Traumatic Brain Injury Induces Tau Aggregation and Spreading

George Edwards 3rd, Jing Zhao, Pramod K Dash, Claudio Soto, Ines Moreno-Gonzalez, George Edwards 3rd, Jing Zhao, Pramod K Dash, Claudio Soto, Ines Moreno-Gonzalez

Abstract

The misfolding and aggregation of tau protein into neurofibrillary tangles is the main underlying hallmark of tauopathies. Most tauopathies have a sporadic origin and can be associated with multiple risk factors. Traumatic brain injury (TBI) has been suggested as a risk factor for tauopathies by triggering disease onset and facilitating its progression. Several studies indicate that TBI seems to be a risk factor to development of Alzheimer disease and chronic traumatic encephalopathy, because there is a relationship of TBI severity and propensity to development of these illnesses. In this study, we evaluated whether moderate to severe TBI can trigger the initial formation of pathological tau that would induce the development of the pathology throughout the brain. To this end, we subjected tau transgenic mice to TBI and assessed tau phosphorylation and aggregation pattern to create a spatial heat map of tau deposition and spreading in the brain. Our results suggest that brain injured tau transgenic mice have an accelerated tau pathology in different brain regions that increases over time compared with sham mice. The appearance of pathological tau occurs in regions distant to the injury area that are connected synaptically, suggesting dissemination of tau aggregates. Overall, this work posits TBI as a risk factor for tauopathies through the induction of tau hyperphosphorylation and aggregation.

Keywords: Alzheimer disease; chronic traumatic encephalopathy; risk factor; tau aggregation; traumatic brain injury.

Conflict of interest statement

No competing financial interests exist.

Figures

FIG. 1.
FIG. 1.
Tau aggregation is induced one day after traumatic brain injury (TBI). Tau hyperphosphorylation was assessed in brain homogenates and coronal tissue sections in three-month-old P301S TBI and sham induced mice one day after the event. (A) Representative bright field microscopy images of AT8 immunoreactivity in TBI and sham mice brain sections. (B) The AT8 burden quantification was obtained by analyzing the percentage of the area reactive with AT8 antibody in relation to the total area analyzed in the cortex/amygdala, hippocampal area, and brainstem. Statistical analysis was performed by Student t test or Mann-Whitney test for the ipsilateral (Ipsi) and contralateral (Contra) areas of impact as well as overall brain region. Ipsilateral and contralateral hippocampus quantifications were analyzed by Mann-Whitney test. (C) Semi-quantitative evaluation (lowest [0] to highest [5]) of tau deposition is captured in the heat map at different distance from the impacted area. The star denotes the estimated placement of impact. (D,E) Insoluble tau levels were evaluated by serial extraction and enzyme-linked immunosorbent assay performed in the formic acid fraction of the brains. Graphs in panels B, D, and E show the mean ± standard error of the mean of the n = 5 animals analyzed per group. Scale bar: 100 μm. *p < 0.05, **p < 0.01, ***p < 0.001.
FIG. 2.
FIG. 2.
Spatial pattern and burden of pathological tau one week after traumatic brain injury (TBI). The P301S mice having either a TBI or sham surgery were assessed for tau aggregate formation and spreading by AT8 immunostaining. (A) Representative images of the TBI and sham animals using AT8 antibody are displayed. (B) The AT8 burden was evaluated in the cortex, hippocampal area, and brainstem in overall, ipsilateral (Ipsi), and contralateral (Contra) side of impact. Ipsilateral and contralateral hippocampus were evaluated by Mann-Whitney test, while all other data underwent Student t test. (C) Four coronal areas per animal were assessed to re-create a heat map to navigate tau spreading after TBI. Heat map scale: lowest (0) to highest (5) score. The star denotes the estimated placement of impact. Scale bar: 100 μm. *p < 0.05, **p < 0.01.
FIG. 3.
FIG. 3.
Tau aggregation is significantly increased two months after traumatic brain injury (TBI) injury. (A) Illustrative microphotography of AT8 immunopositive staining visually displayed growing pathologic tau aggregation. (B) The AT8 burden quantification in the overall, ipsilateral (Ipsi), and contralateral (Contra) impacted area in the three main brain regions analyzed. Ipsilateral cortex and Contra hippocampus were evaluated by Mann-Whitney test, while all other statistical analysis was performed by Student t test. (C) The spatial heat map revealed increasing tau burden and presence of pathogenic tau aggregates in multiple areas in TBI-induced mice having more aggressive pathology compared with sham mice. Scale bar: 100 μm. Heat map scale: lowest (0) to highest (5) score. The star denotes the estimated placement of impact. *p < 0.05, **p < 0.01.
FIG. 4.
FIG. 4.
Traumatic brain injury (TBI) promotes behavior impairment in transgenic tau mice in the chronic stage of injury. Barnes maze test was performed on P301S TBI, P301S sham, and wild-type (WT) littermate TBI mice six months after controlled cortical impact injury. The P301S TBI mice exhibited attenuated learning and memory capabilities observed by the learning curve (A) and long term memory assessment (B) *p < 0.05, **p < 0.01, ***p < 0.001.
FIG. 5.
FIG. 5.
Robust tau pathology six months after traumatic brain injury (TBI). (A) Example AT8 microscope images illustrate substantial tau pathology in TBI mice six months after injury compared with age-matched sham mice. (B) The AT8 burden quantification shows a significant overall increase of tau deposition in TBI mice compared with sham mice in the ipsilateral (Ipsi), contralateral (Contra), and overall cortex and hippocampal area. Statistics for brainstem were calculated by Mann-Whitney test, while all other were performed by Student t test. (C) The AT8 burden scores exhibited in a spatial heat map of four rostrocaudal areas in TBI versus sham mice. Heat map scale: lowest (0) to highest (5) score. Star denotes the estimated placement of impact. Scale bar: 100 μm. *p < 0.05.
FIG. 6.
FIG. 6.
Altered tropism of tau deposition after traumatic brain injury (TBI). Representative figures of pathological tau deposition as evaluated by AT8 immunohistochemistry in hippocampus, cortex, and cerebellar lobes in mice six months after TBI. Age-matched P301S sham animals revealed very low detectability using AT8 at the different times analyzed. Scale bar: 100 μm; insets: 30 μm. CA1, cornu ammonis 1 area; cc, corpus callosum; DG, dentate gyrus; Ctx, cortex; Hp, hippocampus; ml, molecular layer, gl, granular layer; wm, white matter.
FIG. 7.
FIG. 7.
Traumatic brain injury (TBI)-induced tau aggregates fold-change summary. The AT8 burden fold-change is represented in the overall, ipsilateral (Ipsi) and contralateral (Contra) side of impact in the hippocampal area (HPa), cortex/amygdala, and brainstem (BS) in relation to the respective brain areas rostrocaudally to the impacted side in TBI versus sham induced mice. The colored scale exhibits the lowest to highest fold change compared with sham brains. The fold-change values displayed in red were analyzed by Mann-Whitney test while all other data analysis was performed by Student t test. The values below the fold change display the lower to upper 95% confidence interval of the mean of the TBI (top) and sham (bottom) groups. *p < 0.05.

References

    1. Götz J. (2001). Tau and transgenic animal models. Brain Res. Brain Res. Rev. 35, 266–286
    1. Lee V.M., Goedert M., and Trojanowski J.Q. (2001). Neurodegenerative tauopathies. Annu. Rev. Neurosci. 24, 1121–1159
    1. Soto C. (2003). Unfolding the role of protein misfolding in neurodegenerative diseases. Nat. Rev. Neurosci. 4, 49–60
    1. Moreno-Gonzalez I. and Soto C. (2011). Misfolded protein aggregates: mechanisms, structures and potential for disease transmission. Semin. Cell Dev. Biol. 22, 482–487
    1. Hyman B.T., Augustinack J.C., and Ingelsson M. (2005). Transcriptional and conformational changes of the tau molecule in Alzheimer's disease. Biochim. Biophys. Acta 1739, 150–157
    1. Braak H. and Braak E. (1995). Staging of Alzheimer's disease-related neurofibrillary changes. Neurobiol. Aging 16, 271–278
    1. Braak H., Thal D.R., Ghebremedhin E., and Del Tredici K. (2011). Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J. Neuropathol. Exp. Neurol. 70, 960–969
    1. Braak H. and Del Tredici K. (2012). Where, when, and in what form does sporadic Alzheimer's disease begin? Curr. Opin. Neurol. 25, 708–714
    1. McKee A.C., Stein T.D., Kiernan P.T., and Alvarez V.E. (2015). The neuropathology of chronic traumatic encephalopathy. Brain Pathol. 25, 350–364
    1. National Center for Injury Prevention and Conrol. (2003). Report to Congress on Mild Traumatic Brain Injury in the United States: Steps to Prevent a Serious Public Health Problem. Centers for Disease Control and Prevention: Atlanta, GA
    1. Masel B.E. and DeWitt D.S. (2010). Traumatic brain injury: a disease process, not an event. J. Neurotrauma 27, 1529–1540
    1. Blennow K., Brody D.L., Kochanek P.M., Levin H., McKee A., Ribbers G.M., Yaffe K., and Zetterberg H. (2016). Traumatic brain injuries. Nat. Rev. Dis. Primers 2, 16084.
    1. Walker K.R. and Tesco G. (2013). Molecular mechanisms of cognitive dysfunction following traumatic brain injury. Front. Aging Neurosci. 5, 29.
    1. Blennow K., Hardy J., and Zetterberg H. (2012). The neuropathology and neurobiology of traumatic brain injury. Neuron 76, 886–899
    1. Edwards G., Moreno-Gonzalez I., and Soto C. (2017). Amyloid-beta and tau pathology following repetitive mild traumatic brain injury. Biochem. Biophys. Res. Commun. 483, 1137–1142
    1. Sivanandam T.M. and Thakur M.K. (2012). Traumatic brain injury: a risk factor for Alzheimer's disease. Neurosci. Biobehav. Rev. 36, 1376–1381
    1. Mendez M.F., Paholpak P., Lin A., Zhang J.Y., and Teng E. (2015). Prevalence of traumatic brain injury in early versus late-onset Alzheimer's disease. J. Alzheimers. Dis. 47, 985–993
    1. Gilbert M., Snyder C., Corcoran C., Norton M.C., Lyketsos C.G., and Tschanz J.T. (2014). The association of traumatic brain injury with rate of progression of cognitive and functional impairment in a population-based cohort of Alzheimer's disease: the Cache County Dementia Progression Study. Int. Psychogeriatr. 26, 1593–1601
    1. Smith D.H., Uryu K., Saatman K.E., Trojanowski J.Q., and McIntosh T.K. (2003). Protein accumulation in traumatic brain injury. Neuromolecular Med. 4, 59–72
    1. Grady M.S., McLaughlin M.R., Christman C.W., Valadka A.B., Fligner C.L., and Povlishock J.T. (1993). The use of antibodies targeted against the neurofilament subunits for the detection of diffuse axonal injury in humans. J. Neuropathol. Exp. Neurol. 52, 143–152
    1. Johnson V.E., Stewart W., and Smith D.H. (2012). Widespread tau and amyloid-beta pathology many years after a single traumatic brain injury in humans. Brain Pathol. 22, 142–149
    1. Sundman M.H., Hall E.E., and Chen N.K. (2014). Examining the relationship between head trauma and neurodegenerative disease: a review of epidemiology, pathology and neuroimaging techniques. J. Alzheimers Dis. Parkinsonism 4
    1. Kriegel J., Papadopoulos Z., and McKee A.C. (2018). Chronic traumatic encephalopathy: is latency in symptom onset explained by tau propagation? Cold Spring Harb. Perspect. Med. 8
    1. Mez J., Daneshvar D.H., Kiernan P.T., Abdolmohammadi B., Alvarez V.E., Huber B.R., Alosco M.L., Solomon T.M., Nowinski C.J., McHale L., Cormier K.A., Kubilus C.A., Martin B.M., Murphy L., Baugh C.M., Montenigro P.H., Chaisson C.E., Tripodis Y., Kowall N.W., Weuve J., McClean M.D., Cantu R.C., Goldstein L.E., Katz D.I., Stern R.A., Stein T.D., and McKee A.C. (2017). Clinicopathological evaluation of chronic traumatic encephalopathy in players of American football. JAMA 318, 360–370
    1. Frost B. and Diamond M.I. (2009). The expanding realm of prion phenomena in neurodegenerative disease. Prion 3, 74–77
    1. Frost B. and Diamond M.I. (2010). Prion-like mechanisms in neurodegenerative diseases. Nat. Rev. Neurosci. 11, 155–159
    1. Holmes B.B., and Diamond M.I. (2014). Prion-like properties of Tau protein: the importance of extracellular Tau as a therapeutic target. J. Biol. Chem. 289, 19855–19861
    1. Holmes B.B., Furman J.L., Mahan T.E., Yamasaki T.R., Mirbaha H., Eades W.C., Belaygorod L., Cairns N.J., Holtzman D.M., and Diamond M.I. (2014). Proteopathic tau seeding predicts tauopathy in vivo. Proc. Natl. Acad. Sci. U. S. A. 111, E4376–E4385
    1. Sanders D.W., Kaufman S.K., DeVos S.L., Sharma A.M., Mirbaha H., Li A., Barker S.J., Foley A.C., Thorpe J.R., Serpell L.C., Miller T.M., Grinberg L.T., Seeley W.W., and Diamond M.I. (2014). Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 82, 1271–1288
    1. Jucker M. and Walker L.C. (2011). Pathogenic protein seeding in Alzheimer disease and other neurodegenerative disorders. Ann. Neurol. 70, 532–540
    1. Morales R., Callegari K., and Soto C. (2015). Prion-like features of misfolded Aβ and tau aggregates. Virus Res. 207, 106–112
    1. Walker L.C., Diamond M.I., Duff K.E., and Hyman B.T. (2013). Mechanisms of protein seeding in neurodegenerative diseases. JAMA Neurol. 70, 304–310
    1. Morales R., Moreno-Gonzalez I., and Soto C. (2013). Cross-seeding of misfolded proteins: implications for etiology and pathogenesis of protein misfolding diseases. PLoS Pathog. 9, e1003537.
    1. Clavaguera F., Bolmont T., Crowther R.A., Abramowski D., Frank S., Probst A., Fraser G., Stalder A.K., Beibel M., Staufenbiel M., Jucker M., Goedert M., and Tolnay M. (2009). Transmission and spreading of tauopathy in transgenic mouse brain. Nat. Cell Biol. 11, 909–913
    1. Clavaguera F., Akatsu H., Fraser G., Crowther R.A., Frank S., Hench J., Probst A., Winkler D.T., Reichwald J., Staufenbiel M., Ghetti B., Goedert M., and Tolnay M. (2013). Brain homogenates from human tauopathies induce tau inclusions in mouse brain. Proc. Natl. Acad. Sci. U. S. A. 110, 9535–9540
    1. Lasagna-Reeves C.A., Castillo-Carranza D.L., Sengupta U., Guerrero-Munoz M.J., Kiritoshi T., Neugebauer V., Jackson G.R., and Kayed R. (2012). Alzheimer brain-derived tau oligomers propagate pathology from endogenous tau. Sci. Rep. 2, 700.
    1. Iba M., Guo J.L., McBride J.D., Zhang B., Trojanowski J.Q., and Lee V.M. (2013). Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer's-like tauopathy. J. Neurosci. 33, 1024–1037
    1. Hoover B.R., Reed M.N., Su J., Penrod R.D., Kotilinek L.A., Grant M.K., Pitstick R., Carlson G.A., Lanier L.M., Yuan L.L., Ashe K.H., and Liao D. (2010). Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron 68, 1067–1081
    1. Sohn P.D., Tracy T.E., Son H.I., Zhou Y., Leite R.E., Miller B.L., Seeley W.W., Grinberg L.T., and Gan L. (2016). Acetylated tau destabilizes the cytoskeleton in the axon initial segment and is mislocalized to the somatodendritic compartment. Mol. Neurodegener. 11, 47.
    1. Iba M., McBride J.D., Guo J.L., Zhang B., Trojanowski J.Q., and Lee V.M. (2015). Tau pathology spread in PS19 tau transgenic mice following locus coeruleus (LC) injections of synthetic tau fibrils is determined by the LC's afferent and efferent connections. Acta Neuropathol. 130, 349–362
    1. Stancu I.C., Vasconcelos B., Ris L., Wang P., Villers A., Peeraer E., Buist A., Terwel D., Baatsen P., Oyelami T., Pierrot N., Casteels C., Bormans G., Kienlen-Campard P., Octave J.N., Moechars D., and Dewachter I. (2015). Templated misfolding of Tau by prion-like seeding along neuronal connections impairs neuronal network function and associated behavioral outcomes in Tau transgenic mice. Acta Neuropathol. 129, 875–894
    1. Ahmed Z., Cooper J., Murray T.K., Garn K., McNaughton E., Clarke H., Parhizkar S., Ward M.A., Cavallini A., Jackson S., Bose S., Clavaguera F., Tolnay M., Lavenir I., Goedert M., Hutton M.L., and O'Neill M.J. (2014). A novel in vivo model of tau propagation with rapid and progressive neurofibrillary tangle pathology: the pattern of spread is determined by connectivity, not proximity. Acta Neuropathol. 127, 667–683
    1. Vandrovcova J., Anaya F., Kay V., Lees A., Hardy J., and de Silva R. (2010). Disentangling the role of the tau gene locus in sporadic tauopathies. Curr .Alzheimer Res. 7, 726–734
    1. Coria F., Rubio I., and Bayon C. (1994). Alzheimer's disease, beta-amyloidosis, and aging. Rev. Neurosci. 5, 275–292
    1. Yoshiyama Y., Higuchi M., Zhang B., Huang S.M., Iwata N., Saido T.C., Maeda J., Suhara T., Trojanowski J.Q., and Lee V.M. (2007). Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53, 337–351
    1. Zhao J., Hylin M.J., Kobori N., Hood K.N., Moore A.N., and Dash P.K. (2017). Post-injury administration of galantamine reduces traumatic brain injury pathology and improves outcome. J. Neurotrauma 35, 362–374
    1. Osier N.D. and Dixon C.E. (2016). The controlled cortical impact model: applications, considerations for researchers, and future directions. Front. Neurol. 7, 134.
    1. Dixon C.E., Clifton G.L., Lighthall J.W., Yaghmai A.A., and Hayes R.L. (1991). A controlled cortical impact model of traumatic brain injury in the rat. J. Neurosci. Methods 39, 253–262
    1. Moreno-Gonzalez I., Edwards III G., Salvadores N., Shahnawaz M., Diaz-Espinoza R., and Soto C. (2017). Molecular interaction between type 2 diabetes and Alzheimer's disease through cross-seeding of protein misfolding. Mol. Psychiatry. 22, 1327–1334
    1. Moreno-Gonzalez I., Estrada L.D., Sanchez-Mejias E., and Soto C. (2013). Smoking exacerbates amyloid pathology in a mouse model of Alzheimer's disease. Nat. Commun. 4, 1495.
    1. Moreno-Gonzalez I., Baglietto-Vargas D., Sanchez-Varo R., Jimenez S., Trujillo-Estrada L., Sanchez-Mejias E., Del Rio J.C., Torres M., Romero-Acebal M., Ruano D., Vizuete M., Vitorica J., and Gutierrez A. (2009). Extracellular amyloid-β and cytotoxic glial activation induce significant entorhinal neuron loss in young PS1M146L/APP751SL mice. J. Alzheimers Dis. 18, 755–776
    1. Jimenez S., Baglietto-Vargas D., Caballero C., Moreno-Gonzalez I., Torres M., Sanchez-Varo R., Ruano D., Vizuete M., Gutierrez A., and Vitorica J. (2008). Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer's disease: age-dependent switch in the microglial phenotype from alternative to classic. J. Neurosci. 28, 11650–11661
    1. Hurtado D.E., Molina-Porcel L., Iba M., Aboagye A.K., Paul S.M., Trojanowski J.Q., and Lee V.M. (2010). Aβ accelerates the spatiotemporal progression of tau pathology and augments tau amyloidosis in an Alzheimer mouse model. Am. J. Pathol. 177, 1977–1988
    1. Duran-Aniotz C., Morales R., Moreno-Gonzalez I., Hu P.P., Fedynyshyn J., and Soto C. (2014). Aggregate-depleted brain fails to induce abeta deposition in a mouse model of Alzheimer's disease. PLoS One 9, e89014.
    1. Duran-Aniotz C., Morales R., Moreno-Gonzalez I., Hu P.P., and Soto C. (2013). Brains from non-Alzheimer's individuals containing amyloid deposits accelerate Abeta deposition in vivo. Acta Neuropathol. Commun. 1, 76.
    1. Tran H.T., Sanchez L., Esparza T.J., and Brody D.L. (2011). Distinct temporal and anatomical distributions of amyloid-β and tau abnormalities following controlled cortical impact in transgenic mice. PLoS One 6, e25475.
    1. Hoshino S., Tamaoka A., Takahashi M., Kobayashi S., Furukawa T., Oaki Y., Mori O., Matsuno S., Shoji S., Inomata M., and Teramoto A. (1998). Emergence of immunoreactivities for phosphorylated tau and amyloid-β protein in chronic stage of fluid percussion injury in rat brain. Neuroreport 9, 1879–1883
    1. Lv Q., Lan W., Sun W., Ye R., Fan X., Ma M., Yin Q., Jiang Y., Xu G., Dai J., Guo R., and Liu X. (2014). Intranasal nerve growth factor attenuates tau phosphorylation in brain after traumatic brain injury in rats. J. Neurol. Sci. 345, 48–55
    1. Goldstein L., Fisher A., Tagge C., Zhang X.-L., Velisek L., Sullivan J., Upreti C., Kracht J., Ericsson M., Wojnarowicz M., Goletiani C., Maglakelidze G., Casey N., Moncaster J., Minaeva O., Moir R., Nowinski C., Stern R., Cantu R., Geiling J., Blusztajn J., Wolozin B., Ikezu T., Budson A., Kowall N.W., Chargin D., Sharon A., Saman S., Hall G., Moss W., Cleveland R., Tanzi R., Stanton P., and McKee A. (2012). Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci. Transl. Med. 4, 134ra60
    1. Tran H.T., LaFerla F.M., Holtzman D.M., and Brody D.L. (2011). Controlled cortical impact traumatic brain injury in 3xTg-AD mice causes acute intra-axonal amyloid- accumulation and independently accelerates the development of tau abnormalities. J. Neurosci. 31, 9513–9525
    1. Sawmiller D., Li S., Shahaduzzaman M., Smith A.J., Obregon D., Giunta B., Borlongan C. V., Sanberg P.R., and Tan J. (2014). Luteolin reduces Alzheimer's disease pathologies induced by traumatic brain injury. Int. J. Mol. Sci. 15, 895–904
    1. Jankowsky J.L. and Zheng H. (2017). Practical considerations for choosing a mouse model of Alzheimer's disease. Mol. Neurodegener. 12, 89.
    1. Brendel M., Jaworska A., Probst F., Overhoff F., Korzhova V., Lindner S., Carlsen J., Bartenstein P., Harada R., Kudo Y., Haass C., Van Leuven F., Okamura N., Herms J., and Rominger A. (2016). Small-animal PET imaging of tau pathology with 18F-THK5117 in 2 transgenic mouse models. J. Nucl. Med. 57, 792–798
    1. Furman J.L., Vaquer-Alicea J., White C.L., Cairns N.J., Nelson P.T., and Diamond M.I. (2017). Widespread tau seeding activity at early Braak stages. Acta Neuropathol. 133. 91–100
    1. Guo J.L. and Lee V.M. (2011). Seeding of normal Tau by pathological Tau conformers drives pathogenesis of Alzheimer-like tangles. J. Biol. Chem. 286, 15317–15331
    1. De C.A., Polydoro M., Suarez-Calvet M., William C., Adamowicz D.H., Kopeikina K.J., Pitstick R., Sahara N., Ashe K.H., Carlson G.A., Spires-Jones T.L., and Hyman B.T. (2012). Propagation of tau pathology in a model of early Alzheimer's disease. Neuron 73, 685–697
    1. Soto C. (2012). Transmissible proteins: expanding the prion heresy. Cell 149, 968–977
    1. Kaufman S.K., Sanders D.W., Thomas T.L., Ruchinskas A.J., Vaquer-Alicea J., Sharma A.M., Miller T.M., and Diamond M.I. (2016). Tau prion strains dictate patterns of cell pathology, progression rate, and regional vulnerability in vivo. Neuron 92, 796–812
    1. Walker L.C. (2016). Proteopathic strains and the heterogeneity of neurodegenerative diseases. Annu. Rev. Genet. 50, 329–346
    1. Arendt T., Stieler J.T., and Holzer M. (2016). Tau and tauopathies. Brain Res. Bull. 126, 238–292

Source: PubMed

3
Abonneren