Horm Metab Res
DOI: 10.1055/a-2238-1384
Review

Targeting Abnormal Tau Phosphorylation for Alzheimer’s Therapeutics

Aditya Singh
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Vaseem Ahamad Ansari
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Tarique Mahmood
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Syed Misbahul Hasan
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Rufaida Wasim
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Shubhrat Maheshwari
2   Faculty of Pharmaceutical Sciences, Rama University, Kanpur, India
,
Juber Akhtar
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Suvaiv Sheikh
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Vishal Kumar Vishwakarma
3   Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
› Author Affiliations

Abstract

Alzheimer’s disease (AD) is a widespread neurodegenerative disorder characterized by progressive memory and cognitive decline, posing a formidable public health challenge. This review explores the intricate interplay between two pivotal players in AD pathogenesis: β-amyloid (Aβ) and tau protein. While the amyloid cascade theory has long dominated AD research, recent developments have ignited debates about its centrality. Aβ plaques and tau NFTs are hallmark pathologies in AD. Aducanumab and lecanemab, monoclonal antibodies targeting Aβ, have been approved, albeit amidst controversy, raising questions about the therapeutic efficacy of Aβ-focused interventions. On the other hand, tau, specifically its hyperphosphorylation, disrupts microtubule stability and contributes to neuronal dysfunction. Various post-translational modifications of tau drive its aggregation into NFTs. Emerging treatments targeting tau, such as GSK-3β and CDK5 inhibitors, have shown promise in preclinical and clinical studies. Restoring the equilibrium between protein kinases and phosphatases, notably protein phosphatase-2A (PP2A), is a promising avenue for AD therapy, as tau is primarily regulated by its phosphorylation state. Activation of tau-specific phosphatases offers potential for mitigating tau pathology. The evolving landscape of AD drug development emphasizes tau-centric therapies and reevaluation of the amyloid cascade hypothesis. Additionally, exploring the role of neuroinflammation and its interaction with tau pathology present promising research directions.



Publication History

Received: 10 September 2023

Accepted after revision: 15 December 2023

Article published online:
13 February 2024

© 2024. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Scotti L, Bassi L, Soranna D. et al. Association between renin-angiotensin-aldosterone system inhibitors and risk of dementia: a meta-analysis. Pharmacol Res 2021; 166: 105515
  • 2 Ou YN, Tan CC, Shen XN. et al. Blood pressure and risks of cognitive impairment and dementia: a systematic review and meta-analysis of 209 prospective studies. Hypertension 2020; 76: 217-225
  • 3 Mourao RJ, Mansur G, Malloy-Diniz LF. et al. Depressive symptoms increase the risk of progression to dementia in subjects with mild cognitive impairment: systematic review and meta-analysis. Int J Geriatr Psychiatry 2016; 31: 905-911
  • 4 Fu C, Wu Y, Liu S. et al. Rehmannioside A improves cognitive impairment and alleviates ferroptosis via activating PI3K/AKT/Nrf2 and SLC7A11/GPX4 signaling pathway after ischemia. J Ethnopharmacol 2022; 289: 115021
  • 5 Aum S, Choe S, Cai M. et al. Moxibustion for cognitive impairment: a systematic review and meta-analysis of animal studies. Integr Med Res 2021; 10: 100680
  • 6 Schrag M, Mueller C, Zabel M. et al. Oxidative stress in blood in Alzheimer's disease and mild cognitive impairment: a meta-analysis. Neurobiol Dis 2013; 59: 100-110
  • 7 Zhu LN, Mei X, Zhang ZG. et al. Curcumin intervention for cognitive function in different types of people: a systematic review and meta-analysis. Phytother Res 2019; 33: 524-533
  • 8 Sexton CE, Kalu UG, Filippini N. et al. A meta-analysis of diffusion tensor imaging in mild cognitive impairment and Alzheimer’s disease. Neurobiol Aging 2011; 32: 2322-e5
  • 9 Hamilton OK, Backhouse EV, Janssen E. et al. Cognitive impairment in sporadic cerebral small vessel disease: a systematic review and meta-analysis. Alzheimer Dement 2021; 17: 665-685
  • 10 Hampel H, Caraci F, Cuello AC. et al. A path toward precision medicine for neuroinflammatory mechanisms in Alzheimer’s disease. Front Immunol 2020; 11: 456
  • 11 Lacour A, Espinosa A, Louwersheimer E. et al. Genome-wide significant risk factors for Alzheimer’s disease: role in progression to dementia due to Alzheimer’s disease among subjects with mild cognitive impairment. Mol Psychiatry 2017; 22: 153-160
  • 12 Adani G, Filippini T, Michalke B. et al. Selenium and other trace elements in the etiology of Parkinson’s disease: a systematic review and meta-analysis of case-control studies. Neuroepidemiology 2020; 54: 1-23
  • 13 Zhang J, Sun P, Zhou C. et al. Regulatory microRNAs and vascular cognitive impairment and dementia. CNS Neurosci Therap 2020; 26: 1207-1218
  • 14 Su W, Xie M, Li Y. et al. Topiramate reverses physiological and behavioral alterations by postoperative cognitive dysfunction in rat model through inhibiting TNF signaling pathway. NeuroMol Med 2020; 22: 227-238
  • 15 Su C, Zhao K, Xia H. et al. Peripheral inflammatory biomarkers in Alzheimer’s disease and mild cognitive impairment: a systematic review and meta-analysis. Psychogeriatrics 2019; 19: 300-309
  • 16 Wang J, Zhang T, Liu X. et al. Aqueous extracts of se-enriched Auricularia auricular attenuates D-galactose-induced cognitive deficits, oxidative stress and neuroinflammation via suppressing RAGE/MAPK/NF-κB pathway. Neurosci Lett 2019; 704: 106-111
  • 17 Tan MM, Lawton MA, Jabbari E. et al. Genome-wide association studies of cognitive and motor progression in Parkinson’s disease. Movement Disord 2021; 36: 424-433
  • 18 Pang S, Li J, Zhang Y. et al. Meta-analysis of the relationship between the APOE gene and the onset of Parkinson’s disease dementia. Parkinson Dis. 2018
  • 19 Munteanu C, Munteanu D, Onose G. et al. Hydrogen sulfide (H2S)-therapeutic relevance in rehabilitation and balneotherapy. Systematic literature review and meta-analysis based on the PRISMA paradigm. Balneo PRM Res J 2021; 12: 176-195
  • 20 Zhang L, Li B, Yang J. et al. Meta-analysis: resistance training improves cognition in mild cognitive impairment. Int J Sports Med 2020; 41: 815-823
  • 21 Singh A, Ansari VA, Mahmood T. et al. Dendrimers: A neuroprotective lead in alzheimer disease: a review on its synthetic approach and applications. Drug Res (Stuttg) 2022; 72: 417-423
  • 22 Zhang L, Li B, Yang J. et al. Meta-analysis: resistance training improves cognition in mild cognitive impairment. Int J Sports Med 2020; 41: 815-823
  • 23 Mahmoudian Dehkordi S, Arnold M, Nho K. et al. Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease—an emerging role for gut microbiome. Alzheimer Dement 2019; 15: 76-92
  • 24 Liu PP, Xie Y, Meng XY. et al. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduc Target Therapy 2019; 4: 29
  • 25 Hankey GJ, Ford AH, Yi Q. et al. Effect of B vitamins and lowering homocysteine on cognitive impairment in patients with previous stroke or transient ischemic attack: a prespecified secondary analysis of a randomized, placebo-controlled trial and meta-analysis. Stroke 2013; 44: 2232-2239
  • 26 Emamian F, Khazaie H, Tahmasian M. et al. The association between obstructive sleep apnea and Alzheimer’s disease: a meta-analysis perspective. Front Aging Neurosci 2016; 8: 78
  • 27 Tahmasbi F, Mirghafourvand M, Shamekh A. et al. Effects of probiotic supplementation on cognitive function in elderly: a systematic review and meta-analysis. Aging Mental Health 2022; 26: 1778-1786
  • 28 Sherva R, Gross A, Mukherjee S. et al. Genome-wide association study of rate of cognitive decline in Alzheimer’s disease patients identifies novel genes and pathways. Alzheimer Dement 2020; 16: 1134-1145
  • 29 Tang CZ, Yang JT, Liu QH. et al. Up-regulated miR-192-5p expression rescues cognitive impairment and restores neural function in mice with depression via the Fbln2-mediated TGF-β1 signaling pathway. FASEB J 2019; 33: 606-618
  • 30 Sun MK, Alkon DL. et al. Neuro-regeneration therapeutic for Alzheimer’s dementia: perspectives on neurotrophic activity. Trends Pharmacol Sci 2019; 40: 655-668
  • 31 Wu J, Xiong Y, Xia X. et al. Can dementia risk be reduced by following the American Heart Association’s Life’s Simple 7? A systematic review and dose-response meta-analysis. Ageing Res Rev 2022; DOI: 10.1016/j.arr.2022.101788.
  • 32 Hampel H, Vergallo A, Caraci F. et al. Future avenues for Alzheimer’s disease detection and therapy: liquid biopsy, intracellular signaling modulation, systems pharmacology drug discovery. Neuropharmacology 2021; 185: 108081
  • 33 Hughes D, Judge C, Murphy R. et al. Association of blood pressure lowering with incident dementia or cognitive impairment: a systematic review and meta-analysis. JAMA 2020; 323: 1934-1944
  • 34 Van Maurik IS, Bakker ED, Van den Buuse S. et al. Psychosocial effects of corona measures on patients with dementia, mild cognitive impairment and subjective cognitive decline. Front Psychiatry 2020; 11: 585686
  • 35 Shang X, Zhu Z, Wang W. et al. The association between vision impairment and incidence of dementia and cognitive impairment: a systematic review and meta-analysis. Ophthalmology 2021; 128: 1135-1149
  • 36 Åhman HB, Cedervall Y, Kilander L. et al. Dual-task tests discriminate between dementia, mild cognitive impairment, subjective cognitive impairment, and healthy controls–a cross-sectional cohort study. BMC Geriatr 2020; 20: 1
  • 37 Nafti M, Sirois C, Kröger E. et al. Is benzodiazepine use associated with the risk of dementia and cognitive impairment–not dementia in older persons? The Canadian study of health and aging. Ann Pharmacother 2020; 54: 219-225
  • 38 Singh A, Ansari VA, Mahmood T. et al. Neurodegeneration: microglia: Nf-kappab signaling pathways. Drug Res (Stuttg) 2022; 72: 496-499
  • 39 Yim D, Yeo TY, Park MH. et al. Mild cognitive impairment, dementia, and cognitive dysfunction screening using machine learning. J Int Med Res 2020; 48 0300060520936881
  • 40 Qu Y, Hu HY, Ou YN. et al. Association of body mass index with risk of cognitive impairment and dementia: a systematic review and meta-analysis of prospective studies. Neurosci Biobehav Rev 2020; 115: 189-198
  • 41 Uemura MT, Maki T, Ihara M. et al. Brain microvascular pericytes in vascular cognitive impairment and dementia. Front Aging Neurosci 2020; 12: 80
  • 42 Gibson C, Goeman D, Pond D. et al. What is the role of the practice nurse in the care of people living with dementia, or cognitive impairment, and their support person (s)?: a systematic review. BMC Family Pract 2020; 21: 1-8
  • 43 Lyu F, Wu D, Wei C. et al. Vascular cognitive impairment and dementia in type 2 diabetes mellitus: an overview. Life Sci 2020; 254: 117771
  • 44 Rajji TK, Bowie CR, Herrmann N. et al. Design and rationale of the PACt-MD randomized clinical trial: prevention of Alzheimer’s dementia with cognitive remediation plus transcranial direct current stimulation in mild cognitive impairment and depression. J Alzheimer Dis 2020; 76: 733-751
  • 45 Hemmy LS, Linskens EJ, Silverman PC. et al. Brief cognitive tests for distinguishing clinical Alzheimer-type dementia from mild cognitive impairment or normal cognition in older adults with suspected cognitive impairment: a systematic review. Ann Intern Med 2020; 172: 678-687
  • 46 Neopane D, Ansari VA, Singh A. et al. Ferulic Acid: signaling pathways in aging. Drug Res (Stuttg) 2023; 73: 318-324
  • 47 Meiner Z, Ayers E, Verghese J. et al. Motoric cognitive risk syndrome: a risk factor for cognitive impairment and dementia in different populations. Ann Geriatr Med Res 2020; 24: 3
  • 48 Singh A, Ansari VA, Mahmood T. et al. Receptor for advanced glycation end products: dementia and cognitive impairment. Drug Res (Stuttg) 2023; 73: 247-250
  • 49 Singh A, Maheshwari S. et al. Dendrimers for neuro targeting. Int J Pharma Profess Res 2023; 14: 124-130
  • 50 Singh A, Ansari VA, Mahmood T. et al Emerging nanotechnology for the treatment of Alzheimer’s disease. CNS Neurol Disord Drug Targets 2023; DOI: 10.2174/1871527322666230501232815.
  • 51 Singh A, Ansari VA, Ansari TM. et al. Consequence of dementia and cognitive impairment by primary nucleation pathway. Horm Metab Res 2023; 55: 304-314
  • 52 Maheshwari S. AGEs RAGE Pathways: Alzheimer’s Disease. Drug Res (Stuttg) 2023; 73: 251-254
  • 53 Guo Y, Li S, Zeng LH. et al. Tau-targeting therapy in Alzheimer’s disease: critical advances and future opportunities. Ageing Neurodegener Di 2022; 2: 11 DOI: 10.20517/and.2022.16.
  • 54 Chen Y, Yu Y. et al. Tau and neuroinflammation in Alzheimer’s disease: interplay mechanisms and clinical translation. J Neuroinflamm 2023; 20: 1-21
  • 55 Roy RG, Mandal PK, Maroon JC. et al. Oxidative stress occurs prior to amyloid Aβ plaque formation and tau phosphorylation in Alzheimer’s disease: role of glutathione and metal ions. ACS Chem Neurosci 2023; 14: 2944-2954
  • 56 Bueno-Carrasco MT, Cuéllar J, Flydal MI. et al. Structural mechanism for tyrosine hydroxylase inhibition by dopamine and reactivation by Ser40 phosphorylation. Nat Commun 2022; 13: 74
  • 57 Hartz RA, Ahuja VT, Sivaprakasam P. et al. Design, structure–activity relationships, and in vivo evaluation of potent and brain-penetrant imidazo[1,2-b]pyridazines as glycogen synthase kinase-3β (GSK-3β) inhibitors. J Med Chem 2023; 66: 4231-4252
  • 58 Balboni B, Masi M, Rocchia W. et al. GSK-3β allosteric inhibition: a dead end or a new pharmacological frontier?. Int J Mol Sci 2023; 24: 7541
  • 59 Yang W, Xu QQ, Yuan Q. et al. Sulforaphene, a CDK5 inhibitor, attenuates cognitive deficits in a transgenic mouse model of Alzheimer’s disease via reducing Aβ deposition, tau hyperphosphorylation and synaptic dysfunction. Int Immunopharmacol 2023; 114: 109504
  • 60 Pao PC, Seo J, Lee A. et al. A Cdk5-derived peptide inhibits Cdk5/p25 activity and improves neurodegenerative phenotypes. Proc Natl Acad Sci U S A 2023; 120: e2217864120
  • 61 Tang W, Lin C, Yu Q. et al. Novel medicinal chemistry strategies targeting CDK5 for drug discovery. J Med Chem 2023; 66: 7140-7161
  • 62 Batra S, Jahan S, Ashraf A. et al. A review on cyclin-dependent kinase 5: an emerging drug target for neurodegenerative diseases. Int J Biol Macromol. 2023
  • 63 Jahan I, Adachi R, Egawa R. et al. CDK5/p35-dependent microtubule reorganization contributes to homeostatic shortening of the axon initial segment. J Neurosci 2023; 43: 359-372
  • 64 Li H, Zhao H, Hu T. et al. The Cdk5 inhibitor β-butyrolactone impairs reconsolidation of heroin-associated memory in the rat basolateral amygdala. Addict Biol 2023; 28: e13326
  • 65 Requejo-Aguilar R. et al. Cdk5 and aberrant cell cycle activation at the core of neurodegeneration. Neural Regen Res 2023; 18: 1186
  • 66 López-Grueso MJ, Padilla CA, Bárcena JA. et al. Deficiency of Parkinson’s related protein DJ-1 alters Cdk5 Signalling and induces neuronal death by aberrant cell cycle re-entry. Cell Mol Neurobiol 2023; 43: 757-769
  • 67 Eteläinen TS, Silva MC, Uhari-Väänänen JK. et al. A prolyl oligopeptidase inhibitor reduces tau pathology in cellular models and in mice with tauopathy. Sci Transl Med 2023; 15: eabq2915
  • 68 Kaur P, Khera A, Alajangi HK. et al. Role of tau in various tauopathies, treatment approaches, and emerging role of nanotechnology in neurodegenerative disorders. Mol Neurobiol 2023; 60: 1690-1720
  • 69 Christensen KR, Combs B, Richards C. et al. Phosphomimetics at Ser199/Ser202/Thr205 in tau impairs axonal transport in rat hippocampal neurons. Mol Neurobiol 2023; 60: 3423-3438
  • 70 Lv J, Shen X, Shen X. et al. NPLC0393 from Gynostemma pentaphyllum ameliorates Alzheimer’s disease-like pathology in mice by targeting protein phosphatase magnesium-dependent 1A phosphatase. Phytother Res 2023; 37: 4771-4790