Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nature ; 598(7880): 353-358, 2021 10.
Article in English | MEDLINE | ID: mdl-34588695

ABSTRACT

Time-restricted feeding (TRF) has recently gained interest as a potential anti-ageing treatment for organisms from Drosophila to humans1-5. TRF restricts food intake to specific hours of the day. Because TRF controls the timing of feeding, rather than nutrient or caloric content, TRF has been hypothesized to depend on circadian-regulated functions; the underlying molecular mechanisms of its effects remain unclear. Here, to exploit the genetic tools and well-characterized ageing markers of Drosophila, we developed an intermittent TRF (iTRF) dietary regimen that robustly extended fly lifespan and delayed the onset of ageing markers in the muscles and gut. We found that iTRF enhanced circadian-regulated transcription and that iTRF-mediated lifespan extension required both circadian regulation and autophagy, a conserved longevity pathway. Night-specific induction of autophagy was both necessary and sufficient to extend lifespan on an ad libitum diet and also prevented further iTRF-mediated lifespan extension. By contrast, day-specific induction of autophagy did not extend lifespan. Thus, these results identify circadian-regulated autophagy as a critical contributor to iTRF-mediated health benefits in Drosophila. Because both circadian regulation and autophagy are highly conserved processes in human ageing, this work highlights the possibility that behavioural or pharmaceutical interventions that stimulate circadian-regulated autophagy might provide people with similar health benefits, such as delayed ageing and lifespan extension.


Subject(s)
Autophagy/physiology , Circadian Rhythm/physiology , Drosophila melanogaster/physiology , Feeding Behavior/physiology , Longevity/physiology , Aging/genetics , Aging/radiation effects , Animals , Autophagy/genetics , Biomarkers , Circadian Clocks/radiation effects , Circadian Rhythm/genetics , Circadian Rhythm/radiation effects , Darkness , Drosophila melanogaster/genetics , Drosophila melanogaster/radiation effects , Feeding Behavior/radiation effects , Female , Longevity/genetics , Longevity/radiation effects , Male , Time Factors
2.
Nat Methods ; 15(11): 921-923, 2018 11.
Article in English | MEDLINE | ID: mdl-30377360

ABSTRACT

FLIRT (fast local infrared thermogenetics) is a microscopy-based technology to locally and reversibly manipulate protein function while simultaneously monitoring the effects in vivo. FLIRT locally inactivates fast-acting temperature-sensitive mutant proteins. We demonstrate that FLIRT can control temperature-sensitive proteins required for cell division, Delta-Notch cell fate signaling, and germline structure in Caenorhabditis elegans with cell-specific and even subcellular precision.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Genetic Techniques/instrumentation , Infrared Rays , Molecular Imaging/methods , Mutation , Temperature , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/radiation effects , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Gene Expression Regulation , Germ Cells , Microscopy , Receptors, Notch , Signal Transduction
3.
PLoS Biol ; 16(7): e2005206, 2018 07.
Article in English | MEDLINE | ID: mdl-30001323

ABSTRACT

Although sleep appears to be broadly conserved in animals, the physiological functions of sleep remain unclear. In this study, we sought to identify a physiological defect common to a diverse group of short-sleeping Drosophila mutants, which might provide insight into the function and regulation of sleep. We found that these short-sleeping mutants share a common phenotype of sensitivity to acute oxidative stress, exhibiting shorter survival times than controls. We further showed that increasing sleep in wild-type flies using genetic or pharmacological approaches increases survival after oxidative challenge. Moreover, reducing oxidative stress in the neurons of wild-type flies by overexpression of antioxidant genes reduces the amount of sleep. Together, these results support the hypothesis that a key function of sleep is to defend against oxidative stress and also point to a reciprocal role for reactive oxygen species (ROS) in neurons in the regulation of sleep.


Subject(s)
Drosophila melanogaster/physiology , Oxidative Stress , Sleep/physiology , Animals , Antioxidants/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Immunity , Longevity , Mutation/genetics , Neurons/metabolism , Oxidative Stress/genetics , RNA Interference , Reactive Oxygen Species/metabolism
4.
Eur J Neurosci ; 51(1): 494-508, 2020 01.
Article in English | MEDLINE | ID: mdl-30295966

ABSTRACT

A key feature of circadian rhythms is the sleep/wake cycle. Sleep causes reduced responsiveness to the environment, which puts animals in a particularly vulnerable state; yet sleep has been conserved throughout evolution, indicating that it fulfils a vital purpose. A core function of sleep across species has not been identified, but substantial advances in sleep research have been made in recent years using the genetically tractable model organism, Drosophila melanogaster. This review describes the universality of sleep, the regulation of sleep, and current theories on the function of sleep, highlighting a historical and often overlooked theory called the Free Radical Flux Theory of Sleep. Additionally, we summarize our recent work with short-sleeping Drosophila mutants and other genetic and pharmacological tools for manipulating sleep which supports an antioxidant theory of sleep and demonstrates a bi-directional relationship between sleep and oxidative stress.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Circadian Rhythm , Drosophila , Sleep
5.
PLoS Pathog ; 8(1): e1002445, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22253593

ABSTRACT

Survival of bacterial infection is the result of complex host-pathogen interactions. An often-overlooked aspect of these interactions is the circadian state of the host. Previously, we demonstrated that Drosophila mutants lacking the circadian regulatory proteins Timeless (Tim) and Period (Per) are sensitive to infection by S. pneumoniae. Sensitivity to infection can be mediated either by changes in resistance (control of microbial load) or tolerance (endurance of the pathogenic effects of infection). Here we show that Tim regulates resistance against both S. pneumoniae and S. marcescens. We set out to characterize and identify the underlying mechanism of resistance that is circadian-regulated. Using S. pneumoniae, we found that resistance oscillates daily in adult wild-type flies and that these oscillations are absent in Tim mutants. Drosophila have at least three main resistance mechanisms to kill high levels of bacteria in their hemolymph: melanization, antimicrobial peptides, and phagocytosis. We found that melanization is not circadian-regulated. We further found that basal levels of AMP gene expression exhibit time-of-day oscillations but that these are Tim-independent; moreover, infection-induced AMP gene expression is not circadian-regulated. We then show that phagocytosis is circadian-regulated. Wild-type flies exhibit up-regulated phagocytic activity at night; Tim mutants have normal phagocytic activity during the day but lack this night-time peak. Tim appears to regulate an upstream event in phagocytosis, such as bacterial recognition or activation of phagocytic hemocytes. Interestingly, inhibition of phagocytosis in wild type flies results in survival kinetics similar to Tim mutants after infection with S. pneumoniae. Taken together, these results suggest that loss of circadian oscillation of a specific immune function (phagocytosis) can have significant effects on long-term survival of infection.


Subject(s)
Bacteria/immunology , Drosophila Proteins/physiology , Drosophila/genetics , Drosophila/immunology , Phagocytosis/genetics , Animals , Animals, Genetically Modified , Bacteria/growth & development , Bacteria/metabolism , Bacterial Infections/genetics , Bacterial Infections/microbiology , Bacterial Infections/mortality , Base Sequence , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/physiology , Colony Count, Microbial , Drosophila/microbiology , Drosophila Proteins/genetics , Host-Pathogen Interactions , Male , Models, Biological , Molecular Sequence Data , Survival Analysis
6.
bioRxiv ; 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38979134

ABSTRACT

Cytokinesis, the physical division of one cell into two, is typically assumed to use the same molecular process across animal cells. However, regulation of cell division can vary significantly among different cell types, even within the same multicellular organism. Using six fast-acting temperature-sensitive (ts) cytokinesis-defective mutants, we found that each had unique cell type-specific profiles in the early C. elegans embryo. Certain cell types were more sensitive than others to actomyosin and spindle signaling disruptions, disrupting two members of the same complex could result in different phenotypes, and protection against actomyosin inhibition did not always protect against spindle signaling inhibition.

7.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798632

ABSTRACT

Insulin resistance and diabetes are associated with many health issues including higher rates of birth defects and miscarriage during pregnancy. Because insulin resistance and diabetes are both associated with obesity, which also affects fertility, the role of insulin signaling itself in embryo development is not well understood. A key downstream target of the insulin/insulin-like growth factor signaling (IIS) pathway is the forkhead family transcription factor FoxO (DAF-16 in C. elegans ). Here, we used quantitative live imaging to measure the patterning of endogenously tagged FoxO/DAF-16 in the early worm embryo. In 2-4-cell stage embryos, FoxO/DAF-16 initially localized uniformly to all cell nuclei, then became dramatically enriched in germ precursor cell nuclei beginning at the 8-cell stage. This nuclear enrichment in early germ precursor cells required germ fate specification, PI3K (AGE-1)- and PTEN (DAF-18)-mediated phospholipid regulation, and the deubiquitylase USP7 (MATH-33), yet was unexpectedly insulin receptor (DAF-2)- and AKT-independent. Functional analysis revealed that FoxO/DAF-16 acts as a cell cycle pacer for early cleavage divisions-without FoxO/DAF-16 cell cycles were shorter than in controls, especially in germ lineage cells. These results reveal the germ lineage specific patterning, upstream regulation, and cell cycle role for FoxO/DAF-16 during early C. elegans embryogenesis.

8.
Mol Biol Cell ; 35(7): ar94, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38696255

ABSTRACT

Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formincyk-1(ts) mutant Caenorhabditis elegans 4-cell embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide with greatly reduced F-actin levels at the cell division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septinUNC-59 and anillinANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into the regulation of cytokinesis in other cell types, especially in stem cells with high potency.


Subject(s)
Actins , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cell Division , Cytokinesis , Germ Cells , Septins , Animals , Cytokinesis/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Septins/metabolism , Septins/genetics , Germ Cells/metabolism , Germ Cells/cytology , Actins/metabolism , Contractile Proteins/metabolism , Actomyosin/metabolism
9.
Sci Rep ; 14(1): 1541, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38233464

ABSTRACT

Mutations in Cullin-3 (Cul3), a conserved gene encoding a ubiquitin ligase, are strongly associated with autism spectrum disorder (ASD). Here, we characterize ASD-related pathologies caused by neuron-specific Cul3 knockdown in Drosophila. We confirmed that neuronal Cul3 knockdown causes short sleep, paralleling sleep disturbances in ASD. Because sleep defects and ASD are linked to metabolic dysregulation, we tested the starvation response of neuronal Cul3 knockdown flies; they starved faster and had lower triacylglyceride levels than controls, suggesting defects in metabolic homeostasis. ASD is also characterized by increased biomarkers of oxidative stress; we found that neuronal Cul3 knockdown increased sensitivity to hyperoxia, an exogenous oxidative stress. Additional hallmarks of ASD are deficits in social interactions and learning. Using a courtship suppression assay that measures social interactions and memory of prior courtship, we found that neuronal Cul3 knockdown reduced courtship and learning compared to controls. Finally, we found that neuronal Cul3 depletion alters the anatomy of the mushroom body, a brain region required for memory and sleep. Taken together, the ASD-related phenotypes of neuronal Cul3 knockdown flies establish these flies as a genetic model to study molecular and cellular mechanisms underlying ASD pathology, including metabolic and oxidative stress dysregulation and neurodevelopment.


Subject(s)
Autism Spectrum Disorder , Drosophila Proteins , Animals , Autism Spectrum Disorder/genetics , Cullin Proteins/genetics , Cullin Proteins/metabolism , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Neurons/metabolism
10.
Sci Rep ; 13(1): 10411, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37369755

ABSTRACT

Inhibitors of enzymes that inactivate amine neurotransmitters (dopamine, serotonin), such as catechol-O-methyltransferase (COMT) and monoamine oxidase (MAO), are thought to increase neurotransmitter levels and are widely used to treat Parkinson's disease and psychiatric disorders, yet the role of these enzymes in regulating behavior remains unclear. Here, we investigated the genetic loss of a similar enzyme in the model organism Drosophila melanogaster. Because the enzyme Ebony modifies and inactivates amine neurotransmitters, its loss is assumed to increase neurotransmitter levels, increasing behaviors such as aggression and courtship and decreasing sleep. Indeed, ebony mutants have been described since 1960 as "aggressive mutants," though this behavior has not been quantified. Using automated machine learning-based analyses, we quantitatively confirmed that ebony mutants exhibited increased aggressive behaviors such as boxing but also decreased courtship behaviors and increased sleep. Through tissue-specific knockdown, we found that ebony's role in these behaviors was specific to glia. Unexpectedly, direct measurement of amine neurotransmitters in ebony brains revealed that their levels were not increased but reduced. Thus, increased aggression is the anomalous behavior for this neurotransmitter profile. We further found that ebony mutants exhibited increased aggression only when fighting each other, not when fighting wild-type controls. Moreover, fights between ebony mutants were less likely to end with a clear winner than fights between controls or fights between ebony mutants and controls. In ebony vs. control fights, ebony mutants were more likely to win. Together, these results suggest that ebony mutants exhibit prolonged aggressive behavior only in a specific context, with an equally dominant opponent.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Amines , Catechol O-Methyltransferase , DNA-Binding Proteins/genetics , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Neuroglia
11.
bioRxiv ; 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-38014027

ABSTRACT

Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formin cyk-1 (ts) mutant C. elegans embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide without detectable F-actin at the division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septin UNC-59 and anillin ANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into cytokinetic regulation in other cell types, especially in stem cells with high potency.

12.
J Cell Biol ; 221(3)2022 03 07.
Article in English | MEDLINE | ID: mdl-34994802

ABSTRACT

Contractile ring constriction during cytokinesis is thought to compact central spindle microtubules to form the midbody, an antiparallel microtubule bundle at the intercellular bridge. In Caenorhabditis elegans, central spindle microtubule assembly requires targeting of the CLASP family protein CLS-2 to the kinetochores in metaphase and spindle midzone in anaphase. CLS-2 targeting is mediated by the CENP-F-like HCP-1/2, but their roles in cytokinesis and midbody assembly are not known. We found that although HCP-1 and HCP-2 mostly function cooperatively, HCP-1 plays a more primary role in promoting CLS-2-dependent central spindle microtubule assembly. HCP-1/2 codisrupted embryos did not form central spindles but completed cytokinesis and formed functional midbodies capable of supporting abscission. These central spindle-independent midbodies appeared to form via contractile ring constriction-driven bundling of astral microtubules at the furrow tip. This work suggests that, in the absence of a central spindle, astral microtubules can support midbody assembly and that midbody assembly is more predictive of successful cytokinesis than central spindle assembly.


Subject(s)
Spindle Apparatus/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/metabolism , Embryo, Nonmammalian/metabolism , Green Fluorescent Proteins/metabolism , Microtubules/metabolism
13.
Sci Rep ; 12(1): 9523, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35681073

ABSTRACT

Traumatic brain injury (TBI) affects millions annually and is associated with long-term health decline. TBI also shares molecular and cellular hallmarks with neurodegenerative diseases (NDs), typically increasing in prevalence with age, and is a major risk factor for developing neurodegeneration later in life. While our understanding of genes and pathways that underlie neurotoxicity in specific NDs has advanced, we still lack a complete understanding of early molecular and physiological changes that drive neurodegeneration, particularly as an individual ages following a TBI. Recently Drosophila has been introduced as a model organism for studying closed-head TBI. In this paper, we deliver a TBI to flies early in adult life, and then measure molecular and physiological phenotypes at short-, mid-, and long-term timepoints following the injury. We aim to identify the timing of changes that contribute to neurodegeneration. Here we confirm prior work demonstrating a TBI-induced decline in lifespan, and present evidence of a progressive decline in locomotor function, robust acute and modest chronic neuroinflammation, and a late-onset increase in protein aggregation. We also present evidence of metabolic dysfunction, in the form of starvation sensitivity and decreased lipids, that persists beyond the immediate injury response, but does not differ long-term. An intervention of dietary restriction (DR) partially ameliorates some TBI-induced phenotypes, including lifespan and locomotor function, though it does not alter the pattern of starvation sensitivity of injured flies. In the future, molecular pathways identified as altered following TBI-particularly in the short-, or mid-term-could present potential therapeutic targets.


Subject(s)
Brain Injuries, Traumatic , Neurodegenerative Diseases , Animals , Brain Injuries, Traumatic/metabolism , Drosophila , Drosophila melanogaster/physiology , Longevity , Neurodegenerative Diseases/metabolism , Phenotype
14.
Nat Commun ; 11(1): 1927, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32317636

ABSTRACT

Because old age is associated with defects in circadian rhythm, loss of circadian regulation is thought to be pathogenic and contribute to mortality. We show instead that loss of specific circadian clock components Period (Per) and Timeless (Tim) in male Drosophila significantly extends lifespan. This lifespan extension is not mediated by canonical diet-restriction longevity pathways but is due to altered cellular respiration via increased mitochondrial uncoupling. Lifespan extension of per mutants depends on mitochondrial uncoupling in the intestine. Moreover, upregulated uncoupling protein UCP4C in intestinal stem cells and enteroblasts is sufficient to extend lifespan and preserve proliferative homeostasis in the gut with age. Consistent with inducing a metabolic state that prevents overproliferation, mitochondrial uncoupling drugs also extend lifespan and inhibit intestinal stem cell overproliferation due to aging or even tumorigenesis. These results demonstrate that circadian-regulated intestinal mitochondrial uncoupling controls longevity in Drosophila and suggest a new potential anti-aging therapeutic target.


Subject(s)
Circadian Rhythm , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Period Circadian Proteins/metabolism , Animals , CRISPR-Cas Systems , Carcinogenesis , Cell Proliferation , Circadian Clocks , Homeostasis , Intestines/pathology , Longevity , Male , Membrane Potential, Mitochondrial , Mutation , Oxidative Stress/physiology , Oxygen Consumption , Uncoupling Protein 1/metabolism
15.
Curr Biol ; 16(20): 1977-85, 2006 Oct 24.
Article in English | MEDLINE | ID: mdl-17055976

ABSTRACT

BACKGROUND: Studies in Drosophila have taught us a great deal about how animals regulate the immediate innate immune response, but we still know little about how infections cause pathology. Here, we examine the pathogenesis associated with Mycobacterium marinum infection in the fly. M. marinum is closely related to M. tuberculosis, which causes tuberculosis in people. RESULTS: A microarray analysis showed that metabolism is profoundly affected in M. marinum-infected flies. A genetic screen identified foxo mutants as slower-dying after infection than wild-type flies. FOXO activity is inhibited by the insulin effector kinase Akt; we show that Akt activation is systemically reduced as a result of M. marinum infection. Finally, we show that flies infected with Mycobacterium marinum undergo a process like wasting: They progressively lose metabolic stores, in the form of fat and glycogen. They also become hyperglycemic. In contrast, foxo mutants exhibit less wasting. CONCLUSIONS: In people, many infections--including tuberculosis--can cause wasting, much as we see in Drosophila. Our study is the first examination of the metabolic consequences of infection in a genetically tractable invertebrate and gives insight into the metabolic consequences of mycobacterial infection, implicating impaired insulin signaling as a key mediator of these events. These results suggest that the fly can be used to study more than the immediate innate immune response to infection; it can also be used to understand the physiological consequences of infection and the immune response.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/microbiology , Energy Metabolism/physiology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/physiology , Mycobacterium marinum , Proto-Oncogene Proteins c-akt/metabolism , Weight Loss/physiology , Animals , Blotting, Western , DNA Primers , Drosophila melanogaster/immunology , Insulin/metabolism , Longevity , Microarray Analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
PLoS Pathog ; 3(3): e26, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17352533

ABSTRACT

Drosophila melanogaster, like other invertebrates, relies solely on its innate immune response to fight invading microbes; by definition, innate immunity lacks adaptive characteristics. However, we show here that priming Drosophila with a sublethal dose of Streptococcus pneumoniae protects against an otherwise-lethal second challenge of S. pneumoniae. This protective effect exhibits coarse specificity for S. pneumoniae and persists for the life of the fly. Although not all microbial challenges induced this specific primed response, we find that a similar specific protection can be elicited by Beauveria bassiana, a natural fly pathogen. To characterize this primed response, we focused on S. pneumoniae-induced protection. The mechanism underlying this protective effect requires phagocytes and the Toll pathway. However, activation of the Toll pathway is not sufficient for priming-induced protection. This work contradicts the paradigm that insect immune responses cannot adapt and will promote the search for similar responses overlooked in organisms with an adaptive immune response.


Subject(s)
Drosophila melanogaster/immunology , Immunity, Innate , Phagocytes/physiology , Toll-Like Receptors/physiology , Animals , Cells, Cultured , Drosophila Proteins , Drosophila melanogaster/microbiology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Streptococcus pneumoniae/pathogenicity , Vaccination
17.
PLoS Pathog ; 3(3): e41, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17381241

ABSTRACT

We showed previously that eiger, the Drosophila tumor necrosis factor homolog, contributes to the pathology induced by infection with Salmonella typhimurium. We were curious whether eiger is always detrimental in the context of infection or if it plays a role in fighting some types of microbes. We challenged wild-type and eiger mutant flies with a collection of facultative intracellular and extracellular pathogens, including a fungus and Gram-positive and Gram-negative bacteria. The response of eiger mutants divided these microbes into two groups: eiger mutants are immunocompromised with respect to extracellular pathogens but show no change or reduced sensitivity to facultative intracellular pathogens. Hence, eiger helps fight infections but also can cause pathology. We propose that eiger activates the cellular immune response of the fly to aid clearance of extracellular pathogens. Intracellular pathogens, which can already defeat professional phagocytes, are unaffected by eiger.


Subject(s)
Beauveria/pathogenicity , Burkholderia cepacia/pathogenicity , Drosophila Proteins/physiology , Drosophila/microbiology , Gram-Positive Bacteria/pathogenicity , Membrane Proteins/physiology , Animals , Beauveria/immunology , Burkholderia cepacia/immunology , Drosophila/immunology , Drosophila Proteins/genetics , Gene Expression Regulation , Gram-Positive Bacteria/immunology , Immunity, Innate/physiology , Immunocompromised Host/immunology , Membrane Proteins/genetics , Mutation
18.
J Cell Biol ; 161(2): 349-58, 2003 Apr 28.
Article in English | MEDLINE | ID: mdl-12719474

ABSTRACT

Microtubules (MTs) polymerized with GMPCPP, a slowly hydrolyzable GTP analogue, are stable in buffer but are rapidly depolymerized in Xenopus egg extracts. This depolymerization is independent of three previously identified MT destabilizers (Op18, katanin, and XKCM1/KinI). We purified the factor responsible for this novel depolymerizing activity using biochemical fractionation and a visual activity assay and identified it as XMAP215, previously identified as a prominent MT growth-promoting protein in Xenopus extracts. Consistent with the purification results, we find that XMAP215 is necessary for GMPCPP-MT destabilization in extracts and that recombinant full-length XMAP215 as well as an NH2-terminal fragment have depolymerizing activity in vitro. Stimulation of depolymerization is specific for the MT plus end. These results provide evidence for a robust MT-destabilizing activity intrinsic to this microtubule-associated protein and suggest that destabilization may be part of its essential biochemical functions. We propose that the substrate in our assay, GMPCPP-stabilized MTs, serves as a model for the pause state of MT ends and that the multiple activities of XMAP215 are unified by a mechanism of antagonizing MT pauses.


Subject(s)
Cell Extracts/chemistry , Cell Extracts/pharmacology , Guanosine Triphosphate/analogs & derivatives , Microtubule-Associated Proteins/isolation & purification , Microtubules/metabolism , Oocytes/metabolism , Xenopus Proteins , Xenopus laevis/metabolism , Animals , Biological Assay , Female , Guanosine Triphosphate/pharmacology , Microtubule Proteins/biosynthesis , Microtubule Proteins/drug effects , Microtubule Proteins/genetics , Microtubule-Associated Proteins/pharmacology , Microtubules/drug effects , Models, Biological , Oocytes/drug effects , Protein Structure, Tertiary/physiology , Recombinant Fusion Proteins/pharmacology
19.
J Comp Neurol ; 527(3): 508-521, 2019 02 15.
Article in English | MEDLINE | ID: mdl-29744881

ABSTRACT

In the developing mouse optic tract, retinal ganglion cell (RGC) axon position is organized by topography and laterality (i.e., eye-specific or ipsi- and contralateral segregation). Our lab previously showed that ipsilaterally projecting RGCs are segregated to the lateral aspect of the developing optic tract and found that ipsilateral axons self-fasciculate to a greater extent than contralaterally projecting RGC axons in vitro. However, the full complement of axon-intrinsic and -extrinsic factors mediating eye-specific segregation in the tract remain poorly understood. Glia, which are known to express several guidance cues in the visual system and regulate the navigation of ipsilateral and contralateral RGC axons at the optic chiasm, are natural candidates for contributing to eye-specific pre-target axon organization. Here, we investigate the spatiotemporal expression patterns of both putative astrocytes (Aldh1l1+ cells) and microglia (Iba1+ cells) in the embryonic and neonatal optic tract. We quantified the localization of ipsilateral RGC axons to the lateral two-thirds of the optic tract and analyzed glia position and distribution relative to eye-specific axon organization. While our results indicate that glial segregation patterns do not strictly align with eye-specific RGC axon segregation in the tract, we identify distinct spatiotemporal organization of both Aldh1l1+ cells and microglia in and around the developing optic tract. These findings inform future research into molecular mechanisms of glial involvement in RGC axon growth and organization in the developing retinogeniculate pathway.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Neuroglia/metabolism , Optic Tract/embryology , Optic Tract/metabolism , Retinal Dehydrogenase/metabolism , Retinal Ganglion Cells/metabolism , Age Factors , Aldehyde Dehydrogenase 1 Family/analysis , Animals , Axons/metabolism , Mice , Mice, Inbred C57BL , Optic Tract/cytology , Retinal Dehydrogenase/analysis , Visual Pathways/cytology , Visual Pathways/embryology , Visual Pathways/metabolism
20.
Elife ; 82019 10 15.
Article in English | MEDLINE | ID: mdl-31613218

ABSTRACT

In Drosophila, ~150 neurons expressing molecular clock proteins regulate circadian behavior. Sixteen of these neurons secrete the neuropeptide Pdf and have been called 'master pacemakers' because they are essential for circadian rhythms. A subset of Pdf+ neurons (the morning oscillator) regulates morning activity and communicates with other non-Pdf+ neurons, including a subset called the evening oscillator. It has been assumed that the molecular clock in Pdf+ neurons is required for these functions. To test this, we developed and validated Gal4-UAS based CRISPR tools for cell-specific disruption of key molecular clock components, period and timeless. While loss of the molecular clock in both the morning and evening oscillators eliminates circadian locomotor activity, the molecular clock in either oscillator alone is sufficient to rescue circadian locomotor activity in the absence of the other. This suggests that clock neurons do not act in a hierarchy but as a distributed network to regulate circadian activity.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Neurons/metabolism , Neuropeptides/genetics , Period Circadian Proteins/genetics , Animals , Brain/cytology , Brain/metabolism , Brain/radiation effects , CRISPR-Cas Systems , Cell Communication , Cell Lineage/genetics , Circadian Clocks/drug effects , Circadian Rhythm/drug effects , Darkness , Drosophila Proteins/deficiency , Drosophila melanogaster/metabolism , Drosophila melanogaster/radiation effects , Feedback, Physiological , Gene Editing , Gene Expression Regulation , Light Signal Transduction/genetics , Locomotion/genetics , Locomotion/radiation effects , Nerve Net/metabolism , Nerve Net/radiation effects , Neurons/cytology , Neurons/radiation effects , Neuropeptides/deficiency , Period Circadian Proteins/deficiency , Transcription Factors/deficiency , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL