Network


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

Hotspot


Dive into the research topics where Brian Halabisky is active.

Publication


Featured researches published by Brian Halabisky.


The Journal of Neuroscience | 2011

Amyloid-β/Fyn–Induced Synaptic, Network, and Cognitive Impairments Depend on Tau Levels in Multiple Mouse Models of Alzheimer's Disease

Erik D. Roberson; Brian Halabisky; Jong W. Yoo; Jinghua Yao; Jeannie Chin; Fengrong Yan; Tiffany Wu; Patricia Hamto; Nino Devidze; Gui-Qiu Yu; Jorge J. Palop; Jeffrey L. Noebels; Lennart Mucke

Alzheimers disease (AD), the most common neurodegenerative disorder, is a growing public health problem and still lacks effective treatments. Recent evidence suggests that microtubule-associated protein tau may mediate amyloid-β peptide (Aβ) toxicity by modulating the tyrosine kinase Fyn. We showed previously that tau reduction prevents, and Fyn overexpression exacerbates, cognitive deficits in human amyloid precursor protein (hAPP) transgenic mice overexpressing Aβ. However, the mechanisms by which Aβ, tau, and Fyn cooperate in AD-related pathogenesis remain to be fully elucidated. Here we examined the synaptic and network effects of this pathogenic triad. Tau reduction prevented cognitive decline induced by synergistic effects of Aβ and Fyn. Tau reduction also prevented synaptic transmission and plasticity deficits in hAPP mice. Using electroencephalography to examine network effects, we found that tau reduction prevented spontaneous epileptiform activity in multiple lines of hAPP mice. Tau reduction also reduced the severity of spontaneous and chemically induced seizures in mice overexpressing both Aβ and Fyn. To better understand these protective effects, we recorded whole-cell currents in acute hippocampal slices from hAPP mice with and without tau. hAPP mice with tau had increased spontaneous and evoked excitatory currents, reduced inhibitory currents, and NMDA receptor dysfunction. Tau reduction increased inhibitory currents and normalized excitation/inhibition balance and NMDA receptor-mediated currents in hAPP mice. Our results indicate that Aβ, tau, and Fyn jointly impair synaptic and network function and suggest that disrupting the copathogenic relationship between these factors could be of therapeutic benefit.


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.


Nature Neuroscience | 2008

Phospholipase A2 reduction ameliorates cognitive deficits in a mouse model of Alzheimer's disease

Rene O. Sanchez-Mejia; John W. Newman; Sandy Toh; Gui-Qiu Yu; Yungui Zhou; Brian Halabisky; Moustapha Cissé; Kimberly Scearce-Levie; Irene H. Cheng; Li Gan; Jorge J. Palop; Joseph V. Bonventre; Lennart Mucke

Neuronal expression of familial Alzheimers disease–mutant human amyloid precursor protein (hAPP) and hAPP-derived amyloid-β (Aβ) peptides causes synaptic dysfunction, inflammation and abnormal cerebrovascular tone in transgenic mice. Fatty acids may be involved in these processes, but their contribution to Alzheimers disease pathogenesis is uncertain. We used a lipidomics approach to generate a broad profile of fatty acids in brain tissues of hAPP-expressing mice and found an increase in arachidonic acid and its metabolites, suggesting increased activity of the group IV isoform of phospholipase A2 (GIVA-PLA2). The levels of activated GIVA-PLA2 in the hippocampus were increased in individuals with Alzheimers disease and in hAPP mice. Aβ caused a dose-dependent increase in GIVA-PLA2 phosphorylation in neuronal cultures. Inhibition of GIVA-PLA2 diminished Aβ-induced neurotoxicity. Genetic ablation or reduction of GIVA-PLA2 protected hAPP mice against Aβ-dependent deficits in learning and memory, behavioral alterations and premature mortality. Inhibition of GIVA-PLA2 may be beneficial in the treatment and prevention of Alzheimers disease.


Journal of Biological Chemistry | 2011

CX3CR1 protein signaling modulates microglial activation and protects against plaque-independent cognitive deficits in a mouse model of Alzheimer disease.

Seo Hyun Cho; Binggui Sun; Yungui Zhou; Tiina M. Kauppinen; Brian Halabisky; Paul D. Wes; Richard M. Ransohoff; Li Gan

Aberrant microglial activation has been proposed to contribute to the cognitive decline in Alzheimer disease (AD), but the underlying molecular mechanisms remain enigmatic. Fractalkine signaling, a pathway mediating the communication between microglia and neurons, is deficient in AD brains and down-regulated by amyloid-β. Although fractalkine receptor (CX3CR1) on microglia was found to regulate plaque load, no functional effects have been reported. Our study demonstrates that CX3CR1 deficiency worsens the AD-related neuronal and behavioral deficits. The effects were associated with cytokine production but not with plaque deposition. Ablation of CX3CR1 in mice overexpressing human amyloid precursor protein enhanced Tau pathology and exacerbated the depletion of calbindin in the dentate gyrus. The levels of calbindin in the dentate gyrus correlated negatively with those of tumor necrosis factor α and interleukin 6, suggesting neurotoxic effects of inflammatory factors. Functionally, removing CX3CR1 in human amyloid precursor protein mice worsened the memory retention in passive avoidance and novel object recognition tests, and their memory loss in the novel object recognition test is associated with high levels of interleukin 6. Our findings identify CX3CR1 as a key microglial pathway in protecting against AD-related cognitive deficits that are associated with aberrant microglial activation and elevated inflammatory cytokines.


Neuron | 2008

Cystatin C-Cathepsin B Axis Regulates Amyloid Beta Levels and Associated Neuronal Deficits in an Animal Model of Alzheimer's Disease

Binggui Sun; Yungui Zhou; Brian Halabisky; Iris Lo; Seo-hyun Cho; Sarah Mueller-Steiner; Nino Devidze; Xin Wang; Anders Grubb; Li Gan

Impaired degradation of amyloid beta (Abeta) peptides could lead to Abeta accumulation, an early trigger of Alzheimers disease (AD). How Abeta-degrading enzymes are regulated remains largely unknown. Cystatin C (CysC, CST3) is an endogenous inhibitor of cysteine proteases, including cathepsin B (CatB), a recently discovered Abeta-degrading enzyme. A CST3 polymorphism is associated with an increased risk of late-onset sporadic AD. Here, we identified CysC as the key inhibitor of CatB-induced Abeta degradation in vivo. Genetic ablation of CST3 in hAPP-J20 mice significantly lowered soluble Abeta levels, the relative abundance of Abeta1-42, and plaque load. CysC removal also attenuated Abeta-associated cognitive deficits and behavioral abnormalities and restored synaptic plasticity in the hippocampus. Importantly, the beneficial effects of CysC reduction were abolished on a CatB null background, providing direct evidence that CysC regulates soluble Abeta and Abeta-associated neuronal deficits through inhibiting CatB-induced Abeta degradation.


Cell Stem Cell | 2009

GABAergic Interneuron Dysfunction Impairs Hippocampal Neurogenesis in Adult Apolipoprotein E4 Knockin Mice

Gang Li; Nga Bien-Ly; Yaisa Andrews-Zwilling; Qin Xu; Aubrey Bernardo; Karen Ring; Brian Halabisky; Changhui Deng; Robert W. Mahley; Yadong Huang

Apolipoprotein (apo) E, a polymorphic protein with three isoforms (apoE2, apoE3, and apoE4), is essential for lipid homeostasis. Carriers of apoE4 are at higher risk for developing Alzheimers disease. We have investigated adult neurogenesis in mice with knockout (KO) for apoE or with knockin (KI) alleles for human apoE3 or apoE4, and we report that neurogenesis is reduced in both apoE-KO and apoE4-KI mice. In apoE-KO mice, increased BMP signaling promoted glial differentiation at the expense of neurogenesis. In contrast, in apoE4-KI mice, presynaptic GABAergic input-mediated maturation of newborn neurons was diminished. Tau phosphorylation, an Alzheimers disease characteristic, and levels of neurotoxic apoE fragments were both elevated in apoE4-KI hippocampal neurons concomitant with decreased GABAergic interneuron survival. Potentiating GABAergic signaling restored neuronal maturation and neurogenesis in apoE4-KI mice to normal levels. These findings suggest that GABAergic signaling can be targeted to mitigate the deleterious effects of apoE4 on neurogenesis.


Cell Stem Cell | 2009

Imbalance between GABAergic and Glutamatergic Transmission Impairs Adult Neurogenesis in an Animal Model of Alzheimer's Disease

Binggui Sun; Brian Halabisky; Yungui Zhou; Jorge J. Palop; Gui-Qiu Yu; Lennart Mucke; Li Gan

Adult neurogenesis regulates plasticity and function in the hippocampus, which is critical for memory and vulnerable to Alzheimers disease (AD). Promoting neurogenesis may improve hippocampal function in AD brains. However, how amyloid beta (Abeta), the key AD pathogen, affects the development and function of adult-born neurons remains unknown. Adult-born granule cells (GCs) in human amyloid precursor protein (hAPP) transgenic mice, an AD model, showed greater dendritic length, spine density, and functional responses than did controls early in development, but were impaired morphologically and functionally during later maturation. Early inhibition of GABA(A) receptors to suppress GABAergic signaling or late inhibition of calcineurin to enhance glutamatergic signaling normalized the development of adult-born GCs in hAPP mice with high Abeta levels. Abeta-induced increases in GABAergic neurotransmission or an imbalance between GABAergic and glutamatergic neurotransmission may contribute to impaired neurogenesis in AD.


The Journal of Neuroscience | 2010

Many Neuronal and Behavioral Impairments in Transgenic Mouse Models of Alzheimer’s Disease Are Independent of Caspase Cleavage of the Amyloid Precursor Protein

Julie A. Harris; Nino Devidze; Brian Halabisky; Iris Lo; Myo T. Thwin; Gui-Qiu Yu; Dale E. Bredesen; Eliezer Masliah; Lennart Mucke

Previous studies suggested that cleavage of the amyloid precursor protein (APP) at aspartate residue 664 by caspases may play a key role in the pathogenesis of Alzheimers disease. Mutation of this site (D664A) prevents caspase cleavage and the generation of the C-terminal APP fragments C31 and Jcasp, which have been proposed to mediate amyloid-β (Aβ) neurotoxicity. Here we compared human APP transgenic mice with (B254) and without (J20) the D664A mutation in a battery of tests. Before Aβ deposition, hAPP–B254 and hAPP–J20 mice had comparable hippocampal levels of Aβ1-42. At 2–3 or 5–7 months of age, hAPP–B254 and hAPP–J20 mice had similar abnormalities relative to nontransgenic mice in spatial and nonspatial learning and memory, elevated plus maze performance, electrophysiological measures of synaptic transmission and plasticity, and levels of synaptic activity-related proteins. Thus, caspase cleavage of APP at position D664 and generation of C31 do not play a critical role in the development of these abnormalities.


Alzheimers & Dementia | 2008

P3-384: Impaired hippocampal adult neurogenesis in an animal model of Alzheimer's disease

Brian Halabisky; Binggui Sun; Yungui Zhou; Sarah Mueller-Steiner; Xin Wang; Giuqiu Yu; Lennart Mucke; Li Gan

Background: The hippocampal formation is severely affected by Alzheimer’s disease (AD). It contains the dentate gyrus (DG), which is among the few brain regions in adult rodents and humans in which new neurons are continually born and functionally integrated into the neural network. How A and AD affect the newborn neurons remains to be fully elucidated. Our objective was to assess the effect of amyloid(A ) on neurogenesis, determine underlying mechanisms, and develop strategies to counteract relevant pathogenic processes. Methods: We used human amyloid precursor protein (hAPP) transgenic mice from line J20 and nontransgenic controls at 3 weeks, 2-3 months or 6-7 months of age. Newborn neurons in the subgranular zone of their DG were labeled by stereotaxic injection of a GFP-expressing retroviral vector that only labels cycling progenitor cells. Neuronal dendrites and spines were quantitated by computer-assisted analysis of confocal microscopic images and electrophysiological responses by patch-clamp recordings. Results: At 2-3 weeks after the birth of adult-born neurons, when their response to GABAergic stimulation is still excitatory, adult-born granule cells in hAPP mice had more dendritic spines, received more GABAergic input, and showed stronger excitatory synaptic transmission than adult-born granule cells in controls. New granule cells in hAPP mice also exhibited an accelerated switch from a depolarizing to a hyperpolarizing chloride reversal potential. Thus, adult-born neurons in hAPP mice initially appear to undergo an abnormally accelerated development, which may relate to the excessive GABAergic sprouting in the dentate gyrus of hAPP mice (Neuron 55:697). By 4 weeks after the birth of adult-born neurons, when their response to GABAergic stimulation had switched from excitatory to inhibitory, adult-born granule cells in hAPP mice showed shorter dendrites, fewer dendritic spines, and poorer functional integration into the DG circuitry than controls. Importantly, nicotine treatment restored the normal dendritic and functional development of adult-born neurons in hAPP mice. Conclusions: hAPP/A first accelerates and then impairs the development of adult-born granule cells, possibly through increases in GABAergic input. Nicotine treatment blocks this process, possibly by improving the balance between excitatory and inhibitory activities. Supported by Whittier Foundation.


Archive | 2010

Transsynaptic progression of amyloid-beta-induced neuronal dysfunction within the entorhinal-hippoca

John Harris; Nino Devidze; Laure Verret; K. C. Ho; Brian Halabisky; Myo T. Thwin; Dong-Kyun Kim; Patricia Hamto; Irene M. C. Lo; Guan-Qiao Yu; Jorge J. Palop; Eliezer Masliah; Lennart Mucke

Collaboration


Dive into the Brian Halabisky's collaboration.

Top Co-Authors

Avatar

Lennart Mucke

University of California

View shared research outputs
Top Co-Authors

Avatar

Binggui Sun

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

Li Gan

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yungui Zhou

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

Iris Lo

University of California

View shared research outputs
Top Co-Authors

Avatar

Julie A. Harris

Allen Institute for Brain Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge