Association between lower body temperature and increased tau pathology in cognitively normal older adults

Esther M Blessing, Ankit Parekh, Rebecca A Betensky, James Babb, Natalie Saba, Ludovic Debure, Andrew W Varga, Indu Ayappa, David M Rapoport, Tracy A Butler, Mony J de Leon, Thomas Wisniewski, Brian J Lopresti, Ricardo S Osorio, Esther M Blessing, Ankit Parekh, Rebecca A Betensky, James Babb, Natalie Saba, Ludovic Debure, Andrew W Varga, Indu Ayappa, David M Rapoport, Tracy A Butler, Mony J de Leon, Thomas Wisniewski, Brian J Lopresti, Ricardo S Osorio

Abstract

Background: Preclinical studies suggest body temperature (Tb) and consequently brain temperature has the potential to bidirectionally interact with tau pathology in Alzheimer's Disease (AD). Tau phosphorylation is substantially increased by a small (<1 °C) decrease in temperature within the human physiological range, and thermoregulatory nuclei are affected by tau pathology early in the AD continuum. In this study we evaluated whether Tb (as a proxy for brain temperature) is cross-sectionally associated with clinically utilized markers of tau pathology in cognitively normal older adults.

Methods: Tb was continuously measured with ingestible telemetry sensors for 48 h. This period included two nights of nocturnal polysomnography to delineate whether Tb during waking vs sleep is differentially associated with tau pathology. Tau phosphorylation was assessed with plasma and cerebrospinal fluid (CSF) tau phosphorylated at threonine 181 (P-tau), sampled the day following Tb measurement. In addition, neurofibrillary tangle (NFT) burden in early Braak stage regions was imaged with PET-MR using the [18F]MK-6240 radiotracer on average one month later.

Results: Lower Tb was associated with increased NFT burden, as well as increased plasma and CSF P-tau levels (p < 0.05). NFT burden was associated with lower Tb during waking (p < 0.05) but not during sleep intervals. Plasma and CSF P-tau levels were highly correlated with each other (p < 0.05), and both variables were correlated with tau tangle radiotracer uptake (p < 0.05).

Conclusions: These results, the first available for human, suggest that lower Tb in older adults may be associated with increased tau pathology. Our findings add to the substantial preclinical literature associating lower body and brain temperature with tau hyperphosphorylation.

Clinical trial number: NCT03053908.

Keywords: Aging; Alzheimer's disease; Body temperature; Neurofibrillary tangle; Phosphorylation; Tau; [(18)F]MK-6240.

Conflict of interest statement

Declaration of Competing Interest

The authors declare that they have no competing interests.

Copyright © 2022. Published by Elsevier Inc.

Figures

Fig. 1. |
Fig. 1. |
Protocol for integrated measurement of body temperature and sleep. Time of body temperature sensor (pill) ingestion and recording indicated by line continuing from pill. Nocturnal polysomnography (NPSG) on Nights 1 and 2. Sleep interval (grey) included time between lights off (L. Off) and lights on (L. On), occurring in the sleep lab during NPSG. The Waking interval (white) included time on Days 1 and 2 between lights On and lights Off. During this time, subjects were free to return home and behave according to their normal schedule, indicated by activities bracketed under ‘?’.
Fig. 2.
Fig. 2.
Relationships between aggregated and soluble markers of tau pathology. A, Coronal [18F] MK-6240 tau tangle MR-PET images from two subjects: upper, T-negative 65 yo male, lower, T+ positive 63 yo female. B-D, associations between plasma P-tau, CSF P-tau and MK6240 SUVR uptake. Solid lines show linear regression, dashed lines show mean confidence intervals. Spearman’s rank correlation results at lower right of each plot.
Fig. 3. |
Fig. 3. |
Raw and DRAGO processed body temperature data for one participant. Upper panel, raw body temperature (Tb) data, yellow shading indicates data outlier, blue shading indicates data gap. Lower panel, DRAGO processed Tb data divided into Sleep (nights 1 and 2) and Waking (days 1 and 2) intervals. Shaded areas show Tb values under indicated threshold (37 °C). Waking interval Tb metrics were Mean Tb, 37.15 °C, and time Tb under threshold (TimeSubThr), 21.44%.
Fig. 4. |
Fig. 4. |
Correlation matrix of associations between body temperature, MK-6240 SUVR uptake in Braak I-III regions, CSF P-tau and plasma P-tau. Significant correlations are color coded by magnitude according to the indicated scale. Abbreviations: Tb, body temperature; SUVR, standardized uptake value ratio, TimeSubThr, body temperature under threshold.
Fig. 5. |
Fig. 5. |
Associations between Waking Tb and MK-6240 SUVR uptake. A, C associations for mean Tb and MK6240 SUVR uptake in Braak I and parahippocampal gyrus regions, respectively. B, D, equivalent associations for the proportion of time Tb was under threshold during Waking intervals. Results of Spearman’s rank correlations are shown, dashed lines show mean confidence intervals. Abbreviations, PHG, parahippocampal gyrus, Tb, body temperature, TimeSubThr, time Tb was under threshold, MK6240, [18F]MK-6240 tau tangle tracer, SUVR, standardized uptake value ratio.
Fig. 6. |
Fig. 6. |
Bidirectional relationship between body and brain temperature dysregulation and tau pathology. Arm 1 (black arrow) represents a direct effect of body and brain temperature upon tau phosphorylation and other molecular targets affecting tau pathology. Arm 2 (grey arrows) represents a multifactorial pathway by which tau pathology impacts body and brain temperature. Dashed line indicates minimal data available. Abbreviations, TR, thermoregulatory, S/WP, sleep- or wake promoting (neuron), CR, circadian rhythm.

References

    1. Arendt T, Bullmann T, 2013. Neuronal plasticity in hibernation and the proposed role of the microtubule-associated protein tau as a “master switch” regulating synaptic gain in neuronal networks. Am. J. Phys. Regul. Integr. Comp. Phys 305, R478–R489.
    1. Arendt T, Holzer M, Fruth R, Bruckner MK, Gartner U, 1995. Paired helical filament-like phosphorylation of tau, deposition of beta/A4-amyloid and memory impairment in rat induced by chronic inhibition of phosphatase 1 and 2A. Neuroscience 69, 691–698.
    1. Arendt T, et al., 2003. Reversible paired helical filament-like phosphorylation of tau is an adaptive process associated with neuronal plasticity in hibernating animals. J. Neurosci 23, 6972–6981.
    1. Arendt T, Stieler J, Holzer M, 2015. Brain hypometabolism triggers PHF-like phosphorylation of tau, a major hallmark of Alzheimer’s disease pathology. J. Neural Transm. (Vienna) 122, 531–539.
    1. Aschoff J, 1998. Circadian parameters as individual characteristics. J. Biol. Rhythm 13, 123–131.
    1. Attems J, Thal DR, Jellinger KA, 2012. The relationship between subcortical tau pathology and Alzheimer’s disease. Biochem. Soc. Trans 40, 711–715.
    1. Augustinack JC, Schneider A, Mandelkow EM, Hyman BT, 2002. Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer’s disease. Acta Neuropathol 103, 26–35.
    1. Barthelemy NR, et al., 2020. A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited Alzheimer’s disease. Nat. Med 26, 398–407.
    1. Betthauser TJ, et al., 2020. Amyloid and tau imaging biomarkers explain cognitive decline from late middle-age. Brain 143, 320–335.
    1. Biundo F, Del Prete D, Zhang H, Arancio O, D’Adamio L, 2018. A role for tau in learning, memory and synaptic plasticity. Sci. Rep 8, 3184.
    1. Blatteis CM, 2012. Age-dependent changes in temperature regulation - a mini review. Gerontology 58, 289–295.
    1. Braak H, Braak E, 1997. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol. Aging 18, 351–357.
    1. Braak H, Thal DR, Ghebremedhin E, 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. Bretteville A, et al., 2012. Hypothermia-induced hyperphosphorylation: a new model to study tau kinase inhibitors. Sci. Rep 2, 480.
    1. Buhr ED, Yoo SH, Takahashi JS, 2010. Temperature as a universal resetting cue for mammalian circadian oscillators. Science 330, 379–385.
    1. Bunnell DE, Agnew JA, Horvath SM, Jopson L, Wills M, 1988. Passive body heating and sleep: influence of proximity to sleep. Sleep 11, 210–219.
    1. Byrne C, Lim CL, 2007. The ingestible telemetric body core temperature sensor: a review of validity and exercise applications. Br. J. Sports Med 41, 126–133.
    1. Casa DJ, et al., 2007. Validity of devices that assess body temperature during outdoor exercise in the heat. J. Athl. Train 42, 333–342.
    1. Clark C, et al., 2021. Plasma neurofilament light and phosphorylated tau 181 as biomarkers of Alzheimer’s disease pathology and clinical disease progression. Alzheimers Res. Ther 13, 65.
    1. Czeisler CA, Zimmerman JC, Ronda JM, Moore-Ede MC, Weitzman ED, 1980. Timing of REM sleep is coupled to the circadian rhythm of body temperature in man. Sleep 2, 329–346.
    1. Dickey CA, et al., 2007. The high-affinity HSP90-CHIP complex recognizes and selectively degrades phosphorylated tau client proteins. J. Clin. Invest 117, 648–658.
    1. Dixit R, Ross JL, Goldman YE, Holzbaur EL, 2008. Differential regulation of dynein and kinesin motor proteins by tau. Science 319, 1086–1089.
    1. Duffy JF, Dijk DJ, 2002. Getting through to circadian oscillators: why use constant routines? J. Biol. Rhythm 17, 4–13.
    1. Fields PA, Dong Y, Meng X, Somero GN, 2015. Adaptations of protein structure and function to temperature: there is more than one way to ‘skin a cat’. J. Exp. Biol 218, 1801–1811.
    1. Fultz NE, et al., 2019. Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science 366, 628–631.
    1. Gant N, Atkinson G, Williams C, 2006. The validity and reliability of intestinal temperature during intermittent running. Med. Sci. Sports Exerc 38, 1926–1931.
    1. Gogola A, et al., 2021. Direct comparison of the tau PET tracers [(18)F]flortaucipir and [(18)F]MK-6240 in human subjects. J. Nucl. Med 63, 108–116.
    1. Gomolin IH, Aung MM, Wolf-Klein G, Auerbach C, 2005. Older is colder: temperature range and variation in older people. J. Am. Geriatr. Soc 53, 2170–2172.
    1. Gong CX, Iqbal K, 2008. Hyperphosphorylation of microtubule-associated protein tau: a promising therapeutic target for Alzheimer disease. Curr. Med. Chem 15, 2321–2328.
    1. Gong CX, Liu F, Grundke-Iqbal I, Iqbal K, 2005. Post-translational modifications of tau protein in Alzheimer’s disease. J. Neural Transm. (Vienna) 112, 813–838.
    1. Gotz J, Halliday G, Nisbet RM, 2019. Molecular pathogenesis of the Tauopathies. Annu. Rev. Pathol 14, 239–261.
    1. Gratuze M, Julien J, Petry FR, Morin F, Planel E, 2017. Insulin deprivation induces PP2A inhibition and tau hyperphosphorylation in hTau mice, a model of Alzheimer’s disease-like tau pathology. Sci. Rep 7, 46359.
    1. Grinberg LT, et al., 2009. The dorsal raphe nucleus shows phospho-tau neurofibrillary changes before the transentorhinal region in Alzheimer’s disease. A precocious onset? Neuropathol. Appl. Neurobiol 35, 406–416.
    1. Grundke-Iqbal I, et al., 1986. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. U. S. A 83, 4913–4917.
    1. Guisle I, et al., 2020. Circadian and sleep/wake-dependent variations in tau phosphorylation are driven by temperature. Sleep 43, 1–12.
    1. Guisle I, et al., 2022. Sauna-like conditions or menthol treatment reduce tau phosphorylation through mild hyperthermia. Neurobiol. Aging 113, 118–130.
    1. Guo T, Noble W, Hanger DP, 2017. Roles of tau protein in health and disease. Acta Neuropathol 133, 665–704.
    1. Hansson O, et al., 2006. Association between CSF biomarkers and incipient Alzheimer’s disease in patients with mild cognitive impairment: a follow-up study. Lancet Neurol 5, 228–234.
    1. Hansson O, et al., 2018. CSF biomarkers of Alzheimer’s disease concord with amyloid-beta PET and predict clinical progression: a study of fully automated immunoassays in BioFINDER and ADNI cohorts. Alzheimers Dement 14, 1470–1481.
    1. Harper DG, et al., 2004. Dementia severity and Lewy bodies affect circadian rhythms in Alzheimer disease. Neurobiol. Aging 25, 771–781.
    1. Harper DG, et al., 2005. Disturbance of endogenous circadian rhythm in aging and Alzheimer disease. Am. J. Geriatr. Psychiatry 13, 359–368.
    1. Harper DG, et al., 2008. Dorsomedial SCN neuronal subpopulations subserve different functions in human dementia. Brain 131, 1609–1617.
    1. Harrison IF, et al., 2020. Impaired glymphatic function and clearance of tau in an Alzheimer’s disease model. Brain 143, 2576–2593.
    1. Hitrec T, et al., 2021. Reversible tau phosphorylation induced by synthetic torpor in the spinal cord of the rat. Front. Neuroanat 15, 592288.
    1. Hochachka PW, Somero GN, 1968. The adaptation of enzymes to temperature. Comp. Biochem. Physiol 27, 659–668.
    1. Holth JK, et al., 2019. The sleep-wake cycle regulates brain interstitial fluid tau in mice and CSF tau in humans. Science 363, 880–884.
    1. Horne JA, Staff LH, 1983. Exercise and sleep: body-heating effects. Sleep 6, 36–46.
    1. Hostetler ED, et al., 2016. Preclinical characterization of 18F-MK-6240, a promising PET tracer for in vivo quantification of human neurofibrillary tangles. J. Nucl. Med 57, 1599–1606.
    1. Huang Q, Komarzynski S, Bolborea M, Finkenstadt B, Levi FA, 2021. Telemonitored human circadian temperature dynamics during daily routine. Front. Physiol 12, 659973.
    1. Janelidze S, et al., 2020. Plasma P-tau181 in Alzheimer’s disease: relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer’s dementia. Nat. Med 26, 379–386.
    1. Johns MW, 1991. A new method for measuring daytime sleepiness: the Epworth sleepiness scale. Sleep 14, 540–545.
    1. Jordan J, Montgomery I, Trinder J, 1990. The effect of afternoon body heating on body temperature and slow wave sleep. Psychophysiology 27, 560–566.
    1. Karikari TK, et al., 2020. Blood phosphorylated tau 181 as a biomarker for Alzheimer’s disease: a diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol 19, 422–433.
    1. Kelly G, 2006. Body temperature variability (part 1): a review of the history of body temperature and its variability due to site selection, biological rhythms, fitness, and aging. Altern. Med. Rev 11, 278–293.
    1. Kelly GS, 2007. Body temperature variability (part 2): masking influences of body temperature variability and a review of body temperature variability in disease. Altern. Med. Rev 12, 49–62.
    1. Kenney WL, Munce TA, 2003. Invited review: aging and human temperature regulation. J. Appl. Physiol 1985 (95), 2598–2603.
    1. Kim E, Sakata K, Liao FF, 2017. Bidirectional interplay of HSF1 degradation and UPR activation promotes tau hyperphosphorylation. PLoS Genet 13, e1006849.
    1. Kimura T, et al., 2014. Microtubule-associated protein tau is essential for long-term depression in the hippocampus. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci 369, 20130144.
    1. Knekt P, Jarvinen R, Rissanen H, Heliovaara M, Aromaa A, 2020. Does sauna bathing protect against dementia? Prev. Med. Rep 20, 101221.
    1. Krauchi K, Wirz-Justice A, 1994. Circadian rhythm of heat production, heart rate, and skin and core temperature under unmasking conditions in men. Am. J. Phys 267, R819–R829.
    1. Laing KK, et al., 2020. Cerebrovascular disease promotes tau pathology in Alzheimer’s disease. Brain Commun 2, fcaa132.
    1. Lee J, et al., 2020. Cerebrospinal fluid biomarkers for the diagnosis and classification of Alzheimer’s disease Spectrum. J. Korean Med. Sci 35, e361.
    1. Lindwall G, Cole RD, 1984. Phosphorylation affects the ability of tau protein to promote microtubule assembly. J. Biol. Chem 259, 5301–5305.
    1. Luppi M, et al., 2019. Phosphorylation and Dephosphorylation of tau protein during synthetic torpor. Front. Neuroanat 13, 57.
    1. Mishima K, et al., 1997. Different manifestations of circadian rhythms in senile dementia of Alzheimer’s type and multi-infarct dementia. Neurobiol. Aging 18, 105–109.
    1. Monnard CR, et al., 2017. Issues in continuous 24-h Core body temperature monitoring in humans using an ingestible capsule telemetric sensor. Front Endocrinol (Lausanne) 8, 130.
    1. Morrison SF, Nakamura K, 2019. Central mechanisms for thermoregulation. Annu. Rev. Physiol 81, null.
    1. Oh J, et al., 2019. Profound degeneration of wake-promoting neurons in Alzheimer’s disease. Alzheimers Dement 15, 1253–1263.
    1. Okawa M, et al., 1991. Circadian rhythm disorders in sleep-waking and body temperature in elderly patients with dementia and their treatment. Sleep 14, 478–485.
    1. Ossenkoppele R, et al., 2016. Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer’s disease. Brain 139, 1551–1567.
    1. Ossenkoppele R, et al., 2021. Accuracy of tau positron emission tomography as a prognostic marker in preclinical and prodromal Alzheimer disease: a head-to-head comparison against amyloid positron emission tomography and magnetic resonance imaging. JAMA Neurol 78, 961–971.
    1. Parekh A, 2021. In: Obeid I, Selesnick I, Picone J (Eds.), Nonlinear Smoothing of Core Body Temperature Data with Random Gaps and Outliers (DRAGO). In Biomedical Signal Processing Springer International Publishing, pp. 63–84.
    1. Pascoal TA, et al., 2017. Amyloid-beta and hyperphosphorylated tau synergy drives metabolic decline in preclinical Alzheimer’s disease. Mol. Psychiatry 22, 306–311.
    1. Pascoal TA, et al., 2018. In vivo quantification of neurofibrillary tangles with [(18)F] MK-6240. Alzheimers Res. Ther 10, 74.
    1. Pascoal TA, et al., 2020. 18F-MK-6240 PET for early and late detection of neurofibrillary tangles. Brain 143, 2818–2830.
    1. Planel E, Yasutake K, Fujita SC, Ishiguro K, 2001. Inhibition of protein phosphatase 2A overrides tau protein kinase I/glycogen synthase kinase 3 beta and cyclin-dependent kinase 5 inhibition and results in tau hyperphosphorylation in the hippocampus of starved mouse. J. Biol. Chem 276, 34298–34306.
    1. Planel E, et al., 2004. Alterations in glucose metabolism induce hypothermia leading to tau hyperphosphorylation through differential inhibition of kinase and phosphatase activities: implications for Alzheimer’s disease. J. Neurosci 24, 2401–2411.
    1. Planel E, et al., 2007. Anesthesia leads to tau hyperphosphorylation through inhibition of phosphatase activity by hypothermia. J. Neurosci 27, 3090–3097.
    1. Planel E, et al., 2008. Anesthesia-induced hyperphosphorylation detaches 3-repeat tau from microtubules without affecting their stability in vivo. J. Neurosci 28, 12798–12807.
    1. Planel E, et al., 2009. Acceleration and persistence of neurofibrillary pathology in a mouse model of tauopathy following anesthesia. FASEB J 23, 2595–2604.
    1. Preussner M, et al., 2017. Body temperature cycles control rhythmic alternative splicing in mammals. Mol. Cell 67, 433–446 e434.
    1. Prinz PN, et al., 1984. Circadian temperature variation in healthy aged and in Alzheimer’s disease. J. Gerontol 39, 30–35.
    1. Rasmussen MK, Mestre H, Nedergaard M, 2018. The glymphatic pathway in neurological disorders. Lancet Neurol 17, 1016–1024.
    1. Refinetti R, 2010. The circadian rhythm of body temperature. Front Biosci. (Landmark Ed) 15, 564–594.
    1. Refinetti R, Lissen GC, Halberg F, 2007. Procedures for numerical analysis of circadian rhythms. Biol. Rhythm. Res 38, 275–325.
    1. Rousset OG, Ma Y, Evans AC, 1998. Correction for partial volume effects in PET: principle and validation. J. Nucl. Med 39, 904–911.
    1. Rub U, et al., 2016. The brainstem tau cytoskeletal pathology of Alzheimer’s disease: a brief historical overview and description of its anatomical distribution pattern, evolutional features, Pathogenetic and clinical relevance. Curr. Alzheimer Res 13, 1178–1197.
    1. Satlin A, Volicer L, Stopa EG, Harper D, 1995. Circadian locomotor activity and core-body temperature rhythms in Alzheimer’s disease. Neurobiol. Aging 16, 765–771.
    1. Scheltens P, et al., 2021. Alzheimer’s disease. Lancet 397, 1577–1590.
    1. Shapiro CM, Allan M, Driver H, Mitchell D, 1989. Thermal load alters sleep. Biol. Psychiatry 26, 736–740.
    1. Shukla V, Skuntz S, Pant HC, 2012. Deregulated Cdk5 activity is involved in inducing Alzheimer’s disease. Arch. Med. Res 43, 655–662.
    1. Somero GN, 1975. Temperature as a selective factor in protein evolution: the adaptational strategy of “compromise”. J. Exp. Zool 194, 175–188.
    1. Somero GN, Low PS, 1976. Temperature: a ‘shaping force’ in protein evolution. Biochem. Soc. Symp 33–42.
    1. Somero GN, Lockwood BL, Tomanek L, 2017. Biochemical Adaptation : Response to Environmental Challenges, from life’s Origins to the Anthropocene Sinauer Associates, Inc. Publishers, Sunderland, Massachusetts.
    1. Sontag JM, Sontag E, 2014. Protein phosphatase 2A dysfunction in Alzheimer’s disease. Front. Mol. Neurosci 7, 16.
    1. Spiegel J, et al., 2016. Greater specificity for cerebrospinal fluid P-tau231 over P-tau181 in the differentiation of healthy controls from Alzheimer’s disease. J. Alzheimer’s Dis. : JAD 49, 93–100.
    1. Stieler JT, et al., 2011. The physiological link between metabolic rate depression and tau phosphorylation in mammalian hibernation. PLoS One 6, e14530.
    1. Stratmann K, et al., 2016. Precortical phase of Alzheimer’s disease (AD)-related tau cytoskeletal pathology. Brain Pathol 26, 371–386.
    1. Sund-Levander M, Forsberg C, Wahren LK, 2002. Normal oral, rectal, tympanic and axillary body temperature in adult men and women: a systematic literature review. Scand. J. Caring Sci 16, 122–128.
    1. Swaab DF, et al., 1992. Tau and ubiquitin in the human hypothalamus in aging and Alzheimer’s disease. Brain Res 590, 239–249.
    1. Touitou Y, et al., 1986. Age-related changes in both circadian and seasonal rhythms of rectal temperature with special reference to senile dementia of Alzheimer type. Gerontology 32, 110–118.
    1. Tournissac M, Vandal M, Francois A, Planel E, Calon F, 2017. Old age potentiates cold-induced tau phosphorylation: linking thermoregulatory deficit with Alzheimer’s disease. Neurobiol. Aging 50, 25–29.
    1. Tournissac M, et al., 2019. Repeated cold exposures protect a mouse model of Alzheimer’s disease against cold-induced tau phosphorylation. Mol. Metab 22, 110–120.
    1. Travers GJ, Nichols DS, Farooq A, Racinais S, Periard JD, 2016. Validation of an ingestible temperature data logging and telemetry system during exercise in the heat. Temperature (Austin) 3, 208–219.
    1. Tsukamoto D, et al., 2019. Circadian transcription factor HSF1 regulates differential HSP70 gene transcription during the arousal-torpor cycle in mammalian hibernation. Sci. Rep 9, 832.
    1. Tupone D, Madden CJ, Cano G, Morrison SF, 2011. An orexinergic projection from perifornical hypothalamus to raphe pallidus increases rat brown adipose tissue thermogenesis. J. Neurosci 31, 15944–15955.
    1. Uematsu M, et al., 2018. Brainstem tau pathology in Alzheimer’s disease is characterized by increase of three repeat tau and independent of amyloid beta. Acta Neuropathol. Commun 6, 1.
    1. van der Kant R, Goldstein LSB, Ossenkoppele R, 2020. Amyloid-beta-independent regulators of tau pathology in Alzheimer disease. Nat. Rev. Neurosci 21, 21–35.
    1. Van Someren EJ, Raymann RJ, Scherder EJ, Daanen HA, Swaab DF, 2002. Circadian and age-related modulation of thermoreception and temperature regulation: mechanisms and functional implications. Ageing Res. Rev 1, 721–778.
    1. Vandal M, et al., 2016. Impaired thermoregulation and beneficial effects of thermoneutrality in the 3xTg-AD model of Alzheimer’s disease. Neurobiol. Aging 43, 47–57.
    1. Vanderstichele H, et al., 2012. Standardization of preanalytical aspects of cerebrospinal fluid biomarker testing for Alzheimer’s disease diagnosis: a consensus paper from the Alzheimer’s biomarkers standardization initiative. Alzheimers Dement 8, 65–73.
    1. Vershinin M, Carter BC, Razafsky DS, King SJ, Gross SP, 2007. Multiple-motor based transport and its regulation by tau. Proc. Natl. Acad. Sci. U. S. A 104, 87–92.
    1. Volicer L, Harper DG, Stopa EG, 2012. Severe impairment of circadian rhythm in Alzheimer’s disease. J. Nutr. Health Aging 16, 888–890.
    1. Waalen J, Buxbaum JN, 2011. Is older colder or colder older? The association of age with body temperature in 18,630 individuals. J. Gerontol. A Biol. Sci. Med. Sci 66, 487–492.
    1. Wegmann S, Biernat J, Mandelkow E, 2021. A current view on tau protein phosphorylation in Alzheimer’s disease. Curr. Opin. Neurobiol 69, 131–138.
    1. Weinert D, 2010. Circadian temperature variation and ageing. Ageing Res. Rev 9, 51–60.
    1. Weitzman ED, Moline ML, Czeisler CA, Zimmerman JC, 1982. Chronobiology of aging: temperature, sleep-wake rhythms and entrainment. Neurobiol. Aging 3, 299–309.
    1. Yap SY, et al., 2021. Discriminatory ability of next-generation tau PET tracers for Alzheimer’s disease. Brain 144, 2284–2290.
    1. Zhu K, et al., 2019. Primary age-related Tauopathy in human subcortical nuclei. Front. Neurosci 13, 529.

Source: PubMed

3
Subscribe