Network


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

Hotspot


Dive into the research topics where Elizabeta Gjoneska is active.

Publication


Featured researches published by Elizabeta Gjoneska.


Nature | 2015

Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer/'s disease

Elizabeta Gjoneska; Andreas R. Pfenning; Hansruedi Mathys; Gerald Quon; Anshul Kundaje; Li-Huei Tsai; Manolis Kellis

Alzheimer’s disease (AD) is a severe age-related neurodegenerative disorder characterized by accumulation of amyloid-β plaques and neurofibrillary tangles, synaptic and neuronal loss, and cognitive decline. Several genes have been implicated in AD, but chromatin state alterations during neurodegeneration remain uncharacterized. Here we profile transcriptional and chromatin state dynamics across early and late pathology in the hippocampus of an inducible mouse model of AD-like neurodegeneration. We find a coordinated downregulation of synaptic plasticity genes and regulatory regions, and upregulation of immune response genes and regulatory regions, which are targeted by factors that belong to the ETS family of transcriptional regulators, including PU.1. Human regions orthologous to increasing-level enhancers show immune-cell-specific enhancer signatures as well as immune cell expression quantitative trait loci, while decreasing-level enhancer orthologues show fetal-brain-specific enhancer activity. Notably, AD-associated genetic variants are specifically enriched in increasing-level enhancer orthologues, implicating immune processes in AD predisposition. Indeed, increasing enhancers overlap known AD loci lacking protein-altering variants, and implicate additional loci that do not reach genome-wide significance. Our results reveal new insights into the mechanisms of neurodegeneration and establish the mouse as a useful model for functional studies of AD regulatory regions.


Cell | 2015

Activity-Induced DNA Breaks Govern the Expression of Neuronal Early-Response Genes

Ram Madabhushi; Fan Gao; Andreas R. Pfenning; Ling Pan; Satoko Yamakawa; Jinsoo Seo; Richard Rueda; Trongha X. Phan; Hidekuni Yamakawa; Ping Chieh Pao; Ryan T. Stott; Elizabeta Gjoneska; Alexi Nott; Sukhee Cho; Manolis Kellis; Li-Huei Tsai

Neuronal activity causes the rapid expression of immediate early genes that are crucial for experience-driven changes to synapses, learning, and memory. Here, using both molecular and genome-wide next-generation sequencing methods, we report that neuronal activity stimulation triggers the formation of DNA double strand breaks (DSBs) in the promoters of a subset of early-response genes, including Fos, Npas4, and Egr1. Generation of targeted DNA DSBs within Fos and Npas4 promoters is sufficient to induce their expression even in the absence of an external stimulus. Activity-dependent DSB formation is likely mediated by the type II topoisomerase, Topoisomerase IIβ (Topo IIβ), and knockdown of Topo IIβ attenuates both DSB formation and early-response gene expression following neuronal stimulation. Our results suggest that DSB formation is a physiological event that rapidly resolves topological constraints to early-response gene expression in neurons.


Neuron | 2018

APOE4 Causes Widespread Molecular and Cellular Alterations Associated with Alzheimer’s Disease Phenotypes in Human iPSC-Derived Brain Cell Types

Yuan-Ta Lin; Jinsoo Seo; Fan Gao; Heather M. Feldman; Hsin-Lan Wen; Jay Penney; Hugh P. Cam; Elizabeta Gjoneska; Waseem K. Raja; Jemmie Cheng; Richard Rueda; Oleg Kritskiy; Fatema Abdurrob; Zhuyu Peng; Blerta Milo; Chung Jong Yu; Sara Elmsaouri; Dilip Dey; Tak Ko; Bruce A. Yankner; Li-Huei Tsai

The apolipoprotein E4 (APOE4) variant is the single greatest genetic risk factor for sporadic Alzheimers disease (sAD). However, the cell-type-specific functions of APOE4 in relation to AD pathology remain understudied. Here, we utilize CRISPR/Cas9 and induced pluripotent stem cells (iPSCs) to examine APOE4 effects on human brain cell types. Transcriptional profiling identified hundreds of differentially expressed genes in each cell type, with the most affected involving synaptic function (neurons), lipid metabolism (astrocytes), and immune response (microglia-like cells). APOE4 neurons exhibited increased synapse number and elevated Aβ42 secretion relative to isogenic APOE3 cells while APOE4 astrocytes displayed impaired Aβ uptake and cholesterol accumulation. Notably, APOE4 microglia-like cells exhibited altered morphologies, which correlated with reduced Aβ phagocytosis. Consistently, converting APOE4 to APOE3 in brain cell types from sAD iPSCs was sufficient to attenuate multiple AD-related pathologies. Our study establishes a reference for human cell-type-specific changes associated with the APOE4 variant. VIDEO ABSTRACT.


bioRxiv | 2018

Epigenome-wide study uncovers tau pathology-driven changes of chromatin organization in the aging human brain

Hans-Ulrich Klein; Cristin McCabe; Elizabeta Gjoneska; Sarah E. Sullivan; Belinda J. Kaskow; Anna Tang; Robert V Smith; Jishu Xu; Andreas R. Pfenning; Bradley E. Bernstein; Alexander Meissner; Julie A. Schneider; Li-Huei Tsai; Tracy L. Young-Pearse; David A. Bennett; Philip L. De Jager

Accumulation of tau and amyloid-β are two pathologic hallmarks of Alzheimer’s disease (AD). Here, we conducted an epigenome-wide association study using the H3K9 acetylation (H3K9Ac) mark in 669 aged human prefrontal cortices: in contrast to amyloid-β, tau protein burden had a broad effect on the epigenome, affecting 5,590 out of 26,384 H3K9Ac domains. Tau-related alterations aggregated in large genomic segments reflecting spatial chromatin organization, and the magnitude of these effects correlated with the segment’s nuclear lamina association. We confirmed the functional relevance of these chromatin changes by demonstrating (1) consistent transcriptional changes in three independent datasets and (2) similar findings in two AD mouse models. Finally, we found that tau overexpression in iPSC-derived neurons disrupted chromatin organization and that these effects could be blocked by a small molecule predicted to reverse the tau effect. Thus, we report large-scale tau-driven chromatin rearrangements in the aging human brain that may be reversible with HSP90 inhibitors.


Cell Reports | 2017

The Transcription Factor Sp3 Cooperates with HDAC2 to Regulate Synaptic Function and Plasticity in Neurons

Hidekuni Yamakawa; Jemmie Cheng; Jay Penney; Fan Gao; Richard Rueda; Jun Wang; Satoko Yamakawa; Oleg Kritskiy; Elizabeta Gjoneska; Li-Huei Tsai


PMC | 2016

Histone deacetylase 3 associates with MeCP2 to regulate FOXO and social behavior

Feiran Zhang; Peng Jin; Alexander Nott; Jemmie Cheng; Fan Gao; Yuan-Ta Lin; Elizabeta Gjoneska; Tak Ko; Paras Minhas; Alicia Zamudio Montes de Oca; Jia Meng; Li-Huei Tsai


PMC | 2015

Integrative analysis of 111 reference human epigenomes

Anshul Kundaje; Wouter Meuleman; Jason Ernst; Angela Yen; Pouya Kheradpour; Zhizhuo Zhang; Jianrong Wang; Lucas D. Ward; Abhishek Sarkar; Gerald Quon; Matthew L. Eaton; Yi-Chieh Wu; Andreas R. Pfenning; Xinchen Wang; Melina Claussnitzer; Yaping Liu; Mukul S. Bansal; Soheil Feizi-Khankandi; Ah Ram Kim; Richard C. Sallari; Nicholas A Sinnott-Armstrong; Laurie A. Boyer; Elizabeta Gjoneska; Li-Huei Tsai; Manolis Kellis


PMC | 2015

Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease

Elizabeta Gjoneska; Andreas R. Pfenning; Hansruedi Mathys; Gerald Quon; Anshul Kundaje; Li-Huei Tsai; Manolis Kellis

Collaboration


Dive into the Elizabeta Gjoneska's collaboration.

Top Co-Authors

Avatar

Andreas R. Pfenning

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Manolis Kellis

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jemmie Cheng

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hansruedi Mathys

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jay Penney

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jinsoo Seo

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Abhishek Sarkar

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge