29 results on '"Kurti, Aishe"'
Search Results
2. Loss of clusterin shifts amyloid deposition to the cerebrovasculature via disruption of perivascular drainage pathways
- Author
-
Wojtas, Aleksandra M., Kang, Silvia S., Olley, Benjamin M., Gatherer, Maureen, Shinohara, Mitsuru, Lozano, Patricia A., Liu, Chia-Chen, Kurti, Aishe, Baker, Kelsey E., Dickson, Dennis W., Yue, Mei, Petrucelli, Leonard, Bu, Guojun, Carare, Roxana O., and Fryer, John D.
- Published
- 2017
3. C90RF72 repeat expansions in mice cause TDP-43 pathology, neuronal loss, and behavioral deficits
- Author
-
Chew, Jeannie, Gendron, Tania F., Prudencio, Mercedes, Sasaguri, Hiroki, Zhang, Yong-Jie, Castanedes-Casey, Monica, Lee, Chris W., Jansen-West, Karen, Kurti, Aishe, Murray, Melissa E., Bieniek, Kevin F., Bauer, Peter O., Whitelaw, Ena C., Rousseau, Linda, Stankowski, Jeannette N., Stetler, Caroline, Daughrity, Lillian M., Perkerson, Emilie A., Desaro, Pamela, Johnston, Amelia, Overstreet, Karen, Edbauer, Dieter, Rademakers, Rosa, Boylan, Kevin B., Dickson, Dennis W., Fryer, John D., and Petrucelli, Leonard
- Published
- 2015
4. APOE genotype modifies benefits of calorie restriction against aging.
- Author
-
Qiao, Wenhui, Chen, Yixing, Ren, Yingxue, Ikezu, Tadafumi C, Johnson, Stephen, Li, Fuyao, Chen, Kai, Pan, Meixia, Kashyap, Sonu, Bueser, Katrina Rae, Wetmore, Allison, Kurti, Aishe I., Meneses, Axel D., Owens, Jada A., Chini, Eduardo N., Chen, Jun, Han, Xianlin, Bu, Guojun, and Zhao, Na
- Published
- 2024
- Full Text
- View/download PDF
5. Investigating the Pathogenic Interplay of Alpha-Synuclein, Tau, and Amyloid Beta in Lewy Body Dementia: Insights from Viral-Mediated Overexpression in Transgenic Mouse Models.
- Author
-
Lim, Melina J., Boschen, Suelen L., Kurti, Aishe, Castanedes Casey, Monica, Phillips, Virginia R., Fryer, John D., Dickson, Dennis, Jansen-West, Karen R., Petrucelli, Leonard, Delenclos, Marion, and McLean, Pamela J.
- Subjects
LEWY body dementia ,TRANSGENIC mice ,TAU proteins ,ALPHA-synuclein ,LABORATORY mice - Abstract
Lewy body dementia (LBD) is an often misdiagnosed and mistreated neurodegenerative disorder clinically characterized by the emergence of neuropsychiatric symptoms followed by motor impairment. LBD falls within an undefined range between Alzheimer's disease (AD) and Parkinson's disease (PD) due to the potential pathogenic synergistic effects of tau, beta-amyloid (Aβ), and alpha-synuclein (αsyn). A lack of reliable and relevant animal models hinders the elucidation of the molecular characteristics and phenotypic consequences of these interactions. Here, the goal was to evaluate whether the viral-mediated overexpression of αsyn in adult hTau and APP/PS1 mice or the overexpression of tau in Line 61 hThy1-αsyn mice resulted in pathology and behavior resembling LBD. The transgenes were injected intravenously via the tail vein using AAV-PHP.eB in 3-month-old hThy1-αsyn, hTau, or APP/PS1 mice that were then aged to 6-, 9-, and 12-months-old for subsequent phenotypic and histological characterization. Although we achieved the widespread expression of αsyn in hTau and tau in hThy1-αsyn mice, no αsyn pathology in hTau mice and only mild tau pathology in hThy1-αsyn mice was observed. Additionally, cognitive, motor, and limbic behavior phenotypes were not affected by overexpression of the transgenes. Furthermore, our APP/PS1 mice experienced premature deaths starting at 3 months post-injection (MPI), therefore precluding further analyses at later time points. An evaluation of the remaining 3-MPI indicated no αsyn pathology or cognitive and motor behavioral changes. Taken together, we conclude that the overexpression of αsyn in hTau and APP/PS1 mice and tau in hThy1-αsyn mice does not recapitulate the behavioral and neuropathological phenotypes observed in LBD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. APOE2 Exacerbates TDP‐43 Related Toxicity in the Absence of Alzheimer Pathology.
- Author
-
Meneses, Axel D., Koga, Shunsuke, Li, Zonghua, O'Leary, Justin, Li, Fuyao, Chen, Kai, Murakami, Aya, Qiao, Wenhui, Kurti, Aishe, Heckman, Michael G., White, Launia, Xie, Manling, Chen, Yixing, Finch, Nicole A., Lim, Melina J., Delenclos, Marion, DeTure, Michael A., Linares, Cynthia, Martin, Nicholas B., and Ikezu, Tadafumi C.
- Subjects
MOTOR neuron diseases ,FRONTOTEMPORAL lobar degeneration ,AMYOTROPHIC lateral sclerosis ,ALZHEIMER'S disease ,MOTOR cortex - Abstract
Objective: Recent evidence supports a link between increased TDP‐43 burden and the presence of an APOE4 gene allele in Alzheimer's disease (AD); however, it is difficult to conclude the direct effect of APOE on TDP‐43 pathology due to the presence of mixed AD pathologies. The goal of this study is to address how APOE isoforms impact TDP‐43 pathology and related neurodegeneration in the absence of typical AD pathologies. Methods: We overexpressed human TDP‐43 via viral transduction in humanized APOE2, APOE3, APOE4 mice, and murine Apoe‐knockout (Apoe‐KO) mice. Behavior tests were performed across ages. Animals were harvested at 11 months of age and TDP‐43 overexpression‐related neurodegeneration and gliosis were assessed. To further address the human relevance, we analyzed the association of APOE with TDP‐43 pathology in 160 postmortem brains from autopsy‐confirmed amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with motor neuron disease (FTLD‐MND) in the Mayo Clinic Brain Bank. Results: We found that TDP‐43 overexpression induced motor function deficits, neuronal loss, and gliosis in the motor cortex, especially in APOE2 mice, with much milder or absent effects in APOE3, APOE4, or Apoe‐KO mice. In the motor cortex of the ALS and FTLD‐MND postmortem human brains, we found that the APOE2 allele was associated with more severe TDP‐43‐positive dystrophic neurites. Interpretation: Our data suggest a genotype‐specific effect of APOE on TDP‐43 proteinopathy and neurodegeneration in the absence of AD pathology, with the strongest association seen with APOE2. ANN NEUROL 2023;93:830–843 [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Tau deposition drives neuropathological, inflammatory and behavioral abnormalities independently of neuronal loss in a novel mouse model
- Author
-
Cook, Casey, Kang, Silvia S., Carlomagno, Yari, Lin, Wen-Lang, Yue, Mei, Kurti, Aishe, Shinohara, Mitsuru, Jansen-West, Karen, Perkerson, Emilie, Castanedes-Casey, Monica, Rousseau, Linda, Phillips, Virginia, Bu, Guojun, Dickson, Dennis W., Petrucelli, Leonard, and Fryer, John D.
- Published
- 2015
- Full Text
- View/download PDF
8. Two FTD-ALS genes converge on the endosomal pathway to induce TDP-43 pathology and degeneration.
- Author
-
Wei Shao, Todd, Tiffany W., Yanwei Wu, Jones, Caroline Y., Jimei Tong, Jansen-West, Karen, Daughrity, Lillian M., Jinyoung Park, Yuka Koike, Kurti, Aishe, Mei Yue, Castanedes-Casey, Monica, Rosso, Giulia del, Dunmore, Judith A., Alepuz, Desiree Zanetti, Oskarsson, Björn, Dickson, Dennis W., Cook, Casey N., Prudencio, Mercedes, and Gendron, Tania F.
- Published
- 2022
- Full Text
- View/download PDF
9. Amyloid pathology and cognitive impairment in hAβ-KI and APPSAA-KI mouse models of Alzheimer's disease.
- Author
-
Lu, Wenyan, Shue, Francis, Kurti, Aishe, Jeevaratnam, Suren, Macyczko, Jesse R., Roy, Bhaskar, Izhar, Taha, Wang, Ni, Bu, Guojun, Kanekiyo, Takahisa, and Li, Yonghe
- Subjects
- *
AMYLOID plaque , *ALZHEIMER'S disease , *COGNITION disorders , *COGNITIVE ability , *AMYLOID - Abstract
The hAβ-KI and APPSAA-KI are two amyloid models that harbor mutations in the endogenous mouse App gene. Both hAβ-KI and APPSAA-KI mice contain a humanized Aβ sequence, and APPSAA-KI mice carry three additional familial AD mutations. We herein report that the Aβ levels and Aβ42/Aβ40 ratio in APPSAA-KI homozygotes are dramatically higher than those in hAβ-KI homozygotes at 14 months of age. In addition, APPSAA-KI mice display a widespread distribution of amyloid plaques in the brain, whereas the plaques are undetectable in hAβ-KI mice. Moreover, there are no sex differences in amyloid pathology in APPSAA-KI mice. Both APPSAA-KI and hAβ-KI mice exhibit cognitive impairments, wherein no significant differences are found between these two models, although APPSAA KI mice show a trend towards worse cognitive function. Notably, female hAβ-KI and APPSAA-KI mice have a more pronounced cognitive impairments compared to their respective males. Our findings suggest that Aβ humanization contributes to cognitive deficits in APPSAA-KI mice, and that amyloid deposition might not be closely associated with cognitive impairments in APPSAA-KI mice. • Aβ levels in APPSAA-KI mice are dramatically higher than those in hAβ-KI mice. • Aβ42/Aβ40 ratio in APPSAA-KI mice is higher than those in hAβ-KI mice. • No sex differences in Aβ levels and amyloid plaque burden in APPSAA-KI mice. • Insoluble Aβ levels are higher in male than female hAβ-KI mice. • Aβ humanization contributes to cognitive deficits in APPSAA-KI mice. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
10. Loss of Tmem106b leads to cerebellum Purkinje cell death and motor deficits.
- Author
-
Rademakers, Rosa, Nicholson, Alexandra M., Ren, Yingxue, Koga, Shunsuke, Nguyen, Hung Phuoc, Brooks, Mieu, Qiao, Wenhui, Quicksall, Zachary S., Matchett, Billie, Perkerson, Ralph B., Kurti, Aishe, Castanedes‐Casey, Monica, Phillips, Virginia, Librero, Ariston L, Fernandez De Castro, Cristhoper H., Baker, Matthew C., Roemer, Shanu F., Murray, Melissa E., Asmann, Yan, and Fryer, John D.
- Subjects
PURKINJE cells ,CELL death ,CEREBELLUM ,CEREBELLAR cortex ,GENOME-wide association studies ,FRONTOTEMPORAL lobar degeneration - Abstract
Since cerebellar Purkinje cells are known to play an important role in controlling balance, we next investigated motor function in aged I Tmem106b-/-, Tmem106b+/-, i and WT mice (15 months of age) using a dowel test. We next examined Purkinje cell numbers in younger mice; however, no significant difference in terms of Purkinje cell number was observed between I Tmem106b-/- i and WT mice at 3 and 8 months of age (Figure 1B, C). Taken together, these results are in line with Purkinje cell loss in I Tmem106b-/- i cerebellum and further implicate inflammation in Tmem106b deficiency-induced Purkinje cell loss. [Extracted from the article]
- Published
- 2021
- Full Text
- View/download PDF
11. APOE2 is associated with longevity independent of Alzheimer's disease.
- Author
-
Mitsuru Shinohara, Takahisa Kanekiyo, Masaya Tachibana, Kurti, Aishe, Motoko Shinohara, Yuan Fu, Jing Zhao, Xianlin Han, Sullivan, Patrick M., Rebeck, G. William, Fryer, John D., Heckman, Michael G., and Guojun Bu
- Published
- 2020
- Full Text
- View/download PDF
12. Loss of Tmem106b exacerbates FTLD pathologies and causes motor deficits in progranulin‐deficient mice.
- Author
-
Zhou, Xiaolai, Brooks, Mieu, Jiang, Peizhou, Koga, Shunsuke, Zuberi, Aamir R, Baker, Matthew C, Parsons, Tammee M, Castanedes‐Casey, Monica, Phillips, Virginia, Librero, Ariston L, Kurti, Aishe, Fryer, John D, Bu, Guojun, Lutz, Cathleen, Dickson, Dennis W, and Rademakers, Rosa
- Abstract
Progranulin (PGRN) and transmembrane protein 106B (TMEM106B) are important lysosomal proteins implicated in frontotemporal lobar degeneration (FTLD) and other neurodegenerative disorders. Loss‐of‐function mutations in progranulin (GRN) are a common cause of FTLD, while TMEM106B variants have been shown to act as disease modifiers in FTLD. Overexpression of TMEM106B leads to lysosomal dysfunction, while loss of Tmem106b ameliorates lysosomal and FTLD‐related pathologies in young Grn−/− mice, suggesting that lowering TMEM106B might be an attractive strategy for therapeutic treatment of FTLD‐GRN. Here, we generate and characterize older Tmem106b−/−Grn−/− double knockout mice, which unexpectedly show severe motor deficits and spinal cord motor neuron and myelin loss, leading to paralysis and premature death at 11–12 months. Compared to Grn−/−, Tmem106b−/−Grn−/− mice have exacerbated FTLD‐related pathologies, including microgliosis, astrogliosis, ubiquitin, and phospho‐Tdp43 inclusions, as well as worsening of lysosomal and autophagic deficits. Our findings confirm a functional interaction between Tmem106b and Pgrn and underscore the need to rethink whether modulating TMEM106B levels is a viable therapeutic strategy. Synopsis: Loss of TMEM106B has been suggested as therapeutic treatment of FTLD‐GRN. This study shows that loss of Tmem106b exacerbates FTLD pathologies and causes spinal cord motor neuron loss and myelin degeneration leading to motor deficits in Grn deficient mice. Loss of Tmem106b leads to severe motor deficits and premature death in Grn deficient mice.Loss of Tmem106b leads to spinal cord motor neuron loss and myelin degeneration in Grn deficient mice.Loss of Tmem106b exacerbates FTLD‐related pathologies, including microgliosis, astrogliosis, and ubiquitin and phospho‐Tdp43 inclusions in Grn deficient mice.Loss of Tmem106b exacerbates lysosomal and autophagic deficits in Grn deficient mice. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
13. APOE4 exacerbates α-synuclein pathology and related toxicity independent of amyloid.
- Author
-
Zhao, Na, Attrebi, Olivia N., Ren, Yingxue, Qiao, Wenhui, Sonustun, Berkiye, Martens, Yuka A., Meneses, Axel D., Li, Fuyao, Shue, Francis, Zheng, Jiaying, Van Ingelgom, Alexandra J., Davis, Mary D., Kurti, Aishe, Knight, Joshua A., Linares, Cynthia, Chen, Yixing, Delenclos, Marion, Liu, Chia-Chen, Fryer, John D., and Asmann, Yan W.
- Subjects
APOLIPOPROTEIN E4 ,LEWY body dementia ,DISEASE risk factors ,PARKINSON'S disease ,PATHOLOGY ,APOLIPOPROTEIN E - Abstract
APOE4 beyond amyloid: Although several genetic risk factors for neurodegenerative disorders have been identified, often the mechanistic aspect is not clear. Now, Zhao et al. and Davis et al. investigated whether apolipoprotein E4 (APOE4) genotype, a major genetic risk factor for neurodegenerative diseases, affected α-synuclein pathology in mouse models and Parkinson's disease (PD) patients. Zhao et al. generated a mouse model of α-synucleinopathy and showed that APOE4 exacerbated α-synuclein pathology in the absence of amyloid. Davis et al. used a mouse model of PD and analyzed cognition in patients with PD to demonstrate that APOE4 directly regulated α-synuclein pathology and was associated with faster cognitive decline. These results provide insight into the mechanisms linking APOE genotype to neurodegenerative disorders. The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease mainly by driving amyloid-β pathology. Recently, APOE4 has also been found to be a genetic risk factor for Lewy body dementia (LBD), which includes dementia with Lewy bodies and Parkinson's disease dementia. How APOE4 drives risk of LBD and whether it has a direct effect on α-synuclein pathology are not clear. Here, we generated a mouse model of synucleinopathy using an adeno-associated virus gene delivery of α-synuclein in human APOE-targeted replacement mice expressing APOE2, APOE3, or APOE4. We found that APOE4, but not APOE2 or APOE3, increased α-synuclein pathology, impaired behavioral performances, worsened neuronal and synaptic loss, and increased astrogliosis at 9 months of age. Transcriptomic profiling in APOE4-expressing α-synuclein mice highlighted altered lipid and energy metabolism and synapse-related pathways. We also observed an effect of APOE4 on α-synuclein pathology in human postmortem brains with LBD and minimal amyloid pathology. Our data demonstrate a pathogenic role of APOE4 in exacerbating α-synuclein pathology independent of amyloid, providing mechanistic insights into how APOE4 increases the risk of LBD. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
14. Behavioral and transcriptomic analysis of Trem2-null mice: not all knockout mice are created equal.
- Author
-
Kang, Silvia S., Kurti, Aishe, Baker, Kelsey E., Chia-Chen Liu, Colonna, Marco, Ulrich, Jason D., Holtzman, David M., Guojun Bu, and Fryer, John D.
- Published
- 2018
- Full Text
- View/download PDF
15. Neonatal AAV delivery of alpha-synuclein induces pathology in the adult mouse brain.
- Author
-
Delenclos, Marion, Faroqi, Ayman H., Mei Yue, Kurti, Aishe, Castanedes-Casey, Monica, Rousseau, Linda, Phillips, Virginia, Dickson, Dennis W., Fryer, John D., and McLean, Pamela J.
- Subjects
ALPHA-synuclein ,LEWY body dementia ,PARKINSONIAN disorders - Abstract
Abnormal accumulation of alpha-synuclein (asyn) is a pathological hallmark of Lewy body related disorders such as Parkinson's disease and Dementia with Lewy body disease. During the past two decades, a myriad of animal models have been developed to mimic pathological features of synucleinopathies by overexpressing human asyn. Although different strategies have been used, most models have little or no reliable and predictive phenotype. Novel animal models are a valuable tool for understanding neuronal pathology and to facilitate development of new therapeutics for these diseases. Here, we report the development and characterization of a novel model in which mice rapidly express wild-type asyn via somatic brain transgenesis mediated by adeno-associated virus (AAV). At 1, 3, and 6 months of age following intracerebroventricular (ICV) injection, mice were subjected to a battery of behavioral tests followed by pathological analyses of the brains. Remarkably, significant levels of asyn expression are detected throughout the brain as early as 1 month old, including olfactory bulb, hippocampus, thalamic regions and midbrain. Immunostaining with a phospho-asyn (pS129) specific antibody reveals abundant pS129 expression in specific regions. Also, pathologic asyn is detected using the disease specific antibody 5G4. However, this model did not recapitulate behavioral phenotypes characteristic of rodent models of synucleinopathies. In fact no deficits in motor function or cognition were observed at 3 or 6 months of age. Taken together, these findings show that transduction of neonatal mouse with AAV-asyn can successfully lead to rapid, whole brain transduction of wild-type human asyn, but increased levels of wildtype asyn do not induce behavior changes at an early time point (6 months), despite pathological changes in several neurons populations as early as 1 month. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
16. Identification of plexin A4 as a novel clusterin receptor links two Alzheimer's disease risk genes.
- Author
-
Kang, Silvia S., Kurti, Aishe, Wojtas, Aleksandra, Baker, Kelsey E., Liu, Chia-Chen, Kanekiyo, Takahisa, Deming, Yuetiva, Cruchaga, Carlos, Estus, Steven, Bu, Guojun, and Fryer, John D.
- Published
- 2016
- Full Text
- View/download PDF
17. ABCA7 Deficiency Accelerates Amyloid-β Generation and Alzheimer's Neuronal Pathology.
- Author
-
Sakae, Nobutaka, Chia-Chen Liu, Shinohara, Mitsuru, Frisch-Daiello, Jessica, Li Ma, Yu Yamazaki, Tachibana, Masaya, Younkin, Linda, Kurti, Aishe, Carrasquillo, Minerva M., Zou, Fanggeng, Sevlever, Daniel, Bisceglio, Gina, Gan, Ming, Fol, Romain, Knight, Patrick, Miao Wang, Xianlin Han, Fryer, John D., and Fitzgerald, Michael L.
- Subjects
ALZHEIMER'S disease ,AMYLOID ,PEPTIDES ,BRAIN ,PROTEIN precursors - Abstract
In Alzheimer's disease (AD), the accumulation and deposition of amyloid-β (Aβ) peptides in the brain is a central event. Aβ is cleaved from amyloid precursor protein (APP) byβ-secretase andγ-secretase mainly in neurons. Although mutations in APP, PS1, or PS2 cause early-onset familial AD, ABCA7 encoding ATP-binding cassette transporter A7 is one of the susceptibility genes for late-onset AD (LOAD), in which its loss-of-function variants increase the disease risk. ABCA7 is homologous to a major lipid transporter ABCA1 and is highly expressed in neurons and microglia in the brain. Here, we show that ABCA7 deficiency altered brain lipid profile and impaired memory in ABCA7 knock-out (Abca7
-/- ) mice. When bred to amyloid model APP/PS1 mice, plaque burden was exacerbated by ABCA7 deficit. In vivo microdialysis studies indicated that the clearance rate of Aβ was unaltered. Interestingly, ABCA7 deletion facilitated the processing of APP to Aβ by increasing the levels ofβ-site APP cleaving enzyme 1 (BACE1) and sterol regulatory element-binding protein 2 (SREBP2) in primary neurons and mouse brains. Knock-down of ABCA7 expression in neurons caused endoplasmic reticulum stress highlighted by increased level of protein kinase R-like endoplasmic reticulum kinase (PERK) and increased phosphorylation of eukaryotic initiation factor 2α (eIF2α). In the brains of APP/PS1;Abca7-/- mice, the level of phosphorylated extracellular regulated kinase (ERK) was also significantly elevated. Together, our results reveal novel pathways underlying the association of ABCA7 dysfunction and LOAD pathogenesis. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
18. Dietary intervention rescues maternal obesity induced behavior deficits and neuroinflammation in offspring.
- Author
-
Kang, Silvia S., Kurti, Aishe, Fair, Damien A., and Fryer, John D.
- Subjects
- *
AUTISM spectrum disorders , *HYPERACTIVITY , *ATTENTION-deficit hyperactivity disorder , *HIGH-fat diet , *OBESITY - Abstract
Obesity induces a low-grade inflammatory state and has been associated with behavioral and cognitive alterations. Importantly, maternal environmental insults can adversely impact subsequent offspring behavior and have been linked with neurodevelopmental disorders such as autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (AHDH). It is unknown if maternal obesity significantly alters offspring sociability, a key ASD feature, and if altering maternal diet will provide an efficacious intervention paradigm for behavioral deficits. Here we investigated the impact of maternal high fat diet (HFD) and maternal dietary intervention during lactation on offspring behavior and brain inflammation in mice. We found that maternal HFD increased anxiety and decreased sociability in female offspring. Additionally, female offspring from HFD-fed dams also exhibited increased brain IL-1β and TNFα and microglial activation. Importantly, maternal dietary intervention during lactation was sufficient to alleviate social deficits and brain inflammation. Maternal obesity during gestation alone was sufficient to increase hyperactivity in male offspring, a phenotype that was not ameliorated by dietary intervention. These data suggest that maternal HFD acts as a prenatal/perinatal insult that significantly impacts offspring behavior and inflammation and that dietary intervention during lactation may be an easily translatable, efficacious intervention to offset some of these manifestations. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
19. Clusterin ameliorates tau pathology in vivo by inhibiting fibril formation.
- Author
-
Wojtas, Aleksandra M., Carlomagno, Yari, Sens, Jonathon P., Kang, Silvia S., Jensen, Tanner D., Kurti, Aishe, Baker, Kelsey E., Berry, Taylor J., Phillips, Virginia R., Castanedes, Monica Casey, Awan, Ayesha, DeTure, Michael, De Castro, Cristhoper H. Fernandez, Librero, Ariston L., Yue, Mei, Daughrity, Lillian, Jansen-West, Karen R., Cook, Casey N., Dickson, Dennis W., and Petrucelli, Leonard
- Subjects
TAU proteins ,CLUSTERIN ,MOLECULAR chaperones ,ALZHEIMER'S disease ,PATHOLOGY - Abstract
The molecular chaperone Clusterin (CLU) impacts the amyloid pathway in Alzheimer's disease (AD) but its role in tau pathology is unknown. We observed CLU co-localization with tau aggregates in AD and primary tauopathies and CLU levels were upregulated in response to tau accumulation. To further elucidate the effect of CLU on tau pathology, we utilized a gene delivery approach in CLU knock-out (CLU KO) mice to drive expression of tau bearing the P301L mutation. We found that loss of CLU was associated with exacerbated tau pathology and anxiety-like behaviors in our mouse model of tauopathy. Additionally, we found that CLU dramatically inhibited tau fibrilization using an in vitro assay. Together, these results demonstrate that CLU plays a major role in both amyloid and tau pathologies in AD. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
20. Astrocyte-derived clusterin suppresses amyloid formation in vivo.
- Author
-
Wojtas, Aleksandra M., Sens, Jonathon P., Kang, Silvia S., Baker, Kelsey E., Berry, Taylor J., Kurti, Aishe, Daughrity, Lillian, Jansen-West, Karen R., Dickson, Dennis W., Petrucelli, Leonard, Bu, Guojun, Liu, Chia-Chen, and Fryer, John D.
- Subjects
CLUSTERIN ,ASTROCYTES ,AMYLOID ,AMYLOID beta-protein ,AMYLOID plaque ,ALZHEIMER'S disease ,HIPPOCAMPUS (Brain) - Abstract
Background: Accumulation of amyloid-β (Aβ) peptide in the brain is a pathological hallmark of Alzheimer's disease (AD). The clusterin (CLU) gene confers a risk for AD and CLU is highly upregulated in AD patients, with the common non-coding, protective CLU variants associated with increased expression. Although there is strong evidence implicating CLU in amyloid metabolism, the exact mechanism underlying the CLU involvement in AD is not fully understood or whether physiologic alterations of CLU levels in the brain would be protective. Results: We used a gene delivery approach to overexpress CLU in astrocytes, the major source of CLU expression in the brain. We found that CLU overexpression resulted in a significant reduction of total and fibrillar amyloid in both cortex and hippocampus in the APP/PS1 mouse model of AD amyloidosis. CLU overexpression also ameliorated amyloid-associated neurotoxicity and gliosis. To complement these overexpression studies, we also analyzed the effects of haploinsufficiency of Clu using heterozygous (Clu
+/− ) mice and control littermates in the APP/PS1 model. CLU reduction led to a substantial increase in the amyloid plaque load in both cortex and hippocampus in APP/PS1; Clu+/− mice compared to wild-type (APP/PS1; Clu+/+ ) littermate controls, with a concomitant increase in neuritic dystrophy and gliosis. Conclusions: Thus, both physiologic ~ 30% overexpression or ~ 50% reduction in CLU have substantial impacts on amyloid load and associated pathologies. Our results demonstrate that CLU plays a major role in Aβ accumulation in the brain and suggest that efforts aimed at CLU upregulation via pharmacological or gene delivery approaches offer a promising therapeutic strategy to regulate amyloid pathology. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
21. P4‐065: APOE4 CONDITIONALLY EXPRESSED IN CEREBROVASCULATURE IMPAIRS ENDOTHELIAL FUNCTIONS AND INDUCES COGNITIVE DEFICITS.
- Author
-
Yamazaki, Yu, Liu, Chia-Chen, Oue, Hiroshi, Kurti, Aishe, Yamazaki, Akari, Fryer, John D., Kanekiyo, Takahisa, and Bu, Guojun
- Published
- 2018
- Full Text
- View/download PDF
22. Aberrant deposition of stress granule-resident proteins linked to C9orf72-associated TDP-43 proteinopathy.
- Author
-
Chew, Jeannie, Cook, Casey, Gendron, Tania F., Jansen-West, Karen, del Rosso, Giulia, Daughrity, Lillian M., Castanedes-Casey, Monica, Kurti, Aishe, Stankowski, Jeannette N., Disney, Matthew D., Rothstein, Jeffrey D., Dickson, Dennis W., Fryer, John D., Zhang, Yong-Jie, and Petrucelli, Leonard
- Subjects
PROTEINS ,ADENO-associated virus ,FRONTOTEMPORAL dementia ,AMYOTROPHIC lateral sclerosis ,NEURODEGENERATION - Abstract
Background: A G
4 C2 hexanucleotide repeat expansion in the noncoding region of C9orf72 is the major genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis (c9FTD/ALS). Putative disease mechanisms underlying c9FTD/ALS include toxicity from sense G4 C2 and antisense G2 C4 repeat-containing RNA, and from dipeptide repeat (DPR) proteins unconventionally translated from these RNA products. Methods: Intracerebroventricular injections with adeno-associated virus (AAV) encoding 2 or 149 G4 C2 repeats were performed on postnatal day 0, followed by assessment of behavioral and neuropathological phenotypes. Results: Relative to control mice, gliosis and neurodegeneration accompanied by cognitive and motor deficits were observed in (G4 C2 )149 mice by 6 months of age. Recapitulating key pathological hallmarks, we also demonstrate that sense and antisense RNA foci, inclusions of poly(GA), poly(GP), poly(GR), poly(PR), and poly(PA) DPR proteins, and inclusions of endogenous phosphorylated TDP-43 (pTDP-43) developed in (G4 C2 )149 mice but not control (G4 C2 )2 mice. Notably, proteins that play a role in the regulation of stress granules – RNA-protein assemblies that form in response to translational inhibition and that have been implicated in c9FTD/ALS pathogenesis – were mislocalized in (G4 C2 )149 mice as early as 3 months of age. Specifically, we observed the abnormal deposition of stress granule components within inclusions immunopositive for poly(GR) and pTDP-43, as well as evidence of nucleocytoplasmic transport defects. Conclusions: Our in vivo model of c9FTD/ALS is the first to robustly recapitulate hallmark features derived from both sense and antisense C9orf72 repeat-associated transcripts complete with neurodegeneration and behavioral impairments. More importantly, the early appearance of persistent pathological stress granules prior to significant pTDP-43 deposition implicates an aberrant stress granule response as a key disease mechanism driving TDP-43 proteinopathy in c9FTD/ALS. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
23. Enhanced phosphorylation of T153 in soluble tau is a defining biochemical feature of the A152T tau risk variant.
- Author
-
Carlomagno, Yari, Chung, Dah-eun Chloe, Yue, Mei, Kurti, Aishe, Avendano, Nicole M., Castanedes-Casey, Monica, Hinkle, Kelly M., Jansen-West, Karen, Daughrity, Lillian M., Tong, Jimei, Phillips, Virginia, Rademakers, Rosa, DeTure, Michael, Fryer, John D., Dickson, Dennis W., Petrucelli, Leonard, and Cook, Casey
- Subjects
PHOSPHORYLATION ,GENETIC mutation ,TAU proteins - Abstract
Pathogenic mutations in the tau gene (microtubule associated protein tau, MAPT) are linked to the onset of tauopathy, but the A152T variant is unique in acting as a risk factor for a range of disorders including Alzheimer's disease (AD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and dementia with Lewy bodies (DLB). In order to provide insight into the mechanism by which A152T modulates disease risk, we developed a novel mouse model utilizing somatic brain transgenesis with adeno-associated virus (AAV) to drive tau expression in vivo, and validated the model by confirming the distinct biochemical features of A152T tau in postmortem brain tissue from human carriers. Specifically, Tau
A152T -AAV mice exhibited increased tau phosphorylation that unlike animals expressing the pathogenic P301L mutation remained localized to the soluble fraction. To investigate the possibility that the A152T variant might alter the phosphorylation state of tau on T152 or the neighboring T153 residue, we generated a novel antibody that revealed significant accumulation of soluble tau species that were hyperphosphorylated on T153 (pT153) in TauA152T -AAV mice, which were absent the soluble fraction of TauP301L -AAV mice. Providing new insight into the role of A152T in modifying risk of tauopathy, as well as validating the TauA152T -AAV model, we demonstrate that the presence of soluble pT153-positive tau species in human postmortem brain tissue differentiates A152T carriers from noncarriers, independent of disease classification. These results implicate both phosphorylation of T153 and an altered solubility profile in the mechanism by which A152T modulates disease risk. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
24. APOE ε2 is associated with increased tau pathology in primary tauopathy.
- Author
-
Zhao, Na, Liu, Chia-Chen, Van Ingelgom, Alexandra J., Linares, Cynthia, Kurti, Aishe, Knight, Joshua A., Heckman, Michael G., Diehl, Nancy N., Shinohara, Mitsuru, Martens, Yuka A., Attrebi, Olivia N., Petrucelli, Leonard, Fryer, John D., Wszolek, Zbigniew K., Graff-Radford, Neill R., Caselli, Richard J., Sanchez-Contreras, Monica Y., Rademakers, Rosa, Murray, Melissa E., and Koga, Shunsuke
- Abstract
Apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset Alzheimer’s disease mainly by modulating amyloid-β pathology. APOE ε4 is also shown to exacerbate neurodegeneration and neuroinflammation in a tau transgenic mouse model. To further evaluate the association of APOE genotype with the presence and severity of tau pathology, we express human tau via an adeno-associated virus gene delivery approach in human APOE targeted replacement mice. We find increased hyperphosphorylated tau species, tau aggregates, and behavioral abnormalities in mice expressing APOE ε2/ε2. We also show that in humans, the APOE ε2 allele is associated with increased tau pathology in the brains of progressive supranuclear palsy (PSP) cases. Finally, we identify an association between the APOE ε2/ε2 genotype and risk of tauopathies using two series of pathologically-confirmed cases of PSP and corticobasal degeneration. Our data together suggest APOE ε2 status may influence the risk and progression of tauopathy. The APOE ε4 allele is a strong genetic risk factor for Alzheimer’s disease, whereas the APOE ε2 allele is protective. Here the authors show that mice expressing the human APOE ε2/ε2 genotype have increased tau pathology and related behavioral deficits; they also find that the APOE ε2 allele is associated with an increased burden of tau pathology in postmortem human brains with progressive supranuclear palsy. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
25. Loss of Tmem106b is unable to ameliorate frontotemporal dementia-like phenotypes in an AAV mouse model of <italic>C9ORF72</italic>-repeat induced toxicity.
- Author
-
Nicholson, Alexandra M., Zhou, Xiaolai, Perkerson, Ralph B., Parsons, Tammee M., Chew, Jeannie, Brooks, Mieu, DeJesus-Hernandez, Mariely, Finch, NiCole A., Matchett, Billie J., Kurti, Aishe, Jansen-West, Karen R., Perkerson, Emilie, Daughrity, Lillian, Castanedes-Casey, Monica, Rousseau, Linda, Phillips, Virginia, Hu, Fenghua, Gendron, Tania F., Murray, Melissa E., and Dickson, Dennis W.
- Subjects
GENETIC mutation ,FRONTOTEMPORAL dementia ,PHENOTYPES ,HUMAN phenotype - Abstract
Loss-of-function mutations in progranulin (
GRN ) and a non-coding (GGGGCC)n hexanucleotide repeat expansions inC9ORF72 are the two most common genetic causes of frontotemporal lobar degeneration with aggregates of TAR DNA binding protein 43 (FTLD-TDP).TMEM106B encodes a type II transmembrane protein with unknown function. Genetic variants inTMEM106B associated with reduced TMEM106B levels have been identified as disease modifiers in individuals withGRN mutations andC9ORF72 expansions. Recently, loss of Tmem106b has been reported to protect the FTLD-like phenotypes inGrn−/− mice. Here, we generatedTmem106b−/− mice and examined whether loss of Tmem106b could rescue FTLD-like phenotypes in an AAV mouse model ofC9ORF72 -repeat induced toxicity. Our results showed that neither partial nor complete loss of Tmem106b was able to rescue behavioral deficits induced by the expression of (GGGGCC)66 repeats (66R). Loss of Tmem106b also failed to ameliorate 66R-induced RNA foci, dipeptide repeat protein formation and pTDP-43 pathological burden. We further found that complete loss of Tmem106b increased astrogliosis, even in the absence of 66R, and failed to rescue 66R-induced neuronal cell loss, whereas partial loss of Tmem106b significantly rescued the neuronal cell loss but not neuroinflammation induced by 66R. Finally, we showed that overexpression of 66R did not alter expression ofTmem106b and other lysosomal genes in vivo, and subsequent analyses in vitro found that transiently knocking downC9ORF72 , but not overexpression of 66R, significantly increased TMEM106B and other lysosomal proteins. In summary, reducing Tmem106b levels failed to rescue FTLD-like phenotypes in a mouse model mimicking the toxic gain-of-functions associated with overexpression of 66R. Combined with the observation that loss of C9ORF72 and not 66R overexpression was associated with increased levels of TMEM106B, this work suggests that the protectiveTMEM106B haplotype may exert its effect in expansion carriers by counteracting lysosomal dysfunction resulting from a loss of C9ORF72. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
26. THE MOLECULAR CHAPERONE BRICHOS INHIBITS Aβ AGGREGATION AND OTHER NEUROPATHOLOGICAL PHENOTYPES IN A MOUSE MODEL OF Aβ AMYLOIDOSIS.
- Author
-
Kim, Jungsu, Kim, Chaeyoung, Dolfe, Lisa, Belmonte, Krystal, Flores, Luis, Kurti, Aishe, Fryer, John D., Presto, Jenny, and Johansson, Jan
- Published
- 2017
- Full Text
- View/download PDF
27. Severe amygdala dysfunction in a MAPT transgenic mouse model of frontotemporal dementia.
- Author
-
Cook, Casey, Dunmore, Judy H., Murray, Melissa E., Scheffel, Kristyn, Shukoor, Nawsheen, Tong, Jimei, Castanedes-Casey, Monica, Phillips, Virginia, Rousseau, Linda, Penuliar, Michael S., Kurti, Aishe, Dickson, Dennis W., Petrucelli, Leonard, and Fryer, John D.
- Subjects
- *
AMYGDALOID body , *TRANSGENIC mice , *FRONTOTEMPORAL dementia , *PARKINSONIAN disorders , *CHROMOSOMES , *NEURODEGENERATION - Abstract
Abstract: Frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17) is a neurodegenerative tauopathy caused by mutations in the tau gene (MAPT). Individuals with FTDP-17 have deficits in learning, memory, and language, in addition to personality and behavioral changes that are often characterized by a lack of social inhibition. Several transgenic mouse models expressing tau mutations have been tested extensively for memory or motor impairments, though reports of amygdala-dependent behaviors are lacking. To this end, we tested the rTg4510 mouse model on a behavioral battery that included amygdala-dependent tasks of exploration. As expected, rTg4510 mice exhibit profound impairments in hippocampal-dependent learning and memory tests, including contextual fear conditioning. However, rTg4510 mice also display an abnormal hyperexploratory phenotype in the open-field assay, elevated plus maze, light-dark exploration, and cued fear conditioning, indicative of amygdala dysfunction. Furthermore, significant tau burden is detected in the amygdala of both rTg4510 mice and human FTDP-17 patients, suggesting that the rTg4510 mouse model recapitulates the behavioral disturbances and neurodegeneration of the amygdala characteristic of FTDP-17. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
28. Vascular ApoE4 Impairs Behavior by Modulating Gliovascular Function.
- Author
-
Yamazaki, Yu, Liu, Chia-Chen, Yamazaki, Akari, Shue, Francis, Martens, Yuka A., Chen, Yuanxin, Qiao, Wenhui, Kurti, Aishe, Oue, Hiroshi, Ren, Yingxue, Li, Ying, Aikawa, Tomonori, Cherukuri, Yesesri, Fryer, John D., Asmann, Yan W., Kim, Betty Y.S., Kanekiyo, Takahisa, and Bu, Guojun
- Subjects
- *
NEUROGLIA , *RNA sequencing , *APOLIPOPROTEIN E , *BLOOD flow , *ALZHEIMER'S disease - Abstract
The ε4 allele of the apolipoprotein E gene (APOE4) is a strong genetic risk factor for Alzheimer's disease (AD) and multiple vascular conditions. ApoE is abundantly expressed in multiple brain cell types, including astrocytes, microglia, and vascular mural cells (VMCs). Here, we show that VMC-specific expression of apoE4 in mice impairs behavior and cerebrovascular function. Expression of either apoE3 or apoE4 in VMCs was sufficient to rescue the hypercholesterolemia and atherosclerosis phenotypes seen in Apoe knockout mice. Intriguingly, vascular expression of apoE4, but not apoE3, reduced arteriole blood flow, impaired spatial learning, and increased anxiety-like phenotypes. Single-cell RNA sequencing of vascular and glial cells revealed that apoE4 in VMCs was associated with astrocyte activation, while apoE3 was linked to angiogenic signature in pericytes. Together, our data support cell-autonomous effects of vascular apoE on brain homeostasis in an isoform-dependent manner, suggesting a critical contribution of vascular apoE to AD pathogenesis. • Conditional mouse models expressing apoE isoforms in vascular mural cells • Vascular apoE4 is associated with impaired arteriole blood flow and behaviors • Transcriptomic analysis suggests apoE4-related astrogliosis at the vasculature • ApoE3 expressed in vasculature is linked to angiogenic signature in pericytes Yamazaki et al. generated conditional mouse models expressing apoE isoforms uniquely in vascular mural cells on a murine Apoe knockout background. They show that vascular-mural-cell-derived apoE4 impairs vascular function and behavior. Single-cell RNA sequencing reveals apoE4-associated astrogliosis along the vasculature and apoE3-related angiogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
29. TIA1 Mutations in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Promote Phase Separation and Alter Stress Granule Dynamics.
- Author
-
Mackenzie, Ian R., Nicholson, Alexandra M., Sarkar, Mohona, Messing, James, Purice, Maria D., Pottier, Cyril, Annu, Kavya, Baker, Matt, Perkerson, Ralph B., Kurti, Aishe, Matchett, Billie J., Mittag, Tanja, Temirov, Jamshid, Hsiung, Ging-Yuek R., Krieger, Charles, Murray, Melissa E., Kato, Masato, Fryer, John D., Petrucelli, Leonard, and Zinman, Lorne
- Subjects
- *
GENETIC mutation , *AMYOTROPHIC lateral sclerosis , *FRONTOTEMPORAL dementia , *AGE factors in disease , *NEURODEGENERATION , *GENETICS - Abstract
Summary Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are age-related neurodegenerative disorders with shared genetic etiologies and overlapping clinical and pathological features. Here we studied a novel ALS/FTD family and identified the P362L mutation in the low-complexity domain (LCD) of T cell-restricted intracellular antigen-1 (TIA1). Subsequent genetic association analyses showed an increased burden of TIA1 LCD mutations in ALS patients compared to controls (p = 8.7 × 10 −6 ). Postmortem neuropathology of five TIA1 mutations carriers showed a consistent pathological signature with numerous round, hyaline, TAR DNA-binding protein 43 (TDP-43)-positive inclusions. TIA1 mutations significantly increased the propensity of TIA1 protein to undergo phase transition. In live cells, TIA1 mutations delayed stress granule (SG) disassembly and promoted the accumulation of non-dynamic SGs that harbored TDP-43. Moreover, TDP-43 in SGs became less mobile and insoluble. The identification of TIA1 mutations in ALS/FTD reinforces the importance of RNA metabolism and SG dynamics in ALS/FTD pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.