49 results on '"Dagmar E. Ehrnhoefer"'
Search Results
2. Tau seed amplification assay reveals relationship between seeding and pathological forms of tau in Alzheimer’s disease brain
- Author
-
Bryan Frey, David Holzinger, Keenan Taylor, Dagmar E. Ehrnhoefer, Andreas Striebinger, Sandra Biesinger, Laura Gasparini, Michael J. O’Neill, Florian Wegner, Stefan Barghorn, Günter U. Höglinger, and Roland G. Heym
- Subjects
Alzheimer’s disease ,Tau seeding ,Seed amplification assay (SAA) ,Real-time quaking induced conversion assay (RT-QuIC) ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Tau seed amplification assays (SAAs) directly measure the seeding activity of tau and would therefore be ideal biomarkers for clinical trials targeting seeding-competent tau in Alzheimer’s disease (AD). However, the precise relationship between tau seeding measured by SAA and the levels of pathological forms of tau in the AD brain remains unknown. We developed a new tau SAA based on full-length 0N3R tau with sensitivity in the low fg/ml range and used it to characterize 103 brain samples from three independent cohorts. Tau seeding clearly discriminated between AD and control brain samples. Interestingly, seeding was absent in Progressive Supranuclear Palsy (PSP) putamen, suggesting that our tau SAA did not amplify 4R tau aggregates from PSP brain. The specificity of our tau SAA for AD brain was further supported by analysis of matched hippocampus and cerebellum samples. While seeding was detected in hippocampus from Braak stages I-II, no seeding was present in AD cerebellum that is devoid of tau inclusions. Analysis of 40 middle frontal gyrus samples encompassing all Braak stages showed that tau SAA seeding activity gradually increased with Braak stage. This relationship between seeding activity and the presence of tau inclusions in AD brain was further supported by robust correlations between tau SAA results and the levels of phosphorylated tau212/214, phosphorylated tau181, aggregated tau, and sarkosyl-insoluble tau. Strikingly, we detected tau seeding in the middle frontal gyrus already at Braak stage II-III, suggesting that tau SAA can detect tau pathology earlier than conventional immunohistochemical staining. In conclusion, our data suggest a quantitative relationship between tau seeding activity and pathological forms of tau in the human brain and provides an important basis for further development of tau SAA for accessible human samples.
- Published
- 2023
- Full Text
- View/download PDF
3. Fully co-factor-free ClearTau platform produces seeding-competent Tau fibrils for reconstructing pathological Tau aggregates
- Author
-
Galina Limorenko, Meltem Tatli, Rajasekhar Kolla, Sergey Nazarov, Marie-Theres Weil, David C. Schöndorf, Daniela Geist, Peter Reinhardt, Dagmar E. Ehrnhoefer, Henning Stahlberg, Laura Gasparini, and Hilal A. Lashuel
- Subjects
Science - Abstract
Abstract Tau protein fibrillization is implicated in the pathogenesis of several neurodegenerative diseases collectively known as Tauopathies. For decades, investigating Tau fibrillization in vitro has required the addition of polyanions or other co-factors to induce its misfolding and aggregation, with heparin being the most commonly used. However, heparin-induced Tau fibrils exhibit high morphological heterogeneity and a striking structural divergence from Tau fibrils isolated from Tauopathies patients’ brains at ultra- and macro-structural levels. To address these limitations, we developed a quick, cheap, and effective method for producing completely co-factor-free fibrils from all full-length Tau isoforms and mixtures thereof. We show that Tau fibrils generated using this ClearTau method – ClearTau fibrils - exhibit amyloid-like features, possess seeding activity in biosensor cells and hiPSC-derived neurons, retain RNA-binding capacity, and have morphological properties and structures more reminiscent of the properties of the brain-derived Tau fibrils. We present the proof-of-concept implementation of the ClearTau platform for screening Tau aggregation-modifying compounds. We demonstrate that these advances open opportunities to investigate the pathophysiology of disease-relevant Tau aggregates and will facilitate the development of Tau pathology-targeting and modifying therapies and PET tracers that can distinguish between different Tauopathies.
- Published
- 2023
- Full Text
- View/download PDF
4. Correction: Tau seed amplification assay reveals relationship between seeding and pathological forms of tau in Alzheimer’s disease brain
- Author
-
Bryan Frey, David Holzinger, Keenan Taylor, Dagmar E. Ehrnhoefer, Andreas Striebinger, Sandra Biesinger, Laura Gasparini, Michael J. O’Neill, Florian Wegner, Stefan Barghorn, Günter U. Höglinger, and Roland G. Heym
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 - Published
- 2024
- Full Text
- View/download PDF
5. Dual truncation of tau by caspase-2 accelerates its CHIP-mediated degradation
- Author
-
Lydia Reinhardt, Fabrizio Musacchio, Maria Bichmann, Annika Behrendt, Ebru Ercan-Herbst, Juliane Stein, Isabelle Becher, Per Haberkant, Julia Mader, David C. Schöndorf, Melanie Schmitt, Jürgen Korffmann, Peter Reinhardt, Christian Pohl, Mikhail Savitski, Corinna Klein, Laura Gasparini, Martin Fuhrmann, and Dagmar E. Ehrnhoefer
- Subjects
Tau ,Caspase-2 ,Proteolysis ,Tauopathy ,Degradation ,CHIP ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Intraneuronal aggregates of the microtubule binding protein Tau are a hallmark of different neurodegenerative diseases including Alzheimer's disease (AD). In these aggregates, Tau is modified by posttranslational modifications such as phosphorylation as well as by proteolytic cleavage. Here we identify a novel Tau cleavage site at aspartate 65 (D65) that is specific for caspase-2. In addition, we show that the previously described cleavage site at D421 is also efficiently processed by caspase-2, and both sites are cleaved in human brain samples. Caspase-2-generated Tau fragments show increased aggregation potential in vitro, but do not accumulate in vivo after AAV-mediated overexpression in mouse hippocampus. Interestingly, we observe that steady-state protein levels of caspase-2 generated Tau fragments are low in our in vivo model despite strong RNA expression, suggesting efficient clearance. Consistent with this hypothesis, we find that caspase-2 cleavage significantly improves the recognition of Tau by the ubiquitin E3 ligase CHIP, leading to increased ubiquitination and faster degradation of Tau fragments. Taken together our data thus suggest that CHIP-induced ubiquitination is of particular importance for the clearance of caspase-2 generated Tau fragments in vitro and in vivo.
- Published
- 2023
- Full Text
- View/download PDF
6. SETD7-mediated monomethylation is enriched on soluble Tau in Alzheimer’s disease
- Author
-
Maria Bichmann, Nuria Prat Oriol, Ebru Ercan-Herbst, David C. Schöndorf, Borja Gomez Ramos, Vera Schwärzler, Marie Neu, Annabelle Schlüter, Xue Wang, Liang Jin, Chenqi Hu, Yu Tian, Janina S. Ried, Per Haberkant, Laura Gasparini, and Dagmar E. Ehrnhoefer
- Subjects
Lysine methylation ,Protein methyl transferase ,Nuclear tau ,Posttranslational modification ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Human tauopathies including Alzheimer’s disease (AD) are characterized by alterations in the post-translational modification (PTM) pattern of Tau, which parallel the formation of insoluble Tau aggregates, neuronal dysfunction and degeneration. While PTMs on aggregated Tau have been studied in detail, much less is known about the modification patterns of soluble Tau. Furthermore, PTMs other than phosphorylation have only come into focus recently and are still understudied. Soluble Tau species are likely responsible for the spreading of pathology during disease progression and are currently being investigated as targets for immunotherapies. A better understanding of their biochemical properties is thus of high importance. Methods We used a mass spectrometry approach to characterize Tau PTMs on a detergent-soluble fraction of human AD and control brain tissue, which led to the discovery of novel lysine methylation events. We developed specific antibodies against Tau methylated at these sites and biochemically characterized methylated Tau species in extracts from human brain, the rTg4510 mouse model and in hiPSC-derived neurons. Results Our study demonstrates that methylated Tau levels increase with Tau pathology stage in human AD samples as well as in a mouse model of Tauopathy. Methylated Tau is enriched in soluble brain extracts and is not associated with hyperphosphorylated, high molecular weight Tau species. We also show that in hiPSC-derived neurons and mouse brain, methylated Tau preferentially localizes to the cell soma and nuclear fractions and is absent from neurites. Knock down and inhibitor studies supported by proteomics data led to the identification of SETD7 as a novel lysine methyltransferase for Tau. SETD7 specifically methylates Tau at K132, an event that facilitates subsequent methylation at K130. Conclusions Our findings indicate that methylated Tau has a specific somatic and nuclear localization, suggesting that the methylation of soluble Tau species may provide a signal for their translocation to different subcellular compartments. Since the mislocalization and depletion of Tau from axons is associated with tauopathies, our findings may shed light onto this disease-associated phenomenon.
- Published
- 2021
- Full Text
- View/download PDF
7. A post-translational modification signature defines changes in soluble tau correlating with oligomerization in early stage Alzheimer’s disease brain
- Author
-
Ebru Ercan-Herbst, Jens Ehrig, David C. Schöndorf, Annika Behrendt, Bernd Klaus, Borja Gomez Ramos, Nuria Prat Oriol, Christian Weber, and Dagmar E. Ehrnhoefer
- Subjects
Alzheimer’s disease ,Tau ,Posttranslational modifications ,Tau oligomerization ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Tau is a microtubule-binding protein that can receive various post-translational modifications (PTMs) including phosphorylation, methylation, acetylation, glycosylation, nitration, sumoylation and truncation. Hyperphosphorylation of tau is linked to its aggregation and the formation of neurofibrillary tangles (NFTs), which are a hallmark of Alzheimer’s disease (AD). While more than 70 phosphorylation sites have been detected previously on NFT tau, studies of oligomeric and detergent-soluble tau in human brains during the early stages of AD are lacking. Here we apply a comprehensive electrochemiluminescence ELISA assay to analyze twenty-five different PTM sites as well as tau oligomerization in control and sporadic AD brain. The samples were classified as Braak stages 0–I, II or III–IV, corresponding to the progression of microscopically detectable tau pathology throughout different brain regions. We found that soluble tau multimers are strongly increased at Braak stages III–IV in all brain regions under investigation, including the temporal cortex, which does not contain NFTs or misfolded oligomers at this stage of pathology. We additionally identified five phosphorylation sites that are specifically and consistently increased across the entorhinal cortex, hippocampus and temporal cortex in the same donors. Three of these sites correlate with tau multimerization in all three brain regions, but do not overlap with the epitopes of phospho-sensitive antibodies commonly used for the immunohistochemical detection of NFTs. Our results thus suggest that soluble multimers are characterized by a small set of specific phosphorylation events that differ from those dominating in mature NFTs. These findings shed light on early PTM changes of tau during AD pathogenesis in human brains.
- Published
- 2019
- Full Text
- View/download PDF
8. Preventing mutant huntingtin proteolysis and intermittent fasting promote autophagy in models of Huntington disease
- Author
-
Dagmar E. Ehrnhoefer, Dale D. O. Martin, Mandi E. Schmidt, Xiaofan Qiu, Safia Ladha, Nicholas S. Caron, Niels H. Skotte, Yen T. N. Nguyen, Kuljeet Vaid, Amber L. Southwell, Sabine Engemann, Sonia Franciosi, and Michael R. Hayden
- Subjects
Huntington disease ,Autophagy ,Proteolysis ,Caspase ,Mutant huntingtin lowering ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Huntington disease (HD) is caused by the expression of mutant huntingtin (mHTT) bearing a polyglutamine expansion. In HD, mHTT accumulation is accompanied by a dysfunction in basal autophagy, which manifests as specific defects in cargo loading during selective autophagy. Here we show that the expression of mHTT resistant to proteolysis at the caspase cleavage site D586 (C6R mHTT) increases autophagy, which may be due to its increased binding to the autophagy adapter p62. This is accompanied by faster degradation of C6R mHTT in vitro and a lack of mHTT accumulation the C6R mouse model with age. These findings may explain the previously observed neuroprotective properties of C6R mHTT. As the C6R mutation cannot be easily translated into a therapeutic approach, we show that a scheduled feeding paradigm is sufficient to lower mHTT levels in YAC128 mice expressing cleavable mHTT. This is consistent with a previous model, where the presence of cleavable mHTT impairs basal autophagy, while fasting-induced autophagy remains functional. In HD, mHTT clearance and autophagy may become increasingly impaired as a function of age and disease stage, because of gradually increased activity of mHTT-processing enzymes. Our findings imply that mHTT clearance could be enhanced by a regulated dietary schedule that promotes autophagy.
- Published
- 2018
- Full Text
- View/download PDF
9. A validated antibody panel for the characterization of tau post-translational modifications
- Author
-
Ebru Ercan, Sameh Eid, Christian Weber, Alexandra Kowalski, Maria Bichmann, Annika Behrendt, Frank Matthes, Sybille Krauss, Peter Reinhardt, Simone Fulle, and Dagmar E. Ehrnhoefer
- Subjects
Tau ,Post-translational modification ,Antibody validation ,Alzheimer’s disease ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Tau is a microtubule-binding protein, which is subject to various post-translational modifications (PTMs) including phosphorylation, methylation, acetylation, glycosylation, nitration, sumoylation and truncation. Aberrant PTMs such as hyperphosphorylation result in tau aggregation and the formation of neurofibrillary tangles, which are a hallmark of Alzheimer’s disease (AD). In order to study the importance of PTMs on tau function, antibodies raised against specific modification sites are widely used. However, quality control of these antibodies is lacking and their specificity for particular modifications is often unclear. Methods In this study, we first designed an online tool called ‘TauPTM’, which enables the visualization of PTMs and their interactions on human tau. Using TauPTM, we next searched for commercially available antibodies against tau PTMs and characterized their specificity by peptide array, immunoblotting, electrochemiluminescence ELISA and immunofluorescence technologies. Results We demonstrate that commercially available antibodies can show a significant lack of specificity, and PTM-specific antibodies in particular often recognize non-modified versions of the protein. In addition, detection may be hindered by other PTMs in close vicinity, complicating the interpretation of results. Finally, we compiled a panel of specific antibodies and show that they are useful to detect PTM-modified endogenous tau in hiPSC-derived neurons and mouse brains. Conclusion This study has created a platform to reliably and robustly detect changes in localization and abundance of post-translationally modified tau in health and disease. A web-based version of TauPTM is fully available at http://www.tauptm.org .
- Published
- 2017
- Full Text
- View/download PDF
10. Asparagine endopeptidase cleaves tau at N167 after uptake into microglia
- Author
-
Annika Behrendt, Maria Bichmann, Ebru Ercan-Herbst, Per Haberkant, David C. Schöndorf, Michael Wolf, Salma A. Fahim, Enrico Murolo, and Dagmar E. Ehrnhoefer
- Subjects
Tau ,Proteolysis ,Alzheimer's disease ,AEP ,Microglia ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Tau cleavage by different proteolytic enzymes generates short, aggregation-prone fragments that have been implicated in the pathogenesis of Alzheimer's disease (AD). Asparagine endopeptidase (AEP) activity in particular has been associated with tau dysfunction and aggregation, and the activity of the protease is increased in both aging and AD. Using a mass spectrometry approach, we identified a novel tau cleavage site at N167 and confirmed its processing by AEP. In combination with the previously known site at N368, we show that AEP cleavage yields a tau fragment that is present in both control and AD brains at similar levels. AEP is a lysosomal enzyme, and our data suggest that it is expressed in microglia rather than in neurons. Accordingly, we observe tau cleavage at N167 and N368 after endocytotic uptake into microglia, but not neurons. However, tau168–368 does not accumulate in microglia and we thus conclude that the fragment is part of a proteolytic cascade leading to tau degradation. While we confirm previous studies showing increased overall AEP activity in AD brains, our data suggests that AEP-mediated cleavage of tau is a physiological event occurring during microglial degradation of the secreted neuronal protein. As a consequence, we caution against preventing AEP-mediated tau cleavage as a therapeutic approach in AD.
- Published
- 2019
- Full Text
- View/download PDF
11. Generation of an induced pluripotent stem cell cohort suitable to investigate sporadic Alzheimer's Disease
- Author
-
David C. Schöndorf, Myriam Elschami, Maximilian Schieck, Ebru Ercan-Herbst, Christian Weber, Yasmin Riesinger, Sara Kalman, Doris Steinemann, and Dagmar E. Ehrnhoefer
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Alzheimer's Disease (AD) is the major cause of dementia in the elderly, and cortical neurons differentiated from patient-derived induced pluripotent stem cells (iPSCs) can recapitulate disease phenotypes such as tau phosphorylation or amyloid beta (Aß) deposition. Here we describe the generation of an iPSC cohort consisting of 2 sporadic AD cases and 3 controls, derived from dermal fibroblasts. All lines were karyotypically normal, showed expression of stem cell markers and efficiently differentiated into cells of all three germ layers.
- Published
- 2019
- Full Text
- View/download PDF
12. Partial rescue of some features of Huntington Disease in the genetic absence of caspase-6 in YAC128 mice
- Author
-
Bibiana K.Y. Wong, Dagmar E. Ehrnhoefer, Rona K. Graham, Dale D.O. Martin, Safia Ladha, Valeria Uribe, Lisa M. Stanek, Sonia Franciosi, Xiaofan Qiu, Yu Deng, Vlad Kovalik, Weining Zhang, Mahmoud A. Pouladi, Lamya S. Shihabuddin, and Michael R. Hayden
- Subjects
Caspase-6 ,YAC128 ,Huntington Disease ,Autophagy ,p62 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Huntington Disease (HD) is a progressive neurodegenerative disease caused by an elongated CAG repeat in the huntingtin (HTT) gene that encodes a polyglutamine tract in the HTT protein. Proteolysis of the mutant HTT protein (mHTT) has been detected in human and murine HD brains and is implicated in the pathogenesis of HD. Of particular importance is the site at amino acid (aa) 586 that contains a caspase-6 (Casp6) recognition motif.Activation of Casp6 occurs presymptomatically in human HD patients and the inhibition of mHTT proteolysis at aa586 in the YAC128 mouse model results in the full rescue of HD-like phenotypes. Surprisingly, Casp6 ablation in two different HD mouse models did not completely prevent the generation of this fragment, and therapeutic benefits were limited, questioning the role of Casp6 in the disease.We have evaluated the impact of the loss of Casp6 in the YAC128 mouse model of HD. Levels of the mHTT-586 fragment are reduced but not absent in the absence of Casp6 and we identify caspase 8 as an alternate enzyme that can generate this fragment. In vivo, the ablation of Casp6 results in a partial rescue of body weight gain, normalized IGF-1 levels, a reversal of the depression-like phenotype and decreased HTT levels. In the YAC128/Casp6−/− striatum there is a concomitant reduction in p62 levels, a marker of autophagic activity, suggesting increased autophagic clearance. These results implicate the HTT-586 fragment as a key contributor to certain features of HD, irrespective of the enzyme involved in its generation.
- Published
- 2015
- Full Text
- View/download PDF
13. NP03, a novel low-dose lithium formulation, is neuroprotective in the YAC128 mouse model of Huntington disease
- Author
-
Mahmoud A. Pouladi, Elsa Brillaud, Yuanyun Xie, Paola Conforti, Rona K. Graham, Dagmar E. Ehrnhoefer, Sonia Franciosi, Weining Zhang, Patrick Poucheret, Elsa Compte, Jean-Claude Maurel, Chiara Zuccato, Elena Cattaneo, Christian Néri, and Michael R. Hayden
- Subjects
Huntington disease ,Transgenic mouse model ,Lithium ,Caspase-6 ,BDNF ,GSK-3 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Huntington disease (HD), a neurodegenerative disorder caused by an expanded CAG repeat in the HTT gene, remains without a treatment to modify the course of the illness. Lithium, a drug widely used for the treatment of bipolar disorder, has been shown to exert neuroprotective effects in a number of models of neurological disease but may have various toxic effects at conventional therapeutic doses. We examined whether NP03, a novel low-dose lithium microemulsion, would improve the disease phenotypes in the YAC128 mouse model of HD. We demonstrate that NP03 improves motor function, ameliorates the neuropathological deficits in striatal volume, neuronal counts, and DARPP-32 expression, and partially rescues testicular atrophy in YAC128 mice. These positive effects were accompanied by improvements in multiple biochemical endpoints associated with the pathogenesis of HD, including normalization of caspase-6 activation and amelioration of deficits in BDNF levels, and with no lithium-related toxicity. Our findings demonstrate that NP03 ameliorates the motor and neuropathological phenotypes in the YAC128 mouse model of HD, and represents a potential therapeutic approach for HD.
- Published
- 2012
- Full Text
- View/download PDF
14. Tau in the brain interstitial fluid is fragmented and seeding–competent
- Author
-
Florie LePrieult, Ina Mairhofer, Karen Bodie, Jonas Hoppe, Marcus W. Meinhardt, Sonja Julier, Gudrun Plotzky, Ebru Ercan-Herbst, Yulia Mordashova, Sandra Biesinger, Miroslav Cik, Kerstin Schlegel, Dagmar E. Ehrnhoefer, Laura Gasparini, Esther Rodriguez-Correa, Mario Mezler, Corinna Klein, Andreas Striebinger, and Erica Barini
- Subjects
Genetically modified mouse ,Aging ,Microdialysis ,Transgene ,Tau protein ,Mice, Transgenic ,tau Proteins ,Protein Aggregation, Pathological ,In vivo ,Interstitial fluid ,mental disorders ,medicine ,Animals ,Humans ,Phosphorylation ,biology ,Chemistry ,General Neuroscience ,Brain ,Extracellular Fluid ,medicine.disease ,Peptide Fragments ,Cell biology ,Disease Models, Animal ,HEK293 Cells ,Tauopathies ,biology.protein ,Neurology (clinical) ,Tauopathy ,Geriatrics and Gerontology ,Alzheimer's disease ,Developmental Biology - Abstract
In Alzheimer disease, Tau pathology is thought to propagate from cell to cell throughout interconnected brain areas. However, the forms of Tau released into the brain interstitial fluid (ISF) in vivo during the development of Tauopathy and their pathological relevance remain unclear. Combining in vivo microdialysis and biochemical analysis, we find that in Tau transgenic mice, human Tau (hTau) present in brain ISF is truncated and comprises at least 10 distinct fragments spanning the entire Tau protein. The fragmentation pattern is similar across different Tau transgenic models, pathological stages and brain areas. ISF hTau concentration decreases during Tauopathy progression, while its phosphorylation increases. ISF from mice with established Tauopathy induces Tau aggregation in HEK293-Tau biosensor cells. Notably, immunodepletion of ISF phosphorylated Tau, but not Tau fragments, significantly reduces its ability to seed Tau aggregation and only a fraction of Tau, separated by ultracentrifugation, is seeding competent. These results indicate that ISF seeding competence is driven by a small subset of Tau, which potentially contribute to the propagation of Tau pathology.HighlightsIn interstitial fluid (ISF) of transgenic mice, Tau comprises >10 distinct fragmentsISF Tau decreases with Tauopathy progression, while its phosphorylation increasesOnly ISF from mice with established Tauopathy is seeding competent in vitroRemoval of phospho-Tau reduces ISF seeding competenceISF seeding competence is driven by less soluble, aggregated and phosphorylated TauGraphical abstract
- Published
- 2022
- Full Text
- View/download PDF
15. A post-translational modification signature defines changes in soluble tau correlating with oligomerization in early stage Alzheimer’s disease brain
- Author
-
David C. Schöndorf, Borja Gomez Ramos, Bernd Klaus, Annika Behrendt, Jens Ehrig, Nuria Prat Oriol, Christian Weber, Dagmar E. Ehrnhoefer, and Ebru Ercan-Herbst
- Subjects
Male ,Induced Pluripotent Stem Cells ,SUMO protein ,Hippocampus ,Hyperphosphorylation ,tau Proteins ,Epitope ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,mental disorders ,Humans ,Phosphorylation ,lcsh:Neurology. Diseases of the nervous system ,Aged ,Temporal cortex ,Aged, 80 and over ,Chemistry ,Research ,Brain ,Methylation ,Tau oligomerization ,Entorhinal cortex ,Cell biology ,Posttranslational modifications ,Female ,Neurology (clinical) ,Tau ,Protein Processing, Post-Translational ,Alzheimer’s disease - Abstract
Tau is a microtubule-binding protein that can receive various post-translational modifications (PTMs) including phosphorylation, methylation, acetylation, glycosylation, nitration, sumoylation and truncation. Hyperphosphorylation of tau is linked to its aggregation and the formation of neurofibrillary tangles (NFTs), which are a hallmark of Alzheimer’s disease (AD). While more than 70 phosphorylation sites have been detected previously on NFT tau, studies of oligomeric and detergent-soluble tau in human brains during the early stages of AD are lacking. Here we apply a comprehensive electrochemiluminescence ELISA assay to analyze twenty-five different PTM sites as well as tau oligomerization in control and sporadic AD brain. The samples were classified as Braak stages 0–I, II or III–IV, corresponding to the progression of microscopically detectable tau pathology throughout different brain regions. We found that soluble tau multimers are strongly increased at Braak stages III–IV in all brain regions under investigation, including the temporal cortex, which does not contain NFTs or misfolded oligomers at this stage of pathology. We additionally identified five phosphorylation sites that are specifically and consistently increased across the entorhinal cortex, hippocampus and temporal cortex in the same donors. Three of these sites correlate with tau multimerization in all three brain regions, but do not overlap with the epitopes of phospho-sensitive antibodies commonly used for the immunohistochemical detection of NFTs. Our results thus suggest that soluble multimers are characterized by a small set of specific phosphorylation events that differ from those dominating in mature NFTs. These findings shed light on early PTM changes of tau during AD pathogenesis in human brains.
- Published
- 2019
16. Constitutive ablation of caspase-6 reduces the inflammatory response and behavioural changes caused by peripheral pro-inflammatory stimuli
- Author
-
Safia Ladha, Lorenzo Casal, Nicholas S. Caron, Dagmar E. Ehrnhoefer, Michael R. Hayden, and Xiaofan Qiu
- Subjects
0301 basic medicine ,Cancer Research ,Lipopolysaccharide ,Immunology ,Regulator ,Inflammation ,Caspase 6 ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,lcsh:QH573-671 ,Caspase ,biology ,lcsh:Cytology ,Cell Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Pathophysiology ,Cell biology ,030104 developmental biology ,chemistry ,Apoptosis ,biology.protein ,Tumor necrosis factor alpha ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
Traditionally, the family of caspases has been subcategorised according to their respective main roles in mediating apoptosis or inflammation. However, recent studies have revealed that caspases participate in diverse cellular functions beyond their canonical roles. Caspase-6 (C6) is one such protease known for its role as a pro-apoptotic executioner caspase and its aberrant activity in several neurodegenerative diseases. In addition to apoptosis, C6 has been shown to regulate B-cell activation and differentiation in plasma cells as well as macrophage activation. Furthermore, C6 has recently been postulated to play a role in mediating the inflammatory response through the production of TNF-α. In this study we further examine the role of C6 in mediating the inflammatory response and its contribution to the manifestation of behavioural abnormalities in mice. We find that C6 is a positive regulator of TNF-α transcription in macrophages and that ablation of C6 reduces lipopolysaccharide (LPS)-induced TNF-α levels in plasma. Furthermore, loss of C6 attenuates LPS-induced behavioural changes in mice and protects neurons from cytokine-mediated toxicity. These data further support the involvement of C6 in the inflammatory response and point to a previously unknown role for C6 in the pathophysiology of depression.
- Published
- 2018
- Full Text
- View/download PDF
17. HACE1 is essential for astrocyte mitochondrial function and influences Huntington disease phenotypes in vivo
- Author
-
Sabine Waltl, Dagmar E. Ehrnhoefer, Boguslaw Felczak, Lorenzo Casal, Amber L. Southwell, Meenalochani Sivasubramanian, Lisa M. Anderson, Anita Fazeli, Xiaofan Qiu, Erika B. Villanueva, Yuanyun Xie, Michelle Tsang, and Michael R. Hayden
- Subjects
0301 basic medicine ,HECT domain ,Huntingtin ,NF-E2-Related Factor 2 ,Ubiquitin-Protein Ligases ,Nerve Tissue Proteins ,Context (language use) ,Biology ,Mitochondrion ,medicine.disease_cause ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Molecular Biology ,Genetics (clinical) ,Tumor Suppressor Proteins ,Articles ,General Medicine ,medicine.disease ,Corpus Striatum ,Mitochondria ,3. Good health ,Astrogliosis ,Ubiquitin ligase ,Cell biology ,Neostriatum ,Disease Models, Animal ,Oxidative Stress ,Huntington Disease ,030104 developmental biology ,medicine.anatomical_structure ,Astrocytes ,biology.protein ,030217 neurology & neurosurgery ,Oxidative stress ,Astrocyte - Abstract
Oxidative stress is a prominent feature of Huntington disease (HD), and we have shown previously that reduced levels of hace1 (HECT domain and Ankyrin repeat containing E3 ubiquitin protein ligase 1) in patient striatum may contribute to the pathogenesis of HD. Hace1 promotes the stability of Nrf2 and thus plays an important role in antioxidant response mechanisms, which are dysfunctional in HD. Moreover, hace1 overexpression mitigates mutant huntingtin (mHTT)-induced oxidative stress in vitro through promotion of the Nrf2 antioxidant response. Here, we show that the genetic ablation of hace1 in the YAC128 mouse model of HD accelerates motor deficits and exacerbates cognitive and psychiatric phenotypes in vivo. We find that both the expression of mHTT and the ablation of hace1 alone are sufficient to cause deficits in astrocytic mitochondrial respiration. We confirm the crucial role of hace1 in astrocytes in vivo, since its ablation is sufficient to cause dramatic astrogliosis in wild-type FVB/N mice. Astrogliosis is not observed in the presence of mHTT but a strong dysregulation in the expression of astrocytic markers in HACE1-/- x YAC128 striatum suggests an additive effect of mHTT expression and hace1 loss on this cell type. HACE1-/- x YAC128 mice and primary cells derived from these animals therefore provide model systems that will allow for the further dissection of Nrf2 pathways and astrocyte dysfunction in the context of HD.
- Published
- 2017
- Full Text
- View/download PDF
18. SETD7-mediated lysine monomethylation is abundant on non-hyperphosphorylated nuclear Tau
- Author
-
Dagmar E. Ehrnhoefer, Borja Gomez Ramos, David C. Schöndorf, Ebru Ercan-Herbst, Nuria Prat Oriol, Per Haberkant, Laura Gasparini, Vera Schwärzler, and Maria Bichmann
- Subjects
medicine.anatomical_structure ,Chemistry ,Lysine ,medicine ,Chromosomal translocation ,Methylation ,Tauopathy ,Human brain ,Methylation Site ,medicine.disease ,Protein Lysine Methyltransferase ,Function (biology) ,Cell biology - Abstract
Human tauopathies including Alzheimer’s disease (AD) are characterized by alterations in the post-translational modification (PTM) pattern of Tau, leading to the formation of insoluble aggregates, neuronal dysfunction and degeneration. Using a mass spectrometry approach, we identified multiple sites of lysine monomethylation on Tau isolated from a detergent-soluble fraction of human brain, some of which were increased in early AD samples. Brain tissues derived from a mouse model of tauopathy demonstrate an age-dependent increase in methylation at specific sites, with methylated Tau enriched in the soluble nuclear fraction and not associated with hyperphosphorylated, insoluble Tau species. Furthermore, we show that the protein lysine methyltransferase SETD7 methylates Tau at K132 and demonstrate an interaction with K130, an additional methylation site in close vicinity. These findings shed light on the function of a novel type of PTM on Tau that provide a potential signal for its translocation to different subcellular sites. Since the mislocalization and depletion of Tau from axons is associated with tauopathies, our findings may furthermore provide insight into this disease-associated phenomenon.
- Published
- 2020
- Full Text
- View/download PDF
19. Asparagine endopeptidase cleaves tau at N167 after uptake into microglia
- Author
-
David C. Schöndorf, Ebru Ercan-Herbst, Annika Behrendt, Dagmar E. Ehrnhoefer, Salma A. Fahim, Maria Bichmann, Per Haberkant, Enrico Murolo, and Michael Wolf
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Proteolysis ,tau Proteins ,Cleavage (embryo) ,Mass Spectrometry ,lcsh:RC321-571 ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Asparagine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Neurons ,chemistry.chemical_classification ,Protease ,medicine.diagnostic_test ,Microglia ,Chemistry ,Proteolytic enzymes ,AEP ,Brain ,Alzheimer's disease ,Endopeptidase ,Cell biology ,Cysteine Endopeptidases ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,Enzyme ,Neurology ,Tau ,030217 neurology & neurosurgery - Abstract
BackgroundTau cleavage by different proteolytic enzymes generates short, aggregation-prone fragments that have been implicated in the pathogenesis of Alzheimer’s disease (AD). Asparagine endopeptidase (AEP) activity in particular has been associated with tau dysfunction and aggregation, and the activity of the protease is increased in both aging and AD.Methods and ResultsUsing a mass spectrometry approach we identified a novel tau cleavage site at N167 and confirmed its processing by AEP. In combination with the previously known site at N368, we show that AEP cleavage yields a tau fragment that is present in both control and AD brains at similar levels. AEP is a lysosomal enzyme, and our data suggest that it is expressed in microglia rather than in neurons. Accordingly, we observe tau cleavage at N167 and N368 after endocytotic uptake into microglia, but not neurons. However, tau168-368 does not accumulate in microglia and we thus conclude that the fragment is part of a proteolytic cascade leading to tau degradation.ConclusionsWhile we confirm previous studies showing increased overall AEP activity in AD brains, our data suggests that AEP-mediated cleavage of tau is a physiological event occurring during microglial degradation of the secreted neuronal protein. The disease-associated increase in active AEP may thus be related to pro-inflammatory conditions in AD brains, and our findings argue against AEP inhibition as a therapeutic approach in AD.
- Published
- 2019
20. A post-translational modification signature defines changes in soluble tau correlating with oligomerization in early stage Alzheimer’s disease brain
- Author
-
Borja Gomez Ramos, Christian Weber, Bernd Klaus, Dagmar E. Ehrnhoefer, David C. Schöndorf, Ebru Ercan-Herbst, and Annika Behrendt
- Subjects
Temporal cortex ,Chemistry ,mental disorders ,SUMO protein ,Phosphorylation ,Hippocampus ,Hyperphosphorylation ,Methylation ,Entorhinal cortex ,Epitope ,Cell biology - Abstract
Tau is a microtubule-binding protein that can receive various post-translational modifications (PTMs) including phosphorylation, methylation, acetylation, glycosylation, nitration, sumoylation and truncation. Hyperphosphorylation of tau is linked to its aggregation and the formation of neurofibrillary tangles (NFTs), which are a hallmark of Alzheimer’s disease (AD). While more than 70 phosphorylation sites have been detected previously on NFT tau, studies of oligomeric and detergent-soluble tau in human brains during the early stages of AD are lacking. Here we apply a comprehensive electrochemiluminescence ELISA assay to analyze twenty-five different PTM sites as well as tau oligomerization in control and sporadic AD brain. The samples were classified as Braak stages 0-I, II or III-IV, respectively, corresponding to the progression of microscopically detectable tau pathology throughout different brain regions. We find that soluble tau oligomers are strongly increased at Braak stages III-IV in all brain regions under investigation, including the temporal cortex, which does not contain NFTs at this stage of pathology. We additionally identified five phosphorylation sites that are specifically and consistently increased across the entorhinal cortex, hippocampus and temporal cortex in the same donors. Three of these sites correlate with tau oligomerization in all three brain regions, but do not overlap with the epitopes of phospho-sensitive antibodies commonly used for the immunohistochemical detection of NFTs. Our results thus suggest that soluble oligomers are characterized by a small set of specific phosphorylation events that differ from those dominating in mature NFTs and shed light on early PTM changes of tau during AD pathogenesis in human brains.
- Published
- 2019
- Full Text
- View/download PDF
21. Palmitoylation of caspase-6 by HIP14 regulates its activation
- Author
-
Xiaofan Qiu, Dagmar E. Ehrnhoefer, Roshni R. Singaraja, Shaun S. Sanders, Kuljeet Vaid, Michael R. Hayden, Niels H. Skotte, Srinivasaragavan Kannan, and Chandra S. Verma
- Subjects
0301 basic medicine ,Programmed cell death ,Immunoprecipitation ,Lipoylation ,Regulator ,Caspase 6 ,Molecular Dynamics Simulation ,Biology ,Substrate Specificity ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Palmitoylation ,Chlorocebus aethiops ,medicine ,Animals ,Palmitoyl acyltransferase ,Molecular Biology ,Original Paper ,COS cells ,Neurodegeneration ,Brain ,Cell Biology ,medicine.disease ,Protein Structure, Tertiary ,Cell biology ,Disease Models, Animal ,Huntington Disease ,030104 developmental biology ,COS Cells ,Mutagenesis, Site-Directed ,lipids (amino acids, peptides, and proteins) ,Dimerization ,Acyltransferases ,030217 neurology & neurosurgery - Abstract
Caspase-6 (CASP6) has an important role in axonal degeneration during neuronal apoptosis and in the neurodegenerative diseases Alzheimer and Huntington disease. Decreasing CASP6 activity may help to restore neuronal function in these and other diseases such as stroke and ischemia, where increased CASP6 activity has been implicated. The key to finding approaches to decrease CASP6 activity is a deeper understanding of the mechanisms regulating CASP6 activation. We show that CASP6 is posttranslationally palmitoylated by the palmitoyl acyltransferase HIP14 and that the palmitoylation of CASP6 inhibits its activation. Palmitoylation of CASP6 is decreased both in Hip14−/− mice, where HIP14 is absent, and in YAC128 mice, a model of Huntington disease, where HIP14 is dysfunctional and where CASP6 activity is increased. Molecular modeling suggests that palmitoylation of CASP6 may inhibit its activation via steric blockage of the substrate-binding groove and inhibition of CASP6 dimerization, both essential for CASP6 function. Our studies identify palmitoylation as a novel CASP6 modification and as a key regulator of CASP6 activity.
- Published
- 2016
- Full Text
- View/download PDF
22. Compromised IGF signaling causes caspase-6 activation in Huntington disease
- Author
-
Xiaofan Qiu, Troels Tolstrup Nielsen, Signe Marie Borch Nielsen, Dagmar E. Ehrnhoefer, Michael R. Hayden, Anne Nørremølle, Mahmoud A. Pouladi, Niels H. Skotte, and Katie Huynh
- Subjects
0301 basic medicine ,Genetically modified mouse ,Programmed cell death ,Huntingtin ,medicine.medical_treatment ,Mutant ,Mice, Transgenic ,Caspase 6 ,Biology ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,medicine ,Animals ,Humans ,Insulin-Like Growth Factor I ,Huntingtin Protein ,Cell Death ,Growth factor ,Cell biology ,Enzyme Activation ,Huntington Disease ,Insulin-Like Growth Factor Binding Protein 3 ,Neuroprotective Agents ,030104 developmental biology ,Neurology ,Signal transduction ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Huntington disease (HD) is an autosomal dominant neurodegenerative disorder caused by an expansion of a polyglutamine repeat in the huntingtin (HTT) protein. Aberrant activation of caspase-6 and cleavage of mutant HTT generating the toxic N-terminal 586 HTT fragment are important steps in the pathogenesis of HD. Similarly, alterations in the insulin-like growth factor 1 (IGF-1) signaling pathway have been implicated in the disease as a result of decreased plasma IGF-1 levels in HD patients. In addition, two recent studies have demonstrated therapeutic benefit of IGF-1 treatment in mouse models of HD. Since IGF-1 promotes pro-survival pathways, we examined the relationship between IGF-1 signaling and aberrant caspase-6 activation in HD. Using immortalized mouse striatal cells expressing wild-type (STHdhQ7) or mutant HTT (STHdhQ111), we show that reduced levels of IGF-1 are associated with enhanced activation of caspase-6, increased cell death, and mutant HTT cleavage in a cellular stress paradigm. We demonstrate that IGF-1 supplementation reverses these effects and lowers the level of the toxic 586 HTT fragment. In addition, transcriptional analysis in the R6/2 HD transgenic mouse model demonstrated that the IGF-1 signaling system is dysregulated at multiple levels in several tissues including liver, muscle, and brain. Among these changes, we found increased expression of IGF-1 binding protein 3 (IGFBP-3), which may further reduce the bioavailability of IGF-1 as a consequence of increased IGF-1 binding. Our findings thus suggest that the therapeutic benefit of IGF-1 supplementation in HD may be significantly improved if other defects in the IGF-1 signaling pathway are corrected concurrently.
- Published
- 2020
- Full Text
- View/download PDF
23. Activation of Caspase-6 Is Promoted by a Mutant Huntingtin Fragment and Blocked by an Allosteric Inhibitor Compound
- Author
-
Mehdi Khankischpur, Björn Windshügel, Priyadarshini Jaishankar, Adelia Razeto, Olga A. Petina, Ilka Wittig, Adam R. Renslo, Philip Gribbon, Matthias Meyer zu Rheda, Detlef Geffken, Nicholas S. Caron, Jeanette Reinshagen, Safia Ladha, Dagmar E. Ehrnhoefer, Khuong Tuyen Huynh, Niels H. Skotte, Yen T. Nguyen, Yu Deng, Xiaofan Qiu, Sheraz Gul, Michael R. Hayden, and Publica
- Subjects
non-apoptotic ,Huntington's Disease ,Huntingtin ,Clinical Biochemistry ,Allosteric regulation ,Mutant ,Nerve Tissue Proteins ,Apoptosis ,Caspase 6 ,Neurodegenerative ,Biology ,caspase-6 ,Cleavage (embryo) ,01 natural sciences ,Biochemistry ,Article ,allosteric ,Pathogenesis ,Rare Diseases ,Allosteric Regulation ,Zymogen ,Chlorocebus aethiops ,Drug Discovery ,Animals ,Molecular Biology ,Neurons ,Pharmacology ,Huntingtin Protein ,010405 organic chemistry ,Neurosciences ,Nuclear Proteins ,Huntington disease ,Brain Disorders ,3. Good health ,0104 chemical sciences ,Cell biology ,Molecular Docking Simulation ,inhibitor ,Huntington Disease ,Zymogen activation ,COS Cells ,Molecular Medicine - Abstract
Aberrant activation of caspase-6 (C6) in the absence of other hallmarks of apoptosis has been demonstrated in cells and tissues from patients with Huntington disease (HD) and animal models. C6 activity correlates with disease progression in patients with HD and the cleavage of mutant huntingtin (mHTT) protein is thought to strongly contribute to disease pathogenesis. Here we show that the mHTT1-586 fragment generated by C6 cleavage interacts with the zymogen form of the enzyme, stabilizing a conformation that contains an active site and is prone to full activation. This shift toward enhanced activity can be prevented by a small-molecule inhibitor that blocks the interaction between C6 and mHTT1-586. Molecular docking studies suggest that the inhibitor binds an allosteric site in the C6 zymogen. The interaction of mHTT1-586 with C6 may therefore promote a self-reinforcing, feedforward cycle of C6 zymogen activation and mHTT cleavage driving HD pathogenesis.
- Published
- 2019
24. Preventing mutant huntingtin proteolysis and intermittent fasting promote autophagy in models of Huntington disease
- Author
-
Mandi E. Schmidt, Nicholas S. Caron, Dagmar E. Ehrnhoefer, Kuljeet Vaid, Sabine Engemann, Amber L. Southwell, Sonia Franciosi, Safia Ladha, Yen T. Nguyen, Xiaofan Qiu, Niels H. Skotte, Michael R. Hayden, and Dale D.O. Martin
- Subjects
Male ,0301 basic medicine ,Aging ,Huntingtin ,Proteolysis ,Mutant ,Mice, Transgenic ,Neuroprotection ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,Random Allocation ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Chlorocebus aethiops ,Intermittent fasting ,Autophagy ,medicine ,Animals ,Humans ,Cells, Cultured ,lcsh:Neurology. Diseases of the nervous system ,Caspase ,Cerebral Cortex ,Neurons ,Huntingtin Protein ,medicine.diagnostic_test ,biology ,Research ,Fasting ,Huntington disease ,In vitro ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,Liver ,COS Cells ,Mutation ,biology.protein ,Female ,Mutant huntingtin lowering ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Huntington disease (HD) is caused by the expression of mutant huntingtin (mHTT) bearing a polyglutamine expansion. In HD, mHTT accumulation is accompanied by a dysfunction in basal autophagy, which manifests as specific defects in cargo loading during selective autophagy. Here we show that the expression of mHTT resistant to proteolysis at the caspase cleavage site D586 (C6R mHTT) increases autophagy, which may be due to its increased binding to the autophagy adapter p62. This is accompanied by faster degradation of C6R mHTT in vitro and a lack of mHTT accumulation the C6R mouse model with age. These findings may explain the previously observed neuroprotective properties of C6R mHTT. As the C6R mutation cannot be easily translated into a therapeutic approach, we show that a scheduled feeding paradigm is sufficient to lower mHTT levels in YAC128 mice expressing cleavable mHTT. This is consistent with a previous model, where the presence of cleavable mHTT impairs basal autophagy, while fasting-induced autophagy remains functional. In HD, mHTT clearance and autophagy may become increasingly impaired as a function of age and disease stage, because of gradually increased activity of mHTT-processing enzymes. Our findings imply that mHTT clearance could be enhanced by a regulated dietary schedule that promotes autophagy. Electronic supplementary material The online version of this article (10.1186/s40478-018-0518-0) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
25. A Huntingtin-based peptide inhibitor of caspase-6 provides protection from mutant Huntingtin-induced motor and behavioral deficits
- Author
-
Israel Aharony, Dagmar E. Ehrnhoefer, Sonia Franciosi, Adi Shruster, Xiaofan Qiu, Daniel Offen, and Michael R. Hayden
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Proteolysis ,Mutant ,Nerve Tissue Proteins ,Peptide ,Caspase 6 ,Mice ,mental disorders ,Genetics ,Huntingtin Protein ,medicine ,Animals ,Molecular Biology ,Genetics (clinical) ,Caspase ,chemistry.chemical_classification ,Behavior, Animal ,biology ,medicine.diagnostic_test ,Nuclear Proteins ,Articles ,General Medicine ,Caspase Inhibitors ,Molecular biology ,Cell biology ,Enzyme Activation ,Disease Models, Animal ,Huntington Disease ,Neuroprotective Agents ,chemistry ,Mutation ,Toxicity ,biology.protein ,Female ,Peptides ,Psychomotor Performance - Abstract
Over the past decade, increasing evidence has implied a significant connection between caspase-6 activity and the pathogenesis of Huntington's disease (HD). Consequently, inhibiting caspase-6 activity was suggested as a promising therapeutic strategy to reduce mutant Huntingtin toxicity, and to provide protection from mutant Huntingtin-induced motor and behavioral deficits. Here, we describe a novel caspase-6 inhibitor peptide based on the huntingtin caspase-6 cleavage site, fused with a cell-penetrating sequence. The peptide reduces mutant Huntingtin proteolysis by caspase-6, and protects cells from mutant Huntingtin toxicity. Continuous subcutaneous administration of the peptide protected pre-symptomatic BACHD mice from motor deficits and behavioral abnormalities. Moreover, administration of the peptide in an advanced disease state resulted in the partial recovery of motor performance, and an alleviation of depression-related behavior and cognitive deficits. Our findings reveal the potential of substrate-based caspase inhibition as a therapeutic strategy, and present a promising agent for the treatment of HD.
- Published
- 2015
- Full Text
- View/download PDF
26. Feeding schedule and proteolysis regulate autophagic clearance of mutant huntingtin
- Author
-
Dagmar E. Ehrnhoefer, Yen T. Nguyen, Nicholas S. Caron, Safia Ladha, Dale D.O. Martin, Niels H. Skotte, Sabine Engemann, Hayden, Sonia Franciosi, and Xiaofan Qiu
- Subjects
chemistry.chemical_classification ,0303 health sciences ,Huntingtin ,medicine.diagnostic_test ,Proteolysis ,Mutant ,Autophagy ,Biology ,Cleavage (embryo) ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Enzyme ,chemistry ,Biochemistry ,medicine ,030217 neurology & neurosurgery ,030304 developmental biology ,Therapeutic strategy - Abstract
The expression of mutant huntingtin (mHTT) causes Huntington disease (HD), and lowering its levels is therefore an attractive therapeutic strategy. Here we show that scheduled feeding significantly decreases mHTT protein levels through enhanced autophagy in the CNS of an HD mouse model, while short term fasting is sufficient to observe similar effects in peripheral tissue. Furthermore, preventing proteolysis at the caspase-6 cleavage site D586 (C6R) makes mHTT a better substrate for autophagy, additionally increasing its clearance. Mice expressing mutant C6R also exhibit increased autophagy at baseline compared to an HD model with cleavable mHTT, suggesting that the native function of HTT in promoting autophagy is disrupted upon cleavage and re-established by prevention of cleavage by caspase-6. In HD patients, mHTT clearance and autophagy may therefore become increasingly impaired as a function of age and disease stage by gradually increased activity of mHTT-processing enzymes.
- Published
- 2017
- Full Text
- View/download PDF
27. p53 increases caspase-6 expression and activation in muscle tissue expressing mutant huntingtin
- Author
-
Niels H. Skotte, Sabine Engemann, Safia Ladha, Dagmar E. Ehrnhoefer, Khuong Tuyen Huynh, Kristina Becanovic, Yen T. Nguyen, Signe Marie Borch Nielsen, Blair R. Leavitt, Michael R. Hayden, Yu Deng, Xiaofan Qiu, and Lis Hasholt
- Subjects
Male ,Muscle tissue ,Huntingtin ,Mice, Transgenic ,Nerve Tissue Proteins ,Caspase 6 ,Mice ,Genetics ,medicine ,Huntingtin Protein ,Animals ,Humans ,Benzothiazoles ,Muscle, Skeletal ,Molecular Biology ,Cells, Cultured ,Genetics (clinical) ,Caspase ,Neurons ,biology ,Skeletal muscle ,General Medicine ,Fibroblasts ,Lamin Type A ,Molecular biology ,Muscle atrophy ,Disease Models, Animal ,Huntington Disease ,medicine.anatomical_structure ,Mutation ,biology.protein ,Female ,Tumor Suppressor Protein p53 ,medicine.symptom ,Lamin ,Toluene - Abstract
Activation of caspase-6 in the striatum of both presymptomatic and affected persons with Huntington's disease (HD) is an early event in the disease pathogenesis. However, little is known about the role of caspase-6 outside the central nervous system (CNS) and whether caspase activation might play a role in the peripheral phenotypes, such as muscle wasting observed in HD. We assessed skeletal muscle tissue from HD patients and well-characterized mouse models of HD. Cleavage of the caspase-6 specific substrate lamin A is significantly increased in skeletal muscle obtained from HD patients as well as in muscle tissues from two different HD mouse models. p53, a transcriptional activator of caspase-6, is upregulated in neuronal cells and tissues expressing mutant huntingtin. Activation of p53 leads to a dramatic increase in levels of caspase-6 mRNA, caspase-6 activity and cleavage of lamin A. Using mouse embryonic fibroblasts (MEFs) from YAC128 mice, we show that this increase in caspase-6 activity can be mitigated by pifithrin-α (pifα), an inhibitor of p53 transcriptional activity, but not through the inhibition of p53's mitochondrial pro-apoptotic function. Remarkably, the p53-mediated increase in caspase-6 expression and activation is exacerbated in cells and tissues of both neuronal and peripheral origin expressing mutant huntingtin (Htt). These findings suggest that the presence of the mutant Htt protein enhances p53 activity and lowers the apoptotic threshold, which activates caspase-6. Furthermore, these results suggest that this pathway is activated both within and outside the CNS in HD and may contribute to both loss of CNS neurons and muscle atrophy.
- Published
- 2013
- Full Text
- View/download PDF
28. Caspase-6-Resistant Mutant Huntingtin Does not Rescue the Toxic Effects of Caspase-Cleavable Mutant Huntingtin in vivo
- Author
-
Yu Deng, Nagat Bissada, Mahmoud A. Pouladi, Dagmar E. Ehrnhoefer, Sonia Franciosi, Amber L. Southwell, Kuljeet Vaid, Michael R. Hayden, and Rona K. Graham
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Mutant ,Mice, Transgenic ,Nerve Tissue Proteins ,Caspase 6 ,Neuroprotection ,Pathogenesis ,Mice ,Cellular and Molecular Neuroscience ,Species Specificity ,Huntington's disease ,medicine ,Animals ,Caspase ,Huntingtin Protein ,biology ,Nuclear Proteins ,Polyglutamine tract ,medicine.disease ,Corpus Striatum ,Cell biology ,Enzyme Activation ,Disease Models, Animal ,Huntington Disease ,biology.protein ,Female ,Neurology (clinical) - Abstract
Background: The amelioration of behavioral and neuropathological deficits in mice expressing caspase-6-resistant (C6R) mutant huntingtin (mhtt), despite the presence of an expanded polyglutamine tract, highlights proteolysis of htt at the 586aa caspase-6 (casp6) site may be an important mechanism in the pathogenesis of Huntington disease (HD). One possible explanation of these effects is that C6R mhtt could act as a dominant negative on mhtt. Objective and Methods: To determine if the neuroprotective effect observed in the C6R mice is due to dominant negative effects, we crossed the C6R mice to the YAC128 HD mouse model to generate mice expressing both caspase-cleavable and C6R mhtt (YAC/C6R) concurrently and assessed previously defined behavioral and neuropathological endpoints. Results: Our results demonstrate that YAC/C6R animals exhibit similar motor abnormalities and learning deficits as the YAC128 mice. Neuropathological analysis reveals a significant decrease in brain weight and striatal volume in the YAC/C6R mice comparable to the YAC128 mice. In contrast, and similar to previous findings, C6R mice demonstrate preserved brain weight and striatal volume. As expected, body weight is significantly increased in the YAC/C6R mice due to the increased levels of htt. Conclusions: The results of this study suggest that the lack of an HD phenotype in the C6R mice is most likely due to the absence of cleavage of htt and not due to suppression of expression of mhtt.
- Published
- 2012
- Full Text
- View/download PDF
29. Caspase-6 and neurodegeneration
- Author
-
Dagmar E. Ehrnhoefer, Michael R. Hayden, and Rona K. Graham
- Subjects
Models, Molecular ,Proteases ,Caspase 6 ,biology ,Protein Conformation ,General Neuroscience ,Neurodegeneration ,medicine.disease ,Caspase Inhibitors ,Cell biology ,Enzyme Activation ,Disease Models, Animal ,Enzyme activator ,Huntington Disease ,Alzheimer Disease ,Apoptosis ,Nerve Degeneration ,medicine ,biology.protein ,Animals ,Humans ,Nuclear lamina ,Alzheimer's disease ,Caspase - Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
30. Full-length huntingtin levels modulate body weight by influencing insulin-like growth factor 1 expression
- Author
-
Yuanyun Xie, Paolo Paganetti, Nagat Bissada, Dagmar E. Ehrnhoefer, Michael R. Hayden, Mahmoud A. Pouladi, Robert M. Friedlander, Rona K. Graham, X. William Yang, Niels H. Skotte, Jeong Eun Kim, and Blair R. Leavitt
- Subjects
Male ,Genetically modified mouse ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Huntingtin ,animal diseases ,medicine.medical_treatment ,Transgene ,Central nervous system ,Gene Expression ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Mice ,Insulin-like growth factor ,Internal medicine ,mental disorders ,Gene expression ,Genetics ,medicine ,Huntingtin Protein ,Animals ,Humans ,Insulin-Like Growth Factor I ,Molecular Biology ,Genetics (clinical) ,Body Weight ,Brain ,Nuclear Proteins ,Articles ,General Medicine ,nervous system diseases ,Disease Models, Animal ,Huntington Disease ,Endocrinology ,medicine.anatomical_structure ,nervous system ,Signal transduction ,Signal Transduction - Abstract
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.
- Published
- 2010
- Full Text
- View/download PDF
31. Balance between synaptic versus extrasynaptic NMDA receptor activity influences inclusions and neurotoxicity of mutant huntingtin
- Author
-
Dongdong Yao, Dagmar E. Ehrnhoefer, Stuart A. Lipton, Marcus Kaul, Arjay Clemente, Peng Xia, Mahmoud A. Pouladi, H-S Vincent Chen, Michael R. Hayden, Rona K. Graham, Dongxian Zhang, Rameez Zaidi, Gary Tong, Maria Talantova, and Shu-ichi Okamoto
- Subjects
Huntington's Disease ,Patch-Clamp Techniques ,Huntingtin ,Neurodegenerative ,Medical and Health Sciences ,Transgenic ,Mice ,0302 clinical medicine ,Receptors ,Nuclear protein ,Neurons ,Huntingtin Protein ,0303 health sciences ,Cell Death ,Memantine ,Nuclear Proteins ,General Medicine ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Biochemistry ,Neurological ,Artificial ,NMDA receptor ,N-Methyl-D-Aspartate ,medicine.drug ,Programmed cell death ,Immunology ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Receptors, N-Methyl-D-Aspartate ,Neuroprotection ,Chromosomes ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Rare Diseases ,mental disorders ,medicine ,Animals ,Chromosomes, Artificial, Yeast ,030304 developmental biology ,Neurosciences ,Neurotoxicity ,medicine.disease ,Yeast ,Brain Disorders ,nervous system ,Mutation ,Synapses ,Trans-Activators ,Neuroscience ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Huntington's 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-gamma coactivator-1alpha (PGC-1alpha) 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 Huntington's disease.
- Published
- 2009
- Full Text
- View/download PDF
32. Mouse models of Huntington disease: variations on a theme
- Author
-
Dagmar E. Ehrnhoefer, Michael R. Hayden, Mahmoud A. Pouladi, and Stefanie L. Butland
- Subjects
Huntingtin ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,Nerve Tissue Proteins ,Context (language use) ,Disease ,Biology ,medicine.disease_cause ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Immunology and Microbiology (miscellaneous) ,microRNA ,medicine ,Huntingtin Protein ,Animals ,Humans ,Gene ,Genetics ,Mutation ,Binding Sites ,Nuclear Proteins ,Neurodegenerative Diseases ,Phenotype ,Disease Models, Animal ,MicroRNAs ,Huntington Disease ,Commentary ,Transcription Factors - Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
33. Laquinimod decreases Bax expression and reduces caspase-6 activation in neurons
- Author
-
Michael R. Hayden, Yu Deng, Xiaofan Qiu, Michelle Tsang, Nicholas S. Caron, and Dagmar E. Ehrnhoefer
- Subjects
0301 basic medicine ,Time Factors ,Mice, Transgenic ,Caspase 6 ,Biology ,Quinolones ,Neuroprotection ,Pathogenesis ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Developmental Neuroscience ,Downregulation and upregulation ,medicine ,Animals ,Humans ,bcl-2-Associated X Protein ,chemistry.chemical_classification ,Cerebral Cortex ,Neurons ,Protein Synthesis Inhibitors ,Huntingtin Protein ,Microglia ,Dose-Response Relationship, Drug ,Multiple sclerosis ,Tosylphenylalanyl Chloromethyl Ketone ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Enzyme ,Neurology ,chemistry ,Gene Expression Regulation ,COS Cells ,Mutation ,Camptothecin ,Down Syndrome ,Neuroscience ,Laquinimod ,030217 neurology & neurosurgery - Abstract
Laquinimod is an immunomodulatory compound that has shown neuroprotective benefits in clinical trials for multiple sclerosis. Laquinimod ameliorates both white and gray matter damage in human patients, and prevents axonal degeneration in animal models of multiple sclerosis. Axonal damage and white matter loss are a common feature shared between different neurodegenerative diseases. Caspase-6 activation plays an important role in axonal degeneration on the molecular level. Increased activity of caspase-6 has been demonstrated in brain tissue from presymptomatic Huntington disease mutation carriers, and it is an early marker of axonal dysfunction. Since laquinimod is currently undergoing a clinical trial in Huntington disease (LEGATO-HD, clinicaltrials.gov ID: NCT02215616), we set out to evaluate its impact on neuronal caspase-6 activation. We find that laquinimod ameliorates DNA-damage induced activation of caspase-6 in primary neuronal cultures. This is an indirect effect that is not mediated by direct inhibition of the enzyme. The investigation of potential caspase-6 activating mechanisms revealed that laquinimod reduces the expression of Bax, a pro-apoptotic molecule that causes mitochondrial cytochrome c release and caspase activation. Bax expression is furthermore increased in striatal tissues from the YAC128 mouse model of HD in an age-dependent manner. Our results demonstrate that laquinimod can directly downregulate neuronal apoptosis pathways relevant for axonal degeneration in addition to its known effects on astrocytes and microglia in the CNS. It targets a pathway that is relevant for the pathogenesis of HD, supporting the hypothesis that laquinimod may provide clinical benefit.
- Published
- 2016
34. Interactome network analysis identifies multiple caspase-6 interactors involved in the pathogenesis of HD
- Author
-
Dagmar E. Ehrnhoefer, Jean Lainé, Sean-Patrick Riechers, Erich E. Wanker, Michael R. Hayden, Mélissa Laroche, Yu Deng, Rona K. Graham, Stefanie L. Butland, Jenny Russ, Niels H. Skotte, and Mahmoud A. Pouladi
- Subjects
0301 basic medicine ,Huntingtin ,Mutant ,Computational biology ,Caspase 6 ,Protein Serine-Threonine Kinases ,Interactome ,Models, Biological ,Serine-Threonine Kinase 3 ,Cell Line ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Two-Hybrid System Techniques ,Genetics ,medicine ,Humans ,Protein Interaction Maps ,Molecular Biology ,Genetics (clinical) ,Caspase ,Huntingtin Protein ,Binding Sites ,biology ,Kinase ,General Medicine ,medicine.disease ,030104 developmental biology ,Huntington Disease ,Gene Expression Regulation ,biology.protein ,Alzheimer's disease ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery - Abstract
Caspase-6 (CASP6) has emerged as an important player in Huntington disease (HD), Alzheimer disease (AD) and cerebral ischemia, where it is activated early in the disease process. CASP6 also plays a key role in axonal degeneration, further underscoring the importance of this protease in neurodegenerative pathways. As a protein's function is modulated by its protein-protein interactions, we performed a high-throughput yeast-2-hybrid (Y2H) screen against ∼17,000 human proteins to gain further insight into the function of CASP6. We identified a high-confidence list of 87 potential CASP6 interactors. From this list, 61% are predicted to contain a CASP6 recognition site. Of nine candidate substrates assessed, six are cleaved by CASP6. Proteins that did not contain a predicted CASP6 recognition site were assessed using a LUMIER assay approach, and 51% were further validated as interactors by this method. Of note, 54% of the high-confidence interactors identified show alterations in human HD brain at the mRNA level, and there is a significant enrichment for previously validated huntingtin (HTT) interactors. One protein of interest, STK3, a pro-apoptotic kinase, was validated biochemically to be a CASP6 substrate. Furthermore, our results demonstrate that in striatal cells expressing mutant huntingtin (mHTT), an increase in full length and fragment levels of STK3 are observed. We further show that caspase-3 is not essential for the endogenous cleavage of STK3. Characterization of the interaction network provides important new information regarding key pathways of interactors of CASP6 and highlights potential novel therapeutic targets for HD, AD and cerebral ischemia.
- Published
- 2015
35. Autophagy in Huntington disease and huntingtin in autophagy
- Author
-
Dagmar E. Ehrnhoefer, Dale D.O. Martin, Michael R. Hayden, and Safia Ladha
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin Protein ,Huntingtin ,biology ,General Neuroscience ,Neurodegeneration ,Autophagy ,Wild type ,Nerve Tissue Proteins ,Protein aggregation ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,Huntington Disease ,Lysosome ,mental disorders ,Nerve Degeneration ,medicine ,biology.protein ,Animals ,Humans ,Caspase ,Myristoylation - Abstract
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.
- Published
- 2014
36. A new function of TFIIH explains the neurological symptoms in trichothiodystrophy
- Author
-
Dagmar E. Ehrnhoefer
- Subjects
musculoskeletal diseases ,animal structures ,business.industry ,musculoskeletal, neural, and ocular physiology ,fungi ,Trichothiodystrophy ,medicine.disease ,nervous system ,Coactivator ,Genetics ,Transcription factor II H ,medicine ,business ,Neuroscience ,Genetics (clinical) ,Function (biology) - Abstract
Neurological defects in trichothiodystrophy reveal a coactivator function of TFIIH Compe et al. (2007) Nature Neuroscience 10: 1414–1422
- Published
- 2008
- Full Text
- View/download PDF
37. HACE1 reduces oxidative stress and mutant Huntingtin toxicity by promoting the NRF2 response
- Author
-
Jason D. Galpin, Gabriel Leprivier, Dagmar E. Ehrnhoefer, Syam Prakash Somasekharan, Christopher A. Ahern, Liheng Li, Joan Mathers, Roberto Nitsch, Christopher J. Carnie, Sonia Franciosi, Barak Rotblat, Martina Metzler, Amber L. Southwell, Mads Daugaard, Tiffany Tang, Brian Kwok, Yu Deng, Gerry Melino, Niels H. Skotte, Dieter Fink, Adi Barokas, Naniye Malli Cetinbas, Josef M. Penninger, Michael R. Hayden, and Poul H. Sorensen
- Subjects
Huntingtin ,NF-E2-Related Factor 2 ,Ubiquitin-Protein Ligases ,Blotting, Western ,Fluorescent Antibody Technique ,Nerve Tissue Proteins ,Biology ,medicine.disease_cause ,Cell Fractionation ,Real-Time Polymerase Chain Reaction ,chemistry.chemical_compound ,Mice ,ROS ,aging ,glutathione ,transcription factor ,Animals ,Corpus Striatum ,DNA Primers ,HEK293 Cells ,Humans ,Huntington Disease ,Oxidative Stress ,Reactive Oxygen Species ,medicine ,Settore BIO/10 ,Transcription factor ,chemistry.chemical_classification ,Reactive oxygen species ,Huntingtin Protein ,Multidisciplinary ,Blotting ,Neurodegeneration ,Glutathione ,Biological Sciences ,medicine.disease ,Molecular biology ,Cell biology ,chemistry ,Ectopic expression ,Carcinogenesis ,Western ,Oxidative stress - Abstract
Oxidative stress plays a key role in late onset diseases including cancer and neurodegenerative diseases such as Huntington disease. Therefore, uncovering regulators of the antioxidant stress responses is important for understanding the course of these diseases. Indeed, the nuclear factor erythroid 2-related factor 2 (NRF2), a master regulator of the cellular antioxidative stress response, is deregulated in both cancer and neurodegeneration. Similar to NRF2, the tumor suppressor Homologous to the E6-AP Carboxyl Terminus (HECT) domain and Ankyrin repeat containing E3 ubiquitin-protein ligase 1 (HACE1) plays a protective role against stress-induced tumorigenesis in mice, but its roles in the antioxidative stress response or its involvement in neurodegeneration have not been investigated. To this end we examined Hace1 WT and KO mice and found that Hace1 KO animals exhibited increased oxidative stress in brain and that the antioxidative stress response was impaired. Moreover, HACE1 was found to be essential for optimal NRF2 activation in cells challenged with oxidative stress, as HACE1 depletion resulted in reduced NRF2 activity, stability, and protein synthesis, leading to lower tolerance against oxidative stress triggers. Strikingly, we found a reduction of HACE1 levels in the striatum of Huntington disease patients, implicating HACE1 in the pathology of Huntington disease. Moreover, ectopic expression of HACE1 in striatal neuronal progenitor cells provided protection against mutant Huntingtin-induced redox imbalance and hypersensitivity to oxidative stress, by augmenting NRF2 functions. These findings reveal that the tumor suppressor HACE1 plays a role in the NRF2 antioxidative stress response pathway and in neurodegeneration.
- Published
- 2014
38. NP03, a novel low-dose lithium formulation, is neuroprotective in the YAC128 mouse model of Huntington disease
- Author
-
Dagmar E. Ehrnhoefer, Yuanyun Xie, Elsa Brillaud, Elena Cattaneo, Weining Zhang, Chiara Zuccato, Patrick Poucheret, Michael R. Hayden, Elsa Compte, Rona K. Graham, Sonia Franciosi, Christian Neri, Jean-Claude Maurel, Mahmoud A. Pouladi, Paola Conforti, Biologie et Pathologie du Neurone (Brain-C), Adaptation Biologique et Vieillissement = Biological Adaptation and Ageing (B2A), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Biologie Paris Seine (IBPS), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Biologie Paris Seine (IBPS), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Medesis Pharma, Canadian Institute of Health Research, Michael Smith Foundation for Health Research (MSFHR), CIHR, Huntington Society of Canada, Huntington's Disease Society of America, CHDI Foundation, ANR, AP-HP, Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Biologie Paris Seine (IBPS), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Male ,Lithium (medication) ,Immunoblotting ,Disease ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Motor Activity ,Pharmacology ,Lithium ,Neuroprotection ,Transgenic mouse model ,lcsh:RC321-571 ,Treatment of bipolar disorder ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,GSK-3 ,medicine ,Animals ,Humans ,Rats, Wistar ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,030304 developmental biology ,0303 health sciences ,Testicular atrophy ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Brain ,Huntington disease ,medicine.disease ,Rats ,3. Good health ,Disease Models, Animal ,Neuroprotective Agents ,BDNF ,Neurology ,Toxicity ,Female ,Tau ,business ,Neuroscience ,030217 neurology & neurosurgery ,Caspase-6 ,medicine.drug - Abstract
International audience; Huntington disease (HD), a neurodegenerative disorder caused by an expanded CAG repeat in the HTT gene, remains without a treatment to modify the course of the illness. Lithium, a drug widely used for the treatment of bipolar disorder, has been shown to exert neuroprotective effects in a number of models of neurological disease but may have various toxic effects at conventional therapeutic doses. We examined whether NP03, a novel low-dose lithium microemulsion, would improve the disease phenotypes in the YAC128 mouse model of HD. We demonstrate that NP03 improves motor function, ameliorates the neuropathological deficits in striatal volume, neuronal counts, and DARPP-32 expression, and partially rescues testicular atrophy in YAC128 mice. These positive effects were accompanied by improvements in multiple biochemical endpoints associated with the pathogenesis of HD, including normalization of caspase-6-activation and amelioration of deficits in BDNF levels, and with no lithium-related toxicity. Our findings demonstrate that NP03 ameliorates the motor and neuropathological phenotypes in the YAC128 mouse model of HD, and represents a potential therapeutic approach for HD.
- Published
- 2012
- Full Text
- View/download PDF
39. Intrinsic cleavage of receptor-interacting protein kinase-1 by caspase-6
- Author
-
B J van Raam, Guy S. Salvesen, Dagmar E. Ehrnhoefer, and Michael R. Hayden
- Subjects
Programmed cell death ,Necroptosis ,Fas-Associated Death Domain Protein ,Apoptosis ,Caspase 6 ,Caspase 8 ,RIPK1 ,Jurkat Cells ,Mice ,Animals ,Humans ,Molecular Biology ,Caspase ,Mice, Knockout ,Original Paper ,biology ,NLRP1 ,Intrinsic apoptosis ,Cell Biology ,U937 Cells ,Cell biology ,HEK293 Cells ,Receptor-Interacting Protein Serine-Threonine Kinases ,biology.protein ,Signal Transduction - Abstract
Necroptosis is a form of programmed cell death that occurs in the absence of caspase activation and depends on the activity of the receptor-interacting protein kinases. Inactivation of these kinases by caspase-mediated cleavage has been shown to be essential for successful embryonic development, survival and activation of certain cell types. The initiator of extrinsic apoptosis, caspase-8, which has a pro-death as well as a pro-life function, has been assigned this role. In the present study we demonstrate that caspase-6, an executioner caspase, performs this role during apoptosis induced through the intrinsic pathway. In addition, we demonstrate that in the absence of caspase activity, intrinsic triggers of apoptosis induce the receptor-interacting-kinase-1-dependent production of pro-inflammatory cytokines. We show that ubiquitously expressed caspase-6 has a supporting role in apoptosis by cleaving this kinase, thus preventing production of inflammatory cytokines as well as inhibiting the necroptotic pathway. These findings shed new light on the regulation of necroptosis as well as cell death in an inflammatory environment wherein cells receive both intrinsic and extrinsic death signals.
- Published
- 2012
40. Rescue from excitotoxicity and axonal degeneration accompanied by age-dependent behavioral and neuroanatomical alterations in caspase-6-deficient mice
- Author
-
Bibiana K. Y. Wong, Mahmoud A. Pouladi, Michael R. Hayden, Valeria Uribe, Rona K. Graham, Corey L. Cusack, Konstantin Feinberg, Nagat Bissada, Niels H. Skotte, Freda D. Miller, Mohanish Deshmukh, Kuljeet Vaid, Yu Deng, Weining Zhang, David Howland, Yuanyun Xie, Amanda Spreeuw, Yingbin Ouyang, Yimiao Ou, Sonia Franciosi, and Dagmar E. Ehrnhoefer
- Subjects
Programmed cell death ,Aging ,Excitotoxicity ,Cellular homeostasis ,Apoptosis ,Caspase 6 ,Motor Activity ,medicine.disease_cause ,Receptors, N-Methyl-D-Aspartate ,Mice ,Alzheimer Disease ,Genetics ,medicine ,Animals ,Humans ,RNA, Messenger ,Molecular Biology ,Genetics (clinical) ,Caspase ,Mice, Knockout ,Neurons ,biology ,Base Sequence ,Behavior, Animal ,Brain ,General Medicine ,Anatomy ,Articles ,medicine.disease ,Nerve growth factor ,Huntington Disease ,Nerve Degeneration ,biology.protein ,Alzheimer's disease ,Neuroscience - Abstract
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.
- Published
- 2012
41. Convergent pathogenic pathways in Alzheimer's and Huntington's diseases: shared targets for drug development
- Author
-
Bibiana K. Y. Wong, Dagmar E. Ehrnhoefer, and Michael R. Hayden
- Subjects
Pharmacology ,Disease mutation ,TOR Serine-Threonine Kinases ,Disease mechanisms ,Brain ,General Medicine ,Disease ,Biology ,Bioinformatics ,Article ,Drug Delivery Systems ,Huntington Disease ,Drug development ,Alzheimer Disease ,Drug Discovery ,Neural Pathways ,Animals ,Humans ,Protein Kinase Inhibitors - Abstract
Neurodegenerative diseases, exemplified by Alzheimer's disease and Huntington's 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 Huntington's disease has a monogenic cause, it is possible to accurately identify individuals who carry the Huntington's disease mutation but do not yet manifest symptoms. These individuals could act as a model for Alzheimer's disease to test therapeutic interventions that target shared pathogenic pathways.
- Published
- 2011
42. Mice lacking caspase-2 are protected from behavioral changes, but not pathology, in the YAC128 model of Huntington disease
- Author
-
Amber L. Southwell, Dagmar E. Ehrnhoefer, Weining Zhang, Jason P. Lerch, Michael R. Hayden, Jeffrey B. Carroll, Rona K. Graham, Nagat Bissada, R. Mark Henkelman, Yu Deng, and Li-Ping Cao
- Subjects
Huntington's Disease ,Pathology ,medicine.medical_specialty ,caspase ,Caspase 2 ,Clinical Neurology ,Disease ,lcsh:Geriatrics ,lcsh:RC346-429 ,Pathogenesis ,Cellular and Molecular Neuroscience ,Huntington's disease ,medicine ,Huntingtin Protein ,magnetic resonance imaging ,Molecular Biology ,Caspase ,lcsh:Neurology. Diseases of the nervous system ,biology ,business.industry ,Neurodegeneration ,neurodegeneration ,medicine.disease ,Molecular medicine ,lcsh:RC952-954.6 ,biology.protein ,Neurology (clinical) ,business ,Research Article - Abstract
Background Huntington Disease (HD) is a neurodegenerative disorder in which caspase activation and cleavage of substrates, including the huntingtin protein, has been invoked as a pathological mechanism. Specific changes in caspase-2 (casp2) activity have been suggested to contribute to the pathogenesis of HD, however unique casp2 cleavage substrates have remained elusive. We thus utilized mice completely lacking casp2 (casp2-/-) to examine the role played by casp2 in the progression of HD. This 'substrate agnostic' approach allows us to query the effect of casp2 on HD progression without pre-defining proteolytic substrates of interest. Results YAC128 HD model mice lacking casp2 show protection from well-validated motor and cognitive features of HD, including performance on rotarod, swimming T-maze, pre-pulse inhibition, spontaneous alternation and locomotor tasks. However, the specific pathological features of the YAC128 mice including striatal volume loss and testicular degeneration are unaltered in mice lacking casp2. The application of high-resolution magnetic resonance imaging (MRI) techniques validates specific neuropathology in the YAC128 mice that is not altered by ablation of casp2. Conclusions The rescue of behavioral phenotypes in the absence of pathological improvement suggests that different pathways may be operative in the dysfunction of neural circuitry in HD leading to behavioral changes compared to the processes leading to cell death and volume loss. Inhibition of caspase-2 activity may be associated with symptomatic improvement in HD.
- Published
- 2011
43. Small changes, big impact: Posttranslational modifications and function of huntingtin in Huntington disease
- Author
-
Liza M. Sutton, Michael R. Hayden, and Dagmar E. Ehrnhoefer
- Subjects
Huntingtin ,Lipoylation ,SUMO protein ,Nerve Tissue Proteins ,Biology ,Article ,Palmitoylation ,Ubiquitin ,Huntingtin Protein ,medicine ,Animals ,Humans ,Phosphorylation ,Genetics ,General Neuroscience ,Neurodegeneration ,Ubiquitination ,Brain ,Nuclear Proteins ,Sumoylation ,Acetylation ,Polyglutamine tract ,medicine.disease ,Cell biology ,Huntington Disease ,biology.protein ,Neurology (clinical) ,Protein Processing, Post-Translational - Abstract
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.
- Published
- 2011
44. A quantitative method for the specific assessment of caspase-6 activity in cell culture
- Author
-
Safia Ladha, Yen T. Nguyen, Dagmar E. Ehrnhoefer, Li-Ping Cao, Niels H. Skotte, Edie Dullaghan, Michael R. Hayden, and Jane Savill
- Subjects
Cell Extracts ,Luminescence ,medicine.medical_treatment ,Cell ,Cell Culture Techniques ,Fluorescent Antibody Technique ,lcsh:Medicine ,Substrate Specificity ,Mice ,0302 clinical medicine ,Molecular Cell Biology ,Neurobiology of Disease and Regeneration ,Chlorocebus aethiops ,lcsh:Science ,Peptide sequence ,Cellular Stress Responses ,Neurons ,0303 health sciences ,Multidisciplinary ,Cell Death ,Caspase 6 ,Neurodegenerative Diseases ,Lamins ,3. Good health ,Huntington Disease ,medicine.anatomical_structure ,Neurology ,Biochemistry ,COS Cells ,Medicine ,Intracellular ,Research Article ,Proteases ,Immunoprecipitation ,Immunology ,Molecular Sequence Data ,Enzyme-Linked Immunosorbent Assay ,Biology ,Cercopithecus aethiops ,03 medical and health sciences ,Alzheimer Disease ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Immunoassays ,Enzyme Assays ,030304 developmental biology ,Protease ,lcsh:R ,Kinetics ,Cell culture ,Immunologic Techniques ,Dementia ,lcsh:Q ,Peptides ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Aberrant activation of caspase-6 has recently emerged as a major contributor to the pathogeneses of neurodegenerative disorders such as Alzheimer's and Huntington disease. Commercially available assays to measure caspase-6 activity commonly use the VEID peptide as a substrate. However these methods are not well suited to specifically assess caspase-6 activity in the presence of other, confounding protease activities, as often encountered in cell and tissue samples. Here we report the development of a method that overcomes this limitation by using a protein substrate, lamin A, which is highly specific for caspase-6 cleavage at amino acid 230. Using a neo-epitope antibody against cleaved lamin A, we developed an electrochemiluminescence-based ELISA assay that is suitable to specifically detect and quantify caspase-6 activity in highly apoptotic cell extracts. The method is more sensitive than VEID-based assays and can be adapted to a high-content imaging platform for high-throughput screening. This method should be useful to screen for and characterize caspase-6 inhibitor compounds and other interventions to decrease intracellular caspase-6 activity for applications in neurodegenerative disorders.
- Published
- 2011
- Full Text
- View/download PDF
45. M08 Laquinimod Reduces Neuronal Caspase-6 Activation And Axonal Degeneration In Vitro
- Author
-
Xiaofan Qiu, Dagmar E. Ehrnhoefer, M Tsang, S Ladha, and Hayden
- Subjects
Multiple sclerosis ,Inflammation ,Caspase 6 ,Biology ,medicine.disease ,Neuroprotection ,Psychiatry and Mental health ,chemistry.chemical_compound ,nervous system ,chemistry ,Mechanism of action ,Downregulation and upregulation ,In vivo ,medicine ,Surgery ,Neurology (clinical) ,medicine.symptom ,Laquinimod ,Neuroscience - Abstract
Background Laquinimod is an immunomodulatory compound that reduces relapse rate, brain atrophy and disability progression in multiple sclerosis. It has well-documented effects on inflammation, is widely distributed in the CNS and has been shown to ameliorate axonal damage in vitro and in vivo through an unknown mechanism. We have shown recently that caspase-6 is an important mediator in axonal degeneration, since sympathetic neurons derived from caspase-6 -/- mice do not degenerate when cultured in the absence of neuronal growth factor (NGF). We therefore investigated whether the beneficial effect of Laquinimod on axonal damage is mediated by caspase-6. Aim To investigate neuron-specific beneficial effects of Laquinimod. Methods Cell-free and cell-based enzymatic activity assays were used to determine whether Laquinimod directly inhibits caspase-6. To investigate an effect on intraneuronal caspase-6 activity, primary cortical neurons were treated with camptothecin in the presence or absence of Laquinimod, and cleavage of the caspase-6 specific substrate lamin A was quantified by ELISA. Axonal degeneration of primary sympathetic neurons from the superior cerebral ganglion was induced by NGF withdrawal in the presence or absence of Laquinimod. Results Laquinimod did not directly inhibit caspase-6 activity in cell-free or transfection-based cellular systems. However, the presence of Laquinimod in camptothecin-stressed primary cortical neuron cultures led to a significant decrease in caspase-6 activation. In cultures of primary superior cerebral ganglion neurons, Laquinimod partially protected axons from degeneration after NGF withdrawal, a process that is specifically dependent on caspase-6 activity, in agreement with the reduction of caspase-6 activity observed in stressed cortical neurons. Conclusions The beneficial effects of Laquinimod described so far involve neuroprotection through the downregulation of glial activation, whereas our findings represent a novel, purely neuronal mechanism of action for this drug. We propose that by preventing neuronal caspase-6 activation and axonal degeneration, Laquinimod might also provide benefits in other neurodegenerative disorders such as Huntington’s disease that are associated with excessive activation of caspase-6.
- Published
- 2014
- Full Text
- View/download PDF
46. EGCG remodels mature α-synuclein and amyloid-β fibrils and reduces cellular toxicity
- Author
-
Katja Neugebauer, Erich E. Wanker, Jan Bieschke, Jenny Russ, Dagmar E. Ehrnhoefer, Ralf P. Friedrich, and Heike J. Wobst
- Subjects
Conformational change ,Amyloid ,Amyloid β ,Blotting, Western ,CHO Cells ,Protein aggregation ,Fibril ,Amyloid Neuropathies ,Microscopy, Atomic Force ,PC12 Cells ,Catechin ,Chromatography, Affinity ,chemistry.chemical_compound ,Cricetulus ,Microscopy, Electron, Transmission ,Cricetinae ,Escherichia coli ,Animals ,Humans ,Multidisciplinary ,Amyloid beta-Peptides ,Chemistry ,Circular Dichroism ,food and beverages ,Fibrillogenesis ,Biological Sciences ,Rats ,Monomer ,Neuroprotective Agents ,Biochemistry ,Microscopy, Fluorescence ,Toxicity ,Biophysics ,alpha-Synuclein ,Protein folding - Abstract
Protein misfolding and formation of beta-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 alpha-synuclein and amyloid-beta 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 alpha-synuclein and amyloid-beta fibrils into smaller, amorphous protein aggregates that are nontoxic to mammalian cells. Mechanistic studies revealed that the compound directly binds to beta-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.
- Published
- 2010
47. IFRD1 modulates disease severity in cystic fibrosis through the regulation of neutrophil effector function
- Author
-
Dagmar E. Ehrnhoefer
- Subjects
Effector ,business.industry ,respiratory system ,medicine.disease ,Cystic fibrosis ,Article ,IFRD1 ,Disease severity ,Lung disease ,Immunology ,Genetics ,medicine ,business ,Gene ,Genetics (clinical) ,Function (biology) - Abstract
Lung disease is the major cause of morbidity and mortality in cystic fibrosis (CF), an autosomal recessive disease caused by mutations in CFTR. In CF, chronic infection and dysregulated neutrophilic inflammation lead to progressive airway destruction. The severity of CF lung disease has significant heritability, independent of CFTR genotype1. To identify genetic modifiers, we performed a genome-wide single nucleotide polymorphism (SNP) scan in one cohort of CF patients, replicating top candidates in an independent cohort. This approach identified IFRD1 as a modifier of CF lung disease severity. IFRD1 is a histone deacetylase (HDAC)-dependent transcriptional co-regulator expressed during terminal neutrophil differentiation. Neutrophils, but not macrophages, from Ifrd1-deficient mice exhibited blunted effector function, associated with decreased NF-κB p65 transactivation. In vivo, IFRD1 deficiency caused delayed bacterial clearance from the airway, but also less inflammation and disease—a phenotype primarily dependent on hematopoietic cell expression, or lack of expression, of IFRD1. In humans, IFRD1 polymorphisms were significantly associated with variation in neutrophil effector function. These data suggest that IFRD1 modulates the pathogenesis of CF lung disease through regulation of neutrophil effector function.
- Published
- 2009
48. Green tea (-)-epigallocatechin-gallate modulates early events in huntingtin misfolding and reduces toxicity in Huntington's disease models
- Author
-
Paul J. Muchowski, Phoebe Markovic, Susan Lindquist, Margaret Roark, Jennifer L. Wacker, J. Lawrence Marsh, Sabine Engemann, Justin Legleiter, Leslie M. Thompson, Erich E. Wanker, Dagmar E. Ehrnhoefer, and Martin L. Duennwald
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Protein Folding ,Huntingtin ,Protein Conformation ,Transgene ,Recombinant Fusion Proteins ,Nerve Tissue Proteins ,Saccharomyces cerevisiae ,Biology ,Protein aggregation ,Epigallocatechin gallate ,In Vitro Techniques ,medicine.disease_cause ,Microscopy, Atomic Force ,Models, Biological ,Camellia sinensis ,Catechin ,Animals, Genetically Modified ,Exon ,chemistry.chemical_compound ,Huntington's disease ,mental disorders ,Genetics ,Huntingtin Protein ,medicine ,Animals ,Humans ,Protein Structure, Quaternary ,Molecular Biology ,Genetics (clinical) ,Motor Neurons ,Mutation ,food and beverages ,Nuclear Proteins ,General Medicine ,Exons ,medicine.disease ,Cell biology ,Drosophila melanogaster ,Huntington Disease ,chemistry ,Multiprotein Complexes ,Nerve Degeneration ,Photoreceptor Cells, Invertebrate ,Phytotherapy - Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder for which only symptomatic treatments of limited effectiveness are available. Preventing early misfolding steps and thereby aggregation of the polyglutamine (polyQ)-containing protein huntingtin (htt) in neurons of patients may represent an attractive therapeutic strategy to postpone the onset and progression of HD. Here, we demonstrate that the green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) potently inhibits the aggregation of mutant htt exon 1 protein in a dose-dependent manner. Dot-blot assays and atomic force microscopy studies revealed that EGCG modulates misfolding and oligomerization of mutant htt exon 1 protein in vitro, indicating that it interferes with very early events in the aggregation process. Also, EGCG significantly reduced polyQ-mediated htt protein aggregation and cytotoxicity in an yeast model of HD. When EGCG was fed to transgenic HD flies overexpressing a pathogenic htt exon 1 protein, photoreceptor degeneration and motor function improved. These results indicate that modulators of htt exon 1 misfolding and oligomerization like EGCG are likely to reduce polyQ-mediated toxicity in vivo. Our studies may provide the basis for the development of a novel pharmacotherapy for HD and related polyQ disorders.
- Published
- 2006
49. Genetic modifiers of ataxin-3-mediated neurodegeneration: approaching protein misfolding from different angles
- Author
-
Dagmar E. Ehrnhoefer
- Subjects
Genetics ,Ataxin ,Neurodegeneration ,medicine ,Protein folding ,Biology ,Drosophila (subgenus) ,biology.organism_classification ,medicine.disease ,Genetics (clinical) - Abstract
Genome-wide screen for modifiers of ataxin-3 neurodegeneration in Drosophila Bilen et al. (2007) PLoS Genetics 3: 1950–1963
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.