Network


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

Hotspot


Dive into the research topics where Edward Rockenstein is active.

Publication


Featured researches published by Edward Rockenstein.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein.

Paula Desplats; He-Jin Lee; Eunjin Bae; Christina Patrick; Edward Rockenstein; Leslie Crews; Brian Spencer; Eliezer Masliah; Seung-Jae Lee

Neuronal accumulation of α-synuclein and Lewy body formation are characteristic to many neurodegenerative diseases, including Parkinsons disease (PD). This Lewy pathology appears to spread throughout the brain as the disease progresses. Furthermore, recent studies showed the occurrence of Lewy pathology in neurons grafted into the brains of PD patients, suggesting the spread of pathology from the host tissues to the grafts. The mechanism underlying this propagation is unknown. Here, we show that α-synuclein is transmitted via endocytosis to neighboring neurons and neuronal precursor cells, forming Lewy-like inclusions. Moreover, α-synuclein was transmitted from the affected neurons to engrafted neuronal precursor cells in a transgenic model of PD-like pathology. Failure of the protein quality control systems, especially lysosomes, promoted the accumulation of transmitted α-synuclein and inclusion formation. Cells exposed to neuron-derived α-synuclein showed signs of apoptosis, such as nuclear fragmentation and caspase 3 activation, both in vitro and in vivo. These findings demonstrate the cell-to-cell transmission of α-synuclein aggregates and provide critical insights into the mechanism of pathological progression in PD and other proteinopathies.


Journal of Clinical Investigation | 2008

The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid β accumulation in mice

Fiona Pickford; Eliezer Masliah; Markus Britschgi; Kurt M. Lucin; Ramya Narasimhan; Philipp A. Jaeger; Scott A. Small; Brian Spencer; Edward Rockenstein; Beth Levine; Tony Wyss-Coray

Autophagy is the principal cellular pathway for degradation of long-lived proteins and organelles and regulates cell fate in response to stress. Recently, autophagy has been implicated in neurodegeneration, but whether it is detrimental or protective remains unclear. Here we report that beclin 1, a protein with a key role in autophagy, was decreased in affected brain regions of patients with Alzheimer disease (AD) early in the disease process. Heterozygous deletion of beclin 1 (Becn1) in mice decreased neuronal autophagy and resulted in neurodegeneration and disruption of lysosomes. In transgenic mice that express human amyloid precursor protein (APP), a model for AD, genetic reduction of Becn1 expression increased intraneuronal amyloid beta (Abeta) accumulation, extracellular Abeta deposition, and neurodegeneration and caused microglial changes and profound neuronal ultrastructural abnormalities. Administration of a lentiviral vector expressing beclin 1 reduced both intracellular and extracellular amyloid pathology in APP transgenic mice. We conclude that beclin 1 deficiency disrupts neuronal autophagy, modulates APP metabolism, and promotes neurodegeneration in mice and that increasing beclin 1 levels may have therapeutic potential in AD.


Nature Medicine | 2009

Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer's disease

Alan H. Nagahara; David A. Merrill; Giovanni Coppola; Shingo Tsukada; Brock E. Schroeder; Gideon M. Shaked; Ling Wang; Armin Blesch; Albert H. Kim; James M. Conner; Edward Rockenstein; Moses V. Chao; Edward H. Koo; Daniel H. Geschwind; Eliezer Masliah; Andrea A. Chiba; Mark H. Tuszynski

Profound neuronal dysfunction in the entorhinal cortex contributes to early loss of short-term memory in Alzheimers disease. Here we show broad neuroprotective effects of entorhinal brain-derived neurotrophic factor (BDNF) administration in several animal models of Alzheimers disease, with extension of therapeutic benefits into the degenerating hippocampus. In amyloid-transgenic mice, BDNF gene delivery, when administered after disease onset, reverses synapse loss, partially normalizes aberrant gene expression, improves cell signaling and restores learning and memory. These outcomes occur independently of effects on amyloid plaque load. In aged rats, BDNF infusion reverses cognitive decline, improves age-related perturbations in gene expression and restores cell signaling. In adult rats and primates, BDNF prevents lesion-induced death of entorhinal cortical neurons. In aged primates, BDNF reverses neuronal atrophy and ameliorates age-related cognitive impairment. Collectively, these findings indicate that BDNF exerts substantial protective effects on crucial neuronal circuitry involved in Alzheimers disease, acting through amyloid-independent mechanisms. BDNF therapeutic delivery merits exploration as a potential therapy for Alzheimers disease.


American Journal of Pathology | 2000

α-Synuclein Promotes Mitochondrial Deficit and Oxidative Stress

Leigh J. Hsu; Yutaka Sagara; Armando Arroyo; Edward Rockenstein; Abbyann Sisk; Margaret Mallory; Jeff Wong; Takato Takenouchi; Makoto Hashimoto; Eliezer Masliah

Abnormal accumulation of the presynaptic protein α-synuclein has recently been implicated in the pathogenesis of Alzheimer’s and Parkinson’s diseases. Because neurodegeneration in these conditions might be associated with mitochondrial dysfunction and oxidative stress, the effects of α-synuclein were investigated in a hypothalamic neuronal cell line (GT1-7). α-Synuclein overexpression in these cells resulted in formation of α-synuclein-immunopositive inclusion-like structures and mitochondrial alterations accompanied by increased levels of free radicals and decreased secretion of gonadotropin-releasing hormone. These alterations were ameliorated by pretreatment with anti-oxidants such as vitamin E. Taken together these results suggest that abnormal accumulation of α-synuclein could lead to mitochondrial alterations that may result in oxidative stress and, eventually, cell death.


Proceedings of the National Academy of Sciences of the United States of America | 2001

β-Amyloid peptides enhance α-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer's disease and Parkinson's disease

Eliezer Masliah; Edward Rockenstein; Isaac Veinbergs; Yutaka Sagara; Margaret Mallory; Makoto Hashimoto; Lennart Mucke

Alzheimers disease and Parkinsons disease are associated with the cerebral accumulation of β-amyloid and α-synuclein, respectively. Some patients have clinical and pathological features of both diseases, raising the possibility of overlapping pathogenetic pathways. We generated transgenic (tg) mice with neuronal expression of human β-amyloid peptides, α-synuclein, or both. The functional and morphological alterations in doubly tg mice resembled the Lewy-body variant of Alzheimers disease. These mice had severe deficits in learning and memory, developed motor deficits before α-synuclein singly tg mice, and showed prominent age-dependent degeneration of cholinergic neurons and presynaptic terminals. They also had more α-synuclein-immunoreactive neuronal inclusions than α-synuclein singly tg mice. Ultrastructurally, some of these inclusions were fibrillar in doubly tg mice, whereas all inclusions were amorphous in α-synuclein singly tg mice. β-Amyloid peptides promoted aggregation of α-synuclein in a cell-free system and intraneuronal accumulation of α-synuclein in cell culture. β-Amyloid peptides may contribute to the development of Lewy-body diseases by promoting the aggregation of α-synuclein and exacerbating α-synuclein-dependent neuronal pathologies. Therefore, treatments that block the production or accumulation of β-amyloid peptides could benefit a broader spectrum of disorders than previously anticipated.


Neuron | 2005

Effects of α-Synuclein Immunization in a Mouse Model of Parkinson’s Disease

Eliezer Masliah; Edward Rockenstein; Anthony Adame; Michael Alford; Leslie Crews; Makoto Hashimoto; Peter Seubert; Michael K. Lee; Jason Goldstein; Tamie J. Chilcote; Dora Games; Dale Schenk

Abnormal folding of alpha-synuclein (alpha-syn) is thought to lead to neurodegeneration and the characteristic symptoms of Lewy body disease (LBD). Since previous studies suggest that immunization might be a potential therapy for Alzheimers disease, we hypothesized that immunization with human (h)alpha-syn might have therapeutic effects in LBD. For this purpose, halpha-syn transgenic (tg) mice were vaccinated with halpha-syn. In mice that produced high relative affinity antibodies, there was decreased accumulation of aggregated halpha-syn in neuronal cell bodies and synapses that was associated with reduced neurodegeneration. Furthermore, antibodies produced by immunized mice recognized abnormal halpha-syn associated with the neuronal membrane and promoted the degradation of halpha-syn aggregates, probably via lysosomal pathways. Similar effects were observed with an exogenously applied FITC-tagged halpha-syn antibody. These results suggest that vaccination is effective in reducing neuronal accumulation of halpha-syn aggregates and that further development of this approach might have a potential role in the treatment of LBD.


The Journal of Neuroscience | 2009

Beclin 1 Gene Transfer Activates Autophagy and Ameliorates the Neurodegenerative Pathology in α-Synuclein Models of Parkinson's and Lewy Body Diseases

Brian Spencer; Rewati Potkar; Margarita Trejo; Edward Rockenstein; Christina Patrick; Ryan Gindi; Anthony Adame; Tony Wyss-Coray; Eliezer Masliah

Accumulation of the synaptic protein α-synuclein (α-syn) is a hallmark of Parkinsons disease (PD) and Lewy body disease (LBD), a heterogeneous group of disorders with dementia and parkinsonism, where Alzheimers disease and PD interact. Accumulation of α-syn in these patients might be associated with alterations in the autophagy pathway. Therefore, we postulate that delivery of beclin 1, a regulator of the autophagy pathway, might constitute a strategy toward developing a therapy for LBD/PD. Overexpression of α-syn from lentivirus transduction in a neuronal cell line resulted in lysosomal accumulation and alterations in autophagy. Coexpression of beclin 1 activated autophagy, reduced accumulation of α-syn, and ameliorated associated neuritic alterations. The effects of beclin 1 overexpression on LC3 and α-syn accumulation were partially blocked by 3-MA and completely blocked by bafilomycin A1. In contrast, rapamycin enhanced the effects of beclin 1. To evaluate the potential effects of activating autophagy in vivo, a lentivirus expressing beclin 1 was delivered to the brain of a α-syn transgenic mouse. Neuropathological analysis demonstrated that beclin 1 injections ameliorated the synaptic and dendritic pathology in the tg mice and reduced the accumulation of α-syn in the limbic system without any significant deleterious effects. This was accompanied by enhanced lysosomal activation and reduced alterations in the autophagy pathway. Thus, beclin 1 plays an important role in the intracellular degradation of α-syn either directly or indirectly through the autophagy pathway and may present a novel therapeutic target for LBD/PD.


Nature Neuroscience | 2005

Targeting BACE1 with siRNAs ameliorates Alzheimer disease neuropathology in a transgenic model

Oded Singer; Robert A. Marr; Edward Rockenstein; Leslie Crews; Nicole G. Coufal; Fred H. Gage; Inder M. Verma; Eliezer Masliah

In Alzheimer disease, increased β-secretase (BACE1) activity has been associated with neurodegeneration and accumulation of amyloid precursor protein (APP) products. Thus, inactivation of BACE1 could be important in the treatment of Alzheimer disease. In this study, we found that lowering BACE1 levels using lentiviral vectors expressing siRNAs targeting BACE1 reduced amyloid production and the neurodegenerative and behavioral deficits in APP transgenic mice, a model of Alzheimer disease. Our results suggest that lentiviral vector delivery of BACE1 siRNA can specifically reduce the cleavage of APP and neurodegeneration in vivo and indicate that this approach could have potential therapeutic value for treatment of Alzheimer disease.


Neuron | 2001

β-Synuclein Inhibits α-Synuclein Aggregation

Makoto Hashimoto; Edward Rockenstein; Michael Mante; Margaret Mallory; Eliezer Masliah

Abstract We characterized β-synuclein, the non-amyloidogenic homolog of α-synuclein, as an inhibitor of aggregation of α-synuclein, a molecule implicated in Parkinsons disease. For this, doubly transgenic mice expressing human (h) α- and β-synuclein were generated. In doubly transgenic mice, β-synuclein ameliorated motor deficits, neurodegenerative alterations, and neuronal α-synuclein accumulation seen in hα-synuclein transgenic mice. Similarly, cell lines transfected with β-synuclein were resistant to α-synuclein accumulation. hα-synuclein was coimmunoprecipitated with hβ-synuclein in the brains of doubly transgenic mice and in the double-transfected cell lines. Our results raise the possibility that β-synuclein might be a natural negative regulator of α-synuclein aggregation and that a similar class of endogenous factors might regulate the aggregation state of other molecules involved in neurodegeneration. Such an anti-amyloidogenic property of β-synuclein might also provide a novel strategy for the treatment of neurodegenerative disorders.


Journal of Neuroscience Research | 2002

Differential neuropathological alterations in transgenic mice expressing α-synuclein from the platelet-derived growth factor and Thy-1 promoters

Edward Rockenstein; Margaret Mallory; Makoto Hashimoto; David D. Song; Clifford W. Shults; Ingrid Lang; Eliezer Masliah

Accumulation of α‐synuclein has been associated with neurodegenerative disorders, such as Lewy body disease and multiple system atrophy. We previously showed that expression of wild‐type human α‐synuclein in transgenic mice results in motor and dopaminergic deficits associated with inclusion formation. To determine whether different levels of human α‐synuclein expression from distinct promoters might result in neuropathology mimicking other synucleopathies, we compared patterns of human α‐synuclein accumulation in the brains of transgenic mice expressing this molecule from the murine Thy‐1 and platelet‐derived growth factor (PDGF) promoters. In murine Thy‐1‐human α‐synuclein transgenic mice, this protein accumulated in synapses and neurons throughout the brain, including the thalamus, basal ganglia, substantia nigra, and brainstem. Expression of human α‐synuclein from the PDGF promoter resulted in accumulation in synapses of the neocortex, limbic system, and olfactory regions as well as formation of inclusion bodies in neurons in deeper layers of the neocortex. Furthermore, one of the intermediate expresser lines (line M) displayed human α‐synuclein expression in glial cells mimicking some features of multiple system atrophy. These results show a more widespread accumulation of human α‐synuclein in transgenic mouse brains. Taken together, these studies support the contention that human α‐synuclein expression in transgenic mice might mimic some neuropathological alterations observed in Lewy body disease and other synucleopathies, such as multiple system atrophy.

Collaboration


Dive into the Edward Rockenstein's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony Adame

University of California

View shared research outputs
Top Co-Authors

Avatar

Michael Mante

University of California

View shared research outputs
Top Co-Authors

Avatar

Brian Spencer

University of California

View shared research outputs
Top Co-Authors

Avatar

Leslie Crews

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kiren Ubhi

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paula Desplats

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge