Network


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

Hotspot


Dive into the research topics where Christina R. Muratore is active.

Publication


Featured researches published by Christina R. Muratore.


Human Molecular Genetics | 2014

The familial Alzheimer's disease APPV717I mutation alters APP processing and Tau expression in iPSC-derived neurons

Christina R. Muratore; Heather C. Rice; Priya Srikanth; Dana G. Callahan; Taehwan Shin; Lawrence N. P. Benjamin; Dominic M. Walsh; Dennis J. Selkoe; Tracy L. Young-Pearse

Alzheimers disease (AD) is a complex neurodegenerative disorder characterized by extracellular plaques containing amyloid β (Aβ)-protein and intracellular tangles containing hyperphosphorylated Tau protein. Here, we describe the generation of inducible pluripotent stem cell lines from patients harboring the London familial AD (fAD) amyloid precursor protein (APP) mutation (V717I). We examine AD-relevant phenotypes following directed differentiation to forebrain neuronal fates vulnerable in AD. We observe that over differentiation time to mature neuronal fates, APP expression and levels of Aβ increase dramatically. In both immature and mature neuronal fates, the APPV717I mutation affects both β- and γ-secretase cleavage of APP. Although the mutation lies near the γ-secretase cleavage site in the transmembrane domain of APP, we find that β-secretase cleavage of APP is elevated leading to generation of increased levels of both APPsβ and Aβ. Furthermore, we find that this mutation alters the initial cleavage site of γ-secretase, resulting in an increased generation of both Aβ42 and Aβ38. In addition to altered APP processing, an increase in levels of total and phosphorylated Tau is observed in neurons with the APPV717I mutation. We show that treatment with Aβ-specific antibodies early in culture reverses the phenotype of increased total Tau levels, implicating altered Aβ production in fAD neurons in this phenotype. These studies use human neurons to reveal previously unrecognized effects of the most common fAD APP mutation and provide a model system for testing therapeutic strategies in the cell types most relevant to disease processes.


The Journal of Neuroscience | 2015

C-Terminally Truncated Forms of Tau, But Not Full-Length Tau or Its C-Terminal Fragments, Are Released from Neurons Independently of Cell Death

Daniel Kanmert; Adam Cantlon; Christina R. Muratore; Ming Jin; Tiernan T. O'Malley; Gloria Lee; Tracy L. Young-Pearse; Dennis J. Selkoe; Dominic M. Walsh

Recent evidence suggests that tau aggregation may spread via extracellular release and subsequent uptake by synaptically connected neurons, but little is known about the processes by which tau is released or the molecular forms of extracellular tau. To gain insight into the nature of extracellular tau, we used highly sensitive ELISAs, which, when used in tandem, are capable of differentiating between full-length (FL) tau, mid-region-bearing fragments, and C-terminal (CT) fragments. We applied these assays to the systematic study of the conditioned media of N2a cells, induced pluripotent stem cell-derived human cortical neurons, and primary rat cortical neurons, each of which was carefully assessed for viability. In all three neuronal models, the bulk of extracellular tau was free-floating and unaggregated and <0.2% was encapsulated in exosomes. Although most intracellular tau was FL, the majority of extracellular tau was CT truncated and appeared to be released both actively by living neurons and passively by dead cells. In contrast, only a small amount of extracellular tau was aggregation-competent tau (i.e., contained the microtubule-binding regions) and this material appears to be released solely due to a low level of cell death that occurs in all cell culture systems. Importantly, amyloid β-protein (Aβ)-induced neuronal compromise significantly increased the quantity of all forms of extracellular tau, but the presence of Aβ before detectable cell compromise did not increase extracellular tau. Collectively, these results suggest that factors that induce neuronal death are likely to be necessary to initiate the extracellular spread of tau aggregation. SIGNIFICANCE STATEMENT Recent studies suggest that the transfer of tau between neurons underlies the characteristic spatiotemporal progression of neurofibrillary pathology. We searched for tau in the conditioned medium of N2a cells, induced pluripotent stem cell-derived human cortical neurons, and primary rat cortical neurons and analyzed the material present using four different tau ELISAs. We demonstrate that the majority of tau released from healthy neurons is C-terminally truncated and lacks the microtubule-binding region (MTBR) thought necessary for self-aggregation. A small amount of MTBR-containing tau is present outside of cells, but this appears to be solely due to cell death. Therefore, if propagation of tau aggregation is mediated by extracellular tau, our findings suggest that neuronal compromise is required to facilitate this process.


Cell Reports | 2015

Genomic DISC1 Disruption in hiPSCs Alters Wnt Signaling and Neural Cell Fate

Priya Srikanth; Karam Han; Dana G. Callahan; Eugenia Makovkina; Christina R. Muratore; Matthew A. Lalli; Honglin Zhou; Justin D. Boyd; Kenneth S. Kosik; Dennis J. Selkoe; Tracy L. Young-Pearse

Genetic and clinical association studies have identified disrupted in schizophrenia 1 (DISC1) as a candidate risk gene for major mental illness. DISC1 is interrupted by a balanced chr(1;11) translocation in a Scottish family in which the translocation predisposes to psychiatric disorders. We investigate the consequences of DISC1 interruption in human neural cells using TALENs or CRISPR-Cas9 to target the DISC1 locus. We show that disruption of DISC1 near the site of the translocation results in decreased DISC1 protein levels because of nonsense-mediated decay of long splice variants. This results in an increased level of canonical Wnt signaling in neural progenitor cells and altered expression of fate markers such as Foxg1 and Tbr2. These gene expression changes are rescued by antagonizing Wnt signaling in a critical developmental window, supporting the hypothesis that DISC1-dependent suppression of basal Wnt signaling influences the distribution of cell types generated during cortical development.


Journal of Alzheimer's Disease | 2013

Soluble Oligomers of Amyloid-β Cause Changes in Redox State, DNA Methylation, and Gene Transcription by Inhibiting EAAT3 Mediated Cysteine Uptake

Nathaniel Hodgson; Malav Trivedi; Christina R. Muratore; Shaomin Li; Richard C. Deth

Oxidative stress, hyperhomocysteinemia, altered DNA methylation, and insulin resistance in the brain are associated with Alzheimers disease (AD), but the role of amyloid-β (Aβ) in these events remains unclear. Intracellular cysteine is rate-limiting for synthesis of the antioxidant glutathione (GSH), and factors regulating cysteine uptake exert a powerful influence over cellular redox status, especially in mature neurons where cysteine synthesis via transsulfuration of homocysteine (HCY) is restricted. We investigated the effect of soluble Aβ oligomers (oAβ) on basal and insulin-like growth factor-1 (IGF-1)-induced cysteine uptake mediated by the excitatory amino acid transporter 3 (EAAT3) in cultured human neuronal cells. We also examined the effect of oAβ on intracellular thiol metabolite levels, DNA methylation, and the transcription status of redox and methylation-associated genes. oAβ inhibited EAAT3-mediated cysteine uptake, causing a decrease in intracellular cysteine and GSH levels. The ratio of the methyl donor S-adenosylmethionine to the methylation inhibitor S-adenosylhomocysteine was decreased, in association with an increase in HCY and a global decrease in DNA methylation, indicative of decreased activity of the redox-sensitive enzyme methionine synthase. These metabolic effects of oAβ coincided with changes in the expression of redox and methylation pathway genes. The ability of oAβ to modulate gene expression via their redox and methylation-dependent epigenetic effects may contribute to the pathology of AD and recognition of this mechanism may lead to novel treatment approaches. We describe a role of IGF-1 signaling in regulating redox and methylation homeostasis, and propose this to be a pathogenic target of oAβ.


PLOS ONE | 2014

Comparison and Optimization of hiPSC Forebrain Cortical Differentiation Protocols

Christina R. Muratore; Priya Srikanth; Dana G. Callahan; Tracy L. Young-Pearse

Several protocols have been developed for human induced pluripotent stem cell neuronal differentiation. We compare several methods for forebrain cortical neuronal differentiation by assessing cell morphology, immunostaining and gene expression. We evaluate embryoid aggregate vs. monolayer with dual SMAD inhibition differentiation protocols, manual vs. AggreWell aggregate formation, plating substrates, neural progenitor cell (NPC) isolation methods, NPC maintenance and expansion, and astrocyte co-culture. The embryoid aggregate protocol, using a Matrigel substrate, consistently generates a high yield and purity of neurons. NPC isolation by manual selection, enzymatic rosette selection, or FACS all are efficient, but exhibit some differences in resulting cell populations. Expansion of NPCs as neural aggregates yields higher cell purity than expansion in a monolayer. Finally, co-culture of iPSC-derived neurons with astrocytes increases neuronal maturity by day 40. This study directly compares commonly employed methods for neuronal differentiation of iPSCs, and can be used as a resource for choosing between various differentiation protocols.


The Journal of Neuroscience | 2016

Single-Cell Detection of Secreted Aβ and sAPPα from Human IPSC-Derived Neurons and Astrocytes

Meichen Liao; Christina R. Muratore; Todd M. Gierahn; Sarah E. Sullivan; Priya Srikanth; Philip L. De Jager; J. Christopher Love; Tracy L. Young-Pearse

Secreted factors play a central role in normal and pathological processes in every tissue in the body. The brain is composed of a highly complex milieu of different cell types and few methods exist that can identify which individual cells in a complex mixture are secreting specific analytes. By identifying which cells are responsible, we can better understand neural physiology and pathophysiology, more readily identify the underlying pathways responsible for analyte production, and ultimately use this information to guide the development of novel therapeutic strategies that target the cell types of relevance. We present here a method for detecting analytes secreted from single human induced pluripotent stem cell (iPSC)-derived neural cells and have applied the method to measure amyloid β (Aβ) and soluble amyloid precursor protein-alpha (sAPPα), analytes central to Alzheimers disease pathogenesis. Through these studies, we have uncovered the dynamic range of secretion profiles of these analytes from single iPSC-derived neuronal and glial cells and have molecularly characterized subpopulations of these cells through immunostaining and gene expression analyses. In examining Aβ and sAPPα secretion from single cells, we were able to identify previously unappreciated complexities in the biology of APP cleavage that could not otherwise have been found by studying averaged responses over pools of cells. This technique can be readily adapted to the detection of other analytes secreted by neural cells, which would have the potential to open new perspectives into human CNS development and dysfunction. SIGNIFICANCE STATEMENT We have established a technology that, for the first time, detects secreted analytes from single human neurons and astrocytes. We examine secretion of the Alzheimers disease-relevant factors amyloid β (Aβ) and soluble amyloid precursor protein-alpha (sAPPα) and present novel findings that could not have been observed without a single-cell analytical platform. First, we identify a previously unappreciated subpopulation that secretes high levels of Aβ in the absence of detectable sAPPα. Further, we show that multiple cell types secrete high levels of Aβ and sAPPα, but cells expressing GABAergic neuronal markers are overrepresented. Finally, we show that astrocytes are competent to secrete high levels of Aβ and therefore may be a significant contributor to Aβ accumulation in the brain.


Stem cell reports | 2017

Cell-type Dependent Alzheimer's Disease Phenotypes: Probing the Biology of Selective Neuronal Vulnerability

Christina R. Muratore; Constance Zhou; Meichen Liao; Marty A. Fernandez; Walter M. Taylor; Valentina N. Lagomarsino; Richard Pearse; Heather C. Rice; Joseph Negri; Amy He; Priya Srikanth; Dana G. Callahan; Taehwan Shin; Monica Zhou; David A. Bennett; Scott Noggle; J. Christopher Love; Dennis J. Selkoe; Tracy L. Young-Pearse

Summary Alzheimers disease (AD) induces memory and cognitive impairment in the absence of motor and sensory deficits during its early and middle course. A major unresolved question is the basis for this selective neuronal vulnerability. Aβ, which plays a central role in AD pathogenesis, is generated throughout the brain, yet some regions outside of the limbic and cerebral cortices are relatively spared from Aβ plaque deposition and synapse loss. Here, we examine neurons derived from iPSCs of patients harboring an amyloid precursor protein mutation to quantify AD-relevant phenotypes following directed differentiation to rostral fates of the brain (vulnerable) and caudal fates (relatively spared) in AD. We find that both the generation of Aβ and the responsiveness of TAU to Aβ are affected by neuronal cell type, with rostral neurons being more sensitive than caudal neurons. Thus, cell-autonomous factors may in part dictate the pattern of selective regional vulnerability in human neurons in AD.


Translational Psychiatry | 2018

Shared effects of DISC1 disruption and elevated WNT signaling in human cerebral organoids

Priya Srikanth; Valentina N. Lagomarsino; Christina R. Muratore; Steven C. Ryu; Amy He; Walter M. Taylor; Constance Zhou; Marlise Arellano; Tracy L. Young-Pearse

The development of three-dimensional culture methods has allowed for the study of developing cortical morphology in human cells. This provides a new tool to study the neurodevelopmental consequences of disease-associated mutations. Here, we study the effects of isogenic DISC1 mutation in cerebral organoids. DISC1 has been implicated in psychiatric disease based on genetic studies, including its interruption by a balanced translocation that increases the risk of major mental illness. Isogenic wild-type and DISC1-disrupted human-induced pluripotent stem cells were used to generate cerebral organoids, which were then examined for morphology and gene expression. We show that DISC1-mutant cerebral organoids display disorganized structural morphology and impaired proliferation, which is phenocopied by WNT agonism and rescued by WNT antagonism. Furthermore, there are many shared changes in gene expression with DISC1 disruption and WNT agonism, including in neural progenitor and cell fate markers, regulators of neuronal migration, and interneuron markers. These shared gene expression changes suggest mechanisms for the observed morphologic dysregulation with DISC1 disruption and points to new avenues for future studies. The shared changes in three-dimensional cerebral organoid morphology and gene expression with DISC1 interruption and WNT agonism further strengthens the link between DISC1 mutation, abnormalities in WNT signaling, and neuropsychiatric disease.


Alzheimers & Dementia | 2018

AGE-DEPENDENT CHANGES IN HUMAN CORTEX ANTIOXIDANT AND METHYLATION METABOLIC PATHWAYS: IMPLICATIONS AND OPPORTUNITIES FOR ALZHEIMER'S DISEASE RISK REDUCTION

Richard C. Deth; Christina R. Muratore; Nathaniel Hodgson; Yiting Zhang; Malav Trivedi

Background:SORL1 is an established risk gene forAlzheimer’s disease (AD) that encodes the neuronal sortilin related receptor SORLA. Genome-wide association studies showed that SORL1 is associated with late-onset AD through several single nucleotide polymorphisms clustered in two independent haplotype blocks in the 50 and 30 regions of the gene, respectively. Moreover, SORL1 loss-of-functions variants were also recently identified in patients with early-onset AD providing direct evidence that SORL1 defects are causative of the disease. Methods: qPCR was used to quantify the expression of a novel SORL1 splicevariant inhuman tissues.Thecellular localizationof transcript and translation product was investigated by in-situ hybridization and immunohistochemistry, respectively. Characterization of the novel receptor protein was performed by immunocytochemistry, deglycosylation, and pulse-chase maturation studies. Results:We here describe a SORL1 transcript containing a novel exon located between exon38 and exon 39, named 38B, that is located within the 3’ risk haploblock region. We demonstrated the presence of this novel SORL1 transcript in various human tissues, showing the strongest expression in cerebellum. In addition, we found that SORL1-38B is reduced by >50% in the cerebellumof 25ADpatients compared to 25 control cerebellum samples. We observed a strong cellular retention of this novel variant despite the receptor protein lacks a transmembrane segment, but confirm the production of a stable translation product from this transcript. Within the human cerebellum SORL1-38B is mainly found in the soma and dendrites of Purkinje cells. Conclusions: The reduction of SORL1-38B levels in AD brain provides clues of a potential protective role for this alternative transcript in theonset ofAD,and the specific cerebellar expression might be related to new independent physiological functions of this variant. Accordingly, the novel SORL1 transcript represents a new candidateAD risk factor, and thus qualifies for further investigations to elaborate its impacts on AD pathology.


Alzheimers & Dementia | 2017

CELL-TYPE DEPENDENT ALZHEIMER'S DISEASE PHENOTYPES: PROBING THE BIOLOGY OF SELECTIVE NEURONAL VULNERABILITY

Christina R. Muratore

Collaboration


Dive into the Christina R. Muratore's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Priya Srikanth

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Dennis J. Selkoe

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Dana G. Callahan

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Heather C. Rice

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy He

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Constance Zhou

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Dominic M. Walsh

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

J. Christopher Love

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge