Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dis Model Mech ; 17(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38469687

RESUMO

Protein homeostasis is perturbed in aging-related neurodegenerative diseases called tauopathies, which are pathologically characterized by aggregation of the microtubule-associated protein tau (encoded by the human MAPT gene). Transgenic Caenorhabditis elegans serve as a powerful model organism to study tauopathy disease mechanisms, but moderating transgenic expression level has proven problematic. To study neuronal tau proteostasis, we generated a suite of transgenic strains expressing low, medium or high levels of Dendra2::tau fusion proteins by comparing integrated multicopy transgene arrays with single-copy safe-harbor locus strains generated by recombinase-mediated cassette exchange. Multicopy Dendra2::tau strains exhibited expression level-dependent neuronal dysfunction that was modifiable by known genetic suppressors or an enhancer of tauopathy. Single-copy Dendra2::tau strains lacked distinguishable phenotypes on their own but enabled detection of enhancer-driven neuronal dysfunction. We used multicopy Dendra2::tau strains in optical pulse-chase experiments measuring tau turnover in vivo and found that Dendra2::tau turned over faster than the relatively stable Dendra2. Furthermore, Dendra2::tau turnover was dependent on the protein expression level and independent of co-expression with human TDP-43 (officially known as TARDBP), an aggregating protein interacting with pathological tau. We present Dendra2::tau transgenic C. elegans as a novel tool for investigating molecular mechanisms of tau proteostasis.


Assuntos
Proteínas de Caenorhabditis elegans , Proteostase , Proteínas tau , Animais , Humanos , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animais de Doenças , Proteínas tau/genética , Proteínas tau/metabolismo , Tauopatias/metabolismo
2.
Neurobiol Dis ; 193: 106441, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38378122

RESUMO

Alzheimer's disease (AD), the most common aging-associated neurodegenerative dementia disorder, is defined by the presence of amyloid beta (Aß) and tau aggregates in the brain. However, more than half of patients also exhibit aggregates of the protein TDP-43 as a secondary pathology. The presence of TDP-43 pathology in AD is associated with increased tau neuropathology and worsened clinical outcomes in AD patients. Using C. elegans models of mixed pathology in AD, we have previously shown that TDP-43 specifically synergizes with tau but not Aß, resulting in enhanced neuronal dysfunction, selective neurodegeneration, and increased accumulation of pathological tau. However, cellular responses to co-morbid tau and TDP-43 preceding neurodegeneration have not been characterized. In this study, we evaluate transcriptomic changes at time-points preceding frank neuronal loss using a C. elegans model of tau and TDP-43 co-expression (tau-TDP-43 Tg). We find significant differential expression and exon usage in genes enriched in multiple pathways including lipid metabolism and lysosomal degradation. We note that early changes in tau-TDP-43 Tg resemble changes with tau alone, but a unique expression signature emerges during aging. We test loss-of-function mutations in a subset of tau and TDP-43 responsive genes, identifying new modifiers of neurotoxicity. Characterizing early cellular responses to tau and TDP-43 co-pathology is critical for understanding protective and pathogenic responses to mixed proteinopathies, and an important step in developing therapeutic strategies protecting against pathological tau and TDP-43 in AD.


Assuntos
Doença de Alzheimer , Tauopatias , Animais , Humanos , Proteínas tau/genética , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/genética , Caenorhabditis elegans/genética , Tauopatias/genética , Doença de Alzheimer/metabolismo , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica
3.
MicroPubl Biol ; 20232023.
Artigo em Inglês | MEDLINE | ID: mdl-37602280

RESUMO

Expression of human tau in C. elegans neurons causes progressive, age-associated loss of motor coordination, selective neurodegeneration, and shortened lifespan. Loss of function (LOF) mutations in the conserved gene sut-2 protects against progressive motor uncoordination and neurodegeneration in models of tauopathy. To determine whether sut-2 LOF also protects against shortened lifespan of tau transgenic C. elegans , we conducted lifespan assays comparing four different alleles of sut-2 . We found that sut-2 LOF robustly suppresses the shortened lifespan of tau transgenic animals. We also demonstrate that tau transgenic C. elegans exhibit hyperactive pharyngeal pumping, which is restored by sut-2 LOF.

4.
Dis Model Mech ; 15(4)2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35178571

RESUMO

Although amyloid ß (Aß) and tau aggregates define the neuropathology of Alzheimer's disease (AD), TDP-43 has recently emerged as a co-morbid pathology in more than half of patients with AD. Individuals with concomitant Aß, tau and TDP-43 pathology experience accelerated cognitive decline and worsened brain atrophy, but the molecular mechanisms of TDP-43 neurotoxicity in AD are unknown. Synergistic interactions among Aß, tau and TDP-43 may be responsible for worsened disease outcomes. To study the biology underlying this process, we have developed new models of protein co-morbidity using the simple animal Caenorhabditis elegans. We demonstrate that TDP-43 specifically enhances tau but not Aß neurotoxicity, resulting in neuronal dysfunction, pathological tau accumulation and selective neurodegeneration. Furthermore, we find that synergism between tau and TDP-43 is rescued by loss-of-function of the robust tau modifier sut-2. Our results implicate enhanced tau neurotoxicity as the primary driver underlying worsened clinical and neuropathological phenotypes in AD with TDP-43 pathology, and identify cell-type specific sensitivities to co-morbid tau and TDP-43. Determining the relationship between co-morbid TDP-43 and tau is crucial to understand, and ultimately treat, mixed pathology AD.


Assuntos
Doença de Alzheimer , Proteínas de Caenorhabditis elegans , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteínas de Ligação a Poli(A) , Proteínas tau/metabolismo
5.
J Vis Exp ; (175)2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34542535

RESUMO

The neurodegenerative disease amyotrophic lateral sclerosis (ALS) features progressive loss of motor neurons accompanied by muscle weakness and motor impairment that worsens with time. While considerable advances have been made in determining genetic drivers of ALS for a subset of patients, the majority of cases have an unknown etiology. Further, the mechanisms underlying motor neuron dysfunction and degeneration are not well understood; therefore, there is an ongoing need to develop and characterize representative models to study these processes. Caenorhabditis elegans can adapt their movement to the physical constraints of their surroundings, with two primary movement paradigms studied in a laboratory environment- crawling on a solid surface and swimming in liquid. These represent a complex interplay between sensation, motor neurons, and muscles. C. elegans models of ALS can exhibit impairment in one or both of these movement paradigms. This protocol describes two sensitive assays for evaluating motility in C. elegans: an optimized radial locomotion assay measuring crawling on a solid surface and an automated method for tracking and analyzing swimming in liquid (thrashing). In addition to the characterization of baseline motor impairment of ALS models, these assays can detect suppression or enhancement of the phenotypes from genetic or small molecule interventions. Thus, these methods have utility for studying ALS models and any C. elegans strain that exhibits altered motility.


Assuntos
Esclerose Lateral Amiotrófica , Transtornos Motores , Doenças Neurodegenerativas , Esclerose Lateral Amiotrófica/genética , Animais , Caenorhabditis elegans , Modelos Animais de Doenças , Humanos , Neurônios Motores
7.
Acta Neuropathol Commun ; 7(1): 91, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31174609

RESUMO

Alzheimer's disease neuropathologic change (ADNC) is defined by progressive accumulation of ß-amyloid plaques and hyperphosphorylated tau (pTau) neurofibrillary tangles across diverse regions of brain. Non-demented individuals who reach advanced age without significant ADNC are considered to be resistant to AD, while those burdened with ADNC are considered to be resilient. Understanding mechanisms underlying ADNC resistance and resilience may provide important clues to treating and/or preventing AD associated dementia. ADNC criteria for resistance and resilience are not well-defined, so we developed stringent pathologic cutoffs for non-demented subjects to eliminate cases of borderline pathology. We identified 14 resistant (85+ years old, non-demented, Braak stage ≤ III, CERAD absent) and 7 resilient (non-demented, Braak stage VI, CERAD frequent) individuals out of 684 autopsies from the Adult Changes in Thought study, a long-standing community-based cohort. We matched each resistant or resilient subject to a subject with dementia and severe ADNC (Braak stage VI, CERAD frequent) by age, sex, year of death, and post-mortem interval. We expanded the neuropathologic evaluation to include quantitative approaches to assess neuropathology and found that resilient participants had lower neocortical pTau burden despite fulfilling criteria for Braak stage VI. Moreover, limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) was robustly associated with clinical dementia and was more prevalent in cases with high pTau burden, supporting the notion that resilience to ADNC may depend, in part, on resistance to pTDP-43 pathology. To probe for interactions between tau and TDP-43, we developed a C. elegans model of combined human (h) Tau and TDP-43 proteotoxicity, which exhibited a severe degenerative phenotype most compatible with a synergistic, rather than simply additive, interaction between hTau and hTDP-43 neurodegeneration. Pathways that underlie this synergy may present novel therapeutic targets for the prevention and treatment of AD.


Assuntos
Doença de Alzheimer/patologia , Proteínas de Ligação a DNA , Vida Independente , Sistema Límbico/patologia , Neocórtex/patologia , Resiliência Psicológica , Proteínas tau , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Animais , Caenorhabditis elegans , Estudos de Coortes , Proteínas de Ligação a DNA/genética , Feminino , Humanos , Vida Independente/psicologia , Masculino , Testes de Estado Mental e Demência , Estudos Prospectivos , Proteínas tau/genética
9.
Sci Rep ; 8(1): 975, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343694

RESUMO

Parasitic helminths infect over 1 billion people worldwide, while current treatments rely on a limited arsenal of drugs. To expedite drug discovery, we screened a small-molecule library of compounds with histories of use in human clinical trials for anthelmintic activity against the soil nematode Caenorhabditis elegans. From this screen, we found that the neuromodulatory drugs sertraline, paroxetine, and chlorpromazine kill C. elegans at multiple life stages including embryos, developing larvae and gravid adults. These drugs act rapidly to inhibit C. elegans feeding within minutes of exposure. Sertraline, paroxetine, and chlorpromazine also decrease motility of adult Trichuris muris whipworms, prevent hatching and development of Ancylostoma caninum hookworms and kill Schistosoma mansoni flatworms, three widely divergent parasitic helminth species. C. elegans mutants with resistance to known anthelmintic drugs such as ivermectin are equally or more susceptible to these three drugs, suggesting that they may act on novel targets to kill worms. Sertraline, paroxetine, and chlorpromazine have long histories of use clinically as antidepressant or antipsychotic medicines. They may represent new classes of anthelmintic drug that could be used in combination with existing front-line drugs to boost effectiveness of anti-parasite treatment as well as offset the development of parasite drug resistance.


Assuntos
Anti-Helmínticos/farmacologia , Clorpromazina/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Paroxetina/farmacologia , Sertralina/farmacologia , Ancylostoma/efeitos dos fármacos , Animais , Caenorhabditis elegans/efeitos dos fármacos , Descoberta de Drogas/métodos , Reposicionamento de Medicamentos/métodos , Schistosoma mansoni/efeitos dos fármacos
10.
Acta Neuropathol ; 132(4): 545-61, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27473149

RESUMO

Detergent insoluble inclusions of TDP-43 protein are hallmarks of the neuropathology in over 90 % of amyotrophic lateral sclerosis (ALS) cases and approximately half of frontotemporal dementia (FTLD-TDP) cases. In TDP-43 proteinopathy disorders, lesions containing aggregated TDP-43 protein are extensively post-translationally modified, with phosphorylated TDP-43 (pTDP) being the most consistent and robust marker of pathological TDP-43 deposition. Abnormally phosphorylated TDP-43 has been hypothesized to mediate TDP-43 toxicity in many neurodegenerative disease models. To date, several different kinases have been implicated in the genesis of pTDP, but no phosphatases have been shown to reverse pathological TDP-43 phosphorylation. We have identified the phosphatase calcineurin as an enzyme binding to and catalyzing the removal of pathological C-terminal phosphorylation of TDP-43 in vitro. In C. elegans models of TDP-43 proteinopathy, genetic elimination of calcineurin results in accumulation of excess pTDP, exacerbated motor dysfunction, and accelerated neurodegenerative changes. In cultured human cells, treatment with FK506 (tacrolimus), a calcineurin inhibitor, results in accumulation of pTDP species. Lastly, calcineurin co-localizes with pTDP in degenerating areas of the central nervous system in subjects with FTLD-TDP and ALS. Taken together, these findings suggest calcineurin acts on pTDP as a phosphatase in neurons. Furthermore, patient treatment with calcineurin inhibitors may have unappreciated adverse neuropathological consequences.


Assuntos
Calcineurina/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Proteinopatias TDP-43/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Caenorhabditis elegans , Proteínas de Ligação a DNA/metabolismo , Corpos de Inclusão/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Proteinopatias TDP-43/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...