Implication of Thyroid Hormones in the Development of the Pathological Characteristics of Alzheimer’s Disease; A Mini Review

Special Article - Thyroid Gland

Annals Thyroid Res. 2019; 5(2): 211-214.

Implication of Thyroid Hormones in the Development of the Pathological Characteristics of Alzheimer’s Disease; A Mini Review

Carrera-González MP* and Cantón Habas V

Department of Nursing, Reina Sofía University Hospital, Spain

*Corresponding author: Carrera Gonzalez MP, Department of Nursing, Faculty of Medicine and Nursing, Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofía University Hospital, Av. Menéndez Pidal, 7, 14004 Córdoba, Spain

Received: June 10, 2019; Accepted: July 04, 2019; Published: July 11, 2019

Abstract

Thyroid hormones play key roles in the central nervous system. Indeed, thyroid dysfunctions, either in the form of hypothyroidism or hyperthyroidism, have been recognized as risk factors for the progression of irreversible dementia, suggested by the epidemiological findings, being hypothyroidism is a condition that becomes more prevalent with age. The aging of the population is leading to the appearance of neurodegenerative diseases such as Alzheimer’s disease. Alzheimer’s disease is a multifactorial neurodegenerative disease which occurs relatively later in life (onset > 65 years of age, 95% of cases.). It involves a combination of aggregated proteins, chronic neuroinflammation and neuronal cell loss. Several studies have identified an association between dysregulation of thyroid hormones and Alzheimer’s disease dementia.

The aim of this review is to determine the involvement of thyroid hormones in the different hallkmarks that characterize Alzheimer’s disease.

Keywords: Thyroid hormone; Hypothyroidism; Alzheimer disease; Amyloid plaques

Introduction

The most widely identified association between endocrine and cognitive functions involves Thyroid Hormones (THs) [1]. Although the increase in longevity in western populations is our main achievement, it is a challenge for socioeconomic sustainability and a burden for health care systems since we are facing age-related diseases, as is the case of Alzheimer’s disease [2]. In this sense, TH dysregulation in advanced age is associated with risk for dementia [3-6]. Indeed, hypothyroidism is a condition that becomes more prevalent with age. Patients with untreated hypothyroidism have consistently reported symptoms of severe cognitive impairments. In patients suffering hypothyroidism, thyroid hormone supplementation offers the prospect to alleviate the cognitive consequences of hypothyroidism. However, the link between cellular modifications associated with hypothyroidism and neurodegeneration remains to be elucidated [7]. Thereby, T3 supplementation can alleviate hippocampaldependent memory impairments displayed by hypothyroid rats and normalize key markers of thyroid status in the hippocampus, of neuroinflammation, Aβ production, and of cell-signaling pathways known to be involved in synaptic plasticity and memory function [7]. On the other hand, the literature indicates a complicated relationship between TH and dementia risk so hyperthyroidism is usually more strongly associated with cognitive decline, although some reports indicate that hypothyroidism also is a risk factor for dementia [8-12].

Therefore, AD appears to lie on an intricate crosstalk between age-related metabolic, hormonal and specific genetic changes that challenge its traditional view [13].

THs have been implicated in practically all clinical features of AD. However, the mechanism underlying such involvement is unknown. In this review, we intend to describe the aforementioned relationship.

Alzheimer disease

Alzheimer’s Disease (AD) is the leading cause of dementia [14], affecting ~35 million people worldwide [15], ~2/3 of them are women [16]. Clinically, AD is a progressive neurodegenerative disease that typically begins with a subtle decline in the ability to encode new memories, then progressing towards a more profound cognitive, behavioural/personality and adaptive deterioration [17,18]. In this sense, there is evidence that TH regulates two of the main pathogenetic processes in AD, namely, tau protein phosphorylation [19] and the altered metabolism of amyloid precursor protein [20].

pClassic hallmark characteristics of AD pathology include Aβ plaques and oligomers, tau neurofibrillary tangles and progressive cholinergic neuron degeneration [21]. Also, proliferation and activation of microglia in the brain, concentrated around amyloid plaques, is a prominent feature of AD [22].

Amyloid Precursor Protein (app) processing and thyroid hormones

Extracellular senile plaques arises from the amyloidogenic proteolytic processing of Amyloid Precursor Protein (APP), sequentially catalysed by the aspartic protease β-site APP Cleaving Enzyme-1 (β-secretase or BACE1) and then by the γ-secretase complex [17,23,24]. Several studies have indicated that THs regulate the gene expression of APP [25,26]. Belandia et al. [27] reported that T3 negatively regulates APP gene expression in a rat neuroblastoma cell line. The results of various studies suggest that decrease in TH receptor levels occur in Alzheimer hippocampus cells. Moreover, the effects of estradiol or retinoic acid on APP expression and metabolism suggested that members of the nuclear superfamily of receptors and their ligands might play the main role in AD [27]. In this context, it has been suggested that low CNS TH levels may predispose to AD via increasing APP expression and consequently, Aβ peptide and β- amyloid levels [1]. Indeed, preclinical studies have repeatedly found an association between thyroid hormones and brain Aβ deposition in mice [28-30] and in human brain-derived neuroblastoma cells [30].

In the last years different authors found that Aβ impairs hormonemediated signalling, mitochondrial (energy) metabolism and calcium homeostasis, leading to oxidative/endoplasmic reticulum stresses, microglia activation/inflammation and endotelial dysfunction, culminating in synaptic/neuronal loss and cognitive deficits [31]. In this sense, there is evidence that TH regulates the altered metabolism of APP [20]. In addition, hypothyroidism interferes in the maintenance of normal energy (glucose)-consuming processes needed for essential brain functions such as neurotransmission and memory [32], harming cognition.

Recent evidence suggests that microglia form a protective barrier around amyloid deposits, compacting amyloid fibrils into a tightly packed and potentially less toxic form, preventing the accretion of new Aβ onto existing plaques, and reducing axonal dystrophy in the nearby neuropil [33]. On the other hand, there is also evidence that activated microglia can be harmful to neurons [22]. Proliferation and activation of microglia in the brain, concentrated around amyloid plaques, is a prominent feature of Alzheimer’s Disease (AD). There is mounting evidence that microglia protect against the incidence of AD, as impaired microglial activities and altered microglial responses to β-amyloid are associated with increased AD risk [22]. However, we have not found detailed studies of the possible involvement of thyroid hormones in the process of activating microglia in AD, although its relevance in relation to amyloid plaques is definitive [1].

Tau protein phosphorylation and thyroid hormones

AD is determined pathologically by alterations in the brain including the formation of intracellular neurofibrillary tangles of hyperphosphorylated Tau [1]. Tau is an essential protein from the family of Microtubule-Associated Proteins (MAPs) that physiologically interacts with tubulin [34] to maintain axonal diameter [35] and microtubule stability [29,34,36-38], thus maintaining neuronal structure and axonal transport of synaptic vesicles [39]. However, P-Tau tends to aggregate within neuronal perikaryal, interfering with microtubule network, promoting oxidative stress, mitocondrial dysfunction, neurodegeneration and death [24,39]. Strikingly, Aβ may also induce P-Tau, exacerbating neurodegeneration/ neuronal loss and brain atrophy in AD [24,31].

In this context, THs could affect the transcription of APP gene as well as the phosphorylation of Tau [27,28,30,40,41]. Indeed, THs may also interact with the amount and phosphorylation of Tau [41,42], however, the mechanism involved is still unknown.

Cholinergic disfuction and thyroid hormones

The cortical cholinergic dysfunction begins in midlife and is closely followed by increases in deposition of cortical Aβ levels [43- 45]. In neocortex and hippocampus of AD brain, a harm of cholinergic fibers and terminals, declines in cholinergic receptors and signal transduction, and significant decreases in choline acetyltransferase and increase in acetylcholinesterase enzyme activities have been reported [46,47].

The thyroid function has a central role in both neurodevelopment process and neurodegenerative processes. So it has been proposed that the thyroid dysfunction enhances the risk of AD by a direct harmful effect of thyroxine discharge on cholinergic neurons; however, whether the neuropathological mechanism of altered thyrotropin levels happens before or after the beginning of AD is unclear [48].

Bavarsad et al. [1] suggests a relationship between hypothyroidism and AD; reduced thyroxin level is associated with the preservation of cholinergic neurons [48], concentrations of lower T3 in the brain increase the expression of APP [30], and the TH receptor competitively increases the level of the seladin-1 gene which prevents the accumulation of β-amyloid plaques and cell death [49].

Therefore, thyroid dysfunctions, either in the form of hypothyroidism or hyperthyroidism, have been recognized as risk factors for the progression of irreversible dementia as suggested by the epidemiological findings [50,51]. In this sense, several reports suggest that subclinical abnormalities in thyroid function may play a role in AD [52-54].

Recent studies developed by Quinlan et al. [55], showed that serum FT3 levels were inversely associated with the risk of progression to AD, with a more than doubled risk of subsequent AD in the lowest FT3 quartile compared to the highest quartile. However there was no association between serum FT3 and all-cause dementia or vascular dementia, and serum levels of TSH and FT4 did not associate with the risk of conversion to dementia of any type. In this sense, authors suggest that supplementation with THs could be of use in patients with prodromal AD.

Conclusion

In view of the collected studies we can conclude that the alteration of thyroid hormones is involved in the development of Alzheimer’s disease. In fact, they seem to be implicated in the processes underlying the appearance of hallmarks of Alzheimer’s disease. However, based on current lack of knowledge of the specific pathophysiological mechanisms of Alzheimer’s disease, as well as the involvement of thyroid hormones in the related cerebral metabolic pathways, a greater number of studies are needed in our view to delve into each one of these points.

References

  1. Bavarsad K, Hosseini M, Hadjzadeh MA, Sahebkar A. The effects of thyroid hormones on memory impairment and Alzheimer’s disease. Journal of cellular physiology. 2019.
  2. Tortelli R, Lozupone M, Guerra V, Barulli MR, Imbimbo BP, Capozzo R, et al. Midlife Metabolic Profile and the Risk of Late-Life Cognitive Decline. Journal of Alzheimer’s disease: JAD. 2017; 59: 121-130.
  3. Chaker L, Wolters FJ, Bos D, Korevaar TI, Hofman A, van der Lugt A, et al. Thyroid function and the risk of dementia: The Rotterdam Study. Neurology. 2016; 87: 1688-1695.
  4. Mafrica F, Fodale V. Thyroid function, Alzheimer’s disease and postoperative cognitive dysfunction: a tale of dangerous liaisons? Journal of Alzheimer’s disease: JAD. 2008; 14: 95-105.
  5. Sampaolo S, Campos-Barros A, Mazziotti G, Carlomagno S, Sannino V, Amato G, et al. Increased cerebrospinal fluid levels of 3,3’,5’-triiodothyronine in patients with Alzheimer’s disease. The Journal of clinical endocrinology and metabolism. 2005; 90: 198-202.
  6. Tan ZS, Vasan RS. Thyroid function and Alzheimer’s disease. Journal of Alzheimer’s disease: JAD. 2009; 16: 503-507.
  7. Chaalal A, Poirier R, Blum D, Laroche S, Enderlin V. Thyroid Hormone Supplementation Restores Spatial Memory, Hippocampal Markers of Neuroinflammation, Plasticity-Related Signaling Molecules, and beta- Amyloid Peptide Load in Hypothyroid Rats. Molecular neurobiology. 2019; 56: 722-735.
  8. Akintola AA, Jansen SW, van Bodegom D, van der Grond J, Westendorp RG, de Craen AJ, et al. Subclinical hypothyroidism and cognitive function in people over 60 years: a systematic review and meta-analysis. Frontiers in aging neuroscience. 2015; 7: 150.
  9. Moon JH, Ahn S, Seo J, Han JW, Kim KM, Choi SH, et al. The effect of long-term thyroid-stimulating hormone suppressive therapy on the cognitive function of elderly patients with differentiated thyroid carcinoma. The Journal of clinical endocrinology and metabolism. 2014; 99: 3782-3789.
  10. Pasqualetti G, Pagano G, Rengo G, Ferrara N, Monzani F. Subclinical Hypothyroidism and Cognitive Impairment: Systematic Review and Meta- Analysis. The Journal of clinical endocrinology and metabolism. 2015; 100: 4240-4248.
  11. Rieben C, Segna D, da Costa BR, Collet TH, Chaker L, Aubert CE, et al. Subclinical Thyroid Dysfunction and the Risk of Cognitive Decline: a Meta- Analysis of Prospective Cohort Studies. The Journal of clinical endocrinology and metabolism. 2016; 101: 4945-4954.
  12. Yeap BB, Alfonso H, Chubb SA, Puri G, Hankey GJ, Flicker L, et al. Higher free thyroxine levels predict increased incidence of dementia in older men: the Health in Men Study. The Journal of clinical endocrinology and metabolism. 2012; 97: E2230-2237.
  13. Duarte AI, Santos MS, Oliveira CR, Moreira PI. Brain insulin signalling, glucose metabolism and females’ reproductive aging: A dangerous triad in Alzheimer’s disease. Neuropharmacology. 2018; 136: 223-242.
  14. World Health Organization. Dementia Fact Sheet. 2017.
  15. Alzheimer’s Association. 2017 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association. 2017; 13: 325-373.
  16. Brookmeyer R, Evans DA, Hebert L, Langa KM, Heeringa SG, Plassman BL, et al. National estimates of the prevalence of Alzheimer’s disease in the United States. Alzheimer’s & dementia: the journal of the Alzheimer’s Association. 2011; 7: 61-73.
  17. Neth BJ, Craft S. Insulin Resistance and Alzheimer’s Disease: Bioenergetic Linkages. Frontiers in aging neuroscience. 2017; 9: 345.
  18. Yu Q, Du F, Douglas JT, Yu H, Yan SS, Yan SF. Mitochondrial Dysfunction Triggers Synaptic Deficits via Activation of p38 MAP Kinase Signaling in Differentiated Alzheimer’s Disease Trans-Mitochondrial Cybrid Cells. Journal of Alzheimer’s disease: JAD. 2017; 59: 223-239.
  19. Luo L, Stopa EG. Thyrotropin releasing hormone inhibits tau phosphorylation by dual signaling pathways in hippocampal neurons. Journal of Alzheimer’s disease: JAD. 2004; 6: 527-536.
  20. Latasa MJ, Belandia B, Pascual A. Thyroid hormones regulate beta-amyloid gene splicing and protein secretion in neuroblastoma cells. Endocrinology. 1998; 139: 2692-2698.
  21. Singh Y, Gupta G, Shrivastava B, Dahiya R, Tiwari J, Ashwathanarayana M, et al. Calcitonin gene-related peptide (CGRP): A novel target for Alzheimer’s disease. CNS neuroscience & therapeutics. 2017; 23: 457-461.
  22. Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer’s disease. The Journal of cell biology. 2018; 217: 459-472.
  23. Blonz ER. Alzheimer’s Disease as the Product of a Progressive Energy Deficiency Syndrome in the Central Nervous System: The Neuroenergetic Hypothesis. Journal of Alzheimer’s disease: JAD. 2017; 60: 1223-1229.
  24. Kandimalla R, Reddy PH. Therapeutics of Neurotransmitters in Alzheimer’s Disease. Journal of Alzheimer’s disease: JAD. 2017; 57: 1049-1069.
  25. Chen Y, Huang X, Zhang YW, Rockenstein E, Bu G, Golde TE, et al. Alzheimer’s beta-secretase (BACE1) regulates the cAMP/PKA/CREB pathway independently of beta-amyloid. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2012; 32: 11390-11395.
  26. Goumidi L, Flamant F, Lendon C, Galimberti D, Pasquier F, Scarpini E, et al. Study of thyroid hormone receptor alpha gene polymorphisms on Alzheimer’s disease. Neurobiology of aging. 2011; 32: 624-630.
  27. Belandia B, Latasa MJ, Villa A, Pascual A. Thyroid hormone negatively regulates the transcriptional activity of the beta-amyloid precursor protein gene. The Journal of biological chemistry. 1998; 273: 30366-30371.
  28. O’Barr SA, Oh JS, Ma C, Brent GA, Schultz JJ. Thyroid hormone regulates endogenous amyloid-beta precursor protein gene expression and processing in both in vitro and in vivo models. Thyroid: official journal of the American Thyroid Association. 2006; 16: 1207-1213.
  29. Gustke N, Trinczek B, Biernat J, Mandelkow EM, Mandelkow E. Domains of tau protein and interactions with microtubules. Biochemistry. 1994; 33: 9511- 9522.
  30. Belakavadi M, Dell J, Grover GJ, Fondell JD. Thyroid hormone suppression of beta-amyloid precursor protein gene expression in the brain involves multiple epigenetic regulatory events. Molecular and cellular endocrinology. 2011; 339: 72-80.
  31. Bharadwaj P, Wijesekara N, Liyanapathirana M, Newsholme P, Ittner L, Fraser P, et al. The Link between Type 2 Diabetes and Neurodegeneration: Roles for Amyloid-beta, Amylin, and Tau Proteins. Journal of Alzheimer’s disease: JAD. 2017; 59: 421-432.
  32. Begin ME, Langlois MF, Lorrain D, Cunnane SC. Thyroid Function and Cognition during Aging. Current gerontology and geriatrics research. 2008: 474868.
  33. Condello C, Yuan P, Schain A, Grutzendler J. Microglia constitute a barrier that prevents neurotoxic protofibrillar Aβ42 hotspots around plaques. Nature communications. 2015; 6: 6176.
  34. Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW. A protein factor essential for microtubule assembly. Proceedings of the National Academy of Sciences of the United States of America. 1975; 72: 1858-1862.
  35. Chen J, Kanai Y, Cowan NJ, Hirokawa N. Projection domains of MAP2 and tau determine spacings between microtubules in dendrites and axons. Nature. 1992; 360: 674-677.
  36. Cleveland DW, Hwo SY, Kirschner MW. Purification of tau, a microtubuleassociated protein that induces assembly of microtubules from purified tubulin. Journal of molecular biology. 1977; 116: 207-225.
  37. Cleveland DW, Hwo SY, Kirschner MW. Physical and chemical properties of purified tau factor and the role of tau in microtubule assembly. Journal of molecular biology. 1977; 116: 227-247.
  38. Butner KA, Kirschner MW. Tau protein binds to microtubules through a flexible array of distributed weak sites. The Journal of cell biology. 1991; 115: 717-730.
  39. De-Paula VJ, Radanovic M, Diniz BS, Forlenza OV. Alzheimer’s disease. Sub-cellular biochemistry. 2012; 65: 329-352.
  40. Contreras-Jurado C, Pascual A. Thyroid hormone regulation of APP (β-amyloid precursor protein) gene expression in brain and brain cultured cells. Neurochemistry international. 2012; 60: 484-487.
  41. Oyanagi K, Negishi T, Tashiro T. Action of thyroxine on the survival and neurite maintenance of cerebellar granule neurons in culture. Journal of neuroscience research. 2015; 93: 592-603.
  42. O’Dowd ST, Ardah MT, Johansson P, Lomakin A, Benedek GB, Roberts KA, et al. The ELISA-measured increase in cerebrospinal fluid tau that discriminates Alzheimer’s disease from other neurodegenerative disorders is not attributable to differential recognition of tau assembly forms. Journal of Alzheimer’s disease: JAD. 2013; 33: 923-928.
  43. Perry EK, Johnson M, Kerwin JM, Piggott MA, Court JA, Shaw PJ, et al. Convergent cholinergic activities in aging and Alzheimer’s disease. Neurobiology of aging. 1992; 13: 393-400.
  44. Funato H, Yoshimura M, Kusui K, Tamaoka A, Ishikawa K, Ohkoshi N, et al. Quantitation of amyloid beta-protein (A beta) in the cortex during aging and in Alzheimer’s disease. The American journal of pathology. 1998; 152: 1633-1640.
  45. Dewachter I, Van Dorpe J, Smeijers L, Gilis M, Kuiperi C, Laenen I, et al. Aging increased amyloid peptide and caused amyloid plaques in brain of old APP/V717I transgenic mice by a different mechanism than mutant presenilin1. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2000; 20: 6452-6458.
  46. Coyle JT, Price DL, DeLong MR. Alzheimer’s disease: a disorder of cortical cholinergic innervation. Science. 1983; 219: 1184-1190.
  47. Wightman EL. Potential benefits of phytochemicals against Alzheimer’s disease. The Proceedings of the Nutrition Society. 2017; 76: 106-112.
  48. Zhang N, Du HJ, Wang JH, Cheng Y. A pilot study on the relationship between thyroid status and neuropsychiatric symptoms in patients with Alzheimer disease. Chinese medical journal. 2012; 125: 3211-3216.
  49. Benvenuti S, Luciani P, Cellai I, Deledda C, Baglioni S, Saccardi R, et al. Thyroid hormones promote cell differentiation and up-regulate the expression of the seladin-1 gene in in vitro models of human neuronal precursors. The Journal of endocrinology. 2008; 197: 437-446.
  50. Kalmijn S, Mehta KM, Pols HA, Hofman A, Drexhage HA, Breteler MM. Subclinical hyperthyroidism and the risk of dementia. The Rotterdam study. Clinical endocrinology. 2000; 53: 733-737.
  51. van Osch LA, Hogervorst E, Combrinck M, Smith AD. Low thyroid-stimulating hormone as an independent risk factor for Alzheimer disease. Neurology. 2004; 62: 1967-1971.
  52. Volpato S, Guralnik JM, Fried LP, Remaley AT, Cappola AR, Launer LJ. Serum thyroxine level and cognitive decline in euthyroid older women. Neurology. 2002; 58: 1055-1061.
  53. Ganguli M, Burmeister LA, Seaberg EC, Belle S, DeKosky ST. Association between dementia and elevated TSH: a community-based study. Biological psychiatry. 1996; 40: 714-725.
  54. Davis JD, Podolanczuk A, Donahue JE, Stopa E, Hennessey JV, Luo LG, et al. Thyroid hormone levels in the prefrontal cortex of post-mortem brains of Alzheimer’s disease patients. Current aging science. 2008; 1: 175-181.
  55. Quinlan P, Horvath A, Wallin A, Svensson J. Low serum concentration of free triiodothyronine (FT3) is associated with increased risk of Alzheimer’s disease. Psychoneuroendocrinology. 2019; 99: 112-119.

Download PDF

Citation: Carrera-González MP and Cantón Habas V. Implication of Thyroid Hormones in the Development of the Pathological Characteristics of Alzheimer’s Disease; A Mini Review. Annals Thyroid Res. 2019; 5(2): 211-214.

Home
Journal Scope
Online First
Current Issue
Editorial Board
Instruction for Authors
Submit Your Article
Contact Us