Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1579-1591, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30904609

ABSTRACT

Evolutionarily conserved homeostatic systems have been shown to modulate synaptic efficiency at the neuromuscular junctions of organisms. While advances have been made in identifying molecules that function presynaptically during homeostasis, limited information is currently available on how postsynaptic alterations affect presynaptic function. We previously identified a role for postsynaptic Dystrophin in the maintenance of evoked neurotransmitter release. We herein demonstrated that Dystrobrevin, a member of the Dystrophin Glycoprotein Complex, was delocalized from the postsynaptic region in the absence of Dystrophin. A newly-generated Dystrobrevin mutant showed elevated evoked neurotransmitter release, increased bouton numbers, and a readily releasable pool of synaptic vesicles without changes in the function or numbers of postsynaptic glutamate receptors. In addition, we provide evidence to show that the highly conserved Cdc42 Rho GTPase plays a key role in the postsynaptic Dystrophin/Dystrobrevin pathway for synaptic homeostasis. The present results give novel insights into the synaptic deficits underlying Duchenne Muscular Dystrophy affected by a dysfunctional Dystrophin Glycoprotein complex.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Dystrophin-Associated Proteins/genetics , Dystrophin/genetics , Neuromuscular Junction/genetics , cdc42 GTP-Binding Protein/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Dystrophin/deficiency , Dystrophin-Associated Proteins/metabolism , Gene Expression Regulation , Homeostasis/genetics , Humans , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Synaptic Potentials/genetics , Synaptic Transmission , Synaptic Vesicles/metabolism , cdc42 GTP-Binding Protein/metabolism
2.
Cancer Nanotechnol ; 7: 4, 2016.
Article in English | MEDLINE | ID: mdl-27358656

ABSTRACT

On March 4th, 2016, Springer's Cancer Nanotechnology office promoted the launch of the Nanotechnology Startup Challenge in Cancer (NSC2 ). This innovation-development model is a partnership among our company, the Center for Advancing Innovation (CAI), MedImmune, the global biologics arm of AstraZeneca, and multiple institutes at the National Institutes of Health (NIH). NSC2 "crowdsources" talent from around the world to launch startups with near-term, commercially viable cancer nanotechnology inventions, which were developed by the National Cancer Institute (NCI), the National Heart, Lung and Blood Institute (NHLBI), and the National Institute of Biomedical Imaging and Bioengineering (NIBIB). Crowdsourcing is a process in which one uses the internet to engage a large group of people in an activity, such as NSC2 . For this initiative, CAI engaged universities, industry professionals, foundations, investors, relevant media outlets, seasoned entrepreneurs, and life sciences membership organizations to request that they participate in the challenge. From this outreach, fifty-six key thought leaders have enrolled in NSC2 as judges, mentors, and/or advisors to challenge teams (http://www.nscsquared.org/judges.html). Along with crowdsourcing talent to bolt startups around NIH inventions, NSC2 will also catalyze the launch of companies around "third-party" cancer nanotechnology inventions, which were conceived and developed outside of the NIH. Twenty-eight robust teams were accepted to the challenge on March 14th, 2016.

3.
Neuron ; 72(5): 760-75, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-22153373

ABSTRACT

We provide evidence for a prodegenerative, glial-derived signaling framework in the Drosophila neuromuscular system that includes caspase and mitochondria-dependent signaling. We demonstrate that Drosophila TNF-α (eiger) is expressed in a subset of peripheral glia, and the TNF-α receptor (TNFR), Wengen, is expressed in motoneurons. NMJ degeneration caused by disruption of the spectrin/ankyrin skeleton is suppressed by an eiger mutation or by eiger knockdown within a subset of peripheral glia. Loss of wengen in motoneurons causes a similar suppression providing evidence for glial-derived prodegenerative TNF-α signaling. Neither JNK nor NFκß is required for prodegenerative signaling. However, we provide evidence for the involvement of both an initiator and effector caspase, Dronc and Dcp-1, and mitochondrial-dependent signaling. Mutations that deplete the axon and nerve terminal of mitochondria suppress degeneration as do mutations in Drosophila Bcl-2 (debcl), a mitochondria-associated protein, and Apaf-1 (dark), which links mitochondrial signaling with caspase activity in other systems.


Subject(s)
Motor Neuron Disease/pathology , Nerve Degeneration/pathology , Neuroglia/physiology , Neuromuscular Junction/pathology , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Caspases/genetics , Caspases/metabolism , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Excitatory Postsynaptic Potentials/genetics , Fluorescence Recovery After Photobleaching/methods , Green Fluorescent Proteins/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/ultrastructure , Motor Neuron Disease/genetics , Mutation/genetics , Nerve Degeneration/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/ultrastructure , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference/physiology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/genetics
4.
J Cell Biol ; 194(6): 921-35, 2011 Sep 19.
Article in English | MEDLINE | ID: mdl-21930778

ABSTRACT

The dimensions of neuronal dendrites, axons, and synaptic terminals are reproducibly specified for each neuron type, yet it remains unknown how these structures acquire their precise dimensions of length and diameter. Similarly, it remains unknown how active zone number and synaptic strength are specified relative the precise dimensions of presynaptic boutons. In this paper, we demonstrate that S6 kinase (S6K) localizes to the presynaptic active zone. Specifically, S6K colocalizes with the presynaptic protein Bruchpilot (Brp) and requires Brp for active zone localization. We then provide evidence that S6K functions downstream of presynaptic PDK1 to control synaptic bouton size, active zone number, and synaptic function without influencing presynaptic bouton number. We further demonstrate that PDK1 is also a presynaptic protein, though it is distributed more broadly. We present a model in which synaptic S6K responds to local extracellular nutrient and growth factor signaling at the synapse to modulate developmental size specification, including cell size, bouton size, active zone number, and neurotransmitter release.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases/analysis , Synapses/enzymology , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Animals, Genetically Modified , Drosophila/embryology , Drosophila/metabolism , Immunohistochemistry , Microscopy, Electron , Presynaptic Terminals , Protein Serine-Threonine Kinases/genetics , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Synaptic Transmission/physiology
5.
Genetics ; 183(4): 1357-72, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19797046

ABSTRACT

The nerve-cell cytoskeleton is essential for the regulation of intrinsic neuronal activity. For example, neuronal migration defects are associated with microtubule regulators, such as LIS1 and dynein, as well as with actin regulators, including Rac GTPases and integrins, and have been thought to underlie epileptic seizures in patients with cortical malformations. However, it is plausible that post-developmental functions of specific cytoskeletal regulators contribute to the more transient nature of aberrant neuronal activity and could be masked by developmental anomalies. Accordingly, our previous results have illuminated functional roles, distinct from developmental contributions, for Caenorhabditis elegans orthologs of LIS1 and dynein in GABAergic synaptic vesicle transport. Here, we report that C. elegans with function-altering mutations in canonical Rac GTPase-signaling-pathway members demonstrated a robust behavioral response to a GABA(A) receptor antagonist, pentylenetetrazole. Rac mutants also exhibited hypersensitivity to an acetylcholinesterase inhibitor, aldicarb, uncovering deficiencies in inhibitory neurotransmission. RNA interference targeting Rac hypomorphs revealed synergistic interactions between the dynein motor complex and some, but not all, members of Rac-signaling pathways. These genetic interactions are consistent with putative Rac-dependent regulation of actin and microtubule networks and suggest that some cytoskeletal regulators cooperate to uniquely govern neuronal synchrony through dynein-mediated GABAergic vesicle transport in C. elegans.


Subject(s)
Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Pharmacogenetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , gamma-Aminobutyric Acid/metabolism , rac GTP-Binding Proteins/metabolism , Aldicarb/pharmacology , Animals , Biological Transport/drug effects , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Dyneins/metabolism , Integrins/metabolism , Larva/drug effects , Larva/genetics , Larva/growth & development , Larva/physiology , Male , Microtubule-Associated Proteins/metabolism , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/metabolism , Mutation , Pentylenetetrazole/pharmacology , RNA Interference , Seizures/chemically induced , Seizures/metabolism , Seizures/pathology , Seizures/physiopathology , Synaptic Vesicles/drug effects , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism
6.
J Vis Exp ; (18)2008 Aug 18.
Article in English | MEDLINE | ID: mdl-19066504

ABSTRACT

The nematode, Caenorhabditis elegans, has become an expedient model for studying neurotransmission. C. elegans is unique among animal models, as the anatomy and connectivity of its nervous system has been determined from electron micrographs and refined by pharmacological assays. In this video, we describe how two complementary neural stimulants, an acetylcholinesterase inhibitor, called aldicarb, and a gamma-aminobutyric acid (GABA) receptor antagonist, called pentylenetetrazole (PTZ), may be employed to specifically characterize signaling at C. elegans neuromuscular junctions (NMJs) and facilitate our understanding of antagonistic neural circuits. Of 302 C. elegans neurons, nineteen GABAergic D-type motor neurons innervate body wall muscles (BWMs), while four GABAergic neurons, called RMEs, innervate head muscles. Conversely, thirty-nine motor neurons express the excitatory neurotransmitter, acetylcholine (ACh), and antagonize GABA transmission at BWMs to coordinate locomotion. The antagonistic nature of GABAergic and cholinergic motor neurons at body wall NMJs was initially determined by laser ablation and later buttressed by aldicarb exposure. Acute aldicarb exposure results in a time-course or dose-responsive paralysis in wild-type worms. Yet, loss of excitatory ACh transmission confers resistance to aldicarb, as less ACh accumulates at worm NMJs, leading to less stimulation of BWMs. Resistance to aldicarb may be observed with ACh-specific or general synaptic function mutants. Consistent with antagonistic GABA and ACh transmission, loss of GABA transmission, or a failure to negatively regulate ACh release, confers hypersensitivity to aldicarb. Although aldicarb exposure has led to the isolation of numerous worm homologs of neurotransmission genes, aldicarb exposure alone cannot efficiently determine prevailing roles for genes and pathways in specific C. elegans motor neurons. For this purpose, we have introduced a complementary experimental approach, which uses PTZ. Neurotransmission mutants display clear phenotypes, distinct from aldicarb-induced paralysis, in response to PTZ. Wild-type worms, as well as mutants with specific inabilities to release or receive ACh, do not show apparent sensitivity to PTZ. However, GABA mutants, as well as general synaptic function mutants, display anterior convulsions in a time-course or dose-responsive manner. Mutants that cannot negatively regulate general neurotransmitter release and, thus, secrete excessive amounts of ACh onto BWMs, become paralyzed on PTZ. The PTZ-induced phenotypes of discrete mutant classes indicate that a complementary approach with aldicarb and PTZ exposure paradigms in C. elegans may accelerate our understanding of neurotransmission. Moreover, videos demonstrating how we perform pharmacological assays should establish consistent methods for C. elegans research.


Subject(s)
Aldicarb/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Mutation , Pentylenetetrazole/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Animals , Caenorhabditis elegans/genetics , Cholinesterase Inhibitors/pharmacology , GABA Antagonists/pharmacology , Models, Animal , Synaptic Transmission/genetics
7.
Neurosci Lett ; 439(2): 129-33, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18514411

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder with approximately 2% of people over age 65 suffering from this disease. Risk factors for PD involve interplay between still poorly defined genetic and non-genetic contributors, but appear to converge upon cellular pathways that mediate protein misfolding and oxidative stress that lead to dopaminergic neuron loss. The identification of either new or repurposed drugs that exhibit benefit in slowing the age-dependent neuronal damage that occurs in PD is a significant goal of much ongoing research. We have exploited the nematode Caenorhabditis elegans as a model system by which the neuroprotective capacity of acetaminophen could be rapidly evaluated for efficacy in attenuating dopamine (DA) neurodegeneration. Using three independent and established neurodegenerative models in C. elegans, we assayed for acetaminophen-dependent rescue in response to: (1) over-expression of the PD-associated protein, alpha-synuclein; (2) acute exposure to 6-hydroxydopamine (6-OHDA); (3) excess intracellular DA production due to over-expression of the DA biosynthetic enzyme, tyrosine hydroxylase (TH). These data suggest that acetaminophen significantly protected C. elegans DA neurons from stressors related to oxidative damage, but not protein misfolding. Taken together, these studies imply an activity for acetaminophen in the attenuation of DA neuron loss that, following essential corroborative analyses in mammalian systems, may represent a potential benefit for PD.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Dopamine/metabolism , Neurodegenerative Diseases/pathology , Neurons/drug effects , Adrenergic Agents/toxicity , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Cell Count/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Neurodegenerative Diseases/drug therapy , Oxidopamine/toxicity , Parkinson Disease/complications , Parkinson Disease/etiology , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
8.
Brain Res ; 1120(1): 23-34, 2006 Nov 20.
Article in English | MEDLINE | ID: mdl-16996038

ABSTRACT

Epilepsy is estimated to affect 1-2% of the world population, yet remains poorly understood at a molecular level. We have previously established the roundworm Caenorhabditis elegans as a model for investigating genetic susceptibilities to seizure-like convulsions in vivo. Here we investigate the behavioral consequences of decreasing the activity of nematode gene homologs within the LIS1 pathway that are associated with a human cortical malformation termed lissencephaly. Bioinformatic analysis revealed the nud-2 gene, encoding the worm homolog of mammalian effectors of LIS1, termed NDE1 and NDEL1. Phenotypic analysis of animals targeted by RNA interference (RNAi) was performed using a pentylenetetrazole (PTZ) exposure paradigm to induce convulsions. Worms depleted for LIS1 pathway components (NUD-1, NUD-2, DHC-1, CDK-5, and CDKA-1) exhibited significant convulsions following PTZ and RNAi treatment. Strains harboring fluorescent markers for GABAergic neuronal architecture and synaptic vesicle trafficking were employed to discern putative mechanisms accounting for observed convulsion behaviors. We found that depletion of LIS1 pathway components resulted in defective GABA synaptic vesicle trafficking. We also utilized combinations of specific genetic backgrounds to create a sensitized state for convulsion susceptibility and discovered that convulsion effects were significantly enhanced when LIS-1 and other pathway components were compromised within the same animals. Thus, interactions among gene products with LIS-1 may mediate intrinsic thresholds of neuronal synchrony.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Disease Susceptibility , Microtubule-Associated Proteins/genetics , Seizures/genetics , Animals , Animals, Genetically Modified , Behavior, Animal , Caenorhabditis elegans , Caenorhabditis elegans Proteins/chemistry , Cell Movement/drug effects , Cell Movement/physiology , Computational Biology/methods , Dose-Response Relationship, Drug , Gene Expression/physiology , Green Fluorescent Proteins/biosynthesis , Microinjections/methods , Microtubule-Associated Proteins/chemistry , Models, Biological , Molecular Sequence Data , Pentylenetetrazole , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction/methods , Seizures/chemically induced , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , gamma-Aminobutyric Acid/metabolism
9.
Hum Mol Genet ; 13(18): 2043-59, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15254012

ABSTRACT

Cortical malformations are a collection of disorders affecting brain development. Mutations in the LIS1 gene lead to a disorganized and smooth cerebral cortex caused by failure in neuronal migration. Among the clinical consequences of lissencephaly are mental retardation and intractable epilepsy. It remains unclear whether the seizures result from aberrant neuronal placement, disruption of intrinsic properties of neurons, or both. The nematode Caenorhabditis elegans offers an opportunity to study such convulsions in a simple animal with a defined nervous system. Here we show that convulsions mimicking epilepsy can be induced by a mutation in a C. elegans lis-1 allele (pnm-1), in combination with a chemical antagonist of gamma-aminobutyric acid (GABA) neurotransmitter signaling. Identical convulsions were obtained using C. elegans mutants defective in GABA transmission, whereas none of these mutants or the antagonist alone caused convulsions, indicating a threshold was exceeded in response to this combination. Crosses between pnm-1 and fluorescent marker strains designed to exclusively illuminate either the processes of GABAergic neurons or synaptic vesicles surprisingly showed no deviations in neuronal architecture. Instead, presynaptic defects in GABAergic vesicle distribution were clearly evident and could be phenocopied by RNAi directed against cytoplasmic dynein, a known LIS1 interactor. Furthermore, mutations in UNC-104, a neuronal-specific kinesin, and SNB-1, a synaptic vesicle-associated protein termed synaptobrevin, exhibit similar convulsion phenotypes following chemical induction. Taken together, these studies establish C. elegans as a system to investigate subtle cytoskeletal mechanisms regulating intrinsic neuronal activity and suggest that it may be possible to dissociate the epileptic consequences of lissencephaly from the more phenotypically overt cortical defects associated with neuronal migration.


Subject(s)
Caenorhabditis elegans/genetics , Cytoskeleton/physiology , Epilepsy/etiology , Microtubule-Associated Proteins/genetics , gamma-Aminobutyric Acid/physiology , Alleles , Amino Acid Sequence , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins , Codon, Nonsense/genetics , Disease Models, Animal , Epilepsy/genetics , GABA Antagonists/pharmacology , Molecular Sequence Data , Neurons/cytology , Pentylenetetrazole/pharmacology , RNA, Small Interfering/genetics , Seizures/etiology , Seizures/genetics , Sequence Alignment , Synaptic Vesicles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...