Network


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

Hotspot


Dive into the research topics where Natalia Louneva is active.

Publication


Featured researches published by Natalia Louneva.


American Journal of Pathology | 2008

Caspase-3 Is Enriched in Postsynaptic Densities and Increased in Alzheimer's Disease

Natalia Louneva; Julia W. Cohen; Li-Ying Han; Konrad Talbot; Robert S. Wilson; David A. Bennett; John Q. Trojanowski; Steven E. Arnold

Progressive synaptic degeneration and neuron loss are major structural correlates of cognitive impairment in Alzheimers disease (AD). The mechanisms by which synaptic degeneration in AD occurs have not been established. The activation of proteins within the caspase family has been implicated in AD-associated neurodegeneration, and synaptically localized caspase activity could play a role in the synaptic degeneration and loss found in AD. We used synaptosomal fractionation with Western blotting and immunohistochemistry to examine the anatomical, subcellular, and subsynaptic expression patterns of caspase 3 in both the anterior cingulate cortex and hippocampus of control and AD patients. In both control and AD cases, there was a selective enrichment of caspase- 3 at synapses, particularly in the postsynaptic density (PSD) fractions. Compared with controls, AD patients exhibited significant increases in synaptic procaspase- 3 and active caspase-3 expression levels that were most evident in the PSD fractions. These data demonstrate for the first time the preferential localization and increase of caspase-3 in the PSD fractions in AD and suggest an important role for caspase 3 in synapse degeneration during disease progression.


Human Molecular Genetics | 2009

Dysbindin-1 in dorsolateral prefrontal cortex of schizophrenia cases is reduced in an isoform-specific manner unrelated to dysbindin-1 mRNA expression

Junxia Tang; Robert P. LeGros; Natalia Louneva; Lilly Yeh; Julia W. Cohen; Chang-Gyu Hahn; Derek J. Blake; Steven E. Arnold; Konrad Talbot

DTNBP1 (dystrobrevin binding protein 1) remains a top candidate gene in schizophrenia. Reduced expression of this gene and of its encoded protein, dysbindin-1, have been reported in the brains of schizophrenia cases. It has not been established, however, if the protein reductions encompass all dysbindin-1 isoforms or if they are associated with decreased DTNBP1 gene expression. Using a matched pairs design in which each of 28 Caucasian schizophrenia cases was matched in age and sex to a normal Caucasian control, Western blotting of whole-tissue lysates of dorsolateral prefrontal cortex (DLPFC) revealed significant reductions in dysbindin-1C (but not in dysbindin-1A or -1B) in schizophrenia (P = 0.022). These reductions occurred without any significant change in levels of the encoding transcript in the same tissue samples and in the absence of the only DTNBP1 risk haplotype for schizophrenia reported in the USA. Indeed, no significant correlations were found between case-control differences in any dysbindin-1 isoform and the case-control differences in its encoding mRNA. Consequently, the mean 60% decrease in dysbindin-1C observed in 71% of our case-control pairs appears to reflect abnormalities in mRNA translation and/or processes promoting dysbindin-1C degradation (e.g. oxidative stress, phosphorylation and/or ubiquitination). Given the predominantly post-synaptic localization of dysbindin-1C and known post-synaptic effects of dysbindin-1 reductions in the rodent equivalent of the DLPFC, the present findings suggest that decreased dysbindin-1C in the DLPFC may contribute to the cognitive deficits of schizophrenia by promoting NMDA receptor hypofunction in fast-spiking interneurons.


PLOS ONE | 2011

Synaptic Dysbindin-1 Reductions in Schizophrenia Occur in an Isoform-Specific Manner Indicating Their Subsynaptic Location

Konrad Talbot; Natalia Louneva; Julia W. Cohen; Hala Kazi; Derek J. Blake; Steven E. Arnold

Background An increasing number of studies report associations between variation in DTNBP1, a top candidate gene in schizophrenia, and both the clinical symptoms of the disorder and its cognitive deficits. DTNBP1 encodes dysbindin-1, reduced levels of which have been found in synaptic fields of schizophrenia cases. This study determined whether such synaptic reductions are isoform-specific. Methodology/Principal Findings Using Western blotting of tissue fractions, we first determined the synaptic localization of the three major dysbindin-1 isoforms (A, B, and C). All three were concentrated in synaptosomes of multiple brain areas, including auditory association cortices in the posterior half of the superior temporal gyrus (pSTG) and the hippocampal formation (HF). Tests on the subsynaptic tissue fractions revealed that each isoform is predominantly, if not exclusively, associated with synaptic vesicles (dysbindin-1B) or with postsynaptic densities (dysbindin-1A and -1C). Using Western blotting on pSTG (n = 15) and HF (n = 15) synaptosomal fractions from schizophrenia cases and their matched controls, we discovered that synaptic dysbindin-1 is reduced in an isoform-specific manner in schizophrenia without changes in levels of synaptophysin or PSD-95. In pSTG, about 92% of the schizophrenia cases displayed synaptic dysbindin-1A reductions averaging 48% (p = 0.0007) without alterations in other dysbindin-1 isoforms. In the HF, by contrast, schizophrenia cases displayed normal levels of synaptic dysbindin-1A, but 67% showed synaptic reductions in dysbindin-1B averaging 33% (p = 0.0256), while 80% showed synaptic reductions in dysbindin-1C averaging 35% (p = 0.0171). Conclusions/Significance Given the distinctive subsynaptic localization of dysbindin-1A, -1B, and -1C across brain regions, the observed pSTG reductions in dysbindin-1A are postsynaptic and may promote dendritic spine loss with consequent disruption of auditory information processing, while the noted HF reductions in dysbindin-1B and -1C are both presynaptic and postsynaptic and could promote deficits in spatial working memory.


Acta Neuropathologica | 2014

Abnormal serine phosphorylation of insulin receptor substrate 1 is associated with tau pathology in Alzheimer's disease and tauopathies.

Mark Yarchoan; Jon B. Toledo; Edward B. Lee; Zoe Arvanitakis; Hala Kazi; Li.-Ying Han; Natalia Louneva; Virginia M.-Y. Lee; Sangwon F. Kim; John Q. Trojanowski; Steven E. Arnold

Neuronal insulin signaling abnormalities have been associated with Alzheimer’s disease (AD). However, the specificity of this association and its underlying mechanisms have been unclear. This study investigated the expression of abnormal serine phosphorylation of insulin receptor substrate 1 (IRS1) in 157 human brain autopsy cases that included AD, tauopathies, α-synucleinopathies, TDP-43 proteinopathies, and normal aging. IRS1-pS616, IRS1-pS312 and downstream target Akt-pS473 measures were most elevated in AD but were also significantly increased in the tauopathies: Pick’s disease, corticobasal degeneration and progressive supranuclear palsy. Double immunofluorescence labeling showed frequent co-expression of IRS1-pS616 with pathologic tau in neurons and dystrophic neurites. To further investigate an association between tau and abnormal serine phosphorylation of IRS1, we examined the presence of abnormal IRS1-pS616 expression in pathological tau-expressing transgenic mice and demonstrated that abnormal IRS1-pS616 frequently co-localizes in tangle-bearing neurons. Conversely, we observed increased levels of hyperphosphorylated tau in the high-fat diet-fed mouse, a model of insulin resistance. These results provide confirmation and specificity that abnormal phosphorylation of IRS1 is a pathological feature of AD and other tauopathies, and provide support for an association between insulin resistance and abnormal tau as well as amyloid-β.


Neurobiology of Aging | 2014

Neuroprotective effects of the amylin analogue pramlintide on Alzheimer's disease pathogenesis and cognition.

Brittany L. Adler; Mark Yarchoan; Hae Min Hwang; Natalia Louneva; Jeffrey A. Blair; Russell Palm; Mark A. Smith; Hyoung Gon Lee; Steven E. Arnold; Gemma Casadesus

Amylin is a metabolic peptide hormone that is co-secreted with insulin from beta cells in the pancreas and activates many of the downstream targets of insulin. To investigate the relationship between this hormone and Alzheimers disease (AD), we measured plasma human amylin levels in 206 subjects with AD, 64 subjects with mild cognitive impairment, and 111 subjects with no cognitive impairment and found significantly lower amylin levels among subjects with AD and mild cognitive impairment compared with the cognitively intact subjects. To investigate mechanisms underlying amylins effects in the brain, we administered chronic infusions of the amylin analog pramlintide in the senescence-accelerated prone mouse, a mouse model of sporadic AD. Pramlintide administration improved performance in the novel object recognition task, a validated test of memory and cognition. The pramlintide-treated mice had increased expression of the synaptic marker synapsin I and the kinase cyclin-dependent kinase-5 in the hippocampus, as well as decreased oxidative stress and inflammatory markers in the hippocampus. A dose-dependent increase in cyclin-dependent kinase-5 and activation of extracellular-signal-regulated-kinases 1/2 by pramlintide treatment in vitro was also present indicating functionality of the amylin receptor in neurons. Together these results suggest that amylin analogs have neuroprotective properties and might be of therapeutic benefit in AD.


Synapse | 2011

Chronic corticosterone exposure alters postsynaptic protein levels of PSD‐95, NR1, and synaptopodin in the mouse brain

Julia W. Cohen; Natalia Louneva; Li-Ying Han; Georgia E. Hodes; Robert S. Wilson; David A. Bennett; Irwin Lucki; Steven E. Arnold

Animal models provide compelling evidence that chronic stress is associated with biochemical and morphological changes in the brain, many of which are mediated by corticosterone, a principal glucocorticoid synthesized in the rodent adrenal cortex and secreted in response to stress. To better characterize the effects of chronic corticosterone at the synaptic and subsynaptic level, we implanted three‐month‐old male C57B/6 mice with 2 × 5 mg corticosterone pellets (CORT group, n = 14), 21 day release formulation (20 mg/kg/day dose) or placebo pellets (Placebo group, n = 14), 21‐day release formulation. After 20 days, brains were removed. One hemisphere was frozen for biochemical analysis by synaptosomal fractionation with Western blotting, and the other hemisphere was fixed for immunohistochemistry. Localization and expression levels for PSD‐95, NR1, and synaptopodin proteins were assessed. Biochemical analysis revealed lower protein levels of PSD‐95 (32% decrease, P < 0.001), NR1 (47%, P = 0.01), and synaptopodin (65%, P < 0.001) in the postsynaptic density subsynaptic fraction of the CORT group. Optical densitometry in immunohistochemically labeled sections also found lower levels of PSD‐95 in synaptic fields of the dentate gyrus (PSD‐95, 33% decrease, P < 0.001; NR1, 31%, P < 0.001; synaptopodin, 40%, P < 0.001) and the CA3 stratum lucidum (36%, P < 0.001, 40%, P < 0.001, and 35%, P < 0.001) of the CORT group. While mechanistic relationships for these changes are not yet known, we speculate that synaptopodin, which is involved in regulation of spine calcium kinetics and posttranslational modification and transport of locally synthesized proteins, may play an important role in the changes of PSD‐95 and NR1 protein levels and other synaptic alterations. Synapse 2011.


Journal of the Neurological Sciences | 2013

Association of plasma C-reactive protein levels with the diagnosis of Alzheimer's disease.

Mark Yarchoan; Natalia Louneva; Sharon X. Xie; Frank Swenson; William T. Hu; Holly Soares; John Q. Trojanowski; Virginia M.-Y. Lee; Mitchel A. Kling; Leslie M. Shaw; Alice Chen-Plotkin; David A. Wolk; Steven E. Arnold

C-reactive protein (CRP) participates in the systemic response to inflammation. Previous studies report inconsistent findings regarding the relationship between plasma CRP and Alzheimers disease (AD). We measured plasma CRP in 203 subjects with AD, 58 subjects with mild cognitive impairment (MCI) and 117 normal aging subjects and administered annual Mini-Mental State Examinations (MMSE) during a 3-year follow-up period to investigate CRPs relationship with diagnosis and progression of cognitive decline. Adjusted for age, sex, and education, subjects with AD had significantly lower levels of plasma CRP than subjects with MCI and normal aging. However, there was no significant association between plasma CRP at baseline and subsequent cognitive decline as assessed by longitudinal changes in MMSE score. Our results support previous reports of reduced levels of plasma CRP in AD and indicate its potential utility as a biomarker for the diagnosis of AD.


The Journal of Neuroscience | 2015

PDE-4 inhibition rescues aberrant synaptic plasticity in Drosophila and mouse models of fragile X syndrome.

Catherine H. Choi; Brian P. Schoenfeld; Eliana D. Weisz; Aaron J. Bell; Daniel B. Chambers; Joseph Hinchey; Richard J. Choi; Paul Hinchey; Maria Kollaros; Michael Gertner; Neal J. Ferrick; Allison M. Terlizzi; Nicole L. Yohn; Eric Koenigsberg; David A. Liebelt; R. Suzanne Zukin; Newton H. Woo; Michael R. Tranfaglia; Natalia Louneva; Steven E. Arnold; Steven J. Siegel; Francois V. Bolduc; Thomas V. McDonald; Thomas A. Jongens; Sean M.J. McBride

Fragile X syndrome (FXS) is the leading cause of both intellectual disability and autism resulting from a single gene mutation. Previously, we characterized cognitive impairments and brain structural defects in a Drosophila model of FXS and demonstrated that these impairments were rescued by treatment with metabotropic glutamate receptor (mGluR) antagonists or lithium. A well-documented biochemical defect observed in fly and mouse FXS models and FXS patients is low cAMP levels. cAMP levels can be regulated by mGluR signaling. Herein, we demonstrate PDE-4 inhibition as a therapeutic strategy to ameliorate memory impairments and brain structural defects in the Drosophila model of fragile X. Furthermore, we examine the effects of PDE-4 inhibition by pharmacologic treatment in the fragile X mouse model. We demonstrate that acute inhibition of PDE-4 by pharmacologic treatment in hippocampal slices rescues the enhanced mGluR-dependent LTD phenotype observed in FXS mice. Additionally, we find that chronic treatment of FXS model mice, in adulthood, also restores the level of mGluR-dependent LTD to that observed in wild-type animals. Translating the findings of successful pharmacologic intervention from the Drosophila model into the mouse model of FXS is an important advance, in that this identifies and validates PDE-4 inhibition as potential therapeutic intervention for the treatment of individuals afflicted with FXS.


Frontiers in Behavioral Neuroscience | 2016

Multiple Drug Treatments That Increase cAMP Signaling Restore Long-Term Memory and Aberrant Signaling in Fragile X Syndrome Models.

Catherine H. Choi; Brian P. Schoenfeld; Aaron J. Bell; Joseph Hinchey; Cory Rosenfelt; Michael Gertner; Sean R. Campbell; Danielle Emerson; Paul Hinchey; Maria Kollaros; Neal J. Ferrick; Daniel B. Chambers; Steven Langer; Steven Sust; Aatika Malik; Allison M. Terlizzi; David A. Liebelt; David Ferreiro; Ali Sharma; Eric Koenigsberg; Richard J. Choi; Natalia Louneva; Steven E. Arnold; Robert E. Featherstone; Steven J. Siegel; R. Suzanne Zukin; Thomas V. McDonald; Francois V. Bolduc; Thomas A. Jongens; Sean M.J. McBride

Fragile X is the most common monogenic disorder associated with intellectual disability (ID) and autism spectrum disorders (ASD). Additionally, many patients are afflicted with executive dysfunction, ADHD, seizure disorder and sleep disturbances. Fragile X is caused by loss of FMRP expression, which is encoded by the FMR1 gene. Both the fly and mouse models of fragile X are also based on having no functional protein expression of their respective FMR1 homologs. The fly model displays well defined cognitive impairments and structural brain defects and the mouse model, although having subtle behavioral defects, has robust electrophysiological phenotypes and provides a tool to do extensive biochemical analysis of select brain regions. Decreased cAMP signaling has been observed in samples from the fly and mouse models of fragile X as well as in samples derived from human patients. Indeed, we have previously demonstrated that strategies that increase cAMP signaling can rescue short term memory in the fly model and restore DHPG induced mGluR mediated long term depression (LTD) in the hippocampus to proper levels in the mouse model (McBride et al., 2005; Choi et al., 2011, 2015). Here, we demonstrate that the same three strategies used previously with the potential to be used clinically, lithium treatment, PDE-4 inhibitor treatment or mGluR antagonist treatment can rescue long term memory in the fly model and alter the cAMP signaling pathway in the hippocampus of the mouse model.


Nature Precedings | 2009

Dysbindin-1 in dorsolateral prefrontal cortex of schizophrenia cases is reduced in an isoform-specific manner unrelated to altered dysbindin-1 gene expression

Junxia Tang; Robert P. LeGros; Natalia Louneva; Lilly Yeh; Julia W. Cohen; Chang-Gyu Hahn; Derek J. Blake; Steven E. Arnold; Konrad Talbot

Running Title: Dysbindin-1 isoforms in DLPFC of schizophrenia cases Abstract DTNBP1 (dystrobrevin binding protein 1) remains one of the top candidate genes in schizophrenia. Reduced expression of this gene and the protein it encodes, dysbindin-1, has been reported in the dorsolateral prefrontal cortex (DLPFC) of schizophrenia cases. It has not been established, however, if all dysbindin-1 isoforms are reduced in the DLPFC or if the reduction is associated with reduced DTNBP1 gene expression. Using Western blotting of whole-tissue lysates of the DLPFC with antibodies differentially sensitive to the three major isoforms of this protein (dysbindin-1A,-1B, and-1C), we found no significant differences between our schizophrenia cases and matched controls in dysbindin-1A or-1B, but did find a mean 46% reduction in dysbindin-1C in 71% of 28 case-control pairs (p = 0.022). This occurred in the absence of the one DTNBP1 risk haplotype for schizophrenia reported in the US and without alteration in levels of dysbindin-1C transcripts. Conversely, the absence of changes in the dysbindin-1A and-1B isoforms was accompanied by increased levels of their transcripts. We thus found no correspondence between alterations in dysbindin-1 gene and protein expression, the latter of which might be due to posttranslational modifications such as ubiquitination. Reduced DLPFC dysbindin-1C in schizophrenia probably occurs in PSDs, where we find dysbindin-1C to be heavily concentrated in the human brain. Given known postsynaptic effects of dysbindin-1 reductions in the rodent homolog of the prefrontal cortex, these findings suggest that reduced dysbindin-1C in the DLPFC may contribute to cognitive deficits of schizophrenia by promoting NMDA receptor hypofunction.

Collaboration


Dive into the Natalia Louneva's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julia W. Cohen

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Konrad Talbot

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David A. Bennett

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Junxia Tang

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Li-Ying Han

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Mark Yarchoan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Robert S. Wilson

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Aaron J. Bell

Albert Einstein College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge