Network


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

Hotspot


Dive into the research topics where Gaia Skibinski is active.

Publication


Featured researches published by Gaia Skibinski.


Journal of Biological Chemistry | 2011

Direct Membrane Association Drives Mitochondrial Fission by the Parkinson Disease-associated Protein α-Synuclein

Ken Nakamura; Venu M. Nemani; Farnaz Azarbal; Gaia Skibinski; Jon M. Levy; Kiyoshi Egami; Larissa A. Munishkina; Jue Zhang; Brooke M. Gardner; Junko Wakabayashi; Hiromi Sesaki; Yifan Cheng; Steven Finkbeiner; Robert L. Nussbaum; Eliezer Masliah; Robert H. Edwards

The protein α-synuclein has a central role in Parkinson disease, but the mechanism by which it contributes to neural degeneration remains unknown. We now show that the expression of α-synuclein in mammalian cells, including neurons in vitro and in vivo, causes the fragmentation of mitochondria. The effect is specific for synuclein, with more fragmentation by α- than β- or γ-isoforms, and it is not accompanied by changes in the morphology of other organelles or in mitochondrial membrane potential. However, mitochondrial fragmentation is eventually followed by a decline in respiration and neuronal death. The fragmentation does not require the mitochondrial fission protein Drp1 and involves a direct interaction of synuclein with mitochondrial membranes. In vitro, synuclein fragments artificial membranes containing the mitochondrial lipid cardiolipin, and this effect is specific for the small oligomeric forms of synuclein. α-Synuclein thus exerts a primary and direct effect on the morphology of an organelle long implicated in the pathogenesis of Parkinson disease.


The Journal of Neuroscience | 2010

Cytoplasmic Mislocalization of TDP-43 Is Toxic to Neurons and Enhanced by a Mutation Associated with Familial Amyotrophic Lateral Sclerosis

Sami J. Barmada; Gaia Skibinski; Erica Korb; Elizabeth J. Rao; Jane Y. Wu; Steven Finkbeiner

Mutations in the gene encoding TDP-43—the major protein component of neuronal aggregates characteristic of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) with ubiquitin-positive inclusion bodies—have been linked to familial forms of both disorders. Aggregates of TDP-43 in cortical and spinal motorneurons in ALS, or in neurons of the frontal and temporal cortices in FTLD, are closely linked to neuron loss and atrophy in these areas. However, the mechanism by which TDP-43 mutations lead to neurodegeneration is unclear. To investigate the pathogenic role of TDP-43 mutations, we established a model of TDP-43 proteinopathies by expressing fluorescently tagged wild-type and mutant TDP-43 in primary rat cortical neurons. Expression of mutant TDP-43 was toxic to neurons, and mutant-specific toxicity was associated with increased cytoplasmic mislocalization of TDP-43. Inclusion bodies were not necessary for the toxicity and did not affect the risk of cell death. Cellular survival was unaffected by the total amount of exogenous TDP-43 in the nucleus, but the amount of cytoplasmic TDP-43 was a strong and independent predictor of neuronal death. These results suggest that mutant TDP-43 is mislocalized to the cytoplasm, where it exhibits a toxic gain-of-function and induces cell death.


The Journal of Neuroscience | 2014

Mutant LRRK2 Toxicity in Neurons Depends on LRRK2 Levels and Synuclein But Not Kinase Activity or Inclusion Bodies

Gaia Skibinski; Ken Nakamura; Mark R. Cookson; Steven Finkbeiner

By combining experimental neuron models and mathematical tools, we developed a “systems” approach to deconvolve cellular mechanisms of neurodegeneration underlying the most common known cause of Parkinsons disease (PD), mutations in leucine-rich repeat kinase 2 (LRRK2). Neurons ectopically expressing mutant LRRK2 formed inclusion bodies (IBs), retracted neurites, accumulated synuclein, and died prematurely, recapitulating key features of PD. Degeneration was predicted from the levels of diffuse mutant LRRK2 that each neuron contained, but IB formation was neither necessary nor sufficient for death. Genetic or pharmacological blockade of its kinase activity destabilized LRRK2 and lowered its levels enough to account for the moderate reduction in LRRK2 toxicity that ensued. By contrast, targeting synuclein, including neurons made from PD patient-derived induced pluripotent cells, dramatically reduced LRRK2-dependent neurodegeneration and LRRK2 levels. These findings suggest that LRRK2 levels are more important than kinase activity per se in predicting toxicity and implicate synuclein as a major mediator of LRRK2-induced neurodegeneration.


PLOS ONE | 2014

Targeting the Intrinsically Disordered Structural Ensemble of α-Synuclein by Small Molecules as a Potential Therapeutic Strategy for Parkinson’s Disease

Gergely Toth; Shyra J. Gardai; Wagner Zago; Carlos W. Bertoncini; Nunilo Cremades; Susan L. Roy; Mitali A. Tambe; Jean-Christophe Rochet; Céline Galvagnion; Gaia Skibinski; Steven Finkbeiner; Michael P. Bova; Karin Regnstrom; San-San Chiou; Jennifer J. Johnston; Kari Callaway; John P. Anderson; Michael F. Jobling; Alexander K. Buell; Ted Yednock; Tuomas P. J. Knowles; Michele Vendruscolo; John Christodoulou; Christopher M. Dobson; Dale Schenk; Lisa McConlogue

The misfolding of intrinsically disordered proteins such as α-synuclein, tau and the Aβ peptide has been associated with many highly debilitating neurodegenerative syndromes including Parkinson’s and Alzheimer’s diseases. Therapeutic targeting of the monomeric state of such intrinsically disordered proteins by small molecules has, however, been a major challenge because of their heterogeneous conformational properties. We show here that a combination of computational and experimental techniques has led to the identification of a drug-like phenyl-sulfonamide compound (ELN484228), that targets α-synuclein, a key protein in Parkinson’s disease. We found that this compound has substantial biological activity in cellular models of α-synuclein-mediated dysfunction, including rescue of α-synuclein-induced disruption of vesicle trafficking and dopaminergic neuronal loss and neurite retraction most likely by reducing the amount of α-synuclein targeted to sites of vesicle mobilization such as the synapse in neurons or the site of bead engulfment in microglial cells. These results indicate that targeting α-synuclein by small molecules represents a promising approach to the development of therapeutic treatments of Parkinson’s disease and related conditions.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Nrf2 mitigates LRRK2- and α-synuclein–induced neurodegeneration by modulating proteostasis

Gaia Skibinski; Vicky Hwang; Dale Michael Ando; Aaron Daub; Alicia K. Lee; Abinaya Ravisankar; Sara Modan; Mariel M. Finucane; Benjamin A. Shaby; Steven Finkbeiner

Significance The prevailing view of nuclear factor erythroid 2-related factor (Nrf2) function in the central nervous system is that it acts by a cell-nonautonomous mechanism to activate a program of gene expression that mitigates reactive oxygen species and the damage that ensues. Our work significantly expands the biological understanding of Nrf2 by showing that Nrf2 mitigates toxicity induced by α-synuclein and leucine-rich repeat kinase 2 (LRRK2), by potently promoting neuronal protein homeostasis in a cell-autonomous and time-dependent fashion. Nrf2 accelerates the clearance of α-synuclein, shortening its half-life and leading to lower overall levels of α-synuclein. By contrast, Nrf2 promotes the aggregation of LRRK2 into inclusion bodies, leading to a significant reduction in diffuse mutant LRRK2 levels elsewhere in the neuron. Mutations in leucine-rich repeat kinase 2 (LRRK2) and α-synuclein lead to Parkinson’s disease (PD). Disruption of protein homeostasis is an emerging theme in PD pathogenesis, making mechanisms to reduce the accumulation of misfolded proteins an attractive therapeutic strategy. We determined if activating nuclear factor erythroid 2-related factor (Nrf2), a potential therapeutic target for neurodegeneration, could reduce PD-associated neuron toxicity by modulating the protein homeostasis network. Using a longitudinal imaging platform, we visualized the metabolism and location of mutant LRRK2 and α-synuclein in living neurons at the single-cell level. Nrf2 reduced PD-associated protein toxicity by a cell-autonomous mechanism that was time-dependent. Furthermore, Nrf2 activated distinct mechanisms to handle different misfolded proteins. Nrf2 decreased steady-state levels of α-synuclein in part by increasing α-synuclein degradation. In contrast, Nrf2 sequestered misfolded diffuse LRRK2 into more insoluble and homogeneous inclusion bodies. By identifying the stress response strategies activated by Nrf2, we also highlight endogenous coping responses that might be therapeutically bolstered to treat PD.


International Journal of High Throughput Screening | 2011

Drug discovery in Parkinson's disease-Update and developments in the use of cellular models.

Gaia Skibinski; Steven Finkbeiner

Parkinsons disease (PD) is the second most common neurodegenerative disorder and is characterized by the degeneration of dopaminergic (DA) neurons within the substantia nigra. Dopamine replacement drugs remain the most effective PD treatment but only provide temporary symptomatic relief. New therapies are urgently needed, but the search for a disease-modifying treatment and a definitive understanding of the underlying mechanisms of PD has been limited by the lack of physiologically relevant models that recapitulate the disease phenotype. The use of immortalized cell lines as in vitro model systems for drug discovery has met with limited success, since efficacy and safety too often fail to translate successfully in human clinical trials. Drug discoverers are shifting their focus to more physiologically relevant cellular models, including primary neurons and stem cells. The recent discovery of induced pluripotent stem (iPS) cell technology presents an exciting opportunity to derive human DA neurons from patients with sporadic and familial forms of PD. We anticipate that these human DA models will recapitulate key features of the PD phenotype. In parallel, high-content screening platforms, which extract information on multiple cellular features within individual neurons, provide a network-based approach that can resolve temporal and spatial relationships underlying mechanisms of neurodegeneration and drug perturbations. These emerging technologies have the potential to establish highly predictive cellular models that could bring about a desperately needed revolution in PD drug discovery.


FEBS Letters | 2013

Longitudinal measures of proteostasis in live neurons: Features that determine fate in models of neurodegenerative disease

Gaia Skibinski; Steven Finkbeiner

Protein misfolding and proteostasis decline is a common feature of many neurodegenerative diseases. However, modeling the complexity of proteostasis and the global cellular consequences of its disruption is a challenge, particularly in live neurons. Although conventional approaches, based on population measures and single “snapshots”, can identify cellular changes during neurodegeneration, they fail to determine if these cellular events drive cell death or act as adaptive responses. Alternatively, a “systems” cell biology approach known as longitudinal survival analysis enables single neurons to be followed over the course of neurodegeneration. By capturing the dynamics of misfolded proteins and the multiple cellular events that occur along the way, the relationship of these events to each other and their importance and role during cell death can be determined. Quantitative models of proteostasis dysfunction may yield unique insight and novel therapeutic strategies for neurodegenerative disease.


Cell and Tissue Research | 2018

iPS cells in the study of PD molecular pathogenesis

Melanie M. Cobb; Abinaya Ravisankar; Gaia Skibinski; Steven Finkbeiner

Parkinson’s disease (PD) is the second most common neurodegenerative disease and its pathogenic mechanisms are poorly understood. The majority of PD cases are sporadic but a number of genes are associated with familial PD. Sporadic and familial PD have many molecular and cellular features in common, suggesting some shared pathogenic mechanisms. Induced pluripotent stem cells (iPSCs) have been derived from patients harboring a range of different mutations of PD-associated genes. PD patient-derived iPSCs have been differentiated into relevant cell types, in particular dopaminergic neurons and used as a model to study PD. In this review, we describe how iPSCs have been used to improve our understanding of the pathogenesis of PD. We describe what cellular and molecular phenotypes have been observed in neurons derived from iPSCs harboring known PD-associated mutations and what common pathways may be involved.


Biometrics | 2016

A Three-groups Model for High Throughput Survival Screens

Benjamin A. Shaby; Gaia Skibinski; Michael Ando; Eva S. LaDow; Steven Finkbeiner

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative condition characterized by the progressive deterioration of motor neurons in the cortex and spinal cord. Using an automated robotic microscope platform that enables the longitudinal tracking of thousands of single neurons, we examine the effects a large library of compounds on modulating the survival of primary neurons expressing a mutation known to cause ALS. The goal of our analysis is to identify the few potentially beneficial compounds among the many assayed, the vast majority of which do not extend neuronal survival. This resembles the large-scale simultaneous inference scenario familiar from microarray analysis, but transferred to the survival analysis setting due to the novel experimental setup. We apply a three-component mixture model to censored survival times of thousands of individual neurons subjected to hundreds of different compounds. The shrinkage induced by our model significantly improves performance in simulations relative to performing treatment-wise survival analysis and subsequent multiple testing adjustment. Our analysis identified compounds that provide insight into potential novel therapeutic strategies for ALS.


Cell | 2018

In Silico Labeling: Predicting Fluorescent Labels in Unlabeled Images

Eric Martin Christiansen; Samuel J. Yang; D. Michael Ando; Ashkan Javaherian; Gaia Skibinski; Scott Lipnick; Elliot Mount; Alison O’Neil; Kevan Shah; Alicia K. Lee; Piyush Goyal; William Fedus; Ryan Poplin; Andre Esteva; Marc Berndl; Lee L. Rubin; Philip C. Nelson; Steven Finkbeiner

Collaboration


Dive into the Gaia Skibinski's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin A. Shaby

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erica Korb

University of California

View shared research outputs
Top Co-Authors

Avatar

Jane Y. Wu

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Ken Nakamura

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aaron Daub

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge