Network


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

Hotspot


Dive into the research topics where Ying Y. Jean is active.

Publication


Featured researches published by Ying Y. Jean.


Cell | 2014

Axonally Synthesized ATF4 Transmits a Neurodegenerative Signal across Brain Regions

Jimena Baleriola; Chandler A. Walker; Ying Y. Jean; John F. Crary; Carol M. Troy; Peter L. Nagy; Ulrich Hengst

In Alzheimers disease (AD) brain, exposure of axons to Aβ causes pathogenic changes that spread retrogradely by unknown mechanisms, affecting the entire neuron. We found that locally applied Aβ1-42 initiates axonal synthesis of a defined set of proteins including the transcription factor ATF4. Inhibition of local translation and retrograde transport or knockdown of axonal Atf4 mRNA abolished Aβ-induced ATF4 transcriptional activity and cell loss. Aβ1-42 injection into the dentate gyrus (DG) of mice caused loss of forebrain neurons whose axons project to the DG. Protein synthesis and Atf4 mRNA were upregulated in these axons, and coinjection of Atf4 siRNA into the DG reduced the effects of Aβ1-42 in the forebrain. ATF4 protein and transcripts were found with greater frequency in axons in the brain of AD patients. These results reveal an active role for intra-axonal translation in neurodegeneration and identify ATF4 as a mediator for the spread of AD pathology.


Progress in Molecular Biology and Translational Science | 2011

Regulation of caspases in the nervous system implications for functions in health and disease.

Carol M. Troy; Nsikan Akpan; Ying Y. Jean

Caspases, initially identified as a family of proteases regulating cell death, have been found to have nonapoptotic functions as well. Some family members are critical for mediating programmed cell death in development. After development, caspases are downregulated in the nervous system, but continue to perform important nonapoptotic functions relevant for neurogenesis and synaptic plasticity. In neurodegenerative diseases, where aberrant neuronal death is an outstanding feature, there is an increase in caspase activity. The specific caspase death pathways leading to dysfunction and death have still not been fully clarified, despite the plethora of scientific literature addressing these issues. In this chapter, we will present the current knowledge of caspase activation and activity pathways, the current tools for examining caspases, and functions of caspases in the nervous system in health and in disease. Alzheimers Disease, the most common neurodegenerative disorder, and cerebral ischemia, the most common cause of neurologic death, are used to illustrate our current understanding of death signaling in neurodegenerative diseases. A better understanding of how caspases function in health and disease would provide appropriate specific targets for the development of therapeutic interventions for these diseases. Life and death are exquisitely regulated at the cellular level from development through maturity. During development, neuronal death is the major factor shaping the nervous system. This death is mainly caspase-mediated apoptosis. Once the waves of developmental death have passed (death occurs at different times in different parts of the nervous system), there is downregulation of the death machinery, as the postmitotic neurons should live for the life of the organism. Aberrant neuronal death is a major part of neurodegenerative disorders, but there is still no clear understanding of the processes leading to the phenotypes of the various diseases. Even the type of death that occurs continues to be debated, whether it is apoptotic, necrotic, or autophagic, or some combination of these death mechanisms. Here, we will discuss the role that the caspases play in neuronal function, dysfunction, and death. First, we will discuss the regulation of caspase activation and activity. We will examine the current understanding of caspase function in developmental neuronal death and then illustrate the role of caspases in neuronal death in disease employing two diseases of neuronal loss, Alzheimers Disease (AD), which is the most common chronic neurodegenerative disorder, and cerebral ischemia/stroke, the third most common cause of death in Western society, which is an acute neuronal disorder with chronic sequelae.


Biochemical Journal | 2012

Neuronal caspase 2 activity and function requires RAIDD, but not PIDD.

Elena M. Ribe; Ying Y. Jean; Rebecca L. Goldstein; Claudia Manzl; Leonidas Stefanis; Andreas Villunger; Carol M. Troy

Caspase 2 was initially identified as a neuronally expressed developmentally down-regulated gene (HUGO gene nomenclature CASP2) and has been shown to be required for neuronal death induced by several stimuli, including NGF (nerve growth factor) deprivation and Aβ (β-amyloid). In non-neuronal cells the PIDDosome, composed of caspase 2 and two death adaptor proteins, PIDD (p53-inducible protein with a death domain) and RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1β-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, has been proposed as the caspase 2 activation complex, although the absolute requirement for the PIDDosome is not clear. To investigate the requirement for the PIDDosome in caspase-2-dependent neuronal death, we have examined the necessity for each component in induction of active caspase 2 and in execution of caspase-2-dependent neuronal death. We find that both NGF deprivation and Aβ treatment of neurons induce active caspase 2 and that induction of this activity depends on expression of RAIDD, but is independent of PIDD expression. We show that treatment of wild-type or PIDD-null neurons with Aβ or NGF deprivation induces formation of a complex of caspase 2 and RAIDD. We also show that caspase-2-dependent execution of neurons requires RAIDD, not PIDD. Caspase 2 activity can be induced in neurons from PIDD-null mice, and NGF deprivation or Aβ use caspase 2 and RAIDD to execute death of these neurons.


Neurotherapeutics | 2015

Caspases: Therapeutic Targets in Neurologic Disease

Carol M. Troy; Ying Y. Jean

Specific therapies for neurologic diseases such as Alzheimer’s disease provide the potential for better clinical outcomes. Expression of caspases in the brain is developmentally regulated, and dysregulated in neurologic disease, supporting that caspases may be therapeutic targets. The activity of caspases is carefully regulated via binding partners, cleavage, or endogenous inhibitors to prevent spontaneous activation, which could lead to aberrant cell death. This review serves as a brief examination of the current understanding of the regulation and function of caspases, and approaches to specifically target aberrant caspase activity. The use of proper tools to investigate individual caspases is addressed. Moreover, it summarizes the reports of various caspases in Alzheimer’s disease studies. A better understanding of specific caspase pathways in heath and neurodegenerative disease is crucial for identifying specific targets for the development of therapeutic interventions.


Biochemical Journal | 2013

Caspase-2 is essential for c-Jun transcriptional activation and Bim induction in neuron death.

Ying Y. Jean; Elena M. Ribe; Maria Elena Pero; Marina Moskalenko; Zarah Iqbal; Lianna J. Marks; Lloyd A. Greene; Carol M. Troy

Neuronal apoptotic death generally requires de novo transcription, and activation of the transcription factor c-Jun has been shown to be necessary in multiple neuronal death paradigms. Caspase-2 has been implicated in death of neuronal and non-neuronal cells, but its relationship to transcriptional activation has not been clearly elucidated. In the present study, using two different neuronal apoptotic paradigms, β-amyloid treatment and NGF (nerve growth factor) withdrawal, we examined the hierarchical role of caspase-2 activation in the transcriptional control of neuron death. Both paradigms induce rapid activation of caspase-2 as well as activation of the transcription factor c-Jun and subsequent induction of the pro-apoptotic BH3 (Bcl-homology domain 3)-only protein Bim (Bcl-2-interacting mediator of cell death). Caspase-2 activation is dependent on the adaptor protein RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1β-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, and both caspase-2 and RAIDD are required for c-Jun activation and Bim induction. The present study thus shows that rapid caspase-2 activation is essential for c-Jun activation and Bim induction in neurons subjected to apoptotic stimuli. This places caspase-2 at an apical position in the apoptotic cascade and demonstrates for the first time that caspase-2 can regulate transcription.


American Journal of Human Genetics | 2016

Mutations in CRADD Result in Reduced Caspase-2-Mediated Neuronal Apoptosis and Cause Megalencephaly with a Rare Lissencephaly Variant

Nataliya Di Donato; Ying Y. Jean; A. Murat Maga; Briana D. Krewson; Alison B. Shupp; Maria I. Avrutsky; Achira Roy; Sarah Collins; Carissa Olds; Rebecca A. Willert; Agnieszka M. Czaja; Rachel Johnson; Jessi A. Stover; Steven M. Gottlieb; Deborah Bartholdi; Anita Rauch; Amy Goldstein; Victoria Boyd-Kyle; Kimberly A. Aldinger; Ghayda M. Mirzaa; Anke Nissen; Karlla W. Brigatti; Erik G. Puffenberger; Kathleen J. Millen; Kevin A. Strauss; William B. Dobyns; Carol M. Troy; Robert N. Jinks

Lissencephaly is a malformation of cortical development typically caused by deficient neuronal migration resulting in cortical thickening and reduced gyration. Here we describe a “thin” lissencephaly (TLIS) variant characterized by megalencephaly, frontal predominant pachygyria, intellectual disability, and seizures. Trio-based whole-exome sequencing and targeted re-sequencing identified recessive mutations of CRADD in six individuals with TLIS from four unrelated families of diverse ethnic backgrounds. CRADD (also known as RAIDD) is a death-domain-containing adaptor protein that oligomerizes with PIDD and caspase-2 to initiate apoptosis. TLIS variants cluster in the CRADD death domain, a platform for interaction with other death-domain-containing proteins including PIDD. Although caspase-2 is expressed in the developing mammalian brain, little is known about its role in cortical development. CRADD/caspase-2 signaling is implicated in neurotrophic factor withdrawal- and amyloid-β-induced dendritic spine collapse and neuronal apoptosis, suggesting a role in cortical sculpting and plasticity. TLIS-associated CRADD variants do not disrupt interactions with caspase-2 or PIDD in co-immunoprecipitation assays, but still abolish CRADD’s ability to activate caspase-2, resulting in reduced neuronal apoptosis in vitro. Homozygous Cradd knockout mice display megalencephaly and seizures without obvious defects in cortical lamination, supporting a role for CRADD/caspase-2 signaling in mammalian brain development. Megalencephaly and lissencephaly associated with defective programmed cell death from loss of CRADD function in humans implicate reduced apoptosis as an important pathophysiological mechanism of cortical malformation. Our data suggest that CRADD/caspase-2 signaling is critical for normal gyration of the developing human neocortex and for normal cognitive ability.


Journal of Visualized Experiments | 2015

Stereotaxic Infusion of Oligomeric Amyloid-beta into the Mouse Hippocampus

Ying Y. Jean; Jimena Baleriola; Mauro Fa; Ulrich Hengst; Carol M. Troy

Alzheimers disease is a neurodegenerative disease affecting the aging population. A key neuropathological feature of the disease is the over-production of amyloid-beta and the deposition of amyloid-beta plaques in brain regions of the afflicted individuals. Throughout the years scientists have generated numerous Alzheimers disease mouse models that attempt to replicate the amyloid-beta pathology. Unfortunately, the mouse models only selectively mimic the disease features. Neuronal death, a prominent effect in the brains of Alzheimers disease patients, is noticeably lacking in these mice. Hence, we and others have employed a method of directly infusing soluble oligomeric species of amyloid-beta - forms of amyloid-beta that have been proven to be most toxic to neurons - stereotaxically into the brain. In this report we utilize male C57BL/6J mice to document this surgical technique of increasing amyloid-beta levels in a select brain region. The infusion target is the dentate gyrus of the hippocampus because this brain structure, along with the basal forebrain that is connected by the cholinergic circuit, represents one of the areas of degeneration in the disease. The results of elevating amyloid-beta in the dentate gyrus via stereotaxic infusion reveal increases in neuron loss in the dentate gyrus within 1 week, while there is a concomitant increase in cell death and cholinergic neuron loss in the vertical limb of the diagonal band of Broca of the basal forebrain. These effects are observed up to 2 weeks. Our data suggests that the current amyloid-beta infusion model provides an alternative mouse model to address region specific neuron death in a short-term basis. The advantage of this model is that amyloid-beta can be elevated in a spatial and temporal manner.


Journal of Visualized Experiments | 2015

Detection of Axonally Localized mRNAs in Brain Sections Using High-Resolution In Situ Hybridization

Jimena Baleriola; Ying Y. Jean; Carol M. Troy; Ulrich Hengst

mRNAs are frequently localized to vertebrate axons and their local translation is required for axon pathfinding or branching during development and for maintenance, repair or neurodegeneration in postdevelopmental periods. High throughput analyses have recently revealed that axons have a more dynamic and complex transcriptome than previously expected. These analysis, however have been mostly done in cultured neurons where axons can be isolated from the somato-dendritic compartments. It is virtually impossible to achieve such isolation in whole tissues in vivo. Thus, in order to verify the recruitment of mRNAs and their functional relevance in a whole animal, transcriptome analyses should ideally be combined with techniques that allow the visualization of mRNAs in situ. Recently, novel ISH technologies that detect RNAs at a single-molecule level have been developed. This is especially important when analyzing the subcellular localization of mRNA, since localized RNAs are typically found at low levels. Here we describe two protocols for the detection of axonally-localized mRNAs using a novel ultrasensitive RNA ISH technology. We have combined RNAscope ISH with axonal counterstain using fluorescence immunohistochemistry or histological dyes to verify the recruitment of Atf4 mRNA to axons in vivo in the mature mouse and human brains.


Alzheimers & Dementia | 2015

Snps in cugbp2 influence the risk of Alzheimer disease in white and caribbean hispanic elderly and in adults with down syndrome

Joseph H. Lee; Rong Cheng; Ying Y. Jean; Lam-ha T. Dang; Giuseppe Tosto; Badri N. Vardarajan; Wayne Silverman; Nicole Schupf; Richard Mayeux; Carol M. Troy

throughout the genome with LOAD risk in 4,831 LOAD cases and 3,174 cognitively normal controls in the Alzheimer’s Disease Genetics Consortium (ADGC). Methods:We called CNVs using quality-controlled, high-density GWAS genotyping on the Illumina 660w and HumanOmniExpress SNP arrays in 8,005 participants from waves 1-6 of the ADC (Alzheimer’s Disease Center) subsets of the ADGC using the PennCNV program. Quality control excluded variants from telomeric, centromeric, and HLA regions and samples with excess standard deviation of allelic intensity [SD(LRR)>0.3] or G/C base content waviness factor [jGCWFj>0.05 or more than 100 CNVs genome-wide. We analyzed 31,007 duplications (n1⁄47,411) and 63,200 deletions (n1⁄47,628) using joint SNV-CNVassociation in ParseCNV. Results: We observed an average of 12.16 CNVs per individuals (4.01 duplications and 8.16 deletions), and no significant quantitative difference between cases and controls. Joint association testing in ParseCNV identified 3 significant deletions enriched in cases, and 1 deletion and 1 duplication enriched in controls (OR>2; contained 10 more SNPs; uncorrected two-tailed P<5310), all with frequencies<3%. Joint SNV/CNV gene-based testing identified 17 genes in or near deletions and 7 genes in or near duplications associated with LOAD at gene-wide significance (uncorrected two-tailed P<5310). We estimated heterogeneity in associations by genotyping platforms using METAL, which identified CNVassociations of P<5310 in 5 genes in or near deletions (EYS, TYRP1, RAB9BP1, ASCC3, FSTL5) but not duplications. Conclusions: Our findings suggest several rare deletion CNVs with strong effects may contribute to LOAD risk, however confirmation in independent datasets is needed. Additional work to clarify the frequency of these CNVs in large population samples is ongoing using case-only rare CNV annotation in population databases.


Alzheimers & Dementia | 2012

Development of an induced pluripotent stem cell (iPSC) Alzheimer's disease model using PSEN1 mutant fibroblasts

Andrew A. Sproul; Samson Jacob; Michael W. Nestor; Serene Keilani; Ying Y. Jean; David J. Kahler; Ismael Santa-Maria; John W. Steele; John F. Crary; Carol M. Troy; Sam Gandy; Scott Noggle

Collaboration


Dive into the Ying Y. Jean's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John F. Crary

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

A. Murat Maga

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Achira Roy

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Amy Goldstein

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge