Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Pharmacol Rev ; 75(2): 263-308, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36549866

RESUMO

Lysine-selective molecular tweezers (MTs) are supramolecular host molecules displaying a remarkably broad spectrum of biologic activities. MTs act as inhibitors of the self-assembly and toxicity of amyloidogenic proteins using a unique mechanism. They destroy viral membranes and inhibit infection by enveloped viruses, such as HIV-1 and SARS-CoV-2, by mechanisms unrelated to their action on protein self-assembly. They also disrupt biofilm of Gram-positive bacteria. The efficacy and safety of MTs have been demonstrated in vitro, in cell culture, and in vivo, suggesting that these versatile compounds are attractive therapeutic candidates for various diseases, infections, and injuries. A lead compound called CLR01 has been shown to inhibit the aggregation of various amyloidogenic proteins, facilitate their clearance in vivo, prevent infection by multiple viruses, display potent anti-biofilm activity, and have a high safety margin in animal models. The inhibitory effect of CLR01 against amyloidogenic proteins is highly specific to abnormal self-assembly of amyloidogenic proteins with no disruption of normal mammalian biologic processes at the doses needed for inhibition. Therapeutic effects of CLR01 have been demonstrated in animal models of proteinopathies, lysosomal-storage diseases, and spinal-cord injury. Here we review the activity and mechanisms of action of these intriguing compounds and discuss future research directions. SIGNIFICANCE STATEMENT: Molecular tweezers are supramolecular host molecules with broad biological applications, including inhibition of abnormal protein aggregation, facilitation of lysosomal clearance of toxic aggregates, disruption of viral membranes, and interference of biofilm formation by Gram-positive bacteria. This review discusses the molecular and cellular mechanisms of action of the molecular tweezers, including the discovery of distinct mechanisms acting in vitro and in vivo, and the application of these compounds in multiple preclinical disease models.


Assuntos
Produtos Biológicos , COVID-19 , Animais , Organofosfatos/farmacologia , SARS-CoV-2 , Proteínas Amiloidogênicas , Mamíferos
2.
Alzheimers Dement ; 20(3): 2240-2261, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38170841

RESUMO

INTRODUCTION: The pace of innovation has accelerated in virtually every area of tau research in just the past few years. METHODS: In February 2022, leading international tau experts convened to share selected highlights of this work during Tau 2022, the second international tau conference co-organized and co-sponsored by the Alzheimer's Association, CurePSP, and the Rainwater Charitable Foundation. RESULTS: Representing academia, industry, and the philanthropic sector, presenters joined more than 1700 registered attendees from 59 countries, spanning six continents, to share recent advances and exciting new directions in tau research. DISCUSSION: The virtual meeting provided an opportunity to foster cross-sector collaboration and partnerships as well as a forum for updating colleagues on research-advancing tools and programs that are steadily moving the field forward.


Assuntos
Doença de Alzheimer , Tauopatias , Humanos , Proteínas tau
3.
J Biol Chem ; 298(1): 101478, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34896392

RESUMO

Aptamers are oligonucleotides selected from large pools of random sequences based on their affinity for bioactive molecules and are used in similar ways to antibodies. Aptamers provide several advantages over antibodies, including their small size, facile, large-scale chemical synthesis, high stability, and low immunogenicity. Amyloidogenic proteins, whose aggregation is relevant to neurodegenerative diseases, such as Alzheimer's, Parkinson's, and prion diseases, are among the most challenging targets for aptamer development due to their conformational instability and heterogeneity, the same characteristics that make drug development against amyloidogenic proteins difficult. Recently, chemical tethering of aptagens (equivalent to antigens) and advances in high-throughput sequencing-based analysis have been used to overcome some of these challenges. In addition, internalization technologies using fusion to cellular receptors and extracellular vesicles have facilitated central nervous system (CNS) aptamer delivery. In view of the development of these techniques and resources, here we review antiamyloid aptamers, highlighting preclinical application to CNS therapy.


Assuntos
Proteínas Amiloidogênicas , Aptâmeros de Nucleotídeos , Doenças Neurodegenerativas , Doenças Priônicas , Proteínas Amiloidogênicas/metabolismo , Animais , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Aptâmeros de Nucleotídeos/uso terapêutico , Sistema Nervoso Central/metabolismo , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Doenças Priônicas/tratamento farmacológico , Técnica de Seleção de Aptâmeros/métodos
4.
Acta Neuropathol ; 145(5): 515-540, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37012443

RESUMO

Extracellular vesicles (EVs), including exosomes, microvesicles, and oncosomes, are nano-sized particles enclosed by a lipid bilayer. EVs are released by virtually all eukaryotic cells and have been shown to contribute to intercellular communication by transporting proteins, lipids, and nucleic acids. In the context of neurodegenerative diseases, EVs may carry toxic, misfolded forms of amyloidogenic proteins and facilitate their spread to recipient cells in the central nervous system (CNS). CNS-originating EVs can cross the blood-brain barrier into the bloodstream and may be found in other body fluids, including saliva, tears, and urine. EVs originating in the CNS represent an attractive source of biomarkers for neurodegenerative diseases, because they contain cell- and cell state-specific biological materials. In recent years, multiple papers have reported the use of this strategy for identification and quantitation of biomarkers for neurodegenerative diseases, including Parkinson's disease and atypical parkinsonian disorders. However, certain technical issues have yet to be standardized, such as the best surface markers for isolation of cell type-specific EVs and validating the cellular origin of the EVs. Here, we review recent research using CNS-originating EVs for biomarker studies, primarily in parkinsonian disorders, highlight technical challenges, and propose strategies for overcoming them.


Assuntos
Exossomos , Vesículas Extracelulares , Doenças Neurodegenerativas , Doença de Parkinson , Humanos , Vesículas Extracelulares/metabolismo , Sistema Nervoso Central/metabolismo , Exossomos/metabolismo , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/metabolismo , Biomarcadores/metabolismo
5.
Mov Disord ; 37(4): 778-789, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35040506

RESUMO

BACKGROUND: Multiple system atrophy (MSA) is a fatal neurodegenerative disease characterized by the aggregation of α-synuclein in glia and neurons. Sirolimus (rapamycin) is an mTOR inhibitor that promotes α-synuclein autophagy and reduces its associated neurotoxicity in preclinical models. OBJECTIVE: To investigate the efficacy and safety of sirolimus in patients with MSA using a futility design. We also analyzed 1-year biomarker trajectories in the trial participants. METHODS: Randomized, double-blind, parallel group, placebo-controlled clinical trial at the New York University of patients with probable MSA randomly assigned (3:1) to sirolimus (2-6 mg daily) for 48 weeks or placebo. Primary endpoint was change in the Unified MSA Rating Scale (UMSARS) total score from baseline to 48 weeks. (ClinicalTrials.gov NCT03589976). RESULTS: The trial was stopped after a pre-planned interim analysis met futility criteria. Between August 15, 2018 and November 15, 2020, 54 participants were screened, and 47 enrolled and randomly assigned (35 sirolimus, 12 placebo). Of those randomized, 34 were included in the intention-to-treat analysis. There was no difference in change from baseline to week 48 between the sirolimus and placebo in UMSARS total score (mean difference, 2.66; 95% CI, -7.35-6.91; P = 0.648). There was no difference in UMSARS-1 and UMSARS-2 scores either. UMSARS scores changes were similar to those reported in natural history studies. Neuroimaging and blood biomarker results were similar in the sirolimus and placebo groups. Adverse events were more frequent with sirolimus. Analysis of 1-year biomarker trajectories in all participants showed that increases in blood neurofilament light chain (NfL) and reductions in whole brain volume correlated best with UMSARS progression. CONCLUSIONS: Sirolimus for 48 weeks was futile to slow the progression of MSA and had no effect on biomarkers compared to placebo. One-year change in blood NfL and whole brain atrophy are promising biomarkers of disease progression for future clinical trials. © 2022 International Parkinson and Movement Disorder Society.


Assuntos
Atrofia de Múltiplos Sistemas , alfa-Sinucleína , Método Duplo-Cego , Humanos , Futilidade Médica , Atrofia de Múltiplos Sistemas/tratamento farmacológico , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR , Resultado do Tratamento
6.
Acta Neuropathol ; 142(3): 495-511, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33991233

RESUMO

The diagnosis of Parkinson's disease (PD) and atypical parkinsonian syndromes is difficult due to the lack of reliable, easily accessible biomarkers. Multiple system atrophy (MSA) is a synucleinopathy whose symptoms often overlap with PD. Exosomes isolated from blood by immunoprecipitation using CNS markers provide a window into the brain's biochemistry and may assist in distinguishing between PD and MSA. Thus, we asked whether α-synuclein (α-syn) in such exosomes could distinguish among healthy individuals, patients with PD, and patients with MSA. We isolated exosomes from the serum or plasma of these three groups by immunoprecipitation using neuronal and oligodendroglial markers in two independent cohorts and measured α-syn in these exosomes using an electrochemiluminescence ELISA. In both cohorts, α-syn concentrations were significantly lower in the control group and significantly higher in the MSA group compared to the PD group. The ratio between α-syn concentrations in putative oligodendroglial exosomes compared to putative neuronal exosomes was a particularly sensitive biomarker for distinguishing between PD and MSA. Combining this ratio with the α-syn concentration itself and the total exosome concentration, a multinomial logistic model trained on the discovery cohort separated PD from MSA with an AUC = 0.902, corresponding to 89.8% sensitivity and 86.0% specificity when applied to the independent validation cohort. The data demonstrate that a minimally invasive blood test measuring α-syn in blood exosomes immunoprecipitated using CNS markers can distinguish between patients with PD and patients with MSA with high sensitivity and specificity. Future optimization and validation of the data by other groups would allow this strategy to become a viable diagnostic test for synucleinopathies.


Assuntos
Exossomos/imunologia , Atrofia de Múltiplos Sistemas/diagnóstico , Neurônios/metabolismo , Oligodendroglia/metabolismo , Doença de Parkinson/diagnóstico , alfa-Sinucleína/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Área Sob a Curva , Biomarcadores , Estudos de Coortes , Diagnóstico Diferencial , Ensaio de Imunoadsorção Enzimática , Feminino , Voluntários Saudáveis , Humanos , Imunoprecipitação , Masculino , Pessoa de Meia-Idade , Atrofia de Múltiplos Sistemas/sangue , Doença de Parkinson/sangue , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
7.
FASEB J ; 34(9): 11957-11969, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32701214

RESUMO

Small-molecule inhibitors of abnormal protein self-assembly are promising candidates for developing therapy against proteinopathies. Such compounds have been examined primarily as inhibitors of amyloid ß-protein (Aß), whereas testing of inhibitors of other amyloidogenic proteins has lagged behind. An important issue with screening compound libraries is that although an inhibitor suitable for therapy must be both effective and nontoxic, typical screening focuses on efficacy, whereas safety typically is tested at a later stage using cells and/or animals. In addition, typical thioflavin T (ThT)-fluorescence-based screens use the final fluorescence value as a readout, potentially missing important kinetic information. Here, we examined potential inhibitors of superoxide dismutase 1 (SOD1) using ThT-fluorescence including the different phases of fluorescence change and added a parallel screen of SOD1 activity as a potential proxy for compound toxicity. Some compounds previously reported to inhibit other amyloidogenic proteins also inhibited SOD1 aggregation at low micromolar concentrations, whereas others were ineffective. Analysis of the lag phase and exponential slope added important information that could help exclude false-positive or false-negative results. SOD1 was highly resistant to inhibition of its activity, and therefore, did not have the necessary sensitivity to serve as a proxy for examining potential toxicity.


Assuntos
Benzotiazóis/farmacologia , Agregados Proteicos/efeitos dos fármacos , Superóxido Dismutase-1/química , Benzotiazóis/química , Humanos
8.
Mol Ther ; 28(4): 1167-1176, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32087148

RESUMO

Lysosomal storage diseases (LSDs) are inherited disorders caused by lysosomal deficiencies and characterized by dysfunction of the autophagy-lysosomal pathway (ALP) often associated with neurodegeneration. No cure is currently available to treat neuropathology in LSDs. By studying a mouse model of mucopolysaccharidosis (MPS) type IIIA, one of the most common and severe forms of LSDs, we found that multiple amyloid proteins including α-synuclein, prion protein (PrP), Tau, and amyloid ß progressively aggregate in the brain. The amyloid deposits mostly build up in neuronal cell bodies concomitantly with neurodegeneration. Treating MPS-IIIA mice with CLR01, a "molecular tweezer" that acts as a broad-spectrum inhibitor of amyloid protein self-assembly reduced lysosomal enlargement and re-activates autophagy flux. Restoration of the ALP was associated with reduced neuroinflammation and amelioration of memory deficits. Together, these data provide evidence that brain deposition of amyloid proteins plays a gain of neurotoxic function in a severe LSD by affecting the ALP and identify CLR01 as new potent drug candidate for MPS-IIIA and likely for other LSDs.


Assuntos
Autofagia/efeitos dos fármacos , Hidrocarbonetos Aromáticos com Pontes/administração & dosagem , Mucopolissacaridose III/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Organofosfatos/administração & dosagem , Amiloide/antagonistas & inibidores , Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Corpo Celular/metabolismo , Modelos Animais de Doenças , Masculino , Camundongos , Mucopolissacaridose III/complicações , Mucopolissacaridose III/metabolismo , Doenças Neurodegenerativas/etiologia , Organofosfatos/farmacologia , Resultado do Tratamento
9.
J Biol Chem ; 294(10): 3501-3513, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30602569

RESUMO

Mutations in superoxide dismutase 1 (SOD1) cause 15-20% of familial amyotrophic lateral sclerosis (fALS) cases. The resulting amino acid substitutions destabilize SOD1's protein structure, leading to its self-assembly into neurotoxic oligomers and aggregates, a process hypothesized to cause the characteristic motor-neuron degeneration in affected individuals. Currently, effective disease-modifying therapy is not available for ALS. Molecular tweezers prevent formation of toxic protein assemblies, yet their protective action has not been tested previously on SOD1 or in the context of ALS. Here, we tested the molecular tweezer CLR01-a broad-spectrum inhibitor of the self-assembly and toxicity of amyloid proteins-as a potential therapeutic agent for ALS. Using recombinant WT and mutant SOD1, we found that CLR01 inhibited the aggregation of all tested SOD1 forms in vitro Next, we examined whether CLR01 could prevent the formation of misfolded SOD1 in the G93A-SOD1 mouse model of ALS and whether such inhibition would have a beneficial therapeutic effect. CLR01 treatment decreased misfolded SOD1 in the spinal cord significantly. However, these histological findings did not correlate with improvement of the disease phenotype. A small, dose-dependent decrease in disease duration was found in CLR01-treated mice, relative to vehicle-treated animals, yet motor function did not improve in any of the treatment groups. These results demonstrate that CLR01 can inhibit SOD1 misfolding and aggregation both in vitro and in vivo, but raise the question whether such inhibition is sufficient for achieving a therapeutic effect. Additional studies in other less aggressive ALS models may be needed to determine the therapeutic potential of this approach.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Mutação , Organofosfatos/farmacologia , Superóxido Dismutase-1/química , Superóxido Dismutase-1/genética , Sequência de Aminoácidos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Sítios de Ligação , Peso Corporal/efeitos dos fármacos , Hidrocarbonetos Aromáticos com Pontes/metabolismo , Modelos Animais de Doenças , Camundongos , Força Muscular/efeitos dos fármacos , Organofosfatos/metabolismo , Agregados Proteicos/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Superóxido Dismutase-1/metabolismo , Análise de Sobrevida
10.
J Am Chem Soc ; 142(40): 17024-17038, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-32926779

RESUMO

Broad-spectrum antivirals are powerful weapons against dangerous viruses where no specific therapy exists, as in the case of the ongoing SARS-CoV-2 pandemic. We discovered that a lysine- and arginine-specific supramolecular ligand (CLR01) destroys enveloped viruses, including HIV, Ebola, and Zika virus, and remodels amyloid fibrils in semen that promote viral infection. Yet, it is unknown how CLR01 exerts these two distinct therapeutic activities. Here, we delineate a novel mechanism of antiviral activity by studying the activity of tweezer variants: the "phosphate tweezer" CLR01, a "carboxylate tweezer" CLR05, and a "phosphate clip" PC. Lysine complexation inside the tweezer cavity is needed to antagonize amyloidogenesis and is only achieved by CLR01. Importantly, CLR01 and CLR05 but not PC form closed inclusion complexes with lipid head groups of viral membranes, thereby altering lipid orientation and increasing surface tension. This process disrupts viral envelopes and diminishes infectivity but leaves cellular membranes intact. Consequently, CLR01 and CLR05 display broad antiviral activity against all enveloped viruses tested, including herpesviruses, Measles virus, influenza, and SARS-CoV-2. Based on our mechanistic insights, we potentiated the antiviral, membrane-disrupting activity of CLR01 by introducing aliphatic ester arms into each phosphate group to act as lipid anchors that promote membrane targeting. The most potent ester modifications harbored unbranched C4 units, which engendered tweezers that were approximately one order of magnitude more effective than CLR01 and nontoxic. Thus, we establish the mechanistic basis of viral envelope disruption by specific tweezers and establish a new class of potential broad-spectrum antivirals with enhanced activity.


Assuntos
Antivirais/química , Antivirais/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Organofosfatos/farmacologia , Proteínas do Envelope Viral/efeitos dos fármacos , Fosfatase Ácida/química , Fosfatase Ácida/metabolismo , Amiloide/antagonistas & inibidores , Fármacos Anti-HIV/química , Fármacos Anti-HIV/farmacologia , Arginina/química , Betacoronavirus/efeitos dos fármacos , Hidrocarbonetos Aromáticos com Pontes/química , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/virologia , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Humanos , Lipídeos/química , Lisina/química , Espectroscopia de Ressonância Magnética , Organofosfatos/química , SARS-CoV-2 , Proteínas Secretadas pela Vesícula Seminal/química , Proteínas Secretadas pela Vesícula Seminal/metabolismo , Relação Estrutura-Atividade , Proteínas do Envelope Viral/metabolismo , Zika virus/efeitos dos fármacos
12.
J Am Chem Soc ; 139(16): 5640-5643, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28406616

RESUMO

Huntington's disease is a neurodegenerative disorder associated with the expansion of the polyglutamine tract in the exon-1 domain of the huntingtin protein (htte1). Above a threshold of 37 glutamine residues, htte1 starts to aggregate in a nucleation-dependent manner. A 17-residue N-terminal fragment of htte1 (N17) has been suggested to play a crucial role in modulating the aggregation propensity and toxicity of htte1. Here we identify N17 as a potential target for novel therapeutic intervention using the molecular tweezer CLR01. A combination of biochemical experiments and computer simulations shows that binding of CLR01 induces structural rearrangements within the htte1 monomer and inhibits htte1 aggregation, underpinning the key role of N17 in modulating htte1 toxicity.


Assuntos
Hidrocarbonetos Aromáticos com Pontes/farmacologia , Proteína Huntingtina/antagonistas & inibidores , Organofosfatos/farmacologia , Hidrocarbonetos Aromáticos com Pontes/química , Éxons , Humanos , Interações Hidrofóbicas e Hidrofílicas , Simulação de Dinâmica Molecular , Estrutura Molecular , Organofosfatos/química , Agregados Proteicos/efeitos dos fármacos
13.
Anal Biochem ; 518: 78-85, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27810329

RESUMO

Evidence suggests that amyloid ß-protein (Aß) oligomers may be seminal pathogenic agents in Alzheimer's disease (AD). If so, developing oligomer-targeted therapeutics requires an understanding of oligomer structure. This has been difficult due to the instability of these non-covalently associated Aß assemblies. We previously used rapid, zero-length, in situ chemical cross-linking to stabilize oligomers of Aß40. These enabled us to isolate pure, stable populations of dimers, trimers, and tetramers and to determine their structure-activity relationships. However, equivalent methods applied to Aß42 did not produce stable oligomers. We report here that the use of an Aß42 homologue, [F10, Y42]Aß42, coupled with sequential denaturation/dissociation and gel electrophoresis procedures, provides the means to produce highly pure, stable populations of oligomers of sizes ranging from dimer through dodecamer that are suitable for structure-activity relationship determination.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/isolamento & purificação , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/isolamento & purificação , Humanos
16.
Biochemistry ; 54(24): 3729-38, 2015 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-26030124

RESUMO

The tumor suppressor p53 plays a unique role as a central hub of numerous cell proliferation and apoptotic pathways, and its malfunction due to mutations is a major cause of various malignancies. Therefore, it serves as an attractive target for developing novel anticancer therapeutics. Because of its intrinsically unstable DNA binding domain, p53 unfolds rapidly at physiological temperature. Certain mutants shift the equilibrium toward the unfolded state and yield high-molecular weight, nonfunctional, and cytotoxic ß-sheet-rich aggregates that share tinctorial and conformational similarities with amyloid deposits found in various protein misfolding diseases. Here, we examined the effect of a novel protein assembly modulator, the lysine (Lys)-specific molecular tweezer, CLR01, on different aggregation stages of misfolded mutant p53 in vitro and on the cytotoxicity of the resulting p53 aggregates in cell culture. We found that CLR01 induced rapid formation of ß-sheet-rich, intermediate-size p53 aggregates yet inhibited further p53 aggregation and reduced the cytotoxicity of the resulting aggregates. Our data suggest that aggregation modulators, such as CLR01, could prevent the formation of toxic p53 aggregates.


Assuntos
Antineoplásicos/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Modelos Moleculares , Mutação , Organofosfatos/farmacologia , Agregação Patológica de Proteínas/tratamento farmacológico , Proteína Supressora de Tumor p53/antagonistas & inibidores , Substituição de Aminoácidos , Antineoplásicos/química , Sítios de Ligação , Hidrocarbonetos Aromáticos com Pontes/química , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/ultraestrutura , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/ultraestrutura , Microscopia Eletrônica de Transmissão , Mutagênese Sítio-Dirigida , Organofosfatos/química , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Estabilidade Proteica/efeitos dos fármacos , Desdobramento de Proteína/efeitos dos fármacos , Deficiências na Proteostase/tratamento farmacológico , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
17.
J Biol Chem ; 289(15): 10727-10737, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24567327

RESUMO

Recent work on α-synuclein has shown that aggregation is controlled kinetically by the rate of reconfiguration of the unstructured chain, such that the faster the reconfiguration, the slower the aggregation. In this work we investigate this relationship by examining α-synuclein in the presence of a small molecular tweezer, CLR01, which binds selectively to Lys side chains. We find strong binding to multiple Lys within the chain as measured by fluorescence and mass-spectrometry and a linear increase in the reconfiguration rate with concentration of the inhibitor. Top-down mass-spectrometric analysis shows that the main binding of CLR01 to α-synuclein occurs at the N-terminal Lys-10/Lys-12. Photo-induced cross-linking of unmodified proteins (PICUP) analysis shows that under the conditions used for the fluorescence analysis, α-synuclein is predominantly monomeric. The results can be successfully modeled using a kinetic scheme in which two aggregation-prone monomers can form an encounter complex that leads to further oligomerization but can also dissociate back to monomers if the reconfiguration rate is sufficiently high. Taken together, the data provide important insights into the preferred binding site of CLR01 on α-synuclein and the mechanism by which the molecular tweezer prevents self-assembly into neurotoxic aggregates by α-synuclein and presumably other amyloidogenic proteins.


Assuntos
Hidrocarbonetos Aromáticos com Pontes/química , Lisina/química , Organofosfatos/química , alfa-Sinucleína/química , Sequência de Aminoácidos , Sítios de Ligação , Difusão , Humanos , Cinética , Espectrometria de Massas , Dados de Sequência Molecular , Doença de Parkinson/metabolismo , Probabilidade , Ligação Proteica , Estrutura Terciária de Proteína , Espectrometria de Fluorescência , Resultado do Tratamento
18.
Opt Lett ; 39(22): 6517-20, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25490508

RESUMO

We present a semi-analytical method for accurate modeling of wave propagation in cylindrically symmetric subwavelength metal-dielectric multilayers. Utilizing a cylindrical transfer matrix method, we compute the amplitude transfer function of cylindrical hyperlens, simulate the exact field distribution and propagation for a given source and compare it to that in effective hyperbolic medium. We investigate the conditions under which the effective medium theory (EMT) is valid and show that in cylindrical configuration, a new degree of freedom is present in the applicability of the EMT-the ratio between the inner radius of the structure and the unit cell size.


Assuntos
Impedância Elétrica , Metais , Modelos Teóricos , Imagem Óptica
19.
Nat Med ; 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38907160

RESUMO

Sanfilippo syndrome is a fatal childhood neurodegenerative disorder involving neuroinflammation among multiple pathologies. We hypothesized that anakinra, a recombinant interleukin-1 receptor antagonist, could improve neurobehavioral and functional symptoms owing to its capacity to treat neuroinflammation. This phase 1/2 trial aimed to test the safety, tolerability and effects of anakinra on neurobehavioral, functional and quality-of-life outcomes in patients and their caregivers. The primary outcome was the percent of participants requiring a dose increase at week 8 or week 16. Secondary efficacy outcomes included a multi-domain responder index (MDRI). Twenty-three participants (6-26 years of age) were enrolled. Twenty continued treatment to week 8, and 15 (75%) required an increased dose at week 8 or week 16. There was an improvement in at least one domain in the MDRI in 18 of 21 (86%) at week 8 and in 15 of 16 (94%) at week 36. Seven participants withdrew (intolerability of daily injections and lost to follow-up) before week 36. Adverse events occurred in 22 of 23 (96%) participants, most commonly mild injection site reactions. No serious adverse events were related to anakinra. In conclusion, anakinra was safe and associated with improved neurobehavioral and functional outcomes, supporting continued investigation of anakinra in Sanfilippo syndrome and other mucopolysaccharidoses. ClinicalTrials.gov identifier: NCT04018755 .

20.
Biochemistry ; 52(23): 3995-4002, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23679559

RESUMO

The antiallergy and potential anticancer drug tranilast has been patented for treating Alzheimer's disease (AD), in which amyloid ß-protein (Aß) plays a key pathogenic role. We used solution NMR to determine that tranilast binds to Aß40 monomers with ∼300 µM affinity. Remarkably, tranilast increases Aß40 fibrillation more than 20-fold in the thioflavin T assay at a 1:1 molar ratio, as well as significantly reducing the lag time. Tranilast likely promotes fibrillation by shifting Aß monomer conformations to those capable of seed formation and fibril elongation. Molecular docking results qualitatively agree with NMR chemical shift perturbation, which together indicate that hydrophobic interactions are the major driving force of the Aß-tranilast interaction. These data suggest that AD may be a potential complication for tranilast usage in elderly patients.


Assuntos
Peptídeos beta-Amiloides/química , Amiloide/química , Antialérgicos/química , Fragmentos de Peptídeos/química , Multimerização Proteica , ortoaminobenzoatos/química , Antineoplásicos/química , Benzotiazóis , Sítios de Ligação , Corantes Fluorescentes/química , Humanos , Microscopia de Força Atômica , Simulação de Acoplamento Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Estrutura Quaternária de Proteína , Tiazóis/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa