Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41.682
Filter
Add more filters

Publication year range
1.
Cell ; 187(8): 1971-1989.e16, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38521060

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) share many clinical, pathological, and genetic features, but a detailed understanding of their associated transcriptional alterations across vulnerable cortical cell types is lacking. Here, we report a high-resolution, comparative single-cell molecular atlas of the human primary motor and dorsolateral prefrontal cortices and their transcriptional alterations in sporadic and familial ALS and FTLD. By integrating transcriptional and genetic information, we identify known and previously unidentified vulnerable populations in cortical layer 5 and show that ALS- and FTLD-implicated motor and spindle neurons possess a virtually indistinguishable molecular identity. We implicate potential disease mechanisms affecting these cell types as well as non-neuronal drivers of pathogenesis. Finally, we show that neuron loss in cortical layer 5 tracks more closely with transcriptional identity rather than cellular morphology and extends beyond previously reported vulnerable cell types.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Lobar Degeneration , Prefrontal Cortex , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/genetics , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Gene Expression Profiling , Neurons/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Single-Cell Gene Expression Analysis
2.
Cell ; 186(20): 4260-4270, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37729908

ABSTRACT

Recent Aß-immunotherapy trials have yielded the first clear evidence that removing aggregated Aß from the brains of symptomatic patients can slow the progression of Alzheimer's disease. The clinical benefit achieved in these trials has been modest, however, highlighting the need for both a deeper understanding of disease mechanisms and the importance of intervening early in the pathogenic cascade. An immunoprevention strategy for Alzheimer's disease is required that will integrate the findings from clinical trials with mechanistic insights from preclinical disease models to select promising antibodies, optimize the timing of intervention, identify early biomarkers, and mitigate potential side effects.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Alzheimer Disease/prevention & control , Alzheimer Disease/therapy , Amyloid beta-Peptides , Antibodies/therapeutic use , Immunotherapy , Animals
3.
Cell ; 184(17): 4547-4563.e17, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34314701

ABSTRACT

Frontotemporal dementia (FTD) because of MAPT mutation causes pathological accumulation of tau and glutamatergic cortical neuronal death by unknown mechanisms. We used human induced pluripotent stem cell (iPSC)-derived cerebral organoids expressing tau-V337M and isogenic corrected controls to discover early alterations because of the mutation that precede neurodegeneration. At 2 months, mutant organoids show upregulated expression of MAPT, glutamatergic signaling pathways, and regulators, including the RNA-binding protein ELAVL4, and increased stress granules. Over the following 4 months, mutant organoids accumulate splicing changes, disruption of autophagy function, and build-up of tau and P-tau-S396. By 6 months, tau-V337M organoids show specific loss of glutamatergic neurons as seen in individuals with FTD. Mutant neurons are susceptible to glutamate toxicity, which can be rescued pharmacologically by the PIKFYVE kinase inhibitor apilimod. Our results demonstrate a sequence of events that precede neurodegeneration, revealing molecular pathways associated with glutamate signaling as potential targets for therapeutic intervention in FTD.


Subject(s)
Cerebrum/pathology , ELAV-Like Protein 4/genetics , Glutamic Acid/metabolism , Mutation/genetics , Neurons/pathology , Organoids/metabolism , RNA Splicing/genetics , tau Proteins/genetics , Autophagy/drug effects , Autophagy/genetics , Biomarkers/metabolism , Body Patterning/drug effects , Body Patterning/genetics , Cell Death/drug effects , Cell Line , Humans , Hydrazones/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Morpholines/pharmacology , Neurons/drug effects , Neurons/metabolism , Organoids/drug effects , Organoids/ultrastructure , Phosphorylation/drug effects , Pyrimidines/pharmacology , RNA Splicing/drug effects , Signal Transduction/drug effects , Stress Granules/drug effects , Stress Granules/metabolism , Synapses/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
4.
Cell ; 173(3): 665-676.e14, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29551272

ABSTRACT

Class 2 CRISPR-Cas systems endow microbes with diverse mechanisms for adaptive immunity. Here, we analyzed prokaryotic genome and metagenome sequences to identify an uncharacterized family of RNA-guided, RNA-targeting CRISPR systems that we classify as type VI-D. Biochemical characterization and protein engineering of seven distinct orthologs generated a ribonuclease effector derived from Ruminococcus flavefaciens XPD3002 (CasRx) with robust activity in human cells. CasRx-mediated knockdown exhibits high efficiency and specificity relative to RNA interference across diverse endogenous transcripts. As one of the most compact single-effector Cas enzymes, CasRx can also be flexibly packaged into adeno-associated virus. We target virally encoded, catalytically inactive CasRx to cis elements of pre-mRNA to manipulate alternative splicing, alleviating dysregulated tau isoform ratios in a neuronal model of frontotemporal dementia. Our results present CasRx as a programmable RNA-binding module for efficient targeting of cellular RNA, enabling a general platform for transcriptome engineering and future therapeutic development.


Subject(s)
CRISPR-Cas Systems , Computational Biology/methods , Genetic Engineering/methods , Protein Engineering/methods , RNA/analysis , Alternative Splicing , Animals , Bacterial Proteins/metabolism , Cell Differentiation , Escherichia coli/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Lentivirus/genetics , Mice , RNA Interference , RNA, Guide, Kinetoplastida/genetics , Ruminococcus , Sequence Analysis, RNA , Transcriptome
5.
Cell ; 173(3): 720-734.e15, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677515

ABSTRACT

Reversible phase separation underpins the role of FUS in ribonucleoprotein granules and other membrane-free organelles and is, in part, driven by the intrinsically disordered low-complexity (LC) domain of FUS. Here, we report that cooperative cation-π interactions between tyrosines in the LC domain and arginines in structured C-terminal domains also contribute to phase separation. These interactions are modulated by post-translational arginine methylation, wherein arginine hypomethylation strongly promotes phase separation and gelation. Indeed, significant hypomethylation, which occurs in FUS-associated frontotemporal lobar degeneration (FTLD), induces FUS condensation into stable intermolecular ß-sheet-rich hydrogels that disrupt RNP granule function and impair new protein synthesis in neuron terminals. We show that transportin acts as a physiological molecular chaperone of FUS in neuron terminals, reducing phase separation and gelation of methylated and hypomethylated FUS and rescuing protein synthesis. These results demonstrate how FUS condensation is physiologically regulated and how perturbations in these mechanisms can lead to disease.


Subject(s)
Arginine/chemistry , Molecular Chaperones/chemistry , RNA-Binding Protein FUS/chemistry , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cations , DNA Methylation , Frontotemporal Dementia/metabolism , Frontotemporal Lobar Degeneration/metabolism , Humans , Microscopy, Atomic Force , Microscopy, Fluorescence , Protein Binding , Protein Domains , Protein Processing, Post-Translational , Protein Structure, Secondary , RNA-Binding Protein FUS/metabolism , Tyrosine/chemistry , Xenopus laevis
6.
Annu Rev Neurosci ; 47(1): 123-143, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38663088

ABSTRACT

Over 40% of the human genome is composed of retrotransposons, DNA species that hold the potential to replicate via an RNA intermediate and are evolutionarily related to retroviruses. Retrotransposons are most studied for their ability to jump within a genome, which can cause DNA damage and novel insertional mutations. Retrotransposon-encoded products, including viral-like proteins, double-stranded RNAs, and extrachromosomal circular DNAs, can also be potent activators of the innate immune system. A growing body of evidence suggests that retrotransposons are activated in age-related neurodegenerative disorders and that such activation causally contributes to neurotoxicity. Here we provide an overview of retrotransposon biology and outline evidence of retrotransposon activation in age-related neurodegenerative disorders, with an emphasis on those involving TAR-DNA binding protein-43 (TDP-43) and tau. Studies to date provide the basis for ongoing clinical trials and hold promise for innovative strategies to ameliorate the adverse effects of retrotransposon dysregulation in neurodegenerative disorders.


Subject(s)
Aging , Endogenous Retroviruses , Neurodegenerative Diseases , Retroelements , Humans , Neurodegenerative Diseases/genetics , Retroelements/genetics , Endogenous Retroviruses/genetics , Animals , Aging/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , tau Proteins/genetics , tau Proteins/metabolism
7.
Cell ; 171(5): 994-1000, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149615

ABSTRACT

Eukaryotic translation is tightly regulated to ensure that protein production occurs at the right time and place. Recent studies on abnormal repeat proteins, especially in age-dependent neurodegenerative diseases caused by nucleotide repeat expansion, have highlighted or identified two forms of unconventional translation initiation: usage of AUG-like sites (near cognates) or repeat-associated non-AUG (RAN) translation. We discuss how repeat proteins may differ due to not just unconventional initiation, but also ribosomal frameshifting and/or imperfect repeat DNA replication, expansion, and repair, and we highlight how research on translation of repeats may uncover insights into the biology of translation and its contribution to disease.


Subject(s)
Neurodegenerative Diseases/genetics , Protein Biosynthesis , Animals , Codon, Initiator , Frameshifting, Ribosomal , Humans , Neurodegenerative Diseases/metabolism , Open Reading Frames , Regulatory Sequences, Ribonucleic Acid , Trinucleotide Repeat Expansion
8.
CA Cancer J Clin ; 73(3): 320-338, 2023.
Article in English | MEDLINE | ID: mdl-36512303

ABSTRACT

As many countries experience population aging, patients with cancer are becoming older and have more preexisting comorbidities, which include prevalent, age-related, chronic conditions such as dementia. People living with dementia (PLWD) are vulnerable to health disparities, and dementia has high potential to complicate and adversely affect care and outcomes across the cancer trajectory. This report offers an overview of dementia and its prevalence among patients with cancer and a summary of the research literature examining cancer care for PLWD. The reviewed research indicates that PLWD are more likely to have cancer diagnosed at an advanced stage, receive no or less extensive cancer treatment, and have poorer survival after a cancer diagnosis. These cancer disparities do not necessarily signify inappropriately later diagnosis or lower treatment of people with dementia as a group, and they are arguably less feasible and appropriate targets for care optimization. The reviewed research indicates that PLWD also have an increased risk of cancer-related emergency presentations, lower quality processes of cancer-related decision making, accessibility-related barriers to cancer investigations and treatment, higher experienced treatment burden and higher caregiver burden for families, and undertreated cancer-related pain. The authors propose that optimal cancer care for PLWD should focus on proactively minimizing these risk areas and thus must be highly person-centered, with holistic decision making, individualized reasonable adjustments to practice, and strong inclusion and support of family carers. Comprehensive recommendations are made for clinical practice and future research to help clinicians and providers deliver best and equitable cancer care for PLWD and their families.


Subject(s)
Dementia , Neoplasms , Humans , Dementia/complications , Dementia/diagnosis , Dementia/therapy , Caregivers , Neoplasms/complications , Neoplasms/therapy
9.
Physiol Rev ; 101(3): 1047-1081, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33475022

ABSTRACT

The history of Alzheimer's disease (AD) started in 1907, but we needed to wait until the end of the century to identify the components of pathological hallmarks and genetic subtypes and to formulate the first pathogenic hypothesis. Thanks to biomarkers and new technologies, the concept of AD then rapidly changed from a static view of an amnestic dementia of the presenium to a biological entity that could be clinically manifested as normal cognition or dementia of different types. What is clearly emerging from studies is that AD is heterogeneous in each aspect, such as amyloid composition, tau distribution, relation between amyloid and tau, clinical symptoms, and genetic background, and thus it is probably impossible to explain AD with a single pathological process. The scientific approach to AD suffers from chronological mismatches between clinical, pathological, and technological data, causing difficulty in conceiving diagnostic gold standards and in creating models for drug discovery and screening. A recent mathematical computer-based approach offers the opportunity to study AD in real life and to provide a new point of view and the final missing pieces of the AD puzzle.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/metabolism , Brain/pathology , Peptide Fragments/metabolism , Plaque, Amyloid/diagnosis , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Biomarkers/metabolism , Brain/metabolism , Humans , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
10.
Immunol Rev ; 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39351782

ABSTRACT

Within a few years, autoantibodies targeting the nervous system resulted in a novel disease classification. For several of them, which we termed 'established', direct pathogenicity has been proven and now guides diagnostic pathways and early immunotherapy. For a rapidly growing number of further anti-neuronal autoantibodies, the role in disease is less clear. Increasing evidence suggests that they could contribute to disease, by playing a modulating role on brain function. We therefore suggest a three-level classification of neurological autoantibodies according to the degree of experimentally proven pathogenicity and strength of clinical association: established, emerging, explorative. This may facilitate focusing on clinical constellations in which autoantibody-mediated mechanisms have not been assumed previously, including autoimmune psychosis and dementia, cognitive impairment in cancer, and neurodegenerative diseases. Based on recent data reviewed here, humoral autoimmunity may represent an additional "super-system" for brain health. The "brain antibody-ome", that is, the composition of thousands of anti-neuronal autoantibodies, may shape neuronal function not only in disease, but even in healthy aging. Towards this novel concept, extensive research will have to elucidate pathogenicity from the atomic to the clinical level, autoantibody by autoantibody. Such profiling can uncover novel biomarkers, enhance our understanding of underlying mechanisms, and identify selective therapies.

11.
Am J Hum Genet ; 111(9): 1848-1863, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39079537

ABSTRACT

Transcriptome-wide association study (TWAS) tools have been applied to conduct proteome-wide association studies (PWASs) by integrating proteomics data with genome-wide association study (GWAS) summary data. The genetic effects of PWAS-identified significant genes are potentially mediated through genetically regulated protein abundance, thus informing the underlying disease mechanisms better than GWAS loci. However, existing TWAS/PWAS tools are limited by considering only one statistical model. We propose an omnibus PWAS pipeline to account for multiple statistical models and demonstrate improved performance by simulation and application studies of Alzheimer disease (AD) dementia. We employ the Aggregated Cauchy Association Test to derive omnibus PWAS (PWAS-O) p values from PWAS p values obtained by three existing tools assuming complementary statistical models-TIGAR, PrediXcan, and FUSION. Our simulation studies demonstrated improved power, with well-calibrated type I error, for PWAS-O over all three individual tools. We applied PWAS-O to studying AD dementia with reference proteomic data profiled from dorsolateral prefrontal cortex of postmortem brains from individuals of European ancestry. We identified 43 risk genes, including 5 not identified by previous studies, which are interconnected through a protein-protein interaction network that includes the well-known AD risk genes TOMM40, APOC1, and APOC2. We also validated causal genetic effects mediated through the proteome for 27 (63%) PWAS-O risk genes, providing insights into the underlying biological mechanisms of AD dementia and highlighting promising targets for therapeutic development. PWAS-O can be easily applied to studying other complex diseases.


Subject(s)
Alzheimer Disease , Genetic Predisposition to Disease , Genome-Wide Association Study , Proteome , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Apolipoprotein C-I/genetics , Apolipoprotein C-I/metabolism , Polymorphism, Single Nucleotide , Risk Factors , Transcriptome , Mitochondrial Precursor Protein Import Complex Proteins
12.
Proc Natl Acad Sci U S A ; 121(41): e2412017121, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39352934

ABSTRACT

Major initiatives attempt to prevent dementia by targeting modifiable risk factors. Low education is frequently pointed to, due to its relationship with dementia. Impact of education is difficult to assess, however, because of associations with multiple other factors, requiring large population-representative samples to tease the relationships apart. We studied 207,814 Norwegian men born between 1950 and 1959 who underwent compulsory cognitive testing during military conscription as young adults, to systematically test associations of education, cognition, and other important factors. Participants were grouped into five education levels and seven cognitive levels. A total of 1,521 were diagnosed with dementia between ages 60 and 69 y. While having compulsory education only was associated with increased risk (Hazard ratio [HR] = 1.37, CI: 1.17 to 1.60), this association was markedly attenuated when controlling for cognitive test scores (HR = 1.08, CI: 0.91 to 1.28). In contrast, low cognitive score was associated with double risk of later diagnosis, even when controlling for education (HR = 2.00, CI: 1.65 to 2.42). This relationship survived controlling for early-life socioeconomic status and replicated within pairs of brothers. This suggests that genetic and environmental factors shared within families, e.g., common genetics, parental education, socioeconomic status, or other shared experiences, cannot account for the association. Rather, independent, nonfamilial factors are more important. In contrast, within-family factors accounted for the relationship between low education and diagnosis risk. In conclusion, implementing measures to increase cognitive function in childhood and adolescence appears to be a more promising strategy for reducing dementia burden.


Subject(s)
Cognition , Dementia , Educational Status , Humans , Dementia/epidemiology , Dementia/prevention & control , Male , Cognition/physiology , Risk Factors , Middle Aged , Aged , Norway/epidemiology , Adolescent
13.
Proc Natl Acad Sci U S A ; 121(40): e2321078121, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39298474

ABSTRACT

Evidence on cash transfers as a population-level intervention to support healthy cognitive aging in low-income settings is sparse. We assessed the effect of a cash transfer intervention on cognitive aging outcomes in older South African adults. We leveraged the overlap in the sampling frames of a Phase 3 randomized cash transfer trial [HIV Prevention Trial Network (HPTN) 068, 2011-2015] and an aging cohort [Health and Aging in Africa: A Longitudinal Study of an INDEPTH Community (HAALSI), 2014-2022] in rural Mpumalanga Province, South Africa. In 2011/12, young women and their primary caregivers were randomly assigned 1:1 to receive a monthly cash transfer or control. In 2014/2015, 862 adults aged 40+ y living in trial households were enrolled in the HAALSI cohort, with cognitive data collected in three waves over 7 y. We estimated the impact of the intervention on rate of memory decline and dementia probability scores. Memory decline in the cash transfer arm was 0.03 SD units (95% CI: 0.002, 0.05) slower per year than in the control arm. Dementia probability scores were three percentage points lower in the cash transfer arm than the control arm (ß = -0.03; 95% CI: -0.05, -0.001). Effects were consistent across subgroups. A modestly sized household cash transfer delivered over a short period in mid- to later-life led to a meaningful slowing of memory decline and reduction in dementia probability 7 y later. Cash transfer programs could help stem the tide of new dementia cases in economically vulnerable populations in the coming decades.


Subject(s)
Dementia , Rural Population , Humans , South Africa/epidemiology , Female , Male , Dementia/epidemiology , Dementia/economics , Dementia/prevention & control , Middle Aged , Aged , Longitudinal Studies , Poverty , Adult , Memory Disorders/prevention & control , Memory Disorders/epidemiology , Memory Disorders/economics , Caregivers/economics
14.
Pharmacol Rev ; 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013601

ABSTRACT

Over four decades of research support the link between Alzheimer's disease (AD) and somatostatin (somatotropin-releasing inhibitory factor, SRIF). SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-beta peptide (Aß), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation. Whereas, preclinical AD investigations show SRIF or SRIF-receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aß in the brain. Here we review the links between SRIF and AD, along with the therapeutic implications. Significance Statement Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer's disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin mediated processes has significant therapeutic potential for the treatment of Alzheimer's disease.

15.
Pharmacol Rev ; 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164116

ABSTRACT

α-Synuclein (α-Syn) aggregation in Lewy bodies and Lewy neurites has emerged as a key pathogenetic feature in Parkinson's disease (PD), Dementia with Lewy Bodies and Multiple System Atrophy. Various factors, including post-translational modifications (PTMs), can influence the propensity of α-Syn to misfold and aggregate. PTMs are biochemical modifications of a protein that occur during or after translation and are typically mediated by enzymes. PTMs modulate several characteristics of proteins including their structure, activity, localization, and stability. α-Syn undergoes various post-translational modifications, including phosphorylation, ubiquitination, SUMOylation, acetylation, glycation, O-GlcNAcylation, nitration, oxidation, polyamination, arginylation, and truncation. Different PTMs of a protein can physically interact with one another or work together to influence a particular physiological or pathological feature in a process known as PTMs crosstalk. The development of detection techniques for the co-occurrence of PTMs in recent years has uncovered previously unappreciated mechanisms of their crosstalk. This has led to the emergence of evidence supporting an association between α-Syn PTMs crosstalk and synucleinopathies. In this review, we provide a comprehensive evaluation of α-Syn PTMs, their impact on misfolding and pathogenicity, the pharmacological means of targeting them, and their potential as biomarkers of disease. We also highlight the importance of the crosstalk between these PTMs in α-Syn function and aggregation. Insight into these PTMS and the complexities of their crosstalk can improve our understanding of the pathogenesis of synucleinopathies and identify novel targets of therapeutic potential. Significance Statement α-Synuclein as a key pathogenic protein in Parkinson's disease and other synucleinopathies, making it a leading therapeutic target for disease modification. Multiple post-translational modifications occur at various sites in α-Synuclein and alter its biophysical and pathological properties, some interacting with one another to add to the complexity of the pathogenicity of this protein. This review details these modifications, their implications in disease and potential therapeutic opportunities.

16.
Trends Biochem Sci ; 47(1): 6-22, 2022 01.
Article in English | MEDLINE | ID: mdl-34366183

ABSTRACT

RNA-binding proteins (RBPs) are critical players in RNA expression and metabolism, thus, the proper regulation of this class of proteins is critical for cellular health. Regulation of RBPs often occurs through post-translational modifications (PTMs), which allow the cell to quickly and efficiently respond to cellular and environmental stimuli. PTMs have recently emerged as important regulators of RBPs implicated in neurodegenerative disorders, in particular amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here, we summarize how disease-associated PTMs influence the biophysical properties, molecular interactions, subcellular localization, and function of ALS/FTD-linked RBPs, such as FUS and TDP-43. We will discuss how PTMs are believed to play pathological, protective, or ambiguous roles in these neurodegenerative disorders.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Humans , Protein Processing, Post-Translational , RNA-Binding Protein FUS/genetics , RNA-Binding Proteins/metabolism
17.
Annu Rev Med ; 75: 99-111, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38285515

ABSTRACT

When the US Food and Drug Administration used the accelerated approval process to authorize the use of the antiamyloid drug aducanumab to treat Alzheimer's disease (AD), many people hoped this signaled a new era of disease-modifying treatment. But 2 years later, aducanumab's failure to launch provides a cautionary tale about the complexities of dementia and the need for a thorough and transparent review of the role that regulatory agencies and various stakeholders play in approving AD drugs. We highlight the events leading to aducanumab's controversial approval and discuss some of the key lessons learned from the drug's failure to deliver the hoped-for benefits. These lessons include the inherent limitations of antiamyloid strategies for a complex disease in which amyloid is only one of several pathological processes, the need for clinical trials that better reflect the diversity of communities affected by AD, the potential pitfalls of futility analyses in clinical trials, the need for greater transparency and other modifications to the approval process, and the dementia field's unreadiness to move from the highly controlled environment of clinical trials to the widespread and chronic use of resource-intensive, disease-modifying drugs in real-world treatment scenarios. People with dementia desperately need effective therapies. We hope that the aducanumab story will inspire changes to the approval process-changes that restore public trust and improve future efforts to deliver disease-modifying therapies to the clinic.


Subject(s)
Alzheimer Disease , United States , Humans , Alzheimer Disease/drug therapy , Amyloidogenic Proteins , Antibodies, Monoclonal, Humanized/therapeutic use , United States Food and Drug Administration , Amyloid beta-Peptides
18.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39226887

ABSTRACT

Plasma protein biomarkers have been considered promising tools for diagnosing dementia subtypes due to their low variability, cost-effectiveness, and minimal invasiveness in diagnostic procedures. Machine learning (ML) methods have been applied to enhance accuracy of the biomarker discovery. However, previous ML-based studies often overlook interactions between proteins, which are crucial in complex disorders like dementia. While protein-protein interactions (PPIs) have been used in network models, these models often fail to fully capture the diverse properties of PPIs due to their local awareness. This drawback increases the chance of neglecting critical components and magnifying the impact of noisy interactions. In this study, we propose a novel graph-based ML model for dementia subtype diagnosis, the graph propagational network (GPN). By propagating the independent effect of plasma proteins on PPI network, the GPN extracts the globally interactive effects between proteins. Experimental results showed that the interactive effect between proteins yielded to further clarify the differences between dementia subtype groups and contributed to the performance improvement where the GPN outperformed existing methods by 10.4% on average.


Subject(s)
Biomarkers , Blood Proteins , Dementia , Machine Learning , Protein Interaction Maps , Humans , Dementia/metabolism , Dementia/diagnosis , Blood Proteins/metabolism , Protein Interaction Mapping/methods , Algorithms , Computational Biology/methods
19.
Circ Res ; 134(6): 770-790, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38484031

ABSTRACT

Time-of-day significantly influences the severity and incidence of stroke. Evidence has emerged not only for circadian governance over stroke risk factors, but also for important determinants of clinical outcome. In this review, we provide a comprehensive overview of the interplay between chronobiology and cerebrovascular disease. We discuss circadian regulation of pathophysiological mechanisms underlying stroke onset or tolerance as well as in vascular dementia. This includes cell death mechanisms, metabolism, mitochondrial function, and inflammation/immunity. Furthermore, we present clinical evidence supporting the link between disrupted circadian rhythms and increased susceptibility to stroke and dementia. We propose that circadian regulation of biochemical and physiological pathways in the brain increase susceptibility to damage after stroke in sleep and attenuate treatment effectiveness during the active phase. This review underscores the importance of considering circadian biology for understanding the pathology and treatment choice for stroke and vascular dementia and speculates that considering a patient's chronotype may be an important factor in developing precision treatment following stroke.


Subject(s)
Circadian Clocks , Dementia, Vascular , Stroke , Humans , Circadian Rhythm , Sleep/physiology , Risk Factors , Stroke/epidemiology , Stroke/therapy , Circadian Clocks/physiology
20.
Mol Cell ; 69(3): 465-479.e7, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29358076

ABSTRACT

hnRNPA2, a component of RNA-processing membraneless organelles, forms inclusions when mutated in a syndrome characterized by the degeneration of neurons (bearing features of amyotrophic lateral sclerosis [ALS] and frontotemporal dementia), muscle, and bone. Here we provide a unified structural view of hnRNPA2 self-assembly, aggregation, and interaction and the distinct effects of small chemical changes-disease mutations and arginine methylation-on these assemblies. The hnRNPA2 low-complexity (LC) domain is compact and intrinsically disordered as a monomer, retaining predominant disorder in a liquid-liquid phase-separated form. Disease mutations D290V and P298L induce aggregation by enhancing and extending, respectively, the aggregation-prone region. Co-aggregating in disease inclusions, hnRNPA2 LC directly interacts with and induces phase separation of TDP-43. Conversely, arginine methylation reduces hnRNPA2 phase separation, disrupting arginine-mediated contacts. These results highlight the mechanistic role of specific LC domain interactions and modifications conserved across many hnRNP family members but altered by aggregation-causing pathological mutations.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group A-B/chemistry , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Arginine/genetics , Arginine/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Magnetic Resonance Imaging/methods , Methylation , Mutation , Neurons/metabolism , Neurons/pathology , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL