Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35162951

ABSTRACT

Immune-inflammatory activation impacts extracellular vesicles (EVs), including their miRNA cargo. There is evidence for changes in the EV miRNome in inflammation-associated neuropsychiatric disorders. This mouse study investigated: (1) effects of systemic lipopolysaccharide (LPS) and chronic social stress (CSS) on plasma EV miRNome; and (2) physiological, transcriptional, and behavioural effects of peripheral or central delivered LPS-activated EVs in recipient mice. LPS or CSS effects on the plasma EV miRNome were assessed by using microRNA sequencing. Recipient mice received plasma EVs isolated from LPS-treated or SAL-treated donor mice or vehicle only, either intravenously or into the nucleus accumbens (NAc), on three consecutive days. Bodyweight, spleen or NAc transcriptome and reward (sucrose) motivation were assessed. LPS and CSS increased the expression of 122 and decreased expression of 20 plasma EV miRNAs, respectively. Peripheral LPS-EVs reduced bodyweight, and both LPS-EVs and SAL-EVs increased spleen expression of immune-relevant genes. NAc-infused LPS-EVs increased the expression of 10 immune-inflammatory genes. Whereas motivation increased similarly across test days in all groups, the effect of test days was more pronounced in mice that received peripheral or central LPS-EVs compared with other groups. This study provides causal evidence that increased EV levels impact physiological and behavioural processes and are of potential relevance to neuropsychiatric disorders.


Subject(s)
Extracellular Vesicles/genetics , Gene Expression Profiling/methods , Lipopolysaccharides/adverse effects , MicroRNAs/genetics , Stress, Psychological/genetics , Animals , Extracellular Vesicles/drug effects , Gene Expression Regulation/drug effects , Male , Mice , Organ Size/drug effects , Pilot Projects , Sequence Analysis, RNA , Spleen/drug effects , Spleen/metabolism , Stress, Psychological/psychology
2.
Int J Mol Sci ; 23(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35163583

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system (CNS). Its first clinical presentation (clinically isolated syndrome, CIS) is often followed by the development of relapsing-remitting MS (RRMS). The periphery-to-CNS transmission of inflammatory molecules is a major pathophysiological pathway in MS. This could include signalling via extracellular vesicle (EV) microRNAs (miRNAs). In this study, we investigated the serum EV miRNome in CIS and RRMS patients and matched controls, with the aims to identify MS stage-specific differentially expressed miRNAs and investigate their biomarker potential and pathophysiological relevance. miRNA sequencing was conducted on serum EVs from CIS-remission, RRMS-relapse, and viral inflammatory CNS disorder patients, as well as from healthy and hospitalized controls. Differential expression analysis was conducted, followed by predictive power and target-pathway analysis. A moderate number of dysregulated serum EV miRNAs were identified in CIS-remission and RRMS-relapse patients, especially relative to healthy controls. Some of these miRNAs were also differentially expressed between the two MS stages and had biomarker potential for patient-control and CIS-RRMS separations. For the mRNA targets of the RRMS-relapse-specific EV miRNAs, biological processes inherent to MS pathophysiology were identified using in silico analysis. Study findings demonstrate that specific serum EV miRNAs have MS stage-specific biomarker potential and contribute to the identification of potential targets for novel, efficacious therapies.


Subject(s)
Extracellular Vesicles/metabolism , Gene Expression Profiling , Gene Expression Regulation , MicroRNAs/blood , Multiple Sclerosis, Relapsing-Remitting/blood , Adolescent , Adult , Aged , Biomarkers/blood , Female , Humans , Male , Middle Aged
3.
J Med Chem ; 63(18): 10287-10306, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32787079

ABSTRACT

Despite the broad implications of the cannabinoid type 2 receptor (CB2) in neuroinflammatory processes, a suitable CB2-targeted probe is currently lacking in clinical routine. In this work, we synthesized 15 fluorinated pyridine derivatives and tested their binding affinities toward CB2 and CB1. With a sub-nanomolar affinity (Ki for CB2) of 0.8 nM and a remarkable selectivity factor of >12,000 over CB1, RoSMA-18-d6 exhibited outstanding in vitro performance characteristics and was radiofluorinated with an average radiochemical yield of 10.6 ± 3.8% (n = 16) and molar activities ranging from 52 to 65 GBq/µmol (radiochemical purity > 99%). [18F]RoSMA-18-d6 showed exceptional CB2 attributes as demonstrated by in vitro autoradiography, ex vivo biodistribution, and positron emission tomography (PET). Further, [18F]RoSMA-18-d6 was used to detect CB2 upregulation on postmortem human ALS spinal cord tissues. Overall, these results suggest that [18F]RoSMA-18-d6 is a promising CB2 PET radioligand for clinical translation.


Subject(s)
Pyridines/pharmacology , Radiopharmaceuticals/pharmacology , Receptor, Cannabinoid, CB2/metabolism , Animals , Brain/diagnostic imaging , Fluorine Radioisotopes/chemistry , Humans , Ligands , Male , Molecular Docking Simulation , Molecular Structure , Positron-Emission Tomography , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Rats, Wistar , Spinal Cord/diagnostic imaging , Spleen/diagnostic imaging , Structure-Activity Relationship , Tritium/chemistry
4.
J Med Chem ; 62(24): 11165-11181, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31751140

ABSTRACT

The cannabinoid type 2 (CB2) receptor has emerged as a valuable target for therapy and imaging of immune-mediated pathologies. With the aim to find a suitable radiofluorinated analogue of the previously reported CB2 positron emission tomography (PET) radioligand [11C]RSR-056, 38 fluorinated derivatives were synthesized and tested by in vitro binding assays. With a Ki (hCB2) of 6 nM and a selectivity factor of nearly 700 over cannabinoid type 1 receptors, target compound 3 exhibited optimal in vitro properties and was selected for evaluation as a PET radioligand. [18F]3 was obtained in an average radiochemical yield of 11 ± 4% and molar activities between 33 and 114 GBq/µmol. Specific binding of [18F]3 to CB2 was demonstrated by in vitro autoradiography and in vivo PET experiments using the CB2 ligand GW-405 833. Metabolite analysis revealed only intact [18F]3 in the rat brain. [18F]3 detected CB2 upregulation in human amyotrophic lateral sclerosis spinal cord tissue and may thus become a candidate for diagnostic use in humans.


Subject(s)
Brain/metabolism , Fluorine Radioisotopes/metabolism , Neuroimaging/methods , Positron-Emission Tomography/methods , Pyridines/chemistry , Radiopharmaceuticals/metabolism , Receptor, Cannabinoid, CB2/metabolism , Animals , Brain/diagnostic imaging , Cyclic AMP/metabolism , Fluorine Radioisotopes/chemistry , Hepatocytes/metabolism , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Protein Conformation , Radiochemistry , Radiopharmaceuticals/chemistry , Rats , Rats, Wistar , Receptor, Cannabinoid, CB2/chemistry , Structure-Activity Relationship
5.
Nature ; 566(7745): 543-547, 2019 02.
Article in English | MEDLINE | ID: mdl-30747918

ABSTRACT

Oligodendrocyte pathology is increasingly implicated in neurodegenerative diseases as oligodendrocytes both myelinate and provide metabolic support to axons. In multiple sclerosis (MS), demyelination in the central nervous system thus leads to neurodegeneration, but the severity of MS between patients is very variable. Disability does not correlate well with the extent of demyelination1, which suggests that other factors contribute to this variability. One such factor may be oligodendrocyte heterogeneity. Not all oligodendrocytes are the same-those from the mouse spinal cord inherently produce longer myelin sheaths than those from the cortex2, and single-cell analysis of the mouse central nervous system identified further differences3,4. However, the extent of human oligodendrocyte heterogeneity and its possible contribution to MS pathology remain unknown. Here we performed single-nucleus RNA sequencing from white matter areas of post-mortem human brain from patients with MS and from unaffected controls. We identified subclusters of oligodendroglia in control human white matter, some with similarities to mouse, and defined new markers for these cell states. Notably, some subclusters were underrepresented in MS tissue, whereas others were more prevalent. These differences in mature oligodendrocyte subclusters may indicate different functional states of oligodendrocytes in MS lesions. We found similar changes in normal-appearing white matter, showing that MS is a more diffuse disease than its focal demyelination suggests. Our findings of an altered oligodendroglial heterogeneity in MS may be important for understanding disease progression and developing therapeutic approaches.


Subject(s)
Brain/metabolism , Brain/pathology , Multiple Sclerosis/pathology , Oligodendroglia/pathology , Adult , Aged , Aged, 80 and over , Animals , Autopsy , Biomarkers , Case-Control Studies , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation/genetics , Humans , Male , Mice , Middle Aged , Multiple Sclerosis/genetics , Myelin Sheath/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Oligodendroglia/metabolism , Remyelination/genetics , Sequence Analysis, RNA , Transcription, Genetic , White Matter/cytology , White Matter/metabolism , White Matter/pathology
6.
Brain Behav Immun ; 75: 48-59, 2019 01.
Article in English | MEDLINE | ID: mdl-30218784

ABSTRACT

Maternal immune activation (mIA) in rodents is rapidly emerging as a key model for neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia. Here, we optimise a mIA model in rats, aiming to address certain limitations of current work in this field. Specifically, the lack of clear evidence for methodology chosen, identification of successful induction of mIA in the dams and investigation of male offspring only. We focus on gestational and early juvenile changes in offspring following mIA, as detailed information on these critical early developmental time points is sparse. Following strain (Wistar, Lister Hooded, Sprague Dawley) comparison and selection, and polyriboinosinic-polyribocytidylic acid (poly I:C) dose selection (2.5-15 mg/kg single or once daily for 5 days), mIA was induced in pregnant Wistar rats with 10 mg/kg poly I:C i.p. on gestational day (GD) 15. Early morphometric analysis was conducted in male and female offspring at GD21 and postnatal day (PD) 21, eight dams for each treatment at each time point were used, 32 in total. Subsequent microglia analysis was conducted at PD21 in a small group of offspring. Poly I:C at 10 mg/kg i.p. induced a robust, but variable, plasma IL-6 response 3 h post-injection and reduced body weight at 6 h and 24 h post-injection in two separate cohorts of Wistar rats at GD15. Plasma IL-6 was not elevated at PD21 in offspring or dams. Poly I:C-induced mIA did not affect litter numbers, but resulted in PD21 pup, and GD21 placenta growth restriction. Poly I:C significantly increased microglial activation at PD21 in male hippocampi. We have identified 10 mg/kg poly I:C i.p on GD15 as a robust experimental approach for inducing mIA in Wistar rats and used this to identify early neurodevelopmental changes. This work provides a framework to study the developmental trajectory of disease-relevant, sex-specific phenotypic changes in rats.


Subject(s)
Immunity, Active/physiology , Lymphocyte Activation/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Behavior, Animal/physiology , Cytokines/immunology , Disease Models, Animal , Female , Hippocampus/drug effects , Immunity, Active/immunology , Interleukin-6/metabolism , Lymphocyte Activation/physiology , Male , Models, Animal , Motor Activity/drug effects , Neurodevelopmental Disorders , Placenta/metabolism , Poly I-C/pharmacology , Pregnancy , Rats , Rats, Wistar , Schizophrenia/immunology , T-Lymphocytes/immunology
7.
EMBO J ; 37(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29875132

ABSTRACT

Astrocytes are involved in non-cell-autonomous pathogenic cascades in amyotrophic lateral sclerosis (ALS); however, their role is still debated. We show that astrocytic NF-κB activation drives microglial proliferation and leukocyte infiltration in the SOD1 (G93A) ALS model. This response prolongs the presymptomatic phase, delaying muscle denervation and decreasing disease burden, but turns detrimental in the symptomatic phase, accelerating disease progression. The transition corresponds to a shift in the microglial phenotype showing two effects that can be dissociated by temporally controlling NF-κB activation. While NF-κB activation in astrocytes induced a Wnt-dependent microglial proliferation in the presymptomatic phase with neuroprotective effects on motoneurons, in later stage, astrocyte NF-κB-dependent microglial activation caused an accelerated disease progression. Notably, suppression of the early microglial response by CB2R agonists had acute detrimental effects. These data identify astrocytes as important regulators of microglia expansion and immune response. Therefore, stage-dependent microglia modulation may be an effective therapeutic strategy in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Astrocytes/immunology , NF-kappa B/immunology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/therapy , Animals , Astrocytes/pathology , Disease Models, Animal , Mice , Mice, Transgenic , Microglia/immunology , Microglia/pathology , Motor Neurons/immunology , Motor Neurons/pathology , NF-kappa B/genetics , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/immunology , Superoxide Dismutase/genetics , Superoxide Dismutase/immunology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/immunology
8.
Eur J Neurosci ; 47(12): 1534-1562, 2018 06.
Article in English | MEDLINE | ID: mdl-29862588

ABSTRACT

Aberrant epileptic activity is detectable at early disease stages in Alzheimer's disease (AD) patients and in AD mouse models. Here, we investigated in young ArcticAß mice whether AD-like pathology renders neuronal networks more susceptible to the development of acquired epilepsy induced by unilateral intrahippocampal injection of kainic acid (IHK). In this temporal lobe epilepsy model, IHK induces a status epilepticus followed after two weeks by spontaneous recurrent seizures (SRS). ArcticAß mice exhibited more severe status epilepticus and early onset of SRS. This hyperexcitable phenotype was characterized in CA1 neurons by decreased synaptic strength, increased kainic acid-induced LTP and reduced frequency of spontaneous inhibitory currents. However, no difference in neurodegeneration, neuroinflammation, axonal reorganization or adult neurogenesis was observed in ArcticAß mice compared to wild-type littermates following IHK-induced epileptogenesis. Neuropeptide Y (NPY) expression was reduced at baseline and its IHK-induced elevation in mossy fibres and granule cells was attenuated. However, although this alteration might underlie premature seizure onset, neutralization of soluble Aß species by intracerebroventricular Aß-specific antibody application mitigated the hyperexcitable phenotype of ArcticAß mice and prevented early SRS onset. Therefore, the development of seizures at early stages of AD is mediated primarily by Aß species causing widespread changes in synaptic function.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Epilepsy, Temporal Lobe , Hippocampus , Neuronal Plasticity , Seizures , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Dentate Gyrus/metabolism , Dentate Gyrus/physiopathology , Disease Models, Animal , Electroencephalography , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/physiopathology , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/metabolism , Hippocampus/physiopathology , Kainic Acid/pharmacology , Male , Mice , Mice, Transgenic , Neuronal Plasticity/physiology , Seizures/metabolism , Seizures/physiopathology , Status Epilepticus/metabolism , Status Epilepticus/physiopathology
9.
Tissue Eng Part C Methods ; 24(7): 391-398, 2018 07.
Article in English | MEDLINE | ID: mdl-29897015

ABSTRACT

Triggering receptor expressed on myeloid cells-2 (TREM-2), a transmembrane receptor expressed by macrophages, microglia, and osteoclasts (OCs), plays a protective role in late-onset Alzheimer Disease (AD). To validate TREM-2 as a therapeutic target in AD, its potential secondary parallel effect on bone homeostasis should be clarified. However, animal models and monolayer cultures of human cells were shown poorly predictive of TREM-2 function in human. Therefore, this study aimed to engineer a tridimensional in vitro model using human progenitors differentiated into osteoblasts and OCs, recapitulating physiological bone homeostasis. Human bone marrow-derived mesenchymal cells were seeded and cultured under perfusion inside a collagen type I scaffold for 3 weeks, generating osteoblasts and mineralized matrix. Human peripheral blood-derived CD14+ monocytes were subsequently seeded through the generated tissue, thanks to perfusion flow, and further cultured for up to 3 weeks with an inductive medium, generating mature OCs. This culture system supported collagenous matrix deposition and resorption, allowing for the investigation of kinetic of soluble TREM-2 over the coculture time. Agonistic activation of TREM-2 in this model had no effect on OC activity or on mineralized matrix turnover. In conclusion, the engineered culture system represents a tridimensional, in vitro human bone model for drug testing and suggested no effect of TREM-2 agonist on bone resorption.


Subject(s)
Bone and Bones/cytology , Homeostasis , Membrane Glycoproteins/metabolism , Models, Biological , Osteoclasts/cytology , Receptors, Immunologic/metabolism , Tissue Engineering , Bone and Bones/metabolism , Cells, Cultured , Humans , In Vitro Techniques , Osteoclasts/metabolism
10.
EMBO J ; 36(13): 1837-1853, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28559417

ABSTRACT

Genetic variants in the triggering receptor expressed on myeloid cells 2 (TREM2) increase the risk for several neurodegenerative diseases including Alzheimer's disease and frontotemporal dementia (FTD). Homozygous TREM2 missense mutations, such as p.T66M, lead to the FTD-like syndrome, but how they cause pathology is unknown. Using CRISPR/Cas9 genome editing, we generated a knock-in mouse model for the disease-associated Trem2 p.T66M mutation. Consistent with a loss-of-function mutation, we observe an intracellular accumulation of immature mutant Trem2 and reduced generation of soluble Trem2 similar to patients with the homozygous p.T66M mutation. Trem2 p.T66M knock-in mice show delayed resolution of inflammation upon in vivo lipopolysaccharide stimulation and cultured macrophages display significantly reduced phagocytic activity. Immunohistochemistry together with in vivo TSPO small animal positron emission tomography (µPET) demonstrates an age-dependent reduction in microglial activity. Surprisingly, perfusion magnetic resonance imaging and FDG-µPET imaging reveal a significant reduction in cerebral blood flow and brain glucose metabolism. Thus, we demonstrate that a TREM2 loss-of-function mutation causes brain-wide metabolic alterations pointing toward a possible function of microglia in regulating brain glucose metabolism.


Subject(s)
Brain/pathology , Frontotemporal Dementia/pathology , Glucose/metabolism , Membrane Glycoproteins/genetics , Microglia/physiology , Mutation, Missense , Perfusion , Receptors, Immunologic/genetics , Animals , Disease Models, Animal , Gene Knock-In Techniques , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mutant Proteins/genetics , Positron-Emission Tomography
11.
EMBO Mol Med ; 8(9): 992-1004, 2016 09.
Article in English | MEDLINE | ID: mdl-27402340

ABSTRACT

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


Subject(s)
Amyloid/metabolism , Membrane Glycoproteins/deficiency , Neuroglia/immunology , Phagocytosis , Receptors, Immunologic/deficiency , Animals , Cells, Cultured , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/physiology
12.
J Comp Neurol ; 523(3): 406-30, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25271146

ABSTRACT

The olfactory epithelium (OE) of mice deficient in cystic fibrosis transmembrane conductance regulator (CFTR) exhibits ion transport deficiencies reported in human CF airways, as well as progressive neuronal loss, suggesting defects in olfactory neuron homeostasis. Microvillar cells, a specialized OE cell-subtype, have been implicated in maintaining tissue homeostasis. These cells are endowed with a PLCß2/IP3 R3/TRPC6 signal transduction pathway modulating release of neuropeptide Y (NPY), which stimulates OE stem cell activity. It is unknown, however, whether microvillar cells also mediate the deficits observed in CFTR-null mice. Here we show that Cftr mRNA in mouse OE is exclusively localized in microvillar cells and CFTR immunofluorescence is coassociated with the scaffolding protein NHERF-1 and PLCß2 in microvilli. In CFTR-null mice, PLCß2 was undetectable, NHERF-1 mislocalized, and IP3 R3 more intensely stained, along with increased levels of NPY, suggesting profound alteration of the PLCß2/IP3 R3 signaling pathway. In addition, basal olfactory neuron homeostasis was altered, shown by increased progenitor cell proliferation, differentiation, and apoptosis and by reduced regenerative capacity following methimazole-induced neurodegeneration. The importance of CFTR in microvillar cells was further underscored by decreased thickness of the OE mucus layer and increased numbers of immune cells within this tissue in CFTR-KO mice. Finally, we observed enhanced immune responses to an acute viral-like infection, as well as hyper-responsiveness to chemical and physical stimuli applied intranasally. Taken together, these data strengthen the notion that microvillar cells in the OE play a key role in maintaining tissue homeostasis and identify several mechanisms underlying this regulation through the multiple functions of CFTR.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression Regulation/genetics , Homeostasis/physiology , Neurons/physiology , Olfactory Bulb/cytology , Animals , Antigens, CD , Antithyroid Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodeoxyuridine/metabolism , Caspase 3/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelium/metabolism , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Homeostasis/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ki-67 Antigen/metabolism , Methimazole/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred CFTR , Mice, Transgenic , Neurons/drug effects , Polynucleotides/pharmacology
13.
Nat Rev Neurol ; 10(11): 643-60, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25311587

ABSTRACT

Epidemiological studies have shown a clear association between maternal infection and schizophrenia or autism in the progeny. Animal models have revealed maternal immune activation (mIA) to be a profound risk factor for neurochemical and behavioural abnormalities in the offspring. Microglial priming has been proposed as a major consequence of mIA, and represents a critical link in a causal chain that leads to the wide spectrum of neuronal dysfunctions and behavioural phenotypes observed in the juvenile, adult or aged offspring. Such diversity of phenotypic outcomes in the mIA model are mirrored by recent clinical evidence suggesting that infectious exposure during pregnancy is also associated with epilepsy and, to a lesser extent, cerebral palsy in children. Preclinical research also suggests that mIA might precipitate the development of Alzheimer and Parkinson diseases. Here, we summarize and critically review the emerging evidence that mIA is a shared environmental risk factor across CNS disorders that varies as a function of interactions between genetic and additional environmental factors. We also review ongoing clinical trials targeting immune pathways affected by mIA that may play a part in disease manifestation. In addition, future directions and outstanding questions are discussed, including potential symptomatic, disease-modifying and preventive treatment strategies.


Subject(s)
Brain/embryology , Central Nervous System Diseases/epidemiology , Pregnancy Complications, Infectious/epidemiology , Prenatal Exposure Delayed Effects/epidemiology , Alzheimer Disease/epidemiology , Alzheimer Disease/immunology , Animals , Brain/immunology , Central Nervous System Diseases/immunology , Cerebral Palsy/epidemiology , Cerebral Palsy/immunology , Child Development Disorders, Pervasive/epidemiology , Child Development Disorders, Pervasive/immunology , Female , Humans , Parkinson Disease/epidemiology , Parkinson Disease/immunology , Pregnancy , Pregnancy Complications, Infectious/immunology , Prenatal Exposure Delayed Effects/immunology , Schizophrenia/epidemiology , Schizophrenia/immunology
14.
Acta Neuropathol Commun ; 1: 62, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-24252346

ABSTRACT

For the last 20 years, the "amyloid cascade hypothesis" has dominated research aimed at understanding, preventing, and curing Alzheimer's disease (AD). During that time researchers have acquired an enormous amount of data and have been successful, more than 300 times, in curing the disease in animal model systems by treatments aimed at clearing amyloid deposits. However, to date similar strategies have not been successful in human AD patients. Hence, before rushing into further clinical trials with compounds that aim at lowering amyloid-beta (Aß) levels in increasingly younger people, it would be of highest priority to re-assess the initial assumption that accumulation of Aß in the brain is the primary pathological event driving AD. Here we question this assumption by highlighting experimental evidence in support of the alternative hypothesis suggesting that APP and Aß are part of a neuronal stress/injury system, which is up-regulated to counteract inflammation/oxidative stress-associated neurodegeneration that could be triggered by a brain injury, chronic infections, or a systemic disease. In AD, this protective program may be overridden by genetic and other risk factors, or its maintenance may become dysregulated during aging. Here, we provide a hypothetical example of a hypothesis-driven correlation between car accidents and airbag release in analogy to the evolution of the amyloid focus and as a way to offer a potential explanation for the failure of the AD field to translate the success of amyloid-related therapeutic strategies in experimental models to the clinic.


Subject(s)
Alzheimer Disease/physiopathology , Aging/physiology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Humans , Neurons/physiology
15.
Acta Neuropathol Commun ; 1: 27, 2013 Jun 26.
Article in English | MEDLINE | ID: mdl-24252415

ABSTRACT

BACKGROUND: Reelin and its downstream signaling members are important modulators of actin and microtubule cytoskeleton dynamics, a fundamental prerequisite for proper neurodevelopment and adult neuronal functions. Reductions in Reelin levels have been suggested to contribute to Alzheimer's disease (AD) pathophysiology. We have previously reported an age-related reduction in Reelin levels and its accumulation in neuritic varicosities along the olfactory-limbic tracts, which correlated with cognitive impairments in aged mice. Here, we aimed to investigate whether a similar Reelin-associated neuropathology is observed in the aged human hippocampus and whether it correlated with dementia status. RESULTS: Our immunohistochemical stainings revealed the presence of N- and C-terminus-containing Reelin fragments in corpora amylacea (CAm), aging-associated spherical deposits. The density of these deposits was increased in the molecular layer of the subiculum of AD compared to non-demented individuals. Despite the limitation of a small sample size, our evaluation of several neuronal and glial markers indicates that the presence of Reelin in CAm might be related to aging-associated impairments in neuronal transport leading to accumulation of organelles and protein metabolites in neuritic varicosities, as previously suggested by the findings and discussions in rodents and primates. CONCLUSIONS: Our results indicate that aging- and disease-associated changes in Reelin levels and proteolytic processing might play a role in the formation of CAm by altering cytoskeletal dynamics. However, its presence may also be an indicator of a degenerative state of neuritic compartments.


Subject(s)
Alzheimer Disease/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Serine Endopeptidases/metabolism , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Astrocytes/metabolism , Blotting, Western , Cell Adhesion Molecules, Neuronal/cerebrospinal fluid , Extracellular Matrix Proteins/cerebrospinal fluid , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Microglia/metabolism , Nerve Tissue Proteins/cerebrospinal fluid , Reelin Protein , Serine Endopeptidases/cerebrospinal fluid
16.
PLoS One ; 8(6): e66097, 2013.
Article in English | MEDLINE | ID: mdl-23840405

ABSTRACT

Magnetic resonance imaging (MRI) can be used to monitor pathological changes in Alzheimer's disease (AD). The objective of this longitudinal study was to assess the effects of progressive amyloid-related pathology on multiple MRI parameters in transgenic arcAß mice, a mouse model of cerebral amyloidosis. Diffusion-weighted imaging (DWI), T1-mapping and quantitative susceptibility mapping (QSM), a novel MRI based technique, were applied to monitor structural alterations and changes in tissue composition imposed by the pathology over time. Vascular function and integrity was studied by assessing blood-brain barrier integrity with dynamic contrast-enhanced MRI and cerebral microbleed (CMB) load with susceptibility weighted imaging and QSM. A linear mixed effects model was built for each MRI parameter to incorporate effects within and between groups (i.e. genotype) and to account for changes unrelated to the disease pathology. Linear mixed effects modelling revealed a strong association of all investigated MRI parameters with age. DWI and QSM in addition revealed differences between arcAß and wt mice over time. CMBs became apparent in arcAß mice with 9 month of age; and the CMB load reflected disease stage. This study demonstrates the benefits of linear mixed effects modelling of longitudinal imaging data. Moreover, the diagnostic utility of QSM and assessment of CMB load should be exploited further in studies of AD.


Subject(s)
Brain/blood supply , Cerebral Amyloid Angiopathy/diagnostic imaging , Diffusion Magnetic Resonance Imaging/methods , Animals , Blood-Brain Barrier , Brain/diagnostic imaging , Cerebral Amyloid Angiopathy/genetics , Disease Models, Animal , Linear Models , Longitudinal Studies , Male , Mice , Mice, Transgenic
17.
J Neuroimmune Pharmacol ; 8(1): 79-86, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22580757

ABSTRACT

As research into various aging-associated neurodegenerative disorders reveals their immense pathophysiological complexity, the focus is currently shifting from studying changes in an advanced disease state to investigations involving pre-symptomatic periods, possible aberrations in early life, and even abnormalities in brain development. Recent studies on the etiology of schizophrenia and autism spectrum disorders revealed a profound impact of neurodevelopmental disturbances on disease predisposition, onset and progression. Here, we discuss how a prenatal immune challenge can affect the developing brain-with a selective focus on the impact on microglia, the brain's immune cells-and the implications for brain aging and its associated risk of developing Alzheimer's disease.


Subject(s)
Brain/growth & development , Brain/pathology , Immune System Diseases/pathology , Immune System/physiology , Prenatal Exposure Delayed Effects/immunology , Aging/drug effects , Aging/immunology , Aging/pathology , Animals , Brain/drug effects , Brain/immunology , Female , Humans , Immune System/immunology , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Poly I-C/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/pathology
18.
Nat Rev Neurol ; 9(1): 25-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23183882

ABSTRACT

Despite tremendous investments in understanding the complex molecular mechanisms underlying Alzheimer disease (AD), recent clinical trials have failed to show efficacy. A potential problem underlying these failures is the assumption that the molecular mechanism mediating the genetically determined form of the disease is identical to the one resulting in late-onset AD. Here, we integrate experimental evidence outside the 'spotlight' of the genetic drivers of amyloid-ß (Aß) generation published during the past two decades, and present a mechanistic explanation for the pathophysiological changes that characterize late-onset AD. We propose that chronic inflammatory conditions cause dysregulation of mechanisms to clear misfolded or damaged neuronal proteins that accumulate with age, and concomitantly lead to tau-associated impairments of axonal integrity and transport. Such changes have several neuropathological consequences: focal accumulation of mitochondria, resulting in metabolic impairments; induction of axonal swelling and leakage, followed by destabilization of synaptic contacts; deposition of amyloid precursor protein in swollen neurites, and generation of aggregation-prone peptides; further tau hyperphosphorylation, ultimately resulting in neurofibrillary tangle formation and neuronal death. The proposed sequence of events provides a link between Aß and tau-related neuropathology, and underscores the concept that degenerating neurites represent a cause rather than a consequence of Aß accumulation in late-onset AD.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Aged , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Animals , Brain/pathology , Brain/physiopathology , Cell Death/genetics , Cell Death/physiology , Disease Models, Animal , Disease Progression , Humans , Inflammation Mediators/blood , Mice , Neurites/pathology , Neurites/physiology , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/physiology , Proteostasis Deficiencies/diagnosis , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/pathology , Proteostasis Deficiencies/physiopathology , tau Proteins/genetics
19.
PLoS One ; 7(10): e47793, 2012.
Article in English | MEDLINE | ID: mdl-23082219

ABSTRACT

The extracellular signaling protein Reelin, indispensable for proper neuronal migration and cortical layering during development, is also expressed in the adult brain where it modulates synaptic functions. It has been shown that proteolytic processing of Reelin decreases its signaling activity and promotes Reelin aggregation in vitro, and that proteolytic processing is affected in various neurological disorders, including Alzheimer's disease (AD). However, neither the pathophysiological significance of dysregulated Reelin cleavage, nor the involved proteases and their modulators are known. Here we identified the serine protease tissue plasminogen activator (tPA) and two matrix metalloproteinases, ADAMTS-4 and ADAMTS-5, as Reelin cleaving enzymes. Moreover, we assessed the influence of several endogenous protease inhibitors, including tissue inhibitors of metalloproteinases (TIMPs), α-2-Macroglobulin, and multiple serpins, as well as matrix metalloproteinase 9 (MMP-9) on Reelin cleavage, and described their complex interplay in the regulation of this process. Finally, we could demonstrate that in the murine hippocampus, the expression levels and localization of Reelin proteases largely overlap with that of Reelin. While this pattern remained stable during normal aging, changes in their protein levels coincided with accelerated Reelin aggregation in a mouse model of AD.


Subject(s)
ADAM Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Procollagen N-Endopeptidase/metabolism , Proteolysis , Serine Endopeptidases/metabolism , Tissue Plasminogen Activator/metabolism , ADAMTS4 Protein , ADAMTS5 Protein , Animals , Cell Adhesion Molecules, Neuronal/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/chemistry , HEK293 Cells , HeLa Cells , Hippocampus/cytology , Hippocampus/enzymology , Humans , Immunohistochemistry , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Protein Processing, Post-Translational , Protein Transport , Reelin Protein , Serine Endopeptidases/chemistry
20.
Chem Senses ; 37(9): 859-68, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22952298

ABSTRACT

The main olfactory epithelium consists of 4 major cell types: sensory neurons, supporting cells, microvillar cells, and basal progenitor cells. Several populations of microvillar olfactory cells have been described, whose properties are not yet fully understood. In this study, we aimed to clarify the classification of microvillar cells by introducing a specific marker, CD73. Furthermore, we investigated the turnover of CD73-microvillar cells during adult life. Using direct and indirect immunofluorescence in adult main olfactory epithelium, we first demonstrate that ecto-5'-nucleotidase (CD73) is a reliable marker for microvillar cells reported previously to express phospholipase C ß2 (PLC ß2) along with type 3 IP(3) receptors (IP(3)R3) and transient receptor potential channels 6 (TRPC6), as well as for cells labeled by transgenic expression of tauGFP driven by the IP(3)R3 promoter. The ubiquitous CD73 immunoreactivity in the microvilli of these 2 cell populations indicates that they correspond to the same cell type (CD73-microvillar cell), endowed with a signal transduction cascade mobilizing Ca(++) from intracellular stores. These microvillar cells respond to odors, possess a basal process, and do not degenerate after bulbectomy, suggesting that they contribute to cellular homeostasis in the olfactory epithelium. Next, we examined whether CD73-microvillar cells undergo turnover in the adult olfactory epithelium. By combining CD73 immunofluorescence and BrdU pulse labeling, we show delayed BrdU incorporation in a small fraction of CD73-positive microvillar cells, which persists for several weeks after BrdU administration. These findings indicate that CD73-microvillar cells likely differentiate from proliferating progenitor cells and have a slow turnover despite their apical position in the olfactory epithelium. These combined properties are unique among olfactory cells, in line with the possibility that they might regulate cellular homeostasis driven by extracellular ATP and adenosine.


Subject(s)
5'-Nucleotidase/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Microvilli/metabolism , Olfactory Mucosa/metabolism , Animals , Bromodeoxyuridine/chemistry , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Mice, Transgenic , Phospholipase C beta/metabolism , Promoter Regions, Genetic , Signal Transduction , Smell/physiology , TRPC Cation Channels/metabolism , TRPC6 Cation Channel
SELECTION OF CITATIONS
SEARCH DETAIL
...