Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 111
1.
Brain Commun ; 6(3): fcae143, 2024.
Article En | MEDLINE | ID: mdl-38712323

In preclinical models of multiple sclerosis, systemic inflammation has an impact on the compartmentalized inflammatory process within the central nervous system and results in axonal loss. It remains to be shown whether this is the case in humans, specifically whether systemic inflammation contributes to spinal cord or brain atrophy in multiple sclerosis. Hence, an observational longitudinal study was conducted to delineate the relationship between systemic inflammation and atrophy using magnetic resonance imaging: the SIMS (Systemic Inflammation in Multiple Sclerosis) study. Systemic inflammation and progression were assessed in people with progressive multiple sclerosis (n = 50) over two and a half years. Eligibility criteria included: (i) primary or secondary progressive multiple sclerosis; (ii) age ≤ 70; and (iii) Expanded Disability Status Scale ≤ 6.5. First morning urine was collected weekly to quantify systemic inflammation by measuring the urinary neopterin-to-creatinine ratio using a validated ultra-performance liquid chromatography mass spectrometry technique. The urinary neopterin-to-creatinine ratio temporal profile was characterized by short-term responses overlaid on a background level of inflammation, so these two distinct processes were considered as separate variables: background inflammation and inflammatory response. Participants underwent MRI at the start and end of the study, to measure cervical spinal cord and brain atrophy. Brain and cervical cord atrophy occurred on the study, but the most striking change was seen in the cervical spinal cord, in keeping with the corticospinal tract involvement that is typical of progressive disease. Systemic inflammation predicted cervical cord atrophy. An association with brain atrophy was not observed in this cohort. A time lag between systemic inflammation and cord atrophy was evident, suggesting but not proving causation. The association of the inflammatory response with cord atrophy depended on the level of background inflammation, in keeping with experimental data in preclinical models where the effects of a systemic inflammatory challenge on tissue injury depended on prior exposure to inflammation. A higher inflammatory response was associated with accelerated cord atrophy in the presence of background systemic inflammation below the median for the study population. Higher background inflammation, while associated with cervical cord atrophy itself, subdued the association of the inflammatory response with cord atrophy. Findings were robust to sensitivity analyses adjusting for potential confounders and excluding cases with new lesion formation. In conclusion, systemic inflammation associates with, and precedes, multiple sclerosis progression. Further work is needed to prove causation since targeting systemic inflammation may offer novel treatment strategies for slowing neurodegeneration in multiple sclerosis.

2.
J Neuroinflammation ; 20(1): 92, 2023 Apr 09.
Article En | MEDLINE | ID: mdl-37032328

Neuroinflammation is an important component of many neurodegenerative diseases, whether as a primary cause or a secondary outcome. For that reason, either as diagnostic tools or to monitor progression and/or pharmacological interventions, there is a need for robust biomarkers of neuroinflammation in the brain. Mitochondrial TSPO (18 kDa Translocator protein) is one of few available biomarkers of neuroinflammation for which there are clinically available PET imaging agents. In this study, we further characterised neuroinflammation in a mouse model of prion-induced chronic neurodegeneration (ME7) including a pharmacological intervention via a CSF1R inhibitor. This was achieved by autoradiographic binding of the second-generation TSPO tracer, [3H]PBR28, along with a more comprehensive examination of the cellular contributors to the TSPO signal changes by immunohistochemistry. We observed regional increases of TSPO in the ME7 mouse brains, particularly in the hippocampus, cortex and thalamus. This increased TSPO signal was detected in the cells of microglia/macrophage lineage as well as in astrocytes, endothelial cells and neurons. Importantly, we show that the selective CSF1R inhibitor, JNJ-40346527 (JNJ527), attenuated the disease-dependent increase in TSPO signal, particularly in the dentate gyrus of the hippocampus, where JNJ527 attenuated the number of Iba1+ microglia and neurons, but not GFAP+ astrocytes or endothelial cells. These findings suggest that [3H]PBR28 quantitative autoradiography in combination with immunohistochemistry are important translational tools for detecting and quantifying neuroinflammation, and its treatments, in neurodegenerative disease. Furthermore, we demonstrate that although TSPO overexpression in the ME7 brains was driven by various cell types, the therapeutic effect of the CSF1R inhibitor was primarily to modulate TSPO expression in microglia and neurons, which identifies an important route of biological action of this particular CSF1R inhibitor and provides an example of a cell-specific effect of this type of therapeutic agent on the neuroinflammatory process.


Neurodegenerative Diseases , Prion Diseases , Mice , Animals , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Neuroinflammatory Diseases , Endothelial Cells/metabolism , Receptors, GABA/metabolism , Positron-Emission Tomography/methods , Macrophages/metabolism , Brain/diagnostic imaging , Brain/metabolism , Neurons/metabolism , Prion Diseases/metabolism , Biomarkers/metabolism
3.
Front Immunol ; 11: 579000, 2020.
Article En | MEDLINE | ID: mdl-33162994

The proliferation and activation of microglia, the resident macrophages in the brain, is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and prion disease. Colony stimulating factor 1 receptor (CSF1R) is critically involved in regulating microglial proliferation, and CSF1R blocking strategies have been recently used to modulate microglia in neurodegenerative diseases. However, CSF1R is broadly expressed by many cell types and the impact of its inhibition on the innate immune system is still unclear. CSF1R can be activated by two independent ligands, CSF-1 and interleukin 34 (IL-34). Recently, it has been reported that microglia development and maintenance depend on IL-34 signaling. In this study, we evaluate the inhibition of IL-34 as a novel strategy to reduce microglial proliferation in the ME7 model of prion disease. Selective inhibition of IL-34 showed no effects on peripheral macrophage populations in healthy mice, avoiding the side effects observed after CSF1R inhibition on the systemic compartment. However, we observed a reduction in microglial proliferation after IL-34 inhibition in prion-diseased mice, indicating that microglia could be more specifically targeted by reducing IL-34. Overall, our results highlight the challenges of targeting the CSF1R/IL34 axis in the systemic and central compartments, important for framing any therapeutic effort to tackle microglia/macrophage numbers during brain disease.


Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Brain/drug effects , Cell Proliferation/drug effects , Interleukins/antagonists & inhibitors , Microglia/drug effects , Nerve Degeneration , Prion Diseases/drug therapy , Animals , Antibodies, Monoclonal/toxicity , Antibodies, Neutralizing/toxicity , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Disease Models, Animal , Genes, fms , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interleukins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Prion Diseases/metabolism , Prion Diseases/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction
4.
Lancet Neurol ; 19(10): 872-878, 2020 10.
Article En | MEDLINE | ID: mdl-32949547

Studies in experimental animals show transmissibility of amyloidogenic proteins associated with prion diseases, Alzheimer's disease, Parkinson's disease, and other neurodegenerative diseases. Although these data raise potential concerns for public health, convincing evidence for human iatrogenic transmission only exists for prions and amyloid ß after systemic injections of contaminated growth hormone extracts or dura mater grafts derived from cadavers. Even though these procedures are now obsolete, some reports raise the possibility of iatrogenic transmission of amyloid ß through putatively contaminated neurosurgical equipment. Iatrogenic transmission of amyloid ß might lead to amyloid deposition in the brain parenchyma and blood vessel walls, potentially resulting in cerebral amyloid angiopathy after several decades. Cerebral amyloid angiopathy can cause life-threatening brain haemorrhages; yet, there is no proof that the transmission of amyloid ß can also lead to Alzheimer's dementia. Large, long-term epidemiological studies and sensitive, cost-efficient tools to detect amyloid are needed to better understand any potential routes of amyloid ß transmission and to clarify whether other similar proteopathic seeds, such as tau or α-synuclein, can also be transferred iatrogenically.


Amyloid beta-Peptides/metabolism , Neurodegenerative Diseases/metabolism , Population Surveillance , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Creutzfeldt-Jakob Syndrome/transmission , Humans , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Risk Factors
6.
Lancet Psychiatry ; 7(6): 547-560, 2020 06.
Article En | MEDLINE | ID: mdl-32304649

The coronavirus disease 2019 (COVID-19) pandemic is having a profound effect on all aspects of society, including mental health and physical health. We explore the psychological, social, and neuroscientific effects of COVID-19 and set out the immediate priorities and longer-term strategies for mental health science research. These priorities were informed by surveys of the public and an expert panel convened by the UK Academy of Medical Sciences and the mental health research charity, MQ: Transforming Mental Health, in the first weeks of the pandemic in the UK in March, 2020. We urge UK research funding agencies to work with researchers, people with lived experience, and others to establish a high level coordination group to ensure that these research priorities are addressed, and to allow new ones to be identified over time. The need to maintain high-quality research standards is imperative. International collaboration and a global perspective will be beneficial. An immediate priority is collecting high-quality data on the mental health effects of the COVID-19 pandemic across the whole population and vulnerable groups, and on brain function, cognition, and mental health of patients with COVID-19. There is an urgent need for research to address how mental health consequences for vulnerable groups can be mitigated under pandemic conditions, and on the impact of repeated media consumption and health messaging around COVID-19. Discovery, evaluation, and refinement of mechanistically driven interventions to address the psychological, social, and neuroscientific aspects of the pandemic are required. Rising to this challenge will require integration across disciplines and sectors, and should be done together with people with lived experience. New funding will be required to meet these priorities, and it can be efficiently leveraged by the UK's world-leading infrastructure. This Position Paper provides a strategy that may be both adapted for, and integrated with, research efforts in other countries.


Coronavirus Infections/complications , Coronavirus Infections/psychology , Mental Disorders/complications , Mental Disorders/psychology , Pneumonia, Viral/complications , Pneumonia, Viral/psychology , Research , COVID-19 , Humans , Pandemics
7.
Nat Neurosci ; 22(12): 2111-2116, 2019 12.
Article En | MEDLINE | ID: mdl-31659342

Although genetics highlights the role of microglia in Alzheimer's disease, one-third of putative Alzheimer's disease risk genes lack adequate mouse orthologs. Here we successfully engraft human microglia derived from embryonic stem cells in the mouse brain. The cells recapitulate transcriptionally human primary microglia ex vivo and show expression of human-specific Alzheimer's disease risk genes. Oligomeric amyloid-ß induces a divergent response in human versus mouse microglia. This model can be used to study the role of microglia in neurological diseases.


Alzheimer Disease/genetics , Embryonic Stem Cells/cytology , Microglia/metabolism , Microglia/transplantation , Transcriptome , Amyloid beta-Peptides/pharmacology , Animals , Cell Differentiation , Female , Humans , Male , Mice , Mice, Transgenic , Microglia/drug effects
8.
Brain ; 142(10): 3243-3264, 2019 10 01.
Article En | MEDLINE | ID: mdl-31504240

Neuroinflammation and microglial activation are significant processes in Alzheimer's disease pathology. Recent genome-wide association studies have highlighted multiple immune-related genes in association with Alzheimer's disease, and experimental data have demonstrated microglial proliferation as a significant component of the neuropathology. In this study, we tested the efficacy of the selective CSF1R inhibitor JNJ-40346527 (JNJ-527) in the P301S mouse tauopathy model. We first demonstrated the anti-proliferative effects of JNJ-527 on microglia in the ME7 prion model, and its impact on the inflammatory profile, and provided potential CNS biomarkers for clinical investigation with the compound, including pharmacokinetic/pharmacodynamics and efficacy assessment by TSPO autoradiography and CSF proteomics. Then, we showed for the first time that blockade of microglial proliferation and modification of microglial phenotype leads to an attenuation of tau-induced neurodegeneration and results in functional improvement in P301S mice. Overall, this work strongly supports the potential for inhibition of CSF1R as a target for the treatment of Alzheimer's disease and other tau-mediated neurodegenerative diseases.


Imidazoles/pharmacology , Microglia/drug effects , Pyridines/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Alzheimer Disease/pathology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Genome-Wide Association Study , Humans , Imidazoles/metabolism , Mice , Mice, Transgenic , Microglia/physiology , Neurodegenerative Diseases/drug therapy , Neurogenesis , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Pyridines/metabolism , Receptors, GABA/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Tauopathies/drug therapy , tau Proteins/genetics
9.
Cell Rep ; 27(4): 1293-1306.e6, 2019 04 23.
Article En | MEDLINE | ID: mdl-31018141

Gene expression profiles of more than 10,000 individual microglial cells isolated from cortex and hippocampus of male and female AppNL-G-F mice over time demonstrate that progressive amyloid-ß accumulation accelerates two main activated microglia states that are also present during normal aging. Activated response microglia (ARMs) are composed of specialized subgroups overexpressing MHC type II and putative tissue repair genes (Dkk2, Gpnmb, and Spp1) and are strongly enriched with Alzheimer's disease (AD) risk genes. Microglia from female mice progress faster in this activation trajectory. Similar activated states are also found in a second AD model and in human brain. Apoe, the major genetic risk factor for AD, regulates the ARMs but not the interferon response microglia (IRMs). Thus, the ARMs response is the converging point for aging, sex, and genetic AD risk factors.


Aging/pathology , Alzheimer Disease/pathology , Biomarkers/metabolism , Brain/pathology , Disease Models, Animal , Microglia/pathology , Plaque, Amyloid/pathology , Aging/genetics , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , Biomarkers/analysis , Brain/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Presenilins/physiology , Sex Characteristics
10.
Acta Neuropathol Commun ; 7(1): 25, 2019 02 22.
Article En | MEDLINE | ID: mdl-30795807

The synaptic changes underlying the onset of cognitive impairment in Alzheimer's disease (AD) are poorly understood. In contrast to the well documented inhibition of long-term potentiation (LTP) in CA3-CA1 synapses by acute Aß application in adult neurons from rodents, young amyloid precursor protein (APP) transgenic mouse models often, surprisingly, show normal LTP. This suggests that there may be important differences between mature-onset and developmental-onset APP expression/ Aß accumulation and the ensuing synaptic and behavioural phenotype. Here, in agreement with previous studies, we observed that developmental expression of APPSw,Ind (3-4 month old mice from line 102, PLoS Med 2:e355, 2005), resulted in reduced basal synaptic transmission in CA3-CA1 synapses, normal LTP, impaired spatial working memory, but normal spatial reference memory. To analyse early Aß-mediated synaptic dysfunction and cognitive impairment in a more mature brain, we used controllable mature-onset APPSw,Ind expression in line 102 mice. Within 3 weeks of mature-onset APPSw,Ind expression and Aß accumulation, we detected the first synaptic dysfunction: an impairment of LTP in hippocampal CA3-CA1 synapses. Cognitively, at this time point, we observed a deficit in short-term memory. A reduction in basal synaptic strength and deficit in long-term associative spatial memory were only evident following 12 weeks of APPSw,Ind expression. Importantly, the plasticity impairment observed after 3 weeks of mature-onset APP expression is reversible. Together, these findings demonstrate important differences between developmental and mature-onset APP expression. Further research targeted at this early stage of synaptic dysfunction could help identify mechanisms to treat cognitive impairment in mild cognitive impairment (MCI) and early AD.


Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Cognitive Dysfunction/metabolism , Disease Models, Animal , Synapses/metabolism , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Cognitive Dysfunction/genetics , Female , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Organ Culture Techniques , Synapses/genetics
11.
ChemMedChem ; 13(4): 352-359, 2018 02 20.
Article En | MEDLINE | ID: mdl-29232489

Production of a biocompatible hyperpolarized bolus for signal amplification by reversible exchange (SABRE) could open the door to simple clinical diagnosis via magnetic resonance imaging. Essential to successful progression to preclinical/clinical applications is the determination of the toxicology profile of the SABRE reaction mixture. Herein, we exemplify the cytotoxicity of the SABRE approach using in vitro cell assays. We conclude that the main cause of the observed toxicity is due to the SABRE catalyst. We therefore illustrate two catalyst removal methods: one involving deactivation and ion-exchange chromatography, and the second using biphasic catalysis. These routes produce a bolus suitable for future in vivo study.


Coordination Complexes/toxicity , Iridium/chemistry , Toxicity Tests , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Catalysis , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Humans , Nicotinic Acids/metabolism , Nicotinic Acids/toxicity , Solvents/chemistry
12.
Brain Behav Immun ; 68: 11-16, 2018 02.
Article En | MEDLINE | ID: mdl-29107155

The blood-brain interface (BBI) is the subject of a new named series at Brain, Behavior, and Immunity. It is timely to reflect on a number of advances in the field within the last ten years, which may lead to an increased understanding of human behaviour and a wide range of psychiatric and neurological conditions. We cover discoveries made in solute and cell trafficking, endothelial cell and pericyte biology, extracellular matrix and emerging tools, especially those which will enable study of the human BBI. We now recognize the central role of the BBI in a number of immunopsychiatric syndromes, including sickness behaviour, delirium, septic encephalopathy, cognitive side effects of cytokine-based therapies and the frank psychosis observed in neuronal surface antibody syndromes. In addition, we find ourselves interrogating and modulating the brain across the BBI, during diagnostic investigation and treatment of brain disease. The past ten years of BBI research have been exciting but there is more to come.


Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Animals , Basement Membrane , Brain/immunology , Brain/metabolism , Brain Diseases/immunology , Brain Diseases/metabolism , Endothelial Cells , Extracellular Fluid , Glycocalyx , Humans , Nervous System Diseases , Neuroimmunomodulation/physiology , Neurons , Pericytes
13.
Biol Psychiatry ; 83(1): 70-80, 2018 Jan 01.
Article En | MEDLINE | ID: mdl-28688579

BACKGROUND: Peripheral inflammation is often associated with major depressive disorder (MDD), and immunological biomarkers of depression remain a focus of investigation. METHODS: We used microarray data on whole blood from two independent case-control studies of MDD: the GlaxoSmithKline-High-Throughput Disease-specific target Identification Program [GSK-HiTDiP] study (113 patients and 57 healthy control subjects) and the Janssen-Brain Resource Company study (94 patients and 100 control subjects). Genome-wide differential gene expression analysis (18,863 probes) resulted in a p value for each gene in each study. A Bayesian method identified the largest p-value threshold (q = .025) associated with twice the number of genes differentially expressed in both studies compared with the number of coincidental case-control differences expected by chance. RESULTS: A total of 165 genes were differentially expressed in both studies with concordant direction of fold change. The 90 genes overexpressed (or UP genes) in MDD were significantly enriched for immune response to infection, were concentrated in a module of the gene coexpression network associated with innate immunity, and included clusters of genes with correlated expression in monocytes, monocyte-derived dendritic cells, and neutrophils. In contrast, the 75 genes underexpressed (or DOWN genes) in MDD were associated with the adaptive immune response and included clusters of genes with correlated expression in T cells, natural killer cells, and erythroblasts. Consistently, the MDD patients with overexpression of UP genes also had underexpression of DOWN genes (correlation > .70 in both studies). CONCLUSIONS: MDD was replicably associated with proinflammatory activation of the peripheral innate immune system, coupled with relative inactivation of the adaptive immune system, indicating the potential of transcriptional biomarkers for immunological stratification of patients with depression.


Depressive Disorder, Major/blood , Depressive Disorder, Major/immunology , Immunity, Innate , Biomarkers/blood , Case-Control Studies , Depressive Disorder, Major/genetics , Gene Expression , Gene Expression Regulation , Humans , Immunity, Innate/genetics , Microarray Analysis , Transcriptome , Wernicke Encephalopathy
14.
Cell Rep ; 18(2): 391-405, 2017 01 10.
Article En | MEDLINE | ID: mdl-28076784

Microglia play key roles in brain development, homeostasis, and function, and it is widely assumed that the adult population is long lived and maintained by self-renewal. However, the precise temporal and spatial dynamics of the microglial population are unknown. We show in mice and humans that the turnover of microglia is remarkably fast, allowing the whole population to be renewed several times during a lifetime. The number of microglial cells remains steady from late postnatal stages until aging and is maintained by the spatial and temporal coupling of proliferation and apoptosis, as shown by pulse-chase studies, chronic in vivo imaging of microglia, and the use of mouse models of dysregulated apoptosis. Our results reveal that the microglial population is constantly and rapidly remodeled, expanding our understanding of its role in the maintenance of brain homeostasis.


Aging/physiology , Apoptosis , Brain/cytology , Microglia/cytology , Animals , Cell Count , Cell Proliferation , Gene Expression Profiling , Homeostasis , Humans , Mice , Microglia/metabolism , Monocytes/cytology , Monocytes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors
15.
PLoS Biol ; 14(11): e1002579, 2016 Nov.
Article En | MEDLINE | ID: mdl-27880767

Protein misfolding is common across many neurodegenerative diseases, with misfolded proteins acting as seeds for "prion-like" conversion of normally folded protein to abnormal conformations. A central hypothesis is that misfolded protein accumulation, spread, and distribution are restricted to specific neuronal populations of the central nervous system and thus predict regions of neurodegeneration. We examined this hypothesis using a highly sensitive assay system for detection of misfolded protein seeds in a murine model of prion disease. Misfolded prion protein (PrP) seeds were observed widespread throughout the brain, accumulating in all brain regions examined irrespective of neurodegeneration. Importantly, neither time of exposure nor amount of misfolded protein seeds present determined regions of neurodegeneration. We further demonstrate two distinct microglia responses in prion-infected brains: a novel homeostatic response in all regions and an innate immune response restricted to sites of neurodegeneration. Therefore, accumulation of misfolded prion protein alone does not define targeting of neurodegeneration, which instead results only when misfolded prion protein accompanies a specific innate immune response.


Neurodegenerative Diseases/metabolism , Prion Proteins/metabolism , Animals , Brain/metabolism , Mice , Microglia/metabolism , Up-Regulation
16.
Microbiol Spectr ; 4(3)2016 06.
Article En | MEDLINE | ID: mdl-27337461

The concept of the immunological privilege of the central nervous system (CNS) has had a profound influence on studies of interactions between the immune system and the CNS. At one time there was considerable debate as to whether there were any cells in the CNS of myeloid origin, but we now know that there are a number of populations of myeloid cells in specialized compartments of the CNS and that there is an ongoing bidirectional dialogue between the CNS and the immune system. We briefly review what we know of the different myeloid populations, in particular the microglia: their phenotype and function; their role in CNS homeostasis; and also their role in pathology, focusing on chronic neurodegeneration.


Central Nervous System/immunology , Microglia/physiology , Animals , Central Nervous System Diseases/immunology , Central Nervous System Diseases/pathology , Humans
17.
Sci Rep ; 6: 25663, 2016 05 13.
Article En | MEDLINE | ID: mdl-27174644

Inflammation is a common neuropathological feature in several neurological disorders, including amyotrophic lateral sclerosis (ALS). We have studied the contribution of CSF1R signalling to inflammation in ALS, as a pathway previously reported to control the expansion and activation of microglial cells. We found that microglial cell proliferation in the spinal cord of SOD1(G93A) transgenic mice correlates with the expression of CSF1R and its ligand CSF1. Administration of GW2580, a selective CSF1R inhibitor, reduced microglial cell proliferation in SOD1(G93A) mice, indicating the importance of CSF1-CSF1R signalling in microgliosis in ALS. Moreover, GW2580 treatment slowed disease progression, attenuated motoneuron cell death and extended survival of SOD1(G93A) mice. Electrophysiological assessment revealed that GW2580 treatment protected skeletal muscle from denervation prior to its effects on microglial cells. We found that macrophages invaded the peripheral nerve of ALS mice before CSF1R-induced microgliosis occurred. Interestingly, treatment with GW2580 attenuated the influx of macrophages into the nerve, which was partly caused by the monocytopenia induced by CSF1R inhibition. Overall, our findings provide evidence that CSF1R signalling regulates inflammation in the central and peripheral nervous system in ALS, supporting therapeutic targeting of CSF1R in this disease.


Amyotrophic Lateral Sclerosis/metabolism , Macrophages/metabolism , Microglia/metabolism , Peripheral Nerves/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Anisoles/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Disease Progression , Gliosis/genetics , Gliosis/metabolism , Inflammation/genetics , Inflammation/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Motor Neurons/metabolism , Pyrimidines/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
18.
J Neurosci ; 36(13): 3777-88, 2016 Mar 30.
Article En | MEDLINE | ID: mdl-27030762

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.


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
19.
PLoS One ; 11(3): e0151081, 2016.
Article En | MEDLINE | ID: mdl-26963387

Periodontitis is common in the elderly and may become more common in Alzheimer's disease because of a reduced ability to take care of oral hygiene as the disease progresses. Elevated antibodies to periodontal bacteria are associated with an increased systemic pro-inflammatory state. Elsewhere raised serum pro-inflammatory cytokines have been associated with an increased rate of cognitive decline in Alzheimer's disease. We hypothesized that periodontitis would be associated with increased dementia severity and a more rapid cognitive decline in Alzheimer's disease. We aimed to determine if periodontitis in Alzheimer's disease is associated with both increased dementia severity and cognitive decline, and an increased systemic pro inflammatory state. In a six month observational cohort study 60 community dwelling participants with mild to moderate Alzheimer's Disease were cognitively assessed and a blood sample taken for systemic inflammatory markers. Dental health was assessed by a dental hygienist, blind to cognitive outcomes. All assessments were repeated at six months. The presence of periodontitis at baseline was not related to baseline cognitive state but was associated with a six fold increase in the rate of cognitive decline as assessed by the ADAS-cog over a six month follow up period. Periodontitis at baseline was associated with a relative increase in the pro-inflammatory state over the six month follow up period. Our data showed that periodontitis is associated with an increase in cognitive decline in Alzheimer's Disease, independent to baseline cognitive state, which may be mediated through effects on systemic inflammation.


Alzheimer Disease , Cognition Disorders , Periodontitis , Aged , Alzheimer Disease/complications , Alzheimer Disease/epidemiology , Alzheimer Disease/physiopathology , Cognition Disorders/complications , Cognition Disorders/epidemiology , Cognition Disorders/physiopathology , Female , Follow-Up Studies , Humans , Male , Periodontitis/complications , Periodontitis/epidemiology , Periodontitis/physiopathology , Time Factors
20.
Expert Opin Drug Discov ; 11(4): 355-67, 2016.
Article En | MEDLINE | ID: mdl-26878555

INTRODUCTION: Although many disease models exist for neurodegenerative disease, the translation of basic research findings to clinic is very limited. Studies using freshly resected human brain tissue, commonly discarded from neurosurgical procedures, should complement on-going work using stem cell-derived human neurons and glia thus increasing the likelihood of success in clinical trials. AREAS COVERED: Herein, the authors discuss key issues in the lack of translation from basic research to clinic. They also review the evidence that human neurons, both freshly resected brain tissue and stem cell-derived neurons, such as induced pluripotent stem cells (iPSCs), can be used for analysis of physiological and molecular mechanisms in health and disease. Furthermore, the authors compare and contrast studies using live human brain tissue and studies using induced human stem cell-derived neuron models. Using an example from the area of neurodegeneration, the authors suggest that replicating elements of research findings from animals and stem cell models in resected human brain tissue would strengthen our understanding of disease mechanisms and the therapeutic strategies and aid translation. EXPERT OPINION: The use of human brain tissue alongside iPSC-derived neural models can validate molecular mechanisms identified in rodent disease models and strengthen their relevance to humans. If drug target engagement and mechanism of cellular action can be validated in human brain tissue, this will increase the success rate in clinical research. The combined use of resected human brain tissue, alongside iPSC-derived neural models, could be considered a standard step in pre-clinical research and help to bridge the gap to clinical trials.


Dementia/drug therapy , Drug Discovery/methods , Neurons/metabolism , Animals , Brain/physiopathology , Dementia/physiopathology , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Translational Research, Biomedical/methods
...