Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nat Neurosci ; 22(7): 1099-1109, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31235907

RESUMO

Parkinson's disease, the most common age-related movement disorder, is a progressive neurodegenerative disease with unclear etiology. Key neuropathological hallmarks are Lewy bodies and Lewy neurites: neuronal inclusions immunopositive for the protein α-synuclein. In-depth ultrastructural analysis of Lewy pathology is crucial to understanding pathogenesis of this disease. Using correlative light and electron microscopy and tomography on postmortem human brain tissue from Parkinson's disease brain donors, we identified α-synuclein immunopositive Lewy pathology and show a crowded environment of membranes therein, including vesicular structures and dysmorphic organelles. Filaments interspersed between the membranes and organelles were identifiable in many but not all α-synuclein inclusions. Crowding of organellar components was confirmed by stimulated emission depletion (STED)-based super-resolution microscopy, and high lipid content within α-synuclein immunopositive inclusions was corroborated by confocal imaging, Fourier-transform coherent anti-Stokes Raman scattering infrared imaging and lipidomics. Applying such correlative high-resolution imaging and biophysical approaches, we discovered an aggregated protein-lipid compartmentalization not previously described in the Parkinsons' disease brain.


Assuntos
Membranas Intracelulares/ultraestrutura , Corpos de Lewy/ultraestrutura , Doença por Corpos de Lewy/patologia , Lipídeos de Membrana/análise , Organelas/ultraestrutura , Doença de Parkinson/patologia , alfa-Sinucleína/análise , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Hipocampo/química , Hipocampo/ultraestrutura , Humanos , Imageamento Tridimensional , Corpos de Lewy/química , Doença por Corpos de Lewy/metabolismo , Mesencéfalo/química , Mesencéfalo/ultraestrutura , Microscopia Confocal , Microscopia Eletrônica/métodos , Microscopia de Fluorescência , Doença de Parkinson/metabolismo , Substância Negra/química , Substância Negra/ultraestrutura , Sequenciamento do Exoma
2.
Alzheimers Dement ; 15(3): 453-464, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30442540

RESUMO

INTRODUCTION: Murine microglia expressing the Alzheimer's disease-linked TREM2R47H mutation display variable decrease in phagocytosis, while impaired phagocytosis is reported following loss of TREM2. However, no data exist on TREM2+/R47H human microglia. Therefore, we created human pluripotent stem cell (hPSC) monocytes and transdifferentiated microglia-like cells (tMGs) to examine the effect of the TREM2+/R47H mutation and loss of TREM2 on phagocytosis. METHODS: We generated isogenic TREM2+/R47H, TREM2+/-, and TREM2-/- hPSCs using CRISPR/Cas9. Following differentiation to monocytes and tMGs, we studied the uptake of Escherichia coli fragments and analyzed amyloid plaque clearance from cryosections of APP/PS1+/- mouse brains. RESULTS: We demonstrated that tMGs resemble cultured human microglia. TREM2+/- and TREM2-/- hPSC monocytes and tMGs phagocytosed significantly less E. coli fragments and cleared less amyloid plaques than wild-type hPSC progeny, with no difference for TREM2+/R47H progeny. DISCUSSION: In vitro phagocytosis of hPSC monocytes and tMGs was not affected by the TREM2+/R47H mutation but was significantly impaired in TREM2+/- and TREM2-/- progeny.


Assuntos
Glicoproteínas de Membrana/deficiência , Microglia/metabolismo , Monócitos/metabolismo , Placa Amiloide/metabolismo , Receptores Imunológicos/deficiência , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo , Sistemas CRISPR-Cas , Células Cultivadas , Escherichia coli , Glicoproteínas de Membrana/genética , Camundongos Transgênicos , Fagocitose , Células-Tronco Pluripotentes , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores Imunológicos/genética
3.
Sci Rep ; 8(1): 18046, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30575769

RESUMO

Corpora amylacea are cell-derived structures that appear physiologically in the aged human brain. While their histological identification is straightforward, their ultrastructural composition and microenvironment at the nanoscale have remained unclear so far, as has their relevance to aging and certain disease states that involve the sequestration of toxic cellular metabolites. Here, we apply correlative serial block-face scanning electron microscopy and transmission electron tomography to gain three-dimensional insight into the ultrastructure and surrounding microenvironment of cerebral Corpora amylacea in the human brainstem and hippocampal region. We find that cerebral Corpora amylacea are composed of dense labyrinth-like sheets of lipid membranes, contain vesicles as well as morphologically preserved mitochondria, and are in close proximity to blood vessels and the glymphatic system, primarily within the cytoplasm of perivascular glial cells. Our results clarify the nature of cerebral Corpora amylacea and provide first hints on how they may arise and develop in the aging brain.


Assuntos
Encéfalo/patologia , Encéfalo/ultraestrutura , Corpos de Inclusão/patologia , Organelas/patologia , Idoso , Idoso de 80 Anos ou mais , Autopsia , Encéfalo/diagnóstico por imagem , Tronco Encefálico/diagnóstico por imagem , Tronco Encefálico/patologia , Região CA2 Hipocampal/diagnóstico por imagem , Região CA2 Hipocampal/patologia , Humanos , Imageamento Tridimensional , Microscopia Eletrônica/métodos , Doença de Parkinson/patologia , Parte Compacta da Substância Negra/patologia
4.
Theranostics ; 8(18): 4957-4968, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30429879

RESUMO

Beta secretase (BACE) inhibitors are promising therapeutic compounds currently in clinical phase II/III trials. Preclinical [18F]-florbetaben (FBB) amyloid PET imaging facilitates longitudinal monitoring of amyloidosis in Alzheimer's disease (AD) mouse models. Therefore, we applied this theranostic concept to investigate, by serial FBB PET, the efficacy of a novel BACE1 inhibitor in the PS2APP mouse, which is characterized by early and massive amyloid deposition. Methods: PS2APP and C57BL/6 (WT) mice were assigned to treatment (PS2APP: N=13; WT: N=11) and vehicle control (PS2APP: N=13; WT: N=11) groups at the age of 9.5 months. All animals had a baseline PET scan and follow-up scans at two months and after completion of the four-month treatment period. In addition to this longitudinal analysis of cerebral amyloidosis by PET, we undertook biochemical amyloid peptide quantification and histological amyloid plaque analyses after the final PET session. Results: BACE1 inhibitor-treated transgenic mice revealed a progression of the frontal cortical amyloid signal by 8.4 ± 2.2% during the whole treatment period, which was distinctly lower when compared to vehicle-treated mice (15.3 ± 4.4%, p<0.001). A full inhibition of progression was evident in regions with <3.7% of the increase in controls, whereas regions with >10% of the increase in controls showed only 40% attenuation with BACE1 inhibition. BACE1 inhibition in mice with lower amyloidosis at treatment initiation showed a higher efficacy in attenuating progression to PET. A predominant reduction of small plaques in treated mice indicated a main effect of BACE1 on inhibition of de novo amyloidogenesis. Conclusions: This theranostic study with BACE1 treatment in a transgenic AD model together with amyloid PET monitoring indicated that progression of amyloidosis is more effectively reduced in regions with low initial plaque development and revealed the need of an early treatment initiation during amyloidogenesis.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/análise , Compostos de Anilina/administração & dosagem , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Estilbenos/administração & dosagem , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tomografia por Emissão de Pósitrons/métodos , Resultado do Tratamento
5.
Cell Rep ; 22(1): 149-162, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29298417

RESUMO

Receptors show promise for the transport of monoclonal antibodies (mAbs) across the blood-brain barrier. However, safety liabilities associated with peripheral receptor binding and Fc effector function have been reported. We present the Brain Shuttle-mAb (BS-mAb) technology, and we investigate the role of Fc effector function in vitro and in an Fcγ receptor (FcγR)-humanized mouse model. Strong first infusion reactions (FIRs) were observed for a conventional mAb against transferrin receptor (TfR) with a wild-type immunoglobulin G1 (IgG1) Fc. Fc effector-dead constructs completely eliminated all FIRs. Remarkably, no FIR was observed for the BS-mAb construct with a native IgG1 Fc function. Using various BS-mAb constructs, we show that TfR binding through the C-terminal BS module attenuates Fc-FcγR interactions, primarily because of steric hindrance. Nevertheless, BS-mAbs maintain effector function activity when binding their brain target. Thus, mAbs with full effector function can be transported in a stealth mode in the periphery while fully active when engaged with their brain target.


Assuntos
Doença de Alzheimer/metabolismo , Anticorpos Monoclonais , Barreira Hematoencefálica/metabolismo , Sistemas de Liberação de Medicamentos , Imunoglobulina G/farmacologia , Receptores de IgG/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Barreira Hematoencefálica/patologia , Células CHO , Cricetulus , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Receptores de IgG/genética
6.
EBioMedicine ; 27: 258-274, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29269042

RESUMO

Age-related macular degeneration (AMD) is the leading cause of irreversible vision loss. The protein HtrA1 is enriched in retinal pigment epithelial (RPE) cells isolated from AMD patients and in drusen deposits. However, it is poorly understood how increased levels of HtrA1 affect the physiological function of the RPE at the intracellular level. Here, we developed hfRPE (human fetal retinal pigment epithelial) cell culture model where cells fully differentiated into a polarized functional monolayer. In this model, we fine-tuned the cellular levels of HtrA1 by targeted overexpression. Our data show that HtrA1 enzymatic activity leads to intracellular degradation of tubulin with a corresponding reduction in the number of microtubules, and consequently to an altered mechanical cell phenotype. HtrA1 overexpression further leads to impaired apical processes and decreased phagocytosis, an essential function for photoreceptor survival. These cellular alterations correlate with the AMD phenotype and thus highlight HtrA1 as an intracellular target for therapeutic interventions towards AMD treatment.


Assuntos
Polaridade Celular , Serina Peptidase 1 de Requerimento de Alta Temperatura A/metabolismo , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Modelos Biológicos , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Tubulina (Proteína)/metabolismo , Junções Aderentes/metabolismo , Adulto , Feto/metabolismo , Serina Peptidase 1 de Requerimento de Alta Temperatura A/genética , Humanos , Microtúbulos/metabolismo , Mutação/genética , Nanopartículas/química , Fagocitose , Polimerização , Agregados Proteicos , Ligação Proteica , Transcrição Gênica
7.
Reprod Toxicol ; 73: 362-371, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28754630

RESUMO

Gantenerumab is intended for the treatment of Alzheimer's disease. It is a fully human recombinant monoclonal IgG1 which binds aggregated forms of amyloid beta such as Aß oligomers, fibrils and neuritic amyloid plaques. A full package of developmental and reproductive toxicity (DART) studies was performed in the PS2APP double-transgenic mouse model of Alzheimer's disease. A combined fertility and embryo fetal development study and a pre-and post-natal development study were performed. The PS2APP mouse model allowed a more complete DART evaluation than would have been possible in conventional species or strains which do not express the target antigen of gantenerumab. No developmental or reproductive hazards were identified.


Assuntos
Anticorpos Monoclonais/toxicidade , Doença de Alzheimer , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Modelos Animais de Doenças , Embrião de Mamíferos/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Fertilidade/efeitos dos fármacos , Desenvolvimento Fetal/efeitos dos fármacos , Feto/efeitos dos fármacos , Imunoglobulina G/imunologia , Masculino , Camundongos Transgênicos , Presenilina-2/genética
8.
J Pharmacol Exp Ther ; 362(3): 413-423, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28642233

RESUMO

Monoamine oxidase B (MAO-B) has been implicated in the pathogenesis of Alzheimer's disease (AD) and other neurodegenerative disorders. Increased MAO-B expression in astroglia has been observed adjacent to amyloid plaques in AD patient brains. This phenomenon is hypothesized to lead to increased production of hydrogen peroxide and reactive oxygen species (ROS), thereby contributing to AD pathology. Therefore, reduction of ROS-induced oxidative stress via inhibition of MAO-B activity may delay the progression of the disease. In the present study we report the pharmacological properties of sembragiline, a novel selective MAO-B inhibitor specifically developed for the treatment of AD, and on its effect on ROS-mediated neuronal injury and astrogliosis in MAO-B transgenic animals. Sembragiline showed potent and long-lasting MAO-B-selective inhibition and did not inhibit MAO-A at doses where full inhibition of MAO-B was observed. Such selectivity should translate into a favorable clinical safety profile. Indeed, sembragiline neither induced the serotonin syndrome when administered together with the serotonin precursor l-5-hydroxytryptophan in combination with antidepressants such as fluoxetine, nor potentiated the pressor effect of tyramine. Additionally, in experiments using a transgenic animal model conditionally overexpressing MAO-B in astroglia, sembragiline protected against neuronal loss and reduced both ROS formation and reactive astrogliosis. Taken together, these findings warrant further investigation of the potential therapeutic benefit of MAO-B inhibitors in patients with AD and other neurologic disorders.


Assuntos
Acetamidas/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Inibidores da Monoaminoxidase/uso terapêutico , Monoaminoxidase/efeitos dos fármacos , Pirrolidinonas/uso terapêutico , 5-Hidroxitriptofano/farmacologia , Acetamidas/farmacocinética , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Gliose/tratamento farmacológico , Gliose/patologia , Humanos , Hipertensão/induzido quimicamente , Hipertensão/prevenção & controle , Masculino , Monoaminoxidase/genética , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/farmacocinética , Atividade Motora/efeitos dos fármacos , Neurotransmissores/metabolismo , Pirrolidinonas/farmacocinética , Ratos , Ratos Transgênicos , Espécies Reativas de Oxigênio/metabolismo , Especificidade por Substrato , Distribuição Tecidual
9.
EMBO Mol Med ; 8(9): 992-1004, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27402340

RESUMO

Immunotherapeutic approaches are currently the most advanced treatments for Alzheimer's disease (AD). Antibodies against amyloid ß-peptide (Aß) bind to amyloid plaques and induce their clearance by microglia via Fc receptor-mediated phagocytosis. Dysfunctions of microglia may play a pivotal role in AD pathogenesis and could result in reduced efficacy of antibody-mediated Aß clearance. Recently, heterozygous mutations in the triggering receptor expressed on myeloid cells 2 (TREM2), a microglial gene involved in phagocytosis, were genetically linked to late onset AD Loss of TREM2 reduces the ability of microglia to engulf Aß. We have now investigated whether loss of TREM2 affects the efficacy of immunotherapeutic approaches. We show that anti-Aß antibodies stimulate Aß uptake and amyloid plaque clearance in a dose-dependent manner in the presence or absence of TREM2. However, TREM2-deficient N9 microglial cell lines, macrophages as well as primary microglia showed significantly reduced uptake of antibody-bound Aß and as a consequence reduced clearance of amyloid plaques. Titration experiments revealed that reduced efficacy of amyloid plaque clearance by Trem2 knockout cells can be compensated by elevating the concentration of therapeutic antibodies.


Assuntos
Amiloide/metabolismo , Glicoproteínas de Membrana/deficiência , Neuroglia/imunologia , Fagocitose , Receptores Imunológicos/deficiência , Animais , Células Cultivadas , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroglia/fisiologia
10.
Brain ; 139(Pt 5): 1587-604, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26956423

RESUMO

Passive immunization against misfolded toxic proteins is a promising approach to treat neurodegenerative disorders. For effective immunotherapy against Alzheimer's disease, recent clinical data indicate that monoclonal antibodies directed against the amyloid-ß peptide should be administered before the onset of symptoms associated with irreversible brain damage. It is therefore critical to develop technologies for continuous antibody delivery applicable to disease prevention. Here, we addressed this question using a bioactive cellular implant to deliver recombinant anti-amyloid-ß antibodies in the subcutaneous tissue. An encapsulating device permeable to macromolecules supports the long-term survival of myogenic cells over more than 10 months in immunocompetent allogeneic recipients. The encapsulated cells are genetically engineered to secrete high levels of anti-amyloid-ß antibodies. Peripheral implantation leads to continuous antibody delivery to reach plasma levels that exceed 50 µg/ml. In a proof-of-concept study, we show that the recombinant antibodies produced by this system penetrate the brain and bind amyloid plaques in two mouse models of the Alzheimer's pathology. When encapsulated cells are implanted before the onset of amyloid plaque deposition in TauPS2APP mice, chronic exposure to anti-amyloid-ß antibodies dramatically reduces amyloid-ß40 and amyloid-ß42 levels in the brain, decreases amyloid plaque burden, and most notably, prevents phospho-tau pathology in the hippocampus. These results support the use of encapsulated cell implants for passive immunotherapy against the misfolded proteins, which accumulate in Alzheimer's disease and other neurodegenerative disorders.


Assuntos
Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/antagonistas & inibidores , Anticorpos Monoclonais/farmacologia , Implantes de Medicamento , Imunização Passiva/métodos , Tauopatias/prevenção & controle , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/farmacocinética , Encéfalo/metabolismo , Células Cultivadas , Camundongos , Camundongos Transgênicos , Neuroproteção , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Absorção Subcutânea
11.
Brain ; 137(Pt 10): 2834-46, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25085375

RESUMO

The severity of tau pathology in Alzheimer's disease brain correlates closely with disease progression. Tau immunotherapy has therefore been proposed as a new therapeutic approach to Alzheimer's disease and encouraging results have been obtained by active or passive immunization of tau transgenic mice. This work investigates the mechanism by which immunotherapy can impact tau pathology. We demonstrate the development of Alzheimer's disease-like tau pathology in a triple transgenic mouse model of Alzheimer's disease and show that tau/pS422 is present in membrane microdomains on the neuronal cell surface. Chronic, peripheral administration of anti-tau/pS422 antibody reduces the accumulation of tau pathology. The unequivocal presence of anti-tau/pS422 antibody inside neurons and in lysosomes is demonstrated. We propose that anti-tau/pS422 antibody binds to membrane-associated tau/pS422 and that the antigen-antibody complexes are cleared intracellularly, thereby offering one explanation for how tau immunotherapy can ameliorate neuronal tau pathology.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Anticorpos/metabolismo , Proteínas tau/imunologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Western Blotting , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Modelos Animais de Doenças , Progressão da Doença , Imunofluorescência , Humanos , Processamento de Imagem Assistida por Computador , Lisossomos/metabolismo , Lisossomos/patologia , Microdomínios da Membrana/patologia , Camundongos , Camundongos Transgênicos , Fosforilação , Sarcosina/análogos & derivados , Sarcosina/química
12.
J Neurosci ; 34(35): 11621-30, 2014 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-25164658

RESUMO

Therapeutic approaches for prevention or reduction of amyloidosis are currently a main objective in basic and clinical research on Alzheimer's disease. Among the agents explored in clinical trials are anti-Aß peptide antibodies and secretase inhibitors. Most anti-Aß antibodies are considered to act via inhibition of amyloidosis and enhanced clearance of existing amyloid, although secretase inhibitors reduce the de novo production of Aß. Limited information is currently available on the efficacy and potential advantages of combinatorial antiamyloid treatment. We performed a chronic study in APPLondon transgenic mice that received treatment with anti-Aß antibody gantenerumab and BACE inhibitor RO5508887, either as mono- or combination treatment. Treatment aimed to evaluate efficacy on amyloid progression, similar to preexisting amyloidosis as present in Alzheimer's disease patients. Mono-treatments with either compound caused a dose-dependent reduction of total brain Aß and amyloid burden. Combination treatment with both compounds significantly enhanced the antiamyloid effect. The observed combination effect was most pronounced for lowering of amyloid plaque load and plaque number, which suggests effective inhibition of de novo plaque formation. Moreover, significantly enhanced clearance of pre-existing amyloid plaques was observed when gantenerumab was coadministered with RO5508887. BACE inhibition led to a significant time- and dose-dependent decrease in CSF Aß, which was not observed for gantenerumab treatment. Our results demonstrate that combining these two antiamyloid agents enhances overall efficacy and suggests that combination treatments may be of clinical relevance.


Assuntos
Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Anticorpos Monoclonais/administração & dosagem , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Encéfalo/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Anticorpos Monoclonais Humanizados , Encéfalo/patologia , Modelos Animais de Doenças , Quimioterapia Combinada , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placa Amiloide/patologia
13.
Neuron ; 81(1): 49-60, 2014 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-24411731

RESUMO

Although biotherapeutics have vast potential for treating brain disorders, their use has been limited due to low exposure across the blood-brain barrier (BBB). We report that by manipulating the binding mode of an antibody fragment to the transferrin receptor (TfR), we have developed a Brain Shuttle module, which can be engineered into a standard therapeutic antibody for successful BBB transcytosis. Brain Shuttle version of an anti-Aß antibody, which uses a monovalent binding mode to the TfR, increases ß-Amyloid target engagement in a mouse model of Alzheimer's disease by 55-fold compared to the parent antibody. We provide in vitro and in vivo evidence that the monovalent binding mode facilitates transcellular transport, whereas a bivalent binding mode leads to lysosome sorting. Enhanced target engagement of the Brain Shuttle module translates into a significant improvement in amyloid reduction. These findings have major implications for the development of biologics-based treatment of brain disorders.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Transporte Proteico/fisiologia , Anticorpos de Cadeia Única/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Linhagem Celular Transformada , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrolídeos/farmacologia , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Presenilina-1/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/imunologia , Transporte Proteico/efeitos dos fármacos , Receptores da Transferrina/imunologia , Receptores da Transferrina/metabolismo , Anticorpos de Cadeia Única/farmacologia , Anticorpos de Cadeia Única/uso terapêutico , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Fatores de Tempo , Transcitose/efeitos dos fármacos , Transcitose/genética , Transcitose/imunologia
14.
Biomaterials ; 35(2): 792-802, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24344356

RESUMO

The controlled delivery of antibodies by immunoisolated bioimplants containing genetically engineered cells is an attractive and safe approach for chronic treatments. To reach therapeutic antibody levels there is a need to generate renewable cell lines, which can long-term survive in macroencapsulation devices while maintaining high antibody specific productivity. Here we have developed a dual lentiviral vector strategy for the genetic engineering of cell lines compatible with macroencapsulation, using separate vectors encoding IgG light and heavy chains. We show that IgG expression level can be maximized as a function of vector dose and transgene ratio. This approach allows for the generation of stable populations of IgG-expressing C2C12 mouse myoblasts, and for the subsequent isolation of clones stably secreting high IgG levels. Moreover, we demonstrate that cell transduction using this lentiviral system leads to the production of a functional glycosylated antibody by myogenic cells. Subsequent implantation of antibody-secreting cells in a high-capacity macroencapsulation device enables continuous delivery of recombinant antibodies in the mouse subcutaneous tissue, leading to substantial levels of therapeutic IgG detectable in the plasma.


Assuntos
Células Imobilizadas/citologia , Engenharia Genética/métodos , Vetores Genéticos , Imunoglobulina G/administração & dosagem , Lentivirus/genética , Mioblastos/citologia , Animais , Linhagem Celular , Clonagem Molecular , Sistemas de Liberação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Terapia Genética , Glicosilação , Humanos , Camundongos , Regiões Promotoras Genéticas , Proteínas Recombinantes/administração & dosagem , Gordura Subcutânea/metabolismo , Transgenes
15.
J Alzheimers Dis ; 28(1): 49-69, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21955818

RESUMO

The amyloid-ß lowering capacity of anti-Aß antibodies has been demonstrated in transgenic models of Alzheimer's disease (AD) and in AD patients. While the mechanism of immunotherapeutic amyloid-ß removal is controversial, antibody-mediated sequestration of peripheral Aß versus microglial phagocytic activity and disassembly of cerebral amyloid (or a combination thereof) has been proposed. For successful Aß immunotherapy, we hypothesized that high affinity antibody binding to amyloid-ß plaques and recruitment of brain effector cells is required for most efficient amyloid clearance. Here we report the generation of a novel fully human anti-Aß antibody, gantenerumab, optimized in vitro for binding with sub-nanomolar affinity to a conformational epitope expressed on amyloid-ß fibrils using HuCAL(®) phage display technologies. In peptide maps, both N-terminal and central portions of Aß were recognized by gantenerumab. Remarkably, a novel orientation of N-terminal Aß bound to the complementarity determining regions was identified by x-ray analysis of a gantenerumab Fab-Aß(1-11) complex. In functional assays gantenerumab induced cellular phagocytosis of human amyloid-ß deposits in AD brain slices when co-cultured with primary human macrophages and neutralized oligomeric Aß42-mediated inhibitory effects on long-term potentiation in rat brain. In APP751(swedish)xPS2(N141I) transgenic mice, gantenerumab showed sustained binding to cerebral amyloid-ß and, upon chronic treatment, significantly reduced small amyloid-ß plaques by recruiting microglia and prevented new plaque formation. Unlike other Aß antibodies, gantenerumab did not alter plasma Aß suggesting undisturbed systemic clearance of soluble Aß. These studies demonstrated that gantenerumab preferentially interacts with aggregated Aß in the brain and lowers amyloid-ß by eliciting effector cell-mediated clearance.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais/metabolismo , Sequência de Aminoácidos , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Células CHO , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Cricetinae , Cricetulus , Cristalografia por Raios X , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Fagocitose/efeitos dos fármacos , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley
16.
Arch Neurol ; 69(2): 198-207, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21987394

RESUMO

BACKGROUND: Gantenerumab is a fully human anti-Aß monoclonal antibody in clinical development for the treatment of Alzheimer disease (AD). OBJECTIVES: To investigate whether treatment with gantenerumab leads to a measurable reduction in the level of Aß amyloid in the brain and to elucidate the mechanism of amyloid reduction. DESIGN: A multicenter, randomized, double-blind, placebo-controlled, ascending-dose positron emission tomographic study. Additionally, ex vivo studies of human brain slices from an independent sample of patients who had AD were performed. SETTING: Three university medical centers. PATIENTS: Patients with mild-to-moderate AD. INTERVENTION: Two consecutive cohorts of patients received 2 to 7 infusions of intravenous gantenerumab (60 or 200 mg) or placebo every 4 weeks. Brain slices from patients who had AD were coincubated with gantenerumab at increasing concentrations and with human microglial cells. MAIN OUTCOME MEASURES: Percent change in the ratio of regional carbon 11-labeled Pittsburgh Compound B retention in vivo and semiquantitative assessment of gantenerumab-induced phagocytosis ex vivo. RESULTS: Sixteen patients with end-of-treatment positron emission tomographic scans were included in the analysis. The mean (95% CI) percent change from baseline difference relative to placebo (n = 4) in cortical brain amyloid level was -15.6% (95% CI, -42.7 to 11.6) for the 60-mg group (n = 6) and -35.7% (95% CI, -63.5 to -7.9) for the 200-mg group (n = 6). Two patients in the 200-mg group showed transient and focal areas of inflammation or vasogenic edema on magnetic resonance imaging scans at sites with the highest level of amyloid reduction. Gantenerumab induced phagocytosis of human amyloid in a dose-dependent manner ex vivo. CONCLUSION: Gantenerumab treatment resulted in a dose-dependent reduction in brain amyloid level, possibly through an effector cell-mediated mechanism of action.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Vacinas contra Alzheimer/uso terapêutico , Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais/uso terapêutico , Idoso , Doença de Alzheimer/radioterapia , Compostos de Anilina , Anticorpos Monoclonais Humanizados , Química Encefálica/efeitos dos fármacos , Contagem de Células , Estudos de Coortes , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Injeções Intravenosas , Masculino , Microglia/fisiologia , Pessoa de Meia-Idade , Fagocitose/fisiologia , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Tamanho da Amostra , Tiazóis
17.
Mol Cell Biochem ; 357(1-2): 199-207, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21638028

RESUMO

Phosphorylation of Tau at serine 422 promotes Tau aggregation. The kinase that is responsible for this key phosphorylation event has so far not been identified but could be a potential drug target for Alzheimer's disease. We describe here an assay strategy to identify this kinase. Using a combination of screening a library of 65'000 kinase inhibitors and in vitro inhibitor target profiling of the screening hits using the Ambit kinase platform, MKK4 was identified as playing a key role in Tau-S422 phosphorylation in human neuroblastoma cells.


Assuntos
Doença de Alzheimer/metabolismo , MAP Quinase Quinase 4/metabolismo , Fosfotransferases/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Serina/metabolismo , Proteínas tau/metabolismo , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Humanos , Estrutura Molecular , Fosforilação , Fosfotransferases/genética , Fosfotransferases/metabolismo , Serina/química
18.
Brain Res ; 1363: 180-90, 2010 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-20875805

RESUMO

Altered glutamatergic synaptic transmission is among the key events defining the course of Alzheimer's disease (AD). mGlu2 receptors, a subtype of group II metabotropic glutamate receptors, regulate (as autoreceptors) fast synaptic transmission in the CNS via the controlled release of the excitatory amino acid glutamate. Since their pharmacological manipulation in rodents has been reported to affect cognition, they are potential drug targets for AD therapy. We examined the fate of these receptors in cases of AD as well as in aging PS2APP mice--a proposed model of the disease. In vitro binding of [(3)H]LY354740, a selective group II agonist (with selective affinity for mGlu2 receptors, under the assay conditions used) and quantitative radioautography revealed a partial, but highly significant, loss of receptors in amyloid-affected discrete brain regions of AD cases and PS2APP mice. Among the mouse brain regions affected were, above all, the subiculum but also frontolateral cortex, dentate gyrus, lacunosum moleculare and caudate putamen. In AD, significant receptor losses were registered in entorhinal cortex and lacunosum moleculare (40% and 35%, respectively). These findings have implications for the development of selective ligands for symptomatic therapy in AD and for its diagnosis.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Fragmentos de Peptídeos/metabolismo , Presenilina-2/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/farmacologia , Animais , Compostos Bicíclicos com Pontes/metabolismo , Compostos Bicíclicos com Pontes/farmacologia , Agonistas de Aminoácidos Excitatórios/metabolismo , Agonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/patologia , Humanos , Radioisótopos do Iodo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Fragmentos de Peptídeos/farmacologia , Presenilina-2/genética , Ensaio Radioligante , Ratos , Ratos Endogâmicos F344 , Trítio
19.
Neurobiol Dis ; 37(2): 294-306, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19781645

RESUMO

Amyloid beta peptides and microtubule-associated protein Tau are misfolded and form aggregates in brains of Alzheimer's disease patients. To examine their specific roles in the pathogenesis of Alzheimer's disease and their relevance in neurodegenerative processes, we have created TauPS2APP triple transgenic mice that express human mutated Amyloid Precursor Protein, presenilin 2 and Tau. We present a cross-sectional analysis of these mice at 4, 8, 12 and 16 months of age. By comparing with single transgenic Tau mice, we demonstrate that accumulation of Abeta in TauPS2APP triple transgenic mice impacts on Tau pathology by increasing the phosphorylation of Tau at serine 422, as determined by a novel immunodetection method that is able to reliably measure phospho-Tau species in transgenic mouse brains. The TauPS2APP triple transgenic mouse model will be very useful for studying the effect of new therapeutic paradigms on amyloid deposition and downstream neurofibrillary tangle development.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Presenilina-2/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Estudos Transversais , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Marcadores Genéticos/genética , Predisposição Genética para Doença/genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/metabolismo , Fosforilação , Placa Amiloide/metabolismo , Presenilina-2/genética , Regulação para Cima/fisiologia , Proteínas tau/genética
20.
Magn Reson Med ; 62(1): 35-45, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19449370

RESUMO

Function and morphology of the cerebral vasculature were studied in the amyloid (Abeta) plaque-containing double-transgenic (TG) B6.PS2APP Alzheimer's disease (AD) mouse model with MRI at an age range of 10 to 17 months. Perfusion, blood volume, and average vessel geometry were assessed in the brain and compared to age-matched controls (wild-type [WT] C57Bl/6). Additionally, the MR relaxation times T(1), T(2), and T(2)* were measured to detect potential pathological changes that might be associated with Abeta plaque depositions. Both decreased perfusion and decreased blood volume were observed in the occipital cortex in B6.PS2APP mice as compared to controls. A significant decrease in T(1) and T(2) was found in the frontal cortex and in the subiculum/parasubiculum. Immunohistochemistry confirmed plaque depositions in the cortex and in the subiculum/parasubiculum. In summary, our data indicate a reduced blood supply of B6.PS2APP mice in the occipital cortex that parallels the findings in cortical regions of patients with AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Velocidade do Fluxo Sanguíneo , Isquemia Encefálica/fisiopatologia , Encéfalo/fisiopatologia , Circulação Cerebrovascular , Modelos Animais de Doenças , Imageamento por Ressonância Magnética/métodos , Doença de Alzheimer/diagnóstico , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Isquemia Encefálica/diagnóstico , Humanos , Interpretação de Imagem Assistida por Computador/métodos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA