Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Adv ; 8(37): eabq2611, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36103534

ABSTRACT

DNA double-strand breaks occur in many acute and long-term neurological conditions, including neurodegeneration, neurotrauma, and stroke. Nonrepaired breaks chronically activate the DNA damage response in neurons, leading to neural dysfunction and apoptosis. Here, we show that targeting of the central ATM-Chk2 pathway regulating the response to double-strand breaks slows neural decline in Drosophila models of chronic neurodegeneration. Inhibitors of ATM-Chk2, but not the parallel ATR-Chk1 pathway, also promote marked, functional recovery after acute central nervous system injury in rats, suggesting that inhibiting nonhomologous end-joining rather than homologous recombination is crucial for neuroprotection. We demonstrate that the Chk2 inhibitor, prexasertib, which has been evaluated in phase 2 clinical trials for cancer, has potent neuroprotective effects and represents a new treatment option to promote functional recovery after spinal cord or optic nerve injury.


Subject(s)
DNA Damage , Neuroprotection , Animals , Axons , Checkpoint Kinase 1 , Nerve Regeneration , Rats
3.
Biochim Biophys Acta Mol Basis Dis ; 1866(9): 165614, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31783156

ABSTRACT

The NCLs (neuronal ceroid lipofuscinosis) are forms of neurodegenerative disease that affect people of all ages and ethnicities but are most prevalent in children. Commonly known as Batten disease, this debilitating neurological disorder is comprised of 13 different subtypes that are categorized based on the particular gene that is mutated (CLN1-8, CLN10-14). The pathological mechanisms underlying the NCLs are not well understood due to our poor understanding of the functions of NCL proteins. Only one specific treatment (enzyme replacement therapy) is approved, which is for the treating the brain in CLN2 disease. Hence there remains a desperate need for further research into disease-modifying treatments. In this review, we present and evaluate the genes, proteins and studies performed in the social amoeba, nematode, fruit fly, zebrafish, mouse and large animals pertinent to NCL. In particular, we highlight the use of multicellular model organisms to study NCL protein function, pathology and pathomechanisms. Their use in testing novel therapeutic approaches is also presented. With this information, we highlight how future research in these systems may be able to provide new insight into NCL protein functions in human cells and aid in the development of new therapies.


Subject(s)
Biomedical Research , Disease Models, Animal , Membrane Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Animals , Enzyme Replacement Therapy , Humans , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/therapy , Tripeptidyl-Peptidase 1
4.
Sci Rep ; 9(1): 15592, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666534

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs) are a group of fatal, monogenic neurodegenerative disorders with an early onset in infancy or childhood. Despite identification of the genes disrupted in each form of the disease, their normal cellular role and how their deficits lead to disease pathology is not fully understood. Cln7, a major facilitator superfamily domain-containing protein, is affected in a late infantile-onset form of NCL. Cln7 is conserved across species suggesting a common function. Here we demonstrate that Cln7 is required for the normal growth of synapses at the Drosophila larval neuromuscular junction. In a Cln7 mutant, synapses fail to develop fully leading to reduced function and behavioral changes with dysregulation of TOR activity. Cln7 expression is restricted to the post-synaptic cell and the protein localizes to vesicles immediately adjacent to the post-synaptic membrane. Our data suggest an involvement for Cln7 in regulating trans-synaptic communication necessary for normal synapse development.


Subject(s)
Membrane Transport Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Synapses/physiology , Animals , Bone Morphogenetic Proteins/metabolism , Drosophila melanogaster , Mechanistic Target of Rapamycin Complex 1/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/physiopathology , Signal Transduction
5.
J Neurogenet ; 33(3): 190-198, 2019 09.
Article in English | MEDLINE | ID: mdl-31290369

ABSTRACT

The fruit fly, Drosophila, is commonly used to study late-onset neurodegenerative diseases due to the combination of powerful genetic tools, cheap and simple husbandry and short lifespan. One widely-used measure of disease progression is the age-dependent decline in motor performance that manifests in most Drosophila neurodegeneration models. This is usually quantified using a simple climbing assay. However, the standard climbing assay lacks sensitivity and suffers from high variability meaning large numbers of flies are needed or bespoke apparatus and software solutions. Here, we present a modification of the open-source, MATLAB-based, DART software to measure the decline in "startle response" with age. We demonstrate that the DART setup is more sensitive to the motor performance decline induced by adult-onset neuronal expression of amyloid beta (Aß) peptides than a traditional climbing assay despite using smaller cohorts of flies. DART also has the potential to generate multiple metrics of motor behaviour during the startle response. The software requires no coding skills to operate and the required apparatus can be purchased commercially. Therefore, DART is a more useful method than the climbing assay for longitudinal assays of motor performance and will enable higher-throughput screen for genetic and pharmacological modifiers of neurodegeneration. In our proof-of-concept screen for modifiers of Aß-dependent phenotypes, we identified that in vivo knock-down of p53 in adult neurons is neuroprotective. This supports recent work targeting p53 in vitro and demonstrates the potential for DART to be used to screen for targets that ameliorate neurodegeneration.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Locomotion/physiology , Reflex, Startle/physiology , Software , Amyloid beta-Peptides/genetics , Animals , Drosophila
6.
Brain Commun ; 1(1): fcz005, 2019.
Article in English | MEDLINE | ID: mdl-32954257

ABSTRACT

DNA double-strand breaks are a feature of many acute and long-term neurological disorders, including neurodegeneration, following neurotrauma and after stroke. Persistent activation of the DNA damage response in response to double-strand breaks contributes to neural dysfunction and pathology as it can force post-mitotic neurons to re-enter the cell cycle leading to senescence or apoptosis. Mature, non-dividing neurons may tolerate low levels of DNA damage, in which case muting the DNA damage response might be neuroprotective. Here, we show that attenuating the DNA damage response by targeting the meiotic recombination 11, Rad50, Nijmegen breakage syndrome 1 complex, which is involved in double-strand break recognition, is neuroprotective in three neurodegeneration models in Drosophila and prevents Aß1-42-induced loss of synapses in embryonic hippocampal neurons. Attenuating the DNA damage response after optic nerve injury is also neuroprotective to retinal ganglion cells and promotes dramatic regeneration of their neurites both in vitro and in vivo. Dorsal root ganglion neurons similarly regenerate when the DNA damage response is targeted in vitro and in vivo and this strategy also induces significant restoration of lost function after spinal cord injury. We conclude that muting the DNA damage response in the nervous system is neuroprotective in multiple neurological disorders. Our results point to new therapies to maintain or repair the nervous system.

7.
Essays Biochem ; 61(6): 733-749, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29233882

ABSTRACT

The lysosome plays a pivotal role between catabolic and anabolic processes as the nexus for signalling pathways responsive to a variety of factors, such as growth, nutrient availability, energetic status and cellular stressors. Lysosomes are also the terminal degradative organelles for autophagy through which macromolecules and damaged cellular components and organelles are degraded. Autophagy acts as a cellular homeostatic pathway that is essential for organismal physiology. Decline in autophagy during ageing or in many diseases, including late-onset forms of neurodegeneration is considered a major contributing factor to the pathology. Multiple lines of evidence indicate that impairment in autophagy is also a central mechanism underlying several lysosomal storage disorders (LSDs). LSDs are a class of rare, inherited disorders whose histopathological hallmark is the accumulation of undegraded materials in the lysosomes due to abnormal lysosomal function. Inefficient degradative capability of the lysosomes has negative impact on the flux through the autophagic pathway, and therefore dysregulated autophagy in LSDs is emerging as a relevant disease mechanism. Pathology in the LSDs is generally early-onset, severe and life-limiting but current therapies are limited or absent; recognizing common autophagy defects in the LSDs raises new possibilities for therapy. In this review, we describe the mechanisms by which LSDs occur, focusing on perturbations in the autophagy pathway and present the latest data supporting the development of novel therapeutic approaches related to the modulation of autophagy.


Subject(s)
Autophagy/physiology , Lysosomal Storage Diseases/metabolism , Animals , Autophagy/genetics , Humans , Lysosomal Storage Diseases/genetics , Lysosomes/metabolism , Sphingolipidoses/metabolism
8.
Neurobiol Dis ; 103: 123-132, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28365214

ABSTRACT

The neuronal ceroid lipofuscinoses are a group of recessively inherited, childhood-onset neurodegenerative conditions. Several forms are caused by mutations in genes encoding putative lysosomal membrane proteins. Studies of the cell biology underpinning these disorders are hampered by the poor antigenicity of the membrane proteins, which makes visualization of the endogenous proteins difficult. We have used Drosophila to generate knock-in YFP-fusions for two of the NCL membrane proteins: CLN7 and CLN3. The YFP-fusions are expressed at endogenous levels and the proteins can be visualized live without the need for overexpression. Unexpectedly, both CLN7 and CLN3 have restricted expression in the CNS of Drosophila larva and are predominantly expressed in the glia that form the insect blood-brain-barrier. CLN7 is also expressed in neurons in the developing visual system. Analogous with murine CLN3, Drosophila CLN3 is strongly expressed in the excretory and osmoregulatory Malpighian tubules, but the knock-in also reveals unexpected localization of the protein to the apical domain adjacent to the lumen. In addition, some CLN3 protein in the tubules is localized within mitochondria. Our in vivo imaging of CLN7 and CLN3 suggests new possibilities for function and promotes new ideas about the cell biology of the NCLs.


Subject(s)
Blood-Brain Barrier/metabolism , Drosophila Proteins/biosynthesis , Malpighian Tubules/metabolism , Membrane Proteins/biosynthesis , Membrane Transport Proteins/biosynthesis , Neuronal Ceroid-Lipofuscinoses/metabolism , Neurons/metabolism , Animals , Blood-Brain Barrier/chemistry , Blood-Brain Barrier/ultrastructure , Drosophila , Drosophila Proteins/analysis , Drosophila Proteins/genetics , Gene Expression , Gene Knock-In Techniques , Malpighian Tubules/chemistry , Malpighian Tubules/ultrastructure , Membrane Proteins/analysis , Membrane Proteins/genetics , Membrane Transport Proteins/analysis , Membrane Transport Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Neurons/chemistry , Neurons/ultrastructure
9.
Biochim Biophys Acta ; 1852(10 Pt B): 2267-78, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25937302

ABSTRACT

The neuronal ceroid lipofuscinoses are a group of severe and progressive neurodegenerative disorders, generally with childhood onset. Despite the fact that these diseases remain fatal, significant breakthroughs have been made in our understanding of the genetics that underpin these conditions. This understanding has allowed the development of a broad range of models to study disease processes, and to develop new therapeutic approaches. Such models have contributed significantly to our knowledge of these conditions. In this review we will focus on the advantages of each individual model, describe some of the contributions the models have made to our understanding of the broader disease biology and highlight new techniques and approaches relevant to the study and potential treatment of the neuronal ceroid lipofuscinoses. This article is part of a Special Issue entitled: "Current Research on the Neuronal Ceroid Lipofuscinoses (Batten Disease)".

10.
Biol Open ; 3(1): 1-11, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24429107

ABSTRACT

Hyperphosphorylation of the microtubule associated protein, Tau, is the hallmark of a group of neurodegenerative disorders known as the tauopathies which includes Alzheimer's disease. Precisely how and why Tau phosphorylation is increased in disease is not fully understood, nor how individual sites modify Tau function. Several groups have used the Drosophila visual system as an in vivo model to examine how the toxicity of Tau varies with phosphorylation status. This system relies on overexpression of Tau from transgenes but is susceptible to position effects altering expression and activity of the transgenes. We have refined the system by eliminating position effects through the use of site-specific integration. By standardising Tau expression levels we have been able to compare directly the toxicity of different isoforms of Tau and Tau point mutants that abolish important phosphorylation events. We have also examined the importance of human kinases in modulating Tau toxicity in vivo. We were able to confirm that human GSK3ß phosphorylates Tau and increases toxicity but, unexpectedly, we identified that preventing phosphorylation of Ser404 is a protective event. When phosphorylation at this site is prevented, Tau toxicity in the Drosophila visual system is increased in the presence of GSK3ß. Our data suggest that not all phosphorylation events on Tau are associated with toxicity.

11.
Hum Mol Genet ; 20(10): 2037-47, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21372148

ABSTRACT

Mutations in the CLN3 gene cause juvenile neuronal ceroid lipofuscinosis (JNCL or Batten disease), an early onset neurodegenerative disorder. JNCL is the most common of the NCLs, a group of disorders with infant or childhood onset that are caused by single gene mutations. The NCLs, although relatively rare, share many pathological and clinical similarities with the more common late-onset neurodegenerative disorders, while their simple genetic basis makes them an excellent paradigm. The early onset and rapid disease progression in the NCLs suggests that one or more key cellular processes are severely compromised. To identify the functional pathways compromised in JNCL, we have performed a gain-of-function modifier screen in Drosophila. We find that CLN3 interacts genetically with the core stress signalling pathways and components of stress granules, suggesting a function in stress responses. In support of this, we find that Drosophila lacking CLN3 function are hypersensitive to oxidative stress yet they respond normally to other physiological stresses. Overexpression of CLN3 is sufficient to confer increased resistance to oxidative stress. We find that CLN3 mutant flies perceive conditions of increased oxidative stress correctly but are unable to detoxify reactive oxygen species, suggesting that their ability to respond is compromised. Together, our data suggest that the lack of CLN3 function leads to a failure to manage the response to oxidative stress and this may be the key deficit in JNCL that leads to neuronal degeneration.


Subject(s)
Drosophila , Membrane Glycoproteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/physiopathology , Oxidative Stress , Animals , Drosophila/genetics , Drosophila/metabolism , Female , Gene Expression Profiling , Genetic Testing , Male , Membrane Glycoproteins/metabolism , Mutation/genetics , Nerve Degeneration/genetics , Oxidants/pharmacology , Phenotype , Protein Binding , Protein Biosynthesis/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Transcription, Genetic
12.
Hum Mol Genet ; 18(4): 667-78, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19028667

ABSTRACT

Mutations in the gene CLN3 are responsible for the neurodegenerative disorder juvenile neuronal ceroid lipofuscinosis or Batten disease. CLN3 encodes a multi-spanning and hydrophobic transmembrane protein whose function is unclear. As a consequence, the cell biology that underlies the pathology of the disease is not well understood. We have developed a genetic gain-of-function system in Drosophila to identify functional pathways and interactions for CLN3. We have identified previously unknown interactions between CLN3 and the Notch and Jun N-terminal kinase signalling pathways and have uncovered a potential role for the RNA splicing and localization machinery in regulating CLN3 function.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , MAP Kinase Kinase 4/metabolism , Membrane Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Line , Drosophila/genetics , Drosophila Proteins/genetics , Eye/metabolism , Gene Expression , Humans , MAP Kinase Kinase 4/genetics , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Protein Binding , Protein Transport , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Notch/genetics
13.
Development ; 134(21): 3781-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17933789

ABSTRACT

Asymmetric cell divisions generate cell fate diversity during both invertebrate and vertebrate development. Drosophila neural progenitors or neuroblasts (NBs) each divide asymmetrically to produce a larger neuroblast and a smaller ganglion mother cell (GMC). The asymmetric localisation of neural cell fate determinants and their adapter proteins to the neuroblast cortex during mitosis facilitates their preferential segregation to the GMC upon cytokinesis. In this study we report a novel role for the anaphase-promoting complex/cyclosome (APC/C) during this process. Attenuation of APC/C activity disrupts the asymmetric localisation of the adapter protein Miranda and its associated cargo proteins Staufen, Prospero and Brat, but not other components of the asymmetric division machinery. We demonstrate that Miranda is ubiquitylated via its C-terminal domain; removal of this domain disrupts Miranda localisation and replacement of this domain with a ubiquitin moiety restores normal asymmetric Miranda localisation. Our results demonstrate that APC/C activity and ubiquitylation of Miranda are required for the asymmetric localisation of Miranda and its cargo proteins to the NB cortex.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Neurons/cytology , Neurons/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Animals, Genetically Modified , Apc5 Subunit, Anaphase-Promoting Complex-Cyclosome , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Division , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Juvenile Hormones/genetics , Juvenile Hormones/metabolism , Protein Binding , Substrate Specificity , Ubiquitin/metabolism , Ubiquitin-Protein Ligase Complexes/genetics
14.
J Biol Chem ; 280(28): 26557-64, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15826949

ABSTRACT

Myosin VII (M7) plays a role in adhesion in both Dictyostelium and mammalian cells where it is a component of a complex of proteins that serve to link membrane receptors to the underlying actin cytoskeleton. The nature of this complex is not fully known, prompting a search for M7-binding proteins. Co-immunoprecipitation experiments reveal that Dictyostelium M7 (DdM7) interacts with talinA, an actin-binding protein with a known role in cell-substrate adhesion. No additional proteins are observed in the immunoprecipitate, indicating that the interaction is direct. The N-terminal region of the DdM7 tail that lies between the region of predicted coil and the first MyTH4 domain is found to harbor the talinA binding site. Localization experiments reveal that talinA does not serve as a membrane receptor for DdM7 and vice versa. These findings reveal that talinA is a major DdM7 binding partner and suggest that their interaction induces a conformational change in each that, in combination with membrane receptor binding, promotes the assembly of a high avidity receptor complex essential for adhesion of the cell to substrata.


Subject(s)
Dictyostelium/metabolism , Myosins/chemistry , Protozoan Proteins/chemistry , Talin/chemistry , Actins/chemistry , Animals , Cell Adhesion , Cell Membrane/metabolism , Centrifugation, Density Gradient , Cytoskeleton/metabolism , Detergents/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Immunoprecipitation , Plasmids/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sucrose/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...