Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Front Neurosci ; 15: 760721, 2021.
Article in English | MEDLINE | ID: mdl-35058740

ABSTRACT

Bacterial infections are a common cause of morbidity and mortality in the elderly, and particularly in individuals with a neurodegenerative disease. Experimental models of neurodegeneration have shown that LPS-induced systemic inflammation increases neuronal damage, a process thought to be mediated by activation of "primed" microglia. The effects of a real systemic bacterial infection on the innate immune cells in the brain and neuronal networks are less well described, and therefore, in this study we use the ME7 prion model to investigate the alterations in microglia activation and phenotype and synaptic markers in response to a low grade, live bacterial infection. Mice with or without a pre-existing ME7 prion-induced neurodegenerative disease were given a single systemic injection of live Salmonella typhimurium at early or mid-stage of disease progression. Immune activation markers CD11b and MHCII and pro-inflammatory cytokines were analyzed 4 weeks post-infection. Systemic infection with S. typhimurium resulted in an exaggerated inflammatory response when compared to ME7 prion mice treated with saline. These changes to inflammatory markers were most pronounced at mid-stage disease. Analysis of synaptic markers in ME7 prion mice revealed a significant reduction of genes that are associated with early response in synaptic plasticity, extracellular matrix structure and post-synaptic density, but no further reduction following systemic infection. In contrast, analysis of activity-related neuronal receptors involved in development of learning and memory, such as Grm1 and Grin2a, showed a significant decrease in response to systemic bacterial challenge. These changes were observed early in the disease progression and associated with reduced burrowing activity. The exaggerated innate immune activation and altered expression of genes linked to synaptic plasticity may contribute to the onset and/or progression of neurodegeneration.

2.
J Neurosci ; 36(13): 3777-88, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27030762

ABSTRACT

Epilepsy is a chronic disorder characterized by spontaneous recurrent seizures. Brain inflammation is increasingly recognized as a critical factor for seizure precipitation, but the molecular mediators of such proconvulsant effects are only partly understood. The chemokine CCL2 is one of the most elevated inflammatory mediators in patients with pharmacoresistent epilepsy, but its contribution to seizure generation remains unexplored. Here, we show, for the first time, a crucial role for CCL2 and its receptor CCR2 in seizure control. We imposed a systemic inflammatory challenge via lipopolysaccharide (LPS) administration in mice with mesial temporal lobe epilepsy. We found that LPS dramatically increased seizure frequency and upregulated the expression of many inflammatory proteins, including CCL2. To test the proconvulsant role of CCL2, we administered systemically either a CCL2 transcription inhibitor (bindarit) or a selective antagonist of the CCR2 receptor (RS102895). We found that interference with CCL2 signaling potently suppressed LPS-induced seizures. Intracerebral administration of anti-CCL2 antibodies also abrogated LPS-mediated seizure enhancement in chronically epileptic animals. Our results reveal that CCL2 is a key mediator in the molecular pathways that link peripheral inflammation with neuronal hyperexcitability. SIGNIFICANCE STATEMENT: Substantial evidence points to a role for inflammation in epilepsy, but currently there is little insight as to how inflammatory pathways impact on seizure generation. Here, we examine the molecular mediators linking peripheral inflammation with seizure susceptibility in mice with mesial temporal lobe epilepsy. We show that a systemic inflammatory challenge via lipopolysaccharide administration potently enhances seizure frequency and upregulates the expression of the chemokine CCL2. Remarkably, selective pharmacological interference with CCL2 or its receptor CCR2 suppresses lipopolysaccharide-induced seizure enhancement. Thus, CCL2/CCR2 signaling plays a key role in linking systemic inflammation with seizure susceptibility.


Subject(s)
Chemokine CCL2/metabolism , Epilepsy, Temporal Lobe/complications , Inflammation/etiology , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , Benzoxazines/pharmacology , Benzoxazines/therapeutic use , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Disease Models, Animal , Electroencephalography , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/prevention & control , Excitatory Amino Acid Agonists/toxicity , Hippocampus/pathology , Hippocampus/physiopathology , Indazoles/pharmacology , Kainic Acid/toxicity , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Piperidines/therapeutic use , Propionates/pharmacology , RNA, Messenger/metabolism , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
3.
Neurology ; 84(21): 2161-8, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25934853

ABSTRACT

OBJECTIVES: To determine whether the tumor necrosis factor α inhibitor etanercept is well tolerated and obtain preliminary data on its safety in Alzheimer disease dementia. METHODS: In a double-blind study, patients with mild to moderate Alzheimer disease dementia were randomized (1:1) to subcutaneous etanercept (50 mg) once weekly or identical placebo over a 24-week period. Tolerability and safety of this medication was recorded including secondary outcomes of cognition, global function, behavior, and systemic cytokine levels at baseline, 12 weeks, 24 weeks, and following a 4-week washout period. This trial is registered with EudraCT (2009-013400-31) and ClinicalTrials.gov (NCT01068353). RESULTS: Forty-one participants (mean age 72.4 years; 61% men) were randomized to etanercept (n = 20) or placebo (n = 21). Etanercept was well tolerated; 90% of participants (18/20) completed the study compared with 71% (15/21) in the placebo group. Although infections were more common in the etanercept group, there were no serious adverse events or new safety concerns. While there were some interesting trends that favored etanercept, there were no statistically significant changes in cognition, behavior, or global function. CONCLUSIONS: This study showed that subcutaneous etanercept (50 mg/wk) was well tolerated in this small group of patients with Alzheimer disease dementia, but a larger more heterogeneous group needs to be tested before recommending its use for broader groups of patients. CLASSIFICATION OF EVIDENCE: This study shows Class I evidence that weekly subcutaneous etanercept is well tolerated in Alzheimer disease dementia.


Subject(s)
Alzheimer Disease/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunoglobulin G/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Aged , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Double-Blind Method , Etanercept , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/adverse effects , Injections, Subcutaneous , Male , Receptors, Tumor Necrosis Factor/administration & dosage , Treatment Outcome
4.
Acta Neuropathol Commun ; 1: 48, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-24252464

ABSTRACT

BACKGROUND: Basement membranes in the walls of cerebral capillaries and arteries form a major lymphatic drainage pathway for fluid and solutes from the brain. Amyloid-ß (Aß) draining from the brain is deposited in such perivascular pathways as cerebral amyloid angiopathy (CAA) in Alzheimer's disease (AD). CAA increases in severity when Aß is removed from the brain parenchyma by immunotherapy for AD. In this study we investigated the consequences of immune complexes in artery walls upon drainage of solutes similar to soluble Aß. We tested the hypothesis that, following active immunization with ovalbumin, immune complexes form within the walls of cerebral arteries and impair the perivascular drainage of solutes from the brain. Mice were immunized against ovalbumin and then challenged by intracerebral microinjection of ovalbumin. Perivascular drainage of solutes was quantified following intracerebral microinjection of soluble fluorescent 3kDa dextran into the brain at different time intervals after intracerebral challenge with ovalbumin. RESULTS: Ovalbumin, IgG and complement C3 co-localized in basement membranes of artery walls 24 hrs after challenge with antigen; this was associated with significantly reduced drainage of dextran in immunized mice. CONCLUSIONS: Perivascular drainage along artery walls returned to normal by 7 days. These results indicate that immune complexes form in association with basement membranes of cerebral arteries and interfere transiently with perivascular drainage of solutes from the brain. Immune complexes formed during immunotherapy for AD may similarly impair perivascular drainage of soluble Aß and increase severity of CAA.


Subject(s)
Antigen-Antibody Complex/metabolism , Brain/immunology , Alzheimer Disease/immunology , Alzheimer Disease/therapy , Animals , Basement Membrane/immunology , Brain/blood supply , Cerebral Arteries/immunology , Complement C3/metabolism , Dextrans , Immunoglobulin G/metabolism , Immunotherapy , Mice, Inbred BALB C , Neuroimmunomodulation/physiology , Ovalbumin/immunology , Time Factors , Vaccination
5.
J Neuroinflammation ; 9: 146, 2012 Jun 27.
Article in English | MEDLINE | ID: mdl-22738332

ABSTRACT

BACKGROUND: Systemic infection leads to generation of inflammatory mediators that result in metabolic and behavioural changes. Repeated or chronic systemic inflammation leads to a state of innate immune tolerance: a protective mechanism against overactivity of the immune system. In this study, we investigated the immune adaptation of microglia and brain vascular endothelial cells in response to systemic inflammation or bacterial infection. METHODS: Mice were given repeated doses of lipopolysaccharide (LPS) or a single injection of live Salmonella typhimurium. Inflammatory cytokines were measured in serum, spleen and brain, and microglial phenotype studied by immunohistochemistry. To assess priming of the innate immune response in the brain, mice were infected with Salmonella typhimurium and subsequently challenged with a focal unilateral intracerebral injection of LPS. RESULTS: Repeated systemic LPS challenges resulted in increased brain IL-1ß, TNF-α and IL-12 levels, despite attenuated systemic cytokine production. Each LPS challenge induced significant changes in burrowing behaviour. In contrast, brain IL-1ß and IL-12 levels in Salmonella typhimurium-infected mice increased over three weeks, with high interferon-γ levels in the circulation. Behavioural changes were only observed during the acute phase of the infection. Microglia and cerebral vasculature display an activated phenotype, and focal intracerebral injection of LPS four weeks after infection results in an exaggerated local inflammatory response when compared to non-infected mice. CONCLUSIONS: These studies reveal that the innate immune cells in the brain do not become tolerant to systemic infection, but are primed instead. This may lead to prolonged and damaging cytokine production that may have a profound effect on the onset and/or progression of pre-existing neurodegenerative disease.


Subject(s)
Brain/microbiology , Cerebrovascular Circulation/physiology , Lipopolysaccharides/toxicity , Microglia/microbiology , Salmonella Infections/microbiology , Salmonella typhimurium , Animals , Brain/drug effects , Brain/pathology , Cerebrovascular Circulation/drug effects , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Microglia/drug effects , Microglia/pathology , Salmonella Infections/pathology , Time Factors
6.
Blood ; 115(22): 4393-402, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20194898

ABSTRACT

CD20 is an important target for the treatment of B-cell malignancies, including non-Hodgkin lymphoma as well as autoimmune disorders. B-cell depletion therapy using monoclonal antibodies against CD20, such as rituximab, has revolutionized the treatment of these disorders, greatly improving overall survival in patients. Here, we report the development of GA101 as the first Fc-engineered, type II humanized IgG1 antibody against CD20. Relative to rituximab, GA101 has increased direct and immune effector cell-mediated cytotoxicity and exhibits superior activity in cellular assays and whole blood B-cell depletion assays. In human lymphoma xenograft models, GA101 exhibits superior antitumor activity, resulting in the induction of complete tumor remission and increased overall survival. In nonhuman primates, GA101 demonstrates superior B cell-depleting activity in lymphoid tissue, including in lymph nodes and spleen. Taken together, these results provide compelling evidence for the development of GA101 as a promising new therapy for the treatment of B-cell disorders.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antigens, CD20/immunology , B-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Humans , Immunity, Cellular , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Variable Region/genetics , In Vitro Techniques , Lymphocyte Depletion/methods , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy , Macaca fascicularis , Mice , Mice, SCID , Neoplasm Transplantation , Protein Engineering , Receptors, IgG/immunology , Rituximab , Transplantation, Heterologous
7.
Eur J Immunol ; 39(5): 1323-33, 2009 May.
Article in English | MEDLINE | ID: mdl-19337997

ABSTRACT

Gliomas localized within the CNS are generally not rejected by the immune system despite being immunogenic. This failure of the immune system has been associated both with glioma-derived immunosuppressive molecules and the immune-privileged state of the CNS. However, the relative contribution of tumor location to the glioma-mediated immunosuppression, as well as the immune mechanisms involved in the failure of glioma rejection are not fully defined. We report here that syngeneic GL261 gliomas growing either intracranially or subcutaneously in mice are infiltrated by DC and T cells. However, only subcutaneous gliomas elicit an effective anti-tumor immune response. In contrast to DC infiltrating subcutaneously grown GL261 gliomas, tumor-infiltrating DC from intracranial gliomas do not activate antigen-dependent T-cell proliferation in vitro. In addition, brain-localized GL261 gliomas are characterized by significantly higher numbers of Foxp3(+) Treg and higher levels of TGF-beta1 mRNA and protein expression when compared with GL261 gliomas in the skin. Our data show that gliomas in the CNS, but not in the skin, give rise to TGF-beta production and accumulation of both Treg and functionally impaired DC. Thus, not the tumor itself, but its location dictates the efficiency of the anti-tumor immune response.


Subject(s)
Central Nervous System Neoplasms/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Glioma/immunology , Skin Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Cell Line, Tumor , Flow Cytometry , Forkhead Transcription Factors/biosynthesis , Histocytochemistry , Kaplan-Meier Estimate , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
8.
Biotechnol Bioeng ; 93(5): 851-61, 2006 Apr 05.
Article in English | MEDLINE | ID: mdl-16435400

ABSTRACT

The effector functions elicited by IgG antibodies strongly depend on the carbohydrate moiety linked to the Fc region of the protein. Therefore several approaches have been developed to rationally manipulate these glycans and improve the biological functions of the antibody. Overexpression of recombinant beta1,4-N-acetylglucosaminyltransferase III (GnT-III) in production cell lines leads to antibodies enriched in bisected oligosaccharides. Moreover, GnT-III overexpression leads to increases in non-fucosylated and hybrid oligosaccharides. Such antibody glycovariants have increased antibody-dependent cellular cytotoxicity (ADCC). To explore a further variable besides overexpression of GnT-III, we exchanged the localization domain of GnT-III with that of other Golgi-resident enzymes. Our results indicate that chimeric GnT-III can compete even more efficiently against the endogenous core alpha1,6-fucosyltransferase (alpha1,6-FucT) and Golgi alpha-mannosidase II (ManII) leading to higher proportions of bisected non-fucosylated hybrid glycans ("Glyco-1" antibody). The co-expression of GnT-III and ManII led to a similar degree of non-fucosylation as that obtained for Glyco-1, but the majority of the oligosaccharides linked to this antibody ("Glyco-2") are of the complex type. These glycovariants feature strongly increased ADCC activity compared to the unmodified antibody, while Glyco-1 (hybrid-rich) features reduced complement-dependent cytotoxicity (CDC) compared to Glyco-2 or unmodified antibody. We show that apart from GnT-III overexpression, engineering of GnT-III localization is a versatile tool to modulate the biological activities of antibodies relevant for their therapeutic application.


Subject(s)
Immunoglobulin G/metabolism , N-Acetylgalactosaminyltransferases/genetics , Protein Engineering/methods , Protein Sorting Signals/genetics , Recombinant Fusion Proteins/metabolism , alpha-Mannosidase/genetics , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD20/immunology , B-Lymphocytes/immunology , CHO Cells , Cell Line , Cell Line, Tumor , Complement System Proteins/immunology , Cricetinae , Cricetulus , Cytotoxicity, Immunologic/immunology , Genetic Vectors/genetics , Glycosylation , Golgi Apparatus/enzymology , Humans , Immunoglobulin G/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Oligosaccharides/analysis , Protein Processing, Post-Translational , Receptors, IgG/metabolism , Recombinant Fusion Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...