Network


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

Hotspot


Dive into the research topics where Abby Oehler is active.

Publication


Featured researches published by Abby Oehler.


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

Purified and synthetic Alzheimer’s amyloid beta (Aβ) prions

Jan Stöhr; Joel C. Watts; Zachary Mensinger; Abby Oehler; Sunny K. Grillo; Stephen J. DeArmond; Stanley B. Prusiner; Kurt Giles

The aggregation and deposition of amyloid-β (Aβ) peptides are believed to be central events in the pathogenesis of Alzheimer’s disease (AD). Inoculation of brain homogenates containing Aβ aggregates into susceptible transgenic mice accelerated Aβ deposition, suggesting that Aβ aggregates are capable of self-propagation and hence might be prions. Recently, we demonstrated that Aβ deposition can be monitored in live mice using bioluminescence imaging (BLI). Here, we use BLI to probe the ability of Aβ aggregates to self-propagate following inoculation into bigenic mice. We report compelling evidence that Aβ aggregates are prions by demonstrating widespread cerebral β-amyloidosis induced by inoculation of either purified Aβ aggregates derived from brain or aggregates composed of synthetic Aβ. Although synthetic Aβ aggregates were sufficient to induce Aβ deposition in vivo, they exhibited lower specific biological activity compared with brain-derived Aβ aggregates. Our results create an experimental paradigm that should lead to identification of self-propagating Aβ conformations, which could represent novel targets for interrupting the spread of Aβ deposition in AD patients.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Evidence for α-synuclein prions causing multiple system atrophy in humans with parkinsonism

Stanley B. Prusiner; Amanda L. Woerman; Daniel A. Mordes; Joel C. Watts; Ryan Rampersaud; David B. Berry; Smita Patel; Abby Oehler; Jennifer K. Lowe; Stephanie N. Kravitz; Daniel H. Geschwind; David V. Glidden; Glenda M. Halliday; Lefkos Middleton; Steve M. Gentleman; Lea T. Grinberg; Kurt Giles

Significance Prions are proteins that assume alternate shapes that become self-propagating, and while some prions perform normal physiological functions, others cause disease. Prions were discovered while studying the cause of rare neurodegenerative diseases of animals and humans called scrapie and Creutzfeldt–Jakob disease, respectively. We report here the discovery of α-synuclein prions that cause a more common neurodegenerative disease in humans called multiple system atrophy (MSA). In contrast to MSA, brain extracts from Parkinson’s disease (PD) patients were not transmissible to genetically engineered cells or mice, although much evidence argues that PD is also caused by α-synuclein, suggesting that this strain (or variant) is different from those that cause MSA. Prions are proteins that adopt alternative conformations that become self-propagating; the PrPSc prion causes the rare human disorder Creutzfeldt–Jakob disease (CJD). We report here that multiple system atrophy (MSA) is caused by a different human prion composed of the α-synuclein protein. MSA is a slowly evolving disorder characterized by progressive loss of autonomic nervous system function and often signs of parkinsonism; the neuropathological hallmark of MSA is glial cytoplasmic inclusions consisting of filaments of α-synuclein. To determine whether human α-synuclein forms prions, we examined 14 human brain homogenates for transmission to cultured human embryonic kidney (HEK) cells expressing full-length, mutant human α-synuclein fused to yellow fluorescent protein (α-syn140*A53T–YFP) and TgM83+/− mice expressing α-synuclein (A53T). The TgM83+/− mice that were hemizygous for the mutant transgene did not develop spontaneous illness; in contrast, the TgM83+/+ mice that were homozygous developed neurological dysfunction. Brain extracts from 14 MSA cases all transmitted neurodegeneration to TgM83+/− mice after incubation periods of ∼120 d, which was accompanied by deposition of α-synuclein within neuronal cell bodies and axons. All of the MSA extracts also induced aggregation of α-syn*A53T–YFP in cultured cells, whereas none of six Parkinson’s disease (PD) extracts or a control sample did so. Our findings argue that MSA is caused by a unique strain of α-synuclein prions, which is different from the putative prions causing PD and from those causing spontaneous neurodegeneration in TgM83+/+ mice. Remarkably, α-synuclein is the first new human prion to be identified, to our knowledge, since the discovery a half century ago that CJD was transmissible.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Transmission of multiple system atrophy prions to transgenic mice

Joel C. Watts; Kurt Giles; Abby Oehler; Lefkos Middleton; David T. Dexter; Steve M. Gentleman; Stephen J. DeArmond; Stanley B. Prusiner

Significance Multiple system atrophy (MSA) is a neurodegenerative disorder characterized by the accumulation of misfolded α-synuclein protein in glial cells within the brain. Transgenic mice expressing mutant α-synuclein that were inoculated with brain homogenate from MSA patients developed clinical, biochemical, and pathological signs of a neurodegenerative disease, indicating that MSA is transmissible under certain conditions. This transmissibility is reminiscent of the human prion disorders, such as Creutzfeldt–Jakob disease, and suggests that MSA is caused by the accumulation of toxic α-synuclein prions in the brain. Prions are proteins that adopt alternative conformations, which become self-propagating. Increasing evidence argues that prions feature in the synucleinopathies that include Parkinson’s disease, Lewy body dementia, and multiple system atrophy (MSA). Although TgM83+/+ mice homozygous for a mutant A53T α-synuclein transgene begin developing CNS dysfunction spontaneously at ∼10 mo of age, uninoculated TgM83+/− mice (hemizygous for the transgene) remain healthy. To determine whether MSA brains contain α-synuclein prions, we inoculated the TgM83+/− mice with brain homogenates from two pathologically confirmed MSA cases. Inoculated TgM83+/− mice developed progressive signs of neurologic disease with an incubation period of ∼100 d, whereas the same mice inoculated with brain homogenates from spontaneously ill TgM83+/+ mice developed neurologic dysfunction in ∼210 d. Brains of MSA-inoculated mice exhibited prominent astrocytic gliosis and microglial activation as well as widespread deposits of phosphorylated α-synuclein that were proteinase K sensitive, detergent insoluble, and formic acid extractable. Our results provide compelling evidence that α-synuclein aggregates formed in the brains of MSA patients are transmissible and, as such, are prions. The MSA prion represents a unique human pathogen that is lethal upon transmission to Tg mice and as such, is reminiscent of the prion causing kuru, which was transmitted to chimpanzees nearly 5 decades ago.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Serial propagation of distinct strains of Aβ prions from Alzheimer's disease patients.

Joel C. Watts; Carlo Condello; Jan Stöhr; Abby Oehler; Joanne Lee; Stephen J. DeArmond; Lars Lannfelt; Martin Ingelsson; Kurt Giles; Stanley B. Prusiner

Significance The amyloid-β (Aβ) peptide, which plays a central role in Alzheimer’s disease (AD) pathogenesis, exhibits many properties that are reminiscent of prions (self-propagating proteins that cause neurodegenerative disorders, such as mad cow disease). In the human prion diseases, distinct strains of prions can be distinguished, and therefore, we asked whether different strains of Aβ aggregates might exist in the brains of AD patients. Inoculation of transgenic mice with brain samples from patients with two different heritable forms of AD produced two distinct patterns of cerebral Aβ deposition, and these differences were maintained on serial passage. We conclude that distinct strains of Aβ can be discerned in AD patients, which may help to explain the clinical heterogeneity observed in the disease. An increasing number of studies argues that self-propagating protein conformations (i.e., prions) feature in the pathogenesis of several common neurodegenerative diseases. Mounting evidence contends that aggregates of the amyloid-β (Aβ) peptide become self-propagating in Alzheimer’s disease (AD) patients. An important characteristic of prions is their ability to replicate distinct strains, the biological information for which is enciphered within different conformations of protein aggregates. To investigate whether distinct strains of Aβ prions can be discerned in AD patients, we performed transmission studies in susceptible transgenic mice using brain homogenates from sporadic or heritable (Arctic and Swedish) AD cases. Mice inoculated with the Arctic AD sample exhibited a pathology that could be distinguished from mice inoculated with the Swedish or sporadic AD samples, which was judged by differential accumulation of Aβ isoforms and the morphology of cerebrovascular Aβ deposition. Unlike Swedish AD- or sporadic AD-inoculated animals, Arctic AD-inoculated mice, like Arctic AD patients, displayed a prominent Aβ38-containing cerebral amyloid angiopathy. The divergent transmission behavior of the Arctic AD sample compared with the Swedish and sporadic AD samples was maintained during second passage in mice, showing that Aβ strains are serially transmissible. We conclude that at least two distinct strains of Aβ prions can be discerned in the brains of AD patients and that strain fidelity was preserved on serial passage in mice. Our results provide a potential explanation for the clinical and pathological heterogeneity observed in AD patients.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Distinct synthetic Aβ prion strains producing different amyloid deposits in bigenic mice

Jan Stöhr; Carlo Condello; Joel C. Watts; Lillian Bloch; Abby Oehler; Mimi Nick; Stephen J. DeArmond; Kurt Giles; William F. DeGrado; Stanley B. Prusiner

Significance Alzheimer’s disease is the most common neurodegenerative disorder; it is a progressive dementia for which there is currently no effective therapeutic intervention. The brains of patients with Alzheimer’s disease exhibit numerous amyloid β (Aβ) amyloid plaques and tau-laden neurofibrillary tangles. Our studies show that synthetic Aβ peptides can form prions that infect mice and induce Aβ amyloid plaque pathology. Two different Aβ prion strains were produced from Aβ peptides. When injected into transgenic mice, one Aβ strain produced large plaques and the other strain induced small but more numerous plaques. Our findings may help to delineate the molecular pathogenesis of Alzheimer’s disease and the development of anti-Aβ prion therapeutics. An increasing number of studies continue to show that the amyloid β (Aβ) peptide adopts an alternative conformation and acquires transmissibility; hence, it becomes a prion. Here, we report on the attributes of two strains of Aβ prions formed from synthetic Aβ peptides composed of either 40 or 42 residues. Modifying the conditions for Aβ polymerization increased both the protease resistance and prion infectivity compared with an earlier study. Approximately 150 d after intracerebral inoculation, both synthetic Aβ40 and Aβ42 prions produced a sustained rise in the bioluminescence imaging signal in the brains of bigenic Tg(APP23:Gfap-luc) mice, indicative of astrocytic gliosis. Pathological investigations showed that synthetic Aβ40 prions produced amyloid plaques containing both Aβ40 and Aβ42 in the brains of inoculated bigenic mice, whereas synthetic Aβ42 prions stimulated the formation of smaller, more numerous plaques composed predominantly of Aβ42. Synthetic Aβ40 preparations consisted of long straight fibrils; in contrast, the Aβ42 fibrils were much shorter. Addition of 3.47 mM (0.1%) SDS to the polymerization reaction produced Aβ42 fibrils that were indistinguishable from Aβ40 fibrils produced in the absence or presence of SDS. Moreover, the Aβ amyloid plaques in the brains of bigenic mice inoculated with Aβ42 prions prepared in the presence of SDS were similar to those found in mice that received Aβ40 prions. From these results, we conclude that the composition of Aβ plaques depends on the conformation of the inoculated Aβ polymers, and thus, these inocula represent distinct synthetic Aβ prion strains.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Propagation of prions causing synucleinopathies in cultured cells.

Amanda L. Woerman; Jan Stöhr; Atsushi Aoyagi; Ryan Rampersaud; Zuzana Krejciova; Joel C. Watts; Takao Ohyama; Smita Patel; Kartika Widjaja; Abby Oehler; David W. Sanders; Marc I. Diamond; William W. Seeley; Lefkos Middleton; Steve M. Gentleman; Daniel A. Mordes; Thomas C. Südhof; Kurt Giles; Stanley B. Prusiner

Significance Progressive supranuclear palsy (PSP) and multiple system atrophy (MSA) are neurodegenerative diseases caused by tau and α-synuclein prions, respectively. Prions, purified from human brains of deceased patients with PSP and MSA using phosphotungstic acid, were applied to cultured cell models that selectively form aggregates in the presence of tau or α-synuclein prions, respectively. Whereas brain homogenates prepared from two PSP and six MSA patients infected cultured cells, the same approach was unsuccessful with brain samples from three Parkinson’s disease patients. Our findings provide compelling evidence that PSP and MSA are prion diseases, and that MSA is caused by several distinct prion strains. Increasingly, evidence argues that many neurodegenerative diseases, including progressive supranuclear palsy (PSP), are caused by prions, which are alternatively folded proteins undergoing self-propagation. In earlier studies, PSP prions were detected by infecting human embryonic kidney (HEK) cells expressing a tau fragment [TauRD(LM)] fused to yellow fluorescent protein (YFP). Here, we report on an improved bioassay using selective precipitation of tau prions from human PSP brain homogenates before infection of the HEK cells. Tau prions were measured by counting the number of cells with TauRD(LM)–YFP aggregates using confocal fluorescence microscopy. In parallel studies, we fused α-synuclein to YFP to bioassay α-synuclein prions in the brains of patients who died of multiple system atrophy (MSA). Previously, MSA prion detection required ∼120 d for transmission into transgenic mice, whereas our cultured cell assay needed only 4 d. Variation in MSA prion levels in four different brain regions from three patients provided evidence for three different MSA prion strains. Attempts to demonstrate α-synuclein prions in brain homogenates from Parkinson’s disease patients were unsuccessful, identifying an important biological difference between the two synucleinopathies. Partial purification of tau and α-synuclein prions facilitated measuring the levels of these protein pathogens in human brains. Our studies should facilitate investigations of the pathogenesis of both tau and α-synuclein prion disorders as well as help decipher the basic biology of those prions that attack the CNS.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Drug resistance confounding prion therapeutics

David B. Berry; Duo Lu; Michal Geva; Joel C. Watts; Sumita Bhardwaj; Abby Oehler; Adam R. Renslo; Stephen J. DeArmond; Stanley B. Prusiner; Kurt Giles

Significance As people live longer, the prevalence and economic impact of neurodegenerative diseases rise. No cures or effective treatments exist for any of these fatal disorders, so identifying potential therapeutics that extend survival in animal models is vital. Many neurodegenerative illnesses have been shown to be caused by the accumulation of self-propagating misfolded proteins—the hallmark of prion diseases. We report the efficacy of 2-aminothiazoles, which were identified in cell-based screens as antiprion compounds, in extending the lives of prion-infected animals. Efficacy was limited by the development of drug-resistant prions, which is likely to have important implications for creating therapeutics in many different neurodegenerative diseases. There is not a single pharmaceutical that halts or even slows any neurodegenerative disease. Mounting evidence shows that prions cause many neurodegenerative diseases, and arguably, scrapie and Creutzfeldt–Jakob disease prions represent the best therapeutic targets. We report here that the previously identified 2-aminothiazoles IND24 and IND81 doubled the survival times of scrapie-infected, wild-type mice. However, mice infected with Rocky Mountain Laboratory (RML) prions, a scrapie-derived strain, and treated with IND24 eventually exhibited neurological dysfunction and died. We serially passaged their brain homogenates in mice and cultured cells. We found that the prion strain isolated from IND24-treated mice, designated RML[IND24], emerged during a single passage in treated mice. Although RML prions infect both the N2a and CAD5 cell lines, RML[IND24] prions could only infect CAD5 cells. When passaged in CAD5 cells, the prions remained resistant to high concentrations of IND24. However, one passage of RML[IND24] prions in untreated mice restored susceptibility to IND24 in CAD5 cells. Although IND24 treatment extended the lives of mice propagating different prion strains, including RML, another scrapie-derived prion strain ME7, and chronic wasting disease, it was ineffective in slowing propagation of Creutzfeldt–Jakob disease prions in transgenic mice. Our studies demonstrate that prion strains can acquire resistance upon exposure to IND24 that is lost upon passage in mice in the absence of IND24. These data suggest that monotherapy can select for resistance, thus intermittent therapy with mixtures of antiprion compounds may be required to slow or stop neurodegeneration.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Spontaneous generation of anchorless prions in transgenic mice

Jan Stöhr; Joel C. Watts; Giuseppe Legname; Abby Oehler; Azucena Lemus; Hoang-Oanh B. Nguyen; Joshua Sussman; Holger Wille; Stephen J. DeArmond; Stanley B. Prusiner; Kurt Giles

Some prion protein mutations create anchorless molecules that cause Gerstmann–Sträussler–Scheinker (GSS) disease. To model GSS, we generated transgenic mice expressing cellular prion protein (PrPC) lacking the glycosylphosphatidyl inositol (GPI) anchor, denoted PrP(ΔGPI). Mice overexpressing PrP(ΔGPI) developed a late-onset, spontaneous neurologic dysfunction characterized by widespread amyloid deposition in the brain and the presence of a short protease-resistant PrP fragment similar to those found in GSS patients. In Tg(PrP,ΔGPI) mice, disease onset could be accelerated either by inoculation with brain homogenate prepared from spontaneously ill animals or by coexpression of membrane-anchored, full-length PrPC. In contrast, coexpression of N-terminally truncated PrP(Δ23–88) did not affect disease progression. Remarkably, disease from ill Tg(PrP,ΔGPI) mice transmitted to mice expressing wild-type PrPC, indicating the spontaneous generation of prions.


PLOS Pathogens | 2014

Evidence That Bank Vole PrP Is a Universal Acceptor for Prions

Joel C. Watts; Kurt Giles; Smita Patel; Abby Oehler; Stephen J. DeArmond; Stanley B. Prusiner

Bank voles are uniquely susceptible to a wide range of prion strains isolated from many different species. To determine if this enhanced susceptibility to interspecies prion transmission is encoded within the sequence of the bank vole prion protein (BVPrP), we inoculated Tg(M109) and Tg(I109) mice, which express BVPrP containing either methionine or isoleucine at polymorphic codon 109, with 16 prion isolates from 8 different species: humans, cattle, elk, sheep, guinea pigs, hamsters, mice, and meadow voles. Efficient disease transmission was observed in both Tg(M109) and Tg(I109) mice. For instance, inoculation of the most common human prion strain, sporadic Creutzfeldt-Jakob disease (sCJD) subtype MM1, into Tg(M109) mice gave incubation periods of ∼200 days that were shortened slightly on second passage. Chronic wasting disease prions exhibited an incubation time of ∼250 days, which shortened to ∼150 days upon second passage in Tg(M109) mice. Unexpectedly, bovine spongiform encephalopathy and variant CJD prions caused rapid neurological dysfunction in Tg(M109) mice upon second passage, with incubation periods of 64 and 40 days, respectively. Despite the rapid incubation periods, other strain-specified properties of many prion isolates—including the size of proteinase K–resistant PrPSc, the pattern of cerebral PrPSc deposition, and the conformational stability—were remarkably conserved upon serial passage in Tg(M109) mice. Our results demonstrate that expression of BVPrP is sufficient to engender enhanced susceptibility to a diverse range of prion isolates, suggesting that BVPrP may be a universal acceptor for prions.


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

Spontaneous generation of rapidly transmissible prions in transgenic mice expressing wild-type bank vole prion protein

Joel C. Watts; Kurt Giles; Jan Stöhr; Abby Oehler; Sumita Bhardwaj; Sunny K. Grillo; Smita S. Patel; Stephen J. DeArmond; Stanley B. Prusiner

Currently, there are no animal models of the most common human prion disorder, sporadic Creutzfeldt-Jakob disease (CJD), in which prions are formed spontaneously from wild-type (WT) prion protein (PrP). Interestingly, bank voles (BV) exhibit an unprecedented promiscuity for diverse prion isolates, arguing that bank vole PrP (BVPrP) may be inherently prone to adopting misfolded conformations. Therefore, we constructed transgenic (Tg) mice expressing WT BVPrP. Tg(BVPrP) mice developed spontaneous CNS dysfunction between 108 and 340 d of age and recapitulated the hallmarks of prion disease, including spongiform degeneration, pronounced astrogliosis, and deposition of alternatively folded PrP in the brain. Brain homogenates of ill Tg(BVPrP) mice transmitted disease to Tg(BVPrP) mice in ∼35 d, to Tg mice overexpressing mouse PrP in under 100 d, and to WT mice in ∼185 d. Our studies demonstrate experimentally that WT PrP can spontaneously form infectious prions in vivo. Thus, Tg(BVPrP) mice may be useful for studying the spontaneous formation of prions, and thus may provide insight into the etiology of sporadic CJD.

Collaboration


Dive into the Abby Oehler's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kurt Giles

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Smita Patel

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Stöhr

University of California

View shared research outputs
Top Co-Authors

Avatar

Carlo Condello

University of California

View shared research outputs
Top Co-Authors

Avatar

David B. Berry

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge