Network


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

Hotspot


Dive into the research topics where Ayse Ulusoy is active.

Publication


Featured researches published by Ayse Ulusoy.


Brain | 2011

GDNF fails to exert neuroprotection in a rat {alpha}-synuclein model of Parkinson's disease.

Mickael Decressac; Ayse Ulusoy; Bengt Mattsson; Biljana Georgievska; Marina Romero-Ramos; Deniz Kirik; Anders Björklund

The neuroprotective effect of the glial cell line-derived neurotrophic factor has been extensively studied in various toxic models of Parkinsons disease. However, it remains unclear whether this neurotrophic factor can protect against the toxicity induced by the aggregation-prone protein α-synuclein. Targeted overexpression of human wild-type α-synuclein in the nigrostriatal system, using adeno-associated viral vectors, causes a progressive degeneration of the nigral dopamine neurons and the development of axonal pathology in the striatum. In the present study, we investigated, using different paradigms of delivery, whether glial cell line-derived neurotrophic factor can protect against the neurodegenerative changes and the cellular stress induced by α-synuclein. We found that viral vector-mediated delivery of glial cell line-derived neurotrophic factor into substantia nigra and/or striatum, administered 2-3 weeks before α-synuclein, was inefficient in preventing the wild-type α-synuclein-induced loss of dopamine neurons and terminals. In addition, glial cell line-derived neurotrophic factor overexpression did not ameliorate the behavioural deficit in this rat model of Parkinsons disease. Quantification of striatal α-synuclein-positive aggregates revealed that glial cell line-derived neurotrophic factor had no effect on α-synuclein aggregation. These data provide the evidence for the lack of neuroprotective effect of glial cell line-derived neurotrophic factor against the toxicity of human wild-type α-synuclein in an in vivo model of Parkinsons disease. The difference in neuroprotective efficacy of glial cell line-derived neurotrophic factor seen in our model and the commonly used neurotoxin models of Parkinsons disease, raises important issues pertinent to the interpretation of the results obtained in preclinical models of Parkinsons disease, and their relevance for the therapeutic use glial cell line-derived neurotrophic factor in patients with Parkinsons disease.


Human Molecular Genetics | 2012

α-Synuclein expression and Nrf2 deficiency cooperate to aggravate protein aggregation, neuronal death and inflammation in early-stage Parkinson's disease

Isabel Lastres-Becker; Ayse Ulusoy; Nadia G. Innamorato; Gurdal Sahin; Alberto Rábano; Deniz Kirik; Antonio Cuadrado

Although α-synuclein (α-SYN) aggregation is a hallmark of sporadic and familial Parkinsons disease (PD), it is not known how it contributes to early events of PD pathogenesis such as oxidative and inflammatory stress. Here, we addressed this question in a new animal model based on stereotaxic delivery of an adeno-associated viral vector (rAAV) for expression of human α-SYN in the ventral midbrain of mice lacking the transcription factor Nrf2 (Nrf2(-/-)). Two months after surgery, Nrf2(-/-) mice exhibited exacerbated degeneration of nigral dopaminergic neurons and increased dystrophic dendrites, reminiscent of Lewy neurites, which correlated with impaired proteasome gene expression and activity. Dopaminergic neuron loss was associated with an increase in neuroinflammation and gliosis that were intensified in Nrf2(-/-) mice. In response to exogenously added α-SYN, Nrf2(-/-) microglia failed to activate the expression of two anti-inflammatory genes, heme oxygenase-1 (HO-1) and nicotinamide adenine dinucleotide phosphate quinone oxidorreductase-1 (NQO1). This impaired Nrf2 response correlated with a shift in the microglial activation profile, towards increased production of proinflammatory markers, IL-6, IL-1β and iNOS and reduced phagocytic capacity of fluorescent beads, and lower messenger RNA levels for TAM receptors Axl and Mer. Postmortem brain tissue samples from patients in early- to middle-stage progression of PD showed increased HO-1 expression in astrocytes and microglia, suggesting an attempt of the diseased brain to compensate these hallmarks of PD through activation of the Nrf2 pathway. This study demonstrates that α-SYN and Nrf2 deficiency cooperate on protein aggregation, neuroinflammation and neuronal death and provides a bifactorial animal model to study early-stage PD.


Progress in Brain Research | 2010

Viral vector-mediated overexpression of α-synuclein as a progressive model of Parkinson's disease.

Ayse Ulusoy; Mickael Decressac; Deniz Kirik; Anders Björklund

The discovery of the role of α-synuclein in the pathogenesis of Parkinsons disease (PD) has opened new possibilities for the development of more authentic models of Parkinsons disease. Recombinant adeno-associated virus (AAV) and lentivirus (LV) vectors are efficient tools for expression of genes locally in subsets of neurons in the brain and can be used to express human wild-type or mutated α-synuclein selectively in midbrain dopamine neurons. Using this approach, it is possible to trigger extensive PD-like cellular and axonal pathologies in the nigrostriatal projection, involving abnormal protein aggregation, neuronal dysfunction, and cell death that develop progressively over time. Targeted overexpression of human α-synuclein in midbrain dopamine neurons, using AAV vectors, reproduces many of the characteristic features of the human disease and provides, for the first time, a model of progressive PD that can be applied to both rodents and primates.


Molecular Therapy | 2009

Dose Optimization for Long-term rAAV-mediated RNA Interference in the Nigrostriatal Projection Neurons

Ayse Ulusoy; Gurdal Sahin; Tomas Björklund; Patrick Aebischer; Deniz Kirik

Short-hairpin RNA (shRNA)-mediated gene knockdown is a powerful tool for targeted gene silencing and an emerging novel therapeutic strategy. Recent publications, however, reported unexpected toxicity after utilizing viral-mediated shRNA knockdown in vivo. Thus, it is currently unclear whether shRNA-mediated knockdown strategy can be used as a safe and efficient tool for gene silencing. In this study, we have generated rAAV vectors expressing shRNAs targeting the rat tyrosine hydroxylase (TH) mRNA (shTH) for testing the efficacy of in vivo TH knockdown in the nigral dopaminergic neurons. At high titers, not only the shTH vectors but also the scrambled and green fluorescence protein (GFP)-only controls caused cell death. In a dose-response study, we identified a dose window leading to >60% decrease in TH(+) neurons without any change in vesicular monoamine transporter-2 (VMAT2) expression. Moreover, using the safe and efficient dose, we showed that dopamine (DA) synthesis rate was significantly reduced and this lead to emergence of motor deficits in the shTH-expressing rats. Interestingly, these animals showed very robust and long-lasting recovery after a single systemic L-3,4-dihydroxyphenylalanine (L-DOPA) administration beyond what can be achieved in 6-hydroxydopamine (6-OHDA)-lesioned rats. Our results have implications for both mechanistic and therapeutic studies utilizing long-term shRNA-mediated gene silencing in the nigrostriatal projection system.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Presynaptic dopaminergic compartment determines the susceptibility to L-DOPA-induced dyskinesia in rats.

Ayse Ulusoy; Gurdal Sahin; Deniz Kirik

Drug-induced dyskinesias in dopamine-denervated animals are known to depend on both pre- and postsynaptic changes of the nigrostriatal circuitry. In lesion models used thus far, changes occur in both of these compartments and, therefore, it has not been possible to dissect the individual contribution of each compartment in the pathophysiology of dyskinesias. Here we silenced the nigrostriatal dopamine neurotransmission without affecting the anatomical integrity of the presynaptic terminals using a short-hairpin RNA-mediated knockdown of tyrosine hydroxylase enzyme (shTH). This treatment resulted in significant reduction (by about 70%) in extracellular dopamine concentration in the striatum as measured by on-line microdialysis. Under these conditions, the animals remained nondyskinetic after chronic L-DOPA treatment, whereas partial intrastriatal 6-hydoxydopamine lesioned rats with comparable reduction in extracellular dopamine levels developed dyskinesias. On the other hand, apomorphine caused moderate to severe dyskinesias in both groups. Importantly, single-dose L-DOPA challenge in apomorphine-primed shTH animals failed to activate the already established abnormal postsynaptic responses. Taken together, these data provide direct evidence that the status of the presynaptic, DA releasing compartment is a critical determinant of both the induction and maintenance of L-DOPA–induced dyskinesias.


European Journal of Neuroscience | 2010

Co-expression of C-terminal truncated alpha-synuclein enhances full-length alpha-synuclein-induced pathology.

Ayse Ulusoy; Fabia Febbraro; Poul Henning Jensen; Deniz Kirik; Marina Romero-Ramos

Lewy bodies, which are a pathological hallmark of Parkinson’s disease, contain insoluble polymers of alpha‐synuclein (αsyn). Among the different modifications that can promote the formation of toxic αsyn species, C‐terminal truncation is among the most abundant alterations in patients with Parkinson’s disease. In vitro, C‐terminal truncated αsyn aggregates faster and sub‐stoichiometric amounts of C‐terminal truncated αsyn promote aggregation of the full‐length αsyn (αsynFL) and induce neuronal toxicity. To address in vivo the putative stimulation of αsyn‐induced pathology by the presence of truncated αsyn, we used recombinant adeno‐associated virus to express either αsynFL or a C‐terminal truncated αsyn (1‐110) in rats. We adjusted the recombinant adeno‐associated virus vector concentrations so that either protein alone led to only mild to moderate axonal pathology in the terminals of nigrostriatal dopamine neurons without frank cell loss. When these two forms of αsyn were co‐expressed at these pre‐determined levels, it resulted in a more aggressive pathology in fiber terminals as well as dopaminergic cell loss in the substantia nigra. Using an antibody that did not detect the C‐terminal truncated αsyn (1‐110) but only αsynFL, we demonstrated that the co‐expressed truncated protein promoted the progressive accumulation of αsynFL and formation of larger pathological accumulations. Moreover, in the co‐expression group, three of the eight animals showed apomorphine‐induced turning, suggesting prominent post‐synaptic alterations due to impairments in the dopamine release, whereas the mild pathology induced by either form alone did not cause motor abnormalities. Taken together these data suggest that C‐terminal truncated αsyn can interact with and exacerbate the formation of pathological accumulations containing αsynFL in vivo.


Experimental Neurology | 2008

In vivo gene delivery for development of mammalian models for Parkinson's disease

Ayse Ulusoy; Tomas Björklund; Stephan Hermening; Deniz Kirik

During the last decade, identification of the genes involved in familial forms of Parkinsons disease (PD) has advanced our understanding of the mechanisms underlying the development of different aspects of PD. However the available animal models still remain as the main limiting factor for the development of neuroprotective therapies that can halt the progression of the disease, through which we wish to provide a better quality of life for the PD patients. Here, we review the recently developed animal models based on overexpression of PD-associated genes using recombinant viral vectors. Recombinant adeno-associated viral vectors, in particular, have been very useful in targeting the nigral dopamine neurons both in the rodent and the primate brain. In order to provide insights into the establishment of these models in the laboratory, we will not only give an overview of the results from these studies but also cover practical issues related to the production and handling of the viral vectors, which are critical for the successful application of this approach.


Neurobiology of Disease | 2012

Dysregulated dopamine storage increases the vulnerability to alpha-synuclein in nigral neurons

Ayse Ulusoy; Tomas Björklund; Kerstin Buck; Deniz Kirik

Impairments in the capacity of dopaminergic neurons to handle cytoplasmic dopamine may be a critical factor underlying the selective vulnerability of midbrain dopamine neurons in Parkinsons disease. Furthermore, toxicity of α-synuclein in dopaminergic neurons has been suggested to be mediated by direct interaction between dopamine and α-synuclein through formation of abnormal α-synuclein species, although direct in vivo evidence to support this hypothesis is lacking. Here, we investigated the role of dopamine availability on α-synuclein mediated neurodegeneration in vivo. We found that overexpression of α-synuclein in nigral dopamine neurons in mice with deficient vesicular storage of dopamine led to a significant increase in dopaminergic neurodegeneration. Importantly, silencing the tyrosine hydroxylase enzyme - thereby reducing dopamine content in the nigral neurons - reversed the increased vulnerability back to the baseline level observed in wild-type littermates, but failed to eliminate it completely. Importantly, TH knockdown was not effective in altering the toxicity in the wild-type animals. Taken together, our data suggest that under normal circumstances, in healthy dopamine neurons, cytoplasmic dopamine is tightly controlled such that it does not contribute significantly to α-synuclein mediated toxicity. Dysregulation of the dopamine machinery in the substantia nigra, on the other hand, could act as a trigger for induction of increased toxicity in these neurons and could explain how these neurons become more vulnerable and die in the disease process.


Brain | 2016

Brain propagation of transduced α-synuclein involves non-fibrillar protein species and is enhanced in α-synuclein null mice

Michael Helwig; Michael Klinkenberg; Raffaella Rusconi; Ruth E. Musgrove; Nour K. Majbour; Omar M. A. El-Agnaf; Ayse Ulusoy; Donato A. Di Monte

Aggregation and neuron-to-neuron transmission are attributes of α-synuclein relevant to its pathogenetic role in human synucleinopathies such as Parkinsons disease. Intraparenchymal injections of fibrillar α-synuclein trigger widespread propagation of amyloidogenic protein species via mechanisms that require expression of endogenous α-synuclein and, possibly, its structural corruption by misfolded conformers acting as pathological seeds. Here we describe another paradigm of long-distance brain diffusion of α-synuclein that involves inter-neuronal transfer of monomeric and/or oligomeric species and is independent of recruitment of the endogenous protein. Targeted expression of human α-synuclein was induced in the mouse medulla oblongata through an injection of viral vectors into the vagus nerve. Enhanced levels of intra-neuronal α-synuclein were sufficient to initiate its caudo-rostral diffusion that likely involved at least one synaptic transfer and progressively reached specific brain regions such as the locus coeruleus, dorsal raphae and amygdala in the pons, midbrain and forebrain. Transfer of human α-synuclein was compared in two separate lines of α-synuclein-deficient mice versus their respective wild-type controls and, interestingly, lack of endogenous α-synuclein expression did not counteract diffusion but actually resulted in a more pronounced and advanced propagation of exogenous α-synuclein. Self-interaction of adjacent molecules of human α-synuclein was detected in both wild-type and mutant mice. In the former, interaction of human α-synuclein with mouse α-synuclein was also observed and might have contributed to differences in protein transmission. In wild-type and α-synuclein-deficient mice, accumulation of human α-synuclein within recipient axons in the pons, midbrain and forebrain caused morphological evidence of neuritic pathology. Tissue sections from the medulla oblongata and pons were stained with different antibodies recognizing oligomeric, fibrillar and/or total (monomeric and aggregated) α-synuclein. Following viral vector transduction, monomeric, oligomeric and fibrillar protein was detected within donor neurons in the medulla oblongata. In contrast, recipient axons in the pons were devoid of immunoreactivity for fibrillar α-synuclein, indicating that non-fibrillar forms of α-synuclein were primarily transferred from one neuron to the other, diffused within the brain and led to initial neuronal injury. This study elucidates a paradigm of α-synuclein propagation that may play a particularly important role under pathophysiological conditions associated with enhanced α-synuclein expression. Rapid long-distance diffusion and accumulation of monomeric and oligomeric α-synuclein does not necessarily involve pathological seeding but could still result in a significant neuronal burden during the pathogenesis of neurodegenerative diseases.


Experimental Neurology | 2008

Can overexpression of parkin provide a novel strategy for neuroprotection in Parkinson's disease?

Ayse Ulusoy; Deniz Kirik

Parkinsons disease (PD) is a progressive neurodegenerative disorder caused by selective degeneration of the dopamine producing neurons in the substantia nigra. There is currently no clinically applicable therapy for treating or preventing Parkinsonian neurodegeneration. Great effort is put behind the development of novel therapeutic approaches that aim to alter the natural progression of the disease. For example, a disease-modifying strategy based on the use of glial cell line-derived neurotrophic factor family of ligands have yielded successful results in animal models and later in initial clinical trials. More recently, identification of the gene mutations underlying the familial forms of the disease opened new frontiers in tackling the underlying neuropathological changes seen in PD brains. Overexpression of parkin, in particular, emerged as a powerful approach with complementary effects to those described with use of neurotrophic factors. In light of the fact that the mechanism of disease in the affected patient population might be significantly variable, the ability to intervene the disease process at multiple levels should be seen as a key point in devising effective treatments.

Collaboration


Dive into the Ayse Ulusoy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donato A. Di Monte

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Raffaella Rusconi

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nour K. Majbour

United Arab Emirates University

View shared research outputs
Researchain Logo
Decentralizing Knowledge