genetics
6.
J Neurogenet ; 35(3): 192-212, 2021 09.
Article in English | MEDLINE | ID: mdl-34382904

ABSTRACT

The foraging gene in Drosophila melanogaster, which encodes a cGMP-dependent protein kinase, is a highly conserved, complex gene with multiple pleiotropic behavioral and physiological functions in both the larval and adult fly. Adult foraging expression is less well characterized than in the larva. We characterized foraging expression in the brain, gastric system, and reproductive systems using a T2A-Gal4 gene-trap allele. In the brain, foraging expression appears to be restricted to multiple sub-types of glia. This glial-specific cellular localization of foraging was supported by single-cell transcriptomic atlases of the adult brain. foraging is extensively expressed in most cell types in the gastric and reproductive systems. We then mapped multiple cis-regulatory elements responsible for parts of the observed expression patterns by a nested cloned promoter-Gal4 analysis. The mapped cis-regulatory elements were consistently modular when comparing the larval and adult expression patterns. These new data using the T2A-Gal4 gene-trap and cloned foraging promoter fusion GAL4's are discussed with respect to previous work using an anti-FOR antibody, which we show here to be non-specific. Future studies of foraging's function will consider roles for glial subtypes and peripheral tissues (gastric and reproductive systems) in foraging's pleiotropic behavioral and physiological effects.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/biosynthesis , Drosophila Proteins/biosynthesis , Drosophila melanogaster/physiology , Transcriptome , Animals , Brain/metabolism , Genitalia/metabolism , Stomach/metabolism
7.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34074746

ABSTRACT

The construction and maturation of the postsynaptic apparatus are crucial for synapse and dendrite development. The fundamental mechanisms underlying these processes are most often studied in glutamatergic central synapses in vertebrates. Whether the same principles apply to excitatory cholinergic synapses, such as those found in the insect central nervous system, is not known. To address this question, we investigated a group of projection neurons in the Drosophila larval visual system, the ventral lateral neurons (LNvs), and identified nAchRα1 (Dα1) and nAchRα6 (Dα6) as the main functional nicotinic acetylcholine receptor (nAchR) subunits in the larval LNvs. Using morphological analyses and calcium imaging studies, we demonstrated critical roles of these two subunits in supporting dendrite morphogenesis and synaptic transmission. Furthermore, our RNA sequencing analyses and endogenous tagging approach identified distinct transcriptional controls over the two subunits in the LNvs, which led to the up-regulation of Dα1 and down-regulation of Dα6 during larval development as well as to an activity-dependent suppression of Dα1 Additional functional analyses of synapse formation and dendrite dynamics further revealed a close association between the temporal regulation of individual nAchR subunits and their sequential requirements during the cholinergic synapse maturation. Together, our findings support transcriptional control of nAchR subunits as a core element of developmental and activity-dependent regulation of central cholinergic synapses.


Subject(s)
Cholinergic Neurons/metabolism , Dendrites/metabolism , Drosophila Proteins/biosynthesis , Morphogenesis , Receptors, Nicotinic/biosynthesis , Synapses/metabolism , Synaptic Transmission , Animals , Drosophila melanogaster , Larva/metabolism
8.
Open Biol ; 11(5): 200408, 2021 05.
Article in English | MEDLINE | ID: mdl-33947246

ABSTRACT

Linker histones H1 are essential chromatin components that exist as multiple developmentally regulated variants. In metazoans, specific H1s are expressed during germline development in a tightly regulated manner. However, the mechanisms governing their stage-dependent expression are poorly understood. Here, we address this question in Drosophila, which encodes for a single germline-specific dBigH1 linker histone. We show that during female germline lineage differentiation, dBigH1 is expressed in germ stem cells and cystoblasts, becomes silenced during transit-amplifying (TA) cystocytes divisions to resume expression after proliferation stops and differentiation starts, when it progressively accumulates in the oocyte. We find that dBigH1 silencing during TA divisions is post-transcriptional and depends on the tumour suppressor Brain tumour (Brat), an essential RNA-binding protein that regulates mRNA translation and stability. Like other oocyte-specific variants, dBigH1 is maternally expressed during early embryogenesis until it is replaced by somatic dH1 at the maternal-to-zygotic transition (MZT). Brat also mediates dBigH1 silencing at MZT. Finally, we discuss the situation in testes, where Brat is not expressed, but dBigH1 is translationally silenced too.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/metabolism , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Histones/biosynthesis , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Histones/genetics
9.
Neurobiol Dis ; 155: 105390, 2021 07.
Article in English | MEDLINE | ID: mdl-33984508

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) mutations are the most common genetic cause of late-onset Parkinson's disease. The pathogenic G2019S mutation enhances LRRK2 kinase activity and induces neurodegeneration in C. elegans, Drosophila and rodent models through unclear mechanisms. Gene expression profiling has the potential to provide detailed insight into the biological pathways modulated by LRRK2 kinase activity. Prior in vivo studies have surveyed the effects of LRRK2 G2019S on genome-wide mRNA expression in complex brain tissues with high cellular heterogeneity, limiting their power to detect more restricted gene expression changes occurring in a cell type-specific manner. Here, we used translating ribosome affinity purification (TRAP) coupled to RNA-seq to profile dopamine neuron-specific gene expression changes caused by LRRK2 G2019S in the Drosophila CNS. A number of genes were differentially expressed in the presence of mutant LRRK2 that represent a broad range of molecular functions including DNA repair (RfC3), mRNA metabolism and translation (Ddx1 and lin-28), calcium homeostasis (MCU), and other categories (Ugt37c1, disp, l(1)G0196, CG6602, CG1126 and CG11068). Further analysis on a subset of these genes revealed that LRRK2 G2019S did not alter their expression across the whole brain, consistent with dopamine neuron-specific effects uncovered by the TRAP approach that may yield insight into the neurodegenerative process. To our knowledge, this is the first study to profile the effects of LRRK2 G2019S specifically on DA neuron gene expression in vivo. Beyond providing a set of differentially expressed gene candidates relevant to LRRK2, we demonstrate the effective use of TRAP to perform high-resolution assessment of dopamine neuron gene expression for the study of PD.


Subject(s)
Dopaminergic Neurons/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Gene Expression Profiling/methods , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/biosynthesis , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation/physiology , Animals , Animals, Genetically Modified , Drosophila
10.
Metab Brain Dis ; 36(4): 669-683, 2021 04.
Article in English | MEDLINE | ID: mdl-33547995

ABSTRACT

Tauopathies belong to a heterogeneous class of neuronal diseases resulting in the metabolic disturbance. A disulfide natural compound of Alpha-Lipoic acid (ALA) has shown numerous pharmacologic, antioxidant, and neuroprotective activities under neuropathological conditions. The aim of this study was to investigate the neuroprotective effects of ALA on the tauopathy-induced oxidative disturbance and behavioral deficits. The transgenic Drosophila model of tauopathy induced by human tauR406W using GAL4/UAS system and effects of ALA (0.001, 0.005, and 0.025 % w/w of diet) on the neuropathology of tau in younger (20 days) and older (30 days) adults were investigated via biochemical, molecular, behavioral and in-situ tissue analyses. Expression of apoptosis-related proteins involving Drosophila Cyt-c-d (trigger of intrinsic apoptosis) and DrICE (effector caspase) were upregulated in both ages (20 and 30 days) and DIAP1 (caspase inhibitor) has reduced only in older model flies compared to the controls. Remarkably, all doses of ALA increased DIAP1 and glutathione (GSH) as well as reducing Cyt-c-d and lipid peroxidation (LPO) in the younger flies compared to the model flies. Moreover, the higher doses of ALA were able to decrease thiol concentrations, to increase total antioxidant capacity, and to improve the behavioral deficits (locomotor function, olfactory memory, and ethanol sensitivity) in the younger flies. On the other hand, only a higher dose of ALA was able to decrease DrICE, Cyt-c-d, LPO, and thiol as well as increasing antioxidant capacity and decreasing ethanol sensitivity (ST50, RT50) in the older flies. TUNEL assay showed that all doses of ALA could potentially increase the DIAP1/DrICE ratio and exert anti-apoptotic effects on younger, but not on the older adults. Furthermore, data obtained from the in-situ ROS assay confirmed that only a higher dose of ALA significantly decreased the ROS level at both ages. Our data showed that an effective neuroprotective dose of ALA and its mechanism of action on this model of tauopathy could potentially be influenced by longevity. Moreover, it was shown that ALA prevents apoptosis and decreases the redox homeostasis, and this partially explains the mechanism by which ALA diminishes behavioral deficits.


Subject(s)
Caspases/biosynthesis , Drosophila Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/biosynthesis , Locomotion/physiology , Oxidative Stress/physiology , Tauopathies/metabolism , Thioctic Acid/therapeutic use , Age Factors , Animals , Animals, Genetically Modified , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Caspases/genetics , Drosophila , Drosophila Proteins/genetics , Female , Homeostasis/drug effects , Homeostasis/physiology , Inhibitor of Apoptosis Proteins/genetics , Locomotion/drug effects , Male , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Tauopathies/drug therapy , Tauopathies/genetics , Thioctic Acid/pharmacology
11.
Sci Rep ; 11(1): 2645, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514791

ABSTRACT

The initiator caspase Dronc is the only CARD-domain containing caspase in Drosophila and is essential for apoptosis. Here, we report that homozygous dronc mutant adult animals are short-lived due to the presence of a poorly developed, defective and leaky intestine. Interestingly, this mutant phenotype can be significantly rescued by enteroblast-specific expression of dronc+ in dronc mutant animals, suggesting that proper Dronc function specifically in enteroblasts, one of four cell types in the intestine, is critical for normal development of the intestine. Furthermore, enteroblast-specific knockdown of dronc in adult intestines triggers hyperplasia and differentiation defects. These enteroblast-specific functions of Dronc do not require the apoptotic pathway and thus occur in a non-apoptotic manner. In summary, we demonstrate that an apoptotic initiator caspase has a very critical non-apoptotic function for normal development and for the control of the cell lineage in the adult midgut and therefore for proper physiology and homeostasis.


Subject(s)
Caspases/biosynthesis , Drosophila Proteins/biosynthesis , Gene Expression Regulation , Intestinal Mucosa/metabolism , Mutation , Animals , Apoptosis , Caspases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster
12.
Dev Biol ; 469: 1-11, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32950464

ABSTRACT

The regulation of formation of the Drosophila heart by the Nkx 2.5 homologue Tinman is a key event during embryonic development. In this study, we identify the highly conserved transcription cofactor Akirin as a key factor in the earliest induction of tinman by the Twist transcription cofactor. akirin mutant embryos display a variety of morphological defects in the heart, including abnormal spacing between rows of aortic cells and abnormal patterning of the aortic outflow tract. akirin mutant embryos have a greatly reduced level of tinman transcripts, together with a reduction of Tinman protein in the earliest stages of cardiac patterning. Further, akirin mutants have reduced numbers of Tinman-positive cardiomyoblasts, concomitant with disrupted patterning and organization of the heart. Finally, despite the apparent formation of the heart in akirin mutants, these mutant hearts exhibit fewer coordinated contractions in akirin mutants compared with wild-type hearts. These results indicate that Akirin is crucial for the first induction of tinman by the Twist transcription factor, and that the success of the cardiac patterning program is highly dependent upon establishing the proper level of tinman at the earliest steps of the cardiac developmental pathway.


Subject(s)
Drosophila Proteins/biosynthesis , Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Nuclear Proteins/physiology , Repressor Proteins/biosynthesis , Trans-Activators/biosynthesis , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Heart/embryology , Mutation , Myocardial Contraction , Myocardium/metabolism , Myocardium/pathology , Nuclear Proteins/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Trans-Activators/genetics , Twist-Related Protein 1/metabolism
13.
J Neurochem ; 157(6): 2119-2127, 2021 06.
Article in English | MEDLINE | ID: mdl-32915460

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disorder characterized pathologically by motor neuron degeneration and associated with aggregation of RNA-binding proteins. TATA-binding protein-associated factor 15 (TAF15) accumulates as cytoplasmic aggregates in neuronal cells, and clearance of these aggregates is considered a potential therapeutic strategy for ALS. However, the exact pathogenic mechanism of TAF15-induced neurotoxicity remains to be elucidated. Glycogen synthase kinase-3 (GSK-3) plays a critical role in the protection of ALS pathology. In the present study, we use a transgenic fly model over-expressing human TAF15 to study the protective effects of Shaggy/GSK3ß on TAF15-induced neuronal toxicity in Drosophila brain. Transgenic flies were examined for locomotor activity and lithium treatment. The expression level and solubility of TAF15 were assessed with western blotting, whereas immunohistochemistry was used to assess TAF15 aggregation in Drosophila brain. We have revealed that Shaggy/GSK3ß was abnormally activated in neurons of TAF15-expressing flies and its inhibition can suppress the defective phenotypes, thereby preventing retinal degeneration and locomotive activity caused by TAF15. We have also found that Shaggy/GSK3ß inhibition in neuronal cells leads to a reduction in TAF15 levels. Indeed, the F-box proteins Slimb and archipelago genetically interact with TAF15 and control TAF15 protein level in Drosophila. Importantly, SCFslimb is a critical regulator for Shaggy/GSK3ß-mediated suppression of TAF15-induced toxicity in Drosophila. The present study has provided an in vivo evidence supporting the molecular mechanism of GSK3ß inhibition for protection against TAF15-linked proteinopathies.


Subject(s)
Brain/metabolism , Cell Cycle Proteins/biosynthesis , Drosophila Proteins/biosynthesis , Glycogen Synthase Kinase 3 beta/biosynthesis , TATA-Binding Protein Associated Factors/biosynthesis , TATA-Binding Protein Associated Factors/toxicity , Ubiquitin-Protein Ligases/biosynthesis , Animals , Animals, Genetically Modified , Brain/pathology , Cell Cycle Proteins/genetics , Drosophila , Drosophila Proteins/genetics , Glycogen Synthase Kinase 3 beta/genetics , Humans , Locomotion/physiology , Male , TATA-Binding Protein Associated Factors/genetics , Ubiquitin-Protein Ligases/genetics
14.
J BUON ; 25(5): 2160-2170, 2020.
Article in English | MEDLINE | ID: mdl-33277831

ABSTRACT

PURPOSE: To investigate the expressions of caspase-3 and survivin in colorectal cancer patients and their possible associations with clinicopathological parameters and the oncological outcome. METHODS: Between January 2008 and December 2011, 85 patients with sporadic colorectal cancer were submitted to colectomy with curative intent. Postoperatively, all patients were followed every three months up to the 36th month. Immunohistochemical detection of the apoptosis-related proteins was carried out on 4-µm-thick deparaffinized sections from all primary tumors. Univariate and multivariate analyses were performed by using the R software for Windows, version 3.3.2. RESULTS: Setting the cut-off point for caspase-3 positivity at 5%, 48% of the patients were characterized as caspase-3(+). Caspase-3 positivity was not found related either to any clinicopathological parameter or to the oncological outcome. Choosing simple survivin positivity as the cut-off point for its expression, 78% of the patients were considered as survivin(+). Survivin inexpression predisposed to poorly differentiated tumors of advanced T stage. However, neither a dismal nor a favorable prognostic role for survivin expression or inexpression was disclosed. By dividing all enrolled patients in four different groups, a trend for worse 3-year overall survival rate in the caspase-3(-)/survivin(-) subgroup of patients was noticed (p=0.067). CONCLUSION: Caspase-3 expression was unrelated to the oncological outcome in colorectal cancer patients. The proposed favorable prognostic role for survivin inexpression was not confirmed. On the contrary, survivin(-) tumors were mainly of poor differentiation and advanced T stage. An inverse relationship between caspase-3 and survivin expressions was also not confirmed. Future studies focusing on specific survivin isoforms expression or inexpression may give answers on apoptotic-antiapoptotic interactions on cancer cell death.


Subject(s)
Caspase 3/biosynthesis , Colorectal Neoplasms/metabolism , Drosophila Proteins/biosynthesis , Survivin/biosynthesis , Aged , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Caspase 3/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drosophila Proteins/genetics , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Survivin/genetics
15.
Biochimie ; 179: 65-68, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32946989

ABSTRACT

Our understanding of cancer-specific metabolic changes is currently unclear. In recent years, the fruit fly Drosophila melanogaster with its powerful genetic tools has become an attractive model for studying both tumor autonomous and the systemic processes resulting from the tumor growth. Here we investigated the effect of tumorigenesis on the modulation of lipid droplets (LDs) in the larval fat bodies (mammalian equivalent of adipose tissue). We have overexpressed Notch signaling alone or in combination with the developmental regulator Myocyte enhancer factor 2 (Mef2) using wing-specific and eye-specific drivers, quantified the size of LDs in the fat body of the different tumor bearing larvae, and estimated the expression of genes associated with lipolysis and lipogenesis. We have found that hyperplastic and neoplastic tumor induced by overexpression of Notch and co-expression of Notch and Mef2 respectively triggers impaired lipid metabolism marked by increased size of fat body LDs. The impaired lipid metabolism in tumor carrying larvae is linked to the altered expression of genes that participate in lipolysis and lipogenesis. These findings reveal modulation of LDs as one of the host's specific response upon tumor initiation. This information could potentially uncover mechanisms for designing innovative approaches to modulate cancer growth.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epithelium/chemistry , Epithelium/metabolism , Fat Body/metabolism , Imaginal Discs/metabolism , Lipid Droplets/metabolism , Animals , Drosophila Proteins/biosynthesis , Eye/growth & development , Eye/pathology , Fat Body/pathology , Gene Expression Regulation, Neoplastic , Hyperplasia/genetics , Hyperplasia/metabolism , Larva/metabolism , Lipogenesis/genetics , Lipolysis/genetics , Myogenic Regulatory Factors/biosynthesis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Notch/biosynthesis , Wings, Animal/growth & development , Wings, Animal/pathology
16.
J Neurosci ; 40(42): 7999-8024, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32928889

ABSTRACT

In multipolar vertebrate neurons, action potentials (APs) initiate close to the soma, at the axonal initial segment. Invertebrate neurons are typically unipolar with dendrites integrating directly into the axon. Where APs are initiated in the axons of invertebrate neurons is unclear. Voltage-gated sodium (NaV) channels are a functional hallmark of the axonal initial segment in vertebrates. We used an intronic Minos-Mediated Integration Cassette to determine the endogenous gene expression and subcellular localization of the sole NaV channel in both male and female Drosophila, para Despite being the only NaV channel in the fly, we show that only 23 ± 1% of neurons in the embryonic and larval CNS express para, while in the adult CNS para is broadly expressed. We generated a single-cell transcriptomic atlas of the whole third instar larval brain to identify para expressing neurons and show that it positively correlates with markers of differentiated, actively firing neurons. Therefore, only 23 ± 1% of larval neurons may be capable of firing NaV-dependent APs. We then show that Para is enriched in an axonal segment, distal to the site of dendritic integration into the axon, which we named the distal axonal segment (DAS). The DAS is present in multiple neuron classes in both the third instar larval and adult CNS. Whole cell patch clamp electrophysiological recordings of adult CNS fly neurons are consistent with the interpretation that Nav-dependent APs originate in the DAS. Identification of the distal NaV localization in fly neurons will enable more accurate interpretation of electrophysiological recordings in invertebrates.SIGNIFICANCE STATEMENT The site of action potential (AP) initiation in invertebrates is unknown. We tagged the sole voltage-gated sodium (NaV) channel in the fly, para, and identified that Para is enriched at a distal axonal segment. The distal axonal segment is located distal to where dendrites impinge on axons and is the likely site of AP initiation. Understanding where APs are initiated improves our ability to model neuronal activity and our interpretation of electrophysiological data. Additionally, para is only expressed in 23 ± 1% of third instar larval neurons but is broadly expressed in adults. Single-cell RNA sequencing of the third instar larval brain shows that para expression correlates with the expression of active, differentiated neuronal markers. Therefore, only 23 ± 1% of third instar larval neurons may be able to actively fire NaV-dependent APs.


Subject(s)
Axon Initial Segment/metabolism , Drosophila Proteins/biosynthesis , Drosophila/metabolism , Neurons/metabolism , Sodium Channels/biosynthesis , Voltage-Gated Sodium Channels/biosynthesis , Action Potentials/physiology , Animals , Axons/physiology , Dendrites/metabolism , Drosophila Proteins/genetics , Electrophysiological Phenomena , Electroretinography , Gene Expression/genetics , Larva , Neuromuscular Junction/metabolism , Neuromuscular Junction/physiology , Patch-Clamp Techniques , Sodium Channels/genetics , Transcriptome , Voltage-Gated Sodium Channels/genetics
17.
Neurochem Int ; 140: 104825, 2020 11.
Article in English | MEDLINE | ID: mdl-32898622

ABSTRACT

PTEN-induced putative kinase 1 (PINK1) mutation induces autosomal recessive Parkinson's Disease (PD), mitochondrial dysfunction is the central pathogenic process. However, more and more studies presented the bulk of the damage to neurons with mitochondrial dysfunction stems from the endoplasmic reticulum (ER) stress. In mitochondria damaged PINK1B9 fly model how protein kinase RNA-like ER kinase (PERK) arm of ER stress functions remains a mystery. Thus, we generated both PERK overexpressed (PEK OE) and down expressed (PEK RNAi) PINK1B9 flies and monitored their motor activity. We found PEK OE decreased the abnormal wing posture rate and rescued PINK1B9 flies' motor activity. Furthermore, we observed the increased number of dopaminergic neurons of protocerebral posterior lateral 1 (PPL1) and the tyrosine hydroxylase (TH) protein levels in PINK1B9 flies. When testing the mitochondrial morphology in flight muscle with TEM, we found that the shape of the mitochondria became normal. The ATP levels of flight muscle tissues were significantly elevated in PEK OE PINK1B9 flies with the increased function of mitochondrial Electron Transport Chain (ETC) Complex I (CI) but not Complex Ⅱ (CⅡ) which is further confirmed by oxygen consumption experiments, Western Blot, and RT-PCR to examine the corresponding subunits. We suggest that overexpression of PERK can rescue PINK1B9 PD flies' pathogenic phenotypes and it is linked with the improved mitochondrial function especially CI of ETC but not CⅡ. Our findings may pave a way for the target of the drug for alleviating the suffering of PINK1 mutant autosomal recessive PD patients.


Subject(s)
Drosophila Proteins/biosynthesis , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Phenotype , Protein Serine-Threonine Kinases/biosynthesis , Animals , Animals, Genetically Modified , Drosophila , Drosophila Proteins/genetics , Gene Expression , Male , Mitochondria/genetics , Neurodegenerative Diseases/genetics , Protein Serine-Threonine Kinases/genetics
18.
Neurobiol Dis ; 143: 105013, 2020 09.
Article in English | MEDLINE | ID: mdl-32653676

ABSTRACT

Fragile X associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder caused by expansion of CGG repeats in the 5' UTR of the fragile X mental retardation 1 (FMR1) gene. Using the well-established FXTAS Drosophila model, we performed a high-throughput chemical screen using 3200 small molecules. NSC363998 was identified to suppress the neurodegeneration caused by riboCGG (rCGG) repeats. Three predicted targets of a NSC363998 derivative are isopeptidases in the neddylation pathway and could modulate the neurotoxicity caused by the rCGG repeats. Decreasing levels of neddylation resulted in enhancing neurodegeneration phenotypes, while up-regulation could rescue the phenotypes. Furthermore, known neddylation substrates, Cul3 and Vhl, and their downstream target, Sima, were found to modulate rCGG90-dependent neurotoxicity. Our results suggest that altered neddylation activity can modulate the rCGG repeat-mediated toxicity by regulating Sima protein levels, which could serve as a potential therapeutic target for FXTAS.


Subject(s)
Ataxia/metabolism , Fragile X Syndrome/metabolism , Gene Expression Regulation/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Nerve Degeneration/metabolism , Protein Processing, Post-Translational/physiology , Tremor/metabolism , Animals , Ataxia/pathology , Drosophila , Drosophila Proteins/biosynthesis , Fragile X Syndrome/pathology , Humans , NEDD8 Protein , Nerve Degeneration/pathology , Neuroprotective Agents/pharmacology , Protein Processing, Post-Translational/drug effects , Tremor/pathology , Trinucleotide Repeat Expansion
19.
Sheng Wu Gong Cheng Xue Bao ; 36(6): 1198-1208, 2020 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-32597069

ABSTRACT

In this research, we studied the formation of Drosophila melanogaster FADD (Fas-associated death domain-containing protein) amyloid fiber and its influence on signal transduction in IMD (Immune deficiency) signaling pathway to better understand the regulation mechanism of Drosophila innate immune signaling pathway, which will provide reference for the immune regulation in other species. First, we purified dFADD protein expressed in Escherichia coli and performed Sulfur flavin T binding and transmission electron microscopy to identify the dFADD amyloid fibers formed in vitro. Then we investigated the formation of dFADD polymers in S2 cells using SDD-AGE and confocal microscope. We also constructed dFADD mutants to find out which domain is essential to fiber formation and its effect on IMD signal transduction. Our results revealed that dFADD could be polymerized to form amyloid fiber polymers in vitro and inside the cells. Formation of fibers relies on DED (Death-effector domain) domain of dFADD, since DED domain-deleted mutant existed as a monomer. Dual luciferase reporter assay showed that intact DED domain was required for the induction of downstream antimicrobial peptides, indicating that fiber formation was the key to IMD signal transduction. Our study revealed the role of dFADD in mediating the cascade between IMD and Dredd in the IMD signaling pathway by forming amyloid fibers, suggesting an evolutionarily conserved regulatory mechanism of innate immune signaling pathway.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Immunity, Innate , Signal Transduction , Animals , Drosophila Proteins/biosynthesis , Drosophila Proteins/immunology , Drosophila melanogaster/immunology , Fas-Associated Death Domain Protein/biosynthesis , Fas-Associated Death Domain Protein/immunology , Immunity, Innate/immunology
20.
Methods Mol Biol ; 2143: 321-338, 2020.
Article in English | MEDLINE | ID: mdl-32524491

ABSTRACT

Axon degeneration elicits a range of immune responses from local glial cells, including striking changes in glial gene expression, morphology, and phagocytic activity. Here, we describe a detailed set of protocols to assess discrete components of the glial reaction to axotomy in the adult nervous system of Drosophila melanogaster. These methods allow one to visualize and quantify transcriptional, morphological, and functional responses of glia to degenerating axons in a model system that is highly amenable to genetic manipulation.


Subject(s)
Axons/physiology , Drosophila melanogaster/immunology , Intravital Microscopy , Microscopy, Confocal/methods , Neuroglia/immunology , Wallerian Degeneration/immunology , Animals , Axotomy , Central Nervous System/pathology , DNA, Complementary/genetics , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Expression Regulation , Genes, Reporter , Image Processing, Computer-Assisted , Immunohistochemistry/instrumentation , Immunohistochemistry/methods , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroglia/metabolism , Phagocytosis , Polymerase Chain Reaction/methods , Wallerian Degeneration/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL