Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ella Bossy-Wetzel is active.

Publication


Featured researches published by Ella Bossy-Wetzel.


Nature Reviews Neuroscience | 2008

Mitochondrial fragmentation in neurodegeneration

Andrew B. Knott; Guy A. Perkins; Robert Schwarzenbacher; Ella Bossy-Wetzel

Mitochondria are remarkably dynamic organelles that migrate, divide and fuse. Cycles of mitochondrial fission and fusion ensure metabolite and mitochondrial DNA mixing and dictate organelle shape, number and bioenergetic functionality. There is mounting evidence that mitochondrial dysfunction is an early and causal event in neurodegeneration. Mutations in the mitochondrial fusion GTPases mitofusin 2 and optic atrophy 1, neurotoxins and oxidative stress all disrupt the cable-like morphology of functional mitochondria. This results in impaired bioenergetics and mitochondrial migration, and can trigger neurodegeneration. These findings suggest potential new treatment avenues for neurodegenerative diseases.


The EMBO Journal | 2006

Nitric oxide‐induced mitochondrial fission is regulated by dynamin‐related GTPases in neurons

Mark J Barsoum; Hua Yuan; Akos A Gerencser; Géraldine Liot; Yulia Kushnareva; Simone Gräber; Imre Kovacs; Wilson D Lee; Jenna Waggoner; Jiankun Cui; White Ad; Blaise Bossy; Jean-Claude Martinou; Richard J. Youle; Stuart A. Lipton; Mark H. Ellisman; Guy A. Perkins; Ella Bossy-Wetzel

Mitochondria are present as tubular organelles in neuronal projections. Here, we report that mitochondria undergo profound fission in response to nitric oxide (NO) in cortical neurons of primary cultures. Mitochondrial fission by NO occurs long before neurite injury and neuronal cell death. Furthermore, fission is accompanied by ultrastructural damage of mitochondria, autophagy, ATP decline and generation of free radicals. Fission is occasionally asymmetric and can be reversible. Strikingly, mitochondrial fission is also an early event in ischemic stroke in vivo. Mitofusin 1 (Mfn1) or dominant‐negative Dynamin related protein 1 (Drp1K38A) inhibits mitochondrial fission induced by NO, rotenone and Amyloid‐β peptide. Conversely, overexpression of Drp1 or Fis1 elicits fission and increases neuronal loss. Importantly, NO‐induced neuronal cell death was mitigated by Mfn1 and Drp1K38A. Thus, persistent mitochondrial fission may play a causal role in NO‐mediated neurotoxicity.


Neuron | 2004

Crosstalk between Nitric Oxide and Zinc Pathways to Neuronal Cell Death Involving Mitochondrial Dysfunction and p38-Activated K Channels

Ella Bossy-Wetzel; Maria Talantova; Wilson D Lee; Marion N. Schölzke; Anne Harrop; Emily Mathews; Thomas Götz; Jiahuai Han; Mark H. Ellisman; Guy A. Perkins; Stuart A. Lipton

Nitric oxide (NO) and zinc (Zn2+) are implicated in the pathogenesis of cerebral ischemia and neurodegenerative diseases. However, their relationship and the molecular mechanism of their neurotoxic effects remain unclear. Here we show that addition of exogenous NO or NMDA (to increase endogenous NO) leads to peroxynitrite (ONOO-) formation and consequent Zn2+ release from intracellular stores in cerebrocortical neurons. Free Zn2+ in turn induces respiratory block, mitochondrial permeability transition (mPT), cytochrome c release, generation of reactive oxygen species (ROS), and p38 MAP kinase activation. This pathway leads to caspase-independent K+ efflux with cell volume loss and apoptotic-like death. Moreover, Zn2+ chelators, ROS scavengers, Bcl-xL, dominant-interfering p38, or K+ channel blockers all attenuate NO-induced K+ efflux, cell volume loss, and neuronal apoptosis. Thus, these data establish a new form of crosstalk between NO and Zn2+ apoptotic signal transduction pathways that may contribute to neurodegeneration.


Nature Medicine | 2011

Mutant huntingtin binds the mitochondrial fission GTPase dynamin-related protein-1 and increases its enzymatic activity.

Wenjun Song; Jin-jin Chen; Alejandra M. Petrilli; Géraldine Liot; Eva Klinglmayr; Yue-Yue Zhou; Patrick Poquiz; Jonathan Tjong; Mahmoud A. Pouladi; Michael R. Hayden; Eliezer Masliah; Mark H. Ellisman; Isabelle Rouiller; Robert Schwarzenbacher; Blaise Bossy; Guy A. Perkins; Ella Bossy-Wetzel

Huntingtons disease is an inherited and incurable neurodegenerative disorder caused by an abnormal polyglutamine (polyQ) expansion in huntingtin (encoded by HTT). PolyQ length determines disease onset and severity, with a longer expansion causing earlier onset. The mechanisms of mutant huntingtin-mediated neurotoxicity remain unclear; however, mitochondrial dysfunction is a key event in Huntingtons disease pathogenesis. Here we tested whether mutant huntingtin impairs the mitochondrial fission-fusion balance and thereby causes neuronal injury. We show that mutant huntingtin triggers mitochondrial fragmentation in rat neurons and fibroblasts of individuals with Huntingtons disease in vitro and in a mouse model of Huntingtons disease in vivo before the presence of neurological deficits and huntingtin aggregates. Mutant huntingtin abnormally interacts with the mitochondrial fission GTPase dynamin-related protein-1 (DRP1) in mice and humans with Huntingtons disease, which, in turn, stimulates its enzymatic activity. Mutant huntingtin–mediated mitochondrial fragmentation, defects in anterograde and retrograde mitochondrial transport and neuronal cell death are all rescued by reducing DRP1 GTPase activity with the dominant-negative DRP1 K38A mutant. Thus, DRP1 might represent a new therapeutic target to combat neurodegeneration in Huntingtons disease.


Annals of the New York Academy of Sciences | 2008

Impairing the Mitochondrial Fission and Fusion Balance: A New Mechanism of Neurodegeneration

Andrew B. Knott; Ella Bossy-Wetzel

Mitochondrial dysfunction is a common characteristic of all neurodegenerative diseases. However, the cause of this dysfunction remains a mystery. Here, we discuss the potential role of mitochondrial fission and fusion in the onset and progression of neurodegenerative diseases. Specifically, we propose that an imbalance in mitochondrial fission and fusion may underlie both familial and sporadic neurodegenerative disorders. There is substantial evidence that links disruption of the mitochondrial fission and fusion equilibrium, resulting in abnormally long or short mitochondria, to neurodegeneration. First, hereditary mutations in the mitochondrial fusion GTPases optic atrophy‐1 and mitofusin‐2 cause neuropathies in humans. In addition, recent findings report increased mitochondrial fission in Parkinsons disease (PD) models and induction of mitochondrial fission by two proteins, PTEN‐induced kinase 1 and parkin, which are mutant in familial forms of PD. Furthermore, mutant huntingtin, the disease‐causing protein in Huntingtons disease, alters mitochondrial morphology and dynamics. Rotenone, a pesticide and inducer of PD symptoms, and amyloid‐β peptide, which is causally linked to Alzheimers disease, initiate mitochondrial fission. Finally, mitochondrial fission is an early event in ischemic stroke and diabetic neuropathies. In sum, a growing body of research suggests that a better understanding of mitochondrial fission and fusion and the regulatory factors involved may lead to improved treatments and cures for neurodegenerative diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Bcl-xL induces Drp1-dependent synapse formation in cultured hippocampal neurons

Hongmei Li; Yingbei Chen; Adrienne Jones; Richard H. Sanger; Leon P. Collis; Richard J. Flannery; Ewan C. McNay; Tingxi Yu; Robert Schwarzenbacher; Blaise Bossy; Ella Bossy-Wetzel; Marc Pypaert; John Hickman; Peter J. Smith; J. Marie Hardwick; Elizabeth A. Jonas

Maturation of neuronal synapses is thought to involve mitochondria. Bcl-xL protein inhibits mitochondria-mediated apoptosis but may have other functions in healthy adult neurons in which Bcl-xL is abundant. Here, we report that overexpression of Bcl-xL postsynaptically increases frequency and amplitude of spontaneous miniature synaptic currents in rat hippocampal neurons in culture. Bcl-xL, overexpressed either pre or postsynaptically, increases synapse number, the number and size of synaptic vesicle clusters, and mitochondrial localization to vesicle clusters and synapses, likely accounting for the changes in miniature synaptic currents. Conversely, knockdown of Bcl-xL or inhibiting it with ABT-737 decreases these morphological parameters. The mitochondrial fission protein, dynamin-related protein 1 (Drp1), is a GTPase known to localize to synapses and affect synaptic function and structure. The effects of Bcl-xL appear mediated through Drp1 because overexpression of Drp1 increases synaptic markers, and overexpression of the dominant-negative dnDrp1-K38A decreases them. Furthermore, Bcl-xL coimmunoprecipitates with Drp1 in tissue lysates, and in a recombinant system, Bcl-xL protein stimulates GTPase activity of Drp1. These findings suggest that Bcl-xL positively regulates Drp1 to alter mitochondrial function in a manner that stimulates synapse formation.


Cell Death & Differentiation | 2009

Complex II inhibition by 3-NP causes mitochondrial fragmentation and neuronal cell death via an NMDA- and ROS-dependent pathway.

Géraldine Liot; Blaise Bossy; Steven A. Lubitz; Y Kushnareva; N Sejbuk; Ella Bossy-Wetzel

Mitochondrial respiratory complex II inhibition plays a central role in Huntingtons disease (HD). Remarkably, 3-NP, a complex II inhibitor, recapitulates HD-like symptoms. Furthermore, decreases in mitochondrial fusion or increases in mitochondrial fission have been implicated in neurodegenerative diseases. However, the relationship between mitochondrial energy defects and mitochondrial dynamics has never been explored in detail. In addition, the mechanism of neuronal cell death by complex II inhibition remains unclear. Here, we tested the temporal and spatial relationship between energy decline, impairment of mitochondrial dynamics, and neuronal cell death in response to 3-NP using quantitative fluorescence time-lapse microscopy and cortical neurons. 3-NP caused an immediate drop in ATP. This event corresponded with a mild rise in reactive oxygen species (ROS), but mitochondrial morphology remained unaltered. Unexpectedly, several hours after this initial phase, a second dramatic rise in ROS occurred, associated with profound mitochondrial fission characterized by the conversion of filamentous to punctate mitochondria and neuronal cell death. Glutamate receptor antagonist AP5 abolishes the second peak in ROS, mitochondrial fission, and cell death. Thus, secondary excitotoxicity, mediated by glutamate receptor activation of the NMDA subtype, and consequent oxidative and nitrosative stress cause mitochondrial fission, rather than energy deficits per se. These results improve our understanding of the cellular mechanisms underlying HD pathogenesis.


Cell | 2002

Dueling Activities of AIF in Cell Death versus Survival: DNA Binding and Redox Activity

Stuart A. Lipton; Ella Bossy-Wetzel

Apoptosis-inducing factor (AIF) was originally discovered as a mitochondrial protein that, like cytochrome c, is released into the cytoplasm during cell death. New evidence suggests, however, that a redox-active enzymatic region of AIF may be antiapoptotic while a DNA binding region is proapoptotic.


Trends in Neurosciences | 2008

Mutant huntingtin and mitochondrial dysfunction

Ella Bossy-Wetzel; Alejandra M. Petrilli; Andrew B. Knott

Huntingtons disease (HD) is a fatal, inherited neurodegenerative disorder that gradually robs affected individuals of memory, cognitive skills and normal movements. Although research has identified a single faulty gene, the huntingtin gene, as the cause of the disease, a cure remains elusive. Strong evidence indicates that mitochondrial impairment plays a key part in HD pathogenesis. Here, we highlight how mutant huntingtin (mtHtt) might cause mitochondrial dysfunction by either perturbing transcription of nuclear-encoded mitochondrial proteins or by direct interaction with the organelle and modulation of respiration, mitochondrial membrane potential and Ca(2+) buffering. In addition, we propose that mtHtt might convey its neurotoxicity by evoking defects in mitochondrial dynamics, organelle trafficking and fission and fusion, which, in turn, might result in bioenergetic failure and HD-linked neuronal dysfunction and cell death. Finally, we speculate how mitochondria might dictate selective vulnerability of long projection neurons, such as medium spiny neurons, which are particularly affected in HD.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Dominant-interfering forms of MEF2 generated by caspase cleavage contribute to NMDA-induced neuronal apoptosis

Shu-ichi Okamoto; Zhen Li; Chung Ju; Marion N. Schölzke; Emily Mathews; Jiankun Cui; Guy S. Salvesen; Ella Bossy-Wetzel; Stuart A. Lipton

Myocyte enhancer factor-2 (MEF2) transcription factors are activated by p38 mitogen-activated protein kinase during neuronal and myogenic differentiation. Recent work has shown that stimulation of this pathway is antiapoptotic during development but proapoptotic in mature neurons exposed to excitotoxic or other stress. We now report that excitotoxic (N-methyl-D-aspartate) insults to mature cerebrocortical neurons activate caspase-3, -7, in turn cleaving MEF2A, C, and D isoforms. MEF2 cleavage fragments containing a truncated transactivation domain but preserved DNA-binding domain block MEF2 transcriptional activity via dominant interference. Transfection of constitutively active MEF2 (MEF2C-CA) rescues MEF2 transcriptional activity after N-methyl-D-aspartate insult and prevents neuronal apoptosis. Conversely, dominant-interfering MEF2 abrogates neuroprotection by MEF2C-CA. These results define a pathway to excitotoxic neuronal stress/apoptosis via caspase-catalyzed cleavage of MEF2. Additionally, we show that similar MEF2 cleavage fragments are generated in vivo during focal stroke damage. Hence, this pathway appears to have pathophysiological relevance in vivo.

Collaboration


Dive into the Ella Bossy-Wetzel's collaboration.

Top Co-Authors

Avatar

Douglas R. Green

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Shailaja Kasibhatla

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deborah Finucane

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Guy A. Perkins

University of California

View shared research outputs
Top Co-Authors

Avatar

Blaise Bossy

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew B. Knott

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge