Publications

2021

Criado-Marrero, Marangelie, Niat T Gebru, Lauren A Gould, Danielle M Blazier, Yamile Vidal-Aguiar, Taylor M Smith, Salma S Abdelmaboud, et al. (2021) 2021. “FKBP52 Overexpression Accelerates Hippocampal-Dependent Memory Impairments in a Tau Transgenic Mouse Model”. NPJ Aging and Mechanisms of Disease 7 (1): 9. https://doi.org/10.1038/s41514-021-00062-x.

Abnormal accumulation of hyperphosphorylated tau induces pathogenesis in neurodegenerative diseases, like Alzheimer's disease. Molecular chaperones with peptidyl-prolyl cis/trans isomerase (PPIase) activity are known to regulate these processes. Previously, in vitro studies have shown that the 52 kDa FK506-binding protein (FKBP52) interacts with tau inducing its oligomerization and fibril formation to promote toxicity. Thus, we hypothesized that increased expression of FKBP52 in the brains of tau transgenic mice would alter tau phosphorylation and neurofibrillary tangle formation ultimately leading to memory impairments. To test this, tau transgenic (rTg4510) and wild-type mice received bilateral hippocampal injections of virus overexpressing FKBP52 or GFP control. We examined hippocampal-dependent memory, synaptic plasticity, tau phosphorylation status, and neuronal health. This work revealed that rTg4510 mice overexpressing FKBP52 had impaired spatial learning, accompanied by long-term potentiation deficits and hippocampal neuronal loss, which was associated with a modest increase in total caspase 12. Together with previous studies, our findings suggest that FKBP52 may sensitize neurons to tau-mediated dysfunction via activation of a caspase-dependent pathway, contributing to memory and learning impairments.

Criado-Marrero, Marangelie, Niat T Gebru, Danielle M Blazier, Lauren A Gould, Jeremy D Baker, David Beaulieu-Abdelahad, and Laura J Blair. (2021) 2021. “Hsp90 Co-Chaperones, FKBP52 and Aha1, Promote Tau Pathogenesis in Aged Wild-Type Mice”. Acta Neuropathologica Communications 9 (1): 65. https://doi.org/10.1186/s40478-021-01159-w.

The microtubule associated protein tau is an intrinsically disordered phosphoprotein that accumulates under pathological conditions leading to formation of neurofibrillary tangles, a hallmark of Alzheimer's disease (AD). The mechanisms that initiate the accumulation of phospho-tau aggregates and filamentous deposits are largely unknown. In the past, our work and others' have shown that molecular chaperones play a crucial role in maintaining protein homeostasis and that imbalance in their levels or activity can drive tau pathogenesis. We have found two co-chaperones of the 90 kDa heat shock protein (Hsp90), FK506-binding protein 52 (FKBP52) and the activator of Hsp90 ATPase homolog 1 (Aha1), promote tau aggregation in vitro and in the brains of tau transgenic mice. Based on this, we hypothesized that increased levels of these chaperones could promote tau misfolding and accumulation in the brains of aged wild-type mice. We tested this hypothesis by overexpressing Aha1, FKBP52, or mCherry (control) proteins in the hippocampus of 9-month-old wild-type mice. After 7 months of expression, mice were evaluated for cognitive and pathological changes. Our results show that FKBP52 overexpression impaired spatial reversal learning, while Aha1 overexpression impaired associative learning in aged wild-type mice. FKBP52 and Aha1 overexpression promoted phosphorylation of distinct AD-relevant tau species. Furthermore, FKBP52 activated gliosis and promoted neuronal loss leading to a reduction in hippocampal volume. Glial activation and phospho-tau accumulation were also detected in areas adjacent to the hippocampus, including the entorhinal cortex, suggesting that after initiation these pathologies can propagate through other brain regions. Overall, our findings suggest a role for chaperone imbalance in the initiation of tau accumulation in the aging brain.

Sandusky-Beltran, Leslie A, Andrii Kovalenko, Devon S Placides, Kevin Ratnasamy, Chao Ma, Jerry B Hunt, Huimin Liang, et al. (2021) 2021. “Aberrant AZIN2 and Polyamine Metabolism Precipitates Tau Neuropathology”. The Journal of Clinical Investigation 131 (4). https://doi.org/10.1172/JCI126299.

Tauopathies display a spectrum of phenotypes from cognitive to affective behavioral impairments; however, mechanisms promoting tau pathology and how tau elicits behavioral impairment remain unclear. We report a unique interaction between polyamine metabolism, behavioral impairment, and tau fate. Polyamines are ubiquitous aliphatic molecules that support neuronal function, axonal integrity, and cognitive processing. Transient increases in polyamine metabolism hallmark the cell's response to various insults, known as the polyamine stress response (PSR). Dysregulation of gene transcripts associated with polyamine metabolism in Alzheimer's disease (AD) brains were observed, and we found that ornithine decarboxylase antizyme inhibitor 2 (AZIN2) increased to the greatest extent. We showed that sustained AZIN2 overexpression elicited a maladaptive PSR in mice with underlying tauopathy (MAPT P301S; PS19). AZIN2 also increased acetylpolyamines, augmented tau deposition, and promoted cognitive and affective behavioral impairments. Higher-order polyamines displaced microtubule-associated tau to facilitate polymerization but also decreased tau seeding and oligomerization. Conversely, acetylpolyamines promoted tau seeding and oligomers. These data suggest that tauopathies launch an altered enzymatic signature that endorses a feed-forward cycle of disease progression. Taken together, the tau-induced PSR affects behavior and disease continuance, but may also position the polyamine pathway as a potential entry point for plausible targets and treatments of tauopathy, including AD.

Darling, April L, Jan Dahrendorff, Stefan G Creodore, Chad A Dickey, Laura J Blair, and Vladimir N Uversky. (2021) 2021. “Small Heat Shock Protein 22 kDa Can Modulate the Aggregation and Liquid-Liquid Phase Separation Behavior of Tau”. Protein Science : A Publication of the Protein Society 30 (7): 1350-59. https://doi.org/10.1002/pro.4060.

Alzheimer's disease is a progressive fatal neurodegenerative disease with no cure or effective treatments. The hallmarks of disease include extracellular plaques and intracellular tangles of aggregated protein. The intracellular tangles consist of the microtubule associated protein tau. Preventing the pathological aggregation of tau may be an important therapeutic approach to treat disease. In this study we show that small heat shock protein 22 kDa (Hsp22) can prevent the aggregation of tau in vitro. Additionally, tau can undergo liquid-liquid phase separation (LLPS) in the presence of crowding reagents which causes it to have an increased aggregation rate. We show that Hsp22 can modulate both the aggregation and LLPS behavior of tau in vitro.

Criado-Marrero, Marangelie, Niat T Gebru, Danielle M Blazier, Lauren A Gould, Jeremy D Baker, David Beaulieu-Abdelahad, and Laura J Blair. (2021) 2021. “Correction To: Hsp90 co‑chaperones, FKBP52 and Aha1, Promote Tau Pathogenesis in Aged wild‑type Mice”. Acta Neuropathologica Communications 9 (1): 85. https://doi.org/10.1186/s40478-021-01188-5.
Lopez, Abraham, Vinay Dahiya, Florent Delhommel, Lee Freiburger, Ralf Stehle, Sam Asami, Daniel Rutz, Laura Blair, Johannes Buchner, and Michael Sattler. (2021) 2021. “Client Binding Shifts the Populations of Dynamic Hsp90 Conformations through an Allosteric Network”. Science Advances 7 (51): eabl7295. https://doi.org/10.1126/sciadv.abl7295.

Hsp90 is a molecular chaperone that interacts with a specific set of client proteins and assists their folding. The underlying molecular mechanisms, involving dynamic transitions between open and closed conformations, are still enigmatic. Combining nuclear magnetic resonance, small-angle x-ray scattering, and biochemical experiments, we have identified a key intermediate state of Hsp90 induced by adenosine triphosphate (ATP) binding, in which rotation of the Hsp90 N-terminal domain (NTD) yields a domain arrangement poised for closing. This ATP-stabilized NTD rotation is allosterically communicated across the full Hsp90 dimer, affecting distant client sites. By analyzing the interactions of four distinct clients, i.e., steroid hormone receptors (glucocorticoid receptor and mineralocorticoid receptor), p53, and Tau, we show that client-specific interactions with Hsp90 select and enhance the NTD-rotated state and promote closing of the full-length Hsp90 dimer. The p23 co-chaperone shifts the population of Hsp90 toward the closed state, thereby enhancing client interaction and processing.

2020

Doe, Jane, John Smith, Stephanie Black, and Mark White. 2020. “Lorem Ipsum Dolor Sit Amet, Consectetur Adipiscing Elit, Sed Do Eiusmod Tempor Incididunt Ut Labore Et Dolore Magna Aliqua”. Ut Sem Viverra Aliquet Eget Sit Amet Tellus. 32 (7): 1789-1804.
Doe, Jane, John Smith, Stephanie Black, and Mark White. 2020. “Lorem Ipsum Dolor Sit Amet, Consectetur Adipiscing Elit, Sed Do Eiusmod Tempor Incididunt Ut Labore Et Dolore Magna Aliqua”. Ut Sem Viverra Aliquet Eget Sit Amet Tellus. 32 (7): 1789-1804.
Doe, Jane, John Smith, Stephanie Black, and Mark White. 2020. “Lorem Ipsum Dolor Sit Amet, Consectetur Adipiscing Elit, Sed Do Eiusmod Tempor Incididunt Ut Labore Et Dolore Magna Aliqua”. Ut Sem Viverra Aliquet Eget Sit Amet Tellus. 32 (7): 1789-1804.
Doe, Jane, John Smith, Stephanie Black, and Mark White. 2020. “Lorem Ipsum Dolor Sit Amet, Consectetur Adipiscing Elit, Sed Do Eiusmod Tempor Incididunt Ut Labore Et Dolore Magna Aliqua”. Ut Sem Viverra Aliquet Eget Sit Amet Tellus. 32 (7): 1789-1804.