Network


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

Hotspot


Dive into the research topics where Dagmar E. Ehrnhoefer is active.

Publication


Featured researches published by Dagmar E. Ehrnhoefer.


Nature Structural & Molecular Biology | 2008

EGCG redirects amyloidogenic polypeptides into unstructured, off-pathway oligomers.

Dagmar E. Ehrnhoefer; Jan Bieschke; Annett Boeddrich; Martin Herbst; Laura Masino; Rudi Lurz; Sabine Engemann; Annalisa Pastore; Erich E. Wanker

The accumulation of β-sheet–rich amyloid fibrils or aggregates is a complex, multistep process that is associated with cellular toxicity in a number of human protein misfolding disorders, including Parkinsons and Alzheimers diseases. It involves the formation of various transient and intransient, on- and off-pathway aggregate species, whose structure, size and cellular toxicity are largely unclear. Here we demonstrate redirection of amyloid fibril formation through the action of a small molecule, resulting in off-pathway, highly stable oligomers. The polyphenol (−)-epigallocatechin gallate efficiently inhibits the fibrillogenesis of both α-synuclein and amyloid-β by directly binding to the natively unfolded polypeptides and preventing their conversion into toxic, on-pathway aggregation intermediates. Instead of β-sheet–rich amyloid, the formation of unstructured, nontoxic α-synuclein and amyloid-β oligomers of a new type is promoted, suggesting a generic effect on aggregation pathways in neurodegenerative diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2010

EGCG remodels mature α-synuclein and amyloid-β fibrils and reduces cellular toxicity

Jan Bieschke; Jenny Russ; Ralf P. Friedrich; Dagmar E. Ehrnhoefer; Heike Wobst; Katja Neugebauer; Erich E. Wanker

Protein misfolding and formation of β-sheet-rich amyloid fibrils or aggregates is related to cellular toxicity and decay in various human disorders including Alzheimer’s and Parkinson’s disease. Recently, we demonstrated that the polyphenol (-)-epi-gallocatechine gallate (EGCG) inhibits α-synuclein and amyloid-β fibrillogenesis. It associates with natively unfolded polypeptides and promotes the self-assembly of unstructured oligomers of a new type. Whether EGCG disassembles preformed amyloid fibrils, however, remained unclear. Here, we show that EGCG has the ability to convert large, mature α-synuclein and amyloid-β fibrils into smaller, amorphous protein aggregates that are nontoxic to mammalian cells. Mechanistic studies revealed that the compound directly binds to β-sheet-rich aggregates and mediates the conformational change without their disassembly into monomers or small diffusible oligomers. These findings suggest that EGCG is a potent remodeling agent of mature amyloid fibrils.


Nature Medicine | 2009

Balance between synaptic versus extrasynaptic NMDA receptor activity influences inclusions and neurotoxicity of mutant huntingtin

Shu-ichi Okamoto; Mahmoud A. Pouladi; Maria Talantova; Dongdong Yao; Peng Xia; Dagmar E. Ehrnhoefer; Rameez Zaidi; Arjay Clemente; Marcus Kaul; Rona K. Graham; Dongxian Zhang; H-S Vincent Chen; Gary Tong; Michael R. Hayden; Stuart A. Lipton

Huntingtons disease is caused by an expanded CAG repeat in the gene encoding huntingtin (HTT), resulting in loss of striatal and cortical neurons. Given that the gene product is widely expressed, it remains unclear why neurons are selectively targeted. Here we show the relationship between synaptic and extrasynaptic activity, inclusion formation of mutant huntingtin protein (mtHtt) and neuronal survival. Synaptic N-methyl-D-aspartate–type glutamate receptor (NMDAR) activity induces mtHtt inclusions via a T complex-1 (TCP-1) ring complex (TRiC)-dependent mechanism, rendering neurons more resistant to mtHtt-mediated cell death. In contrast, stimulation of extrasynaptic NMDARs increases the vulnerability of mtHtt-containing neurons to cell death by impairing the neuroprotective cyclic AMP response element–binding protein (CREB)–peroxisome proliferator–activated receptor-γ coactivator-1α (PGC-1α) cascade and increasing the level of the small guanine nucleotide–binding protein Rhes, which is known to sumoylate and disaggregate mtHtt. Treatment of transgenic mice expressing a yeast artificial chromosome containing 128 CAG repeats (YAC128) with low-dose memantine blocks extrasynaptic (but not synaptic) NMDARs and ameliorates neuropathological and behavioral manifestations. By contrast, high-dose memantine, which blocks both extrasynaptic and synaptic NMDAR activity, decreases neuronal inclusions and worsens these outcomes. Our findings offer a rational therapeutic approach for protecting susceptible neurons in Huntingtons disease.


The Neuroscientist | 2011

Small Changes, Big Impact: Posttranslational Modifications and Function of Huntingtin in Huntington Disease

Dagmar E. Ehrnhoefer; Liza M. Sutton; Michael R. Hayden

Huntington disease (HD) is a neurodegenerative disorder caused by an elongated polyglutamine tract in huntingtin (htt). htt normally undergoes different posttranslational modifications (PTMs), including phosphorylation, SUMOylation, ubiquitination, acetylation, proteolytic cleavage, and palmitoylation. In the presence of the HD mutation, some PTMs are significantly altered and can result in changes in the clinical phenotype. A rate-limiting PTM is defined as one that can result in significant effects on the phenotype in animal models. For example, the prevention of proteolysis at D586 as well as constitutive phosphorylation at S13 and S16 can obviate the expression of phenotypic features of HD. The enzymes involved in these modifications such as caspase-6, the IκB kinase (IKK) complex, and still to be characterized phosphatases therefore represent promising therapeutic targets for HD. Identifying and testing specific modulators of PTMs now constitute the next big challenges in order to further validate these targets and proceed towards the goal of a mechanism-based treatment for HD.


Trends in Neurosciences | 2015

Autophagy in Huntington disease and huntingtin in autophagy

Dale D. O. Martin; Safia Ladha; Dagmar E. Ehrnhoefer; Michael R. Hayden

Autophagy is an important biological process that is essential for the removal of damaged organelles and toxic or aggregated proteins by delivering them to the lysosome for degradation. Consequently, autophagy has become a primary target for the treatment of neurodegenerative diseases that involve aggregating proteins. In Huntington disease (HD), an expansion of the polyglutamine (polyQ) tract in the N-terminus of the huntingtin (HTT) protein leads to protein aggregation. However, HD is unique among the neurodegenerative proteinopathies in that autophagy is not only dysfunctional but wild type (wt) HTT also appears to play several roles in regulating the dynamics of autophagy. Herein, we attempt to integrate the recently described novel roles of wtHTT and altered autophagy in HD.


Trends in Neurosciences | 2011

Caspase-6 and neurodegeneration.

Rona K. Graham; Dagmar E. Ehrnhoefer; Michael R. Hayden

Caspases are cysteine-aspartic proteases that post-translationally modify their substrates through cleavage at specific sites, which causes either substrate inactivation or a gain of function through the generation of active fragments. Currently, each caspase is categorized as either an initiator of apoptosis or an end-stage executioner. Caspase-6 was originally identified as an executioner caspase owing to its role in cleavage of nuclear lamins. However, it has since been shown that caspase-6 cleaves caspases-2, 3 and 8. Furthermore, active caspase-6 is present in post mortem brains of Huntington and Alzheimer disease subjects that do not yet display apoptotic morphology, which suggests a function distinct from its well-validated executioner role. In this review, we discuss evidence to date regarding the role of caspase-6 in neurodegeneration. The findings suggest that selective inhibitors of caspase-6 may have therapeutic potential for various neurodegenerative disorders.


Disease Models & Mechanisms | 2009

Mouse models of Huntington disease: variations on a theme.

Dagmar E. Ehrnhoefer; Stefanie L. Butland; Mahmoud A. Pouladi; Michael R. Hayden

An accepted prerequisite for clinical trials of a compound in humans is the successful alleviation of the disease in animal models. For some diseases, however, successful translation of drug effects from mouse models to the bedside has been limited. One question is whether the current models accurately reproduce the human disease. Here, we examine the mouse models that are available for therapeutic testing in Huntington disease (HD), a late-onset neurodegenerative disorder for which there is no effective treatment. The current mouse models show different degrees of similarity to the human condition. Significant phenotypic differences are seen in mouse models that express either truncated or full-length human, or full-length mouse, mutant huntingtin (mHTT). These differences in phenotypic expression may be attributable to the influences of protein context, mouse strain and a difference in regulatory sequences between the mouse Htt and human HTT genes.


Human Molecular Genetics | 2010

Full-length huntingtin levels modulate body weight by influencing insulin-like growth factor 1 expression

Mahmoud A. Pouladi; Yuanyun Xie; Niels H. Skotte; Dagmar E. Ehrnhoefer; Rona K. Graham; Jeong Eun Kim; Nagat Bissada; X. William Yang; Paolo Paganetti; Robert M. Friedlander; Blair R. Leavitt; Michael R. Hayden

Levels of full-length huntingtin (FL htt) influence organ and body weight, independent of polyglutamine length. The growth hormone-insulin like growth factor-1 (GH-IGF-1) axis is well established as a regulator of organ growth and body weight. In this study, we investigate the involvement of the IGF-1 pathway in mediating the effect of htt on body weight. IGF-1 expression was examined in transgenic mouse lines expressing different levels of FL wild-type (WT) htt (YAC18 mice), FL mutant htt (YAC128 and BACHD mice) and truncated mutant htt (shortstop mice). We demonstrate that htt influences body weight by modulating the IGF-1 pathway. Plasma IGF-1 levels correlate with body weight and htt levels in the transgenic YAC mice expressing human htt. The effect of htt on IGF-1 expression is independent of CAG size. No effect on body weight is observed in transgenic YAC mice expressing a truncated N-terminal htt fragment (shortstop), indicating that FL htt is required for the modulation of IGF-1 expression. Treatment with 17beta-estradiol (17beta-ED) lowers the levels of circulating IGF-1 in mammals. Treatment of YAC128 with 17beta-ED, but not placebo, reduces plasma IGF-1 levels and decreases the body weight of YAC128 animals to WT levels. Furthermore, given the ubiquitous expression of IGF-1 within the central nervous system, we also examined the impact of FL htt levels on IGF-1 expression in different regions of the brain, including the striatum, cerebellum of YAC18, YAC128 and littermate WT mice. We demonstrate that the levels of FL htt influence IGF-1 expression in striatal tissues. Our data identify a novel function for FL htt in influencing IGF-1 expression.


Human Molecular Genetics | 2012

Rescue from excitotoxicity and axonal degeneration accompanied by age-dependent behavioral and neuroanatomical alterations in caspase-6-deficient mice

Valeria Uribe; Bibiana K.Y. Wong; Rona K. Graham; Corey L. Cusack; Niels H. Skotte; Mahmoud A. Pouladi; Yuanyun Xie; Konstantin Feinberg; Yimiao Ou; Yingbin Ouyang; Yu Deng; Sonia Franciosi; Nagat Bissada; Amanda Spreeuw; Weining Zhang; Dagmar E. Ehrnhoefer; Kuljeet Vaid; Freda D. Miller; Mohanish Deshmukh; David Howland; Michael R. Hayden

Apoptosis, or programmed cell death, is a cellular pathway involved in normal cell turnover, developmental tissue remodeling, embryonic development, cellular homeostasis maintenance and chemical-induced cell death. Caspases are a family of intracellular proteases that play a key role in apoptosis. Aberrant activation of caspases has been implicated in human diseases. In particular, numerous findings implicate Caspase-6 (Casp6) in neurodegenerative diseases, including Alzheimer disease (AD) and Huntington disease (HD), highlighting the need for a deeper understanding of Casp6 biology and its role in brain development. The use of targeted caspase-deficient mice has been instrumental for studying the involvement of caspases in apoptosis. The goal of this study was to perform an in-depth neuroanatomical and behavioral characterization of constitutive Casp6-deficient (Casp6-/-) mice in order to understand the physiological function of Casp6 in brain development, structure and function. We demonstrate that Casp6-/- neurons are protected against excitotoxicity, nerve growth factor deprivation and myelin-induced axonal degeneration. Furthermore, Casp6-deficient mice show an age-dependent increase in cortical and striatal volume. In addition, these mice show a hypoactive phenotype and display learning deficits. The age-dependent behavioral and region-specific neuroanatomical changes observed in the Casp6-/- mice suggest that Casp6 deficiency has a more pronounced effect in brain regions that are involved in neurodegenerative diseases, such as the striatum in HD and the cortex in AD.


Nature Reviews Drug Discovery | 2011

Convergent pathogenic pathways in Alzheimer's and Huntington's diseases: shared targets for drug development

Dagmar E. Ehrnhoefer; Bibiana K.Y. Wong; Michael R. Hayden

Neurodegenerative diseases, exemplified by Alzheimers disease and Huntingtons disease, are characterized by progressive neuropsychiatric dysfunction and loss of specific neuronal subtypes. Although there are differences in the exact sites of pathology, and the clinical profiles of these two conditions only partially overlap, considerable similarities in disease mechanisms and pathogenic pathways can be observed. These shared mechanisms raise the possibility of exploiting common therapeutic targets for drug development. As Huntingtons disease has a monogenic cause, it is possible to accurately identify individuals who carry the Huntingtons disease mutation but do not yet manifest symptoms. These individuals could act as a model for Alzheimers disease to test therapeutic interventions that target shared pathogenic pathways.

Collaboration


Dive into the Dagmar E. Ehrnhoefer's collaboration.

Top Co-Authors

Avatar

Michael R. Hayden

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Xiaofan Qiu

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Niels H. Skotte

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Rona K. Graham

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Safia Ladha

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sonia Franciosi

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Yu Deng

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Mahmoud A. Pouladi

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Amber L. Southwell

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Dale D. O. Martin

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge