Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(11): e0293301, 2023.
Article in English | MEDLINE | ID: mdl-37910561

ABSTRACT

Prion diseases are caused by the misfolding of a normal host protein that leads to gliosis, neuroinflammation, neurodegeneration, and death. Microglia have been shown to be critical for neuroprotection during prion infection of the central nervous system (CNS), and their presence extends survival in mice. How microglia impart these benefits to the infected host are unknown. Previous transcriptomics and bioinformatics studies suggested that signaling through the heterodimeric integrin receptor CD11c/CD18, expressed by microglia in the brain, might be important to microglial function during prion disease. Herein, we intracerebrally challenged CD11c-/- mice with prion strain RML and compared them to similarly infected C57BL/6 mice as controls. We initially assessed changes in the brain that are associated with disease such as astrogliosis, microgliosis, prion accumulation, and survival. Targeted qRT-PCR arrays were used to determine alterations in transcription in mice in response to prion infection. We demonstrate that expression of Itgax (CD11c) and Itgb2 (CD18) increases in the CNS in correlation with advancing prion infection. Gliosis, neuropathology, prion deposition, and disease progression in prion infected CD11c deficient mice were comparable to infected C57BL/6 mice. Additionally, both CD11c deficient and C57BL/6 prion-infected mouse cohorts had a similar consortium of inflammatory- and phagocytosis-associated genes that increased as disease progressed to clinical stages. Ingenuity Pathway Analysis of upregulated genes in infected C57BL/6 mice suggested numerous cell-surface transmembrane receptors signal through Spleen Tyrosine Kinase, a potential key regulator of phagocytosis and innate immune activation in the prion infected brain. Ultimately, the deletion of CD11c did not influence prion pathogenesis in mice and CD11c signaling is not involved in the neuroprotection provided by microglia, but our analysis identified a conspicuous phagocytosis pathway in the CNS of infected mice that appeared to be activated during prion pathogenesis.


Subject(s)
Prion Diseases , Prions , Animals , Mice , Prions/metabolism , Microglia/metabolism , Gliosis/pathology , Neuroprotection , Mice, Inbred C57BL , Prion Diseases/metabolism , Brain/metabolism
2.
PLoS One ; 18(8): e0290325, 2023.
Article in English | MEDLINE | ID: mdl-37616303

ABSTRACT

Prion diseases are transmissible, fatal neurologic diseases that include Creutzfeldt-Jakob Disease (CJD) in humans, chronic wasting disease (CWD) in cervids, bovine spongiform encephalopathy (BSE) in cattle and scrapie in sheep. Prions are extremely difficult to inactivate and established methods to reduce prion infectivity are often dangerous, caustic, expensive, or impractical. Identifying viable and safe methods for treating prion contaminated materials is important for hospitals, research facilities, biologists, hunters, and meat-processors. For three decades, some prion researchers have used a phenolic product called Environ LpH (eLpH) to inactivate prions. ELpH has been discontinued, but a similar product, Wex-cide 128, containing the similar phenolic chemicals as eLpH is now available. In the current study, we directly compared the anti-prion efficacy of eLpH and Wex-cide 128 against prions from four different species (hamster 263K, cervid CWD, mouse 22L and human CJD). Decontamination was performed on either prion infected brain homogenates or prion contaminated steel wires and mouse bioassay was used to quantify the remaining prion infectivity. Our data show that both eLpH and Wex-cide 128 removed 4.0-5.5 logs of prion infectivity from 22L, CWD and 263K prion homogenates, but only about 1.25-1.50 logs of prion infectivity from human sporadic CJD. Wex-cide 128 is a viable substitute for inactivation of most prions from most species, but the resistance of CJD to phenolic inactivation is a concern and emphasizes the fact that inactivation methods should be confirmed for each target prion strain.


Subject(s)
Creutzfeldt-Jakob Syndrome , Deer , Disinfectants , Prions , Scrapie , Sprains and Strains , Cricetinae , Humans , Animals , Cattle , Mice , Sheep , Brain , Disinfectants/pharmacology , Phenols
3.
Vet Res ; 53(1): 111, 2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36527166

ABSTRACT

Chronic wasting disease (CWD) is a prion disease of cervids including deer, elk, reindeer, and moose. Human consumption of cervids is common, therefore assessing the risk potential of CWD transmission to humans is critical. In a previous study, we tested CWD transmission via intracerebral inoculation into transgenic mice (tg66 and tgRM) that over-expressed human prion protein. Mice screened by traditional prion detection assays were negative. However, in a group of 88 mice screened by the ultrasensitive RT-QuIC assay, we identified 4 tg66 mice that produced inconsistent positive RT-QuIC reactions. These data could be false positive reactions, residual input inoculum or indicative of subclinical infections suggestive of cross species transmission of CWD to humans. Additional experiments were required to understand the nature of the prion seeding activity in this model. In this manuscript, second passage experiments using brains from mice with weak prion seeding activity showed they were not infectious to additional recipient tg66 mice. Clearance experiments showed that input CWD prion seeding activity was eliminated by 180 days in tg66 mice and PrPKO mice, which are unable to replicate prion protein, indicating that the weak positive levels of seeding activity detected at later time points was not likely residual inoculum. The failure of CWD prions to cause disease in tg66 after two sequential passages suggested that a strong species barrier prevented CWD infection of mice expressing human prion protein.


Subject(s)
Deer , Prions , Reindeer , Rodent Diseases , Wasting Disease, Chronic , Humans , Animals , Mice , Prion Proteins/genetics , Prions/genetics , Mice, Transgenic
4.
PLoS One ; 17(10): e0276850, 2022.
Article in English | MEDLINE | ID: mdl-36301895

ABSTRACT

Microglia (MG) are critical to host defense during prion infection, but the mechanism(s) of this neuroprotection are poorly understood. To better examine the influence of MG during prion infection, we reduced MG in the brains of C57BL/10 mice using PLX5622 and assessed prion clearance and replication using multiple approaches that included bioassay, immunohistochemistry, and Real-Time Quaking Inducted Conversion (RT-QuIC). We also utilized a strategy of intermittent PLX5622 treatments to reduce MG and allow MG repopulation to test whether new MG could alter prion disease progress. Lastly, we investigated the influence of MG using tga20 mice, a rapid prion model that accumulates fewer pathological features and less PrPres in the infected brain. In C57BL/10 mice we found that MG were excluded from the inoculation site early after infection, but Iba1 positive infiltrating monocytes/macrophage were present. Reducing MG in the brain prior to prion inoculation did not increase susceptibility to prion infection. Short intermittent treatments with PLX5622 in prion infected C57BL/10 mice after 80 dpi were unsuccessful at altering the MG population, gliosis, or survival. Additionally, MG depletion using PLX5622 in tga20 mice had only a minor impact on prion pathogenesis, indicating that the presence of MG might be less important in this fast model with less prion accumulation. In contrast to the benefits of MG against prion disease in late stages of disease, our current experiments suggest MG do not play a role in early prion pathogenesis, clearance, or replication.


Subject(s)
Prion Diseases , Prions , Animals , Mice , Prions/metabolism , Microglia/metabolism , Mice, Inbred C57BL , Prion Diseases/pathology , Brain/metabolism
5.
J Neuroinflammation ; 18(1): 194, 2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34488805

ABSTRACT

BACKGROUND: Past experiments studying innate immunity in the central nervous system (CNS) utilized microglia obtained from neonatal mouse brain, which differ developmentally from adult microglia. These differences might impact our current understanding of the role of microglia in CNS development, function, and disease. METHODS: Cytokine protein secretion was compared in ex vivo P3 and adult microglial cultures after exposure to agonists for three different toll-like receptors (TLR4, lipopolysaccharide [LPS]; TLR7, imiquimod [IMQ]; and TLR9, CpG Oligodeoxynucleotide [CpG-ODN] 1585). In addition, changes in inflammatory gene expression in ex vivo adult microglia in response to the TLR agonists was assessed. Furthermore, in vivo experiments evaluated changes in gene expression associated with inflammation and TLR signaling in brains of mice with or without treatment with PLX5622 to reduce microglia. RESULTS: Ex vivo adult and P3 microglia increased cytokine secretion when exposed to TLR4 agonist LPS and to TLR7 agonist IMQ. However, adult microglia decreased expression of numerous genes after exposure to TLR 9 agonist CpG-ODN 1585. In contrast, in vivo studies indicated a core group of inflammatory and TLR signaling genes increased when each of the TLR agonists was introduced into the CNS. Reducing microglia in the brain led to decreased expression of various inflammatory and TLR signaling genes. Mice with reduced microglia showed extreme impairment in upregulation of genes after exposure to TLR7 agonist IMQ. CONCLUSIONS: Cultured adult microglia were more reactive than P3 microglia to LPS or IMQ exposure. In vivo results indicated microglial influences on neuroinflammation were agonist specific, with responses to TLR7 agonist IMQ more dysregulated in mice with reduced microglia. Thus, TLR7-mediated innate immune responses in the CNS appeared more dependent on the presence of microglia. Furthermore, partial responses to TLR4 and TLR9 agonists in mice with reduced microglia suggested other cell types in the CNS can compensate for their absence.


Subject(s)
Immunity, Innate , Microglia , Animals , Cytokines/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Toll-Like Receptor 4 , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Toll-Like Receptors/agonists
6.
Viruses ; 13(7)2021 07 17.
Article in English | MEDLINE | ID: mdl-34372599

ABSTRACT

In prion diseases, the spread of infectious prions (PrPSc) is thought to occur within nerves and across synapses of the central nervous system (CNS). However, the mechanisms by which PrPSc moves within axons and across nerve synapses remain undetermined. Molecular motors, including kinesins and dyneins, transport many types of intracellular cargo. Kinesin-1C (KIF5C) has been shown to transport vesicles carrying the normal prion protein (PrPC) within axons, but whether KIF5C is involved in PrPSc axonal transport is unknown. The current study tested whether stereotactic inoculation in the striatum of KIF5C knock-out mice (Kif5c-/-) with 0.5 µL volumes of mouse-adapted scrapie strains 22 L or ME7 would result in an altered rate of prion spreading and/or disease timing. Groups of mice injected with each strain were euthanized at either pre-clinical time points or following the development of prion disease. Immunohistochemistry for PrP was performed on brain sections and PrPSc distribution and tempo of spread were compared between mouse strains. In these experiments, no differences in PrPSc spread, distribution or survival times were observed between C57BL/6 and Kif5c-/- mice.


Subject(s)
Brain/virology , Kinesins/genetics , Prion Diseases/physiopathology , Prions/pathogenicity , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Acta Neuropathol Commun ; 9(1): 17, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33509294

ABSTRACT

Accumulation of misfolded host proteins is central to neuropathogenesis of numerous human brain diseases including prion and prion-like diseases. Neurons of retina are also affected by these diseases. Previously, our group and others found that prion-induced retinal damage to photoreceptor cells in mice and humans resembled pathology of human retinitis pigmentosa caused by mutations in retinal proteins. Here, using confocal, epifluorescent and electron microscopy we followed deposition of disease-associated prion protein (PrPSc) and its association with damage to critical retinal structures following intracerebral prion inoculation. The earliest time and place of retinal PrPSc deposition was 67 days post-inoculation (dpi) on the inner segment (IS) of cone photoreceptors. At 104 and 118 dpi, PrPSc was associated with the base of cilia and swollen cone inner segments, suggesting ciliopathy as a pathogenic mechanism. By 118 dpi, PrPSc was deposited in both rods and cones which showed rootlet damage in the IS, and photoreceptor cell death was indicated by thinning of the outer nuclear layer. In the outer plexiform layer (OPL) in uninfected mice, normal host PrP (PrPC) was mainly associated with cone bipolar cell processes, but in infected mice, at 118 dpi, PrPSc was detected on cone and rod bipolar cell dendrites extending into ribbon synapses. Loss of ribbon synapses in cone pedicles and rod spherules in the OPL was observed to precede destruction of most rods and cones over the next 2-3 weeks. However, bipolar cells and horizontal cells were less damaged, indicating high selectivity among neurons for injury by prions. PrPSc deposition in cone and rod inner segments and on the bipolar cell processes participating in ribbon synapses appear to be critical early events leading to damage and death of photoreceptors after prion infection. These mechanisms may also occur in human retinitis pigmentosa and prion-like diseases, such as AD.


Subject(s)
Photoreceptor Connecting Cilium/metabolism , PrPSc Proteins/metabolism , Retinal Bipolar Cells/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Photoreceptor Cell Inner Segment/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Cell Death , Disease Progression , Mice , Microscopy, Confocal , Microscopy, Electron , Microscopy, Fluorescence , Photoreceptor Connecting Cilium/pathology , Photoreceptor Connecting Cilium/ultrastructure , PrPSc Proteins/administration & dosage , Retinal Bipolar Cells/pathology , Retinal Bipolar Cells/ultrastructure , Retinal Cone Photoreceptor Cells/pathology , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Photoreceptor Cell Inner Segment/pathology , Retinal Photoreceptor Cell Inner Segment/ultrastructure , Retinal Photoreceptor Cell Outer Segment/pathology , Retinal Photoreceptor Cell Outer Segment/ultrastructure , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/ultrastructure , Scrapie/metabolism , Scrapie/pathology
8.
Neurobiol Dis ; 144: 105057, 2020 10.
Article in English | MEDLINE | ID: mdl-32829029

ABSTRACT

Tau aggregates consisting of hyperphosphorylated tau fibrils are associated with many neurodegenerative diseases, including Alzheimer's disease, Pick's disease, frontotemporal dementia, and progressive supranuclear palsy. Tau may contribute to the pathogenesis of these diseases, collectively referred to as tauopathies. In human genetic prion diseases, tau aggregates are detected in association with amyloid plaques consisting of prion protein (PrP). However, the role of abnormal tau aggregates in PrP amyloid disease remains unclear. Previously we inoculated scrapie prions into transgenic mice expressing human tau, mouse tau, glycophosphatidylinositol (GPI) anchored PrP, and anchorless PrP. These mice developed both spongiform vacuolar pathology and PrP amyloid pathology, and human tau was detected near PrP amyloid plaques. However, the presence of human tau did not alter the disease tempo or prion-induced neuropathology. In the present study, we tested mice which more closely modeled familial human prion disease. These mice expressed human tau but lacked both mouse tau and GPI-anchored PrP. However, they did produce anchorless PrP, resulting in perivascular PrP amyloid plaques, i.e. cerebral amyloid angiopathy (CAA), without spongiform degeneration. Typical of PrP amyloid disease, the clinical course was very slow in this model. Nevertheless, the accumulation of aggregated, phosphorylated human tau and its association with PrP amyloid plaques failed to alter the timing or course of the clinical disease observed. These data suggest that human tau does not contribute to the pathogenesis of mouse PrP amyloid brain disease and raise the possibility that tau may also not be pathogenic in human PrP amyloid disease.


Subject(s)
Brain/metabolism , Cerebral Amyloid Angiopathy/metabolism , Plaque, Amyloid/metabolism , Prion Proteins/metabolism , Protein Aggregates , Scrapie/metabolism , tau Proteins/metabolism , Animals , Brain/pathology , Cerebral Amyloid Angiopathy/pathology , Disease Progression , Humans , Mice , Mice, Transgenic , Phosphorylation , Plaque, Amyloid/pathology , Scrapie/pathology , tau Proteins/genetics
9.
Mol Brain ; 13(1): 71, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32381108

ABSTRACT

BACKGROUND: Prion diseases and prion-like disorders, including Alzheimer's disease and Parkinson's disease, are characterized by gliosis and accumulation of misfolded aggregated host proteins. Ablating microglia in prion-infected brain by treatment with the colony-stimulating factor-1 receptor (CSF-1R) inhibitor, PLX5622, increased accumulation of misfolded prion protein and decreased survival time. METHODS: To better understand the role of glia during neurodegeneration, we used RNA-seq technology, network analysis, and hierarchical cluster analysis to compare gene expression in brains of prion-infected versus mock-inoculated mice. Comparisons were also made between PLX5622-treated prion-infected mice and untreated prion-infected mice to assess mechanisms involved in disease acceleration in the absence of microglia. RESULTS: RNA-seq and network analysis suggested that microglia responded to prion infection through activation of integrin CD11c/18 and did not adopt the expression signature associated with other neurodegenerative disease models. Instead, microglia acquired an alternative molecular signature late in the disease process. Furthermore, astrocytes expressed a signature pattern of genes which appeared to be specific for prion diseases. Comparisons were also made with prion-infected mice treated with PLX5622 to assess the impact of microglia ablation on astrocyte gene expression during prion infection. In the presence of microglia, a unique mix of transcripts associated with A1- and A2-reactive astrocytes was increased in brains of prion-infected mice. After ablation of microglia, this reactive astrocyte expression pattern was enhanced. Thus, after prion infection, microglia appeared to decrease the overall A1/A2-astrocyte responses which might contribute to increased survival after infection. CONCLUSIONS: RNA-seq analysis indicated dysregulation of over 300 biological processes within the CNS during prion disease. Distinctive microglia- and astrocyte-associated expression signatures were identified during prion infection. Furthermore, astrogliosis and the unique astrocyte-associated expression signature were independent of microglial influences. Astrogliosis and the unique astrocyte-associated gene expression pattern were increased when microglia were ablated. Our findings emphasize the potential existence of alternative pathways for activating the A1/A2 paradigm in astrocytes during neurodegenerative disease.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Organic Chemicals/administration & dosage , Prion Diseases/metabolism , Prions/metabolism , Transcriptome/genetics , Animals , Apoptosis/genetics , Brain/drug effects , Brain/pathology , CD11c Antigen/metabolism , CD18 Antigens/metabolism , Cluster Analysis , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Ontology , Gliosis , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Prion Diseases/drug therapy , Prions/pathogenicity , RNA-Seq , Scrapie/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects
10.
PLoS One ; 14(10): e0223659, 2019.
Article in English | MEDLINE | ID: mdl-31584997

ABSTRACT

Chronic wasting disease (CWD) is a fatal prion disease that can infect deer, elk and moose. CWD has now been detected in 26 states of the USA, 3 Canadian provinces, South Korea, Norway, Sweden and Finland. CWD continues to spread from endemic areas, and new foci of infections are frequently detected. As increasing numbers of cervids become infected, the likelihood for human exposure increases. To date, no cases of CWD infection in humans have been confirmed, but experience with the BSE zoonosis in the United Kingdom suggests exposure to CWD should be minimized. Specifically, hunters, meat processors and others in contact with tissues from potentially CWD-infected cervids need a practical method to decontaminate knives, saws and other equipment. Prions are notoriously difficult to inactivate, and most effective methods require chemicals or sterilization processes that are either dangerous, caustic, expensive or not readily available. Although corrosive, sodium hypochlorite (bleach) is widely available and affordable and has been shown to inactivate prion agents including those that cause scrapie, bovine spongiform encephalopathy and Creutzfeldt-Jakob disease. In the current study, we confirm that bleach is an effective disinfectant for CWD prions and establish minimum times and bleach concentrations to eliminate prion seeding activity from stainless steel and infected brain homogenate solutions. We found that a five-minute treatment with a 40% dilution of household bleach was effective at inactivating CWD seeding activity from stainless-steel wires and CWD-infected brain homogenates. However, bleach was not able to inactivate CWD seeding activity from solid tissues in our studies.


Subject(s)
Prion Diseases/metabolism , Prions/antagonists & inhibitors , Sodium Hypochlorite/pharmacology , Wasting Disease, Chronic/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Decontamination , Deer , Humans , Prion Diseases/diagnosis , Prion Diseases/drug therapy , Prion Diseases/etiology , Wasting Disease, Chronic/diagnosis , Wasting Disease, Chronic/drug therapy , Wasting Disease, Chronic/etiology
11.
Acta Neuropathol Commun ; 7(1): 48, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30909963

ABSTRACT

Degeneration of photoreceptors in the retina is a major cause of blindness in humans. Often retinal degeneration is due to inheritance of mutations in genes important in photoreceptor (PR) function, but can also be induced by other events including retinal trauma, microvascular disease, virus infection or prion infection. The onset of apoptosis and degeneration of PR neurons correlates with invasion of the PR cellular areas by microglia or monocytes, suggesting a causal role for these cells in pathogenesis of PR degenerative disease. To study the role of microglia in prion-induced retinal disease, we fed prion-infected mice a CSF-1 receptor blocking drug, PLX5622, to eliminate microglia in vivo, and the effects on retinal degeneration were analyzed over time. In mice not receiving drug, the main inflammatory cells invading the degenerating PR areas were microglia, not monocytes. Administration of PLX5622 was highly effective at ablating microglia in retina. However, lack of microglia during prion infection did not prevent degeneration of PR cells. Therefore, microglia were not required for the PR damage process during prion infection. Indeed, mice lacking microglia had slightly faster onset of PR damage. Similar results were seen in C57BL/10 mice and transgenic mice expressing GFP or RFP on microglia and monocytes, respectively. These results were supported by experiments using prion-infected Cx3cr1 knockout mice without PLX5622 treatment, where microglial expansion in retina was delayed, but PR degeneration was not. Contrary to predictions, microglia were not a causative factor in retinal damage by prion infection. Instead, newly generated PrPSc accumulated around the inner segment region of the PR cells and appeared to correlate with initiation of the pathogenic process in the absence of microglia.


Subject(s)
Microglia/pathology , Photoreceptor Cells, Vertebrate/pathology , PrPSc Proteins/toxicity , Retinal Degeneration/chemically induced , Retinal Degeneration/pathology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/drug effects , Organic Chemicals/pharmacology , Photoreceptor Cells, Vertebrate/drug effects
12.
Viruses ; 11(1)2019 01 15.
Article in English | MEDLINE | ID: mdl-30650564

ABSTRACT

Prion disorders are transmissible diseases caused by a proteinaceous infectious agent that can infect the lymphatic and nervous systems. The clinical features of prion diseases can vary, but common hallmarks in the central nervous system (CNS) are deposition of abnormally folded protease-resistant prion protein (PrPres or PrPSc), astrogliosis, microgliosis, and neurodegeneration. Numerous proinflammatory effectors expressed by astrocytes and microglia are increased in the brain during prion infection, with many of them potentially damaging to neurons when chronically upregulated. Microglia are important first responders to foreign agents and damaged cells in the CNS, but these immune-like cells also serve many essential functions in the healthy CNS. Our current understanding is that microglia are beneficial during prion infection and critical to host defense against prion disease. Studies indicate that reduction of the microglial population accelerates disease and increases PrPSc burden in the CNS. Thus, microglia are unlikely to be a foci of prion propagation in the brain. In contrast, neurons and astrocytes are known to be involved in prion replication and spread. Moreover, certain astrocytes, such as A1 reactive astrocytes, have proven neurotoxic in other neurodegenerative diseases, and thus might also influence the progression of prion-associated neurodegeneration.


Subject(s)
Inflammation , Microglia/immunology , Prion Diseases/pathology , Prions/immunology , Animals , Astrocytes/immunology , Astrocytes/pathology , Brain/immunology , Brain/pathology , Humans , Mice , Microglia/pathology , Neurodegenerative Diseases/immunology , Neurons/immunology , Neurons/pathology , PrPSc Proteins/immunology , PrPSc Proteins/pathogenicity , Prion Diseases/immunology
13.
Vet Res ; 50(1): 6, 2019 Jan 22.
Article in English | MEDLINE | ID: mdl-30670087

ABSTRACT

Chronic wasting disease (CWD) is a fatal prion disease which infects deer, elk and moose. CWD was first described as a wasting syndrome in captive deer in Colorado and Wyoming wildlife facilities from 1967 to 1979. Currently, CWD has been reported in 26 states of the USA, three Canadian provinces, South Korea, Norway and Finland. Since human consumption of cervids is common, it is critical to determine if CWD can infect humans. Published research, including epidemiologic studies and transmission studies using animal models, including transgenic mice that express human prion protein, have suggested existence of a strong species barrier between cervid CWD and humans. In the current study, we tested CWD transmission into two additional strains of transgenic mice (tg66 and tgRM). These mice over-express human prion protein at high levels and are highly sensitive to infection by human-tropic prions. One hundred and eight mice were inoculated intracerebrally with three different sources of CWD. After long periods of observation, brain tissues from CWD-inoculated mice were screened for evidence of prion infection by RT-QuIC, immunohistochemistry (IHC) and immunoblot. No IHC or immunoblot evidence was found to suggest transmission had occurred, and most mice were negative by RT-QuIC assay. However, four mice with inconsistent positive RT-QuIC reactions were detected. The seeding activity detected in these mice may represent a low level of CWD agent, suggesting a possible transfer of CWD infection. Alternatively, these results might be due to false positive reactions or residual CWD inoculum.


Subject(s)
Deer , Prion Proteins/genetics , Wasting Disease, Chronic/transmission , Animals , Brain/pathology , Disease Models, Animal , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Transgenic , Prion Proteins/isolation & purification , Wasting Disease, Chronic/pathology
14.
J Virol ; 92(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29769333

ABSTRACT

Microglial cells in the central nervous system play important roles in neurodevelopment and resistance to infection, yet microglia can become neurotoxic under some conditions. An early event during prion infection is the activation of microglia and astrocytes in the brain prior to damage or death of neurons. Previous prion disease studies using two different strategies to manipulate signaling through the microglial receptor CSF-1R reported contrary effects on survival from prion disease. However, in these studies, reductions of microglial numbers and function were variable, thus confounding interpretation of the results. In the present work, we used oral treatment with a potent inhibitor of CSF-1R, PLX5622, to eliminate 78 to 90% of microglia from cortex early during the course of prion infection. Oral drug treatment early after infection with the RML scrapie strain significantly accelerated vacuolation, astrogliosis, and deposition of disease-associated prion protein. Furthermore, drug-treated mice had advanced clinical disease requiring euthanasia 31 days earlier than untreated control mice. Similarly, PLX5622 treatment during the preclinical phase at 80 days postinfection with RML scrapie also accelerated disease and resulted in euthanasia of mice 33 days earlier than infected controls. PLX5622 also accelerated clinical disease after infection with scrapie strains ME7 and 22L. Thus, microglia are critical in host defense during prion disease. The early accumulation of PrPSc in the absence of microglia suggested that microglia may function by clearing PrPSc, resulting in longer survival.IMPORTANCE Microglia contribute to many aspects of health and disease. When activated, microglia can be beneficial by repairing damage in the central nervous system (CNS) or they can turn harmful by becoming neurotoxic. In prion and prionlike diseases, the involvement of microglia in disease is unclear. Previous studies suggest that microglia can either speed up or slow down disease. In this study, we infected mice with prions and depleted microglia from the brains of mice using PLX5622, an effective CSF-1R tyrosine kinase inhibitor. Microglia were markedly reduced in brains, and prion disease was accelerated, so that mice needed to be euthanized 20 to 33 days earlier than infected control mice due to advanced clinical disease. Similar results occurred when mice were treated with PLX5622 at 80 days after infection, which was just prior to the start of clinical signs. Thus, microglia are important for removing prions, and the disease is faster when microglia are depleted.


Subject(s)
Microglia/cytology , Microglia/drug effects , Organic Chemicals/adverse effects , PrPSc Proteins/metabolism , Scrapie/metabolism , Administration, Oral , Animals , Apoptosis , Disease Models, Animal , Female , Male , Mice , Microglia/metabolism , Microglia/pathology , Organic Chemicals/administration & dosage , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Scrapie/chemically induced , Scrapie/pathology , Severity of Illness Index
15.
J Virol ; 92(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29695429

ABSTRACT

Chronic wasting disease (CWD) is a fatal prion disease that can infect deer, elk, and moose. CWD was first recognized in captive deer kept in wildlife facilities in Colorado from 1967 to 1979. CWD has now been detected in 25 U.S. states, 2 Canadian provinces, South Korea, Norway, and Finland. It is currently unknown if humans are susceptible to CWD infection. Understanding the health risk from consuming meat and/or products from CWD-infected cervids is a critical human health concern. Previous research using transgenic mouse models and in vitro conversion assays suggests that a significant species barrier exists between CWD and humans. To date, reported epidemiologic studies of humans consuming cervids in areas where CWD is endemic have found no evidence to confirm CWD transmission to humans. Previously, we reported data from ongoing cross-species CWD transmission studies using two species of nonhuman primates as models. Squirrel monkeys (SM) and cynomolgus macaques (CM) were inoculated by either the intracerebral or oral route with brain homogenates from CWD-infected deer and elk containing high levels of infectivity. SM were highly susceptible to CWD infection, while CM were not. In the present study, we present new data for seven CWD-inoculated CM euthanized 11 to 13 years after CWD inoculation and eight additional uninoculated control CM. New and archival CM tissues were screened for prion infection by using the ultrasensitive real-time quaking-induced conversion (RT-QuIC) assay, immunohistochemistry, and immunoblotting. In this study, there was no clinical, pathological, or biochemical evidence suggesting that CWD was transmitted from cervids to CM.IMPORTANCE Chronic wasting disease (CWD) is a fatal prion disease found in deer, elk, and moose. Since it was first discovered in the late 1960s, CWD has now spread to at least 25 U.S. states, 2 Canadian provinces, South Korea, Norway, and Finland. Eradication of CWD from areas of endemicity is very unlikely, and additional spread will occur. As the range and prevalence of CWD increase, so will the potential for human exposure to CWD prions. It is currently unknown if CWD poses a risk to human health. However, determining this risk is critical to preventing a scenario similar to that which occurred when mad cow disease was found to be transmissible to humans. In the present study, we used cynomolgus macaque monkeys as a surrogate model for CWD transmission to humans. After 13 years, no evidence for CWD transmission to macaques was detected clinically or by using highly sensitive prion disease-screening assays.


Subject(s)
Wasting Disease, Chronic/diagnosis , Wasting Disease, Chronic/transmission , Animals , Biological Assay , Deer , Disease Models, Animal , Female , Macaca fascicularis , Male , Species Specificity
16.
Acta Neuropathol Commun ; 6(1): 13, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29458424

ABSTRACT

Human familial prion diseases are associated with mutations at 34 different prion protein (PrP) amino acid residues. However, it is unclear whether infectious prions are found in all cases. Mutant PrP itself may be neurotoxic, or alternatively, PrP mutation might predispose to spontaneous formation of infectious PrP isoforms. Previous reports demonstrated transmission to animal models by human brain tissue expressing 7 different PrP mutations, but 3 other mutations were not transmissible. In the present work, we tested transmission using brain homogenates from patients expressing 3 untested PrP mutants: G131V, Y226X, and Q227X. Human brain homogenates were injected intracerebrally into tg66 transgenic mice overexpressing human PrP. Mice were followed for nearly 800 days.From 593 to 762 dpi, 4 of 8 mice injected with Y226X brain had PrPSc detectable in brain by immunostaining, immunoblot, and PrP amyloid seeding activity assayed by RT-QuIC. From 531 to 784 dpi, 11 of 11 G131V-injected mice had PrPSc deposition in brain, but none were positive by immunoblot or RT-QuIC assay. In contrast, from 529 to 798 dpi, no tg66 mice injected with Q227X brain had PrPSc or PrP amyloid seeding activity detectable by these methods. Y226X is the only one of 4 known PrP truncations associated with familial disease which has been shown to be transmissible. This transmission of prion infectivity from a patient expressing truncated human PrP may have implications for the spread and possible transmission of other aggregated truncated proteins in prion-like diseases such as Alzheimer's disease, Parkinson's disease and tauopathies.


Subject(s)
Mutant Proteins/metabolism , Mutation/genetics , Prion Diseases/genetics , Prion Diseases/pathology , Prion Proteins/genetics , Prion Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Prion Diseases/metabolism
17.
PLoS One ; 13(12): e0208559, 2018.
Article in English | MEDLINE | ID: mdl-30596651

ABSTRACT

Neuroinflammation and neurodegeneration are common during prion infection, but the mechanisms that underlie these pathological features are not well understood. Several components of innate immunity, such as Toll-like receptor (TLR) 4 and Complement C1q, have been shown to influence prion disease. To identify additional components of innate immunity that might impact prion disease within the central nervous system (CNS), we screened RNA from brains of pre-clinical and clinical 22L-infected mice for alterations in genes associated with innate immunity. Transcription of several genes encoding damage-associated molecular pattern (DAMP) proteins and receptors were increased in the brains of prion-infected mice. To investigate the role of some of these proteins in prion disease of the CNS, we infected mice deficient in DAMP receptor genes Tlr2, C3ar1, and C5ar1 with 22L scrapie. Elimination of TLR2 accelerated disease by a median of 10 days, while lack of C3aR1 or C5aR1 had no effect on disease tempo. Histopathologically, all knockout mouse strains tested were similar to infected control mice in gliosis, vacuolation, and PrPSc deposition. Analysis of proinflammatory markers in the brains of infected knockout mice indicated only a few alterations in gene expression suggesting that C5aR1 and TLR2 signaling did not act synergistically in the brains of prion-infected mice. These results indicate that signaling through TLR2 confers partial neuroprotection during prion infection.


Subject(s)
Neuroprotection , Prion Diseases/pathology , Toll-Like Receptor 2/metabolism , Anaphylatoxins/analysis , Animals , Brain/metabolism , Brain/pathology , Chemokines/metabolism , Complement System Proteins/metabolism , Cytokines/metabolism , Disease Susceptibility , Gene Expression , Immunity, Innate/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Prion Diseases/metabolism , Prion Diseases/veterinary , RNA/genetics , RNA/metabolism , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/deficiency , Receptors, Complement/genetics , Receptors, Complement/metabolism , Severity of Illness Index , Signal Transduction , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics
18.
J Gen Virol ; 98(8): 2190-2199, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28758631

ABSTRACT

Neuroinflammation is a prominent component of several neurodegenerative diseases, including multiple sclerosis, Alzheimer's disease, Parkinson's disease, tauopathies, amyotrophic lateral sclerosis and prion diseases. In such conditions, the ability to decrease neuroinflammation by drug therapy may influence disease progression. Statins have been used to treat hyperlipidemia as well as reduce neuroinflammation and oxidative stress in various tissues. In previous studies, treatment of scrapie-infected mice with the type 1 statins, simvastatin or pravastatin, showed a small beneficial effect on survival time. In the current study, to increase the effectiveness of statin therapy, we treated infected mice with atorvastatin, a type 2 statin that has improved pharmacokinetics over many type 1 statins. Treatments with either simvastatin or pravastatin were tested for comparison. We evaluated scrapie-infected mice for protease-resistant PrP (PrPres) accumulation, gliosis, neuroinflammation and time until advanced clinical disease requiring euthanasia. All three statin treatments reduced total serum cholesterol ≥40 % in mice. However, gliosis and PrPres deposition were similar in statin-treated and untreated infected mice. Time to euthanasia due to advanced clinical signs was not changed in statin-treated mice relative to untreated mice, a finding at odds with previous reports. Expression of 84 inflammatory genes involved in neuroinflammation was also quantitated. Seven genes were reduced by pravastatin, and one gene was reduced by atorvastatin. In contrast, simvastatin therapy did not reduce any of the tested genes, but did slightly increase the expression of Ccl2 and Cxcl13. Our studies indicate that none of the three statins tested were effective in reducing scrapie-induced neuroinflammation or neuropathogenesis.


Subject(s)
Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/immunology , Pravastatin/administration & dosage , Simvastatin/administration & dosage , Animals , Humans , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/mortality , Scrapie
19.
Prion ; 11(4): 234-248, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28759310

ABSTRACT

In most human and animal prion diseases the abnormal disease-associated prion protein (PrPSc) is deposited as non-amyloid aggregates in CNS, spleen and lymphoid organs. In contrast, in humans and transgenic mice with PrP mutations which cause expression of PrP lacking a glycosylphosphatidylinositol (GPI)-anchor, most PrPSc is in the amyloid form. In transgenic mice expressing only anchorless PrP (tg anchorless), PrPSc is deposited not only in CNS and lymphoid tissues, but also in extraneural tissues including heart, brown fat, white fat, and colon. In the present paper, we report ultrastructural studies of amyloid PrPSc deposition in extraneural tissues of scrapie-infected tg anchorless mice. Amyloid PrPSc fibrils identified by immunogold-labeling were visible at high magnification in interstitial regions and around blood vessels of heart, brown fat, white fat, colon, and lymphoid tissues. PrPSc amyloid was located on and outside the plasma membranes of adipocytes in brown fat and cardiomyocytes, and appeared to invaginate and disrupt the plasma membranes of these cell types, suggesting cellular damage. In contrast, no cellular damage was apparent near PrPSc associated with macrophages in lymphoid tissues and colon, with enteric neuronal ganglion cells in colon or with adipocytes in white fat. PrPSc localized in macrophage phagolysosomes lacked discernable fibrils and might be undergoing degradation. Furthermore, in contrast to wild-type mice expressing GPI-anchored PrP, in lymphoid tissues of tg anchorless mice, PrPSc was not associated with follicular dendritic cells (FDC), and FDC did not display typical prion-associated pathogenic changes.


Subject(s)
Amyloid/metabolism , Brain/pathology , PrPSc Proteins/metabolism , Rodent Diseases/pathology , Scrapie/pathology , Amyloid/ultrastructure , Animals , Brain/ultrastructure , Gene Expression Regulation , Glycosylphosphatidylinositols/metabolism , Heart/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagosomes , PrPSc Proteins/ultrastructure , Rodent Diseases/metabolism , Scrapie/transmission
20.
Prion ; 10(4): 319-30, 2016 07 03.
Article in English | MEDLINE | ID: mdl-27463540

ABSTRACT

Tauopathies are a family of neurodegenerative diseases in which fibrils of human hyperphosphorylated tau (P-tau) are believed to cause neuropathology. In Alzheimer disease, P-tau associates with A-beta amyloid and contributes to disease pathogenesis. In familial human prion diseases and variant CJD, P-tau often co-associates with prion protein amyloid, and might also accelerate disease progression. To test this latter possibility, here we compared progression of amyloid prion disease in vivo after scrapie infection of mice with and without expression of human tau. The mice used expressed both anchorless prion protein (PrP) and membrane-anchored PrP, that generate disease associated amyloid and non-amyloid PrP (PrPSc) after scrapie infection. Human P-tau induced by scrapie infection was only rarely associated with non-amyloid PrPSc, but abundant human P-tau was detected at extracellular, perivascular and axonal deposits associated with amyloid PrPSc. This pathology was quite similar to that seen in familial prion diseases. However, association of human and mouse P-tau with amyloid PrPSc did not diminish survival time following prion infection in these mice. By analogy, human P-tau may not affect prion disease progression in humans. Alternatively, these results might be due to other factors, including rapidity of disease, blocking effects by mouse tau, or low toxicity of human P-tau in this model.


Subject(s)
Amyloid/metabolism , PrPSc Proteins/metabolism , Prion Proteins/metabolism , Scrapie/metabolism , tau Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Progression , Gene Expression Regulation , Humans , Immunoenzyme Techniques , Mice , Models, Animal , Phosphorylation , Scrapie/mortality , Scrapie/pathology , Survival Rate , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...