Phosphorylation and Dephosphorylation of Tau Protein During Synthetic Torpor

Tau protein is of primary importance for many physiological processes in neurons, where it affects the dynamics of the microtubule system. When hyperphosphorylated (PP-Tau), Tau monomers detach from microtubules and tend to aggregate firstly in oligomers, and then in neurofibrillary tangles, as it occurs in a group of neurodegenerative disorders named thauopathies. A hypothermia-related accumulation of PP-Tau, which is quickly reversed after the return to normothermia, has been shown to occur in the brain of hibernators during torpor. Since, recently, in our lab, a hypothermic torpor-like condition (synthetic torpor, ST) was pharmacologically induced in the rat, a non-hibernator, the aim of the present work was to assess whether ST can lead to a reversible PP-Tau accumulation in the rat brain. PP-Tau was immunohistochemically assessed by staining for AT8 (phosphorylated Tau) and Tau-1 (non-phosphorylated Tau) in 19 brain structures, which were chosen mostly due to their involvement in the regulation of autonomic and cognitive functions in relation to behavioral states. During ST, AT8 staining was strongly expressed throughout the brain, while Tau-1 staining was reduced compared to control conditions. During the following recovery period, AT8 staining progressively reduced close to zero after 6 h from ST. However, Tau-1 staining remained low even after 38 h from ST. Thus, overall, these results show that ST induced an accumulation of PP-Tau that was, apparently, only partially reversed to normal during the recovery period. While the accumulation of PP-Tau may only depend on the physicochemical characteristics of the enzymes regulating Tau phosphorylation, the reverse process of dephosphorylation should be actively regulated, also in non-hibernators. In conclusion, in this work a reversible and widespread PP-Tau accumulation has been induced through a procedure that leads a non-hibernator to a degree of reversible hypothermia, which is comparable to that observed in hibernators. Therefore, the physiological mechanism involved in this process can sustain an adaptive neuronal response to extreme conditions, which may however lead to neurodegeneration when particular intensities and durations are exceeded.

[1]  R. Henning,et al.  Brain inflammatory cytokines and microglia morphology changes throughout hibernation phases in Syrian hamster , 2018, Brain, Behavior, and Immunity.

[2]  Eduardo Colombari,et al.  The nucleus of the solitary tract and the coordination of respiratory and sympathetic activities , 2014, Front. Physiol..

[3]  S. Morrison,et al.  Central Mechanisms for Thermoregulation. , 2019, Annual review of physiology.

[4]  T. Arendt,et al.  Brain hypometabolism triggers PHF-like phosphorylation of tau, a major hallmark of Alzheimer’s disease pathology , 2015, Journal of Neural Transmission.

[5]  E. Mandelkow,et al.  Tau in physiology and pathology , 2015, Nature Reviews Neuroscience.

[6]  George Perry,et al.  Physiological regulation of tau phosphorylation during hibernation , 2008, Journal of neurochemistry.

[7]  M. Billingsley,et al.  Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration. , 1997, The Biochemical journal.

[8]  J. P. Hannon Effect of Temperature on the Heart Rate, Electrocardiogram and Certain Myocardial Oxidations of the Rat , 1958, Circulation research.

[9]  C. Goldsbury,et al.  Microglia show altered morphology and reduced arborization in human brain during aging and Alzheimer's disease , 2017, Brain pathology.

[10]  Seralynne D Vann,et al.  The mammillary bodies and memory: more than a hippocampal relay. , 2015, Progress in brain research.

[11]  J. Filosa,et al.  A quantitative spatiotemporal analysis of microglia morphology during ischemic stroke and reperfusion , 2013, Journal of Neuroinflammation.

[12]  M. King,et al.  Adult-onset focal expression of mutated human tau in the hippocampus impairs spatial working memory of rats , 2012, Behavioural Brain Research.

[13]  J. Hudson,et al.  Daily Torpor in the Laboratory Mouse, Mus musculus Var. Albino , 1979, Physiological Zoology.

[14]  H. Soininen,et al.  Cerebrospinal fluid {beta}-amyloid 42 and tau proteins as biomarkers of Alzheimer-type pathologic changes in the brain. , 2009, Archives of neurology.

[15]  S. Fujita,et al.  Stress‐induced hyperphosphorylation of tau in the mouse brain , 2003, FEBS letters.

[16]  Hai-bing Xiao,et al.  Temperature control can abolish anesthesia‐induced tau hyperphosphorylation and partly reverse anesthesia‐induced cognitive impairment in old mice , 2013, Psychiatry and clinical neurosciences.

[17]  S. Fujita,et al.  Inhibition of Protein Phosphatase 2A Overrides Tau Protein Kinase I/Glycogen Synthase Kinase 3β and Cyclin-dependent Kinase 5 Inhibition and Results in Tau Hyperphosphorylation in the Hippocampus of Starved Mouse* , 2001, The Journal of Biological Chemistry.

[18]  M. Bentivoglio,et al.  Limbic thalamus and state-dependent behavior: The paraventricular nucleus of the thalamic midline as a node in circadian timing and sleep/wake-regulatory networks , 2015, Neuroscience & Biobehavioral Reviews.

[19]  Akihiko Takashima,et al.  Alterations in Glucose Metabolism Induce Hypothermia Leading to Tau Hyperphosphorylation through Differential Inhibition of Kinase and Phosphatase Activities: Implications for Alzheimer's Disease , 2004, The Journal of Neuroscience.

[20]  R. Gross,et al.  Adenosine A2A receptor mediates microglial process retraction , 2009, Nature Neuroscience.

[21]  Jennifer Luebke,et al.  Depletion of microglia and inhibition of exosome synthesis halt tau propagation , 2015, Nature Neuroscience.

[22]  Davide Martelli,et al.  The Inhibition of Neurons in the Central Nervous Pathways for Thermoregulatory Cold Defense Induces a Suspended Animation State in the Rat , 2013, The Journal of Neuroscience.

[23]  E. Camouzis,et al.  On the Dynamics of , 2003 .

[24]  Wolfgang Härtig,et al.  Reversible Paired Helical Filament-Like Phosphorylation of Tau Is an Adaptive Process Associated with Neuronal Plasticity in Hibernating Animals , 2003, The Journal of Neuroscience.

[25]  G. Paxinos,et al.  The Rat Brain in Stereotaxic Coordinates , 1983 .

[26]  Central Mechanisms , 2001, International Journal of Obesity.

[27]  J. Growdon,et al.  Reactive glia not only associates with plaques but also parallels tangles in Alzheimer's disease. , 2011, The American journal of pathology.

[28]  H. Steinbusch,et al.  Vagal efferent projections: viscerotopy, neurochemistry and effects of vagotomy. , 1996, Progress in brain research.

[29]  N. Cairns,et al.  Widespread tau seeding activity at early Braak stages , 2016, Acta Neuropathologica.

[30]  P. Luppi Neurochemical aspects of sleep regulation with specific focus on slow-wave sleep , 2010, The world journal of biological psychiatry : the official journal of the World Federation of Societies of Biological Psychiatry.

[31]  M. Rundgren,et al.  The median preoptic nucleus: front and centre for the regulation of body fluid, sodium, temperature, sleep and cardiovascular homeostasis , 2015, Acta Physiologica.

[32]  Jian-Zhi Wang,et al.  Codonopsis pilosula Polysaccharide Attenuates Tau Hyperphosphorylation and Cognitive Impairments in hTau Infected Mice , 2018, Front. Mol. Neurosci..

[33]  Wendy A Suzuki,et al.  The anatomy, physiology and functions of the perirhinal cortex , 1996, Current Opinion in Neurobiology.

[34]  Javier DeFelipe,et al.  Changes in neocortical and hippocampal microglial cells during hibernation , 2017, Brain Structure and Function.

[35]  M. Luppi MCH and Thermoregulation , 2018 .

[36]  J. Ávila,et al.  Tissue-nonspecific Alkaline Phosphatase Promotes the Neurotoxicity Effect of Extracellular Tau* , 2010, The Journal of Biological Chemistry.

[37]  D. Holtzman,et al.  In Vivo Microdialysis Reveals Age-Dependent Decrease of Brain Interstitial Fluid Tau Levels in P301S Human Tau Transgenic Mice , 2011, The Journal of Neuroscience.

[38]  G. Gilliland,et al.  Epitope mapping and structural basis for the recognition of phosphorylated tau by the anti‐tau antibody AT8 , 2016, Proteins.

[39]  Brian M. Barnes,et al.  The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation , 2011, PloS one.

[40]  Wendy Noble,et al.  Physiological release of endogenous tau is stimulated by neuronal activity , 2013, EMBO reports.

[41]  R. Ransohoff How neuroinflammation contributes to neurodegeneration , 2016, Science.

[42]  L. Binder,et al.  Rapid reversible phosphorylation of rat brain tau proteins in response to cold water stress , 1995, Neuroscience Letters.

[43]  J. Hudson,et al.  DAILY TORPOR IN THE LABORATORY MOUSE , 2016 .

[44]  U. Sengupta,et al.  Tau Oligomers Associate with Inflammation in the Brain and Retina of Tauopathy Mice and in Neurodegenerative Diseases , 2016, Journal of Alzheimer's disease : JAD.

[45]  R. Kayed,et al.  Advances in therapeutics for neurodegenerative tauopathies: moving toward the specific targeting of the most toxic tau species. , 2014, ACS chemical neuroscience.

[46]  M. Dailey,et al.  Dynamics of microglial activation: A confocal time‐lapse analysis in hippocampal slices , 2001, Glia.

[47]  Eduardo E. Benarroch,et al.  Locus coeruleus , 2020, Cell and Tissue Research.

[48]  Virginia M. Y. Lee,et al.  Recognition of the minimal epitope of monoclonal antibody Tau‐1 depends upon the presence of a phosphate group but not its location , 1993, Journal of neuroscience research.

[49]  Z. Andrews,et al.  The temporal pattern of cfos activation in hypothalamic, cortical, and brainstem nuclei in response to fasting and refeeding in male mice. , 2014, Endocrinology.

[50]  H. Braak,et al.  Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry , 2006, Acta Neuropathologica.

[51]  S. Fujita,et al.  Starvation induces tau hyperphosphorylation in mouse brain: implications for Alzheimer's disease , 1999, FEBS letters.

[52]  Matteo Cerri,et al.  The Central Control of Energy Expenditure: Exploiting Torpor for Medical Applications. , 2017, Annual review of physiology.

[53]  D. Baker,et al.  Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline , 2018, Nature.

[54]  E. Brǎiloiu,et al.  Modulation of cardiac vagal tone by bradykinin acting on nucleus ambiguus , 2017, Neuroscience.

[55]  R. Nelson,et al.  Anesthesia Leads to Tau Hyperphosphorylation through Inhibition of Phosphatase Activity by Hypothermia , 2007, The Journal of Neuroscience.

[56]  A. Ittner SITE-SPECIFIC PHOSPHORYLATION OF TAU INHIBITS AMYLOID-β TOXICITY IN ALZHEIMER’S MICE , 2016, Alzheimer's & Dementia.

[57]  Feng Wu,et al.  Expression of Tau Pathology-Related Proteins in Different Brain Regions: A Molecular Basis of Tau Pathogenesis , 2017, Front. Aging Neurosci..

[58]  R. Kayed,et al.  Potential mechanisms and implications for the formation of tau oligomeric strains , 2016, Critical reviews in biochemistry and molecular biology.

[59]  S. Morrison,et al.  Central Activation of the A1 Adenosine Receptor (A1AR) Induces a Hypothermic, Torpor-Like State in the Rat , 2013, The Journal of Neuroscience.