149 results on '"Tania F. Gendron"'
Search Results
2. HDGFL2 cryptic proteins report presence of TDP-43 pathology in neurodegenerative diseases
- Author
-
Anna Calliari, Lillian M. Daughrity, Ellen A. Albagli, Paula Castellanos Otero, Mei Yue, Karen Jansen-West, Naeyma N. Islam, Thomas Caulfield, Bailey Rawlinson, Michael DeTure, Casey Cook, Neill R. Graff-Radford, Gregory S. Day, Bradley F. Boeve, David S. Knopman, Ronald C. Petersen, Keith A. Josephs, Björn Oskarsson, Aaron D. Gitler, Dennis W. Dickson, Tania F. Gendron, Mercedes Prudencio, Michael E. Ward, Yong-Jie Zhang, and Leonard Petrucelli
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract This letter demonstrates the potential of novel cryptic proteins resulting from TAR DNA-binding protein 43 (TDP-43) dysfunction as markers of TDP-43 pathology in neurodegenerative diseases.
- Published
- 2024
- Full Text
- View/download PDF
3. Fluid biomarkers for amyotrophic lateral sclerosis: a review
- Author
-
Katherine E. Irwin, Udit Sheth, Philip C. Wong, and Tania F. Gendron
- Subjects
Amyotrophic lateral sclerosis ,Fluid biomarkers ,Cerebrospinal fluid ,Clinical trial ,Neurofilament ,Plasma ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of upper and lower motor neurons. Presently, three FDA-approved drugs are available to help slow functional decline for patients with ALS, but no cure yet exists. With an average life expectancy of only two to five years after diagnosis, there is a clear need for biomarkers to improve the care of patients with ALS and to expedite ALS treatment development. Here, we provide a review of the efforts made towards identifying diagnostic, prognostic, susceptibility/risk, and response fluid biomarkers with the intent to facilitate a more rapid and accurate ALS diagnosis, to better predict prognosis, to improve clinical trial design, and to inform interpretation of clinical trial results. Over the course of 20 + years, several promising fluid biomarker candidates for ALS have emerged. These will be discussed, as will the exciting new strategies being explored for ALS biomarker discovery and development.
- Published
- 2024
- Full Text
- View/download PDF
4. Correction: HDGFL2 cryptic proteins report presence of TDP-43 pathology in neurodegenerative diseases
- Author
-
Anna Calliari, Lillian M. Daughrity, Ellen A. Albagli, Paula Castellanos Otero, Mei Yue, Karen Jansen-West, Naeyma N. Islam, Thomas Caulfield, Bailey Rawlinson, Michael DeTure, Casey Cook, Neill R. Graff-Radford, Gregory S. Day, Bradley F. Boeve, David S. Knopman, Ronald C. Petersen, Keith A. Josephs, Björn Oskarsson, Aaron D. Gitler, Dennis W. Dickson, Tania F. Gendron, Mercedes Prudencio, Michael E. Ward, Yong-Jie Zhang, and Leonard Petrucelli
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Published
- 2024
- Full Text
- View/download PDF
5. Neurofilament light chain and vaccination status associate with clinical outcomes in severe COVID-19
- Author
-
Young Erben, Mercedes Prudencio, Christopher P. Marquez, Karen R. Jansen-West, Michael G. Heckman, Launia J. White, Judith A. Dunmore, Casey N. Cook, Meredith T. Lilley, Neda Qosja, Yuping Song, Rana Hanna Al Shaikh, Lillian M. Daughrity, Jordan L. Bartfield, Gregory S. Day, Björn Oskarsson, Katharine A. Nicholson, Zbigniew K. Wszolek, Jonathan B. Hoyne, Tania F. Gendron, James F. Meschia, and Leonard Petrucelli
- Subjects
Science - Published
- 2024
- Full Text
- View/download PDF
6. Roadmap for C9ORF72 in Frontotemporal Dementia and Amyotrophic Lateral Sclerosis: Report on the C9ORF72 FTD/ALS Summit
- Author
-
Rita Sattler, Bryan J. Traynor, Janice Robertson, Ludo Van Den Bosch, Sami J. Barmada, Clive N. Svendsen, Matthew D. Disney, Tania F. Gendron, Philip C. Wong, Martin R. Turner, Adam Boxer, Suma Babu, Michael Benatar, Michael Kurnellas, Jonathan D. Rohrer, Christopher J. Donnelly, Lynette M. Bustos, Kendall Van Keuren-Jensen, Penny A. Dacks, Marwan N. Sabbagh, and Attendees of the inaugural C9ORF72 FTD/ALS Summit
- Subjects
Biomarker ,C9ORF72 ,Dipeptide repeat protein ,Gene therapy ,Neurofilament ,Prevention ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract A summit held March 2023 in Scottsdale, Arizona (USA) focused on the intronic hexanucleotide expansion in the C9ORF72 gene and its relevance in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS; C9ORF72-FTD/ALS). The goal of this summit was to connect basic scientists, clinical researchers, drug developers, and individuals affected by C9ORF72-FTD/ALS to evaluate how collaborative efforts across the FTD-ALS disease spectrum might break down existing disease silos. Presentations and discussions covered recent discoveries in C9ORF72-FTD/ALS disease mechanisms, availability of disease biomarkers and recent advances in therapeutic development, and clinical trial design for prevention and treatment for individuals affected by C9ORF72-FTD/ALS and asymptomatic pathological expansion carriers. The C9ORF72-associated hexanucleotide repeat expansion is an important locus for both ALS and FTD. C9ORF72-FTD/ALS may be characterized by loss of function of the C9ORF72 protein and toxic gain of functions caused by both dipeptide repeat (DPR) proteins and hexanucleotide repeat RNA. C9ORF72-FTD/ALS therapeutic strategies discussed at the summit included the use of antisense oligonucleotides, adeno-associated virus (AAV)-mediated gene silencing and gene delivery, and engineered small molecules targeting RNA structures associated with the C9ORF72 expansion. Neurofilament light chain, DPR proteins, and transactive response (TAR) DNA-binding protein 43 (TDP-43)–associated molecular changes were presented as biomarker candidates. Similarly, brain imaging modalities (i.e., magnetic resonance imaging [MRI] and positron emission tomography [PET]) measuring structural, functional, and metabolic changes were discussed as important tools to monitor individuals affected with C9ORF72-FTD/ALS, at both pre-symptomatic and symptomatic disease stages. Finally, summit attendees evaluated current clinical trial designs available for FTD or ALS patients and concluded that therapeutics relevant to FTD/ALS patients, such as those specifically targeting C9ORF72, may need to be tested with composite endpoints covering clinical symptoms of both FTD and ALS. The latter will require novel clinical trial designs to be inclusive of all patient subgroups spanning the FTD/ALS spectrum.
- Published
- 2023
- Full Text
- View/download PDF
7. Targeting the glycine-rich domain of TDP-43 with antibodies prevents its aggregation in vitro and reduces neurofilament levels in vivo
- Author
-
Henrick Riemenschneider, Francesca Simonetti, Udit Sheth, Eszter Katona, Stefan Roth, Saskia Hutten, Daniel Farny, Meike Michaelsen, Brigitte Nuscher, Michael K. Schmidt, Andrew Flatley, Aloys Schepers, Lara A. Gruijs da Silva, Qihui Zhou, Thomas Klopstock, Arthur Liesz, Thomas Arzberger, Jochen Herms, Regina Feederle, Tania F. Gendron, Dorothee Dormann, and Dieter Edbauer
- Subjects
Neurodegeneration ,Amyotrophic lateral sclerosis ,Frontotemporal dementia ,TDP-43 ,Immunotherapy ,Phase separation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Cytoplasmic aggregation and concomitant nuclear clearance of the RNA-binding protein TDP-43 are found in ~ 90% of cases of amyotrophic lateral sclerosis and ~ 45% of patients living with frontotemporal lobar degeneration, but no disease-modifying therapy is available. Antibody therapy targeting other aggregating proteins associated with neurodegenerative disorders has shown beneficial effects in animal models and clinical trials. The most effective epitopes for safe antibody therapy targeting TDP-43 are unknown. Here, we identified safe and effective epitopes in TDP-43 for active and potential future passive immunotherapy. We prescreened 15 peptide antigens covering all regions of TDP-43 to identify the most immunogenic epitopes and to raise novel monoclonal antibodies in wild-type mice. Most peptides induced a considerable antibody response and no antigen triggered obvious side effects. Thus, we immunized mice with rapidly progressing TDP-43 proteinopathy (“rNLS8” model) with the nine most immunogenic peptides in five pools prior to TDP-43ΔNLS transgene induction. Strikingly, combined administration of two N-terminal peptides induced genetic background-specific sudden lethality in several mice and was therefore discontinued. Despite a strong antibody response, no TDP-43 peptide prevented the rapid body weight loss or reduced phospho-TDP-43 levels as well as the profound astrogliosis and microgliosis in rNLS8 mice. However, immunization with a C-terminal peptide containing the disease-associated phospho-serines 409/410 significantly lowered serum neurofilament light chain levels, indicative of reduced neuroaxonal damage. Transcriptomic profiling showed a pronounced neuroinflammatory signature (IL-1β, TNF-α, NfκB) in rNLS8 mice and suggested modest benefits of immunization targeting the glycine-rich region. Several novel monoclonal antibodies targeting the glycine-rich domain potently reduced phase separation and aggregation of TDP-43 in vitro and prevented cellular uptake of preformed aggregates. Our unbiased screen suggests that targeting the RRM2 domain and the C-terminal region of TDP-43 by active or passive immunization may be beneficial in TDP-43 proteinopathies by inhibiting cardinal processes of disease progression. Graphical Abstract
- Published
- 2023
- Full Text
- View/download PDF
8. CRISPR/Cas9-mediated excision of ALS/FTD-causing hexanucleotide repeat expansion in C9ORF72 rescues major disease mechanisms in vivo and in vitro
- Author
-
Katharina E. Meijboom, Abbas Abdallah, Nicholas P. Fordham, Hiroko Nagase, Tomás Rodriguez, Carolyn Kraus, Tania F. Gendron, Gopinath Krishnan, Rustam Esanov, Nadja S. Andrade, Matthew J. Rybin, Melina Ramic, Zachary D. Stephens, Alireza Edraki, Meghan T. Blackwood, Aydan Kahriman, Nils Henninger, Jean-Pierre A. Kocher, Michael Benatar, Michael H. Brodsky, Leonard Petrucelli, Fen-Biao Gao, Erik J. Sontheimer, Robert H. Brown, Zane Zeier, and Christian Mueller
- Subjects
Science - Abstract
A hexanucleotide repeat expansion in C9ORF72 is the most common genetic cause of ALS and FTD. Here, the authors demonstrate CRISPR/Cas9 excision of the expansion results in a rescue of disease mechanisms in vivo and in vitro.
- Published
- 2022
- Full Text
- View/download PDF
9. Poly(GR) interacts with key stress granule factors promoting its assembly into cytoplasmic inclusions
- Author
-
Jinyoung Park, Yanwei Wu, Wei Shao, Tania F. Gendron, Sophie J.F. van der Spek, Grigorii Sultanakhmetov, Avik Basu, Paula Castellanos Otero, Caroline J. Jones, Karen Jansen-West, Lillian M. Daughrity, Sadhna Phanse, Giulia del Rosso, Jimei Tong, Monica Castanedes-Casey, Lulu Jiang, Jenna Libera, Björn Oskarsson, Dennis W. Dickson, David W. Sanders, Clifford P. Brangwynne, Andrew Emili, Benjamin Wolozin, Leonard Petrucelli, and Yong-Jie Zhang
- Subjects
CP: Neuroscience ,CP: Molecular biology ,Biology (General) ,QH301-705.5 - Abstract
Summary: C9orf72 repeat expansions are the most common genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Poly(GR) proteins are toxic to neurons by forming cytoplasmic inclusions that sequester RNA-binding proteins including stress granule (SG) proteins. However, little is known of the factors governing poly(GR) inclusion formation. Here, we show that poly(GR) infiltrates a finely tuned network of protein-RNA interactions underpinning SG formation. It interacts with G3BP1, the key driver of SG assembly and a protein we found is critical for poly(GR) inclusion formation. Moreover, we discovered that N6-methyladenosine (m6A)-modified mRNAs and m6A-binding YTHDF proteins not only co-localize with poly(GR) inclusions in brains of c9FTD/ALS mouse models and patients with c9FTD, they promote poly(GR) inclusion formation via the incorporation of RNA into the inclusions. Our findings thus suggest that interrupting interactions between poly(GR) and G3BP1 or YTHDF1 proteins or decreasing poly(GR) altogether represent promising therapeutic strategies to combat c9FTD/ALS pathogenesis.
- Published
- 2023
- Full Text
- View/download PDF
10. Moderate intrinsic phenotypic alterations in C9orf72 ALS/FTD iPSC-microglia despite the presence of C9orf72 pathological features
- Author
-
Ileana Lorenzini, Eric Alsop, Jennifer Levy, Lauren M. Gittings, Deepti Lall, Benjamin E. Rabichow, Stephen Moore, Ryan Pevey, Lynette M. Bustos, Camelia Burciu, Divya Bhatia, Mo Singer, Justin Saul, Amanda McQuade, Makis Tzioras, Thomas A. Mota, Amber Logemann, Jamie Rose, Sandra Almeida, Fen-Biao Gao, Michael Marks, Christopher J. Donnelly, Elizabeth Hutchins, Shu-Ting Hung, Justin Ichida, Robert Bowser, Tara Spires-Jones, Mathew Blurton-Jones, Tania F. Gendron, Robert H. Baloh, Kendall Van Keuren-Jensen, and Rita Sattler
- Subjects
amyotrophic lateral sclerosis ,C9orf72 ,frontotemporal dementia ,iPSC-microglia ,neuroinflammation ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
While motor and cortical neurons are affected in C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), it remains largely unknown if and how non-neuronal cells induce or exacerbate neuronal damage. We differentiated C9orf72 ALS/FTD patient-derived induced pluripotent stem cells into microglia (iPSC-MG) and examined their intrinsic phenotypes. Similar to iPSC motor neurons, C9orf72 ALS/FTD iPSC-MG mono-cultures form G4C2 repeat RNA foci, exhibit reduced C9orf72 protein levels, and generate dipeptide repeat proteins. Healthy control and C9orf72 ALS/FTD iPSC-MG equally express microglial specific genes and perform microglial functions, including inflammatory cytokine release and phagocytosis of extracellular cargos, such as synthetic amyloid beta peptides and healthy human brain synaptoneurosomes. RNA sequencing analysis revealed select transcriptional changes of genes associated with neuroinflammation or neurodegeneration in diseased microglia yet no significant differentially expressed microglial-enriched genes. Moderate molecular and functional differences were observed in C9orf72 iPSC-MG mono-cultures despite the presence of C9orf72 pathological features suggesting that a diseased microenvironment may be required to induce phenotypic changes in microglial cells and the associated neuronal dysfunction seen in C9orf72 ALS/FTD neurodegeneration.
- Published
- 2023
- Full Text
- View/download PDF
11. Evidence of cerebellar TDP-43 loss of function in FTLD-TDP
- Author
-
Sarah Pickles, Tania F. Gendron, Yuka Koike, Mei Yue, Yuping Song, Jennifer M. Kachergus, J. Shi, Michael DeTure, E. Aubrey Thompson, Björn Oskarsson, Neill R. Graff-Radford, Bradley F. Boeve, Ronald C. Petersen, Zbigniew K. Wszolek, Keith A. Josephs, Dennis W. Dickson, Leonard Petrucelli, Casey N. Cook, and Mercedes Prudencio
- Subjects
Cerebellum ,Frontotemporal lobar degeneration ,Stathmin-2 ,TDP-43 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
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.
- Published
- 2022
- Full Text
- View/download PDF
12. A C. elegans model of C9orf72-associated ALS/FTD uncovers a conserved role for eIF2D in RAN translation
- Author
-
Yoshifumi Sonobe, Jihad Aburas, Gopinath Krishnan, Andrew C. Fleming, Ghanashyam Ghadge, Priota Islam, Eleanor C. Warren, Yuanzheng Gu, Mark W. Kankel, André E. X. Brown, Evangelos Kiskinis, Tania F. Gendron, Fen-Biao Gao, Raymond P. Roos, and Paschalis Kratsios
- Subjects
Science - Abstract
A hexanucleotide repeat expansion of C9orf72 is translated to dipeptide repeat proteins in amyotrophic lateral sclerosis and frontotemporal dementia patients. Here the authors generate a C. elegans model of C9orf72-mediated ALS/FTD and show that translation initiation factor eIF2D regulates the dipeptide repeat protein expression.
- Published
- 2021
- Full Text
- View/download PDF
13. Neurofilament light chain and vaccination status associate with clinical outcomes in severe COVID-19
- Author
-
Young Erben, Mercedes Prudencio, Christopher P. Marquez, Karen R. Jansen-West, Michael G. Heckman, Launia J. White, Judith A. Dunmore, Casey N. Cook, Meredith T. Lilley, Neda Qosja, Yuping Song, Rana Hanna Al Shaikh, Lillian M. Daughrity, Jordan L. Bartfield, Gregory S. Day, Björn Oskarsson, Katharine A. Nicholson, Zbigniew K. Wszolek, Jonathan B. Hoyne, Tania F. Gendron, James F. Meschia, and Leonard Petrucelli
- Subjects
Biological sciences ,Immunology ,Virology ,Science - Abstract
Summary: Blood neurofilament light chain (NFL) is proposed to serve as an estimate of disease severity in hospitalized patients with coronavirus disease 2019 (COVID-19). We show that NFL concentrations in plasma collected from 880 patients with COVID-19 within 5 days of hospital admission were elevated compared to controls. Higher plasma NFL associated with worse clinical outcomes including the need for mechanical ventilation, intensive care, prolonged hospitalization, and greater functional disability at discharge. No difference in the studied clinical outcomes between black/African American and white patients was found. Finally, vaccination associated with less disability at time of hospital discharge. In aggregate, our findings support the utility of measuring NFL shortly after hospital admission to estimate disease severity and show that race does not influence clinical outcomes caused by COVID-19 assuming equivalent access to care, and that vaccination may lessen the degree of COVID-19-caused disability.
- Published
- 2022
- Full Text
- View/download PDF
14. Alterations of mesenchymal stromal cells in cerebrospinal fluid: insights from transcriptomics and an ALS clinical trial
- Author
-
Ashley A. Krull, Deborah O. Setter, Tania F. Gendron, Sybil C. L. Hrstka, Michael J. Polzin, Joseph Hart, Amel Dudakovic, Nicolas N. Madigan, Allan B. Dietz, Anthony J. Windebank, Andre J. van Wijnen, and Nathan P. Staff
- Subjects
Mesenchymal stromal cell ,Gene expression ,Neuroinflammation ,Growth factors ,Cerebrospinal fluid ,Amyotrophic lateral sclerosis ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Mesenchymal stromal cells (MSCs) have been studied with increasing intensity as clinicians and researchers strive to understand the ability of MSCs to modulate disease progression and promote tissue regeneration. As MSCs are used for diverse applications, it is important to appreciate how specific physiological environments may stimulate changes that alter the phenotype of the cells. One need for neuroregenerative applications is to characterize the spectrum of MSC responses to the cerebrospinal fluid (CSF) environment after their injection into the intrathecal space. Mechanistic understanding of cellular biology in response to the CSF environment may predict the ability of MSCs to promote injury repair or provide neuroprotection in neurodegenerative diseases. Methods In this study, we characterized changes in morphology, metabolism, and gene expression occurring in human adipose-derived MSCs cultured in human (hCSF) or artificial CSF (aCSF) as well as examined relevant protein levels in the CSF of subjects treated with MSCs for amyotrophic lateral sclerosis (ALS). Results Our results demonstrated that, under intrathecal-like conditions, MSCs retained their morphology, though they became quiescent. Large-scale transcriptomic analysis of MSCs revealed a distinct gene expression profile for cells cultured in aCSF. The aCSF culture environment induced expression of genes related to angiogenesis and immunomodulation. In addition, MSCs in aCSF expressed genes encoding nutritional growth factors to expression levels at or above those of control cells. Furthermore, we observed a dose-dependent increase in growth factors and immunomodulatory cytokines in CSF from subjects with ALS treated intrathecally with autologous MSCs. Conclusions Overall, our results suggest that MSCs injected into the intrathecal space in ongoing clinical trials remain viable and may provide a therapeutic benefit to patients.
- Published
- 2021
- Full Text
- View/download PDF
15. Dipeptide repeat proteins inhibit homology-directed DNA double strand break repair in C9ORF72 ALS/FTD
- Author
-
Nadja S. Andrade, Melina Ramic, Rustam Esanov, Wenjun Liu, Mathew J. Rybin, Gabriel Gaidosh, Abbas Abdallah, Samuel Del’Olio, Tyler C. Huff, Nancy T. Chee, Sadhana Anatha, Tania F. Gendron, Claes Wahlestedt, Yanbin Zhang, Michael Benatar, Christian Mueller, and Zane Zeier
- Subjects
Amyotrophic lateral sclerosis ,DNA damage ,DNA double strand break repair ,Induced pluripotent stem cells ,CRISPR ,Single-strand annealing ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background The C9ORF72 hexanucleotide repeat expansion is the most common known genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two fatal age-related neurodegenerative diseases. The C9ORF72 expansion encodes five dipeptide repeat proteins (DPRs) that are produced through a non-canonical translation mechanism. Among the DPRs, proline-arginine (PR), glycine-arginine (GR), and glycine-alanine (GA) are the most neurotoxic and increase the frequency of DNA double strand breaks (DSBs). While the accumulation of these genotoxic lesions is increasingly recognized as a feature of disease, the mechanism(s) of DPR-mediated DNA damage are ill-defined and the effect of DPRs on the efficiency of each DNA DSB repair pathways has not been previously evaluated. Methods and results Using DNA DSB repair assays, we evaluated the efficiency of specific repair pathways, and found that PR, GR and GA decrease the efficiency of non-homologous end joining (NHEJ), single strand annealing (SSA), and microhomology-mediated end joining (MMEJ), but not homologous recombination (HR). We found that PR inhibits DNA DSB repair, in part, by binding to the nucleolar protein nucleophosmin (NPM1). Depletion of NPM1 inhibited NHEJ and SSA, suggesting that NPM1 loss-of-function in PR expressing cells leads to impediments of both non-homologous and homology-directed DNA DSB repair pathways. By deleting NPM1 sub-cellular localization signals, we found that PR binds NPM1 regardless of the cellular compartment to which NPM1 was directed. Deletion of the NPM1 acidic loop motif, known to engage other arginine-rich proteins, abrogated PR and NPM1 binding. Using confocal and super-resolution immunofluorescence microscopy, we found that levels of RAD52, a component of the SSA repair machinery, were significantly increased iPSC neurons relative to isogenic controls in which the C9ORF72 expansion had been deleted using CRISPR/Cas9 genome editing. Western analysis of post-mortem brain tissues confirmed that RAD52 immunoreactivity is significantly increased in C9ALS/FTD samples as compared to controls. Conclusions Collectively, we characterized the inhibitory effects of DPRs on key DNA DSB repair pathways, identified NPM1 as a facilitator of DNA repair that is inhibited by PR, and revealed deficits in homology-directed DNA DSB repair pathways as a novel feature of C9ORF72-related disease.
- Published
- 2020
- Full Text
- View/download PDF
16. Transcription elongation factor AFF2/FMR2 regulates expression of expanded GGGGCC repeat-containing C9ORF72 allele in ALS/FTD
- Author
-
Yeliz Yuva-Aydemir, Sandra Almeida, Gopinath Krishnan, Tania F. Gendron, and Fen-Biao Gao
- Subjects
Science - Abstract
Mechanisms of transcription of expanded G4C2 repeats in C9ORF72, associated with ALS/FTD, are not fully understood. Here authors use both Drosophila and C9ORF72 iPSC-derived neurons and identify AFF2/FMR2 as a regulator of poly(GR) toxicity by regulating expression of the expanded G4C2 repeats.
- Published
- 2019
- Full Text
- View/download PDF
17. Extensive transcriptomic study emphasizes importance of vesicular transport in C9orf72 expansion carriers
- Author
-
Dennis W. Dickson, Matthew C. Baker, Jazmyne L. Jackson, Mariely DeJesus-Hernandez, NiCole A. Finch, Shulan Tian, Michael G. Heckman, Cyril Pottier, Tania F. Gendron, Melissa E. Murray, Yingxue Ren, Joseph S. Reddy, Neill R. Graff-Radford, Bradley F. Boeve, Ronald C. Petersen, David S. Knopman, Keith A. Josephs, Leonard Petrucelli, Björn Oskarsson, John W. Sheppard, Yan W. Asmann, Rosa Rademakers, and Marka van Blitterswijk
- Subjects
Frontotemporal dementia ,Frontotemporal lobar degeneration ,Amyotrophic lateral sclerosis ,Motor neuron disease ,C9orf72 ,Transcriptomics ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract The majority of the clinico-pathological variability observed in patients harboring a repeat expansion in the C9orf72-SMCR8 complex subunit (C9orf72) remains unexplained. This expansion, which represents the most common genetic cause of frontotemporal lobar degeneration (FTLD) and motor neuron disease (MND), results in a loss of C9orf72 expression and the generation of RNA foci and dipeptide repeat (DPR) proteins. The C9orf72 protein itself plays a role in vesicular transport, serving as a guanine nucleotide exchange factor that regulates GTPases. To further elucidate the mechanisms underlying C9orf72-related diseases and to identify potential disease modifiers, we performed an extensive RNA sequencing study. We included individuals for whom frontal cortex tissue was available: FTLD and FTLD/MND patients with (n = 34) or without (n = 44) an expanded C9orf72 repeat as well as control subjects (n = 24). In total, 6706 genes were differentially expressed between these groups (false discovery rate [FDR]
- Published
- 2019
- Full Text
- View/download PDF
18. Aberrant deposition of stress granule-resident proteins linked to C9orf72-associated TDP-43 proteinopathy
- Author
-
Jeannie Chew, Casey Cook, Tania F. Gendron, Karen Jansen-West, Giulia del Rosso, Lillian M. Daughrity, Monica Castanedes-Casey, Aishe Kurti, Jeannette N. Stankowski, Matthew D. Disney, Jeffrey D. Rothstein, Dennis W. Dickson, John D. Fryer, Yong-Jie Zhang, and Leonard Petrucelli
- Subjects
C9orf72 ,Stress granules ,Frontotemporal dementia ,Amyotrophic lateral sclerosis ,TDP-43 ,Neurodegeneration ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background A G4C2 hexanucleotide repeat expansion in the noncoding region of C9orf72 is the major genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis (c9FTD/ALS). Putative disease mechanisms underlying c9FTD/ALS include toxicity from sense G4C2 and antisense G2C4 repeat-containing RNA, and from dipeptide repeat (DPR) proteins unconventionally translated from these RNA products. Methods Intracerebroventricular injections with adeno-associated virus (AAV) encoding 2 or 149 G4C2 repeats were performed on postnatal day 0, followed by assessment of behavioral and neuropathological phenotypes. Results Relative to control mice, gliosis and neurodegeneration accompanied by cognitive and motor deficits were observed in (G4C2)149 mice by 6 months of age. Recapitulating key pathological hallmarks, we also demonstrate that sense and antisense RNA foci, inclusions of poly(GA), poly(GP), poly(GR), poly(PR), and poly(PA) DPR proteins, and inclusions of endogenous phosphorylated TDP-43 (pTDP-43) developed in (G4C2)149 mice but not control (G4C2)2 mice. Notably, proteins that play a role in the regulation of stress granules – RNA-protein assemblies that form in response to translational inhibition and that have been implicated in c9FTD/ALS pathogenesis – were mislocalized in (G4C2)149 mice as early as 3 months of age. Specifically, we observed the abnormal deposition of stress granule components within inclusions immunopositive for poly(GR) and pTDP-43, as well as evidence of nucleocytoplasmic transport defects. Conclusions Our in vivo model of c9FTD/ALS is the first to robustly recapitulate hallmark features derived from both sense and antisense C9orf72 repeat-associated transcripts complete with neurodegeneration and behavioral impairments. More importantly, the early appearance of persistent pathological stress granules prior to significant pTDP-43 deposition implicates an aberrant stress granule response as a key disease mechanism driving TDP-43 proteinopathy in c9FTD/ALS.
- Published
- 2019
- Full Text
- View/download PDF
19. Hexanucleotide Repeat Expansions in c9FTD/ALS and SCA36 Confer Selective Patterns of Neurodegeneration In Vivo
- Author
-
Tiffany W. Todd, Zachary T. McEachin, Jeannie Chew, Alexander R. Burch, Karen Jansen-West, Jimei Tong, Mei Yue, Yuping Song, Monica Castanedes-Casey, Aishe Kurti, Judith H. Dunmore, John D. Fryer, Yong-Jie Zhang, Beatriz San Millan, Susana Teijeira Bautista, Manuel Arias, Dennis Dickson, Tania F. Gendron, María-Jesús Sobrido, Matthew D. Disney, Gary J. Bassell, Wilfried Rossoll, and Leonard Petrucelli
- Subjects
C9orf72 ,SCA36 ,ALS ,FTD ,RAN translation ,poly(GP) ,Biology (General) ,QH301-705.5 - Abstract
Summary: A G4C2 hexanucleotide repeat expansion in an intron of C9orf72 is the most common cause of frontal temporal dementia and amyotrophic lateral sclerosis (c9FTD/ALS). A remarkably similar intronic TG3C2 repeat expansion is associated with spinocerebellar ataxia 36 (SCA36). Both expansions are widely expressed, form RNA foci, and can undergo repeat-associated non-ATG (RAN) translation to form similar dipeptide repeat proteins (DPRs). Yet, these diseases result in the degeneration of distinct subsets of neurons. We show that the expression of these repeat expansions in mice is sufficient to recapitulate the unique features of each disease, including this selective neuronal vulnerability. Furthermore, only the G4C2 repeat induces the formation of aberrant stress granules and pTDP-43 inclusions. Overall, our results demonstrate that the pathomechanisms responsible for each disease are intrinsic to the individual repeat sequence, highlighting the importance of sequence-specific RNA-mediated toxicity in each disorder.
- Published
- 2020
- Full Text
- View/download PDF
20. Long-read sequencing across the C9orf72 ‘GGGGCC’ repeat expansion: implications for clinical use and genetic discovery efforts in human disease
- Author
-
Mark T. W. Ebbert, Stefan L. Farrugia, Jonathon P. Sens, Karen Jansen-West, Tania F. Gendron, Mercedes Prudencio, Ian J. McLaughlin, Brett Bowman, Matthew Seetin, Mariely DeJesus-Hernandez, Jazmyne Jackson, Patricia H. Brown, Dennis W. Dickson, Marka van Blitterswijk, Rosa Rademakers, Leonard Petrucelli, and John D. Fryer
- Subjects
C9orf72 ,GGGGCC ,Repeat expansion disorders ,Structural mutations ,Amyotrophic lateral sclerosis (ALS) ,Frontotemporal dementia (FTD) ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Many neurodegenerative diseases are caused by nucleotide repeat expansions, but most expansions, like the C9orf72 ‘GGGGCC’ (G4C2) repeat that causes approximately 5–7% of all amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) cases, are too long to sequence using short-read sequencing technologies. It is unclear whether long-read sequencing technologies can traverse these long, challenging repeat expansions. Here, we demonstrate that two long-read sequencing technologies, Pacific Biosciences’ (PacBio) and Oxford Nanopore Technologies’ (ONT), can sequence through disease-causing repeats cloned into plasmids, including the FTD/ALS-causing G4C2 repeat expansion. We also report the first long-read sequencing data characterizing the C9orf72 G4C2 repeat expansion at the nucleotide level in two symptomatic expansion carriers using PacBio whole-genome sequencing and a no-amplification (No-Amp) targeted approach based on CRISPR/Cas9. Results Both the PacBio and ONT platforms successfully sequenced through the repeat expansions in plasmids. Throughput on the MinION was a challenge for whole-genome sequencing; we were unable to attain reads covering the human C9orf72 repeat expansion using 15 flow cells. We obtained 8× coverage across the C9orf72 locus using the PacBio Sequel, accurately reporting the unexpanded allele at eight repeats, and reading through the entire expansion with 1324 repeats (7941 nucleotides). Using the No-Amp targeted approach, we attained > 800× coverage and were able to identify the unexpanded allele, closely estimate expansion size, and assess nucleotide content in a single experiment. We estimate the individual’s repeat region was > 99% G4C2 content, though we cannot rule out small interruptions. Conclusions Our findings indicate that long-read sequencing is well suited to characterizing known repeat expansions, and for discovering new disease-causing, disease-modifying, or risk-modifying repeat expansions that have gone undetected with conventional short-read sequencing. The PacBio No-Amp targeted approach may have future potential in clinical and genetic counseling environments. Larger and deeper long-read sequencing studies in C9orf72 expansion carriers will be important to determine heterogeneity and whether the repeats are interrupted by non-G4C2 content, potentially mitigating or modifying disease course or age of onset, as interruptions are known to do in other repeat-expansion disorders. These results have broad implications across all diseases where the genetic etiology remains unclear.
- Published
- 2018
- Full Text
- View/download PDF
21. Poly-GR dipeptide repeat polymers correlate with neurodegeneration and Clinicopathological subtypes in C9ORF72-related brain disease
- Author
-
Nobutaka Sakae, Kevin F. Bieniek, Yong-Jie Zhang, Kelly Ross, Tania F. Gendron, Melissa E. Murray, Rosa Rademakers, Leonard Petrucelli, and Dennis W. Dickson
- Subjects
C9ORF72 ,Dipeptide repeat polymers (DPR) ,Poly-GR ,Neurodegeneration ,Dimethylarginine ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Frontotemporal lobar degeneration (FTLD) is heterogeneous in clinical presentation, neuropathological characteristics and genetics. An expanded GGGGCC hexanucleotide repeat in C9ORF72 is the most common genetic cause of both FTLD and motor neuron disease (MND). Dipeptide repeat polymers (DPR) are generated through repeat-associated non-ATG translation, and they aggregate in neuronal inclusions with a distribution distinct from that of TDP-43 pathology. Recent studies from animal and cell culture models suggest that DPR might be toxic, but that toxicity may differ for specific DPR. Arginine containing DPR (poly-GR and poly-PR) have the greatest toxicity and are less frequent than other DPR (poly-GP, poly-GA). A unique feature of arginine-containing DPR is their potential for post-translational modification by methyl-transferases, which produces methylarginine DPR. In this report, we explored the relationship of DPR and methylarginine to markers of neurodegeneration using quantitative digital microscopic methods in 40 patients with C9ORF72 mutations and one of three different clinicopathologic phenotypes, FTLD, FTLD-MND or MND. We find that density and distribution of poly-GR inclusions are different from poly-GA and poly-GP inclusions. We also demonstrate colocalization of poly-GR with asymmetrical dimethylarginine (aDMA) immunoreactivity in regions with neurodegeneration. Differences in aDMA were also noted by clinical phenotype. FTLD-MND had the highest burden of poly-GR pathology compared to FTLD and MND, while FTLD-MND had higher burden of aDMA than FTLD. The results suggest that poly-GR pathology is associated with toxicity and neurodegeneration. It remains to be determined if dimethylarginine modification of poly-GR could contribute to its toxicity.
- Published
- 2018
- Full Text
- View/download PDF
22. Loss of Tmem106b is unable to ameliorate frontotemporal dementia-like phenotypes in an AAV mouse model of C9ORF72-repeat induced toxicity
- Author
-
Alexandra M. Nicholson, Xiaolai Zhou, Ralph B. Perkerson, Tammee M. Parsons, Jeannie Chew, Mieu Brooks, Mariely DeJesus-Hernandez, NiCole A. Finch, Billie J. Matchett, Aishe Kurti, Karen R. Jansen-West, Emilie Perkerson, Lillian Daughrity, Monica Castanedes-Casey, Linda Rousseau, Virginia Phillips, Fenghua Hu, Tania F. Gendron, Melissa E. Murray, Dennis W. Dickson, John D. Fryer, Leonard Petrucelli, and Rosa Rademakers
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Loss-of-function mutations in progranulin (GRN) and a non-coding (GGGGCC)n hexanucleotide repeat expansions in C9ORF72 are the two most common genetic causes of frontotemporal lobar degeneration with aggregates of TAR DNA binding protein 43 (FTLD-TDP). TMEM106B encodes a type II transmembrane protein with unknown function. Genetic variants in TMEM106B associated with reduced TMEM106B levels have been identified as disease modifiers in individuals with GRN mutations and C9ORF72 expansions. Recently, loss of Tmem106b has been reported to protect the FTLD-like phenotypes in Grn−/− mice. Here, we generated Tmem106b−/− mice and examined whether loss of Tmem106b could rescue FTLD-like phenotypes in an AAV mouse model of C9ORF72-repeat induced toxicity. Our results showed that neither partial nor complete loss of Tmem106b was able to rescue behavioral deficits induced by the expression of (GGGGCC)66 repeats (66R). Loss of Tmem106b also failed to ameliorate 66R-induced RNA foci, dipeptide repeat protein formation and pTDP-43 pathological burden. We further found that complete loss of Tmem106b increased astrogliosis, even in the absence of 66R, and failed to rescue 66R-induced neuronal cell loss, whereas partial loss of Tmem106b significantly rescued the neuronal cell loss but not neuroinflammation induced by 66R. Finally, we showed that overexpression of 66R did not alter expression of Tmem106b and other lysosomal genes in vivo, and subsequent analyses in vitro found that transiently knocking down C9ORF72, but not overexpression of 66R, significantly increased TMEM106B and other lysosomal proteins. In summary, reducing Tmem106b levels failed to rescue FTLD-like phenotypes in a mouse model mimicking the toxic gain-of-functions associated with overexpression of 66R. Combined with the observation that loss of C9ORF72 and not 66R overexpression was associated with increased levels of TMEM106B, this work suggests that the protective TMEM106B haplotype may exert its effect in expansion carriers by counteracting lysosomal dysfunction resulting from a loss of C9ORF72.
- Published
- 2018
- Full Text
- View/download PDF
23. A C9ORF72 BAC mouse model recapitulates key epigenetic perturbations of ALS/FTD
- Author
-
Rustam Esanov, Gabriela Toro Cabrera, Nadja S. Andrade, Tania F. Gendron, Robert H. Brown, Michael Benatar, Claes Wahlestedt, Christian Mueller, and Zane Zeier
- Subjects
Amyotrophic lateral sclerosis ,C9ORF72 BAC mouse ,Induced pluripotent stem cells ,DNA methylation ,Hydroxymethylation ,R-loops ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Amyotrophic Lateral Sclerosis (ALS) is a fatal and progressive neurodegenerative disorder with identified genetic causes representing a significant minority of all cases. A GGGGCC hexanucleotide repeat expansion (HRE) mutation within the C9ORF72 gene has recently been identified as the most frequent known cause of ALS. The expansion leads to partial heterochromatinization of the locus, yet mutant RNAs and dipeptide repeat proteins (DPRs) are still produced in sufficient quantities to confer neurotoxicity. The levels of these toxic HRE products positively correlate with cellular toxicity and phenotypic severity across multiple disease models. Moreover, the degree of epigenetic repression inversely correlates with some facets of clinical presentation in C9-ALS patients. Recently, bacterial artificial chromosomes (BAC) have been used to generate transgenic mice that harbor the HRE mutation, complementing other relevant model systems such as patient-derived induced pluripotent stem cells (iPSCs). While epigenetic features of the HRE have been investigated in various model systems and post-mortem tissues, epigenetic dysregulation at the expanded locus in C9-BAC mice remains unexplored. Methods and Results Here, we sought to determine whether clinically relevant epigenetic perturbations caused by the HRE are mirrored in a C9-BAC mouse model. We used complementary DNA methylation assessment and immunoprecipitation methods to demonstrate that epigenetic aberrations caused by the HRE, such as DNA and histone methylation, are recapitulated in the C9-BAC mice. Strikingly, we found that cytosine hypermethylation within the promoter region of the human transgene occurred in a subset of C9-BAC mice similar to what is observed in patient populations. Moreover, we show that partial heterochromatinization of the C9 HRE occurs during the first weeks of the mouse lifespan, indicating age-dependent epigenetic repression. Using iPSC neurons, we found that preventing R-loop formation did not impede heterochromatinization of the HRE. Conclusions Taken together, these observations provide further insight into mechanism and developmental time-course of epigenetic perturbations conferred by the C9ORF72 HRE. Finally, we suggest that epigenetic repression of the C9ORF72 HRE and nearby gene promoter could impede or delay motor neuron degeneration in C9-BAC mouse models of ALS/FTD.
- Published
- 2017
- Full Text
- View/download PDF
24. Biomarkers for Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Associated With Hexanucleotide Expansion Mutations in C9orf72
- Author
-
Mary Kay Floeter and Tania F. Gendron
- Subjects
C9orf72 ,cortical thinning ,diffusion tensor imaging ,dipeptide repeat proteins ,functional connectivity ,motor neuron disease ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Now that genetic testing can identify persons at risk for developing amyotrophic lateral sclerosis (ALS) many decades before symptoms begin, there is a critical need for biomarkers that signal the onset and progression of degeneration. The search for candidate disease biomarkers in patients with mutations in the gene C9orf72 has included imaging, physiology, and biofluid measurements. In cross-sectional imaging studies, C9+ ALS patients display diffuse reductions of gray and white matter integrity compared to ALS patients without mutations. This structural imaging signature overlaps with frontotemporal dementia (FTD), reflecting the frequent co-occurrence of cognitive impairment, even frank FTD, in C9+ ALS patients. Changes in functional connectivity occur as critical components of the networks associated with cognition and behavior degenerate. In presymptomatic C9+carriers, subtle differences in volumes of subcortical structures and functional connectivity can be detected, often decades before the typical family age of symptom onset. Dipeptide repeat proteins produced by the repeat expansion mutation are also measurable in the cerebrospinal fluid (CSF) of presymptomatic gene carriers, possibly throughout their lives. In contrast, a rise in the level of neurofilament proteins in the CSF appears to presage the onset of degeneration in presymptomatic carriers in one longitudinal study. Cross-sectional studies indicate that neurofilament protein levels may provide prognostic information for survival in C9+ ALS patients. Longitudinal studies will be needed to validate the candidate biomarkers discussed here. Understanding how these candidate biomarkers change over time is critical if they are to be used in future therapeutic decisions.
- Published
- 2018
- Full Text
- View/download PDF
25. Two FTD-ALS genes converge on the endosomal pathway to induce TDP-43 pathology and degeneration
- Author
-
Wei Shao, Tiffany W. Todd, Yanwei Wu, Caroline Y. Jones, Jimei Tong, Karen Jansen-West, Lillian M. Daughrity, Jinyoung Park, Yuka Koike, Aishe Kurti, Mei Yue, Monica Castanedes-Casey, Giulia del Rosso, Judith A. Dunmore, Desiree Zanetti Alepuz, Björn Oskarsson, Dennis W. Dickson, Casey N. Cook, Mercedes Prudencio, Tania F. Gendron, John D. Fryer, Yong-Jie Zhang, and Leonard Petrucelli
- Subjects
DNA-Binding Proteins ,Mice ,DNA Repeat Expansion ,Multidisciplinary ,C9orf72 Protein ,Frontotemporal Dementia ,Amyotrophic Lateral Sclerosis ,Mutation ,Animals ,Endosomes ,Protein Serine-Threonine Kinases - Abstract
Frontotemporal dementia and amyotrophic lateral sclerosis (FTD-ALS) are associated with both a repeat expansion in the C9orf72 gene and mutations in the TANK-binding kinase 1 ( TBK1 ) gene. We found that TBK1 is phosphorylated in response to C9orf72 poly(Gly-Ala) [poly(GA)] aggregation and sequestered into inclusions, which leads to a loss of TBK1 activity and contributes to neurodegeneration. When we reduced TBK1 activity using a TBK1-R228H (Arg 228 →His) mutation in mice, poly(GA)-induced phenotypes were exacerbated. These phenotypes included an increase in TAR DNA binding protein 43 (TDP-43) pathology and the accumulation of defective endosomes in poly(GA)-positive neurons. Inhibiting the endosomal pathway induced TDP-43 aggregation, which highlights the importance of this pathway and TBK1 activity in pathogenesis. This interplay between C9orf72 , TBK1 , and TDP-43 connects three different facets of FTD-ALS into one coherent pathway.
- Published
- 2022
26. FUS regulates RAN translation through modulating the G-quadruplex structure of GGGGCC repeat RNA in C9orf72-linked ALS/FTD
- Author
-
Yuzo Fujino, Morio Ueyama, Taro Ishiguro, Daisaku Ozawa, Toshihiko Sugiki, Hayato Ito, Asako Murata, Akira Ishiguro, Tania F. Gendron, Kohji Mori, Eiichi Tokuda, Tomoya Taminato, Takuya Konno, Akihide Koyama, Yuya Kawabe, Toshihide Takeuchi, Yoshiaki Furukawa, Toshimichi Fujiwara, Manabu Ikeda, Toshiki Mizuno, Hideki Mochizuki, Hidehiro Mizusawa, Keiji Wada, Kinya Ishikawa, Osamu Onodera, Kazuhiko Nakatani, Hideki Taguchi, Leonard Petrucelli, and Yoshitaka Nagai
- Abstract
Abnormal expansions of GGGGCC repeat sequence in the noncoding region of the C9orf72 gene is the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). The expanded repeat sequence is translated into dipeptide repeat proteins (DPRs) by noncanonical repeat-associated non-AUG (RAN) translation. Since DPRs play central roles in the pathogenesis of C9-ALS/FTD, we here investigate the regulatory mechanisms of RAN translation, focusing on the effects of RNA-binding proteins (RBPs) targeting GGGGCC repeat RNAs. Using C9-ALS/FTD model flies, we demonstrated that the ALS/FTD-linked RBP FUS suppresses RAN translation and neurodegeneration in an RNA-binding activity-dependent manner. Moreover, we found that FUS directly binds to and modulates the G-quadruplex structure of GGGGCC repeat RNA as an RNA chaperone, resulting in the suppression of RAN translation in vitro. These results reveal a previously unrecognized regulatory mechanism of RAN translation by G-quadruplex-targeting RBPs, providing therapeutic insights for C9-ALS/FTD and other repeat expansion diseases.
- Published
- 2023
27. Single nucleus multiome analysis of the prefrontal cortex fromC9orf72ALS/FTD patients illuminates pathways affected during disease progression
- Author
-
Hsiao-Lin V. Wang, Austin M. Veire, Tania F. Gendron, Marla Gearing, Jonathan D. Glass, Peng Jin, Victor G. Corces, and Zachary T. McEachin
- Abstract
Repeat expansions in theC9orf72gene are the most common genetic cause of amyotrophic lateral sclerosis and familial frontotemporal dementia (ALS/FTD). To identify molecular defects that take place in the dorsolateral frontal cortex of patients withC9orf72ALS/FTD, we compared healthy controls withC9orf72ALS/FTD donor samples staged based on the levels of cortical phosphorylated TAR DNA binding protein (pTDP-43), a neuropathological hallmark of disease progression. We identified distinct molecular changes in different cell types that take place during disease progression. These alterations include downregulation of nuclear and mitochondrial ribosomal protein genes in early disease stages that become upregulated as the disease progresses. High ratios of premature oligodendrocytes expressing low levels of genes encoding major myelin protein components are characteristic of late disease stages and may represent a unique signature ofC9orf72ALS/FTD. Microglia with increased reactivity and astrocyte specific transcriptome changes in genes involved in glucose/glycogen metabolism are also associated with disease progression. Late stages ofC9orf72ALS/FTD correlate with sequential changes in the regulatory landscape of several genes in glial cells, namelyMBP/MAG/MOGin oligodendrocytes,CD83/IRF8in microglia, andGLUT1/GYS2/AGLin astrocytes. Only layer 2-3 cortical projection neurons with high expression ofCUX2/LAMP5are significantly reduced inC9orf72ALS/FTD patients with respect to controls. Our findings reveal previously unknown progressive functional changes in cortical cells ofC9orf72ALS/FTD patients that shed light on the mechanisms underlying the pathology of this disease.
- Published
- 2023
28. Comprehensive evaluation of human-derived anti-poly-GA antibodies in cellular and animal models of
- Author
-
Melanie, Jambeau, Kevin D, Meyer, Marian, Hruska-Plochan, Ricardos, Tabet, Chao-Zong, Lee, Ananya, Ray-Soni, Corey, Aguilar, Kitty, Savage, Nibha, Mishra, Nicole, Cavegn, Petra, Borter, Chun-Chia, Lin, Karen R, Jansen-West, Jie, Jiang, Fernande, Freyermuth, Nan, Li, Pierre, De Rossi, Manuela, Pérez-Berlanga, Xin, Jiang, Lilian M, Daughrity, João, Pereira, Sarav, Narayanan, Yuanzheng, Gu, Shekhar, Dhokai, Isin, Dalkilic-Liddle, Zuzanna, Maniecka, Julien, Weber, Michael, Workman, Melissa, McAlonis-Downes, Eugene, Berezovski, Yong-Jie, Zhang, James, Berry, Brian J, Wainger, Mark W, Kankel, Mia, Rushe, Christoph, Hock, Roger M, Nitsch, Don W, Cleveland, Leonard, Petrucelli, Tania F, Gendron, Fabio, Montrasio, Jan, Grimm, Magdalini, Polymenidou, and Clotilde, Lagier-Tourenne
- Subjects
Mice ,Disease Models, Animal ,C9orf72 Protein ,Genes, Regulator ,Animals ,Humans ,Antigen-Antibody Complex ,Dipeptides ,Poly A - Abstract
Hexanucleotide G
- Published
- 2022
29. A C. elegans model of C9orf72-associated ALS/FTD uncovers a conserved role for eIF2D in RAN translation
- Author
-
Fen-Biao Gao, Evangelos Kiskinis, Jihad Aburas, Mark W. Kankel, Yuanzheng Gu, Tania F. Gendron, Yoshifumi Sonobe, Priota Islam, André Ex Brown, Raymond P. Roos, Paschalis Kratsios, Andrew C. Fleming, Gopinath Krishnan, Eleanor C. Warren, and Ghanashyam D. Ghadge
- Subjects
MOTOR DEFICITS ,General Physics and Astronomy ,medicine.disease_cause ,TOXICITY ,DIPEPTIDE-REPEAT PROTEINS ,INITIATION ,C9orf72 ,Gene Editing ,Motor Neurons ,Genetics ,Mutation ,Alanine ,Multidisciplinary ,Neurodegeneration ,Translation (biology) ,MOUSE MODEL ,Dipeptides ,Middle Aged ,Multidisciplinary Sciences ,Frontotemporal Dementia ,Gene Knockdown Techniques ,Science & Technology - Other Topics ,Female ,MESSENGER-RNA ,Proline ,Science ,Glycine ,Biology ,Arginine ,General Biochemistry, Genetics and Molecular Biology ,C9ORF72 ALS ,Eukaryotic translation ,mental disorders ,medicine ,Animals ,Humans ,Initiation factor ,Caenorhabditis elegans ,Science & Technology ,HEXANUCLEOTIDE REPEAT ,C9orf72 Protein ,ANTISENSE TRANSCRIPTS ,Amyotrophic Lateral Sclerosis ,General Chemistry ,medicine.disease ,HEK293 Cells ,Nerve Degeneration ,Ran ,Trinucleotide repeat expansion ,START - Abstract
A hexanucleotide repeat expansion GGGGCC in the non-coding region of C9orf72 is the most common cause of inherited amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Toxic dipeptide repeats (DPRs) are synthesized from GGGGCC via repeat-associated non-AUG (RAN) translation. Here, we develop C. elegans models that express, either ubiquitously or exclusively in neurons, 75 GGGGCC repeats flanked by intronic C9orf72 sequence. The worms generate DPRs (poly-glycine-alanine [poly-GA], poly-glycine-proline [poly-GP]) and poly-glycine-arginine [poly-GR]), display neurodegeneration, and exhibit locomotor and lifespan defects. Mutation of a non-canonical translation-initiating codon (CUG) upstream of the repeats selectively reduces poly-GA steady-state levels and ameliorates disease, suggesting poly-GA is pathogenic. Importantly, loss-of-function mutations in the eukaryotic translation initiation factor 2D (eif-2D/eIF2D) reduce poly-GA and poly-GP levels, and increase lifespan in both C. elegans models. Our in vitro studies in mammalian cells yield similar results. Here, we show a conserved role for eif-2D/eIF2D in DPR expression.
- Published
- 2021
30. A blood-brain penetrant RNA-targeted small molecule triggers elimination of r(G
- Author
-
Jessica A, Bush, Samantha M, Meyer, Rita, Fuerst, Yuquan, Tong, Yue, Li, Raphael I, Benhamou, Haruo, Aikawa, Patrick R A, Zanon, Quentin M R, Gibaut, Alicia J, Angelbello, Tania F, Gendron, Yong-Jie, Zhang, Leonard, Petrucelli, Torben, Heick Jensen, Jessica L, Childs-Disney, and Matthew D, Disney
- Subjects
Mice ,C9orf72 Protein ,Exosome Multienzyme Ribonuclease Complex ,Blood-Brain Barrier ,Frontotemporal Dementia ,Animals ,RNA ,Brain ,RNA-Binding Proteins ,Exosomes ,RNA, Nuclear - Abstract
A hexanucleotide repeat expansion in intron 1 of the
- Published
- 2022
31. FUS regulates RAN translation through modulating the G-quadruplex structure of GGGGCC repeat RNA inC9orf72-linked ALS/FTD
- Author
-
Yuzo Fujino, Morio Ueyama, Taro Ishiguro, Daisaku Ozawa, Toshihiko Sugiki, Hayato Ito, Asako Murata, Akira Ishiguro, Tania F. Gendron, Kohji Mori, Eiichi Tokuda, Tomoya Taminato, Takuya Konno, Akihide Koyama, Yuya Kawabe, Toshihide Takeuchi, Yoshiaki Furukawa, Toshimichi Fujiwara, Manabu Ikeda, Toshiki Mizuno, Hideki Mochizuki, Hidehiro Mizusawa, Keiji Wada, Kinya Ishikawa, Osamu Onodera, Kazuhiko Nakatani, Hideki Taguchi, Leonard Petrucelli, and Yoshitaka Nagai
- Abstract
Abnormal expansions of GGGGCC repeat sequence in the noncoding region of theC9orf72gene is the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). The expanded repeat sequence is translated into dipeptide repeat proteins (DPRs) by noncanonical repeat-associated non-AUG (RAN) translation. Since DPRs play central roles in the pathogenesis of C9-ALS/FTD, we here investigate the regulatory mechanisms of RAN translation, focusing on the effects of RNA-binding proteins (RBPs) targeting GGGGCC repeat RNAs. Using C9-ALS/FTD model flies, we demonstrated that the ALS/FTD-linked RBP FUS suppresses RAN translation and neurodegeneration in an RNA-binding activity-dependent manner. Moreover, we found that FUS directly binds to and modulates the G-quadruplex structure of GGGGCC repeat RNA as an RNA chaperone, resulting in the suppression of RAN translationin vitro. These results reveal a previously unrecognized regulatory mechanism of RAN translation by G-quadruplex-targeting RBPs, providing therapeutic insights for C9-ALS/FTD and other repeat expansion diseases.
- Published
- 2022
32. A blood-brain penetrant RNA-targeted small molecule triggers elimination of r(G4C2)exp in c9ALS/FTD via the nuclear RNA exosome
- Author
-
Jessica A. Bush, Samantha M. Meyer, Rita Fuerst, Yuquan Tong, Yue Li, Raphael I. Benhamou, Haruo Aikawa, Patrick R. A. Zanon, Quentin M. R. Gibaut, Alicia J. Angelbello, Tania F. Gendron, Yong-Jie Zhang, Leonard Petrucelli, Torben Heick Jensen, Jessica L. Childs-Disney, and Matthew D. Disney
- Subjects
Multidisciplinary ,RNA-targeted degradation ,drug design ,repeat expansion disorders ,RNA ,induced proximity - Abstract
A hexanucleotide repeat expansion in intron 1 of the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia, or c9ALS/FTD. The RNA transcribed from the expansion, r(G 4 C 2 ) exp , causes various pathologies, including intron retention, aberrant translation that produces toxic dipeptide repeat proteins (DPRs), and sequestration of RNA-binding proteins (RBPs) in RNA foci. Here, we describe a small molecule that potently and selectively interacts with r(G 4 C 2 ) exp and mitigates disease pathologies in spinal neurons differentiated from c9ALS patient-derived induced pluripotent stem cells (iPSCs) and in two c9ALS/FTD mouse models. These studies reveal a mode of action whereby a small molecule diminishes intron retention caused by the r(G 4 C 2 ) exp and allows the liberated intron to be eliminated by the nuclear RNA exosome, a multi-subunit degradation complex. Our findings highlight the complexity of mechanisms available to RNA-binding small molecules to alleviate disease pathologies and establishes a pipeline for the design of brain penetrant small molecules targeting RNA with novel modes of action in vivo.
- Published
- 2022
33. Poly(ADP-ribose) promotes toxicity of
- Author
-
Junli, Gao, Quinlan T, Mewborne, Amandeep, Girdhar, Udit, Sheth, Alyssa N, Coyne, Ritika, Punathil, Bong Gu, Kang, Morgan, Dasovich, Austin, Veire, Mariely, DeJesus Hernandez, Shuaichen, Liu, Zheng, Shi, Ruxandra, Dafinca, Elise, Fouquerel, Kevin, Talbot, Tae-In, Kam, Yong-Jie, Zhang, Dennis, Dickson, Leonard, Petrucelli, Marka, van Blitterswijk, Lin, Guo, Ted M, Dawson, Valina L, Dawson, Anthony K L, Leung, Thomas E, Lloyd, Tania F, Gendron, Jeffrey D, Rothstein, and Ke, Zhang
- Subjects
DNA-Binding Proteins ,Poly Adenosine Diphosphate Ribose ,C9orf72 Protein ,Frontotemporal Dementia ,Humans ,Dipeptides ,Arginine - Abstract
Arginine-rich dipeptide repeat proteins (R-DPRs), abnormal translational products of a GGGGCC hexanucleotide repeat expansion in
- Published
- 2022
34. Long-read targeted sequencing uncovers clinicopathological associations for C9orf72-linked diseases
- Author
-
Leonard Petrucelli, Keith A. Josephs, Tania F. Gendron, Bjorn Oskarsson, Ronald C. Petersen, John D. Fryer, Neill R. Graff-Radford, David S. Knopman, Mark T. W. Ebbert, Eric D. Wieben, Ian J. McLaughlin, Ross A. Aleff, Jazmyne L. Jackson, Rosa Rademakers, Bradley F. Boeve, Marka van Blitterswijk, Nicole A. Finch, Dennis W. Dickson, Mariely DeJesus-Hernandez, Matt Baker, John Harting, and Melissa E. Murray
- Subjects
Male ,0301 basic medicine ,amyotrophic lateral sclerosis ,Biology ,03 medical and health sciences ,0302 clinical medicine ,C9orf72 ,Cerebellum ,Report ,medicine ,Humans ,Survival advantage ,Amyotrophic lateral sclerosis ,Aged ,Southern blot ,Genetics ,DNA Repeat Expansion ,C9orf72 Protein ,AcademicSubjects/SCI01870 ,Intron ,Neurodegenerative Diseases ,Sequence Analysis, DNA ,Frontotemporal lobar degeneration ,Middle Aged ,medicine.disease ,Cross-Sectional Studies ,030104 developmental biology ,frontotemporal lobar degeneration ,long-read sequencing ,motor neuron disease ,Female ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery ,GC-content - 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., DeJesus-Hernandez et al. employ an innovative long-read sequencing method to examine the length of the expanded C9orf72 repeat that represents the most common genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis. Smaller expansion size confers a survival benefit for patients.
- Published
- 2021
35. Alterations of mesenchymal stromal cells in cerebrospinal fluid: insights from transcriptomics and an ALS clinical trial
- Author
-
Tania F. Gendron, Amel Dudakovic, Ashley A. Krull, Allan B. Dietz, Andre J. van Wijnen, Deborah O. Setter, Nathan P. Staff, Joseph Hart, Michael J. Polzin, Nicolas N. Madigan, Anthony J. Windebank, and Sybil C. L. Hrstka
- Subjects
Angiogenesis ,Medicine (miscellaneous) ,Mesenchymal Stem Cell Transplantation ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Neuroprotection ,Transcriptome ,Immunomodulation ,lcsh:Biochemistry ,Cerebrospinal fluid ,Neuroinflammation ,Medicine ,Humans ,lcsh:QD415-436 ,lcsh:R5-920 ,Human studies ,business.industry ,Mesenchymal stromal cell ,Research ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Amyotrophic lateral sclerosis ,Phenotype ,Cancer research ,Molecular Medicine ,Cytokines ,Gene expression ,Stem cell ,business ,Growth factors ,lcsh:Medicine (General) - Abstract
Background Mesenchymal stromal cells (MSCs) have been studied with increasing intensity as clinicians and researchers strive to understand the ability of MSCs to modulate disease progression and promote tissue regeneration. As MSCs are used for diverse applications, it is important to appreciate how specific physiological environments may stimulate changes that alter the phenotype of the cells. One need for neuroregenerative applications is to characterize the spectrum of MSC responses to the cerebrospinal fluid (CSF) environment after their injection into the intrathecal space. Mechanistic understanding of cellular biology in response to the CSF environment may predict the ability of MSCs to promote injury repair or provide neuroprotection in neurodegenerative diseases. Methods In this study, we characterized changes in morphology, metabolism, and gene expression occurring in human adipose-derived MSCs cultured in human (hCSF) or artificial CSF (aCSF) as well as examined relevant protein levels in the CSF of subjects treated with MSCs for amyotrophic lateral sclerosis (ALS). Results Our results demonstrated that, under intrathecal-like conditions, MSCs retained their morphology, though they became quiescent. Large-scale transcriptomic analysis of MSCs revealed a distinct gene expression profile for cells cultured in aCSF. The aCSF culture environment induced expression of genes related to angiogenesis and immunomodulation. In addition, MSCs in aCSF expressed genes encoding nutritional growth factors to expression levels at or above those of control cells. Furthermore, we observed a dose-dependent increase in growth factors and immunomodulatory cytokines in CSF from subjects with ALS treated intrathecally with autologous MSCs. Conclusions Overall, our results suggest that MSCs injected into the intrathecal space in ongoing clinical trials remain viable and may provide a therapeutic benefit to patients.
- Published
- 2021
36. Truncated stathmin-2 is a marker of TDP-43 pathology in frontotemporal dementia
- Author
-
Prasanth Sivakumar, Jack Humphrey, Keith A. Josephs, Mercedes Prudencio, E. Aubrey Thompson, Kevin Talbot, Bjorn Oskarsson, Hemali Phatnani, Leonard Petrucelli, Ana Candalija, David S. Knopman, Pietro Fratta, Casey Cook, Yari Carlomagno, Cristhoper H. Fernandez De Castro, Duyang Kim, Neil Graff-Radford, Maria Secrier, Siddharthan Chandran, Mei Yue, Anna-Leigh Brown, Sarah E. Hill, Bhuvaneish T. Selvaraj, Michael G. Heckman, Cristian Bodo, Karen Jansen-West, Michael E. Ward, Demetra Catalano, Samantha Fennessey, Elizabeth M. C. Fisher, Michael DeTure, Ronald C. Petersen, Dennis W. Dickson, Jia Newcombe, Isabel Hubbard, Delphine Fagegaltier, Tania F. Gendron, Ying-Chih Wang, Karen Burr, Lillian M. Daughrity, Tammaryn Lashley, J. Shi, Yuping Song, Jennifer M. Kachergus, Matthew R. Spiegel, Rosa Rademakers, Marka van Blitterswijk, Shunsuke Koga, Bradley F. Boeve, Sarah R. Pickles, Nadia Propp, and Towfique Raj
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Induced Pluripotent Stem Cells ,Stathmin ,Disease ,TARDBP ,Progressive supranuclear palsy ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,Humans ,Medicine ,Dementia ,biology ,business.industry ,nutritional and metabolic diseases ,RNA ,General Medicine ,Human brain ,Middle Aged ,medicine.disease ,Frontal Lobe ,nervous system diseases ,DNA-Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Frontotemporal Dementia ,030220 oncology & carcinogenesis ,Mutation ,Commentary ,biology.protein ,Female ,business ,Biomarkers ,Frontotemporal dementia - Abstract
No treatment for frontotemporal dementia (FTD), the second most common type of early-onset dementia, is available, but therapeutics are being investigated to target the 2 main proteins associated with FTD pathological subtypes: TDP-43 (FTLD-TDP) and tau (FTLD-tau). Testing potential therapies in clinical trials is hampered by our inability to distinguish between patients with FTLD-TDP and FTLD-tau. Therefore, we evaluated truncated stathmin-2 (STMN2) as a proxy of TDP-43 pathology, given the reports that TDP-43 dysfunction causes truncated STMN2 accumulation. Truncated STMN2 accumulated in human induced pluripotent stem cell-derived neurons depleted of TDP-43, but not in those with pathogenic TARDBP mutations in the absence of TDP-43 aggregation or loss of nuclear protein. In RNA-Seq analyses of human brain samples from the NYGC ALS cohort, truncated STMN2 RNA was confined to tissues and disease subtypes marked by TDP-43 inclusions. Last, we validated that truncated STMN2 RNA was elevated in the frontal cortex of a cohort of patients with FTLD-TDP but not in controls or patients with progressive supranuclear palsy, a type of FTLD-tau. Further, in patients with FTLD-TDP, we observed significant associations of truncated STMN2 RNA with phosphorylated TDP-43 levels and an earlier age of disease onset. Overall, our data uncovered truncated STMN2 as a marker for TDP-43 dysfunction in FTD.
- Published
- 2020
37. Sensitive ELISA-based detection method for the mitophagy marker p-S65-Ub in human cells, autopsy brain, and blood samples
- Author
-
Xu Hou, Fabienne C. Fiesel, Jens O. Watzlawik, Michael G. Heckman, Wolfdieter Springer, Owen A. Ross, Matthew S. Goldberg, Zbigniew K. Wszolek, Michael DeTure, Sandeep Kumar Barodia, Dennis W. Dickson, Joanna Siuda, Chloe Ramnarine, Tania F. Gendron, Guojun Bu, Dominika Fricova, and Clemens R. Scherzer
- Subjects
0301 basic medicine ,autophagy ,Ubiquitin-Protein Ligases ,Autopsy ,PINK1 ,Enzyme-Linked Immunosorbent Assay ,Disease ,Parkin ,03 medical and health sciences ,Mice ,Ubiquitin ,Mitophagy ,medicine ,Animals ,Humans ,PRKN ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,Autophagy ,Brain ,Cell Biology ,medicine.disease ,nervous system diseases ,Parkinson disease ,030104 developmental biology ,Cancer research ,biology.protein ,Alzheimer's disease ,Alzheimer disease ,Toolbox ,Protein Kinases ,Research Article - Abstract
Mitochondrial dysfunction is an early, imminent event in neurodegenerative disorders including Parkinson disease (PD) and Alzheimer disease (AD). The enzymatic pair PINK1 and PRKN/Parkin recognize and transiently label damaged mitochondria with ubiquitin (Ub) phosphorylated at Ser65 (p-S65-Ub) as a signal for degradation via the autophagy-lysosome system (mitophagy). Despite its discovery in cell culture several years ago, robust and quantitative detection of altered mitophagy in vivo has remained challenging. Here we developed a sandwich ELISA targeting p-S65-Ub with the goal to assess mitophagy levels in mouse brain and in human clinical and pathological samples. We characterized five total Ub and four p-S65-Ub antibodies by several techniques and found significant differences in their ability to recognize phosphorylated Ub. The most sensitive antibody pair detected recombinant p-S65-Ub chains in the femtomolar to low picomolar range depending on the poly-Ub chain linkage. Importantly, this ELISA was able to assess very low baseline mitophagy levels in unstressed human cells and in brains from wild-type and prkn knockout mice as well as elevated p-S65-Ub levels in autopsied frontal cortex from AD patients vs. control cases. Moreover, the assay allowed detection of p-S65-Ub in blood plasma and was able to discriminate between PINK1 mutation carriers and controls. In summary, we developed a robust and sensitive tool to measure mitophagy levels in cells, tissue, and body fluids. Our data strongly support the idea that the stress-activated PINK1-PRKN mitophagy pathway is constitutively active in mice and humans under unstimulated, physiological and elevated in diseased, pathological conditions. Abbreviations: Ab: antibody; AD: Alzheimer disease; AP: alkaline phosphatase; CV: coefficient of variation; ECL: electrochemiluminescence; KO: knockout; LoB: Limit of Blank; LoD: Limit of Detection; LoQ: Limit of Quantification; MSD: meso scale discovery; PD: Parkinson disease; p-S65-PRKN: phosphorylated PRKN at serine 65; p-S65-Ub: phosphorylated ubiquitin at serine 65; Std.Dev.: standard deviation; Ub: ubiquitin; WT: wild type
- Published
- 2020
38. Reduced C9ORF72 function exacerbates gain of toxicity from ALS/FTD-causing repeat expansion in C9orf72
- Author
-
Don W. Cleveland, Amy Taylor, Albert R. La Spada, Clotilde Lagier-Tourenne, Jie Jiang, Tania F. Gendron, Sandra Diaz Garcia, Huaxi Xu, John Ravits, Somasish Ghosh Dastidar, Lu-Lin Jiang, Leonard Petrucelli, Melissa McAlonis-Downes, María José Cubillas Rodríguez, Patrick King, Sandrine Da Cruz, Qiang Zhu, and Yong Jie Zhang
- Subjects
0301 basic medicine ,Genetically modified mouse ,General Neuroscience ,Transgene ,Autophagy ,Endogeny ,Biology ,medicine.disease ,Cell biology ,C9orf72 Protein ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,C9orf72 ,medicine ,Amyotrophic lateral sclerosis ,Trinucleotide repeat expansion ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Hexanucleotide expansions in C9orf72, which encodes a predicted guanine exchange factor, are the most frequent genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although repeat expansion has been established to generate toxic products, mRNAs encoding the C9ORF72 protein are also reduced in affected individuals. In this study, we tested how C9ORF72 protein levels affected repeat-mediated toxicity. In somatic transgenic mice expressing 66 GGGGCC repeats, inactivation of one or both endogenous C9orf72 alleles provoked or accelerated, respectively, early death. In mice expressing a C9orf72 transgene with 450 repeats that did not encode the C9ORF72 protein, inactivation of one or both endogenous C9orf72 alleles exacerbated cognitive deficits, hippocampal neuron loss, glial activation and accumulation of dipeptide-repeat proteins from translation of repeat-containing RNAs. Reduced C9ORF72 was shown to suppress repeat-mediated elevation in autophagy. These efforts support a disease mechanism in ALS/FTD resulting from reduced C9ORF72, which can lead to autophagy deficits, synergizing with repeat-dependent gain of toxicity.
- Published
- 2020
39. Ribonuclease recruitment using a small molecule reduced c9ALS/FTD r(G(4)C(2)) repeat expansion in vitro and in vivo ALS models
- Author
-
Jeffrey D. Rothstein, Jonathan L. Chen, Montina Van Meter, Tania F. Gendron, Alyssa N. Coyne, Yong Jie Zhang, Eric T. Wang, Yue Li, Ilyas Yildirim, Rita Fuerst, Jessica A. Bush, Leonard Petrucelli, Samantha M. Meyer, Hailey Olafson, Jessica L. Childs-Disney, Kye Won Wang, Yuquan Tong, Kendra K. McKee, Raphael I. Benhamou, Tanya Khan, Andrei Ursu, Matthew D. Disney, Sarah Wagner-Griffin, and Haruo Aikawa
- Subjects
DNA Repeat Expansion ,C9orf72 Protein ,biology ,Amyotrophic Lateral Sclerosis ,RNA ,Chromosome 9 ,General Medicine ,medicine.disease ,Molecular biology ,Article ,Open reading frame ,Ribonucleases ,C9orf72 ,Frontotemporal Dementia ,mental disorders ,biology.protein ,medicine ,Humans ,Ribonuclease ,Amyotrophic lateral sclerosis ,Trinucleotide repeat expansion ,Frontotemporal dementia - Abstract
The most common cause of amyotrophic lateral sclerosis and frontotemporal dementia (c9ALS/FTD) is an expanded G(4)C(2) RNA repeat [r(G(4)C(2))(exp)] in chromosome 9 open reading frame 72 (C9orf72), which elicits pathology through several mechanisms. Here, we developed and characterized a small molecule for targeted degradation of r(G(4)C(2))(exp). The compound was able to selectively bind r(G(4)C(2))(exp)’s structure and to assemble an endogenous nuclease onto the target, provoking removal of the transcript by native RNA quality control mechanisms. In c9ALS patient–derived spinal neurons, the compound selectively degraded the mutant C9orf72 allele with limited off-targets and reduced quantities of toxic dipeptide repeat proteins (DPRs) translated from r(G(4)C(2))(exp). In vivo work in a rodent model showed that abundance of both the mutant allele harboring the repeat expansion and DPRs were selectively reduced by this compound. These results demonstrate that targeted small-molecule degradation of r(G(4)C(2))(exp) is a strategy for mitigating c9ALS/FTD-associated pathologies and studying disease-associated pathways in preclinical models.
- Published
- 2021
40. A small molecule exploits hidden structural features within the RNA repeat expansion that causes c9ALS/FTD and rescues pathological hallmarks
- Author
-
Jessica A. Bush, Andrei Ursu, Amirhossein Taghavi, Matthew D. Disney, Shruti Choudhary, Leonard Petrucelli, Yong Jie Zhang, Jared T. Baisden, Ilyas Yildirim, and Tania F. Gendron
- Subjects
chemistry.chemical_classification ,DNA Repeat Expansion ,C9orf72 Protein ,Physiology ,Cognitive Neuroscience ,Amyotrophic Lateral Sclerosis ,Intron ,RNA ,Translation (biology) ,Cell Biology ,General Medicine ,Biochemistry ,Small molecule ,Article ,Stress granule ,chemistry ,C9orf72 ,Frontotemporal Dementia ,Biophysics ,Humans ,Nucleotide ,Trinucleotide repeat expansion - Abstract
The hexanucleotide repeat expansion GGGGCC [r(G(4)C(2))(exp)] within intron 1 of C9orf72 causes genetically defined amyotrophic lateral sclerosis and frontotemporal dementia, collectively named c9ALS/FTD. , the repeat expansion causes neurodegeneration via deleterious phenotypes stemming from r(G(4)C(2))(exp) RNA gain- and loss-of-function mechanisms. The r(G(4)C(2))(exp) RNA folds into both a hairpin structure with repeating 1 × 1 nucleotide GG internal loops and a G-quadruplex structure. Here, we report the identification of a small molecule (CB253) that selectively binds the hairpin form of r(G(4)C(2))(exp). Interestingly, the small molecule binds to a previously unobserved conformation in which the RNA forms 2 × 2 nucleotide GG internal loops, as revealed by a series of binding and structural studies. NMR and molecular dynamics simulations suggest that the r(G(4)C(2))(exp) hairpin interconverts between 1 × 1 and 2 × 2 internal loops through the process of strand slippage. We provide experimental evidence that CB253 binding indeed shifts the equilibrium toward the 2 × 2 GG internal loop conformation, inhibiting mechanisms that drive c9ALS/FTD pathobiology, such as repeat-associated non-ATG translation formation of stress granules and defective nucleocytoplasmic transport in various cellular models of c9ALS/FTD.
- Published
- 2021
41. Serum neurofilament light protein correlates with unfavorable clinical outcomes in hospitalized patients with COVID-19
- Author
-
Launia J. White, Young Erben, Yuping Song, Meredith T. Lilley, Tania F. Gendron, Leonard Petrucelli, Caroline F. Harlow, Mercedes Prudencio, Bjorn Oskarsson, Zbigniew K. Wszolek, James F. Meschia, La Tonya J. Hickson, Katharine Nicholson, Michael G. Heckman, John C. O’Horo, Jonathan B. Hoyne, Karen Jansen-West, Christopher P. Marquez, Camila Franco-Mesa, Judith A. Dunmore, and Casey Cook
- Subjects
0301 basic medicine ,Tumor Necrosis Factor Ligand Superfamily Member 14 ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Hospitalized patients ,Intermediate Filaments ,Gastroenterology ,Leukoencephalopathy ,03 medical and health sciences ,0302 clinical medicine ,Neuroimaging ,Neurofilament Proteins ,Report ,Internal medicine ,medicine ,Humans ,Prospective Studies ,Prospective cohort study ,medicine.diagnostic_test ,SARS-CoV-2 ,business.industry ,COVID-19 ,Magnetic resonance imaging ,General Medicine ,STM reports ,medicine.disease ,Magnetic Resonance Imaging ,Hyperintensity ,Coronavirus ,030104 developmental biology ,Cohort ,Medicine ,business ,Biomarkers ,030217 neurology & neurosurgery ,Reports - Abstract
Hospitalized patients with COVID-19 show increased serum concentrations of neurofilament light chain that correlate with worse clinical outcomes., Brain damage marker in COVID-19 SARS-CoV-2 infection, the cause of coronavirus disease 2019 (COVID-19), causes neurological manifestations in a substantial proportion of patients. Determining the extent of neuronal injury is essential to better understand disease pathophysiology and to evaluate potential therapies. Prudencio et al. analyzed serum from 142 patients hospitalized with COVID-19 and showed that the expression of the neurofilament light protein (NFL), a marker of neuroaxonal injury, was elevated compared to healthy controls. In addition, serum NFL expression correlated with disease severity and tended to be reduced in subjects treated with remdesivir. The results suggest that serum NFL analysis should be incorporated when evaluating therapeutic trials for COVID-19., Brain imaging studies of patients with COVID-19 show evidence of macro- and microhemorrhagic lesions, multifocal white matter hyperintensities, and lesions consistent with posterior reversible leukoencephalopathy. Imaging studies, however, are subject to selection bias, and prospective studies are challenging to scale. Here, we evaluated whether serum neurofilament light chain (NFL), a neuroaxonal injury marker, could predict the extent of neuronal damage in a cohort of 142 hospitalized patients with COVID-19. NFL was elevated in the serum of patients with COVID-19 compared to healthy controls, including those without overt neurological manifestations. Higher NFL serum concentrations were associated with worse clinical outcomes. In 100 hospitalized patients with COVID-19 treated with remdesivir, a trend toward lower NFL serum concentrations was observed. These data suggest that patients with COVID-19 may experience neuroaxonal injury and may be at risk for long-term neurological sequelae. Neuroaxonal injury should be considered as an outcome in acute pharmacotherapeutic trials for COVID-19.
- Published
- 2021
42. Transcription elongation factor AFF2/FMR2 regulates expression of expanded GGGGCC repeat-containing C9ORF72 allele in ALS/FTD
- Author
-
Gopinath Krishnan, Sandra Almeida, Tania F. Gendron, Fen-Biao Gao, and Yeliz Yuva-Aydemir
- Subjects
0301 basic medicine ,Male ,Transcription, Genetic ,General Physics and Astronomy ,Gene Knockout Techniques ,0302 clinical medicine ,C9orf72 ,Transcription (biology) ,Drosophila Proteins ,Amyotrophic lateral sclerosis ,Induced pluripotent stem cell ,lcsh:Science ,Aged, 80 and over ,Neurons ,Multidisciplinary ,DNA Repeat Expansion ,Nuclear Proteins ,Dipeptides ,Middle Aged ,3. Good health ,Cell biology ,DNA-Binding Proteins ,Frontotemporal Dementia ,Drosophila ,Female ,Locomotion ,Science ,Induced Pluripotent Stem Cells ,Down-Regulation ,Biology ,DNA-binding protein ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,medicine ,Animals ,Humans ,Neurodegeneration ,Aged ,C9orf72 Protein ,Amyotrophic Lateral Sclerosis ,RNA ,General Chemistry ,medicine.disease ,Axons ,GC Rich Sequence ,030104 developmental biology ,lcsh:Q ,030217 neurology & neurosurgery ,Genetic screen ,Transcription Factors - Abstract
Expanded GGGGCC (G4C2) repeats in C9ORF72 cause amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). How RNAs containing expanded G4C2 repeats are transcribed in human neurons is largely unknown. Here we describe a Drosophila model in which poly(GR) expression in adult neurons causes axonal and locomotor defects and premature death without apparent TDP-43 pathology. In an unbiased genetic screen, partial loss of Lilliputian (Lilli) activity strongly suppresses poly(GR) toxicity by specifically downregulating the transcription of GC-rich sequences in Drosophila. Knockout of AFF2/FMR2 (one of four mammalian homologues of Lilli) with CRISPR-Cas9 decreases the expression of the mutant C9ORF72 allele containing expanded G4C2 repeats and the levels of repeat RNA foci and dipeptide repeat proteins in cortical neurons derived from induced pluripotent stem cells of C9ORF72 patients, resulting in rescue of axonal degeneration and TDP-43 pathology. Thus, AFF2/FMR2 regulates the transcription and toxicity of expanded G4C2 repeats in human C9ORF72-ALS/FTD neurons., Mechanisms of transcription of expanded G4C2 repeats in C9ORF72, associated with ALS/FTD, are not fully understood. Here authors use both Drosophila and C9ORF72 iPSC-derived neurons and identify AFF2/FMR2 as a regulator of poly(GR) toxicity by regulating expression of the expanded G4C2 repeats.
- Published
- 2019
43. Extensive transcriptomic study emphasizes importance of vesicular transport in C9orf72 expansion carriers
- Author
-
Joseph S. Reddy, Bjorn Oskarsson, Shulan Tian, Matt Baker, Tania F. Gendron, Michael G. Heckman, Melissa E. Murray, Ni Cole A. Finch, David S. Knopman, Keith A. Josephs, John W. Sheppard, Jazmyne L. Jackson, Yingxue Ren, Leonard Petrucelli, Mariely DeJesus-Hernandez, Dennis W. Dickson, Cyril Pottier, Ronald C. Petersen, Rosa Rademakers, Bradley F. Boeve, Yan W. Asmann, Neill R. Graff-Radford, and Marka van Blitterswijk
- Subjects
Male ,Heterozygote ,Cyclin dependent kinase like 1 ,Biology ,Frontotemporal lobar degeneration ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,C9orf72 ,Machine learning ,Humans ,Gene Regulatory Networks ,Motor neuron disease ,Transcriptomics ,lcsh:Neurology. Diseases of the nervous system ,030304 developmental biology ,Aged ,Aged, 80 and over ,0303 health sciences ,Repeat expansion disorders ,Vesicular transport ,DNA Repeat Expansion ,C9orf72 Protein ,Research ,RNA sequencing ,Middle Aged ,Amyotrophic lateral sclerosis ,Cell biology ,Frontal Lobe ,Vesicular transport protein ,Protein Transport ,RNA splicing ,Female ,Neurology (clinical) ,Synaptic signaling ,Guanine nucleotide exchange factor ,Human medicine ,Trinucleotide repeat expansion ,Transcriptome ,030217 neurology & neurosurgery ,Frontotemporal dementia - Abstract
The majority of the clinico-pathological variability observed in patients harboring a repeat expansion in the C9orf72-SMCR8 complex subunit (C9orf72) remains unexplained. This expansion, which represents the most common genetic cause of frontotemporal lobar degeneration (FTLD) and motor neuron disease (MND), results in a loss of C9orf72 expression and the generation of RNA foci and dipeptide repeat (DPR) proteins. The C9orf72 protein itself plays a role in vesicular transport, serving as a guanine nucleotide exchange factor that regulates GTPases. To further elucidate the mechanisms underlying C9orf72-related diseases and to identify potential disease modifiers, we performed an extensive RNA sequencing study. We included individuals for whom frontal cortex tissue was available: FTLD and FTLD/MND patients with (n = 34) or without (n = 44) an expanded C9orf72 repeat as well as control subjects (n = 24). In total, 6706 genes were differentially expressed between these groups (false discovery rate [FDR] C9orf72 (FDR = 1.41E-14), which was roughly two-fold lower in C9orf72 expansion carriers than in (disease) controls. Co-expression analysis revealed groups of correlated genes (modules) that were enriched for processes such as protein folding, RNA splicing, synaptic signaling, metabolism, and Golgi vesicle transport. Within our cohort of C9orf72 expansion carriers, machine learning uncovered interesting candidates associated with clinico-pathological features, including age at onset (vascular endothelial growth factor A [VEGFA]), C9orf72 expansion size (cyclin dependent kinase like 1 [CDKL1]), DPR protein levels (eukaryotic elongation factor 2 kinase [EEF2K]), and survival after onset (small G protein signaling modulator 3 [SGSM3]). Given the fact that we detected a module involved in vesicular transport in addition to a GTPase activator (SGSM3) as a potential modifier, our findings seem to suggest that the presence of a C9orf72 repeat expansion might hamper vesicular transport and that genes affecting this process may modify the phenotype of C9orf72-linked diseases.
- Published
- 2019
44. Deep Venous Thrombosis and Pulmonary Embolism Among Hospitalized Coronavirus Disease 2019 (COVID-19) Positive Patients Predict Higher Mortality, Prolonged Intensive Care Unit and Hospital Stays in A Multi-Site Healthcare System
- Author
-
Mercedes Prudencio, Melanie R.F. Greenway, Peter Gloviczki, Nancy L. O'Keefe, Houssan Farres, Josephine F. Huang, Jason Siegel, Tania F. Gendron, Yupeng Li, Jonathan B. Hoyne, Alfredo Quinones-Hinojoas, Charles Ritchie, Robert D. McBane, Raymond C. Shields, Osman S. Hamid, Pablo Moreno-Franco, Devang Sanghavi, Beau Toskich, Young Erben, Myung S. Park, Manju Kalra, William M. Stone, Zlatko Devcic, Candido E. Rivera, Christopher P. Marquez, Neethu Gopal, Michelle Lin, Andrew J. Meltzer, James F. Meschia, Albert G. Hakaim, Camila Franco-Mesa, Leonard Petrucelli, Randall R. De Martino, Christopher J. Lamb, and John C. O’Horo
- Subjects
Male ,medicine.medical_specialty ,Critical Care ,Deep vein ,030204 cardiovascular system & hematology ,Article ,law.invention ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,law ,Risk Factors ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Aged ,Venous Thrombosis ,business.industry ,Incidence (epidemiology) ,Incidence ,COVID-19 ,Odds ratio ,Middle Aged ,medicine.disease ,Intensive care unit ,Thrombosis ,Confidence interval ,Pulmonary embolism ,Hospitalization ,Survival Rate ,medicine.anatomical_structure ,Logistic Models ,Case-Control Studies ,Cohort ,Surgery ,Female ,Cardiology and Cardiovascular Medicine ,business ,Pulmonary Embolism - Abstract
OBJECTIVE: We assessed the incidence of deep vein thrombosis (DVT) and pulmonary embolism (PE) in hospitalized patients with coronavirus disease 2019 (COVID-19) compared with that in a matched cohort with similar cardiovascular risk factors and the effects of DVT and PE on the hospital course. METHODS: We performed a retrospective review of prospectively collected data from COVID-19 patients who had been hospitalized from March 11, 2020 to September 4, 2020. The patients were randomly matched in a 1:1 ratio by age, sex, hospital of admission, smoking history, diabetes mellitus, and coronary artery disease with a cohort of patients without COVID-19. The primary endpoint was the incidence of DVT/PE and the odds of developing DVT/PE using a conditional logistic regression model. The secondary endpoint was the hospitalization outcomes for COVID-19 patients with and without DVT/PE, including mortality, intensive care unit (ICU) admission, ICU stay, and length of hospitalization (LOH). Multivariable regression analysis was performed to identify the variables associated with mortality, ICU admission, discharge disposition, ICU duration, and LOH. RESULTS: A total of 13,310 patients had tested positive for COVID-19, 915 of whom (6.9%) had been hospitalized across our multisite health care system. The mean age of the hospitalized patients was 60.8 ± 17.0 years, and 396 (43.3%) were women. Of the 915 patients, 82 (9.0%) had had a diagnosis of DVT/PE confirmed by ultrasound examination of the extremities and/or computed tomography angiography of the chest. The odds of presenting with DVT/PE in the setting of COVID-19 infection was greater than that without COVID-19 infection (0.6% [5 of 915] vs 9.0% [82 of 915]; odds ratio [OR], 18; 95% confidence interval [CI], 8.0-51.2; P < .001). The vascular risk factors were not different between the COVID-19 patients with and without DVT/PE. Mortality (P = .02), the need for ICU stay (P < .001), duration of ICU stay (P < .001), and LOH (P < .001) were greater in the DVT/PE cohort than in the cohort without DVT/PE. On multivariable logistic regression analysis, the hemoglobin (OR, 0.71; 95% CI, 0.46-0.95; P = .04) and D-dimer (OR, 1.0; 95% CI, 0.33-1.56; P = .03) levels were associated with higher mortality. Higher activated partial thromboplastin times (OR, 1.1; 95% CI, 1.00-1.12; P = .03) and higher interleukin-6 (IL-6) levels (OR, 1.0; 95% CI, 1.01-1.07; P = .05) were associated with a greater risk of ICU admission. IL-6 (OR, 1.0; 95% CI, 1.00-1.02; P = .05) was associated with a greater risk of rehabilitation placement after discharge. On multivariable gamma regression analysis, hemoglobin (coefficient, -3.0; 95% CI, 0.03-0.08; P = .005) was associated with a prolonged ICU stay, and the activated partial thromboplastin time (coefficient, 2.0; 95% CI, 0.003-0.006; P = .05), international normalized ratio (coefficient, -3.2; 95% CI, 0.06-0.19; P = .002) and IL-6 (coefficient, 2.4; 95% CI, 0.0011-0.0027; P = .02) were associated with a prolonged LOH. CONCLUSIONS: A significantly greater incidence of DVT/PE occurred in hospitalized COVID-19-positive patients compared with a non-COVID-19 cohort matched for cardiovascular risk factors. Patients affected by DVT/PE were more likely to experience greater mortality, to require ICU admission, and experience prolonged ICU stays and LOH compared with COVID-19-positive patients without DVT/PE. Advancements in DVT/PE prevention are needed for patients hospitalized for COVID-19 infection.
- Published
- 2021
45. Studying the natural history of frontotemporal lobar degeneration (FTLD): The ARTFL LEFFTDS longitudinal FTLD (ALLFTD) protocol
- Author
-
Joanne Taylor, Neill R. Graff-Radford, Kelley Faber, Walter A. Kukull, Tania F. Gendron, Alexander Pantelyat, Daniel I. Kaufer, Emma Lagone, Rosa Rademakers, Bradley F. Boeve, Howard J. Rosen, Leonard Petrucelli, Arthur W. Toga, Marwan N. Sabbagh, Diana Wheaton, Joel H. Kramer, Jeremy Syrjanen, Walter K. Kremers, Diane Lucente, Peter A. Ljubenkov, Anne M. Fagan, Dennis W. Dickson, Zbigniew K. Wszolek, Aaron Ritter, Leah K. Forsberg, Danielle Brushaber, Belen Pascual, Domoto-Reilly Kimiko, Patrick Brannelly, Anna Karydas, Hugo Botha, Daniel H. Geschwind, Bonnie Wong, Erik D. Roberson, Nupur Ghoshal, Doug R. Galasko, David J. Irwin, Jonathan Graff-Radford, Ging-Yuek Robin Hsiung, Murray Grossman, Adam M. Staffaroni, Toji Miyagawa, Edward D. Huey, Chiadi U. Onyike, Eliana Marisa Ramos, Eric J. Huang, John Kornak, Gabriel C. Léger, Carmela Tartaglia, Jessica E. Rexach, Tatiana Foroud, Scott M. McGinnis, Adam L. Boxer, David T.W. Jones, Irene Litvan, Rodney Pearlman, William W. Seeley, Yvette Bordelon, Masood Manoochehri, Julio C. Rojas, Sandra Weintraub, Bruce L. Miller, David S. Knopman, Katherine P. Rankin, Kejal Kantarci, Ian Grant, Ralitza H. Gavrilova, Joseph C. Masdeu, Ann Fishman, David P. Salmon, Julie A. Fields, Kevin M. Nelson, Brad C. Dickerson, Brian S. Appleby, Joanna M. Biernacka, Katya Rascovsky, Jill Goldman, Mario F. Mendez, Hilary W. Heuer, Ian R. A. Mackenzie, Rodolfo Savica, and Nadine Tatton
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Grossman ,Developmental Neuroscience ,Epidemiology ,Health Policy ,media_common.quotation_subject ,Neurology (clinical) ,Art ,Geriatrics and Gerontology ,Humanities ,media_common - Abstract
Author(s): Boeve, Bradley F; Boxer, Adam L; Rosen, Howard J; Forsberg, Leah K; Heuer, Hilary W; Brushaber, Danielle; Appleby, Brian; Biernacka, Joanna M; Bordelon, Yvette M; Botha, Hugo; Brannelly, Patrick; Dickerson, Brad C; Dickson, Dennis W; Kimiko, Domoto‐Reilly; Faber, Kelley; Fagan, Anne; Fields, Julie A; Fishman, Ann; Foroud, Tatiana M; Galasko, Doug R; Gavrilova, Ralitza H; Gendron, Tania F; Geschwind, Daniel H; Ghoshal, Nupur; Goldman, Jill; Graff‐Radford, Jonathan; Graff‐Radford, Neill R; Grant, Ian; Grossman, Murray; Hsiung, Ging‐Yuek Robin; Huang, Eric J; Huey, Edward; Irwin, David J; Jones, David T; Kantarci, Kejal; Karydas, Anna M; Kaufer, Daniel; Knopman, David S; Kramer, Joel H; Kremers, Walter K; Kornak, John; Kukull, Walter A; Lagone, Emma; Leger, Gabriel C; Litvan, Irene; Ljubenkov, Peter A; Lucente, Diane E; Mackenzie, Ian R; Manoochehri, Masood; Masdeu, Joseph C; McGinnis, Scott; Mendez, Mario F; Miller, Bruce L; Miyagawa, Toji; Nelson, Kevin M; Onyike, Chiadi U; Pantelyat, Alex; Pascual, Belen; Pearlman, Rodney; Petrucelli, Leonard; Rademakers, Rosa; Ramos, Eliana Marisa; Rankin, Katherine; Rascovsky, Katya; Rexach, Jessica E; Ritter, Aaron; Roberson, Erik D; Rojas, Julio C; Sabbagh, Marwan N; Salmon, David P; Savica, Rodolfo; Seeley, William W; Staffaroni, Adam M; Syrjanen, Jeremy; Tartaglia, Carmela; Tatton, Nadine; Taylor, Joanne; Toga, Arthur W; Weintraub, Sandra; Wheaton, Diana; Wong, Bonnie; Wszolek, Zbigniew
- Published
- 2020
46. Comprehensive cross-sectional and longitudinal analyses of plasma neurofilament light across FTD spectrum disorders
- Author
-
Tania F. Gendron, Michael G. Heckman, Launia J. White, Austin M. Veire, Otto Pedraza, Alexander R. Burch, Andrea C. Bozoki, Bradford C. Dickerson, Kimiko Domoto-Reilly, Tatiana Foroud, Leah K. Forsberg, Douglas R. Galasko, Nupur Ghoshal, Neill R. Graff-Radford, Murray Grossman, Hilary W. Heuer, Edward D. Huey, Ging-Yuek R. Hsiung, David J. Irwin, Daniel I. Kaufer, Gabriel C. Leger, Irene Litvan, Joseph C. Masdeu, Mario F. Mendez, Chiadi U. Onyike, Belen Pascual, Aaron Ritter, Erik D. Roberson, Julio C. Rojas, Maria Carmela Tartaglia, Zbigniew K. Wszolek, Howard Rosen, Bradley F. Boeve, Adam L. Boxer, Leonard Petrucelli, Brian S. Appleby, Sami Barmada, Yvette Bordelon, Hugo Botha, Danielle Brushaber, David Clark, Giovanni Coppola, Ryan Darby, Katrina Devick, Dennis Dickson, Kelley Faber, Anne Fagan, Julie A. Fields, Ralitza Gavrilova, Daniel Geschwind, Jill Goldman, Jonathon Graff-Radford, Ian Grant, David T. Jones, Kejal Kantarci, Diana Kerwin, David S. Knopman, John Kornak, Walter Kremers, Maria Lapid, Argentina Lario Lago, Peter Ljubenkov, Diane Lucente, Ian R. Mackenzie, Scott McGinnis, Carly Mester, Bruce L. Miller, Peter Pressman, Rosa Rademakers, Vijay K. Ramanan, E. Marisa Ramos, Katherine P. Rankin, Meghana Rao, Katya Rascovsky, Rodolfo Savica, William Seeley, Adam M. Staffaroni, Jeremy Syrjanen, Jack Taylor, Lawren VandeVrede, Sandra Weintraub, and Bonnie Wong
- Subjects
Cross-Sectional Studies ,Pick Disease of the Brain ,Neurofilament Proteins ,Frontotemporal Dementia ,Intermediate Filaments ,Humans ,Syndrome ,General Biochemistry, Genetics and Molecular Biology - Abstract
Frontotemporal dementia (FTD) therapy development is hamstrung by a lack of susceptibility, diagnostic, and prognostic biomarkers. Blood neurofilament light (NfL) shows promise as a biomarker, but studies have largely focused only on core FTD syndromes, often grouping patients with different diagnoses. To expedite the clinical translation of NfL, we avail ARTFL LEFFTDS Longitudinal Frontotemporal Lobar Degeneration (ALLFTD) study resources and conduct a comprehensive investigation of plasma NfL across FTD syndromes and in presymptomatic FTD mutation carriers. We find plasma NfL is elevated in all studied syndromes, including mild cases; increases in presymptomatic mutation carriers prior to phenoconversion; and associates with indicators of disease severity. By facilitating the identification of individuals at risk of phenoconversion, and the early diagnosis of FTD, plasma NfL can aid in participant selection for prevention or early treatment trials. Moreover, its prognostic utility would improve patient care, clinical trial efficiency, and treatment outcome estimations.
- Published
- 2022
47. Structural Features of Small Molecules Targeting the RNA Repeat Expansion That Causes Genetically Defined ALS/FTD
- Author
-
Andrei Ursu, Tania F. Gendron, Ilyas Yildirim, Kye Won Wang, Jonathan L. Chen, Shruti Choudhary, Jessica A. Bush, Jared T. Baisden, Yong Jie Zhang, Matthew D. Disney, and Leonard Petrucelli
- Subjects
0301 basic medicine ,Biology ,Molecular Dynamics Simulation ,01 natural sciences ,Biochemistry ,Article ,Small Molecule Libraries ,03 medical and health sciences ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Genetics ,C9orf72 Protein ,Molecular Structure ,010405 organic chemistry ,Extramural ,Amyotrophic Lateral Sclerosis ,RNA ,General Medicine ,medicine.disease ,Small molecule ,0104 chemical sciences ,High-Throughput Screening Assays ,G-Quadruplexes ,030104 developmental biology ,Frontotemporal Dementia ,Molecular Medicine ,Trinucleotide repeat expansion ,Frontotemporal dementia - Abstract
Genetically defined amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), collectively named c9ALS/FTD, are triggered by hexanucleotide GGGGCC repeat expansions [r(G(4)C(2))(exp)] within the C9orf 72 gene. In these diseases, neuronal loss occurs through an interplay of deleterious phenotypes, including r(G(4)C(2))(exp) RNA gain-of-function mechanisms. Herein, we identified a benzimidazole derivative, CB096, that specifically binds to a repeating 1×1 GG internal loop structure, 5′CGG/3′GGC, that is formed when r(G(4)C(2))(exp) folds. Structure−activity relationship (SAR) studies and molecular dynamics (MD) simulations were used to define the molecular interactions formed between CB096 and r(G(4)C(2))(exp) that results in the rescue of disease-associated pathways. Overall, this study reveals a unique structural feature within r(G(4)C(2))(exp) that can be exploited for the development of lead medicines and chemical probes.
- Published
- 2020
48. Plasma neurofilament light predicts mortality in patients with stroke
- Author
-
Karen Jansen-West, Bjorn Oskarsson, Alexander R. Burch, Zbigniew K. Wszolek, George K. Vilanilam, James D. Berry, Kevin N. Sheth, Michael G. Heckman, Owen A. Ross, Mohammed K. Badi, James F. Meschia, Guido J. Falcone, Thomas G. Brott, Timothy M. Miller, Ronald L. Walton, Brett Cucchiara, Tania F. Gendron, Patrick W. Johnson, Leonard Petrucelli, Lauren H Sansing, and Katharine Nicholson
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Subarachnoid hemorrhage ,medicine.medical_treatment ,Intermediate Filaments ,Article ,Brain Ischemia ,03 medical and health sciences ,0302 clinical medicine ,Neurofilament Proteins ,Modified Rankin Scale ,Internal medicine ,medicine ,Humans ,In patient ,cardiovascular diseases ,Stroke ,Intracerebral hemorrhage ,Rehabilitation ,business.industry ,General Medicine ,medicine.disease ,Clinical trial ,030104 developmental biology ,Cardiology ,Biomarker (medicine) ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Given the substantial heterogeneity of stroke brain injury, there is a clear need for a biomarker that determines the degree of neuroaxonal injury across diverse stroke types. We evaluated whether blood neurofilament light (NFL), a major component of neuronal axons, would fulfill this purpose for patients with acute cerebral infarction (ACI, N=227), aneurysmal subarachnoid hemorrhage (aSAH, N=58) or non-traumatic intracerebral hemorrhage (ICH, N=29). We additionally validated our findings in two independent cohorts of patients with ICH (N=96 and N=54) given the scarcity of blood biomarker studies for this deadliest stroke type. Compared to healthy individuals (N=79 and 48 for the discovery and validation cohorts, respectively), NFL was higher for all stroke types. NFL associated with radiographic markers of brain tissue damage. It correlated with the extent of early ischemic injury in patients with ACI, hemorrhage severity in patients with aSAH, and intracranial hemorrhage volume in patients with ICH. In all patients, NFL independently correlated with scores from the NIH Stroke Scale, the modified Rankin Scale, and the Mini-Mental State Examination at blood draw, which respectively assess neurological, functional, and cognitive status. Furthermore, higher NFL concentrations independently associated with 3- or 6-month functional disability and higher all-cause mortality. These data support NFL as a uniform method to estimate neuroaxonal injury and forecast mortality regardless of stroke mechanism. As a prognostic biomarker, blood NFL has the potential to assist with planning supportive and rehabilitation services, and improving the efficiency of acute therapeutic drug and device trials, and of recovery trials.
- Published
- 2020
49. Toward allele-specific targeting therapy and pharmacodynamic marker for spinocerebellar ataxia type 3
- Author
-
Yari Carlomagno, Paola Giunti, Yuping Song, Bjorn Oskarsson, Jan O. Aasly, Rana Hanna Al-Shaikh, Robin Labrum, Zbigniew K. Wszolek, James M. Polke, João Lemos, Henry L. Paulson, Guojun Bu, Eric R. Eggenberger, Karen Jansen-West, William D. Freeman, Hector Garcia-Moreno, Mercedes Prudencio, Marka van Blitterswijk, Osamu Onodera, Joseph H. Friedman, Ryan J. Uitti, Inês Gomes, Hayley S. McLoughlin, Mark S. LeDoux, Takuya Konno, Venka Veerappan, Nathan P. Staff, Leonard Petrucelli, John N. Caviness, Cristina Januário, Tania F. Gendron, Lillian M. Daughrity, Mari Tada, Iris Vanessa Marin Collazo, Andreas Puschmann, Takeshi Ikeuchi, Katharine Nicholson, Josephine F. Huang, Klaas J. Wierenga, Sorina Gorcenco, Christin Karremo, Matthew R. Spiegel, Akiyoshi Kakita, Jay A. van Gerpen, Judith A. Dunmore, Ronald F. Pfeiffer, Philip W. Tipton, John D. Fryer, Mark R. Pittelkow, Vikram G. Shakkottai, Natalie Byron, and Michael G. Heckman
- Subjects
Neurons ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Neurology ,business.industry ,Mutant ,Machado-Joseph Disease ,General Medicine ,medicine.disease ,Bioinformatics ,Article ,Repressor Proteins ,Clinical trial ,Polymorphism (computer science) ,Spinocerebellar ataxia ,medicine ,Humans ,Allele ,Ataxin-3 ,business ,Trinucleotide repeat expansion ,Gene ,Alleles - Abstract
Spinocerebellar ataxia type 3 (SCA3), caused by a CAG repeat expansion in the ataxin-3 gene (ATXN3), is characterized by neuronal polyglutamine (polyQ) ATXN3 protein aggregates. Although there is no cure for SCA3, gene-silencing approaches to reduce toxic polyQ ATXN3 showed promise in preclinical models. However, a major limitation in translating putative treatments for this rare disease to the clinic is the lack of pharmacodynamic markers for use in clinical trials. Here, we developed an immunoassay that readily detects polyQ ATXN3 proteins in human biological fluids and discriminates patients with SCA3 from healthy controls and individuals with other ataxias. We show that polyQ ATXN3 serves as a marker of target engagement in human fibroblasts, which may bode well for its use in clinical trials. Last, we identified a single-nucleotide polymorphism that strongly associates with the expanded allele, thus providing an exciting drug target to abrogate detrimental events initiated by mutant ATXN3. Gene-silencing strategies for several repeat diseases are well under way, and our results are expected to improve clinical trial preparedness for SCA3 therapies.
- Published
- 2020
50. Cellular and molecular phenotypes of C9orf72 ALS/FTD patient derived iPSC-microglia mono-cultures
- Author
-
Ileana Lorenzini, Eric Alsop, Jennifer Levy, Lauren M Gittings, Deepti Lall, Benjamin E Rabichow, Stephen Moore, Ryan Pevey, Lynette Bustos, Camelia Burciu, Divya Bhatia, Mo Singer, Justin Saul, Amanda McQuade, Makis Tzioras, Thomas A Mota, Amber Logemann, Jamie Rose, Sandra Almeida, Fen-Biao Gao, Michael Marks, Christopher J Donnelly, Elizabeth Hutchins, Shu-Ting Hung, Justin Ichida, Robert Bowser, Tara Spires-Jones, Mathew Blurton-Jones, Tania F Gendron, Robert H Baloh, Kendall Van Keuren-Jensen, and Rita Sattler
- Subjects
Mutation ,Microglia ,Neurodegeneration ,C9orf72 Gene ,Biology ,medicine.disease_cause ,medicine.disease ,Phenotype ,Cell biology ,Transcriptome ,medicine.anatomical_structure ,C9orf72 ,medicine ,Induced pluripotent stem cell - Abstract
Background A mutation in the C9orf72 gene is the most common genetic mutation of familial and sporadic ALS, as well as familial FTD. While prior studies have focused on elucidating the mechanisms of neuronal dysfunction and neurodegeneration associated with this genetic mutation, the contribution of microglia to disease pathogenesis in the ALS/FTD disease spectrum remains poorly understood. Methods Here, we generated a new disease model consisting of cultured C9orf72 ALS/FTD patient-derived induced pluripotent stem cells differentiated into microglia (iPSC-MG). We used this model to study the intrinsic cellular and molecular phenotypes of microglia triggered by the C9orf72 gene mutation. Results We show that C9orf72 ALS/FTD iPSC-MG have a similar transcriptional profile compared to control iPSC-MG, despite the presence of C9orf72-associated phenotypes including reduced C9orf72 protein levels and dipeptide-repeat protein translation. Interestingly, C9orf72 ALS/FTD iPSC-MG exhibit intrinsic dysfunction of phagocytic activity upon exposure to Aβ or brain synaptoneurosomes and display a heightened inflammatory response. Detailed analysis of the endosomal and lysosomal pathways revealed altered expression of endosomal marker early endosome antigen 1 and lysosomal associated membrane protein 1 in C9orf72 ALS/FTD iPSC-MG, which was confirmed in patient postmortem tissues. Conclusions These findings demonstrate that unstimulated C9orf72 iPSC-MG mono-cultures share a largely similar transcriptome profile with control microglia, despite the presence of C9orf72 disease phenotypes. The dysfunction of the endosomal-lysosomal pathway as demonstrated by aberrant microglia phagocytosis and engulfment of cellular debris and brain pathogens suggests that disease-related microglia phenotypes are not intrinsic but instead require microglia to be activated. In summary, the C9orf72 iPSC-MG culture system provides a novel human disease model to study the role of microglia in C9orf72 ALS/FTD disease pathogenesis.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.