Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jan 1.
Published in final edited form as: Cytoskeleton (Hoboken). 2023 Dec 21;81(1):116–121. doi: 10.1002/cm.21822

Tau and Alzheimer’s Disease: Past, Present and Future

Khalid Iqbal 1
PMCID: PMC10977900  NIHMSID: NIHMS1950846  PMID: 38126608

Abstract

My journey with tau started when in 1974 for the first time I isolated neurofibrillary tangles of paired helical filaments (PHFs) from autopsied Alzheimer’s disease (AD) brains and discovered that they were made up of a ~50–70 KDa protein on SDS-polyacrylamide gels. Subsequently my team discovered that this PHF protein and the microtubule-associated factor called tau were one and the same protein. However, we found that tau in neurofibrillary tangles/PHFs in AD brain was abnormally hyperphosphorylated, and unlike normal tau, which promoted the assembly of tubulin into microtubules, the AD-hyperphosphorylated tau inhibited microtubule assembly. These discoveries of tau pathology in AD opened a new and a major area of research on tau and on the molecular pathology of this major cause of dementia in middle- and old-age individuals. Tau pathology, which without fail is made up of the aggregated hyperphosphorylated state of the protein, is also the hallmark lesion of a family of around 20 related neurodegenerative diseases, called tauopathies. Currently, tau pathology is a major drug target for the treatment of AD and related tauopathies. Both active and passive tau immunization human clinical trials at various stages are underway. Initial results range from negative to partially promising. Future studies will reveal whether tau therapy alone or in combination with drugs targeting Aβ and/or neurodegeneration will be required to achieve the most effective treatment for AD and related disorders.

INTRODUCTION

Alzheimer’s disease (AD) was discovered by Alois Alzheimer in 1907 as a neurodegenerative disorder with numerous neurons with neurofibrillary tangles and extracellular neuritic plaques in the brain associated with dementia (Alzheimer, 1907). Clinical-pathological correlation studies revealed that the density of neurofibrillary tangles and not Aβ plaques correlates with dementia (Tomlinson et al., 1970). In 1974, for the first time, neurofibrillary tangles were bulk-isolated from autopsied AD brains and were found to be constituted of a ~50-KDa protein (Iqbal et al., 1974). Subsequent studies led to the discovery that the neurofibrillary tangle protein and the microtubule-associated protein tau reported from porcine brain in 1975 (Weingarten et al., 1975) were one and the same (Grundke-Iqbal, Iqbal, Quinlan, et al., 1986), but in AD, tau was abnormally hyperphosphorylated (Grundke-Iqbal, Iqbal, Tung, et al., 1986; Iqbal et al., 1986). Furthermore, unlike normal tau, which stimulated microtubule assembly, the AD hyperphosphorylated tau was found to inhibit the assembly by sequestering normal tau in a prion-like fashion (Alonso et al., 1996; Alonso et al., 1994; Iqbal et al., 1986). A detailed history of development of the tau field was reviewed previously (Iqbal et al., 2016). In this article, the status of tau-based drug development for the treatment of AD and related tauopathies is described.

AD: A VICIOUS CYCLE OF NEURODEGENERATION, TAU PATHOLOGY, AND Aβ PATHOLOGY

Neurodegeneration, tau pathology, and Aβ pathology are the three histopathological hallmarks of AD, each of which can lead to the other two, forming a vicious cycle (Fig. 1). After many failures over a period of almost 30 years and an expense of ~$20 billion, the U.S. Food and Drug Administration (FDA) approved two Aβ immunotherapies: aducanumab/Aduhelm® developed by Biogen Pharma, on June 7, 2021, and lecanemab/Lecambi® developed by Eisai Pharma, on January 7, 2023. A third Aβ immunotherapeutic drug, donanemab developed by Eli Lilly, is expected to receive approval from the FDA later in 2023. However, for reasons currently not clearly understood, in Phase III clinical trials in mild cognitive impairment (MCI) to mild AD cases, these three Aβ immunotherapies, although very effective in clearing Aβ plaques from the brain in AD patients, only reduced the rate of cognitive decline by ~27–40% as compared with placebo-treated controls over a period of 18 months (Soderberg et al., 2023; van Dyck et al., 2023). The small beneficial effect of Aβ immunotherapies on cognitive function suggests that inhibition of either both Aβ and tau pathologies or in addition, the inhibition of neurodegeneration, might be required for the most effective therapeutic effect.

Figure 1. Alzheimer’s disease (AD) pathology involves a vicious cycle of neurodegeneration, Aβ and hyperphosphorylated tau pathologies.

Figure 1.

AD is a multifactorial disorder that appears to involve several different mechanisms. Independent of the etiology, whether familial or sporadic forms of the disease, it is characterized by brain neurodegeneration associated with Aβ and tau pathologies. Aβ immunotherapies have shown, at best, only a partial rescue of the disease. At present, it is not known whether inhibition of both tau and Aβ pathologies alone or also combined with inhibition of neurodegeneration will be required for the most effective treatment of AD. Our hypothesis is that highly effective inhibition and prevention of AD can be achieved by shifting the balance from neurodegeneration to neural regeneration and inhibition of associated tau and Aβ pathologies. Both physiological and reparative regeneration occurs throughout life in human brain to sustain tissue homeostasis and repair as well as synaptic renewal through synaptic turnover (Martino et al., 2011; Sudhof, 2021). AD brain responds to neurodegeneration by stimulating neural regeneration, but the diseased brain lacks sufficient appropriate neurotrophic activity, and hence this attempt fails (Li et al., 2008). Although studies have implicated both Aβ and tau pathologies in causing cognitive impairment, neurodegeneration is the most proximal pathology to the development of dementia in AD patients. Substantial evidence suggests that the Aβ and hyperphosphorylated tau (ptau) oligomers, rather than Aβ plaques and tau neurofibrillary tangles (NFTs), are the primary cause of neurodegeneration and dementia. Plaques and tangles are defence responses of the diseased brain whereby it forms, respectively polymers of Aβ and hyperphoshorylated tau, which are inert space occupying lesions; in the case of NFTs as intraneuronal lesions, they appear to eventually choke the cells to death. Development of compounds such as P021 that can shift the balance in the treatment of AD from neurodegeneration to neural regeneration and consequently inhibit both tau and Aβ pathologies represents a novel and promising therapeutic approach. A recent study showed the therapeutic benefit of inhibition of neurodegeneration by reduction of long non-coding RNA MEG3 in a human neuron xenograft mouse model of AD (Balusu et al., 2023).

TAU-BASED DRUG DEVELOPMENTS

Tau has been a major drug target for the treatment of AD and related tauopathies for the last several years, resulting in over 30 drugs to inhibit or prevent aggregation and expression of tau, and promotion of its clearance from the brain. So far, none of these studies have shown significant clinical effect (Cummings et al., 2022; Imbimbo et al. 2022). The reasons for the failures of these drugs are uncertain, but one reason could be the selection of the wrong targets in the wrong patients. The two major therapeutic approaches to inhibiting tau pathology are (1) to inhibit hyperphosphorylation of tau and (2) to clear tau pathology by active or passive immunization.

INHIBITION OF HYPERPHOSPHORYLATION OF TAU

Tau is hyperphosphorylated both by the action of one or a combination of proline-dependent protein kinases (PDPKs), such as glycogen synthase kinase-3 (GSK-3) and cycline-dependent kinase-5 (cdk-5), and non-PDPKs, such as protein kinase A (PKA) and calcium, calmodulin-dependent kinase II (CamK-II) (Sironi et al., 1998; Wang et al., 2007). A Phase II clinical trial in patients with mild to moderate AD to inhibit hyperphosphorylation of tau by inhibiting GSK-3 activity with oral administration of tideglusib was unsuccessful (Del Ser, 2010). The hyperphosphorylation of tau is regulated mainly by protein phosphatase 2A (PP2A) (Bennecib et al., 2000; Gong et al., 2000; F. Liu et al., 2005), and the activity of this enzyme is compromised in AD brain (Gong et al., 1993; 1994; 1995). However, to date, no AD clinical trial with an effective PP2A activator has been reported, and this potential therapeutic approach deserves further investigation.

TAU IMMUNOTHERAPY

Tau pathology is understood only as the intracellular accumulation of hyperphosphorylated protein, except (1) that by an unconventional mechanism, tau is released from neurons in the extracellular space, and (2) that after neuronal death, the neurofibrillary tangles remain as tombstones in the extracellular space. The hyperphosphorylated tau from the tombstones, also known as ghost tangles, can become the source of tau seeds for the seeding and spread of tau pathology (Hu et al., 2016). Seeding of tau pathology is believed to spread from neuron to neuron, and thus extracellular tau can serve as a drug target for AD and related tauopathies. Although about 0.1–0.3% of an antibody can pass through the blood-brain barrier, only a small fraction of tau antibodies to Ser 396/404 were reported to enter the neuron via clathrin-dependent Fcγ receptor endocytosis (Congdon et al. 2013; Gu et al., 2013). Thus, unless tau antibodies can enter neurons in sufficient quantities, primarily the only target of tau immunotherapy is the extracellular tau (Ji & Sigurdsson, 2021). The level of extracellular tau in aged human brain is around 300 pg/ml in the cerebrospinal fluid (CSF), and this level is elevated by about 2- to 3-fold in the brains of AD patients. Thus, only about 200 ng (1 ng/ml x ~200 ml CSF = 200 ng) of extracellular tau is targeted by a tau antibody, suggesting that only a few micrograms of the antibody in the brain should be sufficient to inhibit the seeding and spread of tau pathology. Both active and passive immunization studies by several laboratories in transgenic rodent animal models were found to be very encouraging (Asuni et al., 2007; Ayalon et al., 2021; Boimel et al., 2010; Boutajangout et al., 2011; Chai et al., 2011; Collin et al., 2014; Dai et al., 2015; Dai et al., 2018; Dai et al., 2017; Gu et al., 2013; Sankaranarayanan et al., 2015; Theunis et al., 2013; Troquier et al., 2012; Umeda et al., 2015; Yanamandra et al., 2013). Based on these studies, one active and several passive immunotherapy clinical trials in humans are currently underway (Cummings et al., 2022). Results of a phase II clinical trial of active immunization against tau 294–305 carried out by Axon Neuroscience (Bratislava, Slovakia) in patients with mild to moderate AD were overall negative, but post hoc analysis revealed its effectiveness in young patients with recent onset and a high level of tau in the CSF Novak et al., 2021). Results of a Phase II clinical trial of passive immunization with an antibody to tau 25–30 using dosages of ~250–750 mg in patients with progressive supernuclear palsy, and of a Phase II clinical trial in patients with mild AD, both carried out by AbbVie Pharma, were negative (Florian et al., 2023). Results of a Phase II clinical trial with semorinemab, an antibody to tau 9–23, carried out by Genetech and AC Immune at dosages of 1,500–8,100 mg in patients with MCI to mild AD, were negative (Teng et al., 2022). All of these tau immunotherapy trials were found to be safe, with no serious side effects, such as the brain edema and microhemorrhages found with Aβ immunotherapy. However, for reasons currently not clearly understood, these tau immunotherapy trials failed to show any significant therapeutic effect. In the case of semorinemab, even at a dosage of 8,100 mg, no decrease in tau pathology was found with tau positron emission tomography. One possibility is that the tau antibodies used so far in the human clinical trials had low affinity and specificity. Alternatively, because the extracellular tau targeted by the antibodies used in the clinical trials was a non-toxic waste product, its removal had no significant therapeutic effect.

We generated several high-affinity mouse monoclonal antibodies to tau and found that antibody 43D against Tau6–18 could inhibit both tau and Aβ pathologies and reverse cognitive impairment in the 3xTg-AD transgenic mouse model of AD (Dai et al., 2017). Furthermore, we found that antibody 43D could inhibit the seeding and spread of tau pathology induced with hyperphosphorylated tau isolated from AD brain tissue (AD ptau) in 3xTg-AD mice (Dai et al., 2018). We have successfully humanized and optimized antibody 43D, and its further development is currently underway. Based on preclinical studies in mice, its calculated therapeutic dose for humans is only ~50 μg/Kg, which is a small fraction of the ~5 mg–~111.5 mg/Kg used in Phase II AD clinical trials by some pharmaceutical companies.

PREVENTION OF NEURODEGENERATION AND TAU AND Aβ PATHOLOGIES WITH A NEUROTROPHIC COMPOUND: A NOVEL THERAPEUTIC APPROACH

Both tau and Aβ pathologies are associated with neurodegeneration in AD. We have discovered that a small peptidergic compound, P021, which promotes neurogenesis and neural plasticity, can inhibit neurodegeneration and both tau and Aβ pathologies in the 3xTg-AD transgenic mouse model of AD (Baazaoui & Iqbal, 2017a, 2017b; Kazim et al., 2014; Li et al., 2010). P021 is orally bioavailable, has ~90% stability in gastric juice, and ~> 97% stability in intestinal fluid at 37° C. It has a plasma half-life of over 3 hours and is blood-brain barrier–permeable. P021 competitively inhibits leukemia inhibitory factor (LIF) in JAK-STAT3 signaling and increases the CREB activity and the transcription and expression of brain-derived neurotrophic factor (BDNF). BDNF binds to its receptor, TrkB, which leads to activation of the PI3Kinase pathway, and downstream inhibitory phosphorylation of GSK-3 kinase at Ser 9 and, consequently, prevention of hyperphosphorylation of tau and tau pathology. The inhibition of GSK-3 activity also leads to reduction of Aβ pathology through prevention of phosphorylation of APP at Thr 668 (Kazim et al., 2014).

In preliminary studies, P021 did not show any off-target binding activity or any adverse effects, even at ~550-fold the therapeutic dose in rats (unpublished data). Because of the ability of P021 to inhibit neurodegeneration and promote neural regeneration, it showed therapeutic efficacy in animal models of Down syndrome, traumatic brain injury, autism, macular degeneration, and cognitive aging (Bolognin et al., 2014; Chohan et al., 2011; Kazim et al., 2017; Kazim et al., 2014; Kazim & Iqbal, 2016; Khatoon et al., 2015; Y. Liu et al., 2019). Currently, further development of this compound for human clinical trials is underway.

ACKNOWLEDGEMENTS

I thank Dr. Wen Hu, Head of the Neuroregeneration Laboratory at the New York State Institute for Basic Research in Developmental Disabilities (IBR), for his help in preparation of Figure 1 and the bibliography, and Maureen Marlow, also of IBR, for copy editing the manuscript. Studies in my laboratory are supported in part by NIH grant AG079671, Phanes Biotech, Inc., Malvern, PA, and the New York State Office for People With Developmental Disabilities (OPWDD) Research Foundation for Mental Hygiene (RFMH).

Footnotes

CONFLICT OF INTEREST STATEMENT

Dr. Khalid Iqbal is Co-Founder and Chief Scientific Officer of Phanes Biotech, Inc., which is in the process of developing P021 as a therapeutic drug for AD and related neurodegenerative conditions.

DATA AVAILABILITY STATEMENT

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

REFERENCES

  1. Alonso AC, Grundke-Iqbal I, & Iqbal K (1996). Alzheimer’s disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules. Nature Medicine, 2(7), 783–787. doi: 10.1038/nm0796-783 [DOI] [PubMed] [Google Scholar]
  2. Alonso AC, Zaidi T, Grundke-Iqbal I, & Iqbal K (1994). Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease. Proceedings of the National Academy of Sciences of the United States of America, 91(12), 5562–5566. doi: 10.1073/pnas.91.12.5562 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alzheimer A (1907). Über eine eigenartige Erkankung der Hirnrinde. Allgemeine Zeitschrift für Psychiatrie und phychish-Gerichtliche Medizin (Berlin), 64, 146–148. [Google Scholar]
  4. Asuni AA, Boutajangout A, Quartermain D, & Sigurdsson EM (2007). Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. Journal of Neuroscience, 27(34), 9115–9129. doi: 10.1523/JNEUROSCI.2361-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ayalon G, Lee SH, Adolfsson O, Foo-Atkins C, Atwal JK, Blendstrup M, … Kerchner GA (2021). Antibody semorinemab reduces tau pathology in a transgenic mouse model and engages tau in patients with Alzheimer’s disease. Science Translational Medicine, 13(593). doi: 10.1126/scitranslmed.abb2639 [DOI] [PubMed] [Google Scholar]
  6. Baazaoui N, & Iqbal K (2017a). Prevention of amyloid-beta and tau pathologies, associated neurodegeneration, and cognitive deficit by early treatment with a neurotrophic compound. Journal of Alzheimers Disease, 58(1), 215–230. doi: 10.3233/JAD-170075 [DOI] [PubMed] [Google Scholar]
  7. Baazaoui N, & Iqbal K (2017b). Prevention of dendritic and synaptic deficits and cognitive impairment with a neurotrophic compound. Alzheimer’s Research & Therapy, 9(1), 45. doi: 10.1186/s13195-017-0273-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Balusu S, Horré K, Thrupp N, Craessaerts K, Snellinx A, Serneels L, … De Strooper B (2023). MEG3 activates necroptosis in human neuron xenografts modeling Alzheimer’s disease. Science, 381(6663), 1176–1182. doi: 10.1126/science.abp9556 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bennecib M, Gong CX, Grundke-Iqbal I, & Iqbal K (2000). Role of protein phosphatase-2A and −1 in the regulation of GSK-3, cdk5 and cdc2 and the phosphorylation of tau in rat forebrain. FEBS Letters, 485(1), 87–93. doi: 10.1016/s0014-5793(00)02203-1 [DOI] [PubMed] [Google Scholar]
  10. Boimel M, Grigoriadis N, Lourbopoulos A, Haber E, Abramsky O, & Rosenmann H (2010). Efficacy and safety of immunization with phosphorylated tau against neurofibrillary tangles in mice. Experimental Neurology, 224(2), 472–485. doi: 10.1016/j.expneurol.2010.05.010 [DOI] [PubMed] [Google Scholar]
  11. Bolognin S, Buffelli M, Puolivali J, & Iqbal K (2014). Rescue of cognitive-aging by administration of a neurogenic and/or neurotrophic compound. Neurobiology of Aging, 35(9), 2134–2146. doi: 10.1016/j.neurobiolaging.2014.02.017 [DOI] [PubMed] [Google Scholar]
  12. Boutajangout A, Ingadottir J, Davies P, & Sigurdsson EM (2011). Passive immunization targeting pathological phospho-tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain. Journal of Neurochemistry, 118(4), 658–667. doi: 10.1111/j.1471-4159.2011.07337.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Chai X, Wu S, Murray TK, Kinley R, Cella CV, Sims H, … Citron M (2011). Passive immunization with anti-Tau antibodies in two transgenic models: reduction of Tau pathology and delay of disease progression. Journal of Biological Chemistry, 286(39), 34457–34467. doi: 10.1074/jbc.M111.229633 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Chohan MO, Li B, Blanchard J, Tung YC, Heaney AT, Rabe A, … Grundke-Iqbal I (2011). Enhancement of dentate gyrus neurogenesis, dendritic and synaptic plasticity and memory by a neurotrophic peptide. Neurobiology of Aging, 32(8), 1420–1434. doi: 10.1016/j.neurobiolaging.2009.08.008 [DOI] [PubMed] [Google Scholar]
  15. Collin L, Bohrmann B, Gopfert U, Oroszlan-Szovik K, Ozmen L, & Gruninger F (2014). Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimer’s disease. Brain, 137(Pt 10), 2834–2846. doi: 10.1093/brain/awu213 [DOI] [PubMed] [Google Scholar]
  16. Congdon EE, Gu J, Sait HB, Sigurdsson EM (2013). Antibody uptake into neurons occurs primarily via clathrin-dependent Fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance. Journal of Biological Chemistry, 288(49), 35452–35465. doi: 10.1074/jbc.M113.491001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Cummings J, Lee G, Nahed P, Kambar M, Zhong K, Fonseca J, & Taghva K (2022). Alzheimer’s disease drug development pipeline: 2022. Alzheimer’s & Dementia (N Y), 8(1), e12295. doi: 10.1002/trc2.12295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Dai CL, Chen X, Kazim SF, Liu F, Gong CX, Grundke-Iqbal I, & Iqbal K (2015). Passive immunization targeting the N-terminal projection domain of tau decreases tau pathology and improves cognition in a transgenic mouse model of Alzheimer disease and tauopathies. Journal of Neural Transmission (Vienna), 122(4), 607–617. doi: 10.1007/s00702-014-1315-y [DOI] [PubMed] [Google Scholar]
  19. Dai CL, Hu W, Tung YC, Liu F, Gong CX, & Iqbal K (2018). Tau passive immunization blocks seeding and spread of Alzheimer hyperphosphorylated Tau-induced pathology in 3 x Tg-AD mice. Alzheimer’s Research & Therapy, 10(1), 13. doi: 10.1186/s13195-018-0341-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Dai CL, Tung YC, Liu F, Gong CX, & Iqbal K (2017). Tau passive immunization inhibits not only tau but also Abeta pathology. Alzheimer’s Research & Therapy, 9(1), 1. doi: 10.1186/s13195-016-0227-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Del Ser T (2010). (2010). Phase IIa clinical trial on Alzheimer’s disease with NP12, a GSK3 inhibitor. Alzheimer’s & Dementia, 6(4S), S147. doi: 10.1016/j.jalz.2010.05.455 [DOI] [Google Scholar]
  22. Florian H, Wang D, Arnold SE, Boada M, Guo Q, Jin Z, … Gold M (2023). Tilavonemab in early Alzheimer’s disease: results from a phase 2, randomized, double-blind study. Brain, 146(6), 2275–2284. doi: 10.1093/brain/awad024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Gong CX, Grundke-Iqbal I, & Iqbal K (1994). Dephosphorylation of Alzheimer’s disease abnormally phosphorylated tau by protein phosphatase-2A. Neuroscience, 61(4), 765–772. doi: 10.1016/0306-4522(94)90400-6 [DOI] [PubMed] [Google Scholar]
  24. Gong CX, Lidsky T, Wegiel J, Zuck L, Grundke-Iqbal I, & Iqbal K (2000). Phosphorylation of microtubule-associated protein tau is regulated by protein phosphatase 2A in mammalian brain. Implications for neurofibrillary degeneration in Alzheimer’s disease. Journal of Biological Chemistry, 275(8), 5535–5544. doi: 10.1074/jbc.275.8.5535 [DOI] [PubMed] [Google Scholar]
  25. Gong CX, Shaikh S, Wang JZ, Zaidi T, Grundke-Iqbal I, & Iqbal K (1995). Phosphatase activity toward abnormally phosphorylated tau: decrease in Alzheimer disease brain. Journal of Neurochemistry, 65(2), 732–738. doi: 10.1046/j.1471-4159.1995.65020732.x [DOI] [PubMed] [Google Scholar]
  26. Gong CX, Singh TJ, Grundke-Iqbal I, & Iqbal K (1993). Phosphoprotein phosphatase activities in Alzheimer disease brain. Journal of Neurochemistry, 61(3), 921–927. doi: 10.1111/j.1471-4159.1993.tb03603.x [DOI] [PubMed] [Google Scholar]
  27. Grundke-Iqbal I, Iqbal K, Quinlan M, Tung YC, Zaidi MS, & Wisniewski HM (1986). Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. Journal of Biological Chemistry, 261(13), 6084–6089. [PubMed] [Google Scholar]
  28. Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, & Binder LI (1986). Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proceedings of the National Academy of Sciences of the United States of America, 83(13), 4913–4917. doi: 10.1073/pnas.83.13.4913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Gu J, Congdon EE, & Sigurdsson EM (2013). Two novel Tau antibodies targeting the 396/404 region are primarily taken up by neurons and reduce Tau protein pathology. Journal of Biological Chemistry, 288(46), 33081–33095. doi: 10.1074/jbc.M113.494922 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Hu W, Zhang X, Tung YC, Xie S, Liu F, & Iqbal K (2016). Hyperphosphorylation determines both the spread and the morphology of tau pathology. Alzheimer’s & Dementia (NY), 12(10), 1066–1077. doi: 10.1016/j.jalz.2016.01.014 [DOI] [PubMed] [Google Scholar]
  31. Imbimbo BP, Ippati S, Watling M, & Balducci C (2022). A critical appraisal of tau-targeting therapies for primary and secondary tauopathies. Alzheimer’s & Dementia (NY), 18(5), 1008–1037. doi: 10.1002/alz.12453 [DOI] [PubMed] [Google Scholar]
  32. Iqbal K, Grundke-Iqbal I, Zaidi T, Merz PA, Wen GY, Shaikh SS, … Winblad B (1986). Defective brain microtubule assembly in Alzheimer’s disease. Lancet, 2(8504), 421–426. doi: 10.1016/s0140-6736(86)92134-3 [DOI] [PubMed] [Google Scholar]
  33. Iqbal K, Liu F, & Gong CX (2016). Tau and neurodegenerative disease: the story so far. Nature Reviews Neurology, 12(1), 15–27. doi: 10.1038/nrneurol.2015.225 [DOI] [PubMed] [Google Scholar]
  34. Iqbal K, Wisniewski HM, Shelanski ML, Brostoff S, Liwnicz BH, & Terry RD (1974). Protein changes in senile dementia. Brain Research, 77(2), 337–343. doi: 10.1016/0006-8993(74)90798-7 [DOI] [PubMed] [Google Scholar]
  35. Ji C, & Sigurdsson EM (2021). Current status of clinical trials on tau immunotherapies. Drugs, 81(10), 1135–1152. doi: 10.1007/s40265-021-01546-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Kazim SF, Blanchard J, Bianchi R, & Iqbal K (2017). Early neurotrophic pharmacotherapy rescues developmental delay and Alzheimer’s-like memory deficits in the Ts65Dn mouse model of Down syndrome. Scientific Reports, 7, 45561. doi: 10.1038/srep45561 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Kazim SF, Blanchard J, Dai CL, Tung YC, LaFerla FM, Iqbal IG, & Iqbal K (2014). Disease modifying effect of chronic oral treatment with a neurotrophic peptidergic compound in a triple transgenic mouse model of Alzheimer’s disease. Neurobiology of Disease, 71, 110–130. doi: 10.1016/j.nbd.2014.07.001 [DOI] [PubMed] [Google Scholar]
  38. Kazim SF, & Iqbal K (2016). Neurotrophic factor small-molecule mimetics mediated neuroregeneration and synaptic repair: emerging therapeutic modality for Alzheimer’s disease. Molecular Neurodegeneration, 11(1), 50. doi: 10.1186/s13024-016-0119-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Khatoon S, Chalbot S, Bolognin S, Puolivali J, & Iqbal K (2015). Elevated tau level in aged rat cerebrospinal fluid reduced by treatment with a neurotrophic compound. Journal of Alzheimer’s Disease, 47(3), 557–564. doi: 10.3233/JAD-142799 [DOI] [PubMed] [Google Scholar]
  40. Li B, Wanka L, Blanchard J, Liu F, Chohan MO, Iqbal K, & Grundke-Iqbal I (2010). Neurotrophic peptides incorporating adamantane improve learning and memory, promote neurogenesis and synaptic plasticity in mice. FEBS Letters, 584(15), 3359–3365. doi: 10.1016/j.febslet.2010.06.025 [DOI] [PubMed] [Google Scholar]
  41. Li B, Yamamori H, Tatebayashi Y, Shafit-Zagardo B, Tanimukai H, Chen S, … Grundke-Iqbal I (2008). Failure of neuronal maturation in Alzheimer disease dentate gyrus. Journal of Neuropathology and Experimental Neurology, 67(1), 78–84. doi: 10.1097/nen.0b013e318160c5db [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Liu F, Grundke-Iqbal I, Iqbal K, & Gong CX (2005). Contributions of protein phosphatases PP1, PP2A, PP2B and PP5 to the regulation of tau phosphorylation. European Journal of Neuroscience, 22(8), 1942–1950. doi: 10.1111/j.1460-9568.2005.04391.x [DOI] [PubMed] [Google Scholar]
  43. Liu Y, Wei W, Baazaoui N, Liu F, & Iqbal K (2019). Inhibition of AMD-like pathology with a neurotrophic compound in aged rats and 3xTg-AD mice. Frontiers in Aging Neuroscience, 11, 309. doi: 10.3389/fnagi.2019.00309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Martino G, Pluchino S, Bonfanti L, & Schwartz M (2011). Brain regeneration in physiology and pathology: the immune signature driving therapeutic plasticity of neural stem cells. Physiological Reviews, 91(4), 1281–1304. doi: 10.1152/physrev.00032.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Novak P, Kovacech B, Katina S, Schmidt R, Scheltens P, Kontsekova E, … Zilka N (2021). ADAMANT: a placebo-controlled randomized phase 2 study of AADvac1, an active immunotherapy against pathological tau in Alzheimer’s disease. Nature Aging, 1(6), 521–534. doi: 10.1038/s43587-021-00070-2 [DOI] [PubMed] [Google Scholar]
  46. Sankaranarayanan S, Barten DM, Vana L, Devidze N, Yang L, Cadelina G, … Ahlijanian M (2015). Passive immunization with phospho-tau antibodies reduces tau pathology and functional deficits in two distinct mouse tauopathy models. PLoS One, 10(5), e0125614. doi: 10.1371/journal.pone.0125614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Sironi JJ, Yen SH, Gondal JA, Wu Q, Grundke-Iqbal I, & Iqbal K (1998). Ser-262 in human recombinant tau protein is a markedly more favorable site for phosphorylation by CaMKII than PKA or PhK. FEBS Letters, 436(3), 471–475. doi: 10.1016/s0014-5793(98)01185-5 [DOI] [PubMed] [Google Scholar]
  48. Soderberg L, Johannesson M, Nygren P, Laudon H, Eriksson F, Osswald G, … Lannfelt L (2023). lecanemab, aducanumab, and gantenerumab - Binding profiles to different forms of amyloid-beta might explain efficacy and side effects in clinical trials for Alzheimer’s disease. Neurotherapeutics, 20(1), 195–206. doi: 10.1007/s13311-022-01308-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Sudhof TC (2021). The cell biology of synapse formation. Journal of Cell Biology, 220(7). doi: 10.1083/jcb.202103052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Teng E, Manser PT, Pickthorn K, Brunstein F, Blendstrup M, Sanabria Bohorquez S, … Tauriel I (2022). Safety and Efficacy of Semorinemab in Individuals With Prodromal to Mild Alzheimer Disease: A Randomized Clinical Trial. JAMA Neurology, 79(8), 758–767. doi: 10.1001/jamaneurol.2022.1375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Theunis C, Crespo-Biel N, Gafner V, Pihlgren M, Lopez-Deber MP, Reis P, … Muhs A (2013). Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau.P301L mice that model tauopathy. PLoS One, 8(8), e72301. doi: 10.1371/journal.pone.0072301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Tomlinson BE, Blessed G, & Roth M (1970). Observations on the brains of demented old people. Journal of Neurological Sciences, 11(3), 205–242. doi: 10.1016/0022-510x(70)90063-8 [DOI] [PubMed] [Google Scholar]
  53. Troquier L, Caillierez R, Burnouf S, Fernandez-Gomez FJ, Grosjean ME, Zommer N, … Buee L (2012). Targeting phospho-Ser422 by active Tau Immunotherapy in the THYTau22 mouse model: a suitable therapeutic approach. Current Alzheimer Research, 9(4), 397–405. doi: 10.2174/156720512800492503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Umeda T, Eguchi H, Kunori Y, Matsumoto Y, Taniguchi T, Mori H, & Tomiyama T (2015). Passive immunotherapy of tauopathy targeting pSer413-tau: a pilot study in mice. Annals of Clinical and Translational Neurology, 2(3), 241–255. doi: 10.1002/acn3.171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, Gee M, … Iwatsubo T (2023). Lecanemab in Early Alzheimer’s Disease. New England Journal of Medicine, 388(1), 9–21. doi: 10.1056/NEJMoa2212948 [DOI] [PubMed] [Google Scholar]
  56. Wang JZ, Grundke-Iqbal I, & Iqbal K (2007). Kinases and phosphatases and tau sites involved in Alzheimer neurofibrillary degeneration. European Journal of Neuroscience, 25(1), 59–68. doi: 10.1111/j.1460-9568.2006.05226.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Weingarten MD, Lockwood AH, Hwo SY, & Kirschner MW (1975). A protein factor essential for microtubule assembly. Proceedings of the Nationall Academy of Sciences of the United States of America, 72(5), 1858–1862. doi: 10.1073/pnas.72.5.1858 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Yanamandra K, Kfoury N, Jiang H, Mahan TE, Ma S, Maloney SE, … Holtzman DM (2013). Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron, 80(2), 402–414. doi: 10.1016/j.neuron.2013.07.046 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

RESOURCES