Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Cell ; 187(8): 1936-1954.e24, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38490196

RESUMO

Microglia are brain-resident macrophages that shape neural circuit development and are implicated in neurodevelopmental diseases. Multiple microglial transcriptional states have been defined, but their functional significance is unclear. Here, we identify a type I interferon (IFN-I)-responsive microglial state in the developing somatosensory cortex (postnatal day 5) that is actively engulfing whole neurons. This population expands during cortical remodeling induced by partial whisker deprivation. Global or microglial-specific loss of the IFN-I receptor resulted in microglia with phagolysosomal dysfunction and an accumulation of neurons with nuclear DNA damage. IFN-I gain of function increased neuronal engulfment by microglia in both mouse and zebrafish and restricted the accumulation of DNA-damaged neurons. Finally, IFN-I deficiency resulted in excess cortical excitatory neurons and tactile hypersensitivity. These data define a role for neuron-engulfing microglia during a critical window of brain development and reveal homeostatic functions of a canonical antiviral signaling pathway in the brain.


Assuntos
Encéfalo , Interferon Tipo I , Microglia , Animais , Camundongos , Interferon Tipo I/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Peixe-Zebra , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento
2.
Proc Natl Acad Sci U S A ; 120(12): e2300769120, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36927157

RESUMO

In neurodegenerative diseases, proteins fold into amyloid structures with distinct conformations (strains) that are characteristic of different diseases. However, there is a need to rapidly identify amyloid conformations in situ. Here, we use machine learning on the full information available in fluorescent excitation/emission spectra of amyloid-binding dyes to identify six distinct different conformational strains in vitro, as well as amyloid-ß (Aß) deposits in different transgenic mouse models. Our EMBER (excitation multiplexed bright emission recording) imaging method rapidly identifies conformational differences in Aß and tau deposits from Down syndrome, sporadic and familial Alzheimer's disease human brain slices. EMBER has in situ identified distinct conformational strains of tau inclusions in astrocytes, oligodendrocytes, and neurons from Pick's disease. In future studies, EMBER should enable high-throughput measurements of the fidelity of strain transmission in cellular and animal neurodegenerative diseases models, time course of amyloid strain propagation, and identification of pathogenic versus benign strains.


Assuntos
Doença de Alzheimer , Doença de Pick , Camundongos , Animais , Humanos , Microscopia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doença de Pick/metabolismo , Amiloide/metabolismo , Encéfalo/metabolismo , Camundongos Transgênicos , Proteínas tau/metabolismo , Placa Amiloide/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(13): e2220984120, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36952379

RESUMO

The amyotrophic lateral sclerosis-parkinsonism dementia complex (ALS-PDC) of Guam is an endemic neurodegenerative disease that features widespread tau tangles, occasional α-synuclein Lewy bodies, and sparse ß-amyloid (Aß) plaques distributed in the central nervous system. Extensive studies of genetic or environmental factors have failed to identify a cause of ALS-PDC. Building on prior work describing the detection of tau and Aß prions in Alzheimer's disease (AD) and Down syndrome brains, we investigated ALS-PDC brain samples for the presence of prions. We obtained postmortem frozen brain tissue from 26 donors from Guam with ALS-PDC or no neurological impairment and 71 non-Guamanian donors with AD or no neurological impairment. We employed cellular bioassays to detect the prion conformers of tau, α-synuclein, and Aß proteins in brain extracts. In ALS-PDC brain samples, we detected high titers of tau and Aß prions, but we did not detect α-synuclein prions in either cohort. The specific activity of tau and Aß prions was increased in Guam ALS-PDC compared with sporadic AD. Applying partial least squares regression to all biochemical and prion infectivity measurements, we demonstrated that the ALS-PDC cohort has a unique molecular signature distinguishable from AD. Our findings argue that Guam ALS-PDC is a distinct double-prion disorder featuring both tau and Aß prions.


Assuntos
Doença de Alzheimer , Esclerose Lateral Amiotrófica , Demência , Doenças Neurodegenerativas , Transtornos Parkinsonianos , Doenças Priônicas , Príons , Humanos , alfa-Sinucleína , Esclerose Lateral Amiotrófica/metabolismo , Demência/metabolismo , Transtornos Parkinsonianos/metabolismo , Proteínas tau/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35115402

RESUMO

The α-synuclein protein can adopt several different conformations that cause neurodegeneration. Different α-synuclein conformers cause at least three distinct α-synucleinopathies: multiple system atrophy (MSA), dementia with Lewy bodies (DLB), and Parkinson's disease (PD). In earlier studies, we transmitted MSA to transgenic (Tg) mice and cultured HEK cells both expressing mutant α-synuclein (A53T) but not to cells expressing α-synuclein (E46K). Now, we report that DLB is caused by a strain of α-synuclein prions that is distinct from MSA. Using cultured HEK cells expressing mutant α-synuclein (E46K), we found that DLB prions could be transmitted to these HEK cells. Our results argue that a third strain of α-synuclein prions likely causes PD, but further studies are needed to identify cells and/or Tg mice that express a mutant α-synuclein protein that is permissive for PD prion replication. Our findings suggest that other α-synuclein mutants should give further insights into α-synuclein prion replication, strain formation, and disease pathogenesis, all of which are likely required to discover effective drugs for the treatment of PD as well as the other α-synucleinopathies.


Assuntos
Demência/metabolismo , Doença por Corpos de Lewy/metabolismo , Atrofia de Múltiplos Sistemas/metabolismo , Príons/metabolismo , alfa-Sinucleína/metabolismo , Idoso , Linhagem Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/metabolismo , Sinucleinopatias/metabolismo
5.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35082148

RESUMO

Triggering receptor expressed on myeloid cells 2 (TREM2) is a single-pass transmembrane receptor of the immunoglobulin superfamily that is secreted in a soluble (sTREM2) form. Mutations in TREM2 have been linked to increased risk of Alzheimer's disease (AD). A prominent neuropathological component of AD is deposition of the amyloid-ß (Aß) into plaques, particularly Aß40 and Aß42. While the membrane-bound form of TREM2 is known to facilitate uptake of Aß fibrils and the polarization of microglial processes toward amyloid plaques, the role of its soluble ectodomain, particularly in interactions with monomeric or fibrillar Aß, has been less clear. Our results demonstrate that sTREM2 does not bind to monomeric Aß40 and Aß42, even at a high micromolar concentration, while it does bind to fibrillar Aß42 and Aß40 with equal affinities (2.6 ± 0.3 µM and 2.3 ± 0.4 µM). Kinetic analysis shows that sTREM2 inhibits the secondary nucleation step in the fibrillization of Aß, while having little effect on the primary nucleation pathway. Furthermore, binding of sTREM2 to fibrils markedly enhanced uptake of fibrils into human microglial and neuroglioma derived cell lines. The disease-associated sTREM2 mutant, R47H, displayed little to no effect on fibril nucleation and binding, but it decreased uptake and functional responses markedly. We also probed the structure of the WT sTREM2-Aß fibril complex using integrative molecular modeling based primarily on the cross-linking mass spectrometry data. The model shows that sTREM2 binds fibrils along one face of the structure, leaving a second, mutation-sensitive site free to mediate cellular binding and uptake.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Amiloide/genética , Peptídeos beta-Amiloides/genética , Animais , Humanos , Cinética , Glicoproteínas de Membrana/genética , Camundongos , Microglia/metabolismo , Mutação/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/genética , Placa Amiloide/metabolismo , Receptores Imunológicos/genética , Proteínas tau/genética , Proteínas tau/metabolismo
6.
Proc Natl Acad Sci U S A ; 119(46): e2212954119, 2022 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-36343257

RESUMO

Down syndrome (DS) is caused by the triplication of chromosome 21 and is the most common chromosomal disorder in humans. Those individuals with DS who live beyond age 40 y develop a progressive dementia that is similar to Alzheimer's disease (AD). Both DS and AD brains exhibit numerous extracellular amyloid plaques composed of Aß and intracellular neurofibrillary tangles composed of tau. Since AD is a double-prion disorder, we asked if both Aß and tau prions feature in DS. Frozen brains from people with DS, familial AD (fAD), sporadic AD (sAD), and age-matched controls were procured from brain biorepositories. We selectively precipitated Aß and tau prions from DS brain homogenates and measured the number of prions using cellular bioassays. In brain extracts from 28 deceased donors with DS, ranging in age from 19 to 65 y, we found nearly all DS brains had readily measurable levels of Aß and tau prions. In a cross-sectional analysis of DS donor age at death, we found that the levels of Aß and tau prions increased with age. In contrast to DS brains, the levels of Aß and tau prions in the brains of 37 fAD and sAD donors decreased as a function of age at death. Whether DS is an ideal model for assessing the efficacy of putative AD therapeutics remains to be determined.


Assuntos
Doença de Alzheimer , Síndrome de Down , Príons , Adulto , Humanos , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Estudos Transversais , Síndrome de Down/patologia , Príons/metabolismo , Proteínas tau/metabolismo
7.
Acta Neuropathol ; 148(1): 25, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39160375

RESUMO

Both wild-type and mutant tau proteins can misfold into prions and self-propagate in the central nervous system of animals and people. To extend the work of others, we investigated the molecular basis of tau prion-mediated neurodegeneration in transgenic (Tg) rats expressing mutant human tau (P301S); this line of Tg rats is denoted Tg12099. We used the rat Prnp promoter to drive the overexpression of mutant tau (P301S) in the human 0N4R isoform. In Tg12099(+/+) rats homozygous for the transgene, ubiquitous expression of mutant human tau resulted in the progressive accumulation of phosphorylated tau inclusions, including silver-positive tangles in the frontal cortices and limbic system. Signs of central nervous system dysfunction were found in terminal Tg12099(+/+) rats exhibiting severe neurodegeneration and profound atrophy of the amygdala and piriform cortex. The greatest increases in tau prion activity were found in the corticolimbic structures. In contrast to the homozygous Tg12099(+/+) rats, we found lower levels of mutant tau in the hemizygous rats, resulting in few neuropathologic changes up to 2 years of age. Notably, these hemizygous rats could be infected by intracerebral inoculation with recombinant tau fibrils or precipitated tau prions from the brain homogenates of sick, aged homozygous Tg12099(+/+) rats. Our studies argue that the regional propagation of tau prions and neurodegeneration in the Tg12099 rats resembles that found in human primary tauopathies. These findings seem likely to advance our understanding of human tauopathies and may lead to effective therapeutics for Alzheimer's disease and other tau prion disorders.


Assuntos
Encéfalo , Ratos Transgênicos , Proteínas tau , Animais , Proteínas tau/metabolismo , Proteínas tau/genética , Humanos , Ratos , Encéfalo/patologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Príons/metabolismo , Príons/genética , Tauopatias/patologia , Tauopatias/metabolismo , Tauopatias/genética , Degeneração Neural/patologia , Degeneração Neural/genética , Degeneração Neural/metabolismo , Mutação
8.
J Proteome Res ; 22(11): 3475-3488, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37847596

RESUMO

Numerous Aß proteoforms, identified in the human brain, possess differential neurotoxic and aggregation propensities. These proteoforms contribute in unknown ways to the conformations and resultant pathogenicity of oligomers, protofibrils, and fibrils in Alzheimer's disease (AD) manifestation owing to the lack of molecular-level specificity to the exact chemical composition of underlying protein products with widespread interrogating techniques, like immunoassays. We evaluated Aß proteoform flux using quantitative top-down mass spectrometry (TDMS) in a well-studied 5xFAD mouse model of age-dependent Aß-amyloidosis. Though the brain-derived Aß proteoform landscape is largely occupied by Aß1-42, 25 different forms of Aß with differential solubility were identified. These proteoforms fall into three natural groups defined by hierarchical clustering of expression levels in the context of mouse age and proteoform solubility, with each group sharing physiochemical properties associated with either N/C-terminal truncations or both. Overall, the TDMS workflow outlined may hold tremendous potential for investigating proteoform-level relationships between insoluble fibrils and soluble Aß, including low-molecular-weight oligomers hypothesized to serve as the key drivers of neurotoxicity. Similarly, the workflow may also help to validate the utility of AD-relevant animal models to recapitulate amyloidosis mechanisms or possibly explain disconnects observed in therapeutic efficacy in animal models vs humans.


Assuntos
Doença de Alzheimer , Amiloidose , Camundongos , Humanos , Animais , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/metabolismo , Camundongos Transgênicos , Modelos Animais de Doenças , Espectrometria de Massas
9.
Proc Natl Acad Sci U S A ; 115(4): E782-E791, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29311311

RESUMO

Point mutations in the amyloid-ß (Aß) coding region produce a combination of mutant and WT Aß isoforms that yield unique clinicopathologies in familial Alzheimer's disease (fAD) and cerebral amyloid angiopathy (fCAA) patients. Here, we report a method to investigate the structural variability of amyloid deposits found in fAD, fCAA, and sporadic AD (sAD). Using this approach, we demonstrate that mutant Aß determines WT Aß conformation through prion template-directed misfolding. Using principal component analysis of multiple structure-sensitive fluorescent amyloid-binding dyes, we assessed the conformational variability of Aß deposits in fAD, fCAA, and sAD patients. Comparing many deposits from a given patient with the overall population, we found that intrapatient variability is much lower than interpatient variability for both disease types. In a given brain, we observed one or two structurally distinct forms. When two forms coexist, they segregate between the parenchyma and cerebrovasculature, particularly in fAD patients. Compared with sAD samples, deposits from fAD patients show less intersubject variability, and little overlap exists between fAD and sAD deposits. Finally, we examined whether E22G (Arctic) or E22Q (Dutch) mutants direct the misfolding of WT Aß, leading to fAD-like plaques in vivo. Intracerebrally injecting mutant Aß40 fibrils into transgenic mice expressing only WT Aß induced the deposition of plaques with many biochemical hallmarks of fAD. Thus, mutant Aß40 prions induce a conformation of WT Aß similar to that found in fAD deposits. These findings indicate that diverse AD phenotypes likely arise from one or more initial Aß prion conformations, which kinetically dominate the spread of prions in the brain.


Assuntos
Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Conformação Proteica , Dobramento de Proteína , Peptídeos beta-Amiloides/genética , Animais , Camundongos Transgênicos , Mutação Puntual
10.
Proc Natl Acad Sci U S A ; 114(51): E11029-E11036, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29203673

RESUMO

Misfolding of tau proteins into prions and their propagation along neural circuits are thought to result in neurodegeneration causing Alzheimer's disease, progressive supranuclear palsy, chronic traumatic encephalopathy, and other tauopathies. Little is known about the molecular processes mediating tau prion replication and spreading in different brain regions. Using transgenic (Tg) mice with a neuronal promoter driving expression of human mutant (P301S) tau, we found that tau prion formation and histopathologic deposition is largely restricted to the hindbrain. Unexpectedly, tau mRNA and protein levels did not differ between the forebrain and hindbrain, suggesting that other factors modulating the conversion of tau into a prion exist and are region specific. Using a cell-based prion propagation assay, we discovered that tau prion replication is suppressed by forebrain-derived inhibitors, one of which is sortilin, a lysosomal sorting receptor. We also show that sortilin expression is higher in the forebrain than the hindbrain across the life span of the Tg mice, suggesting that sortilin, at least in part, inhibits forebrain tau prion replication in vivo. Our findings provide evidence for selective vulnerability in mice resulting in highly regulated levels of tau prion propagation, thus affording a model for identification of additional molecules that could mitigate the levels of tau prions in human tauopathies.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Fosforilação , Ligação Proteica , Rombencéfalo/metabolismo , Rombencéfalo/patologia
11.
J Autoimmun ; 101: 56-69, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31010726

RESUMO

The molecular events underlying the transition from initial inflammatory flares to the progressive phase of multiple sclerosis (MS) remain poorly understood. Here, we report that the microtubule-associated protein (MAP) Tau exerts a gender-specific protective function on disease progression in the MS model experimental autoimmune encephalomyelitis (EAE). A detailed investigation of the autoimmune response in Tau-deficient mice excluded a strong immunoregulatory role for Tau, suggesting that its beneficial effects are presumably exerted within the central nervous system (CNS). Spinal cord transcriptomic data show increased synaptic dysfunctions and alterations in the NF-kB activation pathway upon EAE in Tau-deficient mice as compared to wildtype animals. We also performed the first comprehensive characterization of Tau post-translational modifications (PTMs) in the nervous system upon EAE. We report that the methylation levels of the conserved lysine residue K306 are significantly decreased in the chronic phase of the disease. By combining biochemical assays and molecular dynamics (MD) simulations, we demonstrate that methylation at K306 decreases the affinity of Tau for the microtubule network. Thus, the down-regulation of this PTM might represent a homeostatic response to enhance axonal stability against an autoimmune CNS insult. The results, altogether, position Tau as key mediator between the inflammatory processes and neurodegeneration that seems to unify many CNS diseases.


Assuntos
Regulação da Expressão Gênica , Esclerose Múltipla/etiologia , Esclerose Múltipla/metabolismo , Neurônios/metabolismo , Sinapses/genética , Sinapses/metabolismo , Proteínas tau/metabolismo , Animais , Autoimunidade , Linhagem Celular , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental , Feminino , Redes Reguladoras de Genes , Masculino , Metilação , Camundongos , Camundongos Knockout , Modelos Moleculares , Esclerose Múltipla/patologia , Transdução de Sinais , Relação Estrutura-Atividade , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transcrição Gênica , Proteínas tau/química
12.
Neurobiol Dis ; 109(Pt B): 191-200, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28359847

RESUMO

The progressive nature of Alzheimer's disease (AD) is thought to occur, at least in part, by the self-replication and spreading of Aß and Tau aggregates through a prion mechanism. Evidence now exists that structural variants of Aß prions can propagate their distinct conformations through template-directed folding of naïve Aß peptides. This notion implicates that the first self-propagating Aß assembly to emerge in the brain dictates the conformation, anatomical spread and pace of subsequently formed deposits. It is hypothesized that a prion mechanism defines the molecular basis underlying the diverse clinicopathologic phenotypes observed across the spectrum of AD patients. Thus, distinct AD strains might require further sub-classification based on biochemical and structural characterization of aggregated Aß. Here, we review the evidence for distinct, self-propagating Aß strains, and discuss potential cellular mechanisms that might contribute to their manifestation. From this perspective, we also explore the implications of Aß strains for current FDA-approved medical imaging probes and therapies for amyloid. Ultimately, the discovery of new molecular tools to differentiate Aß strains and dissect the heterogeneity of AD may lead to the development of more informative diagnostics and strain-specific therapeutics.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Animais , Humanos , Fenótipo , Conformação Proteica
13.
Proc Natl Acad Sci U S A ; 111(28): 10329-34, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982137

RESUMO

An increasing number of studies continue to show that the amyloid ß (Aß) peptide adopts an alternative conformation and acquires transmissibility; hence, it becomes a prion. Here, we report on the attributes of two strains of Aß prions formed from synthetic Aß peptides composed of either 40 or 42 residues. Modifying the conditions for Aß polymerization increased both the protease resistance and prion infectivity compared with an earlier study. Approximately 150 d after intracerebral inoculation, both synthetic Aß40 and Aß42 prions produced a sustained rise in the bioluminescence imaging signal in the brains of bigenic Tg(APP23:Gfap-luc) mice, indicative of astrocytic gliosis. Pathological investigations showed that synthetic Aß40 prions produced amyloid plaques containing both Aß40 and Aß42 in the brains of inoculated bigenic mice, whereas synthetic Aß42 prions stimulated the formation of smaller, more numerous plaques composed predominantly of Aß42. Synthetic Aß40 preparations consisted of long straight fibrils; in contrast, the Aß42 fibrils were much shorter. Addition of 3.47 mM (0.1%) SDS to the polymerization reaction produced Aß42 fibrils that were indistinguishable from Aß40 fibrils produced in the absence or presence of SDS. Moreover, the Aß amyloid plaques in the brains of bigenic mice inoculated with Aß42 prions prepared in the presence of SDS were similar to those found in mice that received Aß40 prions. From these results, we conclude that the composition of Aß plaques depends on the conformation of the inoculated Aß polymers, and thus, these inocula represent distinct synthetic Aß prion strains.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Encéfalo/metabolismo , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Príons , Animais , Humanos , Camundongos Transgênicos , Fatores de Tempo
14.
Proc Natl Acad Sci U S A ; 111(28): 10323-8, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982139

RESUMO

An increasing number of studies argues that self-propagating protein conformations (i.e., prions) feature in the pathogenesis of several common neurodegenerative diseases. Mounting evidence contends that aggregates of the amyloid-ß (Aß) peptide become self-propagating in Alzheimer's disease (AD) patients. An important characteristic of prions is their ability to replicate distinct strains, the biological information for which is enciphered within different conformations of protein aggregates. To investigate whether distinct strains of Aß prions can be discerned in AD patients, we performed transmission studies in susceptible transgenic mice using brain homogenates from sporadic or heritable (Arctic and Swedish) AD cases. Mice inoculated with the Arctic AD sample exhibited a pathology that could be distinguished from mice inoculated with the Swedish or sporadic AD samples, which was judged by differential accumulation of Aß isoforms and the morphology of cerebrovascular Aß deposition. Unlike Swedish AD- or sporadic AD-inoculated animals, Arctic AD-inoculated mice, like Arctic AD patients, displayed a prominent Aß38-containing cerebral amyloid angiopathy. The divergent transmission behavior of the Arctic AD sample compared with the Swedish and sporadic AD samples was maintained during second passage in mice, showing that Aß strains are serially transmissible. We conclude that at least two distinct strains of Aß prions can be discerned in the brains of AD patients and that strain fidelity was preserved on serial passage in mice. Our results provide a potential explanation for the clinical and pathological heterogeneity observed in AD patients.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Encéfalo/metabolismo , Príons , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Humanos , Camundongos , Camundongos Transgênicos
15.
J Pharmacol Exp Ther ; 358(3): 537-47, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27317802

RESUMO

Developing therapeutics for neurodegenerative diseases (NDs) prevalent in the aging population remains a daunting challenge. With the growing understanding that many NDs progress by conformational self-templating of specific proteins, the prototypical prion diseases offer a platform for ND drug discovery. We evaluated high-throughput screening hits with the aryl amide scaffold and explored the structure-activity relationships around three series differing in their N-aryl core: benzoxazole, benzothiazole, and cyano. Potent anti-prion compounds were advanced to pharmacokinetic studies, and the resulting brain-penetrant leads from each series, together with a related N-aryl piperazine lead, were escalated to long-term dosing and efficacy studies. Compounds from each of the four series doubled the survival of mice infected with a mouse-passaged prion strain. Treatment with aryl amides altered prion strain properties, as evidenced by the distinct patterns of neuropathological deposition of prion protein and associated astrocytic gliosis in the brain; however, none of the aryl amide compounds resulted in drug-resistant prion strains, in contrast to previous studies on compounds with the 2-aminothiazole (2-AMT) scaffold. As seen with 2-AMTs and other effective anti-prion compounds reported to date, the novel aryl amides reported here were ineffective in prolonging the survival of transgenic mice infected with human prions. Most encouraging is our discovery that aryl amides show that the development of drug resistance is not an inevitable consequence of efficacious anti-prion therapeutics.


Assuntos
Amidas/química , Amidas/farmacologia , Descoberta de Drogas , Doenças Priônicas/tratamento farmacológico , Amidas/metabolismo , Amidas/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Síndrome de Creutzfeldt-Jakob/tratamento farmacológico , Feminino , Camundongos , Doenças Priônicas/metabolismo , Relação Estrutura-Atividade , Análise de Sobrevida
16.
J Pharmacol Exp Ther ; 355(1): 2-12, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26224882

RESUMO

Because no drug exists that halts or even slows any neurodegenerative disease, developing effective therapeutics for any prion disorder is urgent. We recently reported two compounds (IND24 and IND81) with the 2-aminothiazole (2-AMT) chemical scaffold that almost doubled the incubation times in scrapie prion-infected, wild-type (wt) FVB mice when given in a liquid diet. Remarkably, oral prophylactic treatment with IND24 beginning 14 days prior to intracerebral prion inoculation extended survival from ∼120 days to over 450 days. In addition to IND24, we evaluated the pharmacokinetics and efficacy of five additional 2-AMTs; one was not followed further because its brain penetration was poor. Of the remaining four new 2-AMTs, IND114338 doubled and IND125 tripled the incubation times of RML-inoculated wt and Tg4053 mice overexpressing wt mouse prion protein (PrP), respectively. Neuropathological examination of the brains from untreated controls showed a widespread deposition of self-propagating, ß-sheet-rich "scrapie" isoform (PrP(Sc)) prions accompanied by a profound astrocytic gliosis. In contrast, mice treated with 2-AMTs had lower levels of PrP(Sc) and associated astrocytic gliosis, with each compound resulting in a distinct pattern of deposition. Notably, IND125 prevented both PrP(Sc) accumulation and astrocytic gliosis in the cerebrum. Progressive central nervous system dysfunction in the IND125-treated mice was presumably due to the PrP(Sc) that accumulated in their brainstems. Disappointingly, none of the four new 2-AMTs prolonged the lives of mice expressing a chimeric human/mouse PrP transgene inoculated with Creutzfeldt-Jakob disease prions.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Proteínas PrPSc/metabolismo , Tiazóis/química , Tiazóis/farmacologia , Animais , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos , Proteínas PrPSc/genética , Scrapie/patologia , Especificidade da Espécie , Análise de Sobrevida , Taxa de Sobrevida , Tiazóis/farmacocinética , Tiazóis/uso terapêutico , Transgenes/genética , Resultado do Tratamento
17.
Chem Sci ; 15(22): 8414-8421, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38846390

RESUMO

Insoluble amyloids rich in cross-ß fibrils are observed in a number of neurodegenerative diseases. Depending on the clinicopathology, the amyloids can adopt distinct supramolecular assemblies, termed conformational strains. However, rapid methods to study amyloids in a conformationally specific manner are lacking. We introduce a novel computational method for de novo design of peptides that tile the surface of α-synuclein fibrils in a conformationally specific manner. Our method begins by identifying surfaces that are unique to the conformational strain of interest, which becomes a "target backbone" for the design of a peptide binder. Next, we interrogate structures in the PDB with high geometric complementarity to the target. Then, we identify secondary structural motifs that interact with this target backbone in a favorable, highly occurring geometry. This method produces monomeric helical motifs with a favorable geometry for interaction with the strands of the underlying amyloid. Each motif is then symmetrically replicated to form a monolayer that tiles the amyloid surface. Finally, amino acid sequences of the peptide binders are computed to provide a sequence with high geometric and physicochemical complementarity to the target amyloid. This method was applied to a conformational strain of α-synuclein fibrils, resulting in a peptide with high specificity for the target relative to other amyloids formed by α-synuclein, tau, or Aß40. This designed peptide also markedly slowed the formation of α-synuclein amyloids. Overall, this method offers a new tool for examining conformational strains of amyloid proteins.

18.
bioRxiv ; 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38617229

RESUMO

Down syndrome (DS) is a common genetic condition caused by trisomy of chromosome 21. Among the complex clinical features including musculoskeletal, neurological and cardiovascular disabilities, individuals with DS have an increased risk of developing progressive dementia and early onset Alzheimer's Disease (AD). This is attributed to the increased gene dosage of amyloid-ß (Aß) precursor protein gene, the formation of self-propagating Aß and tau prion conformers, and the deposition of neurotoxic Aß plaques and tau neurofibrillary tangles. Tau amyloid fibrils have previously been established to adopt many distinct conformations across different neurodegenerative conditions. Here we report the characterization of brain samples from four DS cases spanning 36 to 63 years of age by spectral confocal imaging with conformation-specific dyes and cryo-electron microscopy (cryo-EM) to determine structures of isolated tau fibrils. High-resolution structures reveal paired helical filament (PHF) and straight filament (SF) conformations of tau that are identical to those determined from AD. The PHFs and SFs are made of two C-shaped protofilaments with a cross-ß/ß-helix motif. Similar to filaments from AD cases, most filaments from the DS cases adopted the PHF form, while a minority (~20%) formed SFs. Samples from the youngest individual with no documented dementia had sparse tau deposits. To isolate tau for cryo-EM from this challenging sample we used a novel affinity-grid method involving a graphene-oxide surface derivatized with anti-tau antibodies. This improved isolation and revealed primarily tau PHFs and a minor population of chronic traumatic encephalopathy type II-like filaments were present in this youngest case. These findings expand the similarities between AD and DS to the molecular level, providing insight into their related pathologies and the potential for targeting common tau filament folds by small-molecule therapeutics and diagnostics.

19.
Acta Neuropathol Commun ; 12(1): 94, 2024 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-38867338

RESUMO

Down syndrome (DS) is a common genetic condition caused by trisomy of chromosome 21. Among their complex clinical features, including musculoskeletal, neurological, and cardiovascular disabilities, individuals with DS have an increased risk of developing progressive dementia and early-onset Alzheimer's disease (AD). This dementia is attributed to the increased gene dosage of the amyloid-ß (Aß) precursor protein gene, the formation of self-propagating Aß and tau prion conformers, and the deposition of neurotoxic Aß plaques and tau neurofibrillary tangles. Tau amyloid fibrils have previously been established to adopt many distinct conformations across different neurodegenerative conditions. Here, we report the characterization of brain samples from four DS cases spanning 36-63 years of age by spectral confocal imaging with conformation-specific dyes and cryo-electron microscopy (cryo-EM) to determine structures of isolated tau fibrils. High-resolution structures revealed paired helical filament (PHF) and straight filament (SF) conformations of tau that were identical to those determined from AD cases. The PHFs and SFs are made of two C-shaped protofilaments, each containing a cross-ß/ß-helix motif. Similar to filaments from AD cases, most filaments from the DS cases adopted the PHF form, while a minority (approximately 20%) formed SFs. Samples from the youngest individual with no documented dementia had sparse tau deposits. To isolate tau for cryo-EM from this challenging sample we used a novel affinity-grid method involving a graphene oxide surface derivatized with anti-tau antibodies. This method improved isolation and revealed that primarily tau PHFs and a minor population of chronic traumatic encephalopathy type II-like filaments were present in this youngest case. These findings expand the similarities between AD and DS to the molecular level, providing insight into their related pathologies and the potential for targeting common tau filament folds by small-molecule therapeutics and diagnostics.


Assuntos
Doença de Alzheimer , Microscopia Crioeletrônica , Síndrome de Down , Proteínas tau , Humanos , Síndrome de Down/patologia , Síndrome de Down/metabolismo , Proteínas tau/metabolismo , Proteínas tau/ultraestrutura , Microscopia Crioeletrônica/métodos , Pessoa de Meia-Idade , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Feminino , Adulto , Masculino , Emaranhados Neurofibrilares/patologia , Emaranhados Neurofibrilares/metabolismo , Encéfalo/patologia , Encéfalo/metabolismo , Encéfalo/ultraestrutura
20.
bioRxiv ; 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39282355

RESUMO

Aggregation of microtubule-associated protein tau (MAPT/tau) into conformationally distinct fibrils underpins neurodegenerative tauopathies. Fluorescent probes (fluoroprobes), such as thioflavin T (ThT), have been essential tools for studying tau aggregation; however, most of them do not discriminate between amyloid fibril conformations (polymorphs). This gap is due, in part, to a lack of high-throughput methods for screening large, diverse chemical collections. Here, we leverage advances in protein adaptive differential scanning fluorimetry (paDSF) to screen the Aurora collection of 300+ fluorescent dyes against multiple synthetic tau fibril polymorphs. This screen, coupled with orthogonal secondary assays, revealed pan-fibril binding chemotypes, as well as fluoroprobes selective for subsets of fibrils. One fluoroprobe recognized tau pathology in ex vivo brain slices from Alzheimer's disease patients. We propose that these scaffolds represent entry points for development of selective fibril ligands and, more broadly, that high throughput, fluorescence-based dye screening is a platform for their discovery.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA