90 results on '"Petrucelli, Leonard"'
Search Results
2. p53 is a central regulator driving neurodegeneration caused by C9orf72 poly(PR)
- Author
-
Maor-Nof, Maya, Shipony, Zohar, Lopez-Gonzalez, Rodrigo, Nakayama, Lisa, Zhang, Yong-Jie, Couthouis, Julien, Blum, Jacob A, Castruita, Patricia A, Linares, Gabriel R, Ruan, Kai, Ramaswami, Gokul, Simon, David J, Nof, Aviv, Santana, Manuel, Han, Kyuho, Sinnott-Armstrong, Nasa, Bassik, Michael C, Geschwind, Daniel H, Tessier-Lavigne, Marc, Attardi, Laura D, Lloyd, Thomas E, Ichida, Justin K, Gao, Fen-Biao, Greenleaf, William J, Yokoyama, Jennifer S, Petrucelli, Leonard, and Gitler, Aaron D
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research ,Brain Disorders ,ALS ,Genetics ,Frontotemporal Dementia (FTD) ,Neurodegenerative ,Acquired Cognitive Impairment ,Rare Diseases ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Dementia ,Stem Cell Research - Induced Pluripotent Stem Cell ,Neurosciences ,1.1 Normal biological development and functioning ,Underpinning research ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Animals ,Apoptosis Regulatory Proteins ,Axons ,C9orf72 Protein ,Cell Death ,Cells ,Cultured ,Cerebral Cortex ,Chromatin ,DNA Damage ,DNA Repeat Expansion ,Disease Models ,Animal ,Drosophila ,Mice ,Inbred C57BL ,Nerve Degeneration ,Protein Stability ,Transcription ,Genetic ,Tumor Suppressor Protein p53 ,Tumor Suppressor Proteins ,Mice ,ATAC-seq ,C9orf72 ,TDP-43 ,amyotrophic lateral sclerosis ,axonal degeneration ,neurodegeneration ,p53 ,puma ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is a GGGGCC repeat expansion in the C9orf72 gene. We developed a platform to interrogate the chromatin accessibility landscape and transcriptional program within neurons during degeneration. We provide evidence that neurons expressing the dipeptide repeat protein poly(proline-arginine), translated from the C9orf72 repeat expansion, activate a highly specific transcriptional program, exemplified by a single transcription factor, p53. Ablating p53 in mice completely rescued neurons from degeneration and markedly increased survival in a C9orf72 mouse model. p53 reduction also rescued axonal degeneration caused by poly(glycine-arginine), increased survival of C9orf72 ALS/FTD-patient-induced pluripotent stem cell (iPSC)-derived motor neurons, and mitigated neurodegeneration in a C9orf72 fly model. We show that p53 activates a downstream transcriptional program, including Puma, which drives neurodegeneration. These data demonstrate a neurodegenerative mechanism dynamically regulated through transcription-factor-binding events and provide a framework to apply chromatin accessibility and transcription program profiles to neurodegeneration.
- Published
- 2021
3. ADAR2 mislocalization and widespread RNA editing aberrations in C9orf72-mediated ALS/FTD
- Author
-
Moore, Stephen, Alsop, Eric, Lorenzini, Ileana, Starr, Alexander, Rabichow, Benjamin E., Mendez, Emily, Levy, Jennifer L., Burciu, Camelia, Reiman, Rebecca, Chew, Jeannie, Belzil, Veronique V., W. Dickson, Dennis, Robertson, Janice, Staats, Kim A., Ichida, Justin K., Petrucelli, Leonard, Van Keuren-Jensen, Kendall, and Sattler, Rita
- Published
- 2019
- Full Text
- View/download PDF
4. TIA1 regulates the generation and response to toxic tau oligomers
- Author
-
Jiang, Lulu, Ash, Peter E. A., Maziuk, Brandon F., Ballance, Heather I., Boudeau, Samantha, Abdullatif, Ali Al, Orlando, Marcello, Petrucelli, Leonard, Ikezu, Tsuneya, and Wolozin, Benjamin
- Published
- 2019
- Full Text
- View/download PDF
5. eIF4B and eIF4H mediate GR production from expanded G4C2 in a Drosophila model for C9orf72-associated ALS
- Author
-
Goodman, Lindsey D., Prudencio, Mercedes, Srinivasan, Ananth R., Rifai, Olivia M., Lee, Virginia M.-Y., Petrucelli, Leonard, and Bonini, Nancy M.
- Published
- 2019
- Full Text
- View/download PDF
6. Aberrant deposition of stress granule-resident proteins linked to C9orf72-associated TDP-43 proteinopathy
- Author
-
Chew, Jeannie, Cook, Casey, Gendron, Tania F., Jansen-West, Karen, del Rosso, Giulia, Daughrity, Lillian M., Castanedes-Casey, Monica, Kurti, Aishe, Stankowski, Jeannette N., Disney, Matthew D., Rothstein, Jeffrey D., Dickson, Dennis W., Fryer, John D., Zhang, Yong-Jie, and Petrucelli, Leonard
- Published
- 2019
- Full Text
- View/download PDF
7. Spinal poly-GA inclusions in a C9orf72 mouse model trigger motor deficits and inflammation without neuron loss
- Author
-
Schludi, Martin H., Becker, Lore, Garrett, Lillian, Gendron, Tania F., Zhou, Qihui, Schreiber, Franziska, Popper, Bastian, Dimou, Leda, Strom, Tim M., Winkelmann, Juliane, von Thaden, Anne, Rentzsch, Kristin, May, Stephanie, Michaelsen, Meike, Schwenk, Benjamin M., Tan, Jing, Schoser, Benedikt, Dieterich, Marianne, Petrucelli, Leonard, Hölter, Sabine M., Wurst, Wolfgang, Fuchs, Helmut, Gailus-Durner, Valerie, de Angelis, Martin Hrabe, Klopstock, Thomas, Arzberger, Thomas, and Edbauer, Dieter
- Published
- 2017
- Full Text
- View/download PDF
8. Poly-GR dipeptide repeat polymers correlate with neurodegeneration and Clinicopathological subtypes in C9ORF72-related brain disease
- Author
-
Sakae, Nobutaka, Bieniek, Kevin F., Zhang, Yong-Jie, Ross, Kelly, Gendron, Tania F., Murray, Melissa E., Rademakers, Rosa, Petrucelli, Leonard, and Dickson, Dennis W.
- Published
- 2018
- Full Text
- View/download PDF
9. TDP‐1, the Caenorhabditis elegans ortholog of TDP‐43, limits the accumulation of double‐stranded RNA
- Author
-
Saldi, Tassa K, Ash, Peter EA, Wilson, Gavin, Gonzales, Patrick, Garrido‐Lecca, Alfonso, Roberts, Christine M, Dostal, Vishantie, Gendron, Tania F, Stein, Lincoln D, Blumenthal, Thomas, Petrucelli, Leonard, and Link, Christopher D
- Published
- 2014
- Full Text
- View/download PDF
10. Modeling key pathological features of frontotemporal dementia with C9ORF72 repeat expansion in iPSC-derived human neurons
- Author
-
Almeida, Sandra, Gascon, Eduardo, Tran, Hélène, Chou, Hsin Jung, Gendron, Tania F., DeGroot, Steven, Tapper, Andrew R., Sellier, Chantal, Charlet-Berguerand, Nicolas, Karydas, Anna, Seeley, William W., Boxer, Adam L., Petrucelli, Leonard, Miller, Bruce L., and Gao, Fen-Biao
- Published
- 2013
- Full Text
- View/download PDF
11. Rodent Models of TDP-43 Proteinopathy: Investigating the Mechanisms of TDP-43-Mediated Neurodegeneration
- Author
-
Gendron, Tania F. and Petrucelli, Leonard
- Published
- 2011
- Full Text
- View/download PDF
12. Evidence of cerebellar TDP-43 loss of function in FTLD-TDP.
- Author
-
Pickles, Sarah, Gendron, Tania F., Koike, Yuka, Yue, Mei, Song, Yuping, Kachergus, Jennifer M., Shi, J., DeTure, Michael, Thompson, E. Aubrey, Oskarsson, Björn, Graff-Radford, Neill R., Boeve, Bradley F., Petersen, Ronald C., Wszolek, Zbigniew K., Josephs, Keith A., Dickson, Dennis W., Petrucelli, Leonard, Cook, Casey N., and Prudencio, Mercedes
- Subjects
FRONTOTEMPORAL lobar degeneration ,DEGENERATION (Pathology) ,NEURODEGENERATION ,CEREBELLUM ,CEREBELLAR cortex ,AGE of onset - Abstract
Frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP) is a neurodegenerative disease primarily affecting the frontal and/or temporal cortices. However, a growing body of evidence suggests that the cerebellum contributes to biochemical, cognitive, and behavioral changes in FTLD-TDP. To evaluate cerebellar TDP-43 expression and function in FTLD-TDP, we analyzed TDP-43 protein levels and the splicing of a TDP-43 target, STMN2, in the cerebellum of 95 FTLD-TDP cases and 25 non-neurological disease controls. Soluble TDP-43 was decreased in the cerebellum of FTLD-TDP cases but a concomitant increase in insoluble TDP-43 was not seen. Truncated STMN2 transcripts, an indicator of TDP-43 dysfunction, were elevated in the cerebellum of FTLD-TDP cases and inversely associated with TDP-43 levels. Additionally, lower cerebellar TDP-43 associated with a younger age at disease onset. We provide evidence of TDP-43 loss of function in the cerebellum in FTLD-TDP, supporting further investigation into this understudied brain region. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
13. Long-read targeted sequencing uncovers clinicopathological associations for C9orf72-linked diseases.
- Author
-
DeJesus-Hernandez, Mariely, Aleff, Ross A, Jackson, Jazmyne L, Finch, NiCole A, Baker, Matthew C, Gendron, Tania F, Murray, Melissa E, McLaughlin, Ian J, Harting, John R, Graff-Radford, Neill R, Oskarsson, Björn, Knopman, David S, Josephs, Keith A, Boeve, Bradley F, Petersen, Ronald C, Fryer, John D, Petrucelli, Leonard, Dickson, Dennis W, Rademakers, Rosa, and Ebbert, Mark T W
- Subjects
FRONTOTEMPORAL lobar degeneration ,MOTOR neuron diseases ,AMYOTROPHIC lateral sclerosis ,NEURODEGENERATION - Abstract
To examine the length of a hexanucleotide expansion in C9orf72, which represents the most frequent genetic cause of frontotemporal lobar degeneration and motor neuron disease, we employed a targeted amplification-free long-read sequencing technology: No-Amp sequencing. In our cross-sectional study, we assessed cerebellar tissue from 28 well-characterized C9orf72 expansion carriers. We obtained 3507 on-target circular consensus sequencing reads, of which 814 bridged the C9orf72 repeat expansion (23%). Importantly, we observed a significant correlation between expansion sizes obtained using No-Amp sequencing and Southern blotting (P = 5.0 × 10-4). Interestingly, we also detected a significant survival advantage for individuals with smaller expansions (P = 0.004). Additionally, we uncovered that smaller expansions were significantly associated with higher levels of C9orf72 transcripts containing intron 1b (P = 0.003), poly(GP) proteins (P = 1.3 × 10- 5), and poly(GA) proteins (P = 0.005). Thorough examination of the composition of the expansion revealed that its GC content was extremely high (median: 100%) and that it was mainly composed of GGGGCC repeats (median: 96%), suggesting that expanded C9orf72 repeats are quite pure. Taken together, our findings demonstrate that No-Amp sequencing is a powerful tool that enables the discovery of relevant clinicopathological associations, highlighting the important role played by the cerebellar size of the expanded repeat in C9orf72-linked diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
14. Protein contributions to brain atrophy acceleration in Alzheimer's disease and primary age-related tauopathy.
- Author
-
Josephs, Keith A, Martin, Peter R, Weigand, Stephen D, Tosakulwong, Nirubol, Buciuc, Marina, Murray, Melissa E, Petrucelli, Leonard, Senjem, Matthew L, Spychalla, Anthony J, Knopman, David S, Boeve, Bradley F, Petersen, Ronald C, Parisi, Joseph E, Dickson, Dennis W, Jack, Clifford R, and Whitwell, Jennifer L
- Subjects
CEREBRAL atrophy ,ALZHEIMER'S disease ,DNA-binding proteins ,AGE factors in disease ,CEREBRAL amyloid angiopathy ,BRAIN ,NEURONS ,HIPPOCAMPUS (Brain) ,AUTOPSY ,RETROSPECTIVE studies ,MAGNETIC resonance imaging ,ATROPHY ,RESEARCH funding ,NEURODEGENERATION ,LONGITUDINAL method ,CEREBRAL cortex - Abstract
Alzheimer's disease is characterized by the presence of amyloid-β and tau deposition in the brain, hippocampal atrophy and increased rates of hippocampal atrophy over time. Another protein, TAR DNA binding protein 43 (TDP-43) has been identified in up to 75% of cases of Alzheimer's disease. TDP-43, tau and amyloid-β have all been linked to hippocampal atrophy. TDP-43 and tau have also been linked to hippocampal atrophy in cases of primary age-related tauopathy, a pathological entity with features that strongly overlap with those of Alzheimer's disease. At present, it is unclear whether and how TDP-43 and tau are associated with early or late hippocampal atrophy in Alzheimer's disease and primary age-related tauopathy, whether either protein is also associated with faster rates of atrophy of other brain regions and whether there is evidence for protein-associated acceleration/deceleration of atrophy rates. We therefore aimed to model how these proteins, particularly TDP-43, influence non-linear trajectories of hippocampal and neocortical atrophy in Alzheimer's disease and primary age-related tauopathy. In this longitudinal retrospective study, 557 autopsied cases with Alzheimer's disease neuropathological changes with 1638 ante-mortem volumetric head MRI scans spanning 1.0-16.8 years of disease duration prior to death were analysed. TDP-43 and Braak neurofibrillary tangle pathological staging schemes were constructed, and hippocampal and neocortical (inferior temporal and middle frontal) brain volumes determined using longitudinal FreeSurfer. Bayesian bivariate-outcome hierarchical models were utilized to estimate associations between proteins and volume, early rate of atrophy and acceleration in atrophy rates across brain regions. High TDP-43 stage was associated with smaller cross-sectional brain volumes, faster rates of brain atrophy and acceleration of atrophy rates, more than a decade prior to death, with deceleration occurring closer to death. Stronger associations were observed with hippocampus compared to temporal and frontal neocortex. Conversely, low TDP-43 stage was associated with slower early rates but later acceleration. This later acceleration was associated with high Braak neurofibrillary tangle stage. Somewhat similar, but less striking, findings were observed between TDP-43 and neocortical rates. Braak stage appeared to have stronger associations with neocortex compared to TDP-43. The association between TDP-43 and brain atrophy occurred slightly later in time (∼3 years) in cases of primary age-related tauopathy compared to Alzheimer's disease. The results suggest that TDP-43 and tau have different contributions to acceleration and deceleration of brain atrophy rates over time in both Alzheimer's disease and primary age-related tauopathy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
15. Elevated methylation levels, reduced expression levels, and frequent contractions in a clinical cohort of C9orf72 expansion carriers.
- Author
-
Jackson, Jazmyne L., Finch, NiCole A., Baker, Matthew C., Kachergus, Jennifer M., DeJesus-Hernandez, Mariely, Pereira, Kimberly, Christopher, Elizabeth, Prudencio, Mercedes, Heckman, Michael G., Thompson, E. Aubrey, Dickson, Dennis W., Shah, Jaimin, Oskarsson, Björn, Petrucelli, Leonard, Rademakers, Rosa, and van Blitterswijk, Marka
- Subjects
METHYLATION ,DNA methylation ,AMYOTROPHIC lateral sclerosis ,MOTOR neuron diseases ,FRONTOTEMPORAL dementia ,NEURODEGENERATION - Abstract
Background: A repeat expansion in the C9orf72-SMCR8 complex subunit (C9orf72) is the most common genetic cause of two debilitating neurodegenerative diseases: amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Currently, much remains unknown about which variables may modify these diseases. We sought to investigate associations between C9orf72 promoter methylation, RNA expression levels, and repeat length, their potential effects on disease features, as well as changes over time and within families. Methods: All samples were obtained through the ALS Center at Mayo Clinic Florida. Our primary cohort included 75 unrelated patients with an expanded C9orf72 repeat, 33 patients who did not possess this expansion, and 20 control subjects without neurodegenerative diseases. Additionally, 67 members from 17 independent C9orf72 families were selected of whom 33 harbored this expansion. Longitudinally collected samples were available for 35 C9orf72 expansion carriers. To increase our understanding of C9orf72-related diseases, we performed quantitative methylation-sensitive restriction enzyme-based assays, digital molecular barcoding, quantitative real-time PCR, and Southern blotting. Results: In our primary cohort, higher methylation levels were observed in patients with a C9orf72 repeat expansion than in patients without this expansion (p = 1.7e-13) or in control subjects (p = 3.3e-07). Moreover, we discovered that an increase in methylation levels was associated with a decrease in total C9orf72 transcript levels (p = 5.5e-05). These findings aligned with our observation that C9orf72 expansion carriers had lower expression levels of total C9orf72 transcripts than patients lacking this expansion (p = 3.7e-07) or control subjects (p = 9.1e-05). We also detected an elevation of transcripts containing intron 1a (upstream of the repeat) in patients carrying a C9orf72 repeat expansion compared to (disease) controls (p ≤ 0.01), an indication of abortive transcripts and/or a switch in transcription start site usage. While methylation and expression levels were relatively stable over time, fluctuations were seen in repeat length. Interestingly, contractions occurred frequently in parent-offspring transmissions (> 50%), especially in paternal transmissions. Furthermore, smaller repeat lengths were detected in currently unaffected individuals than in affected individuals (p = 8.9e-04) and they were associated with an earlier age at collection (p = 0.008). Conclusions: In blood from C9orf72 expansion carriers, we found elevated methylation levels, reduced expression levels, and unstable expansions that tend to contract in successive generations, arguing against anticipation. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
16. ADAR2 mislocalization and widespread RNA editing aberrations in C9orf72-mediated ALS/FTD.
- Author
-
Lorenzini, Ileana, Starr, Alexander, Rabichow, Benjamin E., Mendez, Emily, Levy, Jennifer L., Burciu, Camelia, Sattler, Rita, Moore, Stephen, Alsop, Eric, Reiman, Rebecca, Van Keuren-Jensen, Kendall, Chew, Jeannie, Belzil, Veronique V., W. Dickson, Dennis, Petrucelli, Leonard, Robertson, Janice, Staats, Kim A., and Ichida, Justin K.
- Subjects
RNA editing ,AMYOTROPHIC lateral sclerosis ,RNA-binding proteins ,DOUBLE-stranded RNA ,CATALYTIC RNA ,ADENOSINE deaminase - Abstract
The hexanucleotide repeat expansion GGGGCC (G
4 C2 )n in the C9orf72 gene is the most common genetic abnormality associated with amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Recent findings suggest that dysfunction of nuclear-cytoplasmic trafficking could affect the transport of RNA binding proteins in C9orf72 ALS/FTD. Here, we provide evidence that the RNA editing enzyme adenosine deaminase acting on RNA 2 (ADAR2) is mislocalized in C9orf72 repeat expansion mediated ALS/FTD. ADAR2 is responsible for adenosine (A) to inosine (I) editing of double-stranded RNA, and its function has been shown to be essential for survival. Here we show the mislocalization of ADAR2 in human induced pluripotent stem cell-derived motor neurons (hiPSC-MNs) from C9orf72 patients, in mice expressing (G4 C2 )149 , and in C9orf72 ALS/FTD patient postmortem tissue. As a consequence of this mislocalization we observe alterations in RNA editing in our model systems and across multiple brain regions. Analysis of editing at 408,580 known RNA editing sites indicates that there are vast RNA A to I editing aberrations in C9orf72-mediated ALS/FTD. These RNA editing aberrations are found in many cellular pathways, such as the ALS pathway and the crucial EIF2 signaling pathway. Our findings suggest that the mislocalization of ADAR2 in C9orf72 mediated ALS/FTD is responsible for the alteration of RNA processing events that may impact vast cellular functions, including the integrated stress response (ISR) and protein translation. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
17. The Caenorhabditis elegans Ortholog of TDP-43 Regulates the Chromatin Localization of the Heterochromatin Protein 1 Homolog HPL-2.
- Author
-
Saldi, Tassa K., Gonzales, Patrick, Garrido-Lecca, Alfonso, Dostal, Vishantie, Roberts, Christine M., Petrucelli, Leonard, and Link, Christopher D.
- Subjects
CAENORHABDITIS elegans ,TDP-43 proteinopathies ,HETEROCHROMATIN ,FRONTOTEMPORAL dementia ,AMYOTROPHIC lateral sclerosis - Abstract
TDP-1 is the Caenorhabditis elegans ortholog of mammalian TDP-43, which is strongly implicated in the etiology of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). We discovered that deletion of the tdp-1 gene results in enhanced nuclear RNA interference (RNAi). As nuclear RNAi in C. elegans involves chromatin changes moderated by HPL-2, a homolog of heterochromatin protein 1 (HP1), we investigated the interaction of TDP-1 and HPL-2. We found that TDP-1 and HPL-2 interact directly and that loss of TDP-1 dramatically alters the chromatin association of HPL-2. We showed previously that deletion of the tdp-1 gene results in transcriptional alterations and the accumulation of double-stranded RNA (dsRNA). These molecular changes are replicated in an hpl-2 deletion strain, consistent with HPL-2 acting in consort with TDP-1 to modulate these aspects of RNA metabolism. Our observations identify novel mechanisms by which HP1 homologs can be recruited to chromatin and by which nuclear depletion of human TDP-43 may lead to changes in RNA metabolism that are relevant to disease. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
18. A zebrafish model for C9orf72 ALS reveals RNA toxicity as a pathogenic mechanism.
- Author
-
Swinnen, Bart, Bento-Abreu, Andre, Gendron, Tania F., Boeynaems, Steven, Bogaert, Elke, Nuyts, Rik, Timmers, Mieke, Scheveneels, Wendy, Hersmus, Nicole, Wang, Jiou, Mizielinska, Sarah, Isaacs, Adrian M., Petrucelli, Leonard, Lemmens, Robin, Van Damme, Philip, Van Den Bosch, Ludo, and Robberecht, Wim
- Subjects
AMYOTROPHIC lateral sclerosis ,NEURODEGENERATION - Abstract
The exact mechanism underlying amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) associated with the GGGGCC repeat expansion in
C9orf72 is still unclear. Two gain-of-function mechanisms are possible: repeat RNA toxicity and dipeptide repeat protein (DPR) toxicity. We here dissected both possibilities using a zebrafish model for ALS. Expression of two DPRs, glycine–arginine and proline–arginine, induced a motor axonopathy. Similarly, expanded sense and antisense repeat RNA also induced a motor axonopathy and formed mainly cytoplasmic RNA foci. However, DPRs were not detected in these conditions. Moreover, stop codon-interrupted repeat RNA still induced a motor axonopathy and a synergistic role of low levels of DPRs was excluded. Altogether, these results show that repeat RNA toxicity is independent of DPR formation. This RNA toxicity, but not the DPR toxicity, was attenuated by the RNA-binding protein Pur-alpha and the autophagy-related protein p62. Our findings demonstrate that RNA toxicity, independent of DPR toxicity, can contribute to the pathogenesis ofC9orf72 -associated ALS/FTD. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
19. ALS and FTD: an epigenetic perspective.
- Author
-
Belzil, Veronique, Katzman, Rebecca, and Petrucelli, Leonard
- Subjects
GENETICS of amyotrophic lateral sclerosis ,FRONTOTEMPORAL dementia ,EPIGENETICS ,NEURODEGENERATION ,COMORBIDITY ,DNA methylation ,GENETICS - Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two fatal neurodegenerative diseases seen in comorbidity in up to 50 % of cases. Despite tremendous efforts over the last two decades, no biomarkers or effective therapeutics have been identified to prevent, decelerate, or stop neuronal death in patients. While the identification of multiple mutations in more than two dozen genes elucidated the involvement of several mechanisms in the pathogenesis of both diseases, identifying the hexanucleotide repeat expansion in C9orf72, the most common genetic abnormality in ALS and FTD, opened the door to the discovery of several novel pathogenic biological routes, including chromatin remodeling and transcriptome alteration. Epigenetic processes regulate DNA replication and repair, RNA transcription, and chromatin conformation, which in turn further dictate transcriptional regulation and protein translation. Transcriptional and post-transcriptional epigenetic regulation is mediated by enzymes and chromatin-modifying complexes that control DNA methylation, histone modifications, and RNA editing. While the alteration of DNA methylation and histone modification has recently been reported in ALS and FTD, the assessment of epigenetic involvement in both diseases is still at an early stage, and the involvement of multiple epigenetic players still needs to be evaluated. As the epigenome serves as a way to alter genetic information not only during aging, but also following environmental signals, epigenetic mechanisms might play a central role in initiating ALS and FTD, especially for sporadic cases. Here, we provide a review of what is currently known about altered epigenetic processes in both ALS and FTD and discuss potential therapeutic strategies targeting epigenetic mechanisms. As approximately 85 % of ALS and FTD cases are still genetically unexplained, epigenetic therapeutics explored for other diseases might represent a profitable direction for the field. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
20. Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat form nuclear RNA foci and undergo repeat-associated non-ATG translation in c9FTD/ALS.
- Author
-
Gendron, Tania F., Bieniek, Kevin F., Zhang, Yong-Jie, Jansen-West, Karen, Ash, Peter E. A., Caulfield, Thomas, Daughrity, Lillian, Dunmore, Judith H., Castanedes-Casey, Monica, Chew, Jeannie, Cosio, Danielle M., van Blitterswijk, Marka, Lee, Wing C., Rademakers, Rosa, Boylan, Kevin B., Dickson, Dennis W., and Petrucelli, Leonard
- Subjects
FRONTOTEMPORAL dementia ,AMYOTROPHIC lateral sclerosis ,NEURODEGENERATION ,NUCLEOTIDES ,MESSENGER RNA - Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are devastating neurodegenerative disorders with clinical, genetic, and neuropathological overlap. A hexanucleotide (GGGGCC) repeat expansion in a non-coding region of C9ORF72 is the major genetic cause of both diseases. The mechanisms by which this repeat expansion causes “c9FTD/ALS” are not definitively known, but RNA-mediated toxicity is a likely culprit. RNA transcripts of the expanded GGGGCC repeat form nuclear foci in c9FTD/ALS, and also undergo repeat-associated non-ATG (RAN) translation resulting in the production of three aggregation-prone proteins. The goal of this study was to examine whether antisense transcripts resulting from bidirectional transcription of the expanded repeat behave in a similar manner. We show that ectopic expression of (CCCCGG)
66 in cultured cells results in foci formation. Using novel polyclonal antibodies for the detection of possible (CCCCGG)exp RAN proteins [poly(PR), poly(GP) and poly(PA)], we validated that (CCCCGG)66 is also subject to RAN translation in transfected cells. Of importance, foci composed of antisense transcripts are observed in the frontal cortex, spinal cord and cerebellum of c9FTD/ALS cases, and neuronal inclusions of poly(PR), poly(GP) and poly(PA) are present in various brain tissues in c9FTD/ALS, but not in other neurodegenerative diseases, including CAG repeat disorders. Of note, RNA foci and poly(GP) inclusions infrequently co-occur in the same cell, suggesting these events represent two distinct ways in which the C9ORF72 repeat expansion may evoke neurotoxic effects. These findings provide mechanistic insight into the pathogenesis of c9FTD/ALS, and have significant implications for therapeutic strategies. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
21. Robust cytoplasmic accumulation of phosphorylated TDP-43 in transgenic models of tauopathy.
- Author
-
Clippinger, Amy, D'Alton, Simon, Lin, Wen-Lang, Gendron, Tania, Howard, John, Borchelt, David, Cannon, Ashley, Carlomagno, Yari, Chakrabarty, Paramita, Cook, Casey, Golde, Todd, Levites, Yona, Ranum, Laura, Schultheis, Patrick, Xu, Guilian, Petrucelli, Leonard, Sahara, Naruhiko, Dickson, Dennis, Giasson, Benoit, and Lewis, Jada
- Subjects
DNA-binding proteins ,NEURODEGENERATION ,TRANSGENIC mice ,NEUROLOGICAL disorders ,PHOSPHORYLATION ,GENETICS - Abstract
Frontotemporal lobar degeneration (FTLD) has been subdivided based on the main pathology found in the brains of affected individuals. When the primary pathology is aggregated, hyperphosphorylated tau, the pathological diagnosis is FTLD-tau. When the primary pathology is cytoplasmic and/or nuclear aggregates of phosphorylated TAR-DNA-binding protein (TDP-43), the pathological diagnosis is FTLD-TDP. Notably, TDP-43 pathology can also occur in conjunction with a number of neurodegenerative disorders; however, unknown environmental and genetic factors may regulate this TDP-43 pathology. Using transgenic mouse models of several diseases of the central nervous system, we explored whether a primary proteinopathy might secondarily drive TDP-43 proteinopathy. We found abnormal, cytoplasmic accumulation of phosphorylated TDP-43 specifically in two tau transgenic models, but TDP-43 pathology was absent in mouse models of Aβ deposition, α-synucleinopathy or Huntington's disease. Though tau pathology showed considerable overlap with cytoplasmic, phosphorylated TDP-43, tau pathology generally preceded TDP-43 pathology. Biochemical analysis confirmed the presence of TDP-43 abnormalities in the tau mice, which showed increased levels of high molecular weight, soluble TDP-43 and insoluble full-length and ~35 kD TDP-43. These data demonstrate that the neurodegenerative cascade associated with a primary tauopathy in tau transgenic mice can also promote TDP-43 abnormalities. These findings provide the first in vivo models to understand how TDP-43 pathology may arise as a secondary consequence of a primary proteinopathy. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
22. Tau Triage Decisions Mediated by the Chaperone Network.
- Author
-
Cook, Casey and Petrucelli, Leonard
- Subjects
- *
NEURODEGENERATION , *ALZHEIMER'S disease , *DEMENTIA , *MOLECULAR chaperones , *DEGENERATION (Pathology) - Abstract
The pathological accumulation of the microtubule-binding protein tau is linked to an increasing number of neurodegenerative conditions associated with aging, though the mechanisms by which tau accumulates in disease are unclear. In this review, we will summarize our previous research assessing the mechanism of action, as well as the therapeutic potential of Hsp90 inhibition for the treatment of tauopathies. Specifically, we describe the development of a high-throughput screening approach to identify and rank compounds, and demonstrate the selective elimination of aberrant p-tau species in the brain following treatment with an Hsp90 inhibitor. Additionally, we identify CHIP as an essential component of the Hsp90 chaperone complex that mediates tau degradation, and present evidence to suggest that CHIP functions to identify and sequester neurotoxic tau species. Finally, we discuss recent data identifying an additional mechanism by which CHIP modulates protein triage decisions involving Hsp90. Specifically, CHIP indirectly regulates Hsp90 chaperone activity by modulating steady-state levels of the Hsp90 deacetylase, HDAC6, thus influencing both the acetylation state and function of Hsp90. Thus future research directions will focus on the manipulation of this network to promote degradation of pathogenic tau species in disease. [ABSTRACT FROM AUTHOR]
- Published
- 2013
23. TARDBP Mutation Analysis in TDP-43 Proteinopathies and Deciphering the Toxicity of Mutant TDP-43.
- Author
-
Gendron, Tania F., Rademakers, Rosa, and Petrucelli, Leonard
- Subjects
AMYOTROPHIC lateral sclerosis ,GENETIC mutation ,UBIQUITIN ,DEGENERATION (Pathology) ,LYMPHOBLASTOID cell lines - Abstract
The identification of TAR DNA-binding protein 43 (TDP-43) as the major disease protein in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with ubiquitin inclusions has defined a new class of neurodegenerative conditions: the TDP-43 proteinopathies. This breakthrough was quickly followed by mutation analysis of TARDBP, the gene encoding TDP-43. Herein, we provide a review of our previously published efforts that led to the identification of 3 TARDBP mutations (p.M337V, p.N345K, and p.I383V) in familial ALS patients, two of which were novel. With over 40 TARDBP mutations now discovered, there exists conclusive evidence that TDP-43 plays a direct role in neurodegeneration. The onus is now on researchers to elucidate the mechanisms by which mutant TDP-43 confers toxicity, and to exploit these findings to gain a better understanding of how TDP-43 contributes to the pathogenesis of disease. Our biochemical analysis of TDP-43 in ALS patient lymphoblastoid cell lines revealed a substantial increase in TDP-43 truncation products, including a ~25 kDa fragment, compared to control lymphoblastoid cell lines. We discuss the putative harmful consequence of abnormal TDP-43 fragmentation, as well as highlight additional mechanisms of toxicity associated with mutant TDP-43. [ABSTRACT FROM AUTHOR]
- Published
- 2013
24. Misregulation of human sortilin splicing leads to the generation of a nonfunctional progranulin receptor.
- Author
-
Prudencio, Mercedes, Jansen-West, Karen R., Lee, Wing C., Gendron, Tania F., Yong-Jie Zhang, Ya-Fei Xu, Gass, Jennifer, Stuani, Cristiana, Stetler, Caroline, Rademakers, Rosa, Dickson, Dennis W., Buratti, Emanuele, and Petrucelli, Leonard
- Subjects
PROGRANULIN ,NEUROTROPHIC functions ,DEGENERATION (Pathology) ,MESSENGER RNA ,GENETIC code - Abstract
Sortilin 1 regulates the levels of brain progranulin (PGRN), a neurotrophic growth factor that, when deficient, is linked to cases of frontotemporal lobar degeneration with TAR DNA-binding protein-43 (TDP-43)-positive inclusions (FTLD-TDP). We identified a specific splicing enhancer element that regulates the inclusion of a sortilin exon cassette (termed Ex17b) not normally present in the mature sortilin mRNA. This enhancer element is consistently present in sortilin RNA of mice and other species but absent in primates, which carry a premature stop codon within the Ex17b sequence. In the absence of TDP-43, which acts as a regulatory inhibitor, Ex17b is included in the sortilin mRNA. In humans, in contrast to mice, the inclusion of Ex17b in sortilin mRNA generates a truncated, nonfunctional, extracellularly released protein that binds to but does not internalize PGRN, essentially acting as a decoy receptor. Based on these results, we propose a potential mechanism linking misregulation of sortilin splicing with altered PGRN metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
25. TDP-1/TDP-43 Regulates Stress Signaling and Age Dependent Proteotoxicity in Caenorhabditis elegans.
- Author
-
Vaccaro, Alexandra, Tauffenberger, Arnaud, Ash, Peter E. A., Carlomagno, Yari, Petrucelli, Leonard, and Parker, J. Alex
- Subjects
CAENORHABDITIS elegans ,CARRIER proteins ,NEURODEGENERATION ,AMYOTROPHIC lateral sclerosis ,FRONTOTEMPORAL dementia ,TRANSCRIPTION factors - Abstract
TDP-43 is a multifunctional nucleic acid binding protein linked to several neurodegenerative diseases including Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia. To learn more about the normal biological and abnormal pathological role of this protein, we turned to Caenorhabditis elegans and its orthologue TDP-1. We report that TDP-1 functions in the Insulin/IGF pathway to regulate longevity and the oxidative stress response downstream from the forkhead transcription factor DAF-16/FOXO3a. However, although tdp-1 mutants are stress-sensitive, chronic upregulation of tdp-1 expression is toxic and decreases lifespan. ALS--associated mutations in TDP-43 or the related RNA binding protein FUS activate the unfolded protein response and generate oxidative stress leading to the daf-16--dependent upregulation of tdp-1 expression with negative effects on neuronal function and lifespan. Consistently, deletion of endogenous tdp-1 rescues mutant TDP-43 and FUS proteotoxicity in C. elegans. These results suggest that chronic induction of wild-type TDP- 1/TDP-43 by cellular stress may propagate neurodegeneration and decrease lifespan. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
26. Targeting Aβ and tau in Alzheimer's disease, an early interim report
- Author
-
Golde, Todd E., Petrucelli, Leonard, and Lewis, Jada
- Subjects
- *
ALZHEIMER'S disease research , *AMYLOID beta-protein , *NERVE tissue proteins , *NEURODEGENERATION , *GENETIC mutation , *FRONTOTEMPORAL dementia , *CLINICAL trials , *MEDICAL triage - Abstract
Abstract: The amyloid β (Aβ) and tau proteins, which misfold, aggregate, and accumulate in the Alzheimer''s disease (AD) brain, are implicated as central factors in a complex neurodegenerative cascade. Studies of mutations that cause early onset AD and promote Aβ accumulation in the brain strongly support the notion that inhibiting Aβ aggregation will prevent AD. Similarly, genetic studies of frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17 MAPT) showing that mutations in the MAPT gene encoding tau lead to abnormal tau accumulation and neurodegeneration. Such genetic studies clearly show that tau dysfunction and aggregation can be central to neurodegeneration, however, most likely in a secondary fashion in relation to AD. Additional pathologic, biochemical, and modeling studies further support the concept that Aβ and tau are prime targets for disease modifying therapies in AD. Treatment strategies aimed at preventing the aggregation and accumulation of Aβ, tau, or both proteins should therefore be theoretically possible, assuming that treatment can be initiated before either irreversible damage is present or downstream, self-sustaining, pathological cascades have been initiated. Herein, we will review recent advances and also potential setbacks with respect to the myriad of therapeutic strategies that are designed to slow down, prevent, or clear the accumulation of either “pathological” Aβ or tau. We will also discuss the need for thoughtful prioritization with respect to clinical development of the preclinically validated modifiers of Aβ and tau pathology. The current number of candidate therapies targeting Aβ is becoming so large that a triage process is clearly needed to insure that resources are invested in a way such that the best candidates for disease modifying therapy are rapidly moved toward clinical trials. Finally, we will discuss the challenges for an appropriate “triage” after potential disease modifying therapies targeting tau and Aβ have entered clinical trials. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
27. Phosphorylation regulates proteasomal-mediated degradation and solubility of TAR DNA binding protein-43 C-terminal fragments.
- Author
-
Yong-Jie Zhang, Gendron, Tania F., Ya-Fei Xu, Li-Wen Ko, Shu-Hui Yen, and Petrucelli, Leonard
- Subjects
GREEN fluorescent protein ,PHOSPHORYLATION ,NEURODEGENERATION ,FLUORESCENT polymers ,NEUROBLASTOMA - Abstract
Background: Inclusions of TAR DNA binding protein-43 (TDP-43) are the defining histopathological feature of several neurodegenerative diseases collectively referred to as TDP-43 proteinopathies. These diseases are characterized by the presence of cellular aggregates composed of abnormally phosphorylated, N-terminally truncated and ubiquitinated TDP-43 in the spinal cord and/or brain. Recent studies indicate that C-terminal fragments of TDP-43 are aggregation-prone and induce cytotoxicity. However, little is known regarding the pathways responsible for the degradation of these fragments and how their phosphorylation contributes to the pathogenesis of disease. Results: Herein, we established a human neuroblastoma cell line (M17D3) that conditionally expresses an enhanced green fluorescent protein (GFP)-tagged caspase-cleaved C-terminal TDP-43 fragment (GFP-TDP
220-414 ). We report that expression of this fragment within cells leads to a time-dependent formation of inclusions that are immunoreactive for both ubiquitin and phosphorylated TDP-43, thus recapitulating pathological hallmarks of TDP-43 proteinopathies. Phosphorylation of GFP-TDP220-414 renders it resistant to degradation and enhances its accumulation into insoluble aggregates. Nonetheless, GFP-TDP220-414 inclusions are reversible and can be cleared through the ubiquitin proteasome system. Moreover, both Hsp70 and Hsp90 bind to GFP-TDP220-414 and regulate its degradation. Conclusions: Our data indicates that inclusions formed from TDP-43 C-terminal fragments are reversible. Given that TDP-43 inclusions have been shown to confer toxicity, our findings have important therapeutic implications and suggest that modulating the phosphorylation state of TDP-43 C-terminal fragments may be a promising therapeutic strategy to clear TDP-43 inclusions. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
28. Aging Is Not Associated with Proteasome Impairment in UPS Reporter Mice.
- Author
-
Cook, Casey, Gass, Jennifer, Dunmore, Judith, Tong, Jimei, Taylor, Julie, Eriksen, Jason, McGowan, Eileen, Lewis, Jada, Johnston, Jennifer, and Petrucelli, Leonard
- Subjects
UBIQUITIN ,PROTEINS ,ECOLOGICAL disturbances ,CELL physiology ,NEURODEGENERATION ,AGING ,PHARMACOLOGY ,NEURONS ,DEGENERATION (Pathology) - Abstract
Background: Covalent linkage of ubiquitin regulates the function and, ultimately, the degradation of many proteins by the ubiquitin-proteasome system (UPS). Given its essential role in protein regulation, even slight perturbations in UPS activity can substantially impair cellular function. Methodology/Principal Findings: We have generated and characterized a novel transgenic mouse model which expresses a previously described reporter for UPS function. This UPS reporter contains a degron sequence attached to the C-terminus of green fluorescent protein, and is predominantly expressed in neurons throughout the brain of our transgenic model. We then demonstrated that this reporter system is sensitive to UPS inhibition in vivo. Conclusions/Significance: Given the obstacles associated with evaluating proteasomal function in the brain, our mouse model uniquely provides the capability to monitor UPS function in real time in individual neurons of a complex organism. Our novel mouse model now provides a useful resource with which to evaluate the impact of aging, as well as various genetic and/or pharmacological modifiers of neurodegenerative disease(s). [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
29. The role of tau in neurodegeneration.
- Author
-
Gendron, Tania F. and Petrucelli, Leonard
- Subjects
- *
HUNTINGTON disease , *ALZHEIMER'S disease , *BRAIN diseases , *NEURODEGENERATION , *DEMENTIA - Abstract
Since the identification of tau as the main component of neurofibrillary tangles in Alzheimer's disease and related tauopathies, and the discovery that mutations in the tau gene cause frontotemporal dementia, much effort has been directed towards determining how the aggregation of tau into fibrillar inclusions causes neuronal death. As evidence emerges that tau-mediated neuronal death can occur even in the absence of tangle formation, a growing number of studies are focusing on understanding how abnormalities in tau (e.g. aberrant phosphorylation, glycosylation or truncation) confer toxicity. Though data obtained from experimental models of tauopathies strongly support the involvement of pathologically modified tau and tau aggregates in neurodegeneration, the exact neurotoxic species remain unclear, as do the mechanism(s) by which they cause neuronal death. Nonetheless, it is believed that tau-mediated neurodegeneration is likely to result from a combination of toxic gains of function as well as from the loss of normal tau function. To truly appreciate the detrimental consequences of aberrant tau function, a better understanding of all functions carried out by tau, including but not limited to the role of tau in microtubule assembly and stabilization, is required. This review will summarize what is currently known regarding the involvement of tau in the initiation and development of neurodegeneration in tauopathies, and will also highlight some of the remaining questions in need of further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
30. Aβ Puts the Alpha in Synuclein.
- Author
-
Cook, Casey and Petrucelli, Leonard
- Subjects
- *
PARKINSON'S disease , *ALZHEIMER'S disease , *NEURODEGENERATION - Abstract
Neurodegenerative diseases like Alzheimer's disease and Parkinson's disease are characterized pathologically by aberrant protein accumulation, such as Aβ or α-synuclein deposition. In this issue of Neuron , Bassil et al. (2020) observed an exacerbation of α-syn pathology in the presence of Aβ plaques in vivo , with comorbid pathologies associated with greater neurodegeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
31. Pharmacologic reductions of total tau levels; implications for the role of microtubule dynamics in regulating tau expression.
- Author
-
Dickey, Chad A., Ash, Peter, Klosak, Natalia, Lee, Wing C., Petrucelli, Leonard, Hutton, Michael, and Eckman, Christopher B.
- Subjects
DEMENTIA ,NEURODEGENERATION ,PROTEINS ,NEUROBLASTOMA ,MEDICINE - Abstract
The microtubule-associated protein tau (MAPT) is a pathological component of several neurodegenerative diseases and clinical dementias. Here, we have investigated the effects of a series of commercially available FDA-approved compounds and natural products on total tau protein levels using a cell-based approach that allows for the rapid and efficient measurement of changes in protein expression. Results: The compounds that reduced tau largely fell within 3 functional categories with the largest percentage being microtubule regulators. Several of these candidates were validated in both a human neuroglioma and a human neuroblastoma cell line. While these drugs lead to a rapid reduction in tau protein levels, a selective decrease in MAPT mRNA expression was also observed. Conclusion: These findings suggest that the identified compounds that reduce tau levels may act either through direct effects on the MAPT promoter itself or by altering a feedback transcriptional mechanism regulating MAPT transcription. This is particularly interesting in light of recent evidence suggesting that MAPT 5′ UTR mutations in late-onset PD and PSP cases alter the expression of tau mRNA. In fact, one of the compounds we identified, rotenone, has been used extensively to model PD in rodents. These observations may provide key insights into the mechanism of tau turnover within the neuron while also providing the first evidence that selectively reducing tau protein levels may be possible using compounds that are FDA-approved for other uses. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
32. Molecular Pathogenesis of Parkinson Disease.
- Author
-
Eriksen, Jason L., Wszolek, Zbigniew, and Petrucelli, Leonard
- Subjects
ETIOLOGY of diseases ,PARKINSON'S disease ,SYMPTOMS ,NEURODEGENERATION ,SUBSTANTIA nigra ,DOPAMINERGIC neurons - Abstract
Parkinson disease (PD), the most common neurodegenerative movement disorder, is characterized by an extensive and progressive loss of dopaminergic neurons in the substantia nigra pars compacta. One of the pathological hallmarks of PD is the presence of Lewy bodies, intracellular inclusions of aggregated α-synuclein. Although the cause and pathogenesis of selective loss of dopamine neurons and the accumulation of α-synuclein in PD remain elusive, growing lines of evidence from environmental risk factors and early-onset genetics point to a convergence between energy metabolism and the disposal of damaged proteins in the development of PD. These findings suggest that impairments in mitochondrial and ubiquitin-proteasome system function can significantly contribute to the pathogenesis of PD. This review will summarize recent insights gained from genetic and environmental studies of PD that underscore this association. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
33. Alterations in microRNA-124 and AMPA receptors contribute to social behavioral deficits in frontotemporal dementia
- Author
-
Gascon, Eduardo, Lynch, Kelleen, Ruan, Hongyu, Almeida, Sandra, Verheyden, Jamie, Seeley, William W., Dickson, Dennis W., Petrucelli, Leonard, Sun, Danqiong, Jiao, Jian, Zhou, Hongru, Jakovcevski, Mira, Akbarian, Schahram, Yao, Wei-Dong, and Gao, Fen-Biao
- Subjects
AMPA receptor ,CHMP2B ,frontotemporal dementia ,microRNA ,neurodegeneration ,social behavior - Abstract
Many neurodegenerative diseases, such as frontotemporal dementia (FTD), are associated with behavioral deficits, but the anatomical and molecular bases remain poorly understood. Here we show that forebrain-specific expression of FTD-associated mutant CHMP2B causes several age-dependent neurodegenerative phenotypes, including social behavioral impairments. The social deficits were accompanied by a change in AMPA receptor (AMPAR) composition, leading to imbalance between Ca2+-permeable and -impermeable AMPARs. Expression of most AMPAR subunits was regulated by the brain-enriched microRNA (miR-124), whose abundance was markedly decreased in the superficial layers of cerebral cortex of FTD mice. We found similar changes in miR-124 and AMPAR levels in the frontal cortex and iPSC-derived neurons of subjects with behavioral variant FTD. Moreover, miR-124 expression in the medial prefrontal cortex decreased AMPAR levels and partially rescued behavioral deficits. Knockdown of Gria2 also alleviated social impairments in FTD mice. Our results identify a novel mechanism involving miR-124 and AMAPRs in regulating social behavior in FTD and suggest a potential therapeutic avenue.
- Published
- 2014
- Full Text
- View/download PDF
34. Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson's disease.
- Author
-
Lee, Byoung Dae, Shin, Joo-Ho, VanKampen, Jackalina, Petrucelli, Leonard, West, Andrew B., Ko, Han Seok, Lee, Yun-Il, Maguire-Zeiss, Kathleen A., Bowers, William J., Federoff, Howard J., Dawson, Valina L., and Dawson, Ted M.
- Subjects
PARKINSON'S disease treatment ,LEUCINE ,GENETIC mutation ,NEURODEGENERATION ,NEURONS - Abstract
Leucine-rich repeat kinase-2 (LRRK2) mutations are a common cause of Parkinson's disease. Here we identify inhibitors of LRRK2 kinase that are protective in in vitro and in vivo models of LRRK2-induced neurodegeneration. These results establish that LRRK2-induced degeneration of neurons in vivo is kinase dependent and that LRRK2 kinase inhibition provides a potential new neuroprotective paradigm for the treatment of Parkinson's disease. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
35. Cryo-EM structures of pathogenic fibrils and their impact on neurodegenerative disease research.
- Author
-
Todd, Tiffany W., Islam, Naeyma N., Cook, Casey N., Caulfield, Thomas R., and Petrucelli, Leonard
- Subjects
- *
NEURODEGENERATION , *DNA-binding proteins , *MEMBRANE proteins , *ALPHA-synuclein , *AMYLOID beta-protein , *TAU proteins , *DISEASE progression , *AMYLOID - Abstract
Neurodegenerative diseases are commonly associated with the formation of aberrant protein aggregates within the brain, and ultrastructural analyses have revealed that the proteins within these inclusions often assemble into amyloid filaments. Cryoelectron microscopy (cryo-EM) has emerged as an effective method for determining the near-atomic structure of these disease-associated filamentous proteins, and the resulting structures have revolutionized the way we think about aberrant protein aggregation and propagation during disease progression. These structures have also revealed that individual fibril conformations may dictate different disease conditions, and this newfound knowledge has improved disease modeling in the lab and advanced the ongoing pursuit of clinical tools capable of distinguishing and targeting different pathogenic entities within living patients. In this review, we summarize some of the recently developed cryo-EM structures of ex vivo α-synuclein, tau, β-amyloid (Aβ), TAR DNA-binding protein 43 (TDP-43), and transmembrane protein 106B (TMEM106B) fibrils and discuss how these structures are being leveraged toward mechanistic research and therapeutic development. Neurodegenerative diseases are often characterized by the deposition of amyloidogenic proteins in the brain. Todd et al. review the structures of disease-associated fibrils as determined by cryoelectron microscopy and discuss how these structures provide mechanistic and translational insight. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Tau exhibits unique seeding properties in globular glial tauopathy.
- Author
-
Chung, Dah-eun Chloe, Carlomagno, Yari, Cook, Casey N., Jansen-West, Karen, Daughrity, Lillian, Lewis-Tuffin, Laura J., Castanedes-Casey, Monica, DeTure, Michael, Dickson, Dennis W., and Petrucelli, Leonard
- Subjects
SOWING ,NEURODEGENERATION ,MICROTUBULES - Abstract
Tauopathies are neurodegenerative disorders characterized by aggregation of microtubule associated tau protein in neurons and glia. They are clinically and pathologically heterogeneous depending on the isoform of tau protein that accumulates (three or four 31-to-32-amino-acid repeats [3R or 4R] in the microtubule binding domain), as well as the cellular and neuroanatomical distribution of tau pathology. Growing evidence suggests that distinct tau conformers may contribute to the characteristic features of various tauopathies. Globular glial tauopathy (GGT) is a rare 4R tauopathy with globular cytoplasmic inclusions within neurons and glial cells. Given the unique cellular distribution and morphology of tau pathology in GGT, we sought to determine if tau species in GGT had distinctive biological properties. To address this question, we performed seeding analyses with postmortem brain tissues using a commercial tau biosensor cell line. We found that brain lysates from GGT cases had significantly higher seeding competency than other tauopathies, including corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), and Alzheimer's disease (AD). The robust seeding activity of GGT brain lysates was independent of phosphorylated tau burden and diminished upon removal of tau from samples, suggesting that seeding properties were indeed mediated by tau in the lysates. In addition, cellular inclusions in the tau biosensor cell line induced by GGT had a distinct, globular morphology that was markedly different from inclusions induced by other tauopathies, further highlighting the unique nature of tau species in GGT. Characterization of different tau species in GGT showed that detergent-insoluble, fibril-like tau contained the highest seeding activity, as reflected in its ability to increase tau aggregation in primary glial cultures. Taken together, our data suggest that unique seeding properties differentiate GGT-tau from other tauopathies, which provides new insight into pathogenic heterogeneity of primary neurodegenerative tauopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
37. Replication of progressive supranuclear palsy genome-wide association study identifies <italic>SLCO1A2</italic> and <italic>DUSP10</italic> as new susceptibility loci.
- Author
-
Sanchez-Contreras, Monica Y., Kouri, Naomi, Cook, Casey N., Serie, Daniel J., Heckman, Michael G., Finch, NiCole A., Caselli, Richard J., Uitti, Ryan J., Wszolek, Zbigniew K., Graff-Radford, Neill, Petrucelli, Leonard, Wang, Li-San, Schellenberg, Gerard D., Dickson, Dennis W., Rademakers, Rosa, and Ross, Owen A.
- Subjects
PROGRESSIVE supranuclear palsy ,COGNITION disorders ,SINGLE nucleotide polymorphisms ,GENETIC polymorphisms ,NEURODEGENERATION ,GENETICS - Abstract
Background: Progressive supranuclear palsy (PSP) is a parkinsonian neurodegenerative tauopathy affecting brain regions involved in motor function, including the basal ganglia, diencephalon and brainstem. While PSP is largely considered to be a sporadic disorder, cases with suspected familial inheritance have been identified and the common MAPT H1haplotype is a major genetic risk factor. Due to the relatively low prevalence of PSP, large sample sizes can be difficult to achieve, and this has limited the ability to detect true genetic risk factors at the genome-wide statistical threshold for significance in GWAS data. With this in mind, in this study we genotyped the genetic variants that displayed the strongest degree of association with PSP (
P <1E-4) in the previous GWAS in a new cohort of 533 pathologically-confirmed PSP cases and 1172 controls, and performed a combined analysis with the previous GWAS data. Results: Our findings validate the known association of loci at MAPT, MOBP, EIF2AK3 and STX6 with risk of PSP, and uncover novel associations with SLCO1A2 (rs11568563) and DUSP10 (rs6687758) variants, both of which were classified as non-significant in the original GWAS. Conclusions: Resolving the genetic architecture of PSP will provide mechanistic insights and nominate candidate genes and pathways for future therapeutic intervention strategies. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
38. The endolysosomal pathway and ALS/FTD.
- Author
-
Todd, Tiffany W., Shao, Wei, Zhang, Yong-jie, and Petrucelli, Leonard
- Subjects
- *
AMYOTROPHIC lateral sclerosis , *FRONTOTEMPORAL dementia , *DNA-binding proteins , *RILUZOLE , *MEMBRANE proteins , *NEURODEGENERATION , *GLYCOGEN storage disease type II - Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD) patient cells and tissue show evidence of a disrupted endolysosomal system, including enlarged endosomes and changes in the expression of endolysosomal genes. The discovery of transmembrane protein 106B (TMEM106B) filaments in insoluble brain extracts from several neurodegenerative disorders further highlights the potential importance of the lysosome in aging and disease. Multiple disease-associated genes play a role in the endolysosomal system, and emerging evidence suggests these roles may be particularly important for pathogenesis. Although autophagy is intrinsically linked to the endolysosomal system, some studies suggest that inhibiting this clearance pathway alone is insufficient to induce TAR DNA-binding protein 43 (TDP-43) proteinopathy in laboratory models. However, inhibiting the endolysosomal system does induce TDP-43 inclusion formation in cells. The non-autophagy-associated functions of this pathway in ALS/FTD should be further explored. Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are considered to be part of a disease spectrum that is associated with causative mutations and risk variants in a wide range of genes. Mounting evidence indicates that several of these genes are linked to the endolysosomal system, highlighting the importance of this pathway in ALS/FTD. Although many studies have focused on how disruption of this pathway impacts on autophagy, recent findings reveal that this may not be the whole picture: specifically, disrupting autophagy may not be sufficient to induce disease, whereas disrupting the endolysosomal system could represent a crucial pathogenic driver. In this review we discuss the connections between ALS/FTD and the endolysosomal system, including a breakdown of how disease-associated genes are implicated in this pathway. We also explore the potential downstream consequences of disrupting endolysosomal activity in the brain, outside of an effect on autophagy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
39. RNA-mediated toxicity in neurodegenerative disease.
- Author
-
Belzil, Veronique V., Gendron, Tania F., and Petrucelli, Leonard
- Subjects
- *
NEURODEGENERATION , *DNA-binding proteins , *GENETIC code , *GENETIC transcription , *ANIMAL models in research , *NEUROTOXICOLOGY - Abstract
Abstract: Cellular viability depends upon the well-orchestrated functions carried out by numerous protein-coding and non-coding RNAs, as well as RNA-binding proteins. During the last decade, it has become increasingly evident that abnormalities in RNA processing represent a common feature among many neurodegenerative diseases. In “RNAopathies”, which include diseases caused by non-coding repeat expansions, RNAs exert toxicity via diverse mechanisms: RNA foci formation, bidirectional transcription, and the production of toxic RNAs and proteins by repeat associated non-ATG translation. The mechanisms of toxicity in “RNA-binding proteinopathies”, diseases in which RNA-binding proteins like TDP-43 and FUS play a prominent role, have yet to be fully elucidated. Nonetheless, both loss of function of the RNA binding protein, and a toxic gain of function resulting from its aggregation, are thought to be involved in disease pathogenesis. As part of the special issue on RNA and Splicing Regulation in Neurodegeneration, this review intends to explore the diverse RNA-related mechanisms contributing to neurodegeneration, with a special emphasis on findings emerging from animal models. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
40. Linking the VPS35 and EIF4G1 Pathways in Parkinson’s Disease.
- Author
-
Ross, Owen A., Cook, Casey, and Petrucelli, Leonard
- Subjects
- *
TRANSLATION initiation factors (Biochemistry) , *PARKINSON'S disease treatment , *TARGETED drug delivery , *DRUG development , *SYNUCLEINS , *NEURODEGENERATION - Abstract
Elucidating the underlying pathogenic pathways in Parkinson’s disease will be critical for targeted drug development. In this issue of Neuron , Dhungel et al. (2015) utilize a yeast model to establish a link between VPS35 and EIF4G1 in α-synuclein-related neurodegeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
41. An acetylation-phosphorylation switch that regulates tau aggregation propensity and function.
- Author
-
Carlomagno, Yari, Dah-eun Chloe Chung, Mei Yue, Castanedes-Casey, Monica, Madden, Benjamin J., Dunmore, Judy, Jimei Tong, DeTure, Michael, Dickson, Dennis W., Petrucelli, Leonard, and Cook, Casey
- Subjects
- *
TAU proteins , *ACETYLATION , *POST-translational modification , *NEURODEGENERATION , *HISTONE deacetylase - Abstract
The aberrant accumulation of tau protein is a pathological hallmark of a class of neurodegenerative diseases known as tauopathies, including Alzheimer's disease and related dementias. On the basis of previous observations that tau is a direct substrate of histone deacetylase 6 (HDAC6), we sought to map all HDAC6-responsive sites in tau and determine how acetylation in a site-specific manner affects tau's biophysical properties in vitro. Our findings indicate that several acetylation sites in tau are responsive to HDAC6 and that acetylation on Lys-321 (within aKCGSmotif) is both essential for acetylation-mediated inhibition of tau aggregation in vitro and a molecular tactic for preventing phosphorylation on the downstream Ser-324 residue. To determine the functional consequence of this HDAC6-regulated phosphorylation event, we examined tau's ability to promote microtubule assembly and found that phosphorylation of Ser-324 interferes with the normal microtubule-stabilizing function of tau. Tau phosphorylation of Ser-324 (pSer-324) has not previously been evaluated in the context of tauopathy, and here we observed increased deposition of pSer-324-positive tau both in mouse models of tauopathy and in patients with Alzheimer's disease. These findings uncover a novel acetylation-phosphorylation switch at Lys-321/Ser-324 that coordinately regulates tau polymerization and function. Because the disease relevance of this finding is evident, additional studies are needed to examine the role of pSer-324 in tau pathobiology and to determine whether therapeutically modulating this acetylation-phosphorylation switch affects disease progression in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
42. Gasdermin-E mediates mitochondrial damage in axons and neurodegeneration.
- Author
-
Neel, Dylan V., Basu, Himanish, Gunner, Georgia, Bergstresser, Matthew D., Giadone, Richard M., Chung, Haeji, Miao, Rui, Chou, Vicky, Brody, Eliza, Jiang, Xin, Lee, Edward, Watts, Michelle E., Marques, Christine, Held, Aaron, Wainger, Brian, Lagier-Tourenne, Clotilde, Zhang, Yong-Jie, Petrucelli, Leonard, Young-Pearse, Tracy L., and Chen-Plotkin, Alice S.
- Subjects
- *
CELL death , *MOTOR neuron diseases , *AXONS , *CENTRAL nervous system , *AMYOTROPHIC lateral sclerosis , *MOTOR neurons - Abstract
Mitochondrial dysfunction and axon loss are hallmarks of neurologic diseases. Gasdermin (GSDM) proteins are executioner pore-forming molecules that mediate cell death, yet their roles in the central nervous system (CNS) are not well understood. Here, we find that one GSDM family member, GSDME, is expressed by both mouse and human neurons. GSDME plays a role in mitochondrial damage and axon loss. Mitochondrial neurotoxins induced caspase-dependent GSDME cleavage and rapid localization to mitochondria in axons, where GSDME promoted mitochondrial depolarization, trafficking defects, and neurite retraction. Frontotemporal dementia (FTD)/amyotrophic lateral sclerosis (ALS)-associated proteins TDP-43 and PR-50 induced GSDME-mediated damage to mitochondria and neurite loss. GSDME knockdown protected against neurite loss in ALS patient iPSC-derived motor neurons. Knockout of GSDME in SOD1G93A ALS mice prolonged survival, ameliorated motor dysfunction, rescued motor neuron loss, and reduced neuroinflammation. We identify GSDME as an executioner of neuronal mitochondrial dysfunction that may contribute to neurodegeneration. [Display omitted] • GSDME is expressed by neurons and activated by mitochondrial toxins • Neuronal GSDME drives local mitochondrial damage and axon loss prior to cell death • ALS/FTD proteins activate GSDME and drive neurite loss in mouse and human neurons • SOD1G93A GSDME KO mice have rescued disease progression and motor neuron loss Gasdermin-E is a pore-forming molecule that is expressed in neurons. When activated, this molecule rapidly targets and destroys mitochondria. In neurons, toxin exposure activates GSDME and drives mitochondrial and axonal destruction. The genetic inhibition of GSDME rescues axon loss in amyotrophic lateral sclerosis (ALS) patient-derived motor neurons and ameliorates disease in a mouse model of ALS. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
43. C9orf72 BAC Transgenic Mice Display Typical Pathologic Features of ALS/FTD.
- Author
-
O’Rourke, Jacqueline G., Bogdanik, Laurent, Muhammad, A.K.M.G., Gendron, Tania F., Kim, Kevin J., Austin, Andrew, Cady, Janet, Liu, Elaine Y., Zarrow, Jonah, Grant, Sharday, Ho, Ritchie, Bell, Shaughn, Carmona, Sharon, Simpkinson, Megan, Lall, Deepti, Wu, Kathryn, Daughrity, Lillian, Dickson, Dennis W., Harms, Matthew B., and Petrucelli, Leonard
- Subjects
- *
TRANSGENIC mice , *AMYOTROPHIC lateral sclerosis , *RNA analysis , *NEURODEGENERATION , *DIPEPTIDES - Abstract
Summary Noncoding expansions of a hexanucleotide repeat (GGGGCC) in the C9orf72 gene are the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia. Here we report transgenic mice carrying a bacterial artificial chromosome (BAC) containing the full human C9orf72 gene with either a normal allele (15 repeats) or disease-associated expansion (∼100–1,000 repeats; C9-BACexp). C9-BACexp mice displayed pathologic features seen in C9orf72 expansion patients, including widespread RNA foci and repeat-associated non-ATG (RAN) translated dipeptides, which were suppressed by antisense oligonucleotides targeting human C9orf72 . Nucleolin distribution was altered, supporting that either C9orf72 transcripts or RAN dipeptides promote nucleolar dysfunction. Despite early and widespread production of RNA foci and RAN dipeptides in C9-BACexp mice, behavioral abnormalities and neurodegeneration were not observed even at advanced ages, supporting the hypothesis that RNA foci and RAN dipeptides occur presymptomatically and are not sufficient to drive neurodegeneration in mice at levels seen in patients. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
44. Characterization of DNA hypermethylation in the cerebellum of c9FTD/ALS patients.
- Author
-
Belzil, Veronique V., Bauer, Peter O., Gendron, Tania F., Murray, Melissa E., Dickson, Dennis, and Petrucelli, Leonard
- Subjects
- *
DNA methylation , *AMYOTROPHIC lateral sclerosis , *FRONTOTEMPORAL dementia , *NUCLEOTIDE sequence , *GENE expression , *NEURODEGENERATION , *LYSINE , *PATIENTS - Abstract
A significant number of patients suffering from amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two diseases commonly seen in comorbidity, carry an expanded noncoding hexanucleotide repeat in the C9orf72 gene, a condition collectively referred to as c9FTD/ALS. Repeat expansions, also present in other neurodegenerative diseases, have been shown to alter epigenetic mechanisms and consequently lead to decreased gene expression, while also leading to toxic RNA gain-of-function. As expression of multiple C9orf72 transcript variants is known to be reduced in c9FTD/ALS cases, our group and others have sought to uncover the mechanisms causing this reduction. We recently demonstrated that histones H3 and H4 undergo trimethylation at lysines 9 (H3K9), 27 (H3K27), 79 (H3K79), and 20 (H4K20) in all pathogenic repeat carrier brain samples, confirming the role of altered histone methylation in disease. It was also reported that about 40% of c9ALS cases show hypermethylation of the CpG island located at the 5' end of the repeat expansion in blood, frontal cortex, and spinal cord. To determine whether the same CpG island is hypermethylated in the cerebella of cases in whom aberrant histone methylation has been identified, we bisulfite-modified the extracted DNA and PCR-amplified 26 CpG sites within the C9orf72 promoter region. Among the ten c9FTD/ALS (4 c9ALS, 6 c9FTD), nine FTD/ALS, and eight disease control samples evaluated, only one c9FTD sample was found to be hypermethylated within the C9orf72 promoter region. This study is the first to report cerebellar hypermethylation in c9FTD/ALS, and the first to identify a c9FTD patient with aberrant DNA methylation. Future studies will need to evaluate hypermethylation of the C9orf72 promoter in a larger cohort of c9FTD patients, and to assess whether DNA methylation variation across brain regions reflects disease phenotype. This article is part of a Special Issue entitled RNA Metabolism 2013 . [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
45. Severe amygdala dysfunction in a MAPT transgenic mouse model of frontotemporal dementia.
- Author
-
Cook, Casey, Dunmore, Judy H., Murray, Melissa E., Scheffel, Kristyn, Shukoor, Nawsheen, Tong, Jimei, Castanedes-Casey, Monica, Phillips, Virginia, Rousseau, Linda, Penuliar, Michael S., Kurti, Aishe, Dickson, Dennis W., Petrucelli, Leonard, and Fryer, John D.
- Subjects
- *
AMYGDALOID body , *TRANSGENIC mice , *FRONTOTEMPORAL dementia , *PARKINSONIAN disorders , *CHROMOSOMES , *NEURODEGENERATION - Abstract
Abstract: Frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17) is a neurodegenerative tauopathy caused by mutations in the tau gene (MAPT). Individuals with FTDP-17 have deficits in learning, memory, and language, in addition to personality and behavioral changes that are often characterized by a lack of social inhibition. Several transgenic mouse models expressing tau mutations have been tested extensively for memory or motor impairments, though reports of amygdala-dependent behaviors are lacking. To this end, we tested the rTg4510 mouse model on a behavioral battery that included amygdala-dependent tasks of exploration. As expected, rTg4510 mice exhibit profound impairments in hippocampal-dependent learning and memory tests, including contextual fear conditioning. However, rTg4510 mice also display an abnormal hyperexploratory phenotype in the open-field assay, elevated plus maze, light-dark exploration, and cued fear conditioning, indicative of amygdala dysfunction. Furthermore, significant tau burden is detected in the amygdala of both rTg4510 mice and human FTDP-17 patients, suggesting that the rTg4510 mouse model recapitulates the behavioral disturbances and neurodegeneration of the amygdala characteristic of FTDP-17. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
46. Homotypic fibrillization of TMEM106B across diverse neurodegenerative diseases.
- Author
-
Chang, Andrew, Xiang, Xinyu, Wang, Jing, Lee, Carolyn, Arakhamia, Tamta, Simjanoska, Marija, Wang, Chi, Carlomagno, Yari, Zhang, Guoan, Dhingra, Shikhar, Thierry, Manon, Perneel, Jolien, Heeman, Bavo, Forgrave, Lauren M., DeTure, Michael, DeMarco, Mari L., Cook, Casey N., Rademakers, Rosa, Dickson, Dennis W., and Petrucelli, Leonard
- Subjects
- *
NEURODEGENERATION , *LEWY body dementia , *DEGENERATION (Pathology) , *PROGRESSIVE supranuclear palsy , *FRONTOTEMPORAL lobar degeneration , *PENNING trap mass spectrometry , *FORENSIC pathology - Abstract
Misfolding and aggregation of disease-specific proteins, resulting in the formation of filamentous cellular inclusions, is a hallmark of neurodegenerative disease with characteristic filament structures, or conformers, defining each proteinopathy. Here we show that a previously unsolved amyloid fibril composed of a 135 amino acid C-terminal fragment of TMEM106B is a common finding in distinct human neurodegenerative diseases, including cases characterized by abnormal aggregation of TDP-43, tau, or α-synuclein protein. A combination of cryoelectron microscopy and mass spectrometry was used to solve the structures of TMEM106B fibrils at a resolution of 2.7 Å from postmortem human brain tissue afflicted with frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP, n = 8), progressive supranuclear palsy (PSP, n = 2), or dementia with Lewy bodies (DLB, n = 1). The commonality of abundant amyloid fibrils composed of TMEM106B, a lysosomal/endosomal protein, to a broad range of debilitating human disorders indicates a shared fibrillization pathway that may initiate or accelerate neurodegeneration. [Display omitted] • Cryo-EM structures of brain-derived TMEM106B fibrils from neurodegenerative diseases • Endolysosomal membrane protein TMEM106B C-terminal fragment forms amyloid fibrils • TMEM106B fibrillization is widespread among diverse neurodegenerative proteinopathies • Identification of fibrillization pathway potentially implicated in neurodegeneration Cryo-EM and mass spectrometry-based proteomics of insoluble amyloid fibrils derived from postmortem human brains afflicted with diverse neurodegenerative diseases reveals widespread fibrillization of an endolysosomal membrane protein, TMEM106B, pointing toward a potentially pathogenic commonality between distinct proteinopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
47. Unconventional Translation of C9ORF72 GGGGCC Expansion Generates Insoluble Polypeptides Specific to c9FTD/ALS
- Author
-
Ash, Peter E.A., Bieniek, Kevin F., Gendron, Tania F., Caulfield, Thomas, Lin, Wen-Lang, DeJesus-Hernandez, Mariely, van Blitterswijk, Marka M., Jansen-West, Karen, Paul, Joseph W., Rademakers, Rosa, Boylan, Kevin B., Dickson, Dennis W., and Petrucelli, Leonard
- Subjects
- *
GENETIC translation , *POLYPEPTIDES , *FRONTOTEMPORAL dementia , *NEURODEGENERATION , *GENETICS of amyotrophic lateral sclerosis , *NUCLEOTIDE sequence , *BIOMARKERS - Abstract
Summary: Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are devastating neurodegenerative disorders with clinical, genetic, and neuropathological overlap. Hexanucleotide (GGGGCC) repeat expansions in a noncoding region of C9ORF72 are the major genetic cause of FTD and ALS (c9FTD/ALS). The RNA structure of GGGGCC repeats renders these transcripts susceptible to an unconventional mechanism of translation—repeat-associated non-ATG (RAN) translation. Antibodies generated against putative GGGGCC repeat RAN-translated peptides (anti-C9RANT) detected high molecular weight, insoluble material in brain homogenates, and neuronal inclusions throughout the CNS of c9FTD/ALS cases. C9RANT immunoreactivity was not found in other neurodegenerative diseases, including CAG repeat disorders, or in peripheral tissues of c9FTD/ALS. The specificity of C9RANT for c9FTD/ALS is a potential biomarker for this most common cause of FTD and ALS. These findings have significant implications for treatment strategies directed at RAN-translated peptides and their aggregation and the RNA structures necessary for their production. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
48. Progranulin: An emerging target for FTLD therapies
- Author
-
Gass, Jennifer, Prudencio, Mercedes, Stetler, Caroline, and Petrucelli, Leonard
- Subjects
- *
PROGRANULIN , *NEURODEGENERATION , *FRONTOTEMPORAL dementia , *GENETIC mutation , *EMBRYOLOGY , *INFLAMMATION - Abstract
Abstract: Frontotemporal lobar degeneration (FTLD), a neurodegenerative disease primarily affecting the frontal and temporal lobes, is one of the most common types of dementia. While the majority of FTLD cases are sporadic, approximately 10–40% of patients have an inherited form of FTLD. Mutations in the progranulin gene (GRN) have recently been identified as a major cause of FTLD with ubiquitin positive inclusions (FTLD-U). Because over 70 disease-linked GRN mutations cause abnormal deficiencies in the production of PGRN, a protein that plays a crucial role in embryogenesis, cell growth and survival, wound repair and inflammation, researchers now aim to design therapies that would increase PGRN levels in affected individuals, thereby alleviating the symptoms associated with disease. Several compounds and genetic factors, as well as PGRN receptors, have recently been identified because of their ability to regulate PGRN levels. Strict quality control measures are needed given that extreme PGRN levels at either end of the spectrum – too low or too high – can lead to neurodegeneration or cancer, respectively. The aim of this review is to highlight what is known regarding PGRN biology; to improve understanding of the mechanisms involved in regulating PGRN levels and highlight studies that are laying the groundwork for the development of effective therapeutic modulators of PGRN. This article is part of a Special Issue entitled RNA-Binding Proteins. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
49. Identification of potential protein interactors of Lrrk2
- Author
-
Dächsel, Justus C., Taylor, Julie P., Mok, Su San, Ross, Owen A., Hinkle, Kelly M., Bailey, Rachel M., Hines, Jacob H., Szutu, Jennifer, Madden, Benjamin, Petrucelli, Leonard, Farrer, Matthew J., and Dächsel, Justus C
- Subjects
- *
PARKINSON'S disease , *NEURODEGENERATION , *PROTEIN binding , *MOLECULAR chaperones - Abstract
Abstract: Pathogenic substitutions in the Lrrk2 protein have been shown to be an important cause of both familial and sporadic parkinsonism. The molecular pathway involved in Lrrk2 dopaminergic neuron degeneration remains elusive. Employing a combination of Lrrk2-mediated protein precipitation and tandem mass spectrometry, we identified 14 potential Lrrk2 binding partners. The majority of these interactions may be subgrouped into three functional cellular pathways: (i) chaperone-mediated response, (ii) proteins associated with the cytoskeleton and trafficking and (iii) phosphorylation and kinase activity. Future investigation of these candidates is now warranted and may help resolve the pathomechanism behind Lrrk2 neurodegeneration. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
50. The high-affinity HSP90-CHIP complex recognizes and selectively degrades phosphorylated tau client proteins.
- Author
-
Dickey, Chad A., Kamal, Adeela, Lundgren, Karen, Klosak, Natalia, Bailey, Rachel M., Dunmore, Judith, Ash, Peter, Shoraka, Sareh, Zlatkovic, Jelena, Eckman, Christopher B., Patterson, Cam, Dennis W. Dickson,1, Nahman Jr., N. Stanley, Hutton, Michael, Burrows, Francis, Petrucelli, Leonard, Dickson, Dennis W, and Nahman, N Stanley Jr
- Subjects
- *
ALZHEIMER'S disease , *NEURODEGENERATION , *UBIQUITIN , *MOLECULAR chaperones , *ADENOSINE triphosphatase , *LABORATORY mice - Abstract
A primary pathologic component of Alzheimer's disease (AD) is the formation of neurofibrillary tangles composed of hyperphosphorylated tau (p-tau). Expediting the removal of these p-tau species may be a relevant therapeutic strategy. Here we report that inhibition of Hsp90 led to decreases in p-tau levels independent of heat shock factor 1 (HSF1) activation. A critical mediator of this mechanism was carboxy terminus of Hsp70-interacting protein (CHIP), a tau ubiquitin ligase. Cochaperones were also involved in Hsp90-mediated removal of p-tau, while those of the mature Hsp90 refolding complex prevented this effect. This is the first demonstration to our knowledge that blockade of the refolding pathway promotes p-tau turnover through degradation. We also show that peripheral administration of a novel Hsp90 inhibitor promoted selective decreases in p-tau species in a mouse model of tauopathy, further suggesting a central role for the Hsp90 complex in the pathogenesis of tauopathies. When taken in the context of known high-affinity Hsp90 complexes in affected regions of the AD brain, these data implicate a central role for Hsp90 in the development of AD and other tauopathies and may provide a rationale for the development of novel Hsp90-based therapeutic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.