Network


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

Hotspot


Dive into the research topics where Xiao-Han Chen is active.

Publication


Featured researches published by Xiao-Han Chen.


Neurology | 1997

Genetic association of the low-density lipoprotein receptor-related protein gene (LRP), and apolipoprotein E receptor, with late-onset Alzheimer's disease

David E. Kang; Tsunao Saitoh; Xiao-Han Chen; Yu Xia; E. Masliah; L. A. Hansen; R. G. Thomas; L. J. Thal; Robert Katzman

The presence of the APOE ϵ4 allele encoding apolipoprotein E4 (apoE4) is the major genetic risk factor for late-onset Alzheimers disease (AD). However, the molecular and cellular mechanisms by which APOE ϵ4 renders AD risk are unclear. In this report, we present genetic evidence that an apoE receptor, LRP, may be associated with the expression of late-onset AD. Using a biallelic genetic marker in exon 3 LRP, late-onset AD cases markedly differed from the control subjects in the distribution of LRP genotypes, and this difference was highly accentuated among AD cases with positive family history of senile dementia. Furthermore, the numbers of neuritic plaques were significantly altered as a consequence of different LRP genotypes in postmortem AD cases. Taken together, our results implicate the pathophysiology of LRP in the expression of late-onset AD.


Experimental Neurology | 2007

Multiple proteins implicated in neurodegenerative diseases accumulate in axons after brain trauma in humans

Kunihiro Uryu; Xiao-Han Chen; Dan Martinez; Kevin D. Browne; Victoria E. Johnson; David I. Graham; Virginia M.-Y. Lee; John Q. Trojanowski; Douglas H. Smith

Studies in animal models have shown that traumatic brain injury (TBI) induces the rapid accumulation of many of the same key proteins that form pathologic aggregates in neurodegenerative diseases. Here, we examined whether this rapid process also occurs in humans after TBI. Brain tissue from 18 cases who died after TBI and from 6 control cases was examined using immunohistochemistry. Following TBI, widespread axonal injury was persistently identified by the accumulation of neurofilament protein and amyloid precursor protein (APP) in axonal bulbs and varicosities. Axonal APP was found to co-accumulate with its cleavage enzymes, beta-site APP cleaving enzyme (BACE), presenilin-1 (PS1) and their product, amyloid-beta (Abeta). In addition, extensive accumulation of alpha-synuclein (alpha-syn) was found in swollen axons and tau protein was found to accumulate in both axons and neuronal cell bodies. These data show rapid axonal accumulation of proteins implicated in neurodegenerative diseases including Alzheimers disease and the synucleinopathies. The cause of axonal pathology can be attributed to disruption of axons due to trauma, or as a secondary effect of raised intracranial pressure or hypoxia. Such axonal pathology in humans may provide a unique environment whereby co-accumulation of APP, BACE, and PS1 leads to intra-axonal production of Abeta as well as accumulation of alpha-syn and tau. This process may have important implications for survivors of TBI who have been shown to be at greater risk of developing neurodegenerative diseases.


Journal of Neuropathology and Experimental Neurology | 1999

Accumulation of Amyloid β and Tau and the Formation of Neurofilament Inclusions Following Diffuse Brain Injury in the Pig

Douglas H. Smith; Xiao-Han Chen; Masahiro Nonaka; John Q. Trojanowski; Virginia M.-Y. Lee; Kathryn E. Saatman; Matthew J. Leoni; Bai-Nan Xu; John A. Wolf; David F. Meaney

Brain trauma in humans increases the risk for developing Alzheimer disease (AD) and may induce the acute formation of AD-like plaques containing amyloid beta (A beta). To further explore the potential link between brain trauma and neurodegeneration, we conducted neuropathological studies using a pig model of diffuse brain injury. Brain injury was induced in anesthetized animals via nonimpact head rotational acceleration of 110 degrees over 20 ms in the coronal plane (n = 15 injured, n = 3 noninjured). At 1, 3, 7, and 10 days post-trauma, control and injured animals were euthanized and immunohistochemical analysis was performed on brain sections using antibodies specific for A beta, beta-amyloid precursor protein (betaPP), tau, and neurofilament (NF) proteins. In addition to diffuse axonal pathology, we detected accumulation of A beta and tau that colocalized with immunoreactive betaPP and NF in damaged axons throughout the white matter in all injured animals at 3-10 days post-trauma. In a subset of brain injured animals, diffuse A beta-containing plaque-like profiles were found in both the gray and white matter, and accumulations of tau and NF rich inclusions were observed in neuronal perikarya. These results show that this pig model of diffuse brain injury is characterized by accumulations of proteins that also form pathological aggregates in AD and related neurodegenerative diseases.


American Journal of Pathology | 2004

Long-term accumulation of amyloid-β, β-secretase, presenilin-1, and caspase-3 in damaged axons following brain trauma

Xiao-Han Chen; Robert Siman; Akira Iwata; David F. Meaney; John Q. Trojanowski; Douglas H. Smith

Plaques composed of amyloid β (Aβ) have been found within days following brain trauma in humans, similar to the hallmark plaque pathology of Alzheimers disease (AD). Here, we evaluated the potential source of this Aβ and long-term mechanisms that could lead to its production. Inertial brain injury was induced in pigs via head rotational acceleration of 110° over 20 ms in the coronal plane. Animals were euthanized at 3 hours, 3 days, 7 days, and 6 months post-injury. Immunohistochemistry and Western blot analyses of the brains were performed using antibodies specific for amyloid precursor protein (APP), Aβ peptides, β-site APP-cleaving enzyme (BACE), presenilin-1 (PS-1), caspase-3, and caspase-mediated cleavage of APP (CCA). Substantial co-accumulation for all of these factors was found in swollen axons at all time points up to 6 months following injury. Western blot analysis of injured brains confirmed a substantial increase in the protein levels of these factors, particularly in the white matter. These data suggest that impaired axonal transport due to trauma induces long-term pathological co-accumulation of APP with BACE, PS-1, and activated caspase. The abnormal concentration of these factors may lead to APP proteolysis and Aβ formation within the axonal membrane compartment.


Journal of Neuropathology and Experimental Neurology | 1997

Characterization of diffuse axonal pathology and selective hippocampal damage following inertial brain trauma in the pig

Douglas H. Smith; Xiao-Han Chen; Bai-Nan Xu; Tracy K. McIntosh; Thomas A. Gennarelli; David E. Meaney

Dynamic deformation applied to white matter tracts is a common feature of human brain trauma, and may result in diffuse axonal injury (DAI). To produce DAI in an experimental model, we have utilized nonimpact inertial loading to induce brain trauma in miniature swine. This species was chosen due to its large gyrencephalic brain with substantial white matter domains. Twenty anesthetized (2% isoflurane) miniature swine were subjected to pure impulsive centroidal rotation 110 degrees in the coronal plane in 4 to 6 ms; peak accelerations ranged from 0.6 to 1.7 x 10(5) rad/s2. Seven days following injury, the brains were fixed (4% paraformaldehyde). Histopathologic examination was performed on 40 microns sections stained with cresyl violet (Nissl), antibodies targeting neurofilament (axonal damage), GFAP (astrocytes), and pig IgG (protein extravasation). Widespread multifocal axonal injury was observed in combination with gliosis throughout the brain, most commonly in the root of gyri and at the interface of the gray and white matter. Very little vascular disruption was noted in regions of axonal injury. Neuronal damage was primarily found in the CA1 and CA3 subfields of the hippocampus. These results suggest that this model is clinically relevant and useful for evaluating mechanisms of inertial brain trauma.


The Journal of Neuroscience | 2004

Traumatic Axonal Injury Induces Proteolytic Cleavage of the Voltage-Gated Sodium Channels Modulated by Tetrodotoxin and Protease Inhibitors

Akira Iwata; Peter K. Stys; John A. Wolf; Xiao-Han Chen; Andrew G. Taylor; David F. Meaney; Douglas H. Smith

We demonstrated previously that dynamic stretch injury of cultured axons induces structural changes and Ca2+ influx modulated by tetrodotoxin (TTX)-sensitive voltage-gated sodium channels (NaChs). In the present study, we evaluated potential damage to the NaCh α-subunit, which can cause noninactivation of NaChs. In addition, we explored the effects of pre-injury and post-injury treatment with TTX and protease inhibition on proteolysis of the NaCh α-subunit and intra-axonal calcium levels ([Ca2+]i) over 60 min after trauma. After stretch injury, we found that [Ca2+]i continued to increase in untreated axons for at least 60 min. We also observed that the III-IV intra-axonal loop of the NaCh α-subunit was proteolyzed between 5 and 20 min after trauma. Pre-injury treatment of the axons with TTX completely abolished the posttraumatic increase in [Ca2+]i and proteolysis of the NaCh α-subunit. In addition, both pre-injury and post-injury inhibition of protease activity attenuated long-term increases in [Ca2+]i as well as mitigating degradation of the NaCh α-subunit. These results suggest a unique “feed-forward” deleterious process initiated by mechanical trauma of axons. Na+ influx through NaChs resulting from axonal deformation triggers initial increases in [Ca2+]i and subsequent proteolysis of the NaChα-subunit. In turn, degradation of the α-subunit promotes persistent elevations in [Ca2+]i, fueling additional pathologic changes. These observations may have important implications for developing therapeutic strategies for axonal trauma.


Brain Pathology | 2009

A Lack of Amyloid β Plaques Despite Persistent Accumulation of Amyloid β in Axons of Long-Term Survivors of Traumatic Brain Injury

Xiao-Han Chen; Victoria E. Johnson; Kunihiro Uryu; John Q. Trojanowski; Douglas H. Smith

Traumatic brain injury (TBI) is a risk factor for developing Alzheimers disease (AD). Additionally, TBI induces AD‐like amyloid β (Aβ) plaque pathology within days of injury potentially resulting from massive accumulation of amyloid precursor protein (APP) in damaged axons. Here, progression of Aβ accumulation was examined using brain tissue from 23 cases with post‐TBI survival of up to 3 years. Even years after injury, widespread axonal pathology was consistently observed and was accompanied by intra‐axonal co‐accumulations of APP with its cleavage enzymes, beta‐site APP cleaving enzyme and presenilin‐1 and their product, Aβ. However, in marked contrast to the plaque pathology noted in short‐term cases post TBI, virtually no Aβ plaques were found in long‐term survivors. A potential mechanism for Aβ plaque regression was suggested by the post‐injury accumulation of an Aβ degrading enzyme, neprilysin. These findings fail to support the premise that progressive plaque pathology after TBI ultimately results in AD.


American Journal of Pathology | 1998

Brain Trauma Induces Massive Hippocampal Neuron Death Linked to a Surge in β-Amyloid Levels in Mice Overexpressing Mutant Amyloid Precursor Protein

Douglas H. Smith; Michio Nakamura; Tracy K. McIntosh; Jun Wang; Amarís Rodríguez; Xiao-Han Chen; Ramesh Raghupathi; Kathryn E. Saatman; James A. Clemens; M. Luise Schmidt; Virginia M.-Y. Lee; John Q. Trojanowski

Although brain trauma is a risk factor for Alzheimers disease, no experimental model has been generated to explore this relationship. We developed a model of brain trauma in transgenic mice that overexpress mutant human amyloid precursor protein (PDAPP) leading to the appearance of Alzheimers disease-like beta-amyloid (Abeta) plaques beginning at 6 months of age. We induced cortical impact brain injury in the PDAPP animals and their wild-type littermates at 4 months of age, ie, before Abeta plaque formation, and evaluated changes in posttraumatic memory function, histopathology, and regional tissue levels of the Abeta peptides Abeta1-40 and Abeta1-42. We found that noninjured PDAPP mice had impaired memory function compared to noninjured wild-type littermates (P < 0.01) and that brain-injured PDAPP mice had more profound memory dysfunction than brain-injured wild-type littermates (P < 0.001). Although no augmentation of Abeta plaque formation was observed in brain-injured PDAPP mice, a substantial exacerbation of neuron death was found in the hippocampus (P < 0.001) in association with an acute threefold increase in Abeta1-40 and sevenfold increase in Abeta1-42 levels selectively in the hippocampus (P < 0.01). These data suggest a mechanistic link between brain trauma and Abeta levels and the death of neurons.


Publication of: Society of Automotive Engineers | 1998

FINITE ELEMENT MODELING APPROACHES FOR PREDICTING INJURY IN AN EXPERIMENTAL MODEL OF SEVERE DIFFUSE AXONAL INJURY

Reid T. Miller; Susan S. Margulies; Matt Leoni; Masahiro Nonaka; Xiao-Han Chen; Douglas H. Smith; David F. Meaney

Traumatic brain injury (TBI) finite element (FE) analyses have evolved from crude geometric representations of the skull and brain system into sophisticated models which take into account distinct anatomical features. Two distinct FE modeling approaches have evolved to account for the relative motion that occurs between the skull and cerebral cortex during TBI. The first approach assumes that the relative motion can be estimated by representing the cerebrospinal fluid inside the subarachnoid space as a low shear modulus, virtually incompressible solid. The second approach assumes that the relative motion can be approximated by defining a frictional interface between the cerebral cortex and dura mater. This study presents data from an experimental model of TBI coupled with FE analyses to evaluate the modeling approachs ability to predict specific forms of TBI. Axial plane rotational accelerations produced prolonged traumatic coma in the miniature pig, axonal injury throughout regions of the white matter, and macroscopic hemorrhagic cortical contusions. Results from 2-dimensional FE analyses of the miniature pig showed that the manner in which the modeling approach accounts for the relative motions occurring between the skull and cerebral cortex can dramatically influence the outcome of an analysis. This study clearly demonstrated that the modeling approach which represented the relative motion between the skull and cerebral cortex as a frictional interface best predicted the resulting injury pattern in a 5th axial plane animal experiment.


Neurosurgery | 2006

Thromboembolism and delayed cerebral ischemia after subarachnoid hemorrhage: an autopsy study.

Sherman C. Stein; Kevin D. Browne; Xiao-Han Chen; Douglas H. Smith; David I. Graham

OBJECTIVE:Recent findings have cast doubt on vasospasm as the sole cause of delayed cerebral ischemia after subarachnoid hemorrhage. METHODS:We reviewed the medical records of 29 patients who died after subarachnoid hemorrhage. Brain sections were taken from the insula, cingulate gyrus, and hippocampus. Adjacent sections were stained with hematoxylin-eosin and immunostained for thromboemboli. The density (burden) of the latter was calculated blindly and correlated with evidence for ischemia and with the amount of subarachnoid blood. RESULTS:There is a strong correlation between microclot burden and delayed cerebral ischemia. Patients with clinical or radiological evidence of delayed ischemia had mean microclot burdens of 10.0/cm2 (standard deviation [SD], ±6.6); those without had mean burdens of 2.8 (SD, ±2.6), a highly significant difference (P = 0.002). There is also significant association (P = 0.001) between microclot burden and histological evidence of ischemia, with the mean burdens being 10.9 in sections exhibiting severe ischemia and 4.1 in those in which ischemia was absent. Microclot burden is high in patients who died within 2 days of hemorrhage, decreasing on Days 3 and 4. In delayed ischemia, the numbers rise again late in the first week and remain high until after the second week. In contrast, the average clot burden is low in patients dying without developing delayed ischemia. The amount of blood on an individual slide influenced the microclot burden on that slide to a highly significant extent (P < 0.001). CONCLUSION:Thromboembolism after subarachnoid hemorrhage may contribute to delayed cerebral ischemia, which parallels that caused by vasospasm. The pathogenesis of thromboembolism is discussed.

Collaboration


Dive into the Xiao-Han Chen's collaboration.

Top Co-Authors

Avatar

Douglas H. Smith

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David F. Meaney

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin D. Browne

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Tracy K. McIntosh

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Masahiro Nonaka

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

David I. Graham

Southern General Hospital

View shared research outputs
Top Co-Authors

Avatar

Sherman C. Stein

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

John A. Wolf

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Leoni

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge