1
|
Rokad D, Harischandra DS, Samidurai M, Chang YT, Luo J, Lawana V, Sarkar S, Palanisamy BN, Manne S, Kim D, Zenitsky G, Jin H, Anantharam V, Willette A, Kanthasamy A, Kanthasamy AG. Manganese Exposure Enhances the Release of Misfolded α-Synuclein via Exosomes by Impairing Endosomal Trafficking and Protein Degradation Mechanisms. Int J Mol Sci 2024; 25:12207. [PMID: 39596274 PMCID: PMC11594990 DOI: 10.3390/ijms252212207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Excessive exposure to manganese (Mn) increases the risk of chronic neurological diseases, including Parkinson's disease (PD) and other related Parkinsonisms. Aggregated α-synuclein (αSyn), a hallmark of PD, can spread to neighboring cells by exosomal release from neurons. We previously discovered that Mn enhances its spread, triggering neuroinflammatory and neurodegenerative processes. To better understand the Mn-induced release of exosomal αSyn, we examined the effect of Mn on endosomal trafficking and misfolded protein degradation. Exposing MN9D dopaminergic neuronal cells stably expressing human wild-type (WT) αSyn to 300 μM Mn for 24 h significantly suppressed protein and mRNA expression of Rab11a, thereby downregulating endosomal recycling, forcing late endosomes to mature into multivesicular bodies (MVBs). Ectopic expression of WT Rab11a significantly mitigated exosome release, whereas ectopic mutant Rab11a (S25N) increased it. Our in vitro and in vivo studies reveal that Mn exposure upregulated (1) mRNA and protein levels of endosomal Rab27a, which mediates the fusion of MVBs with the plasma membrane; and (2) expression of the autophagosomal markers Beclin-1 and p62, but downregulated the lysosomal marker LAMP2, thereby impairing autophagolysosome formation as confirmed by LysoTracker, cathepsin, and acridine orange assays. Our novel findings demonstrate that Mn promotes the exosomal release of misfolded αSyn by impairing endosomal trafficking and protein degradation.
Collapse
Affiliation(s)
- Dharmin Rokad
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Dilshan S. Harischandra
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Manikandan Samidurai
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Yuan-Teng Chang
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Jie Luo
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Vivek Lawana
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Souvarish Sarkar
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Bharathi N. Palanisamy
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Sireesha Manne
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Dongsuk Kim
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Auriel Willette
- Department of Neurology, Rutgers University, New Brunswick, NJ 07101, USA;
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| | - Anumantha G. Kanthasamy
- Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; (D.R.); (D.S.H.); (J.L.); (V.L.); (S.S.); (B.N.P.); (S.M.); (D.K.)
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA; (M.S.); (Y.-T.C.); (G.Z.); (H.J.); (V.A.); (A.K.)
| |
Collapse
|
2
|
Goodwill VS, Dryden I, Choi J, De Lillo C, Soldau K, Llibre-Guerra J, Sanchez H, Sigurdson CJ, Lin JH. Minimal change prion retinopathy: Morphometric comparison of retinal and brain prion deposits in Creutzfeldt-Jakob disease. Exp Eye Res 2022; 222:109172. [PMID: 35803332 PMCID: PMC9946801 DOI: 10.1016/j.exer.2022.109172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023]
Abstract
Sporadic Creutzfeldt-Jakob disease (sCJD) is the most commonly diagnosed human prion disease caused by the abnormal misfolding of the 'cellular' prion protein (PrPC) into the transmissible 'scrapie-type' prion form (PrPSc). Neuropathologic evaluation of brains with sCJD reveals abnormal PrPSc deposits primarily in grey matter structures, often associated with micro-vacuolar spongiform changes in neuropil, neuronal loss, and gliosis. Abnormal PrPSc deposits have also been reported in the retina of patients with sCJD, but few studies have characterized the morphology of these retinal PrPSc deposits or evaluated for any retinal neurodegenerative changes. We performed histopathologic and morphometric analyses of retinal and brain prion deposits in 14 patients with sCJD. Interestingly, we discovered that the morphology of retinal PrPSc deposits generally differs from that of brain PrPSc deposits in terms of size and shape. We found that retinal PrPSc deposits consistently localize to the outer plexiform layer of the retina. Additionally, we observed that the retinal PrPSc deposits are not associated with the spongiform change, neuronal loss, and gliosis often seen in the brain. The stereotypic morphology and location of PrPSc deposits in sCJD retinas may help guide the use of ocular imaging devices in the detection of these deposits for a clinical diagnosis.
Collapse
Affiliation(s)
- Vanessa S Goodwill
- Department of Pathology, University of California, San Diego, CA, 92093, USA.
| | - Ian Dryden
- Departments of Pathology and Ophthalmology, Stanford University, CA, 94305, USA; VA Palo Alto Healthcare System, Palo Alto, CA, 94304, USA
| | - Jihee Choi
- Departments of Pathology and Ophthalmology, Stanford University, CA, 94305, USA; VA Palo Alto Healthcare System, Palo Alto, CA, 94304, USA
| | - Chiara De Lillo
- Departments of Pathology and Ophthalmology, Stanford University, CA, 94305, USA
| | - Katrin Soldau
- Department of Pathology, University of California, San Diego, CA, 92093, USA
| | - Jorge Llibre-Guerra
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63108, USA; Global Brain Health Institute, University of California, San Francisco, CA, 94143, USA
| | - Henry Sanchez
- Department of Neurology, University of California, San Francisco, CA, 94143, USA
| | | | - Jonathan H Lin
- Departments of Pathology and Ophthalmology, Stanford University, CA, 94305, USA; VA Palo Alto Healthcare System, Palo Alto, CA, 94304, USA.
| |
Collapse
|
3
|
Differential Accumulation of Misfolded Prion Strains in Natural Hosts of Prion Diseases. Viruses 2021; 13:v13122453. [PMID: 34960722 PMCID: PMC8706046 DOI: 10.3390/v13122453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 01/01/2023] Open
Abstract
Prion diseases, also known as transmissible spongiform encephalopathies (TSEs), are a group of neurodegenerative protein misfolding diseases that invariably cause death. TSEs occur when the endogenous cellular prion protein (PrPC) misfolds to form the pathological prion protein (PrPSc), which templates further conversion of PrPC to PrPSc, accumulates, and initiates a cascade of pathologic processes in cells and tissues. Different strains of prion disease within a species are thought to arise from the differential misfolding of the prion protein and have different clinical phenotypes. Different strains of prion disease may also result in differential accumulation of PrPSc in brain regions and tissues of natural hosts. Here, we review differential accumulation that occurs in the retinal ganglion cells, cerebellar cortex and white matter, and plexuses of the enteric nervous system in cattle with bovine spongiform encephalopathy, sheep and goats with scrapie, cervids with chronic wasting disease, and humans with prion diseases. By characterizing TSEs in their natural host, we can better understand the pathogenesis of different prion strains. This information is valuable in the pursuit of evaluating and discovering potential biomarkers and therapeutics for prion diseases.
Collapse
|
4
|
Moons L, De Groef L. Multimodal retinal imaging to detect and understand Alzheimer's and Parkinson's disease. Curr Opin Neurobiol 2021; 72:1-7. [PMID: 34399146 DOI: 10.1016/j.conb.2021.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/01/2021] [Accepted: 07/14/2021] [Indexed: 12/28/2022]
Abstract
Retinal neurodegeneration and visual dysfunctions have been reported in a majority of Alzheimer's and Parkinson's patients, and, in light of the quest for novel biomarkers for these neurodegenerative proteinopathies, the retina has been receiving increasing attention as an organ for diagnosing, monitoring, and understanding disease. Thinning of retinal layers, abnormalities in vasculature, and protein deposition can be imaged at unprecedented resolution, which offers a unique systems biology view on the cellular and molecular changes underlying these pathologies. It makes the retina not only a promising target for biomarker development, but it also suggests that novel fundamental insights into the pathophysiology of Alzheimer's and Parkinson's disease can be obtained by studying the retina-brain axis.
Collapse
Affiliation(s)
- Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Biology Department, University of Leuven, Naamsestraat 61 Box 2464, Leuven, 3000, Belgium; Leuven Brain Institute, Leuven, 3000, Belgium.
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Biology Department, University of Leuven, Naamsestraat 61 Box 2464, Leuven, 3000, Belgium; Leuven Brain Institute, Leuven, 3000, Belgium
| |
Collapse
|
5
|
Kushwaha R, Sinha A, Makarava N, Molesworth K, Baskakov IV. Non-cell autonomous astrocyte-mediated neuronal toxicity in prion diseases. Acta Neuropathol Commun 2021; 9:22. [PMID: 33546775 PMCID: PMC7866439 DOI: 10.1186/s40478-021-01123-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/14/2021] [Indexed: 02/08/2023] Open
Abstract
Under normal conditions, astrocytes perform a number of important physiological functions centered around neuronal support and synapse maintenance. In neurodegenerative diseases including Alzheimer’s, Parkinson’s and prion diseases, astrocytes acquire reactive phenotypes, which are sustained throughout the disease progression. It is not known whether in the reactive states associated with prion diseases, astrocytes lose their ability to perform physiological functions and whether the reactive states are neurotoxic or, on the contrary, neuroprotective. The current work addresses these questions by testing the effects of reactive astrocytes isolated from prion-infected C57BL/6J mice on primary neuronal cultures. We found that astrocytes isolated at the clinical stage of the disease exhibited reactive, pro-inflammatory phenotype, which also showed downregulation of genes involved in neurogenic and synaptogenic functions. In astrocyte-neuron co-cultures, astrocytes from prion-infected animals impaired neuronal growth, dendritic spine development and synapse maturation. Toward examining the role of factors secreted by reactive astrocytes, astrocyte-conditioned media was found to have detrimental effects on neuronal viability and synaptogenic functions via impairing synapse integrity, and by reducing spine size and density. Reactive microglia isolated from prion-infected animals were found to induce phenotypic changes in primary astrocytes reminiscent to those observed in prion-infected mice. In particular, astrocytes cultured with reactive microglia-conditioned media displayed hypertrophic morphology and a downregulation of genes involved in neurogenic and synaptogenic functions. In summary, the current study provided experimental support toward the non-cell autonomous mechanisms behind neurotoxicity in prion diseases and demonstrated that the astrocyte reactive phenotype associated with prion diseases is synaptotoxic.
Collapse
|
6
|
Striebel JF, Race B, Leung JM, Schwartz C, Chesebro B. Prion-induced photoreceptor degeneration begins with misfolded prion protein accumulation in cones at two distinct sites: cilia and ribbon synapses. Acta Neuropathol Commun 2021; 9:17. [PMID: 33509294 PMCID: PMC7845122 DOI: 10.1186/s40478-021-01120-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
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.
Collapse
|
7
|
Veys L, Van Houcke J, Aerts J, Van Pottelberge S, Mahieu M, Coens A, Melki R, Moechars D, De Muynck L, De Groef L. Absence of Uptake and Prion-Like Spreading of Alpha-Synuclein and Tau After Intravitreal Injection of Preformed Fibrils. Front Aging Neurosci 2021; 12:614587. [PMID: 33519421 PMCID: PMC7843377 DOI: 10.3389/fnagi.2020.614587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/24/2020] [Indexed: 12/26/2022] Open
Abstract
Although very different in etiology and symptoms, numerous neurodegenerative diseases can be classified as proteinopathies. More so, evidence indicates that the key misfolded proteins at the basis of different neuropathies might share common mechanisms of propagation. As such, the prion-like spreading of protein aggregates through the neural network is subject of intensive research focus and requires adequate models. Here, we made use of the well-defined architecture and large accessibility of the visual system, of which the retinotopic connections represent a simple route of anterograde signaling and an elegant model to investigate transsynaptic, prion-like spreading. In two independent studies, uptake and seeding of alpha-synuclein and tau were examined after intravitreal injection of preformed fibrils. However, extracellular matrix components in the vitreous space and at the vitreoretinal surface appeared to act as a barrier for the entry of both fibrils into the retina. These results show that further experimental refinement is needed to fully realize the potential of the visual system as a model for studying the molecular and cellular mechanisms of anterograde, transsynaptic spreading of prion-like proteins.
Collapse
Affiliation(s)
- Lien Veys
- Laboratory Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium
| | - Jessie Van Houcke
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Jeroen Aerts
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Sophie Van Pottelberge
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Michel Mahieu
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Audrey Coens
- Laboratory of Neurodegenerative Disease, Institute François Jacob, MIRCen, CEA-CNRS, Fontenay aux Roses, France
| | - Ronald Melki
- Laboratory of Neurodegenerative Disease, Institute François Jacob, MIRCen, CEA-CNRS, Fontenay aux Roses, France
| | - Dieder Moechars
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Louis De Muynck
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Lies De Groef
- Laboratory Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Manne S, Kondru N, Jin H, Serrano GE, Anantharam V, Kanthasamy A, Adler CH, Beach TG, Kanthasamy AG. Blinded RT-QuIC Analysis of α-Synuclein Biomarker in Skin Tissue From Parkinson's Disease Patients. Mov Disord 2020; 35:2230-2239. [PMID: 32960470 PMCID: PMC7749035 DOI: 10.1002/mds.28242] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/15/2020] [Accepted: 07/29/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND An unmet clinical need in Parkinson's disease (PD) is to identify biomarkers for diagnosis, preferably in peripherally accessible tissues such as skin. Immunohistochemical studies have detected pathological α-synuclein (αSyn) in skin biopsies from PD patients albeit sensitivity needs to be improved. OBJECTIVE Our study provides the ultrasensitive detection of pathological αSyn present in the skin of PD patients, and thus, pathological αSyn in skin could be a potential biomarker for PD. METHODS The real-time quaking-induced conversion assay was used to detect pathological αSyn present in human skin tissues. Further, we optimized this ultra-sensitive and specific assay for both frozen and formalin-fixed paraffin-embedded sections of skin tissues. We determined the seeding kinetics of the αSyn present in the skin from autopsied subjects consisting of frozen skin tissues from 25 PD and 25 controls and formalin-fixed paraffin-embedded skin sections from 12 PD and 12 controls. RESULTS In a blinded study of skin tissues from autopsied subjects, we correctly identified 24/25 PD and 24/25 controls using frozen skin tissues (96% sensitivity and 96% specificity) compared to 9/12 PD and 10/12 controls using formalin-fixed paraffin-embedded skin sections (75% sensitivity and 83% specificity). CONCLUSIONS Our blinded study results clearly demonstrate the feasibility of using skin tissues for clinical diagnosis of PD by detecting pathological αSyn. Moreover, this peripheral biomarker discovery study may have broader translational value in detecting misfolded proteins in skin samples as a longitudinal progression marker. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sireesha Manne
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Naveen Kondru
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | | | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Charles H. Adler
- Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259
| | | | - Anumantha G. Kanthasamy
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| |
Collapse
|
9
|
Mammadova N, West Greenlee MH, Moore SJ, Hwang S, Lehmkuhl AD, Nicholson EM, Greenlee JJ. Evaluation of Antemortem Diagnostic Techniques in Goats Naturally Infected With Scrapie. Front Vet Sci 2020; 7:517862. [PMID: 33240943 PMCID: PMC7677257 DOI: 10.3389/fvets.2020.517862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
Scrapie is a naturally occurring transmissible spongiform encephalopathy (TSE) that affects sheep and goats. Sheep and goats can be infected with scrapie as lambs or kids via contact with the placenta or placental fluids, or from ingestion of prions shed in the environment and/or bodily fluids (e.g., saliva, urine, and feces). Like other TSEs, scrapie is generally not diagnosed before extensive and irreversible brain damage has occurred. Therefore, a reliable method to screen animals may facilitate diagnosis. Additionally, while natural scrapie in sheep has been widely described, naturally acquired goat scrapie is less well-characterized. The purpose of this study was to better understand natural goat scrapie in regard to disease phenotype (i.e., incubation period, clinical signs, neuroanatomical deposition patterns of PrPSc, and molecular profile as detected by Western blot) and to evaluate the efficacy of antemortem tests to detect scrapie-positive animals in a herd of goats. Briefly, 28 scrapie-exposed goats were removed from a farm depopulated due to previous diagnoses of scrapie on the premises and observed daily for 30 months. Over the course of the observation period, antemortem biopsies of recto-anal mucosa-associated lymphoid tissue (RAMALT) were taken and tested using immunohistochemistry and real-time quaking-induced conversion (RT-QuIC), and retinal thickness was measured in vivo using optical coherence tomography (OCT). Following the observation period, immunohistochemistry and Western blot were performed to assess neuroanatomical deposition patterns of PrPSc and molecular profile. Our results demonstrate that antemortem rectal biopsy was 77% effective in identifying goats naturally infected with scrapie and that a positive antemortem rectal biopsy was associated with the presence of clinical signs of neurologic disease and a positive dam status. We report that changes in retinal thickness are not detectable over the course of the observation period in goats naturally infected with scrapie. Finally, our results indicate that the accumulation of PrPSc in central nervous system (CNS) and non-CNS tissues is consistent with previous reports of scrapie in sheep and goats.
Collapse
Affiliation(s)
- Najiba Mammadova
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - M Heather West Greenlee
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - S Jo Moore
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Soyoun Hwang
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Aaron D Lehmkuhl
- National Veterinary Services Laboratories (NVSL) Diagnostic Bacteriology and Pathology Laboratory, Animal and Plant Health Inspection Service, United States Department of Agriculture, Ames, IA, United States
| | - Eric M Nicholson
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Justin J Greenlee
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| |
Collapse
|
10
|
Microglia in Prion Diseases: Angels or Demons? Int J Mol Sci 2020; 21:ijms21207765. [PMID: 33092220 PMCID: PMC7589037 DOI: 10.3390/ijms21207765] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
Prion diseases are rare transmissible neurodegenerative disorders caused by the accumulation of a misfolded isoform (PrPSc) of the cellular prion protein (PrPC) in the central nervous system (CNS). Neuropathological hallmarks of prion diseases are neuronal loss, astrogliosis, and enhanced microglial proliferation and activation. As immune cells of the CNS, microglia participate both in the maintenance of the normal brain physiology and in driving the neuroinflammatory response to acute or chronic (e.g., neurodegenerative disorders) insults. Microglia involvement in prion diseases, however, is far from being clearly understood. During this review, we summarize and discuss controversial findings, both in patient and animal models, suggesting a neuroprotective role of microglia in prion disease pathogenesis and progression, or—conversely—a microglia-mediated exacerbation of neurotoxicity in later stages of disease. We also will consider the active participation of PrPC in microglial functions, by discussing previous reports, but also by presenting unpublished results that support a role for PrPC in cytokine secretion by activated primary microglia.
Collapse
|
11
|
Singh N, Chaudhary S, Ashok A, Lindner E. Prions and prion diseases: Insights from the eye. Exp Eye Res 2020; 199:108200. [PMID: 32858007 DOI: 10.1016/j.exer.2020.108200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/24/2020] [Accepted: 08/21/2020] [Indexed: 12/30/2022]
Abstract
Prion diseases are invariably fatal neurodegenerative disorders that have gained much publicity due to their transmissible nature. Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common human prion disorder, with an incidence of 1 in a million. Inherited prion disorders are relatively rare, and associated with mutations in the prion protein gene. More than 50 different point mutations, deletions, and insertions have been identified so far. Most are autosomal dominant and fully penetrant. Prion disorders also occur in animals, and are of major concern because of the potential for spreading to humans. The principal pathogenic event underlying all prion disorders is a change in the conformation of prion protein (PrPC) from a mainly α-helical to a β-sheet rich isoform, PrP-scrapie (PrPSc). Accumulation of PrPSc in the brain parenchyma is the major cause of neuronal degeneration. The mechanism by which PrPSc is transmitted, propagates, and causes neurodegenerative changes has been investigated over the years, and several clues have emerged. Efforts are also ongoing for identifying specific and sensitive diagnostic tests for sCJD and animal prion disorders, but success has been limited. The eye is suitable for these evaluations because it shares several anatomical and physiological features with the brain, and can be observed in vivo during disease progression. The retina, considered an extension of the central nervous system, is involved extensively in prion disorders. Accordingly, Optical Coherence Tomography and electroretinogram have shown some promise as pre-mortem diagnostic tests for human and animal prion disorders. However, a complete understanding of the physiology of PrPC and pathobiology of PrPSc in the eye is essential for developing specific and sensitive tests. Below, we summarize recent progress in ocular physiology and pathology in prion disorders, and the eye as an anatomically accessible site to diagnose, monitor disease progression, and test therapeutic options.
Collapse
Affiliation(s)
- Neena Singh
- Departments of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Suman Chaudhary
- Departments of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ajay Ashok
- Departments of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ewald Lindner
- Department of Ophthalmology, Medical University of Graz, Auenbruggerplatz 4, 8036, Graz, Austria
| |
Collapse
|
12
|
Lychee seed polyphenol inhibits Aβ-induced activation of NLRP3 inflammasome via the LRP1/AMPK mediated autophagy induction. Biomed Pharmacother 2020; 130:110575. [PMID: 32768883 DOI: 10.1016/j.biopha.2020.110575] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/14/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence indicates that the enhancement of microglial autophagy inhibits the NLRP3 inflammasome mediated neuroinflammation in Alzheimer's disease (AD). Meanwhile, low density lipoprotein receptor-related protein 1 (LRP1) highly expressed in microglia is able to negatively regulate neuroinflammation and positively regulate autophagy. In addition, we have previously reported that an active lychee seed fraction enriching polyphenol (LSP) exhibits anti-neuroinflammation in Aβ-induced BV-2 cells. However, its molecular mechanism of action is still unclear. In this study, we aim to investigate whether LSP inhibits the NLRP3 inflammasome mediated neuroinflammation and clarify its molecular mechanism in Aβ-induced BV-2 cells and APP/PS1 mice. The results showed that LSP dose- and time-dependently activated autophagy by increasing the expression of Beclin 1 and LC3II in BV-2 cells, which was regulated by the upregulation of LRP1 and its mediated AMPK signaling pathway. In addition, both the Western blotting and fluorescence microscopic results demonstrated that LSP could significantly suppress the activation of NLRP3 inflammasome by inhibiting the expression of NLRP3, ASC, the cleavage of caspase-1, and the release of IL-1β in Aβ(1-42)-induced BV-2 cells. In addition, the siRNA LRP1 successfully abolished the effect of LSP on the activation of AMPK and its mediated autophagy, as well as the inhibition of NLRP3 inflammasome. Furthermore, LSP rescued PC-12 cells which were induced by the conditioned medium from Aβ(1-42)-treated BV-2 cells. Moreover, LSP improved the cognitive function and inhibited the NLRP3 inflammasome in APP/PS1 mice. Taken together, LSP inhibited the NLRP3 inflammasome-mediated neuroinflammation in the in vitro and in vivo models of AD, which was closely associated with the LRP1/AMPK-mediated autophagy. Thus, the findings from this study further provide evidences for LSP serving as a potential drug for the treatment of AD in the future.
Collapse
|
13
|
Kondru N, Manne S, Kokemuller R, Greenlee J, Greenlee MHW, Nichols T, Kong Q, Anantharam V, Kanthasamy A, Halbur P, Kanthasamy AG. An Ex Vivo Brain Slice Culture Model of Chronic Wasting Disease: Implications for Disease Pathogenesis and Therapeutic Development. Sci Rep 2020; 10:7640. [PMID: 32376941 PMCID: PMC7203233 DOI: 10.1038/s41598-020-64456-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 04/12/2020] [Indexed: 12/03/2022] Open
Abstract
Chronic wasting disease (CWD) is a rapidly spreading prion disease of cervids, yet antemortem diagnosis, treatment, and control remain elusive. We recently developed an organotypic slice culture assay for sensitive detection of scrapie prions using ultrasensitive prion seeding. However, this model was not established for CWD prions due to their strong transmission barrier from deer (Odocoileus spp) to standard laboratory mice (Mus musculus). Therefore, we developed and characterized the ex vivo brain slice culture model for CWD, using a transgenic mouse model (Tg12) that expresses the elk (Cervus canadensis) prion protein gene (PRNP). We tested for CWD infectivity in cultured slices using sensitive seeding assays such as real-time quaking-induced conversion (RT-QuIC) and protein misfolding cyclic amplification (PMCA). Slice cultures from Tg12, but not from prnp-/- mice, tested positive for CWD. Slice-generated CWD prions transmitted efficiently to Tg12 mice. Furthermore, we determined the activity of anti-prion compounds and optimized a screening protocol for the infectivity of biological samples in this CWD slice culture model. Our results demonstrate that this integrated brain slice model of CWD enables the study of pathogenic mechanisms with translational implications for controlling CWD.
Collapse
Affiliation(s)
- Naveen Kondru
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Sireesha Manne
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Robyn Kokemuller
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Justin Greenlee
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - M Heather West Greenlee
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Tracy Nichols
- Surveillance, Preparedness and Response Services, Veterinary Services, United States Department of Agriculture, Fort Collins, CO, USA
| | - Qingzhong Kong
- Departments of Pathology and Neurology, Case Western Reserve University, Cleveland, OH, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Patrick Halbur
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| |
Collapse
|
14
|
Experimental Study Using Multiple Strains of Prion Disease in Cattle Reveals an Inverse Relationship between Incubation Time and Misfolded Prion Accumulation, Neuroinflammation, and Autophagy. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1461-1473. [PMID: 32259521 DOI: 10.1016/j.ajpath.2020.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 03/11/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023]
Abstract
Proteinopathies result from aberrant folding and accumulation of specific proteins. Currently, there is a lack of knowledge about the factors that influence disease progression, making this a key challenge for the development of therapies for proteinopathies. Because of the similarities between transmissible spongiform encephalopathies (TSEs) and other protein misfolding diseases, TSEs can be used to understand other proteinopathies. Bovine spongiform encephalopathy (BSE) is a TSE that occurs in cattle and can be subdivided into three strains: classic BSE and atypical BSEs (H and L types) that have shorter incubation periods. The NACHT, LRR, and PYD domains-containing protein 3 inflammasome is a critical component of the innate immune system that leads to release of IL-1β. Macroautophagy is an intracellular mechanism that plays an essential role in protein clearance. In this study, the retina was used as a model to investigate the relationship between disease incubation period, prion protein accumulation, neuroinflammation, and changes in macroautophagy. We demonstrate that atypical BSEs present with increased prion protein accumulation, neuroinflammation, and decreased autophagy. This work suggests a relationship between disease time course, neuroinflammation, and the autophagic stress response, and may help identify novel therapeutic biomarkers that can delay or prevent the progression of proteinopathies.
Collapse
|
15
|
Iwamaru Y, Matsuura Y, Miyazawa K. PrPSc with Seeding Activity Extensively Overlaps with Proteinase-Resistant PrPSc Rather than Infectious PrPSc. Pathogens 2020; 9:pathogens9030241. [PMID: 32213939 PMCID: PMC7157578 DOI: 10.3390/pathogens9030241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
The disease-associated prion protein (PrPSc) has the ability to seed the conformational conversion of normal prion proteins into the amyloid fibril form. This prion seeding activity can be measured using an in vitro amplification assay termed real-time quaking-induced conversion (RT-QuIC). There is a strong correlation between RT-QuIC positivity and prion infection; however, the relationship between seeding activity and infectivity remains elusive. In this study, we used endpoint dilution RT-QuIC on the brain homogenates from wild-type mice with mouse-adopted bovine spongiform encephalopathy (mBSE) at defined intervals during the incubation period and evaluated the temporal relationship among prion seeding dose, levels of proteinase-resistant PrPSc (PrPres), and infectious titer. We found that the infectious titer reached a plateau by 100 days postinfection, whereas seeding dose and PrPres levels were continuously elevated. Our calculation showed that the doubling time (dt) for seeding dose from 40 to 100 days postinoculation was closer to the dt for PrPres levels than to the dt for prion titer. Although an uncoupling of seeding doses and PrPres levels was observed at end-stage disease in this model, our findings suggest that there is substantial but not complete overlap between PrPSc with seeding activity and PrPres rather than infectious PrPSc.
Collapse
|
16
|
Moore J, Tatum T, Hwang S, Vrentas C, West Greenlee MH, Kong Q, Nicholson E, Greenlee J. Novel Strain of the Chronic Wasting Disease Agent Isolated From Experimentally Inoculated Elk With LL132 Prion Protein. Sci Rep 2020; 10:3148. [PMID: 32081886 PMCID: PMC7035384 DOI: 10.1038/s41598-020-59819-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/04/2020] [Indexed: 02/05/2023] Open
Abstract
Chronic wasting disease (CWD) is a fatal, progressive disease that affects cervid species, including Rocky mountain elk (Cervus elaphus nelsoni). There are 2 allelic variants in the elk prion protein gene: L132 (leucine) and M132 (methionine). Following experimental oral challenge with the CWD agent incubation periods are longest in LL132 elk, intermediate in ML132 elk, and shortest in MM132 elk. In order to ascertain whether such CWD-infected elk carry distinct prion strains, groups of Tg12 mice that express M132 elk prion protein were inoculated intracranially with brain homogenate from individual CWD-infected elk of various genotypes (LL132, LM132, or MM132). Brain samples were examined for microscopic changes and assessment of the biochemical properties of disease-associated prion protein (PrPSc). On first passage, mice challenged with LL132 elk inoculum had prolonged incubation periods and greater PrPSc fibril stability compared to mice challenged with MM132 or LM132 inoculum. On second passage, relative incubation periods, western blot profiles, and neuropathology were maintained. These results suggest that the CWD prion isolated from LL132 elk is a novel CWD strain and that M132 PrPC is able to propagate some biophysical properties of the L132 PrPSc conformation.
Collapse
Affiliation(s)
- Jo Moore
- USDA, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, 50010, USA
| | - Trudy Tatum
- USDA, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, 50010, USA
| | - Soyoun Hwang
- USDA, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, 50010, USA
| | - Catherine Vrentas
- USDA, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, 50010, USA
| | | | - Qingzhong Kong
- Case Western Reserve University, Departments of Pathology and Neurology, Cleveland, 44106, USA
| | - Eric Nicholson
- USDA, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, 50010, USA
| | - Justin Greenlee
- USDA, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, 50010, USA.
| |
Collapse
|
17
|
Manne S, Kondru N, Jin H, Anantharam V, Huang X, Kanthasamy A, Kanthasamy AG. α-Synuclein real-time quaking-induced conversion in the submandibular glands of Parkinson's disease patients. Mov Disord 2020; 35:268-278. [PMID: 31758740 PMCID: PMC7102508 DOI: 10.1002/mds.27907] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Identification of a peripheral biomarker is a major roadblock in the diagnosis of PD. Immunohistological identification of p-serine 129 α-synuclein in the submandibular gland tissues of PD patients has been recently reported. OBJECTIVE We report on a proof-of-principle study for using an ultra-sensitive and specific, real-time quaking-induced conversion assay to detect pathological α-synuclein in the submandibular gland tissues of PD patients. METHODS The α-synuclein real-time quaking-induced conversion assay was used to detect and quantify pathological α-synuclein levels in PD, incidental Lewy body disease, and control submandibular gland tissues as well as in formalin-fixed paraffin-embedded sections. RESULTS We determined the quantitative seeding kinetics of pathological α-synuclein present in submandibular gland tissues from autopsied subjects using the α-synuclein real-time quaking-induced conversion assay. A total of 32 cases comprising 13 PD, 3 incidental Lewy body disease, and 16 controls showed 100% sensitivity and 94% specificity. Interestingly, both PD and incidental Lewy body disease tissues showed 100% concordance for elevated levels of pathological α-synuclein seeding activity compared to control tissues. End-point dilution kinetic analyses revealed that the submandibular gland had a wide dynamic range of pathological α-synuclein seeding activity. CONCLUSIONS Our results are the first to demonstrate the utility of using the real-time quaking-induced conversion assay on peripherally accessible submandibular gland tissues and formalin-fixed paraffin-embedded tissue sections to detect PD-related pathological changes with high sensitivity and specificity. Additionally, the detection of seeding activity from incidental Lewy body disease cases containing immunohistochemically undetected pathological α-synuclein demonstrates the α-synuclein real-time quaking-induced conversion assay's potential utility for identifying prodromal PD in submandibular gland tissues. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sireesha Manne
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Naveen Kondru
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Xuemei Huang
- Departments of Neurology and Pharmacology, Neurosurgery, Radiology, and Kinesiology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Anumantha G. Kanthasamy
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| |
Collapse
|
18
|
Region-specific glial homeostatic signature in prion diseases is replaced by a uniform neuroinflammation signature, common for brain regions and prion strains with different cell tropism. Neurobiol Dis 2020; 137:104783. [PMID: 32001329 DOI: 10.1016/j.nbd.2020.104783] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 02/08/2023] Open
Abstract
Chronic neuroinflammation is recognized as a major neuropathological hallmark in a broad spectrum of neurodegenerative diseases including Alzheimer's, Parkinson's, Frontal Temporal Dementia, Amyotrophic Lateral Sclerosis, and prion diseases. Both microglia and astrocytes exhibit region-specific homeostatic transcriptional identities, which under chronic neurodegeneration, transform into reactive phenotypes in a region- and disease-specific manner. Little is known about region-specific identity of glia in prion diseases. The current study was designed to determine whether the region-specific homeostatic signature of glia changes with the progression of prion diseases, and whether these changes occur in a region-dependent or universal manner. Also of interest was whether different prion strains give rise to different reactive phenotypes. To answer these questions, we analyzed gene expression in the thalamus, cortex, hypothalamus and hippocampus of mice infected with 22L and ME7 prion strains using a Nanostring Neuroinflammation panel at the subclinical, early clinical and advanced stages of the disease. We found that at the preclinical stage of the disease, the region-specific homeostatic identities were preserved. However, with the appearance of clinical signs, the region-specific signatures were partially lost and replaced with a neuroinflammation signature. While the same sets of genes were activated by both prion strains, the timing of neuroinflammation and the degree of activation in different brain regions was strain-specific. Changes in astrocyte function scored at the top of the activated pathways. Moreover, clustering analysis suggested that the astrocyte function pathway responded to prion infection prior to the Activated Microglia or Neuron and Neurotransmission pathways. The current work established neuroinflammation gene expression signature associated with prion diseases. Our results illustrate that with the disease progression, the region-specific homeostatic transcriptome signatures are replaced by the region-independent neuroinflammation signature, which is common for prion strains with different cell tropism. The prion-associated neuroinflammation signature identified in the current study overlapped only partially with the microglia degenerative phenotype and the disease-associated microglia phenotype reported for animal models of other neurodegenerative diseases.
Collapse
|
19
|
Makarava N, Chang JCY, Kushwaha R, Baskakov IV. Region-Specific Response of Astrocytes to Prion Infection. Front Neurosci 2019; 13:1048. [PMID: 31649496 PMCID: PMC6794343 DOI: 10.3389/fnins.2019.01048] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022] Open
Abstract
Chronic neuroinflammation involves reactive microgliosis and astrogliosis, and is regarded as a common pathological hallmark of neurodegenerative diseases including Alzheimer’s, Parkinson’s, ALS and prion diseases. Reactive astrogliosis, routinely observed immunohistochemically as an increase in glial fibrillary acidic protein (GFAP) signal, is a well-documented feature of chronic neuroinflammation associated with neurodegenerative diseases. Recent studies on single-cell transcriptional profiling of a mouse brain revealed that, under normal conditions, several distinct subtypes of astrocytes with regionally specialized distribution exist. However, it remains unclear whether astrocytic response to pro-inflammatory pathological conditions is uniform across whole brain or is region-specific. The current study compares the response of microglia and astrocytes to prions in mice infected with 22L mouse-adapted prion strain. While the intensity of reactive microgliosis correlated well with the extent of PrPSc deposition, reactive astrogliosis displayed a different, region-specific pattern. In particular, the thalamus and stratum oriens of hippocampus, which are both affected by 22L prions, displayed strikingly different response of astrocytes to PrPSc. Astrocytes in stratum oriens of hippocampus responded to accumulation of PrPSc with visible hypertrophy and increased GFAP, while in the thalamus, despite stronger PrPSc signal, the increase of GFAP was milder than in hippocampus, and the change in astrocyte morphology was less pronounced. The current study suggests that astrocyte response to prion infection is heterogeneous and, in part, defined by brain region. Moreover, the current work emphasizes the needs for elucidating region-specific changes in functional states of astrocytes and exploring the impact of these changes to chronic neurodegeneration.
Collapse
Affiliation(s)
- Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jennifer Chen-Yu Chang
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rajesh Kushwaha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ilia V Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
20
|
Manne S, Kondru N, Hepker M, Jin H, Anantharam V, Lewis M, Huang X, Kanthasamy A, Kanthasamy AG. Ultrasensitive Detection of Aggregated α-Synuclein in Glial Cells, Human Cerebrospinal Fluid, and Brain Tissue Using the RT-QuIC Assay: New High-Throughput Neuroimmune Biomarker Assay for Parkinsonian Disorders. J Neuroimmune Pharmacol 2019; 14:423-435. [PMID: 30706414 PMCID: PMC6669119 DOI: 10.1007/s11481-019-09835-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/10/2019] [Indexed: 01/23/2023]
Abstract
Adult-onset neurodegenerative disorders, like Parkinson's disease (PD) and dementia with Lewy bodies (DLB), that share the accumulation of aggregated α-synuclein (αSynagg) as their hallmark molecular pathology are collectively known as α-synucleinopathies. Diagnosing α-synucleinopathies requires the post-mortem detection of αSynagg in various brain regions. Recent efforts to measure αSynagg in living patients include quantifying αSynagg in different biofluids as a biomarker for PD. We adopted the real-time quaking-induced conversion (RT-QuIC) assay to detect very low levels of αSynagg. We first optimized RT-QuIC for sensitivity, specificity, and reproducibility by using monomeric recombinant human wild-type αSyn as a substrate and αSynagg as the seed. Next, we exposed mouse microglia to αSyn pre-formed fibrils (αSynPFF) for 24 h. RT-QuIC assay revealed that the αSynPFF is taken up rapidly by mouse microglia, within 30 min, and cleared within 24 h. We then evaluated the αSyn RT-QuIC assay for detecting αSynagg in human PD, DLB, and Alzheimer's disease (AD) post-mortem brain homogenates (BH) along with PD and progressive supranuclear palsy (PSP) cerebrospinal fluid (CSF) samples and then determined protein aggregation rate (PAR) for αSynagg. The PD and DLB BH samples not only showed significantly higher αSynagg PAR compared to age-matched healthy controls and AD, but RT-QuIC was also highly reproducible with 94% sensitivity and 100% specificity. Similarly, PD CSF samples demonstrated significantly higher αSynagg PAR compared to age-matched healthy controls, with 100% sensitivity and specificity. Overall, the RT-QuIC assay accurately detects αSynagg seeding activity, offering a potential tool for antemortem diagnosis of α-synucleinopathies and other protein-misfolding disorders. Graphical Abstract A schematic representation of αSyn RT-QuIC assay.
Collapse
Affiliation(s)
- Sireesha Manne
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Naveen Kondru
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Monica Hepker
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Mechelle Lewis
- Departments of Neurology and Pharmacology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033
| | - Xuemei Huang
- Departments of Neurology and Pharmacology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033
- Neurosurgery, Radiology, and Kinesiology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| | - Anumantha G. Kanthasamy
- Department of Biomedical Sciences, Parkinson’s Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011
| |
Collapse
|
21
|
Michael AV, Greenlee JJ, Harm TA, Moore SJ, Zhang M, Lind MS, Greenlee MHW, Smith JD. In Situ Temporospatial Characterization of the Glial Response to Prion Infection. Vet Pathol 2019; 57:90-107. [PMID: 31331254 DOI: 10.1177/0300985819861708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Mammalian transmissible spongiform encephalopathies (TSEs) display marked activation of astrocytes and microglia that precedes neuronal loss. Investigation of clinical parallels between TSEs and other neurodegenerative protein misfolding diseases, such as Alzheimer's disease, has revealed similar patterns of neuroinflammatory responses to the accumulation of self-propagating amyloids. The contribution of glial activation to the progression of protein misfolding diseases is incompletely understood, with evidence for mediation of both protective and deleterious effects. Glial populations are heterogeneously distributed throughout the brain and capable of dynamic transitions along a spectrum of functional activation states between pro- and antiinflammatory polarization extremes. Using a murine model of Rocky Mountain Laboratory scrapie, the neuroinflammatory response to prion infection was characterized by evaluating glial activation across 15 brain regions over time and correlating it to traditional markers of prion neuropathology, including vacuolation and PrPSc deposition. Quantitative immunohistochemistry was used to evaluate glial expression of iNOS and Arg1, markers of classical and alternative glial activation, respectively. The results indicate progressive upregulation of iNOS in microglia and a mixed astrocytic profile featuring iNOS expression in white matter tracts and detection of Arg1-positive populations throughout the brain. These data establish a temporospatial lesion profile for this prion infection model and demonstrate evidence of multiple glial activation states.
Collapse
Affiliation(s)
- Alyona V Michael
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Justin J Greenlee
- US Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, IA, USA
| | - Tyler A Harm
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - S Jo Moore
- US Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, IA, USA
| | - Min Zhang
- Department of Statistics, College of Liberal Arts and Sciences, Iowa State University, Ames, IA, USA
| | - Melissa S Lind
- US Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, IA, USA.,BluePearl Pet Hospital, Des Moines, IA, USA
| | - M Heather West Greenlee
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Jodi D Smith
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| |
Collapse
|
22
|
Eyk CLV, Samaraweera SE, Scott A, Webber DL, Harvey DP, Mecinger O, O’Keefe LV, Cropley JE, Young P, Ho J, Suter C, Richards RI. Non-self mutation: double-stranded RNA elicits antiviral pathogenic response in a Drosophila model of expanded CAG repeat neurodegenerative diseases. Hum Mol Genet 2019; 28:3000-3012. [DOI: 10.1093/hmg/ddz096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/11/2019] [Accepted: 05/06/2019] [Indexed: 12/16/2022] Open
Abstract
Abstract
Inflammation is activated prior to symptoms in neurodegenerative diseases, providing a plausible pathogenic mechanism. Indeed, genetic and pharmacological ablation studies in animal models of several neurodegenerative diseases demonstrate that inflammation is required for pathology. However, while there is growing evidence that inflammation-mediated pathology may be the common mechanism underlying neurodegenerative diseases, including those due to dominantly inherited expanded repeats, the proximal causal agent is unknown. Expanded CAG.CUG repeat double-stranded RNA causes inflammation-mediated pathology when expressed in Drosophila. Repeat dsRNA is recognized by Dicer-2 as a foreign or ‘non-self’ molecule triggering both antiviral RNA and RNAi pathways. Neither of the RNAi pathway cofactors R2D2 nor loquacious are necessary, indicating antiviral RNA activation. RNA modification enables avoidance of recognition as ‘non-self’ by the innate inflammatory surveillance system. Human ADAR1 edits RNA conferring ‘self’ status and when co-expressed with expanded CAG.CUG dsRNA in Drosophila the pathology is lost. Cricket Paralysis Virus protein CrPV-1A is a known antagonist of Argonaute-2 in Drosophila antiviral defense. CrPV-1A co-expression also rescues pathogenesis, confirming anti-viral-RNA response. Repeat expansion mutation therefore confers ‘non-self’ recognition of endogenous RNA, thereby providing a proximal, autoinflammatory trigger for expanded repeat neurodegenerative diseases.
Collapse
Affiliation(s)
- Clare L van Eyk
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Saumya E Samaraweera
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Andrew Scott
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Dani L Webber
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - David P Harvey
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Olivia Mecinger
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Louise V O’Keefe
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Jennifer E Cropley
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Paul Young
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Joshua Ho
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Catherine Suter
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Robert I Richards
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| |
Collapse
|
23
|
Striebel JF, Race B, Williams K, Carroll JA, Klingeborn M, Chesebro B. Microglia are not required for prion-induced retinal photoreceptor degeneration. Acta Neuropathol Commun 2019; 7:48. [PMID: 30909963 PMCID: PMC6432762 DOI: 10.1186/s40478-019-0702-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/16/2019] [Indexed: 12/28/2022] Open
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.
Collapse
|
24
|
Orrù CD, Soldau K, Cordano C, Llibre-Guerra J, Green AJ, Sanchez H, Groveman BR, Edland SD, Safar JG, Lin JH, Caughey B, Geschwind MD, Sigurdson CJ. Prion Seeds Distribute throughout the Eyes of Sporadic Creutzfeldt-Jakob Disease Patients. mBio 2018; 9:e02095-18. [PMID: 30459197 PMCID: PMC6247090 DOI: 10.1128/mbio.02095-18] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022] Open
Abstract
Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common prion disease in humans and has been iatrogenically transmitted through corneal graft transplantation. Approximately 40% of sCJD patients develop visual or oculomotor symptoms and may seek ophthalmological consultation. Here we used the highly sensitive real-time quaking-induced conversion (RT-QuIC) assay to measure postmortem prion seeding activities in cornea, lens, ocular fluid, retina, choroid, sclera, optic nerve, and extraocular muscle in the largest series of sCJD patient eyes studied by any assay to date. We detected prion seeding activity in 100% of sCJD eyes, representing three common sCJD subtypes, with levels varying by up to 4 log-fold among individuals. The retina consistently showed the highest seed levels, which in some cases were only slightly lower than brain. Within the retina, prion deposits were detected by immunohistochemistry (IHC) in the retinal outer plexiform layer in most sCJD cases, and in some eyes the inner plexiform layer, consistent with synaptic prion deposition. Prions were not detected by IHC in any other eye region. With RT-QuIC, prion seed levels generally declined in eye tissues with increased distance from the brain, and yet all corneas had prion seeds detectable. Prion seeds were also present in the optic nerve, extraocular muscle, choroid, lens, vitreous, and sclera. Collectively, these results reveal that sCJD patients accumulate prion seeds throughout the eye, indicating the potential diagnostic utility as well as a possible biohazard.IMPORTANCE Cases of iatrogenic prion disease have been reported from corneal transplants, yet the distribution and levels of prions throughout the eye remain unknown. This study probes the occurrence, level, and distribution of prions in the eyes of patients with sporadic Creutzfeldt-Jakob disease (sCJD). We tested the largest series of prion-infected eyes reported to date using an ultrasensitive technique to establish the prion seed levels in eight regions of the eye. All 11 cases had detectable prion seeds in the eye, and in some cases, the seed levels in the retina approached those in brain. In most cases, prion deposits could also be seen by immunohistochemical staining of retinal tissue; other ocular tissues were negative. Our results have implications for estimating the risk for iatrogenic transmission of sCJD as well as for the development of antemortem diagnostic tests for prion diseases.
Collapse
Affiliation(s)
- Christina D Orrù
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, Montana, USA
| | - Katrin Soldau
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Christian Cordano
- Department of Neurology, Multiple Sclerosis Center, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Jorge Llibre-Guerra
- Cognitive and Behavioral Research Unit, National Institute of Neurology, Havana, Cuba
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Ari J Green
- Department of Neurology, Multiple Sclerosis Center, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Henry Sanchez
- Department of Pathology, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Bradley R Groveman
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, Montana, USA
| | - Steven D Edland
- Department of Family Medicine & Public Health, University of California, San Diego, La Jolla, California, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Jiri G Safar
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jonathan H Lin
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, Montana, USA
| | - Michael D Geschwind
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Christina J Sigurdson
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
- Department of Pathology, Immunology, and Microbiology, University of California, Davis, Davis, California, USA
| |
Collapse
|
25
|
Mammadova N, Summers CM, Kokemuller RD, He Q, Ding S, Baron T, Yu C, Valentine RJ, Sakaguchi DS, Kanthasamy AG, Greenlee JJ, Heather West Greenlee M. Accelerated accumulation of retinal α-synuclein (pSer129) and tau, neuroinflammation, and autophagic dysregulation in a seeded mouse model of Parkinson's disease. Neurobiol Dis 2018; 121:1-16. [PMID: 30218757 DOI: 10.1016/j.nbd.2018.09.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/05/2018] [Accepted: 09/11/2018] [Indexed: 01/08/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by accumulation of misfolded α-synuclein within the central nervous system (CNS). Visual problems in PD patients are common, although retinal pathology associated with PD is not well understood. The purpose of this study was to investigate retinal pathology in a transgenic mouse model (TgM83) expressing the human A53T α-synuclein mutation and assess the effect of α-synuclein "seeding" on the development of retinal pathology. Two-month-old TgM83 mice were intracerebrally inoculated with brain homogenate from old (12-18 months) TgM83 mice. Retinas were then analyzed at 5 months of age. We analyzed retinas from 5-month-old and 8-month-old uninoculated healthy TgM83 mice, and old (12-18 months) mice that were euthanized following the development of clinical signs. Retinas of B6C3H mice (genetic background of the TgM83 mouse) served as control. We used immunohistochemistry and western blot analysis to detect accumulation of α-synuclein, pTauThr231, inflammation, changes in macroautophagy, and cell death. Raman spectroscopy was used to test the potential to differentiate between retinal tissues of healthy mice and diseased mice. This work demonstrates retinal changes associated with the A53T mutation. Retinas of non-inoculated TgM83 mice had accumulation of α-synuclein, "pre-tangle" tau, activation of retinal glial cells, and photoreceptor cell loss by 8 months of age. The development of these changes is accelerated by inoculation with brain homogenate from clinically ill TgM83 mice. Compared to non-inoculated 5-month-old TgM83 mice, retinas of inoculated 5-month-old mice had increased accumulation of α-synuclein (pSer129) and pTauThr231 proteins, upregulated microglial activation, and dysregulated macroautophagy. Raman spectroscopic analysis was able to discriminate between healthy and diseased mice. This study describes retinal pathology resulting from the A53T mutation. We show that seeding with brain homogenates from old TgM83 mice accelerates retinal pathology. We demonstrate that Raman spectroscopy can be used to accurately identify a diseased retina based on its biochemical profile, and that α-synuclein accumulation may contribute to accumulation of pTauThr231 proteins, neuroinflammation, metabolic dysregulation, and photoreceptor cell death. Our work provides insight into retinal changes associated with Parkinson's disease, and may contribute to a better understanding of visual symptoms experienced by patients.
Collapse
Affiliation(s)
- Najiba Mammadova
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, United States; Immunobiology Graduate Program, Iowa State University, United States; Neuroscience Graduate Program, Iowa State University, United States.
| | - Corey M Summers
- Immunobiology Graduate Program, Iowa State University, United States; Department of Kinesiology, Iowa State University, United States.
| | - Robyn D Kokemuller
- Neuroscience Graduate Program, Iowa State University, United States; Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, United States; Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.
| | - Qing He
- Department of Agriculture and Biosystems Engineering, Iowa State University, Ames, IA, United States.
| | - Shaowei Ding
- Department of Mechanical Engineering, Iowa State University, Ames, IA, United States.
| | - Thierry Baron
- Anses, Laboratoire de Lyon, Unité Maladies Neurodégénératives, Lyon, France.
| | - Chenxu Yu
- Department of Agriculture and Biosystems Engineering, Iowa State University, Ames, IA, United States.
| | - Rudy J Valentine
- Immunobiology Graduate Program, Iowa State University, United States; Department of Kinesiology, Iowa State University, United States.
| | - Donald S Sakaguchi
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, United States; Neuroscience Graduate Program, Iowa State University, United States.
| | - Anumantha G Kanthasamy
- Immunobiology Graduate Program, Iowa State University, United States; Neuroscience Graduate Program, Iowa State University, United States; Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.
| | - Justin J Greenlee
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, United States.
| | - M Heather West Greenlee
- Immunobiology Graduate Program, Iowa State University, United States; Neuroscience Graduate Program, Iowa State University, United States; Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.
| |
Collapse
|
26
|
Richards RI, Robertson SA, Kastner DL. Neurodegenerative diseases have genetic hallmarks of autoinflammatory disease. Hum Mol Genet 2018; 27:R108-R118. [PMID: 29684205 PMCID: PMC6061832 DOI: 10.1093/hmg/ddy139] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 03/12/2018] [Accepted: 04/16/2018] [Indexed: 12/25/2022] Open
Abstract
The notion that one common pathogenic pathway could account for the various clinically distinguishable, typically late-onset neurodegenerative diseases might appear unlikely given the plethora of diverse primary causes of neurodegeneration. On the contrary, an autoinflammatory pathogenic mechanism allows diverse genetic and environmental factors to converge into a common chain of causality. Inflammation has long been known to correlate with neurodegeneration. Until recently this relationship was seen as one of consequence rather than cause-with inflammatory cells and events acting to 'clean up the mess' after neurological injury. This explanation is demonstrably inadequate and it is now clear that inflammation is at the very least, rate-limiting for neurodegeneration (and more likely, a principal underlying cause in most if not all neurodegenerative diseases), protective in its initial acute phase, but pernicious in its latter chronic phase.
Collapse
Affiliation(s)
- Robert I Richards
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Sarah A Robertson
- Robinson Research Institute, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Daniel L Kastner
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Inflammatory response of microglia to prions is controlled by sialylation of PrP Sc. Sci Rep 2018; 8:11326. [PMID: 30054538 PMCID: PMC6063910 DOI: 10.1038/s41598-018-29720-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
Neuroinflammation is recognized as one of the obligatory pathogenic features of neurodegenerative diseases including Alzheimer’s, Parkinson’s or prion diseases. In prion diseases, space and time correlations between deposition of disease-associated, pathogenic form of the prion protein or PrPSc and microglial-mediated neuroinflammation has been established. Yet, it remains unclear whether activation of microglia is triggered directly by a contact with PrPSc, and what molecular features of PrPSc microglia sense and respond to that drive microglia to inflammatory states. The current study asked the questions whether PrPSc can directly trigger activation of microglia and whether the degree of microglia response depends on the nature of terminal carbohydrate groups on the surface of PrPSc particles. PrPSc was purified from brains of mice infected with mouse-adapted prion strain 22L or neuroblastoma N2a cells stably infected with 22L. BV2 microglial cells or primary microglia were cultured in the presence of purified 22L. We found that exposure of BV2 cells or primary microglia to purified PrPSc triggered proinflammatory responses characterized by an increase in the levels of TNFα, IL6, nitric oxide (NO) and expression of inducible Nitric Oxide Synthase (iNOS). Very similar patterns of inflammatory response were induced by PrPSc purified from mouse brains and neuroblastoma cells arguing that microglia response is independent of the source of PrPSc. To test whether the microglial response is mediated by carbohydrate epitopes on PrPSc surface, the levels of sialylation of PrPSc N-linked glycans was altered by treatment of purified PrPSc with neuraminidase. Partial cleavage of sialic acid residues was found to boost the inflammatory response of microglia to PrPSc. Moreover, transient degradation of Iκβα observed upon treatment with partially desialylated PrPSc suggests that canonical NFκB activation pathway is involved in inflammatory response. The current study is the first to demonstrate that PrPSc can directly trigger inflammatory response in microglia. In addition, this work provides direct evidence that the chemical nature of the carbohydrate groups on PrPSc surface is important for microglial activation.
Collapse
|
28
|
Hwang S, West Greenlee MH, Balkema-Buschmann A, Groschup MH, Nicholson EM, Greenlee JJ. Real-Time Quaking-Induced Conversion Detection of Bovine Spongiform Encephalopathy Prions in a Subclinical Steer. Front Vet Sci 2018; 4:242. [PMID: 29404344 PMCID: PMC5780402 DOI: 10.3389/fvets.2017.00242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/20/2017] [Indexed: 01/05/2023] Open
Abstract
Bovine spongiform encephalopathy (BSE) belongs to a group of fatal prion diseases that result from the misfolding of the cellular prion protein (PrPC) into a pathogenic form (PrPSc) that accumulates in the brain. In vitro assays such as serial protein misfolding amplification and real-time quaking-induced conversion (RT-QuIC) allow assessment of the conversion of PrPC to PrPSc. RT-QuIC can be used for the detection of prions in a variety of biological tissues from humans and animals. However, there is no such comparison of RT-QuIC data between BSE positive and presymptomatic cattle. Further, the current study assesses prion distribution in multiple brain regions of clinically ill or subclinical animals. Here, we compare RT-QuIC reactions seeded with brain samples collected from experimentally inoculated cattle that were clinically ill or subclinically affected with BSE. The results demonstrate RT-QuIC seeding in various brain regions of an animal with subclinical BSE despite being determined negative by immunohistochemistry. Bioassay of the subclinical animal and RT-QuIC of brainstem from inoculated knockout (PRNP-/-) cattle were used to confirm infectivity in the subclinical animal and determine that RT-QuIC reactions were not the result of residual inoculum, respectively. These results confirm that RT-QuIC is a highly sensitive prion detection assay that can detect prions in a steer prior to the onset of clinical signs of BSE.
Collapse
Affiliation(s)
- Soyoun Hwang
- United States Department of Agriculture, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, IA, United States
| | - M Heather West Greenlee
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - Anne Balkema-Buschmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Martin H Groschup
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Eric M Nicholson
- United States Department of Agriculture, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, IA, United States
| | - Justin J Greenlee
- United States Department of Agriculture, Agricultural Research Service, National Animal Disease Center, Virus and Prion Research Unit, Ames, IA, United States
| |
Collapse
|
29
|
Manne S, Kondru N, Nichols T, Lehmkuhl A, Thomsen B, Main R, Halbur P, Dutta S, Kanthasamy AG. Ante-mortem detection of chronic wasting disease in recto-anal mucosa-associated lymphoid tissues from elk (Cervus elaphus nelsoni) using real-time quaking-induced conversion (RT-QuIC) assay: A blinded collaborative study. Prion 2017; 11:415-430. [PMID: 29098931 DOI: 10.1080/19336896.2017.1368936] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Prion diseases are transmissible spongiform encephalopathies (TSEs) characterized by fatal, progressive neurologic diseases with prolonged incubation periods and an accumulation of infectious misfolded prion proteins. Antemortem diagnosis is often difficult due to a long asymptomatic incubation period, differences in the pathogenesis of different prions, and the presence of very low levels of infectious prion in easily accessible samples. Chronic wasting disease (CWD) is a TSE affecting both wild and captive populations of cervids, including mule deer, white-tailed deer, elk, moose, muntjac, and most recently, wild reindeer. This study represents a well-controlled evaluation of a newly developed real-time quaking-induced conversion (RT-QuIC) assay as a potential CWD diagnostic screening test using rectal biopsy sections from a depopulated elk herd. We evaluated 69 blinded samples of recto-anal mucosa-associated lymphoid tissue (RAMALT) obtained from USDA Veterinary Services. The results were later un-blinded and statistically compared to immunohistochemical (IHC) results from the USDA National Veterinary Services Laboratories (NVSL) for RAMALT, obex, and medial retropharyngeal lymph node (MRPLN). Comparison of RAMALT RT-QuIC assay results with the IHC results of RAMALT revealed 92% relative sensitivity (95% confidence limits: 61.52-99.8%) and 95% relative specificity (95% confidence limits: 85.13-99%). Collectively, our results show a potential utility of the RT-QuIC assay to advance the development of a rapid, sensitive, and specific prion diagnostic assay for CWD prions.
Collapse
Affiliation(s)
- Sireesha Manne
- a Department of Biomedical Sciences , College of Veterinary Medicine, Iowa State University , Ames , IA , USA
| | - Naveen Kondru
- a Department of Biomedical Sciences , College of Veterinary Medicine, Iowa State University , Ames , IA , USA
| | - Tracy Nichols
- b United States Department of Agriculture (USDA) , National Wildlife Research Center, Wildlife Services , Fort Collins , CO , USA
| | - Aaron Lehmkuhl
- c USDA, National Veterinary Services Laboratories (NVSL), Veterinary Services , Ames , IA , USA
| | - Bruce Thomsen
- c USDA, National Veterinary Services Laboratories (NVSL), Veterinary Services , Ames , IA , USA
| | - Rodger Main
- d Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine , Iowa State University , Ames , IA , USA
| | - Patrick Halbur
- d Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine , Iowa State University , Ames , IA , USA
| | - Somak Dutta
- e Department of Statistics , Iowa State University , Ames , IA , USA
| | - Anumantha G Kanthasamy
- a Department of Biomedical Sciences , College of Veterinary Medicine, Iowa State University , Ames , IA , USA
| |
Collapse
|
30
|
Moore SJ, West Greenlee MH, Kondru N, Manne S, Smith JD, Kunkle RA, Kanthasamy A, Greenlee JJ. Experimental Transmission of the Chronic Wasting Disease Agent to Swine after Oral or Intracranial Inoculation. J Virol 2017; 91:e00926-17. [PMID: 28701407 PMCID: PMC5599732 DOI: 10.1128/jvi.00926-17] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/06/2017] [Indexed: 01/01/2023] Open
Abstract
Chronic wasting disease (CWD) is a naturally occurring, fatal neurodegenerative disease of cervids. The potential for swine to serve as hosts for the agent of CWD is unknown. The purpose of this study was to investigate the susceptibility of swine to the CWD agent following experimental oral or intracranial inoculation. Crossbred piglets were assigned to three groups, intracranially inoculated (n = 20), orally inoculated (n = 19), and noninoculated (n = 9). At approximately the age at which commercial pigs reach market weight, half of the pigs in each group were culled ("market weight" groups). The remaining pigs ("aged" groups) were allowed to incubate for up to 73 months postinoculation (mpi). Tissues collected at necropsy were examined for disease-associated prion protein (PrPSc) by Western blotting (WB), antigen capture enzyme immunoassay (EIA), immunohistochemistry (IHC), and in vitro real-time quaking-induced conversion (RT-QuIC). Brain samples from selected pigs were also bioassayed in mice expressing porcine prion protein. Four intracranially inoculated aged pigs and one orally inoculated aged pig were positive by EIA, IHC, and/or WB. By RT-QuIC, PrPSc was detected in lymphoid and/or brain tissue from one or more pigs in each inoculated group. The bioassay was positive in four out of five pigs assayed. This study demonstrates that pigs can support low-level amplification of CWD prions, although the species barrier to CWD infection is relatively high. However, detection of infectivity in orally inoculated pigs with a mouse bioassay raises the possibility that naturally exposed pigs could act as a reservoir of CWD infectivity.IMPORTANCE We challenged domestic swine with the chronic wasting disease agent by inoculation directly into the brain (intracranially) or by oral gavage (orally). Disease-associated prion protein (PrPSc) was detected in brain and lymphoid tissues from intracranially and orally inoculated pigs as early as 8 months of age (6 months postinoculation). Only one pig developed clinical neurologic signs suggestive of prion disease. The amount of PrPSc in the brains and lymphoid tissues of positive pigs was small, especially in orally inoculated pigs. Regardless, positive results obtained with orally inoculated pigs suggest that it may be possible for swine to serve as a reservoir for prion disease under natural conditions.
Collapse
Affiliation(s)
- S Jo Moore
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - M Heather West Greenlee
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa, USA
| | - Naveen Kondru
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa, USA
| | - Sireesha Manne
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa, USA
| | - Jodi D Smith
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, USA
| | - Robert A Kunkle
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, USA
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa, USA
| | - Justin J Greenlee
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, Iowa, USA
| |
Collapse
|
31
|
Asthana A, Baksi S, Ashok A, Karmakar S, Mammadova N, Kokemuller R, Greenlee MH, Kong Q, Singh N. Prion protein facilitates retinal iron uptake and is cleaved at the β-site: Implications for retinal iron homeostasis in prion disorders. Sci Rep 2017; 7:9600. [PMID: 28851903 PMCID: PMC5575325 DOI: 10.1038/s41598-017-08821-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/17/2017] [Indexed: 12/22/2022] Open
Abstract
Prion disease-associated retinal degeneration is attributed to PrP-scrapie (PrPSc), a misfolded isoform of prion protein (PrPC) that accumulates in the neuroretina. However, a lack of temporal and spatial correlation between PrPSc and cytotoxicity suggests the contribution of host factors. We report retinal iron dyshomeostasis as one such factor. PrPC is expressed on the basolateral membrane of retinal-pigment-epithelial (RPE) cells, where it mediates uptake of iron by the neuroretina. Accordingly, the neuroretina of PrP-knock-out mice is iron-deficient. In RPE19 cells, silencing of PrPC decreases ferritin while over-expression upregulates ferritin and divalent-metal-transporter-1 (DMT-1), indicating PrPC-mediated iron uptake through DMT-1. Polarization of RPE19 cells results in upregulation of ferritin by ~10-fold and β-cleavage of PrPC, the latter likely to block further uptake of iron due to cleavage of the ferrireductase domain. A similar β-cleavage of PrPC is observed in mouse retinal lysates. Scrapie infection causes PrPSc accumulation and microglial activation, and surprisingly, upregulation of transferrin despite increased levels of ferritin. Notably, detergent-insoluble ferritin accumulates in RPE cells and correlates temporally with microglial activation, not PrPSc accumulation, suggesting that impaired uptake of iron by PrPSc combined with inflammation results in retinal iron-dyshomeostasis, a potentially toxic host response contributing to prion disease-associated pathology.
Collapse
Affiliation(s)
- Abhishek Asthana
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Shounak Baksi
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Ajay Ashok
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Shilpita Karmakar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Najiba Mammadova
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa, 50010, USA
| | - Robyn Kokemuller
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa, 50010, USA
| | - Mary Heather Greenlee
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa, 50010, USA
| | - Qingzhong Kong
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Neena Singh
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA.
| |
Collapse
|
32
|
Kondru N, Manne S, Greenlee J, West Greenlee H, Anantharam V, Halbur P, Kanthasamy A, Kanthasamy A. Integrated Organotypic Slice Cultures and RT-QuIC (OSCAR) Assay: Implications for Translational Discovery in Protein Misfolding Diseases. Sci Rep 2017; 7:43155. [PMID: 28233859 PMCID: PMC5324099 DOI: 10.1038/srep43155] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/19/2017] [Indexed: 01/13/2023] Open
Abstract
Protein misfolding is a key pathological event in neurodegenerative diseases like prion diseases, synucleinopathies, and tauopathies that are collectively termed protein misfolding disorders. Prions are a prototypic model to study protein aggregation biology and therapeutic development. Attempts to develop anti-prion therapeutics have been impeded by the lack of screening models that faithfully replicate prion diseases and the lack of rapid, sensitive biological screening systems. Therefore, a sensitive model encompassing prion replication and neurotoxicity would be indispensable to the pursuit of intervention strategies. We present an ultra-sensitive screening system coupled to an ex vivo prion organotypic slice culture model to rapidly advance rationale-based high-throughput therapeutic strategies. This hybrid Organotypic Slice Culture Assay coupled with RT-QuIC (OSCAR) permits sensitive, specific and quantitative detection of prions from an infectious slice culture model on a reduced time scale. We demonstrate that the anti-prion activity of test compounds can be readily resolved based on the power and kinetics of seeding activity in the OSCAR screening platform and that the prions generated in slice cultures are biologically active. Collectively, our results imply that OSCAR is a robust model of prion diseases that offers a promising platform for understanding prion proteinopathies and advancing anti-prion therapeutics.
Collapse
Affiliation(s)
- Naveen Kondru
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Sireesha Manne
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Justin Greenlee
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50011, USA
| | - Heather West Greenlee
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Patrick Halbur
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|