Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Alzheimers Dement (N Y) ; 10(1): e12452, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38356474

RESUMO

INTRODUCTION: Heterozygous mutations in the GRN gene lead to reduced progranulin (PGRN) levels in plasma and cerebrospinal fluid (CSF) and are causative of frontotemporal dementia (FTD) with > 90% penetrance. Latozinemab is a human monoclonal immunoglobulin G1 antibody that is being developed to increase PGRN levels in individuals with FTD caused by heterozygous loss-of-function GRN mutations. METHODS: A first-in-human phase 1 study was conducted to evaluate the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of multiple-dose intravenous administration of latozinemab in eight symptomatic participants with FTD caused by a heterozygous loss-of-function GRN mutation (FTD-GRN). RESULTS: Latozinemab demonstrated favorable safety and PK/PD profiles. Multiple-dose administration of latozinemab increased plasma and CSF PGRN levels in participants with FTD-GRN to levels that approximated those seen in healthy volunteers. DISCUSSION: Data from the first-in-human phase 1 study support further development of latozinemab for the treatment of FTD-GRN. Highlights: GRN mutations decrease progranulin (PGRN) and cause frontotemporal dementia (FTD).Latozinemab is being developed as a PGRN-elevating therapy.Latozinemab demonstrated a favorable safety profile in a phase 1 clinical trial.Latozinemab increased PGRN levels in the CNS of symptomatic FTD-GRN participants.

2.
Int J Mol Sci ; 24(21)2023 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-37958929

RESUMO

Immuno-neurology is an emerging therapeutic strategy for dementia and neurodegeneration designed to address immune surveillance failure in the brain. Microglia, as central nervous system (CNS)-resident myeloid cells, routinely perform surveillance of the brain and support neuronal function. Loss-of-function (LOF) mutations causing decreased levels of progranulin (PGRN), an immune regulatory protein, lead to dysfunctional microglia and are associated with multiple neurodegenerative diseases, including frontotemporal dementia caused by the progranulin gene (GRN) mutation (FTD-GRN), Alzheimer's disease (AD), Parkinson's disease (PD), limbic-predominant age-related transactivation response deoxyribonucleic acid binding protein 43 (TDP-43) encephalopathy (LATE), and amyotrophic lateral sclerosis (ALS). Immuno-neurology targets immune checkpoint-like proteins, offering the potential to convert aging and dysfunctional microglia into disease-fighting cells that counteract multiple disease pathologies, clear misfolded proteins and debris, promote myelin and synapse repair, optimize neuronal function, support astrocytes and oligodendrocytes, and maintain brain vasculature. Several clinical trials are underway to elevate PGRN levels as one strategy to modulate the function of microglia and counteract neurodegenerative changes associated with various disease states. If successful, these and other immuno-neurology drugs have the potential to revolutionize the treatment of neurodegenerative disorders by harnessing the brain's immune system and shifting it from an inflammatory/pathological state to an enhanced physiological/homeostatic state.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Humanos , Progranulinas/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Demência Frontotemporal/genética , Neurônios/patologia
3.
J Transl Med ; 21(1): 387, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37322482

RESUMO

BACKGROUND: Heterozygous loss-of-function mutations in the progranulin (PGRN) gene (GRN) cause a reduction in PGRN and lead to the development of frontotemporal dementia (FTD-GRN). PGRN is a secreted lysosomal chaperone, immune regulator, and neuronal survival factor that is shuttled to the lysosome through multiple receptors, including sortilin. Here, we report the characterization of latozinemab, a human monoclonal antibody that decreases the levels of sortilin, which is expressed on myeloid and neuronal cells and shuttles PGRN to the lysosome for degradation, and blocks its interaction with PGRN. METHODS: In vitro characterization studies were first performed to assess the mechanism of action of latozinemab. After the in vitro studies, a series of in vivo studies were performed to assess the efficacy of a mouse-cross reactive anti-sortilin antibody and the pharmacokinetics, pharmacodynamics, and safety of latozinemab in nonhuman primates and humans. RESULTS: In a mouse model of FTD-GRN, the rodent cross-reactive anti-sortilin antibody, S15JG, decreased total sortilin levels in white blood cell (WBC) lysates, restored PGRN to normal levels in plasma, and rescued a behavioral deficit. In cynomolgus monkeys, latozinemab decreased sortilin levels in WBCs and concomitantly increased plasma and cerebrospinal fluid (CSF) PGRN by 2- to threefold. Finally, in a first-in-human phase 1 clinical trial, a single infusion of latozinemab caused a reduction in WBC sortilin, tripled plasma PGRN and doubled CSF PGRN in healthy volunteers, and restored PGRN to physiological levels in asymptomatic GRN mutation carriers. CONCLUSIONS: These findings support the development of latozinemab for the treatment of FTD-GRN and other neurodegenerative diseases where elevation of PGRN may be beneficial. Trial registration ClinicalTrials.gov, NCT03636204. Registered on 17 August 2018, https://clinicaltrials.gov/ct2/show/NCT03636204 .


Assuntos
Demência Frontotemporal , Humanos , Camundongos , Animais , Progranulinas/genética , Demência Frontotemporal/tratamento farmacológico , Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Mutação/genética
4.
J Immunol ; 210(2): 204-215, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36480261

RESUMO

Antagonizing the CD47-signal regulatory protein (SIRP)α pathway, a critical myeloid checkpoint, promotes antitumor immunity. In this study, we describe the development of AL008, a pan-allelic, SIRPα-specific Ab that triggers the degradation of SIRPα and, concurrently, stimulates FcγR activation of myeloid cells through an engineered Fc domain. AL008 showed superior enhancement of phagocytosis of tumor cells opsonized with antitumor Ag Abs compared with another SIRPα Ab tested. Unlike ligand-blocking SIRPα Abs, AL008 demonstrated single-agent activity by increasing tumor cell engulfment by human monocyte-derived macrophages even in the absence of opsonizing agents. This effect was due to enhanced Fc function, as blocking FcγR2A abrogated AL008-mediated phagocytic activity. AL008 also promoted human monocyte-derived dendritic cell-mediated T cell proliferation. In humanized mouse models, AL008 induced internalization of SIRPα and increased expression of CD86 and HLA-DR on human tumor-associated macrophages, confirming that the mechanism of action is retained in vivo. Monotherapy treatment with AL008 significantly reduced tumor growth in humanized mice implanted with human MDA-MB-231 tumor cells. AL008 also significantly potentiated the effects of T cell checkpoint blockade with anti-programmed death ligand-1 in syngeneic tumor models. This dual and specific mechanism of AL008, to our knowledge, provides a novel therapeutic strategy for targeting myeloid cells for immune activation.


Assuntos
Neoplasias , Receptores Fc , Humanos , Camundongos , Animais , Receptores Fc/metabolismo , Imunoterapia , Fagocitose , Macrófagos , Neoplasias/patologia , Antígenos de Diferenciação , Antígeno CD47/metabolismo
6.
Trends Pharmacol Sci ; 43(8): 641-652, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35039149

RESUMO

Progranulin (PGRN, encoded by the GRN gene) plays a key role in the development, survival, function, and maintenance of neurons and microglia in the mammalian brain. It regulates lysosomal biogenesis, inflammation, repair, stress response, and aging. GRN loss-of-function mutations cause neuronal ceroid lipofuscinosis or frontotemporal dementia-GRN (FTD-GRN) in a gene dosage-dependent manner. Mutations that reduce PGRN levels increase the risk for developing Alzheimer's disease, Parkinson's disease, and limbic-predominant age-related transactivation response DNA-binding protein 43 encephalopathy, as well as exacerbate the progression of amyotrophic lateral sclerosis (ALS) and FTD caused by the hexanucleotide repeat expansion in the C9orf72 gene. Elevating and/or restoring PGRN levels is an attractive therapeutic strategy and is being investigated for neurodegenerative diseases through multiple mechanisms of action.


Assuntos
Demência Frontotemporal , Doenças Neurodegenerativas , Progranulinas , Demência Frontotemporal/tratamento farmacológico , Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Humanos , Microglia , Mutação , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Progranulinas/genética , Progranulinas/metabolismo
7.
Acta Neuropathol ; 140(4): 513-534, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32772264

RESUMO

Multiple sclerosis (MS) is an inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS) triggered by autoimmune mechanisms. Microglia are critical for the clearance of myelin debris in areas of demyelination, a key step to allow remyelination. TREM2 is expressed by microglia and promotes microglial survival, proliferation, and phagocytic activity. Herein we demonstrate that TREM2 was highly expressed on myelin-laden phagocytes in active demyelinating lesions in the CNS of subjects with MS. In gene expression studies, macrophages from subjects with TREM2 genetic deficiency displayed a defect in phagocytic pathways. Treatment with a new TREM2 agonistic antibody promoted the clearance of myelin debris in the cuprizone model of CNS demyelination. Effects included enhancement of myelin uptake and degradation, resulting in accelerated myelin debris removal by microglia. Most importantly, antibody-dependent TREM2 activation on microglia increased density of oligodendrocyte precursors in areas of demyelination, as well as the formation of mature oligodendrocytes thus enhancing remyelination and axonal integrity. These results are relevant as they propose TREM2 on microglia as a potential new target to promote remyelination.


Assuntos
Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Bainha de Mielina/patologia , Receptores Imunológicos/metabolismo , Remielinização/fisiologia , Adulto , Idoso , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Esclerose Múltipla/patologia , Bainha de Mielina/metabolismo , Fagocitose/fisiologia
8.
J Exp Med ; 217(9)2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32579671

RESUMO

TREM2 is a receptor for lipids expressed in microglia. The R47H variant of human TREM2 impairs ligand binding and increases Alzheimer's disease (AD) risk. In mouse models of amyloid ß (Aß) accumulation, defective TREM2 function affects microglial response to Aß plaques, exacerbating tissue damage, whereas TREM2 overexpression attenuates pathology. Thus, AD may benefit from TREM2 activation. Here, we examined the impact of an anti-human TREM2 agonistic mAb, AL002c, in a mouse AD model expressing either the common variant (CV) or the R47H variant of TREM2. Single-cell RNA-seq of microglia after acute systemic administration of AL002c showed induction of proliferation in both CV- and R47H-transgenic mice. Prolonged administration of AL002c reduced filamentous plaques and neurite dystrophy, impacted behavior, and tempered microglial inflammatory response. We further showed that a variant of AL002c is safe and well tolerated in a first-in-human phase I clinical trial and engages TREM2 based on cerebrospinal fluid biomarkers. We conclude that AL002 is a promising candidate for AD therapy.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Glicoproteínas de Membrana/metabolismo , Microglia/patologia , Receptores Imunológicos/metabolismo , Doença de Alzheimer/líquido cefalorraquidiano , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Ansiedade/patologia , Biomarcadores/líquido cefalorraquidiano , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Glicoproteínas de Membrana/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/patologia , Osteopontina/metabolismo , Conformação Proteica , Receptores Imunológicos/imunologia , Transdução de Sinais , Solubilidade
9.
Science ; 352(6286): 712-716, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-27033548

RESUMO

Synapse loss in Alzheimer's disease (AD) correlates with cognitive decline. Involvement of microglia and complement in AD has been attributed to neuroinflammation, prominent late in disease. Here we show in mouse models that complement and microglia mediate synaptic loss early in AD. C1q, the initiating protein of the classical complement cascade, is increased and associated with synapses before overt plaque deposition. Inhibition of C1q, C3, or the microglial complement receptor CR3 reduces the number of phagocytic microglia, as well as the extent of early synapse loss. C1q is necessary for the toxic effects of soluble ß-amyloid (Aß) oligomers on synapses and hippocampal long-term potentiation. Finally, microglia in adult brains engulf synaptic material in a CR3-dependent process when exposed to soluble Aß oligomers. Together, these findings suggest that the complement-dependent pathway and microglia that prune excess synapses in development are inappropriately activated and mediate synapse loss in AD.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Complemento C1q/imunologia , Microglia/imunologia , Fagocitose/imunologia , Sinapses/imunologia , Sinapses/patologia , Peptídeos beta-Amiloides/imunologia , Animais , Região CA1 Hipocampal/imunologia , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Transtornos Cognitivos/imunologia , Transtornos Cognitivos/patologia , Complemento C1q/genética , Via Clássica do Complemento/imunologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases/imunologia , Potenciação de Longa Duração , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Placa Amiloide/imunologia , Sinaptofisina/imunologia , Regulação para Cima
10.
Acta Neuropathol ; 125(6): 829-40, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23677375

RESUMO

Neuromyelitis optica (NMO) is an autoimmune disorder with inflammatory demyelinating lesions in the central nervous system, particularly in the spinal cord and optic nerve. NMO pathogenesis is thought to involve binding of anti-aquaporin-4 (AQP4) autoantibodies to astrocytes, which causes complement-dependent cytotoxicity (CDC) and downstream inflammation leading to oligodendrocyte and neuronal injury. Vasculocentric deposition of activated complement is a prominent feature of NMO pathology. Here, we show that a neutralizing monoclonal antibody against the C1q protein in the classical complement pathway prevents AQP4 autoantibody-dependent CDC in cell cultures and NMO lesions in ex vivo spinal cord slice cultures and in mice. A monoclonal antibody against human C1q with 11 nM binding affinity prevented CDC caused by NMO patient serum in AQP4-transfected cells and primary astrocyte cultures, and prevented complement-dependent cell-mediated cytotoxicity (CDCC) produced by natural killer cells. The anti-C1q antibody prevented astrocyte damage and demyelination in mouse spinal cord slice cultures exposed to AQP4 autoantibody and human complement. In a mouse model of NMO produced by intracerebral injection of AQP4 autoantibody and human complement, the inflammatory demyelinating lesions were greatly reduced by intracerebral administration of the anti-C1q antibody. These results provide proof-of-concept for C1q-targeted monoclonal antibody therapy in NMO. Targeting of C1q inhibits the classical complement pathway directly and causes secondary inhibition of CDCC and the alternative complement pathway. As C1q-targeted therapy leaves the lectin complement activation pathway largely intact, its side-effect profile is predicted to differ from that of therapies targeting downstream complement proteins.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C1q/antagonistas & inibidores , Fatores Imunológicos/uso terapêutico , Neuromielite Óptica/patologia , Neuromielite Óptica/prevenção & controle , Animais , Aquaporina 4/fisiologia , Técnicas de Cultura de Células , Ativação do Complemento , Cricetulus , Modelos Animais de Doenças , Humanos , Camundongos , Neuromielite Óptica/etiologia
11.
J Neuroimmune Pharmacol ; 7(1): 202-14, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22198698

RESUMO

One vexing issue in biomedical research is the failure of many therapies to translate from success in animal models to effective treatment of human disease. One significant difference between the animal models and the human disease is the age of the subject. Cancer, stroke and Alzheimer's occur mainly in humans beyond the 75% mean survival age, while most mouse models use juvenile or young adult animals. Here we compare two mouse models of amyloid deposition, the Tg2576 APP model and the more aggressive APP+PS1 model in which a mutant presenilin1 gene is overexpressed with Tg2576. Middle-aged APP+PS1 mice and aged APP mice have similar degrees of amyloid pathology with a few differences that may partially explain some of the differences between the two age cohorts. The first study evaluated production of microhemorrhage by a monoclonal anti-Aß antibody. We found that in spite of greater amyloid clearance in middle-aged APP+PS1 mice than aged APP mice, the microhemorrhage only developed in old animals. This argues that preclinical studies of immunotherapy in young or middle-aged mice may not predict this potential liability in clinical trials. A second study evaluated the infiltration of systemically injected GFP labeled monocytes into the CNS. Here we find that infiltration is greater in aged mice than middle-aged mice, in spite of greater total Aß staining in the middle-aged animals. We conclude that preclinical studies should be conducted in aged animal models as well as young mice to better prepare for unintended consequences in the human trial.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Modelos Animais de Doenças , Imunoterapia/métodos , Monócitos/imunologia , Fatores Etários , Doença de Alzheimer/imunologia , Amiloide , Animais , Encéfalo/imunologia , Movimento Celular , Ensaio de Imunoadsorção Enzimática , Humanos , Camundongos , Camundongos Transgênicos
12.
J Biomed Inform ; 43(2): 342-53, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19715773

RESUMO

We examine how the biomedical informatics (BMI) community, especially consortia that share data and applications, can take advantage of a new resource called "cloud computing". Clouds generally offer resources on demand. In most clouds, charges are pay per use, based on large farms of inexpensive, dedicated servers, sometimes supporting parallel computing. Substantial economies of scale potentially yield costs much lower than dedicated laboratory systems or even institutional data centers. Overall, even with conservative assumptions, for applications that are not I/O intensive and do not demand a fully mature environment, the numbers suggested that clouds can sometimes provide major improvements, and should be seriously considered for BMI. Methodologically, it was very advantageous to formulate analyses in terms of component technologies; focusing on these specifics enabled us to bypass the cacophony of alternative definitions (e.g., exactly what does a cloud include) and to analyze alternatives that employ some of the component technologies (e.g., an institution's data center). Relative analyses were another great simplifier. Rather than listing the absolute strengths and weaknesses of cloud-based systems (e.g., for security or data preservation), we focus on the changes from a particular starting point, e.g., individual lab systems. We often find a rough parity (in principle), but one needs to examine individual acquisitions--is a loosely managed lab moving to a well managed cloud, or a tightly managed hospital data center moving to a poorly safeguarded cloud?


Assuntos
Redes de Comunicação de Computadores , Sistemas de Gerenciamento de Base de Dados , Armazenamento e Recuperação da Informação/métodos , Internet , Informática Médica , Pesquisa Biomédica , Segurança Computacional , Disseminação de Informação , Software
13.
Cell ; 139(2): 380-92, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19818485

RESUMO

Synapses are asymmetric cellular adhesions that are critical for nervous system development and function, but the mechanisms that induce their formation are not well understood. We have previously identified thrombospondin as an astrocyte-secreted protein that promotes central nervous system (CNS) synaptogenesis. Here, we identify the neuronal thrombospondin receptor involved in CNS synapse formation as alpha2delta-1, the receptor for the anti-epileptic and analgesic drug gabapentin. We show that the VWF-A domain of alpha2delta-1 interacts with the epidermal growth factor-like repeats common to all thrombospondins. alpha2delta-1 overexpression increases synaptogenesis in vitro and in vivo and is required postsynaptically for thrombospondin- and astrocyte-induced synapse formation in vitro. Gabapentin antagonizes thrombospondin binding to alpha2delta-1 and powerfully inhibits excitatory synapse formation in vitro and in vivo. These findings identify alpha2delta-1 as a receptor involved in excitatory synapse formation and suggest that gabapentin may function therapeutically by blocking new synapse formation.


Assuntos
Antígenos CD36/metabolismo , Canais de Cálcio/metabolismo , Neurogênese , Sinapses , Aminas/farmacologia , Animais , Canais de Cálcio Tipo L , Ácidos Cicloexanocarboxílicos/farmacologia , Gabapentina , Camundongos , Plasticidade Neuronal , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Sinapses/efeitos dos fármacos , Ácido gama-Aminobutírico/farmacologia
14.
J Neurosci ; 29(15): 4964-71, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19369565

RESUMO

In amyloid precursor protein (APP) models of amyloid deposition, the amount of amyloid deposits increase with mouse age. At a first approximation, the extent of amyloid accumulation may either reflect small excesses of production over clearance that accumulate over time or, alternatively, indicate a steady-state equilibrium at that age, reflecting the instantaneous excess of production over clearance, which increases as the organism ages. To discriminate between these options, we reversibly suppressed amyloid deposition in Tg2576 mice with the anti-Abeta antibody 2H6, starting at 8 months, just before the first histological deposits can be discerned. Six months later, we stopped the suppression and monitored the progression of amyloid accumulation in control APP mice and suppressed APP mice over the next 3 months. The accumulation hypothesis would predict that the rate of amyloid from 14 to 17 months would be similar in the suppressed and control mice, while the equilibrium hypothesis would predict that the increase would be faster in the suppressed group, possibly catching up completely with the control mice. The results strongly support the accumulation hypothesis, with no evidence of the suppressed mice catching up with the control mice as predicted by equilibrium models. If anything, there was a slower rate of increase in the suppressed APP mice than the control mice, suggesting that a slow seeding mechanism likely precedes a rapid fibrillogenesis in determining the extent of amyloid deposition.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Doença de Alzheimer/prevenção & controle , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/imunologia , Animais , Anticorpos/administração & dosagem , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Drosophila/administração & dosagem , Camundongos , Camundongos Transgênicos , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Fatores de Tempo
15.
J Neuroimmune Pharmacol ; 3(3): 187-97, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18607758

RESUMO

Anti-Abeta antibody administration to amyloid-depositing transgenic mice can reverse amyloid pathology and restore memory function. However, in old mice, these treatments also increase vascular leakage and promote formation of vascular amyloid deposits. Deglycosylated antibodies with reduced affinity for Fcgamma receptors and complement are associated with reduced vascular amyloid and microhemorrhage while retaining amyloid-clearing and memory-enhancing properties of native intact antibodies. In the current experiment, we investigated the effect of 3, 10, or 30 mg/kg of deglycosylated antibody (D-2H6) on amyloid pathology and cognitive behavior in old Tg2576 mice. We found that low doses of deglycosylated antibody appear more efficacious than higher doses in reducing pathology and memory loss in amyloid precursor protein (APP) transgenic mice. These data suggest that excess antibody unbound to antigen can interfere with antibody-mediated Abeta clearance, possibly by saturating the FcRn antibody transporter.


Assuntos
Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Anticorpos/administração & dosagem , Anticorpos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Memória/fisiologia , Envelhecimento/genética , Amiloide/antagonistas & inibidores , Amiloide/metabolismo , Animais , Relação Dose-Resposta Imunológica , Glicosilação , Camundongos , Camundongos Transgênicos
16.
PLoS One ; 3(4): e1900, 2008 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-18382675

RESUMO

Loss of function mutations in the receptor tyrosine kinase TrkB pathway resulted in hyperphagia and morbid obesity in human and rodents. Conversely, peripheral or central stimulation of TrkB by its natural ligands BDNF or NT4 reduced body weight and food intake in mice, supporting the idea that TrkB is a key anorexigenic signal downstream of the melanocortin-4 receptor (Mc4r) system. Here we show that in non-human primates TrkB agonists were anorexigenic when applied centrally, but surprisingly orexigenic, leading to gain in appetite, body weight, fat deposits and serum leptin levels, when given peripherally. The orexigenic and pro-obesity effects of peripherally administered TrkB agonists appear to be dose dependent, not associated with fluid retention nor with evidence of receptor down regulation. Our findings revealed that TrkB signaling exerts dual control on energy homeostasis in the primates that could be targeted for the treatment of either wasting disorders or obesity.


Assuntos
Apetite , Mutação , Receptor trkB/agonistas , Aumento de Peso , Animais , Peso Corporal , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Deleção de Genes , Leptina/sangue , Ligantes , Macaca mulatta , Obesidade/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais
17.
Endocrinology ; 149(3): 1038-48, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18063676

RESUMO

Mutations in the tyrosine kinase receptor trkB or in one of its natural ligands, brain-derived neurotrophic factor (BDNF), lead to severe hyperphagia and obesity in rodents and/or humans. Here, we show that peripheral administration of neurotrophin-4 (NT4), the second natural ligand for trkB, suppresses appetite and body weight in a dose-dependent manner in several murine models of obesity. NT4 treatment increased lipolysis, reduced body fat content and leptin, and elicited long-lasting amelioration of hypertriglyceridemia and hyperglycemia. After treatment termination, body weight gradually recovered to control levels in obese mice with functional leptin receptor. A single intrahypothalamic application of minute amounts of NT4 or an agonist trkB antibody also reduced food intake and body weight in mice. Taken together with the genetic evidence, our findings support the concept that trkB signaling, which originates in the hypothalamus, directly modulates appetite, metabolism, and taste preference downstream of the leptin and melanocortin 4 receptor. The trkB agonists mediate anorexic and weight-reducing effects independent of stress induction, visceral discomfort, or pain sensitization and thus emerge as a potential therapeutic for metabolic disorders.


Assuntos
Fatores de Crescimento Neural/farmacologia , Obesidade/metabolismo , Receptor trkB/agonistas , Animais , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Glucose/metabolismo , Homeostase/efeitos dos fármacos , Leptina/metabolismo , Cloreto de Lítio/farmacologia , Masculino , Melanocortinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Receptores para Leptina/metabolismo , Paladar/efeitos dos fármacos , Triglicerídeos/metabolismo
18.
J Neuroinflammation ; 3: 11, 2006 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-16686956

RESUMO

BACKGROUND: Antibodies against the Ass peptide clear Ass deposits when injected intracranially. Deglycosylated antibodies have reduced effector functions compared to their intact counterparts, potentially avoiding immune activation. METHODS: Deglycosylated or intact C-terminal specific high affinity anti-Abeta antibody (2H6) were intracranially injected into the right frontal cortex and hippocampus of amyloid precursor protein (APP) transgenic mice. The untreated left hemisphere was used to normalize for the extent of amyloid deposition present in each mouse. Control transgenic mice were injected with an antibody against a drosophila-specific protein (amnesiac). Tissues were examined for brain amyloid deposition and microglial responses 3 days after the injection. RESULTS: The deglycosylated 2H6 antibody had lower affinity for several murine Fcgamma receptors and human complement than intact 2H6 without a change in affinity for Ass. Immunohistochemistry for Abeta and thioflavine-S staining revealed that both diffuse and compact deposits were reduced by both antibodies. In animals treated with the intact 2H6 antibody, a significant increase in Fcgamma-receptor II/III immunostaining was observed compared to animals treated with the control IgG antibody. No increase in Fcgamma-receptor II/III was found with the deglycosylated 2H6 antibody. Immunostaining for the microglial activation marker CD45 demonstrated a similar trend. CONCLUSION: These findings suggest that the deglycosylated 2H6 is capable of removing both compact and diffuse plaques without activating microglia. Thus, antibodies with reduced effector functions may clear amyloid without concomitant immune activation when tested as immunotherapy for Alzheimer's disease.

19.
J Neurosci ; 26(20): 5340-6, 2006 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-16707786

RESUMO

Systemic administration of anti-amyloid-beta (Abeta) antibodies results in reduced parenchymal amyloid but increased vascular amyloid and microhemorrhage in amyloid precursor protein (APP) transgenic mice. Here, we evaluate the effects of reducing effector interactions of the antibody via deglycosylation. Mice aged 20 months were treated weekly for 4 months and tested behaviorally before they were killed. APP transgenic mice receiving either anti-Abeta (2H6) or deglycosylated anti-Abeta (de-2H6) showed significant improvement in radial arm water maze performance compared with mice receiving a control antibody. Both groups receiving anti-Abeta antibodies showed significant reductions in total Abeta immunochemistry and Congo red. Significantly fewer vascular amyloid deposits and microhemorrhages were observed in mice administered the de-2H6 antibody compared with those receiving unmodified 2H6 antibody. Deglycosylated anti-Abeta antibodies may be preferable to unmodified IgG because they retain the cognition-enhancing and amyloid-reducing properties of anti-Abeta immunotherapy, while greatly attenuating the increased vascular amyloid deposition and microhemorrhage observed with unmodified IgG.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Anticorpos/farmacologia , Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Placa Amiloide/efeitos dos fármacos , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Anticorpos/uso terapêutico , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Doenças Arteriais Cerebrais/tratamento farmacológico , Doenças Arteriais Cerebrais/fisiopatologia , Doenças Arteriais Cerebrais/prevenção & controle , Transtornos Cognitivos/fisiopatologia , Transtornos Cognitivos/prevenção & controle , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Glicosilação , Imunoterapia/métodos , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Transgênicos , Placa Amiloide/genética , Placa Amiloide/metabolismo , Resultado do Tratamento
20.
Trends Pharmacol Sci ; 27(2): 85-91, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16376998

RESUMO

Nerve growth factor (NGF) was identified originally as a survival factor for sensory and sympathetic neurons in the developing nervous system. In adults, NGF is not required for survival but it has a crucial role in the generation of pain and hyperalgesia in several acute and chronic pain states. The expression of NGF is high in injured and inflamed tissues, and activation of the NGF receptor tyrosine kinase trkA on nociceptive neurons triggers and potentiates pain signalling by multiple mechanisms. Inhibition of NGF function and signalling blocks pain sensation as effectively as cyclooxygenase inhibitors and opiates in rodent models of pain. Several pharmaceutical companies have active drug-discovery and development programs that are based on a variety of approaches to antagonise NGF, including NGF 'capture', blocking the binding of NGF to trkA and inhibiting trkA signalling. NGF antagonism is expected to be a highly effective therapeutic approach in many pain states, and to be free of the adverse effects of traditional analgesic drugs.


Assuntos
Analgésicos/farmacologia , Fatores de Crescimento Neural/antagonistas & inibidores , Dor/fisiopatologia , Animais , Humanos , Hiperalgesia/fisiopatologia , Dor/tratamento farmacológico , Receptor trkA/antagonistas & inibidores , Receptores de Fator de Crescimento Neural/antagonistas & inibidores , Receptores de Fator de Crescimento Neural/efeitos dos fármacos , Receptores de Fator de Crescimento Neural/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...