Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Sci Rep ; 9(1): 16947, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31740740

RESUMO

The over-expression and aggregation of α-synuclein (αSyn) are linked to the onset and pathology of Parkinson's disease. Native monomeric αSyn exists in an intrinsically disordered ensemble of interconverting conformations, which has made its therapeutic targeting by small molecules highly challenging. Nonetheless, here we successfully target the monomeric structural ensemble of αSyn and thereby identify novel drug-like small molecules that impact multiple pathogenic processes. Using a surface plasmon resonance high-throughput screen, in which monomeric αSyn is incubated with microchips arrayed with tethered compounds, we identified novel αSyn interacting drug-like compounds. Because these small molecules could impact a variety of αSyn forms present in the ensemble, we tested representative hits for impact on multiple αSyn malfunctions in vitro and in cells including aggregation and perturbation of vesicular dynamics. We thereby identified a compound that inhibits αSyn misfolding and is neuroprotective, multiple compounds that restore phagocytosis impaired by αSyn overexpression, and a compound blocking cellular transmission of αSyn. Our studies demonstrate that drug-like small molecules that interact with native αSyn can impact a variety of its pathological processes. Thus, targeting the intrinsically disordered ensemble of αSyn offers a unique approach to the development of small molecule research tools and therapeutics for Parkinson's disease.


Assuntos
Bibliotecas de Moléculas Pequenas/farmacologia , alfa-Sinucleína/metabolismo , Amiloide/antagonistas & inibidores , Amiloide/metabolismo , Linhagem Celular , Transferência Ressonante de Energia de Fluorescência , Ensaios de Triagem em Larga Escala/métodos , Humanos , Proteínas Intrinsicamente Desordenadas/metabolismo , Fagocitose/efeitos dos fármacos , Dobramento de Proteína , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/toxicidade , Ressonância de Plasmônio de Superfície , alfa-Sinucleína/química , alfa-Sinucleína/efeitos dos fármacos
2.
JAMA Neurol ; 75(10): 1206-1214, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29913017

RESUMO

Importance: Aggregated α-synuclein is believed to be central to the pathogenesis of Parkinson disease (PD). PRX002/RG7935 (PRX002) is a humanized monoclonal antibody designed to target aggregated forms of α-synuclein, thereby inhibiting neuron-to-neuron transfer of presumed pathogenic forms of α-synuclein, potentially resulting in neuronal protection and slowing disease progression. Objective: To evaluate the safety and tolerability of multiple intravenous infusions of PRX002 in patients with idiopathic PD. Design, Setting, and Participants: Multicenter, randomized, double-blind, placebo-controlled, multiple ascending-dose trial at 8 US study centers from July 2014 to September 2016. Eligible participants were aged 40 to 80 years with mild to moderate idiopathic PD (Hoehn and Yahr stages 1-3). Interventions: Participants were enrolled into 6 ascending-dose cohorts and randomly assigned to receive PRX002 (0.3 mg/kg, 1.0 mg/kg, 3.0 mg/kg, 10 mg/kg, 30 mg/kg, or 60 mg/kg) or placebo. Participants received 3 intravenous infusions every 4 weeks of PRX002 or placebo and were monitored during a 24-week observational period. Main Outcomes and Measures: Safety and tolerability assessments included physical and neurological examinations, laboratory tests, vital signs, and adverse events. Pharmacokinetic parameters included maximum PRX002 concentration, area under the curve, and half-life. Results: Of the 80 participants, most were white (97.5%; n = 78) and male (80%; n = 64); median (SD) age was 58 (8.4) years. PRX002 was generally safe and well tolerated; no serious or severe PRX002-related treatment-emergent adverse events (TEAEs) were reported. The TEAEs experienced by at least 5% of patients receiving PRX002, irrespective of relatedness to study drug, were constipation (9.1%; n = 5), infusion reaction (7.3%; n = 4), diarrhea (5.5%; n = 3), headache (5.5%; n = 3), peripheral edema (5.5%; n = 3), post-lumbar puncture syndrome (5.5%; n = 3), and upper respiratory tract infection (5.5%; n = 3). No antidrug antibodies were detected. Serum PRX002 levels increased in an approximately dose-proportional manner; mean terminal elimination half-life was similar across all doses (10.2 days). Rapid dose- and time-dependent mean reductions from baseline vs placebo in free serum α-synuclein levels of up to 97% were seen after a single infusion at the highest dose (F78,284 = 1.66; P = .002), with similar reductions after 2 additional infusions. Mean cerebrospinal fluid PRX002 concentration increased with PRX002 dose and was approximately 0.3% relative to serum across all dose cohorts. Conclusions and Relevance: Single and multiple doses of PRX002 were generally safe and well tolerated and resulted in robust binding of peripheral α-synuclein and dose-dependent increases of PRX002 in cerebrospinal fluid, reaching cerebrospinal fluid concentrations that may be expected to engage extracellular aggregated α-synuclein in the brain. Findings support the design of an ongoing phase 2 clinical study (NCT03100149). Trial Registration: ClinicalTrials.gov Identifier: NCT02157714.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Doença de Parkinson/tratamento farmacológico , alfa-Sinucleína/efeitos dos fármacos , alfa-Sinucleína/imunologia , Idoso , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Método Duplo-Cego , Esquema de Medicação , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade
3.
Neurology ; 90(2): 74-82, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29237794

RESUMO

Multiple system atrophy (MSA) is a rare neurodegenerative disorder with substantial knowledge gaps despite recent gains in basic and clinical research. In order to make further advances, concerted international collaboration is vital. In 2014, an international meeting involving leaders in the field and MSA advocacy groups was convened in Las Vegas, Nevada, to identify critical research areas where consensus and progress was needed to improve understanding, diagnosis, and treatment of the disease. Eight topic areas were defined: pathogenesis, preclinical modeling, target identification, endophenotyping, clinical measures, imaging biomarkers, nonimaging biomarkers, treatments/trial designs, and patient advocacy. For each topic area, an expert served as a working group chair and each working group developed priority-ranked research recommendations with associated timelines and pathways to reach the intended goals. In this report, each groups' recommendations are provided.


Assuntos
Atrofia de Múltiplos Sistemas , Humanos , Atrofia de Múltiplos Sistemas/diagnóstico , Atrofia de Múltiplos Sistemas/fisiopatologia , Atrofia de Múltiplos Sistemas/terapia , Nevada , Defesa do Paciente , Projetos de Pesquisa
4.
Acta Neuropathol Commun ; 5(1): 7, 2017 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-28086964

RESUMO

Neurodegenerative disorders such as Parkinson's Disease (PD), PD dementia (PDD) and Dementia with Lewy bodies (DLB) are characterized by progressive accumulation of α-synuclein (α-syn) in neurons. Recent studies have proposed that neuron-to-neuron propagation of α-syn plays a role in the pathogenesis of these disorders. We have previously shown that antibodies against the C-terminus of α-syn reduce the intra-neuronal accumulation of α-syn and related deficits in transgenic models of synucleinopathy, probably by abrogating the axonal transport and accumulation of α-syn in in vivo models. Here, we assessed the effect of passive immunization against α-syn in a new mouse model of axonal transport and accumulation of α-syn. For these purpose, non-transgenic, α-syn knock-out and mThy1-α-syn tg (line 61) mice received unilateral intra-cerebral injections with a lentiviral (LV)-α-syn vector construct followed by systemic administration of the monoclonal antibody 1H7 (recognizes amino acids 91-99) or control IgG for 3 months. Cerebral α-syn accumulation and axonopathy was assessed by immunohistochemistry and effects on behavior were assessed by Morris water maze. Unilateral LV-α-syn injection resulted in axonal propagation of α-syn in the contra-lateral site with subsequent behavioral deficits and axonal degeneration. Passive immunization with 1H7 antibody reduced the axonal accumulation of α-syn in the contra-lateral side and ameliorated the behavioral deficits. Together this study supports the notion that immunotherapy might improve the deficits in models of synucleinopathy by reducing the axonal propagation and accumulation of α-syn. This represents a potential new mode of action through which α-syn immunization might work.


Assuntos
Axônios/patologia , Encéfalo/patologia , Imunização Passiva , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , alfa-Sinucleína/imunologia , alfa-Sinucleína/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Transporte Axonal , Axônios/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Lateralidade Funcional , Vetores Genéticos , Humanos , Lentivirus , Aprendizagem em Labirinto/fisiologia , Camundongos Transgênicos , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/psicologia , alfa-Sinucleína/deficiência , alfa-Sinucleína/genética
5.
Mov Disord ; 32(2): 211-218, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27886407

RESUMO

BACKGROUND: α-Synuclein is a major component of pathologic inclusions that characterize Parkinson's disease. PRX002 is an antibody that targets α-synuclein, and its murine parent antibody 9E4 has been shown in preclinical studies to reduce α-synuclein pathology and to protect against cognitive and motor deteriorations and progressive neurodegeneration in human α-synuclein transgenic mice. METHODS: This first-in-human, randomized, double-blind, placebo-controlled, phase 1 study assessed the impact of PRX002 administered to 40 healthy participants in 5 ascending-dose cohorts (n = 8/cohort) in which participants were randomly assigned to receive a single intravenous infusion of study drug (0.3, 1, 3, 10, or 30 mg/kg; n = 6/cohort) or placebo (n = 2/cohort). RESULTS: PRX002 demonstrated favorable safety, tolerability, and pharmacokinetic profiles at all doses tested, with no immunogenicity. No serious adverse events, discontinuations as a result of adverse events, or dose-limiting toxicities were reported. Serum PRX002 exposure was dose proportional; the average terminal half-life across all doses was 18.2 days. A significant dose-dependent reduction in free serum α-synuclein (unbound to PRX002) was apparent within 1 hour after PRX002 administration, whereas total α-synuclein (free plus bound) increased dose-dependently, presumably because of the expected change in kinetics following antibody binding. CONCLUSIONS: This study demonstrates that serum α-synuclein can be safely modulated in a dose-dependent manner after single intravenous infusions of an anti-α-synuclein antibody. These findings support continued development of PRX002, including further characterization of its safety, tolerability, pharmacokinetics, and pharmacodynamic effects in the central nervous system in patients with Parkinson's disease. © 2016 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , alfa-Sinucleína/sangue , alfa-Sinucleína/efeitos dos fármacos , alfa-Sinucleína/imunologia , Adulto , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Imunoglobulina G/imunologia , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Adulto Jovem
6.
Amyloid ; 23(3): 168-177, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27494229

RESUMO

Amyloid light chain (AL) amyloidosis is characterized by misfolded light chain (LC) (amyloid) deposition in various peripheral organs, leading to progressive dysfunction and death. There are no regulatory agency-approved treatments for AL amyloidosis, and none of the available standard of care approaches directly targets the LC protein that constitutes the amyloid. NEOD001, currently in late-stage clinical trials, is a conformation-specific, anti-LC antibody designed to specifically target misfolded LC aggregates and promote phagocytic clearance of AL amyloid deposits. The present study demonstrated that the monoclonal antibody 2A4, the murine form of NEOD001, binds to patient-derived soluble and insoluble LC aggregates and induces phagocytic clearance of AL amyloid in vitro. 2A4 specifically labeled all 21 fresh-frozen organ samples studied, which were derived from 10 patients representing both κ and λ LC amyloidosis subtypes. 2A4 immunoreactivity largely overlapped with thioflavin T-positive labeling, and 2A4 bound both soluble and insoluble LC aggregates extracted from patient tissue. Finally, 2A4 induced macrophage engagement and phagocytic clearance of AL amyloid deposits in vitro. These findings provide further evidence that 2A4/NEOD001 can effectively clear and remove human AL-amyloid from tissue and further support the rationale for the evaluation of NEOD001 in patients with AL amyloidosis.


Assuntos
Proteínas Amiloidogênicas/imunologia , Amiloidose/imunologia , Anticorpos Monoclonais/química , Complexo Antígeno-Anticorpo/química , Cadeias Leves de Imunoglobulina/química , Fagocitose , Proteínas Amiloidogênicas/química , Proteínas Amiloidogênicas/isolamento & purificação , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Anticorpos Monoclonais/biossíntese , Benzotiazóis , Linhagem Celular , Humanos , Cadeias Leves de Imunoglobulina/isolamento & purificação , Camundongos , Monócitos/citologia , Monócitos/imunologia , Agregados Proteicos/imunologia , Ligação Proteica , Coloração e Rotulagem/métodos , Tiazóis/química
7.
J Clin Oncol ; 34(10): 1097-103, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26858336

RESUMO

PURPOSE: Light chain (AL) amyloidosis is caused by the accumulation of misfolded proteins, which induces the dysfunction of vital organs. NEOD001 is a monoclonal antibody targeting these misfolded proteins. We report interim data from a phase I/II dose-escalation/expansion study of NEOD001 in patients with AL amyloidosis and persistent organ dysfunction (NCT01707264). PATIENTS AND METHODS: Patients who had completed at least one previous anti-plasma cell-directed therapy, had partial hematologic response or better, and had persistent organ dysfunction received NEOD001 intravenously every 28 days. Dose levels of 0.5, 1, 2, 4, 8, 16, and 24 mg/kg were evaluated (3 + 3 study design). Primary objectives were to determine the maximum tolerated dose and the recommended dose for future studies and to evaluate safety/tolerability. Secondary and exploratory objectives included pharmacokinetics, immunogenicity, and organ responses on the basis of published consensus criteria. RESULTS: Twenty-seven patients were enrolled in seven cohorts (dose-escalation component). No drug-related serious adverse events (AEs), discontinuations because of drug-related AEs, dose-limiting toxicities, or antidrug antibodies were reported. The most frequent AEs were fatigue, upper respiratory tract infection, cough, and dyspnea. Recommended dosing was 24 mg/kg. Pharmacokinetics support intravenous dosing every 28 days. Of 14 cardiac-evaluable patients, eight (57%) met the criteria for cardiac response and six (43%) had stable disease. Of 15 renal-evaluable patients, nine (60%) met the criteria for renal response and six (40%) had stable disease. CONCLUSION: Monthly infusions of NEOD001 were safe and well tolerated. Recommended future dosing was 24 mg/kg. Organ response rates compared favorably with those reported previously for chemotherapy. A phase II expansion is ongoing. A global phase III study (NCT02312206) has been initiated. Antibody therapy targeting misfolded proteins is a potential new therapy for the management of AL amyloidosis.


Assuntos
Amiloidose/tratamento farmacológico , Amiloidose/imunologia , Anticorpos Monoclonais Humanizados/administração & dosagem , Cadeias Leves de Imunoglobulina , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/prevenção & controle , Plasmócitos/efeitos dos fármacos , Adulto , Idoso , Amiloidose/complicações , Amiloidose/etnologia , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Tosse/induzido quimicamente , Relação Dose-Resposta a Droga , Esquema de Medicação , Dispneia/induzido quimicamente , Fadiga/induzido quimicamente , Feminino , Coração/efeitos dos fármacos , Coração/fisiopatologia , Humanos , Cadeias Leves de Imunoglobulina/imunologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/fisiopatologia , Plasmócitos/imunologia , Infecções Respiratórias/induzido quimicamente , Resultado do Tratamento
8.
Curr Alzheimer Res ; 12(9): 814-28, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26510979

RESUMO

A potential strategy to alleviate the aggregation of intrinsically disordered proteins (IDPs) is to maintain the native functional state of the protein by small molecule binding. However, the targeting of the native state of IDPs by small molecules has been challenging due to their heterogeneous conformational ensembles. To tackle this challenge, we applied a high-throughput chemical microarray surface plasmon resonance imaging screen to detect the binding between small molecules and monomeric full-length Tau, a protein linked with the onset of a range of Tauopathies. The screen identified a novel set of drug-like fragment and lead-like compounds that bound to Tau. We verified that the majority of these hit compounds reduced the aggregation of different Tau constructs in vitro and in N2a cells. These results demonstrate that Tau is a viable receptor of drug-like small molecules. The drug discovery approach that we present can be applied to other IDPs linked to other misfolding diseases such as Alzheimer's and Parkinson's diseases.


Assuntos
Fármacos Neuroprotetores/farmacologia , Tauopatias/tratamento farmacológico , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Benzotiazóis , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Corantes Fluorescentes , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Análise em Microsséries , Microscopia de Fluorescência , Estrutura Molecular , Fármacos Neuroprotetores/química , Agregados Proteicos/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Tiazóis , Proteínas tau/genética
9.
J Neurosci ; 34(28): 9441-54, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25009275

RESUMO

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are common neurodegenerative disorders of the aging population, characterized by progressive and abnormal accumulation of α-synuclein (α-syn). Recent studies have shown that C-terminus (CT) truncation and propagation of α-syn play a role in the pathogenesis of PD/DLB. Therefore, we explored the effect of passive immunization against the CT of α-syn in the mThy1-α-syn transgenic (tg) mouse model, which resembles the striato-nigral and motor deficits of PD. Mice were immunized with the new monoclonal antibodies 1H7, 5C1, or 5D12, all directed against the CT of α-syn. CT α-syn antibodies attenuated synaptic and axonal pathology, reduced the accumulation of CT-truncated α-syn (CT-α-syn) in axons, rescued the loss of tyrosine hydroxylase fibers in striatum, and improved motor and memory deficits. Among them, 1H7 and 5C1 were most effective at decreasing levels of CT-α-syn and higher-molecular-weight aggregates. Furthermore, in vitro studies showed that preincubation of recombinant α-syn with 1H7 and 5C1 prevented CT cleavage of α-syn. In a cell-based system, CT antibodies reduced cell-to-cell propagation of full-length α-syn, but not of the CT-α-syn that lacked the 118-126 aa recognition site needed for antibody binding. Furthermore, the results obtained after lentiviral expression of α-syn suggest that antibodies might be blocking the extracellular truncation of α-syn by calpain-1. Together, these results demonstrate that antibodies against the CT of α-syn reduce levels of CT-truncated fragments of the protein and its propagation, thus ameliorating PD-like pathology and improving behavioral and motor functions in a mouse model of this disease.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Encéfalo/imunologia , Transtornos dos Movimentos/imunologia , Transtornos dos Movimentos/terapia , Transtornos Parkinsonianos/imunologia , Transtornos Parkinsonianos/terapia , alfa-Sinucleína/imunologia , Animais , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Imunoterapia/métodos , Camundongos , Camundongos Transgênicos , Distribuição Tecidual , Resultado do Tratamento
10.
Alzheimers Res Ther ; 6(3): 31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25024748

RESUMO

INTRODUCTION: Immunotherapy targeting amyloid-ß peptide is under active clinical investigation for treatment of Alzheimer's disease (AD). Among the hypotheses being investigated for impact on clinical outcome are the preferred epitope or conformation of amyloid-ß to target for treatment, and the mechanism of action underlying immunotherapy. Bapineuzumab (humanized 3D6), a neo-epitope specific antibody recognizing amyloid-ß1-5 with strong preference for an exposed Asp residue at the N-terminus of the peptide, has undergone advanced clinical testing for treatment of AD. METHODS: To gain further insight into the epitope conformation, we interrogated structural details of amino-terminal epitopes in amyloid-ß using x-ray crystallography of 3D6Fab:amyloid-ß complexes. Humanization of 3D6 was carried out using standard procedures integrating recombinant methods, sequence informatics, and homology modeling predictions to identify important mouse framework residues for retention in the finished humanized product. RESULTS: Here we report the crystal structure of a recombinant Fab fragment of 3D6 in complex with amyloid-ß1-7 solved at 2.0 Å resolution. The N-terminus of amyloid-ß is bound to 3D6 as a 310 helix. The amino-terminal Asp residue is buried deepest in the antibody binding pocket, with the Cß atom of residue 6 visible at the entrance to the binding pocket near the surface of the antibody. We further evaluate homology model based predictions used to guide humanization of 3D6 to bapineuzumab, with actual structure of the Fab. The structure of the Fab:amyloid-ß complex validates design of the humanized antibody, and confirms the amyloid-ß epitope recognized by 3D6 as previously mapped by ELISA. CONCLUSIONS: The conformation of amyloid-ß antigen recognized by 3D6 is novel and distinct from other antibodies recognizing N-terminal epitopes. Our result provides the first report demonstrating structural conservation of antigen contact residues, and conformation of antigen recognized, between the parent murine antibody and its humanized version.

11.
Neuron ; 82(6): 1334-45, 2014 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-24945775

RESUMO

It has been widely reported that ß-amyloid peptide (Aß) blocks long-term potentiation (LTP) of hippocampal synapses. Here, we show evidence that Aß more potently blocks the potentiation of excitatory postsynaptic potential (EPSP)-spike coupling (E-S potentiation). This occurs, not by direct effect on excitatory synapses or postsynaptic neurons, but rather through an indirect mechanism: reduction of endocannabinoid-mediated peritetanic disinhibition. During high-frequency (tetanic) stimulation, somatic synaptic inhibition is suppressed by endocannabinoids. We find that Aß prevents this endocannabinoid-mediated disinhibition, thus leaving synaptic inhibition more intact during tetanic stimulation. This intact inhibition opposes the normal depolarization of hippocampal pyramidal neurons that occurs during tetanus, thus opposing the induction of synaptic plasticity. Thus, a pathway through which Aß can act to modulate neural activity is identified, relevant to learning and memory and how it may mediate aspects of the cognitive decline seen in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Inibição Neural/fisiologia , Fragmentos de Peptídeos/fisiologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/fisiologia , Sinapses/fisiologia , Animais , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar
12.
PLoS One ; 9(2): e87133, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551051

RESUMO

The misfolding of intrinsically disordered proteins such as α-synuclein, tau and the Aß peptide has been associated with many highly debilitating neurodegenerative syndromes including Parkinson's and Alzheimer's diseases. Therapeutic targeting of the monomeric state of such intrinsically disordered proteins by small molecules has, however, been a major challenge because of their heterogeneous conformational properties. We show here that a combination of computational and experimental techniques has led to the identification of a drug-like phenyl-sulfonamide compound (ELN484228), that targets α-synuclein, a key protein in Parkinson's disease. We found that this compound has substantial biological activity in cellular models of α-synuclein-mediated dysfunction, including rescue of α-synuclein-induced disruption of vesicle trafficking and dopaminergic neuronal loss and neurite retraction most likely by reducing the amount of α-synuclein targeted to sites of vesicle mobilization such as the synapse in neurons or the site of bead engulfment in microglial cells. These results indicate that targeting α-synuclein by small molecules represents a promising approach to the development of therapeutic treatments of Parkinson's disease and related conditions.


Assuntos
Proteínas Intrinsicamente Desordenadas/antagonistas & inibidores , Terapia de Alvo Molecular , Doença de Parkinson/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico , alfa-Sinucleína/antagonistas & inibidores , Animais , Sítios de Ligação , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Humanos , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/metabolismo , Camundongos , Modelos Biológicos , Modelos Moleculares , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Doença de Parkinson/patologia , Fagócitos/efeitos dos fármacos , Fagócitos/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo
13.
PLoS One ; 8(8): e71634, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058406

RESUMO

Alpha-synuclein protein is strongly implicated in the pathogenesis Parkinson's disease. Increased expression of α-synuclein due to genetic multiplication or point mutations leads to early onset disease. While α-synuclein is known to modulate membrane vesicle dynamics, it is not clear if this activity is involved in the pathogenic process or if measurable physiological effects of α-synuclein over-expression or mutation exist in vivo. Macrophages and microglia isolated from BAC α-synuclein transgenic mice, which overexpress α-synuclein under regulation of its own promoter, express α-synuclein and exhibit impaired cytokine release and phagocytosis. These processes were affected in vivo as well, both in peritoneal macrophages and microglia in the CNS. Extending these findings to humans, we found similar results with monocytes and fibroblasts isolated from idiopathic or familial Parkinson's disease patients compared to age-matched controls. In summary, this paper provides 1) a new animal model to measure α-synuclein dysfunction; 2) a cellular system to measure synchronized mobilization of α-synuclein and its functional interactions; 3) observations regarding a potential role for innate immune cell function in the development and progression of Parkinson's disease and other human synucleinopathies; 4) putative peripheral biomarkers to study and track these processes in human subjects. While altered neuronal function is a primary issue in PD, the widespread consequence of abnormal α-synuclein expression in other cell types, including immune cells, could play an important role in the neurodegenerative progression of PD and other synucleinopathies. Moreover, increased α-synuclein and altered phagocytosis may provide a useful biomarker for human PD.


Assuntos
Imunidade Inata , Doença de Parkinson/diagnóstico , Doença de Parkinson/imunologia , alfa-Sinucleína/imunologia , Idoso , Idoso de 80 Anos ou mais , Animais , Células Cultivadas , Citocinas/imunologia , Feminino , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Transgênicos , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , Pessoa de Meia-Idade , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Fagocitose , Regulação para Cima , alfa-Sinucleína/genética
14.
Alzheimers Dement ; 9(4): 452-458.e1, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23809366

RESUMO

For decades, researchers have focused primarily on a pathway initiated by amyloid beta aggregation, amyloid deposition, and accumulation in the brain as the key mechanism underlying the disease and the most important treatment target. However, evidence increasingly suggests that amyloid is deposited early during the course of disease, even prior to the onset of clinical symptoms. Thus, targeting amyloid in patients with mild to moderate Alzheimer's disease (AD), as past failed clinical trials have done, may be insufficient to halt further disease progression. Scientists are investigating other molecular and cellular pathways and processes that contribute to AD pathogenesis. Thus, the Alzheimer's Association's Research Roundtable convened a meeting in April 2012 to move beyond amyloid and explore AD as a complex multifactorial disease, with the goal of using a more inclusive perspective to identify novel treatment strategies.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Terapia de Alvo Molecular , Nootrópicos/uso terapêutico , Envelhecimento , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Animais , Autofagia/efeitos dos fármacos , Biomarcadores , Encéfalo/metabolismo , Ciclo Celular/efeitos dos fármacos , Comportamento Cooperativo , Diabetes Mellitus Tipo 2/complicações , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Humanos , Inflamação , Resistência à Insulina , Lisossomos/efeitos dos fármacos , Lisossomos/fisiologia , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Modelos Neurológicos , Neuroimagem , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Nootrópicos/farmacologia , Parcerias Público-Privadas , Alocação de Recursos , Proteínas tau/efeitos dos fármacos , Proteínas tau/fisiologia
15.
J Chem Phys ; 139(3): 035101, 2013 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-23883055

RESUMO

The aggregation of intrinsically disordered peptides and proteins is associated with a wide range of highly debilitating neurological and systemic disorders. In this work we explored the potential of a structure-based drug discovery procedure to target one such system, the soluble monomeric form of the Aß42 peptide. We utilised for this purpose a set of structures of the Aß42 peptide selected from clusters of conformations within an ensemble generated by molecular dynamics simulations. Using these structures we carried out fragment mapping calculations to identify binding "hot spots" on the monomeric form of the Aß42 peptide. This procedure provided a set of hot spots with ligand efficiencies comparable to those observed for structured proteins, and clustered into binding pockets. Such binding pockets exhibited a propensity to bind small molecules known to interact with the Aß42 peptide. Taken together these results provide an initial indication that fragment-based drug discovery may represent a potential therapeutic strategy for diseases associated with the aggregation of intrinsically disordered proteins.


Assuntos
Peptídeos beta-Amiloides/química , Simulação de Dinâmica Molecular , Fragmentos de Peptídeos/química , Sequência de Aminoácidos , Peptídeos beta-Amiloides/metabolismo , Sítios de Ligação , Desenho de Fármacos , Dados de Sequência Molecular , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Solubilidade
16.
Amyloid ; 20(3): 179-87, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23829200

RESUMO

Abstract Amyloid ß-peptide (Aß) is a key molecule in Alzheimer's disease (AD). Reliable immunohistochemical (IHC) methods to detect Aß and Aß-associated factors (AAF) in brain specimens are needed to determine their role in AD pathophysiology. Formic acid (FA) pre-treatment, which is generally used to enable efficient detection of Aß with IHC, induces structural modifications within the Aß, as well as in AAF. Consequently, interpretation of double IHC stainings becomes difficult. Therefore, serial stainings of two newly produced monoclonal antibodies (mAbs) VU-17 and IC16 and two other mAbs (6E10 and 3D6) were performed with four different pre-treatments (no pre-treatment, Tris/EDTA, citrate and FA) and additionally six IHC characteristics were scored: diffuse/compact/classic plaques, arteries with cerebral Aß angiopathy, dyshoric angiopathy, capillaries with dyshoric angiopathy. Subsequently, these stainings were compared with IHC procedures, which are frequently used in a diagnostic setting, employing mAbs 4G8 and 6F/3D with FA pre-treatment. IHC Aß patterns obtained with VU-17 and, IC16 and 3D6 without the use of FA pre-treatment were comparable to those obtained with 4G8 and 6F/3D upon FA pre-treatment. Omission of FA pre-treatment gives the advantage to allow double IHC stainings, detecting both Aß and AAF that otherwise would have been structural modificated upon FA pre-treatment.


Assuntos
Doença de Alzheimer/diagnóstico , Peptídeos beta-Amiloides/análise , Anticorpos Monoclonais , Angiopatia Amiloide Cerebral/diagnóstico , Placa Amiloide/diagnóstico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Capilares/metabolismo , Capilares/patologia , Angiopatia Amiloide Cerebral/metabolismo , Angiopatia Amiloide Cerebral/patologia , Formiatos/química , Humanos , Hibridomas/imunologia , Imuno-Histoquímica , Camundongos , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Estrutura Terciária de Proteína , Sensibilidade e Especificidade
17.
Alzheimers Dement ; 9(5 Suppl): S105-15, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23583235

RESUMO

BACKGROUND: Clinical studies of ß-amyloid (Aß) immunotherapy in Alzheimer's disease (AD) patients have demonstrated reduction of central Aß plaque by positron emission tomography (PET) imaging and the appearance of amyloid-related imaging abnormalities (ARIA). To better understand the relationship between ARIA and the pathophysiology of AD, we undertook a series of studies in PDAPP mice evaluating vascular alterations in the context of central Aß pathology and after anti-Aß immunotherapy. METHODS: We analyzed PDAPP mice treated with either 3 mg/kg/week of 3D6, the murine form of bapineuzumab, or isotype control antibodies for periods ranging from 1 to 36 weeks and evaluated the vascular alterations in the context of Aß pathology and after anti-Aß immunotherapy. The number of mice in each treatment group ranged from 26 to 39 and a total of 345 animals were analyzed. RESULTS: The central vasculature displayed morphological abnormalities associated with vascular Aß deposits. Treatment with 3D6 antibody induced clearance of vascular Aß that was spatially and temporally associated with a transient increase in microhemorrhage and in capillary Aß deposition. Microhemorrhage resolved over a time period that was associated with a recovery of vascular morphology and a decrease in capillary Aß accumulation. CONCLUSIONS: These data suggest that vascular leakage events, such as microhemorrhage, may be related to the removal of vascular Aß. With continued treatment, this initial susceptibility period is followed by restoration of vascular morphology and reduced vulnerability to further vascular leakage events. The data collectively suggested a vascular amyloid clearance model of ARIA, which accounts for the currently known risk factors for the incidence of ARIA in clinical studies.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Vasos Sanguíneos/patologia , Encéfalo/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Aquaporina 4/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/ultraestrutura , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Hemorragias Intracranianas/etiologia , Meninges/patologia , Meninges/ultraestrutura , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutação/genética , Fatores de Tempo
18.
Am J Pathol ; 182(3): 940-53, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23313024

RESUMO

Progressive accumulation of α-synuclein (α-syn) in limbic and striatonigral systems is associated with the neurodegenerative processes in dementia with Lewy bodies (DLB) and Parkinson's disease (PD). The murine Thy-1 (mThy1)-α-syn transgenic (tg) model recapitulates aspects of degenerative processes associated with α-syn accumulation in these disorders. Given that axonal and synaptic pathologies are important features of DLB and PD, we sought to investigate the extent and characteristics of these alterations in mThy1-α-syn tg mice and to determine the contribution of α-syn c-terminally cleaved at amino acid 122 (CT α-syn) to these abnormalities. We generated a novel polyclonal antibody (SYN105) against the c-terminally truncated sequence (amino acids 121 to 123) of α-syn (CT α-syn) and performed immunocytochemical and ultrastructural analyses in mThy1-α-syn tg mice. We found abundant clusters of dystrophic neurites in layers 2 to 3 of the neocortex, the stratum lacunosum, the dentate gyrus, and cornu ammonis 3 of the hippocampus, striatum, thalamus, midbrain, and pons. Dystrophic neurites displayed intense immunoreactivity detected with the SYN105 antibody. Double-labeling studies with antibodies to phosphorylated neurofilaments confirmed the axonal location of full-length and CT α-syn. α-Syn immunoreactive dystrophic neurites contained numerous electrodense laminated structures. These results show that neuritic dystrophy is a prominent pathologic feature of the mThy1-α-syn tg model and suggest that CT α-syn might play an important role in the process of axonal damage in these mice as well as in DLB and PD.


Assuntos
Axônios/patologia , Doença por Corpos de Lewy/patologia , Proteínas Mutantes/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Axônios/metabolismo , Axônios/ultraestrutura , Biomarcadores/metabolismo , Demografia , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Neuritos/metabolismo , Neuritos/patologia , Neuritos/ultraestrutura , Transporte Proteico , Sinapses/metabolismo , Sinapses/patologia , Sinapses/ultraestrutura , Antígenos Thy-1/metabolismo , alfa-Sinucleína/imunologia
19.
Curr Alzheimer Res ; 10(1): 4-10, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22950863

RESUMO

The γ-secretase complex cleaves the carboxy-terminal 99 residue (C99) fragment of the amyloid precursor protein (APP) to generate the amyloid-ß (Aß) peptide. The catalytic activity of this complex is mediated either by the presenilin- 1 (PS1) or the presenilin-2 (PS2) subunit. In vitro and in vivo studies have demonstrated that PS1-containing complexes generate more total Aß product than PS2-containing complexes, indicating greater cleavage activity by PS1- containing γ-secretase complexes at the APP γ-site. However, it remains untested whether γ-secretase cleavage at the APP -site, which precedes γ-site cleavage and produces the physiologically active APP intracellular domain (AICD), follows the same rule. Using a novel Swedish APP-GVP substrate to facilitate the parallel detection of Aß and AICD products from PS1-/-/PS2-/- cells co-transfected with either PS1 or PS2, we observed that while PS1 generates more total Aß product than PS2, consistent with published reports, PS1 and PS2 unexpectedly generate equal amounts of AICD product. We also observed that PS1 and PS2 produce equivalent amounts of Notch intracellular domain (NICD), indicating equal cleavage activity at the Notch S3-site (the corollary of the APP -site). Our findings suggest that processivity differences between PS1 and PS2 underlie the differential production of Aß peptide. Taken together these findings offer novel insights into γ- secretase biology and have important implications for therapeutically targeting γ-secretase.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Presenilina-1/metabolismo , Presenilina-2/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Células Cultivadas , Embrião de Mamíferos , Inibidores Enzimáticos , Fibroblastos , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Fragmentos de Peptídeos , Presenilina-1/genética , Presenilina-2/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores Notch/genética , Transfecção , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
20.
Alzheimers Dement ; 8(5): 445-52, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22959699

RESUMO

To address the pending public health crisis due to Alzheimer's disease (AD) and related neurodegenerative disorders, the Marian S. Ware Alzheimer Program at the University of Pennsylvania held a meeting entitled "State of the Science Conference on the Advancement of Alzheimer's Diagnosis, Treatment and Care," on June 21-22, 2012. The meeting comprised four workgroups focusing on Biomarkers; Clinical Care and Health Services Research; Drug Development; and Health Economics, Policy, and Ethics. The workgroups shared, discussed, and compiled an integrated set of priorities, recommendations, and action plans, which are presented in this article.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/terapia , Política de Saúde , Pesquisa sobre Serviços de Saúde , Doença de Alzheimer/epidemiologia , Análise Custo-Benefício/estatística & dados numéricos , Feminino , Humanos , Masculino , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...