Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
PLoS One ; 19(6): e0304603, 2024.
Article in English | MEDLINE | ID: mdl-38870196

ABSTRACT

Iatrogenic transmission of prions, the infectious agents of fatal Creutzfeldt-Jakob disease, through inefficiently decontaminated medical instruments remains a critical issue. Harsh chemical treatments are effective, but not suited for routine reprocessing of reusable surgical instruments in medical cleaning and disinfection processes due to material incompatibilities. The identification of mild detergents with activity against prions is therefore of high interest but laborious due to the low throughput of traditional assays measuring prion infectivity. Here, we report the establishment of TESSA (sTainlESs steel-bead Seed Amplification assay), a modified real-time quaking induced cyclic amplification (RT-QuIC) assay that explores the propagation activity of prions with stainless steel beads. TESSA was applied for the screening of about 70 different commercially available and novel formulations and conditions for their prion inactivation efficacy. One hypochlorite-based formulation, two commercially available alkaline formulations and a manual alkaline pre-cleaner were found to be highly effective in inactivating prions under conditions simulating automated washer-disinfector cleaning processes. The efficacy of these formulations was confirmed in vivo in a murine prion infectivity bioassay, yielding a reduction of the prion titer for bead surface adsorbed prions below detectability. Our data suggest that TESSA represents an effective method for a rapid screening of prion-inactivating detergents, and that alkaline and oxidative formulations are promising in reducing the risk of potential iatrogenic prion transmission through insufficiently decontaminated instrument surfaces.


Subject(s)
Prions , Stainless Steel , Surgical Instruments , Animals , Mice , Stainless Steel/chemistry , Decontamination/methods , Creutzfeldt-Jakob Syndrome/transmission , Creutzfeldt-Jakob Syndrome/prevention & control , Disinfection/methods , Detergents/chemistry , Detergents/pharmacology , Humans , Disinfectants/pharmacology , Oxidation-Reduction
2.
Nat Struct Mol Biol ; 29(8): 831-840, 2022 08.
Article in English | MEDLINE | ID: mdl-35948768

ABSTRACT

Prion infections cause conformational changes of the cellular prion protein (PrPC) and lead to progressive neurological impairment. Here we show that toxic, prion-mimetic ligands induce an intramolecular R208-H140 hydrogen bond ('H-latch'), altering the flexibility of the α2-α3 and ß2-α2 loops of PrPC. Expression of a PrP2Cys mutant mimicking the H-latch was constitutively toxic, whereas a PrPR207A mutant unable to form the H-latch conferred resistance to prion infection. High-affinity ligands that prevented H-latch induction repressed prion-related neurodegeneration in organotypic cerebellar cultures. We then selected phage-displayed ligands binding wild-type PrPC, but not PrP2Cys. These binders depopulated H-latched conformers and conferred protection against prion toxicity. Finally, brain-specific expression of an antibody rationally designed to prevent H-latch formation prolonged the life of prion-infected mice despite unhampered prion propagation, confirming that the H-latch is an important reporter of prion neurotoxicity.


Subject(s)
PrPC Proteins , Prions , Animals , Antibodies/metabolism , Cerebellum/metabolism , Ligands , Mice , PrPC Proteins/chemistry , PrPC Proteins/genetics , Prion Proteins/chemistry , Prion Proteins/genetics , Prion Proteins/metabolism , Prions/metabolism , Prions/toxicity
3.
J Fungi (Basel) ; 8(5)2022 May 20.
Article in English | MEDLINE | ID: mdl-35628780

ABSTRACT

In vitro interactions of broad-spectrum azole isavuconazole with flavonoid isoquercitrin were evaluated by a broth microdilution checkerboard technique based on the European Committee on Antimicrobial Susceptibility Testing (EUCAST) reference methodology for antifungal susceptibility testing against 60 Candida strains belonging to the species Candida albicans (n = 10), Candida glabrata (n = 30), Candida kefyr (n = 6), Candida krusei (n = 5), Candida parapsilosis (n = 4), and Candida tropicalis (n = 5). The results were analyzed with the fractional inhibitory concentration index and by response surface analysis based on the Bliss model. Synergy was found for all C. glabrata strains, when the results were interpreted by the fractional inhibitory concentration index, and for 60% of the strains when response surface analysis was used. Interaction for all other species was indifferent for all strains tested, whatever interpretation model used. Importantly, antagonistic interaction was never observed.

4.
Brain Pathol ; 32(5): e13056, 2022 09.
Article in English | MEDLINE | ID: mdl-35178783

ABSTRACT

Although prion infections cause cognitive impairment and neuronal death, transcriptional and translational profiling shows progressive derangement within glia but surprisingly little changes within neurons. Here we expressed PrPC selectively in neurons and astrocytes of mice. After prion infection, both astrocyte and neuron-restricted PrPC expression led to copious brain accumulation of PrPSc . As expected, neuron-restricted expression was associated with typical prion disease. However, mice with astrocyte-restricted PrPC expression experienced a normal life span, did not develop clinical disease, and did not show astro- or microgliosis. Besides confirming that PrPSc is innocuous to PrPC -deficient neurons, these results show that astrocyte-born PrPSc does not activate the extreme neuroinflammation that accompanies the onset of prion disease and precedes any molecular changes of neurons. This points to a nonautonomous mechanism by which prion-infected neurons instruct astrocytes and microglia to acquire a specific cellular state that, in turn, drives neural dysfunction.


Subject(s)
Prion Diseases , Prions , Animals , Astrocytes/metabolism , Mice , Neuroglia/metabolism , Neurons/metabolism , Prion Diseases/metabolism , Prions/metabolism
5.
Nat Commun ; 12(1): 3027, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021139

ABSTRACT

Mutations disrupting the nuclear localization of the RNA-binding protein FUS characterize a subset of amyotrophic lateral sclerosis patients (ALS-FUS). FUS regulates nuclear RNAs, but its role at the synapse is poorly understood. Using super-resolution imaging we determined that the localization of FUS within synapses occurs predominantly near the vesicle reserve pool of presynaptic sites. Using CLIP-seq on synaptoneurosomes, we identified synaptic FUS RNA targets, encoding proteins associated with synapse organization and plasticity. Significant increase of synaptic FUS during early disease in a mouse model of ALS was accompanied by alterations in density and size of GABAergic synapses. mRNAs abnormally accumulated at the synapses of 6-month-old ALS-FUS mice were enriched for FUS targets and correlated with those depicting increased short-term mRNA stability via binding primarily on multiple exonic sites. Our study indicates that synaptic FUS accumulation in early disease leads to synaptic impairment, potentially representing an initial trigger of neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , RNA-Binding Protein FUS/metabolism , RNA/metabolism , Synapses/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Nucleus/metabolism , Cerebral Cortex , Disease Models, Animal , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Protein FUS/genetics
6.
Elife ; 92020 09 22.
Article in English | MEDLINE | ID: mdl-32960170

ABSTRACT

Prion diseases are caused by PrPSc, a self-replicating pathologically misfolded protein that exerts toxicity predominantly in the brain. The administration of PrPSc causes a robust, reproducible and specific disease manifestation. Here, we have applied a combination of translating ribosome affinity purification and ribosome profiling to identify biologically relevant prion-induced changes during disease progression in a cell-type-specific and genome-wide manner. Terminally diseased mice with severe neurological symptoms showed extensive alterations in astrocytes and microglia. Surprisingly, we detected only minor changes in the translational profiles of neurons. Prion-induced alterations in glia overlapped with those identified in other neurodegenerative diseases, suggesting that similar events occur in a broad spectrum of pathologies. Our results suggest that aberrant translation within glia may suffice to cause severe neurological symptoms and may even be the primary driver of prion disease.


Subject(s)
Neuroglia , Neurons/metabolism , Prion Diseases , Ribosomal Proteins , Ribosomes , Animals , Green Fluorescent Proteins , Mice , Mice, Transgenic , Neuroglia/metabolism , Neuroglia/pathology , Prion Diseases/metabolism , Prion Diseases/pathology , Recombinant Fusion Proteins , Ribosomal Proteins/analysis , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/chemistry , Ribosomes/metabolism
7.
PLoS Pathog ; 16(6): e1008653, 2020 06.
Article in English | MEDLINE | ID: mdl-32598380

ABSTRACT

The clinical course of prion diseases is accurately predictable despite long latency periods, suggesting that prion pathogenesis is driven by precisely timed molecular events. We constructed a searchable genome-wide atlas of mRNA abundance and splicing alterations during the course of disease in prion-inoculated mice. Prion infection induced PrP-dependent transient changes in mRNA abundance and processing already at eight weeks post inoculation, well ahead of any neuropathological and clinical signs. In contrast, microglia-enriched genes displayed an increase simultaneous with the appearance of clinical signs, whereas neuronal-enriched transcripts remained unchanged until the very terminal stage of disease. This suggests that glial pathophysiology, rather than neuronal demise, could be the final driver of disease. The administration of young plasma attenuated the occurrence of early mRNA abundance alterations and delayed signs in the terminal phase of the disease. The early onset of prion-induced molecular changes might thus point to novel biomarkers and potential interventional targets.


Subject(s)
Genome-Wide Association Study , Microglia/metabolism , Neurons/metabolism , Prion Diseases , RNA, Messenger , Transcriptome , Animals , Male , Mice , Mice, Knockout , Prion Diseases/genetics , Prion Diseases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
PLoS One ; 14(9): e0216013, 2019.
Article in English | MEDLINE | ID: mdl-31513666

ABSTRACT

Prions cause transmissible infectious diseases in humans and animals and have been found to be transmissible by blood transfusion even in the presymptomatic stage. However, the concentration of prions in body fluids such as blood and urine is extremely low; therefore, direct diagnostic tests on such specimens often yield false-negative results. Quantitative preanalytical prion enrichment may significantly improve the sensitivity of prion assays by concentrating trace amounts of prions from large volumes of body fluids. Here, we show that beads conjugated to positively charged peptoids not only captured PrP aggregates from plasma of prion-infected hamsters, but also adsorbed prion infectivity in both the symptomatic and preclinical stages of the disease. Bead absorbed prion infectivity efficiently transmitted disease to transgenic indicator mice. We found that the readout of the peptoid-based misfolded protein assay (MPA) correlates closely with prion infectivity in vivo, thereby validating the MPA as a simple, quantitative, and sensitive surrogate indicator of the presence of prions. The reliable and sensitive detection of prions in plasma will enable a wide variety of applications in basic prion research and diagnostics.


Subject(s)
Biological Assay/methods , Microspheres , Peptoids , Prion Diseases/blood , Prion Diseases/diagnosis , Prions/blood , Animals , Biomarkers , Cerebral Cortex/metabolism , Cricetinae , Disease Models, Animal , Mice , Peptoids/chemistry , Prion Diseases/mortality , Sensitivity and Specificity
9.
Sci Transl Med ; 11(495)2019 06 05.
Article in English | MEDLINE | ID: mdl-31167929

ABSTRACT

Parkinson's disease (PD) is a neurological disorder characterized by the progressive accumulation of neuronal α-synuclein (αSyn) inclusions called Lewy bodies. It is believed that Lewy bodies spread throughout the nervous system due to the cell-to-cell propagation of αSyn via cycles of secretion and uptake. Here, we investigated the internalization and intracellular accumulation of exogenous αSyn, two key steps of Lewy body pathogenesis, amplification and spreading. We found that stable αSyn fibrils substantially accumulate in different cell lines upon internalization, whereas αSyn monomers, oligomers, and dissociable fibrils do not. Our data indicate that the uptake-mediated accumulation of αSyn in a human-derived neuroblastoma cell line triggered an adaptive response that involved proteins linked to ubiquitin ligases of the S-phase kinase-associated protein 1 (SKP1), cullin-1 (Cul1), and F-box domain-containing protein (SCF) family. We found that SKP1, Cul1, and the F-box/LRR repeat protein 5 (FBXL5) colocalized and physically interacted with internalized αSyn in cultured cells. Moreover, the SCF containing the F-box protein FBXL5 (SCFFBXL5) catalyzed αSyn ubiquitination in reconstitution experiments in vitro using recombinant proteins and in cultured cells. In the human brain, SKP1 and Cul1 were recruited into Lewy bodies from brainstem and neocortex of patients with PD and related neurological disorders. In both transgenic and nontransgenic mice, intracerebral administration of exogenous αSyn fibrils triggered a Lewy body-like pathology, which was amplified by SKP1 or FBXL5 loss of function. Our data thus indicate that SCFFXBL5 regulates αSyn in vivo and that SCF ligases may constitute targets for the treatment of PD and other α-synucleinopathies.


Subject(s)
Lewy Bodies/metabolism , Lewy Bodies/pathology , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/metabolism , Animals , Benzothiazoles/metabolism , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Humans , Mice , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Proteome/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin/metabolism
10.
J Antimicrob Chemother ; 74(7): 1921-1927, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30934052

ABSTRACT

OBJECTIVES: To evaluate the in vitro interactions of isavuconazole with immune suppressors (tacrolimus, cyclosporin A or sirolimus) against 30 Mucorales isolates belonging to the most common species responsible for mucormycosis in humans (Rhizopus arrhizus, Rhizopus delemar, Rhizopus microsporus, Lichtheimia corymbifera, Lichtheimia ramosa, Mucor circinelloides and Rhizomucor pusillus). METHODS: In vitro interaction was evaluated by a microdilution chequerboard technique. RESULTS: Combination of isavuconazole with tacrolimus, cyclosporin A or sirolimus, was synergistic for 50%, 46% and 7% of the isolates, respectively. Antagonistic interaction was observed for 4% of the isolates for the combination with cyclosporin A (one R. arrhizus isolate) and for 32% of the isolates for the combination with sirolimus (six R. arrhizus isolates and three R. pusillus isolates). CONCLUSIONS: These in vitro data show that calcineurin inhibitors are more likely than inhibitors of the mTOR pathway to enhance the activity of isavuconazole against Mucorales. These in vitro results warrant further animal experiments.


Subject(s)
Antifungal Agents/pharmacology , Cyclosporine/pharmacology , Mucorales/drug effects , Nitriles/pharmacology , Pyridines/pharmacology , Sirolimus/pharmacology , Tacrolimus/pharmacology , Triazoles/pharmacology , Immunosuppressive Agents/pharmacology , Microbial Sensitivity Tests , Mucormycosis/drug therapy , Mucormycosis/microbiology
11.
BMC Health Serv Res ; 19(1): 158, 2019 Mar 12.
Article in English | MEDLINE | ID: mdl-30866908

ABSTRACT

BACKGROUND: The medical discharge letter is an important communication tool between hospitals and other healthcare providers. Despite its high status, it often does not meet the desired requirements in everyday clinical practice. Occurring risks create barriers for patients and doctors. This present review summarizes risks of the medical discharge letter. METHODS: The research question was answered with a systematic literature research and results were summarized narratively. A literature search in the databases PubMed and Cochrane Library for Studies between January 2008 and May 2018 was performed. Two authors reviewed the full texts of potentially relevant studies to determine eligibility for inclusion. Literature on possible risks associated with the medical discharge letter was discussed. RESULTS: In total, 29 studies were included in this review. The major identified risk factors are the delayed sending of the discharge letter to doctors for further treatments, unintelligible (not patient-centered) medical discharge letters, low quality of the discharge letter, and lack of information as well as absence of training in writing medical discharge letters during medical education. CONCLUSIONS: Multiple risks factors are associated with the medical discharge letter. There is a need for further research to improve the quality of the medical discharge letter to minimize risks and increase patients' safety.


Subject(s)
Medical Records/standards , Patient Discharge/standards , Patient Safety/standards , Communication , Health Personnel , Hospitals , Humans , Narration , Professional Practice/standards
13.
Plant Biotechnol J ; 16(5): 1046-1056, 2018 05.
Article in English | MEDLINE | ID: mdl-29024288

ABSTRACT

The Qfhs.ifa-5A allele, contributing to enhanced Fusarium head blight resistance in wheat, resides in a low-recombinogenic region of chromosome 5A close to the centromere. A near-isogenic RIL population segregating for the Qfhs.ifa-5A resistance allele was developed and among 3650 lines as few as four recombined within the pericentromeric C-5AS1-0.40 bin, yielding only a single recombination point. Genetic mapping of the pericentromeric region using a recombination-dependent approach was thus not successful. To facilitate fine-mapping the physically large Qfhs.ifa-5A interval, two gamma-irradiated deletion panels were generated: (i) seeds of line NIL3 carrying the Qfhs.ifa-5A resistance allele in an otherwise susceptible background were irradiated and plants thereof were selfed to obtain deletions in homozygous state and (ii) a radiation hybrid panel was produced using irradiated pollen of the wheat line Chinese Spring (CS) for pollinating the CS-nullisomic5Atetrasomic5B. In total, 5157 radiation selfing and 276 radiation hybrid plants were screened for deletions on 5AS and plants containing deletions were analysed using 102 5AS-specific markers. Combining genotypic information of both panels yielded an 817-fold map improvement (cR/cM) for the centromeric bin and was 389-fold increased across the Qfhs.ifa-5A interval compared to the genetic map, with an average map resolution of 0.77 Mb/cR. We successfully proved that the RH mapping technique can effectively resolve marker order in low-recombining regions, including pericentromeric intervals, and simultaneously allow developing an in vivo panel of sister lines differing for induced deletions across the Qfhs.ifa-5A interval that can be used for phenotyping.


Subject(s)
Chromosomes, Plant/genetics , Disease Resistance/genetics , Fusarium/physiology , Plant Diseases/immunology , Quantitative Trait Loci/genetics , Triticum/genetics , Chromosome Mapping , Plant Diseases/microbiology , Triticum/immunology , Triticum/microbiology
14.
PLoS One ; 12(2): e0170503, 2017.
Article in English | MEDLINE | ID: mdl-28207746

ABSTRACT

Resistance to proteolytic digestion has long been considered a defining trait of prions in tissues of organisms suffering from transmissible spongiform encephalopathies. Detection of proteinase K-resistant prion protein (PrPSc) still represents the diagnostic gold standard for prion diseases in humans, sheep and cattle. However, it has become increasingly apparent that the accumulation of PrPSc does not always accompany prion infections: high titers of prion infectivity can be reached also in the absence of protease resistant PrPSc. Here, we describe a structural basis for the phenomenon of protease-sensitive prion infectivity. We studied the effect on proteinase K (PK) resistance of the amino acid substitution Y169F, which removes a single oxygen atom from the ß2-α2 loop of the cellular prion protein (PrPC). When infected with RML or the 263K strain of prions, transgenic mice lacking wild-type (wt) PrPC but expressing MoPrP169F generated prion infectivity at levels comparable to wt mice. The newly generated MoPrP169F prions were biologically indistinguishable from those recovered from prion-infected wt mice, and elicited similar pathologies in vivo. Surprisingly, MoPrP169F prions showed greatly reduced PK resistance and density gradient analyses showed a significant reduction in high-density aggregates. Passage of MoPrP169F prions into mice expressing wt MoPrP led to full recovery of protease resistance, indicating that no strain shift had taken place. We conclude that a subtle structural variation in the ß2-α2 loop of PrPC affects the sensitivity of PrPSc to protease but does not impact prion replication and infectivity. With these findings a specific structural feature of PrPC can be linked to a physicochemical property of the corresponding PrPSc.


Subject(s)
Brain/metabolism , Endopeptidase K/metabolism , PrPSc Proteins/metabolism , Prion Diseases/transmission , Prions/pathogenicity , Amino Acid Substitution , Animals , Brain/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Mutation/genetics , PrPSc Proteins/chemistry , PrPSc Proteins/genetics , Prion Diseases/pathology , Protein Conformation , Proteolysis
15.
PLoS One ; 11(9): e0163601, 2016.
Article in English | MEDLINE | ID: mdl-27684562

ABSTRACT

Prions are the infectious agents causing transmissible spongiform encephalopathies (TSE), progressive, inexorably lethal neurological diseases. Antibodies targeting the globular domain (GD) of the cellular prion protein PrPC trigger a neurotoxic syndrome morphologically and molecularly similar to prion disease. This phenomenon raises the question whether such antibodies induce infectious prions de novo. Here we exposed cerebellar organotypic cultured slices (COCS) to the neurotoxic antibody, POM1. We then inoculated COCS homogenates into tga20 mice, which overexpress PrPC and are commonly utilized as sensitive indicators of prion infectivity. None of the mice inoculated with COCS-derived lysates developed any signs of disease, and all mice survived for at least 200 days post-inoculation. In contrast, all mice inoculated with bona fide prions succumbed to TSE after 55-95 days. Post-mortem analyses did not reveal any signs of prion pathology in mice inoculated with POM1-COCS lysates. Also, lysates from POM1-exposed COCS were unable to convert PrP by quaking. Hence, anti-GD antibodies do not catalyze the generation of prion infectivity. These data indicate that prion replication can be separated from prion toxicity, and suggest that anti-GD antibodies exert toxicity by acting downstream of prion replication.

16.
J Exp Med ; 213(6): 1047-59, 2016 05 30.
Article in English | MEDLINE | ID: mdl-27185853

ABSTRACT

Microglial activation is a hallmark of most neurodegenerative disorders, and is particularly conspicuous in prion diseases. However, the role of microglia, which function as both primary immune effector cells and professional phagocytes in the central nervous system, remains contentious in the context of neurodegeneration. Here, we evaluated the effect of microglial depletion/deficiency on prion pathogenesis. We found that ganciclovir-mediated microglial ablation on tga20/CD11b-thymidine kinase of Herpes simplex virus (HSVTK) cerebellar organotypic cultured slices markedly aggravated prion-induced neurotoxicity. A similar deterioration of disease was recapitulated in in vivo microglial depletion in prion-infected tga20/CD11b-HSVTK mice. Additionally, deficiency of microglia in interleukin 34 knockout (IL34(-/-)) mice again resulted in significantly augmented proteinase K-resistant prion protein deposition and accelerated prion disease progression. These results provide unambiguous evidence for a general protective role of microglia in prion pathogenesis.


Subject(s)
Interleukins/metabolism , Microglia/metabolism , Prion Diseases/metabolism , Prions/metabolism , Animals , Interleukins/genetics , Mice , Mice, Knockout , Microglia/pathology , Prion Diseases/genetics , Prion Diseases/pathology , Prions/genetics
17.
J Exp Med ; 213(3): 313-27, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26926995

ABSTRACT

Although its involvement in prion replication and neurotoxicity during transmissible spongiform encephalopathies is undisputed, the physiological role of the cellular prion protein (PrP(C)) remains enigmatic. A plethora of functions have been ascribed to PrP(C) based on phenotypes of Prnp(-/-) mice. However, all currently available Prnp(-/-) lines were generated in embryonic stem cells from the 129 strain of the laboratory mouse and mostly crossed to non-129 strains. Therefore, Prnp-linked loci polymorphic between 129 and the backcrossing strain resulted in systematic genetic confounders and led to erroneous conclusions. We used TALEN-mediated genome editing in fertilized mouse oocytes to create the Zurich-3 (ZH3) Prnp-ablated allele on a pure C57BL/6J genetic background. Genomic, transcriptional, and phenotypic characterization of Prnp(ZH3/ZH3) mice failed to identify phenotypes previously described in non-co-isogenic Prnp(-/-) mice. However, aged Prnp(ZH3/ZH3) mice developed a chronic demyelinating peripheral neuropathy, confirming the crucial involvement of PrP(C) in peripheral myelin maintenance. This new line represents a rigorous genetic resource for studying the role of PrP(C) in physiology and disease.


Subject(s)
Prions/metabolism , Animals , Base Sequence , Chromosomes, Mammalian , Endonucleases/metabolism , Female , Gene Deletion , Macrophages/metabolism , Male , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Degeneration/pathology , Open Reading Frames/genetics , Phagocytosis , Phenotype , Polyradiculoneuropathy/pathology , RNA/metabolism , Sequence Analysis, RNA , Trans-Activators/genetics
19.
PLoS One ; 10(12): e0144983, 2015.
Article in English | MEDLINE | ID: mdl-26658276

ABSTRACT

Epidemiological, clinical, and experimental animal studies suggest a strong correlation between insulin resistance and Alzheimer's disease. In fact, type-2 diabetes is considered an important risk factor of developing Alzheimer's disease. In addition, impaired insulin signaling in the Alzheimer's disease brain may promote Aß production, impair Aß clearance and induce tau hyperphosphorylation, thereby leading to deterioration of the disease. The pathological prion protein, PrPSc, deposits in the form of extracellular aggregates and leads to dementia, raising the question as to whether prion pathogenesis may also be affected by insulin resistance. We therefore established high-fat diet-induced insulin resistance in tga20 mice, which overexpress the prion protein. We then inoculated the insulin-resistant mice with prions. We found that insulin resistance in tga20 mice did not affect prion disease progression, PrPSc deposition, astrogliosis or microglial activation, and had no effect on survival. Our study demonstrates that in a mouse model, insulin resistance does not significantly contribute to prion pathogenesis.


Subject(s)
Amyloid beta-Peptides/metabolism , Insulin Resistance/physiology , Prion Diseases/metabolism , Prions/pathogenicity , Alzheimer Disease/metabolism , Animals , Brain/cytology , Brain/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Insulin/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prions/metabolism
20.
Neurobiol Aging ; 36(5): 1994-2003, 2015 May.
Article in English | MEDLINE | ID: mdl-25816748

ABSTRACT

Dysfunctional variants of the innate immune cell surface receptor TREM2 (triggering receptor expressed on myeloid cells-2) were identified as major genetic risk factors for Alzheimer's disease and other neurodegenerative conditions. Here we assessed a possible involvement of TREM2 in prion disease. We report that TREM2 expression by microglia is significantly up-regulated upon prion infection. However, depletion of TREM2 did not affect disease incubation time and survival after intracerebral prion infection. Interestingly, markers of microglial activation were attenuated in prion-infected TREM2(-/-) mice, suggesting an involvement of TREM2 in prion-induced microglial activation. Further phenotype profiling of microglia revealed that TREM2 deficiency did not change microglial phenotypes. We conclude that TREM2 is involved in prion-induced microglial activation but does not noticeably modulate the pathogenesis of experimental prion infections.


Subject(s)
Gene Expression/genetics , Membrane Glycoproteins/physiology , Microglia/metabolism , Microglia/pathology , Prion Diseases/genetics , Receptors, Immunologic/physiology , Up-Regulation/genetics , Animals , Mice, Transgenic , Phenotype , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...