Network


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

Hotspot


Dive into the research topics where Kaitlyn Ho is active.

Publication


Featured researches published by Kaitlyn Ho.


Neuron | 2007

Aberrant Excitatory Neuronal Activity and Compensatory Remodeling of Inhibitory Hippocampal Circuits in Mouse Models of Alzheimer's Disease

Jorge J. Palop; Jeannie Chin; Erik D. Roberson; Jun Wang; Myo T. Thwin; Nga Bien-Ly; Jong Yoo; Kaitlyn Ho; Gui-Qiu Yu; Anatol C. Kreitzer; Steven Finkbeiner; Jeffrey L. Noebels; Lennart Mucke

Neural network dysfunction may play an important role in Alzheimers disease (AD). Neuronal circuits vulnerable to AD are also affected in human amyloid precursor protein (hAPP) transgenic mice. hAPP mice with high levels of amyloid-beta peptides in the brain develop AD-like abnormalities, including cognitive deficits and depletions of calcium-related proteins in the dentate gyrus, a region critically involved in learning and memory. Here, we report that hAPP mice have spontaneous nonconvulsive seizure activity in cortical and hippocampal networks, which is associated with GABAergic sprouting, enhanced synaptic inhibition, and synaptic plasticity deficits in the dentate gyrus. Many Abeta-induced neuronal alterations could be simulated in nontransgenic mice by excitotoxin challenge and prevented in hAPP mice by blocking overexcitation. Aberrant increases in network excitability and compensatory inhibitory mechanisms in the hippocampus may contribute to Abeta-induced neurological deficits in hAPP mice and, possibly, also in humans with AD.


Nature | 2011

Reversing EphB2 depletion rescues cognitive functions in Alzheimer model

Moustapha Cissé; Brian Halabisky; Julie A. Harris; Nino Devidze; Dena B. Dubal; Binggui Sun; Anna G. Orr; Gregor Lotz; Daniel H. Kim; Patricia Hamto; Kaitlyn Ho; Gui-Qiu Yu; Lennart Mucke

Amyloid-β oligomers may cause cognitive deficits in Alzheimer’s disease by impairing neuronal NMDA-type glutamate receptors, whose function is regulated by the receptor tyrosine kinase EphB2. Here we show that amyloid-β oligomers bind to the fibronectin repeats domain of EphB2 and trigger EphB2 degradation in the proteasome. To determine the pathogenic importance of EphB2 depletions in Alzheimer’s disease and related models, we used lentiviral constructs to reduce or increase neuronal expression of EphB2 in memory centres of the mouse brain. In nontransgenic mice, knockdown of EphB2 mediated by short hairpin RNA reduced NMDA receptor currents and impaired long-term potentiation in the dentate gyrus, which are important for memory formation. Increasing EphB2 expression in the dentate gyrus of human amyloid precursor protein transgenic mice reversed deficits in NMDA receptor-dependent long-term potentiation and memory impairments. Thus, depletion of EphB2 is critical in amyloid-β-induced neuronal dysfunction. Increasing EphB2 levels or function could be beneficial in Alzheimer’s disease.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Levetiracetam suppresses neuronal network dysfunction and reverses synaptic and cognitive deficits in an Alzheimer’s disease model

Pascal E. Sanchez; Lei Zhu; Laure Verret; Keith A. Vossel; Anna G. Orr; John R. Cirrito; Nino Devidze; Kaitlyn Ho; Gui Qiu Yu; Jorge J. Palop; Lennart Mucke

In light of the rising prevalence of Alzheimer’s disease (AD), new strategies to prevent, halt, and reverse this condition are needed urgently. Perturbations of brain network activity are observed in AD patients and in conditions that increase the risk of developing AD, suggesting that aberrant network activity might contribute to AD-related cognitive decline. Human amyloid precursor protein (hAPP) transgenic mice simulate key aspects of AD, including pathologically elevated levels of amyloid-β peptides in brain, aberrant neural network activity, remodeling of hippocampal circuits, synaptic deficits, and behavioral abnormalities. Whether these alterations are linked in a causal chain remains unknown. To explore whether hAPP/amyloid-β–induced aberrant network activity contributes to synaptic and cognitive deficits, we treated hAPP mice with different antiepileptic drugs. Among the drugs tested, only levetiracetam (LEV) effectively reduced abnormal spike activity detected by electroencephalography. Chronic treatment with LEV also reversed hippocampal remodeling, behavioral abnormalities, synaptic dysfunction, and deficits in learning and memory in hAPP mice. Our findings support the hypothesis that aberrant network activity contributes causally to synaptic and cognitive deficits in hAPP mice. LEV might also help ameliorate related abnormalities in people who have or are at risk for AD.


Nature Neuroscience | 2013

Physiologic brain activity causes DNA double-strand breaks in neurons, with exacerbation by amyloid-β.

Elsa Suberbielle; Pascal E. Sanchez; Alexxai V. Kravitz; Xin Wang; Kaitlyn Ho; Nino Devidze; Anatol C. Kreitzer; Lennart Mucke

We show that a natural behavior, exploration of a novel environment, causes DNA double-strand breaks (DSBs) in neurons of young adult wild-type mice. DSBs occurred in multiple brain regions, were most abundant in the dentate gyrus, which is involved in learning and memory, and were repaired within 24 h. Increasing neuronal activity by sensory or optogenetic stimulation increased neuronal DSBs in relevant but not irrelevant networks. Mice transgenic for human amyloid precursor protein (hAPP), which simulate key aspects of Alzheimers disease, had increased neuronal DSBs at baseline and more severe and prolonged DSBs after exploration. Interventions that suppress aberrant neuronal activity and improve learning and memory in hAPP mice normalized their levels of DSBs. Blocking extrasynaptic NMDA-type glutamate receptors prevented amyloid-β (Aβ)-induced DSBs in neuronal cultures. Thus, transient increases in neuronal DSBs occur as a result of physiological brain activity, and Aβ exacerbates DNA damage, most likely by eliciting synaptic dysfunction.


The Journal of Neuroscience | 2009

Neprilysin Overexpression Inhibits Plaque Formation But Fails to Reduce Pathogenic Aβ Oligomers and Associated Cognitive Deficits in Human Amyloid Precursor Protein Transgenic Mice

William J. Meilandt; Moustapha Cissé; Kaitlyn Ho; Tiffany Wu; Luke Esposito; Kimberly Scearce-Levie; Irene H. Cheng; Gui-Qiu Yu; Lennart Mucke

The accumulation of amyloid-β (Aβ) peptides in the brain of patients with Alzheimers disease (AD) may arise from an imbalance between Aβ production and clearance. Overexpression of the Aβ-degrading enzyme neprilysin in brains of human amyloid precursor protein (hAPP) transgenic mice decreases overall Aβ levels and amyloid plaque burdens. Because AD-related synaptic and cognitive deficits appear to be more closely related to Aβ oligomers than to plaques, it is important to determine whether increased neprilysin activity also diminishes the levels of pathogenic Aβ oligomers and related neuronal deficits in vivo. To address this question, we crossed hAPP transgenic mice with neprilysin transgenic mice and analyzed their offspring. Neprilysin overexpression reduced soluble Aβ levels by 50% and effectively prevented early Aβ deposition in the neocortex and hippocampus. However, it did not reduce levels of Aβ trimers and Aβ*56 or improve deficits in spatial learning and memory. The differential effect of neprilysin on plaques and oligomers suggests that neprilysin-dependent degradation of Aβ affects plaques more than oligomers and that these structures may form through distinct assembly mechanisms. Neprilysins inability to prevent learning and memory deficits in hAPP mice may be related to its inability to reduce pathogenic Aβ oligomers. Reduction of Aβ oligomers will likely be required for anti-Aβ treatments to improve cognitive functions.


Cell Reports | 2014

Life Extension Factor Klotho Enhances Cognition

Dena B. Dubal; Jennifer S. Yokoyama; Lei Zhu; Lauren Broestl; Kurtresha Worden; Dan Wang; Virginia E. Sturm; Daniel H. Kim; Eric Klein; Gui Qiu Yu; Kaitlyn Ho; Lei Yu; Makoto Kuro-o; Philip L. De Jager; Giovanni Coppola; Gary W. Small; David A. Bennett; Joel H. Kramer; Carmela R. Abraham; Bruce L. Miller; Lennart Mucke

Aging is the primary risk factor for cognitive decline, an emerging health threat to aging societies worldwide. Whether anti-aging factors such as klotho can counteract cognitive decline is unknown. We show that a lifespan-extending variant of the human KLOTHO gene, KL-VS, is associated with enhanced cognition in heterozygous carriers. Because this allele increased klotho levels in serum, we analyzed transgenic mice with systemic overexpression of klotho. They performed better than controls in multiple tests of learning and memory. Elevating klotho in mice also enhanced long-term potentiation, a form of synaptic plasticity, and enriched synaptic GluN2B, an N-methyl-D-aspartate receptor (NMDAR) subunit with key functions in learning and memory. Blockade of GluN2B abolished klotho-mediated effects. Surprisingly, klotho effects were evident also in young mice and did not correlate with age in humans, suggesting independence from the aging process. Augmenting klotho or its effects may enhance cognition and counteract cognitive deficits at different life stages.


Nature Neuroscience | 2015

Astrocytic adenosine receptor A2A and Gs-coupled signaling regulate memory

Anna G. Orr; Edward C. Hsiao; Max M Wang; Kaitlyn Ho; Daniel H. Kim; Xin Wang; Weikun Guo; Jing Kang; Gui-Qiu Yu; Anthony Adame; Nino Devidze; Dena B. Dubal; Eliezer Masliah; Bruce R. Conklin; Lennart Mucke

Astrocytes express a variety of G protein–coupled receptors and might influence cognitive functions, such as learning and memory. However, the roles of astrocytic Gs-coupled receptors in cognitive function are not known. We found that humans with Alzheimers disease (AD) had increased levels of the Gs-coupled adenosine receptor A2A in astrocytes. Conditional genetic removal of these receptors enhanced long-term memory in young and aging mice and increased the levels of Arc (also known as Arg3.1), an immediate-early gene that is required for long-term memory. Chemogenetic activation of astrocytic Gs-coupled signaling reduced long-term memory in mice without affecting learning. Like humans with AD, aging mice expressing human amyloid precursor protein (hAPP) showed increased levels of astrocytic A2A receptors. Conditional genetic removal of these receptors enhanced memory in aging hAPP mice. Together, these findings establish a regulatory role for astrocytic Gs-coupled receptors in memory and suggest that AD-linked increases in astrocytic A2A receptor levels contribute to memory loss.


The Journal of Neuroscience | 2011

Ablation of Cellular Prion Protein Does Not Ameliorate Abnormal Neural Network Activity or Cognitive Dysfunction in the J20 Line of Human Amyloid Precursor Protein Transgenic Mice

Moustapha Cissé; Pascal E. Sanchez; Daniel H. Kim; Kaitlyn Ho; Gui-Qiu Yu; Lennart Mucke

Previous studies suggested that the cellular prion protein (PrPc) plays a critical role in the pathogenesis of Alzheimers disease (AD). Specifically, amyloid-β (Aβ) oligomers were proposed to cause synaptic and cognitive dysfunction by binding to PrPc. To test this hypothesis, we crossed human amyloid precursor protein (hAPP) transgenic mice from line J20 onto a PrPc-deficient background. Ablation of PrPc did not prevent the premature mortality and abnormal neural network activity typically seen in hAPPJ20 mice. Furthermore, hAPPJ20 mice with or without PrPc expression showed comparably robust abnormalities in learning and memory and in other behavioral domains at 6–8 months of age. Notably, these abnormalities are not refractory to therapeutic manipulations in general: they can be effectively prevented by interventions that prevent Aβ-dependent neuronal dysfunction also in other lines of hAPP transgenic mice. Thus, at least in this model, PrPc is not an important mediator of Aβ-induced neurological impairments.


PLOS ONE | 2012

Human P301L-mutant tau expression in mouse entorhinal-hippocampal network causes tau aggregation and presynaptic pathology but no cognitive deficits.

Julie A. Harris; Akihiko Koyama; Sumihiro Maeda; Kaitlyn Ho; Nino Devidze; Dena B. Dubal; Gui Qiu Yu; Eliezer Masliah; Lennart Mucke

Accumulation of hyperphosphorylated tau in the entorhinal cortex (EC) is one of the earliest pathological hallmarks in patients with Alzheimer’s disease (AD). It can occur before significant Aβ deposition and appears to “spread” into anatomically connected brain regions. To determine whether this early-stage pathology is sufficient to cause disease progression and cognitive decline in experimental models, we overexpressed mutant human tau (hTauP301L) predominantly in layer II/III neurons of the mouse EC. Cognitive functions remained normal in mice at 4, 8, 12 and 16 months of age, despite early and extensive tau accumulation in the EC. Perforant path (PP) axon terminals within the dentate gyrus (DG) contained abnormal conformations of tau even in young EC-hTau mice, and phosphorylated tau increased with age in both the EC and PP. In old mice, ultrastructural alterations in presynaptic terminals were observed at PP-to-granule cell synapses. Phosphorylated tau was more abundant in presynaptic than postsynaptic elements. Human and pathological tau was also detected within hippocampal neurons of this mouse model. Thus, hTauP301L accumulation predominantly in the EC and related presynaptic pathology in hippocampal circuits was not sufficient to cause robust cognitive deficits within the age range analyzed here.


The Journal of Neuroscience | 2015

Life extension factor klotho prevents mortality and enhances cognition in hAPP transgenic mice

Dena B. Dubal; Lei Zhu; Pascal E. Sanchez; Kurtresha Worden; Lauren Broestl; Erik Johnson; Kaitlyn Ho; Gui Qiu Yu; Daniel H. Kim; Alexander Betourne; Makoto Kuro-o; Eliezer Masliah; Carmela R. Abraham; Lennart Mucke

Aging is the principal demographic risk factor for Alzheimer disease (AD), the most common neurodegenerative disorder. Klotho is a key modulator of the aging process and, when overexpressed, extends mammalian lifespan, increases synaptic plasticity, and enhances cognition. Whether klotho can counteract deficits related to neurodegenerative diseases, such as AD, is unknown. Here we show that elevating klotho expression decreases premature mortality and network dysfunction in human amyloid precursor protein (hAPP) transgenic mice, which simulate key aspects of AD. Increasing klotho levels prevented depletion of NMDA receptor (NMDAR) subunits in the hippocampus and enhanced spatial learning and memory in hAPP mice. Klotho elevation in hAPP mice increased the abundance of the GluN2B subunit of NMDAR in postsynaptic densities and NMDAR-dependent long-term potentiation, which is critical for learning and memory. Thus, increasing wild-type klotho levels or activities improves synaptic and cognitive functions, and may be of therapeutic benefit in AD and other cognitive disorders.

Collaboration


Dive into the Kaitlyn Ho's collaboration.

Top Co-Authors

Avatar

Lennart Mucke

University of California

View shared research outputs
Top Co-Authors

Avatar

Gui-Qiu Yu

University of California

View shared research outputs
Top Co-Authors

Avatar

Daniel H. Kim

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dena B. Dubal

University of California

View shared research outputs
Top Co-Authors

Avatar

Jorge J. Palop

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gui Qiu Yu

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge