Fetal Tau Bias in iPSC -Derived Neurons, MAPT Mutant Mouse  
Models and Molecular Mechanisms Assessment for Integrative  
Analysis of Tau Pathology  
Aditi Kaushik1,2*, Richa Mor1, Sushila Kaura3, Apurv Kaushik4, Sapna Sharma5  
1Department of Biotechnology, NIILM University, Kaithal, India,  
2 European Cooperation in Science & Technology (COST), Brussels, Belgium  
3Department of Pharmacology, OSG University, Hisar, India  
4Department of Medicine, RPS Group, Mahendergarh, India  
5Department of Public Health, PGIVER, Jaipur, India  
*Corresponding Author  
Received: 03 December 2025; Accepted: 10 December 2025; Published: 18 December 2025  
ABSTRACT  
Tauopathies, such as Alzheimer's disease and Frontotemporal Dementia, are caused by complex interactions  
between tau isoform imbalance, MAPT mutations, and harmful post-translational modifications. Despite  
significant breakthroughs, current human cellular models may not accurately reflect adult tau biology, limiting  
mechanistic knowledge and therapeutic translation. In this review, we draw on new knowledge from stem cell  
platforms, MAPT mutant mice models, and multi-omics investigations to identify important gaps and stakes in  
modeling tau pathology. Recent efforts using CRISPR-engineered human stem cell lines, NGN3-induced i3  
neurons, and patient-derived iPSCs show great promise, but they consistently retain a fetal-like predominance  
of 3-repeat (3R) tau, limiting the ability to recapitulate adult 3R/4R tau ratios and age-associated tauopathy  
phenotypes. Organoid and multi-cell co-culture methods (such as RenVM and tri-cellular constructions) boost  
microenvironmental complexity, but are limited by developmental immaturity and variable tau isoform  
flipping. Complementary MAPT knock-in and transgenic mice models (e.g., P301L, P301S, V337M, S320F)  
provide robust in vivo mechanisms for tau misfolding, seeding, and propagation, while also revealing species-  
specific compensatory processes that differ from human neurodegeneration. At the molecular level, mass  
spectrometry based phosphoproteomics has revealed a coordinated network of tau post-translational  
modifications; phosphorylation, acetylation, and ubiquitination that converge on proline-rich and C-terminal  
regions to cause tau detachment from microtubules and aggregate. These findings highlight the necessity for  
integrated models that can capture both isoform regulation and combinatorial PTM landscapes. Together, these  
findings highlight a key translational gap that most human model systems fail to reach adult tau maturation, as  
animal models cannot fully mimic human-specific tau biology. We propose a paradigm that integrates  
sophisticated stem cell engineering, MAPT mutation-aligned mice models, and multi-omics profiling to create  
next-generation platforms for understanding tauopathy processes and accelerating therapeutics development.  
Keywords: Tauopathy, Alzheimer’s disease, Frontotemporal Dementia, MAPT mutations, induced pluripotent  
stem cells (iPSCs), tau phosphorylation  
INTRODUCTION  
Alzheimer's disease (AD) and similar tauopathies are distinguished by aberrant aggregation of the  
microtubule-associated protein tau, which impairs neuronal function and causes neurodegeneration. While  
classic conceptions of AD emphasize the amyloid-β (Aβ) cascade, emerging evidence reveals a more  
complicated interplay between Aβ and tau disorders, suggesting that tau may actively contribute to disease  
progression, rather than just following amyloid. For instance, in a recently created APP^NL-G-  
Page 1347  
F/MAPT^P301S double-knockin mice, researchers found that β-amyloid buildup increases tau pathology,  
inflammation, and neurodegeneration. This supports a synergistic paradigm where Aβ promotes tau misfolding  
and aggregation [1,2]. This model emphasizes the importance of combining amyloid and tau pathology in vivo  
to more accurately mimic human AD. Despite breakthroughs in animal models, applying findings from these  
systems to human disease remains difficult. A detailed mass-spectrometry-based comparison of tau post-  
translational modifications (PTMs) revealed that commonly used tauopathy mouse models (such as the P301S  
and P301L mutant lines) mimic early phosphorylation events in human disease but fail to reflect late-stage  
modifications, such as ubiquitination and acetylation, which are common in advanced Alzheimer's [3]. This  
gap highlights the critical need for models that better reproduce the entire spectrum of tau disease. Another  
dimension of tauopathies confuses the scenario even more. Human MAPT knock-in mouse models, which bear  
combinations of pathogenic mutations (such as P301S; Int10+3; S320F), show age-dependent tau buildup,  
behavioral impairments, and brain region-specific susceptibility [4]. These animal models, which closely  
reproduce human tau isoform ratios (3R/4R), provide an effective tool for investigating how individual  
mutations influence disease pathways in vivo.  
Tau's molecular toxicity is caused by more than merely aggregation. Pathological tau experiences a network of  
post-translational modifications (PTMs), including phosphorylation, acetylation, and ubiquitination, all of  
which impact its structure and interactions. A recent review revealed how such alterations impair tau's  
microtubule-stabilizing function, disrupt axonal transport, and activate neuroinflammatory pathways [5].  
Furthermore, mass-spectrometry validation in animal models demonstrated that phosphorylation in the proline-  
rich and C-terminal domains is a prominent early cause of aggregation [3]. Despite these developments,  
human cellular models of tauopathy continue to be a significant challenge. Induced pluripotent stem cell  
(iPSC)-derived neurons and organoids are commonly employed to research tau, however they frequently retain  
fetal-like 3R tau isoforms, failing to fully replicate the adult 3R/4R tau equilibrium found in the human brain.  
This immaturity restricts their usefulness in simulating adult-onset tauopathies and testing treatments.  
To address these constraints, researchers are increasingly using multimodal platforms, including as CRISPR-  
engineered human neurons, patient-derived iPSCs, and sophisticated in vivo models, to capture the genetic and  
metabolic complexity of tauopathies. Such collaboration may be critical for closing the translational gap. Only  
by better simulating adult-like tau biology can the field create next-generation therapeutics that effectively  
target disease-causing tau pathologies. In this review, we summarize recent achievements in stem cell  
modeling, MAPT-mutant mice lines, tau PTM mapping, and amyloid-tau interactions, and propose a unified  
paradigm for future tauopathy research. A conceptual overview of the primary biological and technology  
elements influencing tau pathology modelling is depicted in figure 1.  
Figure 1. The significant aspects influencing tau research, such as fetal tau bias in iPSC-derived neurons,  
organoid maturation restrictions, MAPT mutant mice models, post-translational modification complexity, prion  
seeding & clearance, and biomarker frameworks. Together, these components illustrate the multifaceted  
difficulty of adequately modeling human tauopathy.  
Page 1348  
Stem-Cell Models of Tauopathy  
Human pluripotent stem cell (PSC) based systems, such as induced pluripotent stem cell (iPSC)-derived  
neurons, CRISPR-engineered neuronal lines, and multicellular brain organoids, have emerged as critical tools  
for modeling tau's complex biology in Alzheimer's disease (AD) and frontotemporal lobar degeneration  
(FTLD-tau). These platforms have a distinct advantage, they capture human-specific genetic settings while  
providing temporal control over tau expression, phosphorylation, and aggregation. Recent engineering  
breakthroughs have produced PSC-derived neurons capable of expressing elevated levels of 4-repeat (4R) tau,  
a key requirement for replicating adult tauopathies, and have shown that these cultures can support tau seeding,  
propagation, and conformational diversification under controlled conditions [6]. However, the developmental  
immaturity of most stem-cell-derived neural systems continues to be a major concern. Standard differentiation  
techniques produce neurons that mostly express 3-repeat (3R) tau, indicating a fetal or early postnatal  
phenotype rather than the mature tau landscape found in adult human cortex. This mismatch severely limits the  
ability to mimic diseases characterized by pathological 3R/4R tau imbalances, such as Alzheimer's disease,  
primary age-related tauopathy (PART), and various MAPT mutation-driven types of FTLD-tau. Despite  
ongoing efforts to manipulate tau isoform ratios through CRISPR-editing of MAPT splice-regulatory elements  
such as S305 and intron 10 mutations, the field is still refining methods for achieving stable, adult-like 4R tau  
expression without compromising neuronal viability or synaptic maturation [7,8]. Nonetheless, current PSC  
have proven invaluable for examining early tau post-translational modifications (PTMs), kinase-substrate  
interactions, and cell-intrinsic stress responses. Advanced phosphoproteomic techniques allow for the  
simultaneous assessment of wide PTM landscapes, including phosphorylation, ubiquitination, and acetylation,  
as well as their dynamic interplay during the early phases of tau misfolding [8]. Complementary seeding and  
biosensor assays have shown that human neurons can internalize exogenous pathological tau species, amplify  
disease-linked conformers, and spread them across neural networks, though the efficiency of these processes is  
highly dependent on isoform maturation status. These findings highlight the importance of multimodal  
validation, which involves cross-checking results in iPSC neurons with more mature humanized mouse  
models, long-term organoids, and ex-vivo tissue systems [9].  
Stem-cell based platforms are altering the study of tau biology by linking molecular pathways to human  
genetics. However, their best application necessitates an understanding of critical constraints, including tau  
isoform immaturity, as well as deliberate integration with more mature in vivo systems. As differentiation  
methods, CRISPR designs, and phospho-multiomics technologies advance, PSC models will play an  
increasingly important role in mechanistic discovery, therapeutic screening, and individualized tauopathy  
modeling.  
Amyloid-Tau Interactions  
Emerging data suggests a synergistic link between amyloid-β (Aβ) and tau diseases. The two proteins appear to  
promote each other through molecular interactions, localized microenvironments, and network-level  
dissemination, rather than Aβ alone causing tau aggregation [10]. Recent molecular and computational  
investigations provide detailed insights into how Aβ and tau interact to allow pathological seeding. Tau's  
microtubule-binding repeats (R1-R4) have different affinities for different surfaces of Aβ fibrils. R1 and R3  
preferentially bind the lateral surfaces, while R2 and R4 show higher affinity for the fibril elongation ends,  
promoting β-sheet formation and nucleation of tau aggregation [11,12]. Biochemical investigations show that  
certain areas of the Aβ core can bind to tau epitopes, speeding up the spread of tau species. In vivo, the Aβ  
plaque microenvironment promotes tau aggregation. When pathogenic tau seeds from AD brains are injected  
into amyloid-bearing mice, dystrophic neurites surrounding plaques rapidly aggregate, generating "neuritic-  
plaque tau" before neurofibrillary tangles (NFTs) emerge [11]. This phenomenon aligns with human imaging  
and connectivity studies, for instance, tau spreading in Alzheimer's disease correlates with regions of high local  
Aβ/tau interaction (e.g., inferior temporal gyrus), suggesting that anatomical connectivity and amyloid burden  
both influence tau propagations.  
These molecular findings challenge the traditional amyloid cascade concept and propose a more integrated  
model where Aβ and tau work together rather than sequentially [13,14]. Recent evaluations suggest that  
targeting common epitopes or interaction interfaces may improve therapeutic outcomes compared to focusing  
Page 1349  
solely on Aβ or tau [12]. The molecular interactions of Aβ and tau, especially cross-seeding interfaces, provide  
promise for drug discovery.  
Translational Perspectives  
The molecular intricacy of tau-driven neurodegeneration has prompted a wide range of treatment methods,  
including passive immunotherapy and seed inhibition, as well as manipulation of extracellular clearance  
mechanisms. Success in preclinical models has been uneven, and new translational research highlights the  
importance of aligning therapeutic methods with the biology of tau proliferation, post-translational  
modification (PTM), and clearance. One intriguing therapeutic path is to directly target tau's seeding and self-  
propagation activity. Recent research has improved seed amplification assays (SAA) to find small compounds  
or antibodies that prevent tau aggregation. For example, a newly developed tau SAA platform exhibited the  
ability to detect physiological quantities of tau seeds in Alzheimer's brains while also identifying clinically  
approved drugs that drastically diminish seeding efficiency [15]. Because tau seeding is critical to disease  
dissemination, such tests offer a scalable and sensitive approach to identifying aggregation inhibitors with  
translational potential. Beyond intracellular pathways, the glymphatic clearance system has been identified as a  
regulator of tau disease. In a recent in vivo study, pharmacological suppression of aquaporin-4 (AQP4), a key  
component of the glymphatic system, greatly increased tau aggregation and propagation, worsening cognitive  
impairment in tau-seeded animal models [16]. These findings indicate that therapeutic augmentation of  
glymphatic function or avoidance of age-related decline could be a unique and underutilized method for  
mitigating tau distribution.  
Therapeutic efforts must take into account the extensive network of tau PTMs and proteolytic fragments, both  
of which influence tau toxicity, aggregation, and cellular trafficking. Recent reviews have highlighted that  
certain truncated tau species generated by protease cleavage and aberrant phosphorylation or ubiquitination  
patterns significantly contribute to pathology, and suggest that inhibiting specific proteases, blocking fragment  
formation, or modulating PTM machinery are potential interventions [17]. However, accuracy is essential.  
Given the diversity of tau species, therapies will most likely need to distinguish between pathological and  
healthy isoforms or alterations. To translate preclinical achievements into human trials, robust biomarkers and  
patient classification are required. High-sensitivity assays are currently being used to detect phospho-tau in  
biofluids (e.g., plasma), which informs not just diagnosis but also treatment engagement [18, 19]. A recent  
review highlights that biomarker-led trial designs, such to those utilized in amyloid-targeting therapy, are  
critical for tau-directed therapeutics [20, 21]. Such trials should include biomarkers for seeding activity, PTM  
patterns, and clearance pathways to ensure precise target engagement and treatment efficacy monitoring.  
While these therapy procedures are promising, there are still significant challenges. Tau exists in several  
conformational and PTM states. Therapies must selectively eliminate pathogenic species while preserving  
physiologically functioning tau. Preclinical achievements often come from overexpression or inoculation  
models. Translating these discoveries to clinical disease necessitates ensuring therapeutic efficacy in models  
that mimic adult tau biology, including isoform balance, PTM topography, and propagation dynamics.  
Combination treatments, such as seed inhibitors and glymphatic enhancers, may be more effective than  
monotherapies due to their dual involvement in intracellular seeding and extracellular clearance. Designing  
clinical trials with biomarker techniques, such as plasma p-tau, seeding assays, and neuroimaging, can speed  
the process from preclinical discovery to disease-modifying therapies.  
DISCUSSION  
Tau pathology is caused by a combination of internal molecular vulnerabilities (misfolding, aberrant PTMs,  
proteolytic cleavage, and poor proteostasis) and external modulators such amyloid-β (Aβ), inflammation, and  
lymphatic dysfunction. An increasing collection of research helps to refine how these factors interact across  
disease phases. Jiang et al. (2024) found that Aβ accumulation in APPNL-G-F/MAPTP301S mice leads to  
faster tau aggregation and neuritic plaque-associated seeding, indicating a clear relationship between amyloid  
burden and early amplification of tauopathy. He et al. previously demonstrated that Aβ plaques can increase  
tau-seeded disease, indicating an early amyloid-tau synergy. These findings highlight that amyloid and tau  
Page 1350  
pathology are mutually accelerating processes, which is consistent with the cross-talk outlined by Avila et al.  
(Avila et al., 2025) and reviewed by Haut et al. (Haut et al., 2024).  
Model Fidelity  
Despite significant advances, no single experimental model accurately reproduces the whole intricacy of  
clinical tauopathies. Wenger et al. demonstrated that commonly used mice lines reproduce early but not late  
human pathology, limiting their translational power (Wenger et al., 2023). The key constraint remains  
throughout stem-cell-derived neurons, which continue to exhibit fetal-like tau expression patterns, specifically  
an underrepresentation of 4R tau isoforms, concealing late-stage conformational alterations important to  
Alzheimer's disease (Kühn et al., 2021; Baumann, 2024). Morito et al.'s recent humanized MAPT knock-in  
mice represent a significant step forward, allowing for physiological expression of human tau without  
overexpression artifacts. Representational depiction of tau accumulation can be observed through figure 2.  
Human iPSC systems have emerged as powerful platforms for detecting cell-intrinsic tau disease modifiers.  
Parra Bravo et al. discovered genetic regulators of tau propagation in a 4R-dominant iPSC model, showing  
novel modulators of transcellular dissemination (Parra Bravo et al., 2024). This complements Samelson et al.'s  
proteostasis-focused CRISPR screens, which identified ER-Golgi trafficking and lysosomal nodes as major  
regulators of tau turnover. These findings demonstrate that mature, adult-like human models with physiologic  
3R/4R balance, intact splicing, and complicated PTM dynamics should remain the top goal for future tau  
research.  
Figure 2. Diverse MAPT mutations and primary tauopathies cause varied geographic distributions of tau  
aggregates throughout the mice brain. These varying deposition and transmission patterns highlight the  
biological variety of tau species, as well as the necessity for model systems that incorporate regional  
vulnerability.  
Mechanistic Layers  
Tau toxicity cannot be linked to a particular molecular event. Instead, pathogenic conformer production is  
driven by certain combinations of PTMs (phosphorylation, acetylation, and ubiquitination), as well as  
protease-generated fragments. Yang et al. described how these PTM signatures interact to produce unique,  
highly seeding-competent tau species, while shortened fragments promote fibrillization and propagation (Yang  
et al., 2024). Song et al. (2023) found that tau microtubule-binding repeats interact differently with amyloid-β  
fibrils, indicating a structural foundation for amyloid-accelerated tau templating. These findings support Yan  
and Cook's assertion that mass spectrometry-based PTM mapping is crucial for validating mechanistic insights  
and improving model fidelity.  
Page 1351  
Therapeutic Pathways  
Given the prion-like proliferation of pathogenic tau, seeding inhibition has emerged as a key treatment  
strategy. Gorski et al. recently developed the Tau Seed Amplification Assay (Tau-SAA), which is a sensitive  
and scalable technique for screening for seeding and transmission modifiers. Extracellular fluid dynamics also  
influence tau dispersion. Lopes et al. found that glymphatic inhibition significantly exacerbates tau  
propagation in mouse models, implying that improving glymphatic and interstitial clearance could provide  
considerable therapeutic synergy (Lopes et al., 2024). This is consistent with the larger notions of proteostasis  
and clearance pathways emphasized in tau-targeting techniques (Mohan Kumar & Talwar, 2025).  
Biomarkers and Trial Design  
Rapid advancements in fluid biomarkers have transformed the translational environment. Plasma phospho-tau  
species have shown good diagnostic and staging potential, and Gonzalez-Ortiz et al. confirmed their suitability  
for treatment trial applications. Therriault et al.'s recent comparisons of immunoassay- and mass spectrometry-  
derived p-tau levels underscore the importance of standardization and multi-platform techniques. Penny et al.  
underlined that future studies should use biomarker-anchored designs that incorporate p-tau measurements,  
Tau-SAA, and multimodal imaging to evaluate real-time target engagement and disease change (Penny et al.,  
2024).  
Thus, it can be inferenced that current tauopathy models, which include rodent models, iPSC-derived neurons,  
and 3D organoids, jointly recreate many aspects of human tau biology. Wenger et al. demonstrated and  
Baumann confirmed that widely utilized models efficiently reproduce early-stage tau alterations but are limited  
in their ability to depict late-stage, adult human tau conformers. As a result, capturing the entire illness  
spectrum requires the integration of different complimentary systems, particularly human MAPT knock-in  
models (Morito et al., 2025). iPSC-derived neurons often retain fetal tau expression and do not achieve the  
physiological 3R/4R tau ratio found in adult human brains (Kühn et al., 2021). This immature splicing profile  
obscures important disease-relevant vulnerabilities in tau behavior, slowing mechanistic research and  
decreasing translational accuracy for drug testing. Accumulating data suggested that toxic tau species are  
caused by complicated combinations of post-translational modifications; phosphorylation, acetylation, and  
ubiquitination rather than single erroneous events. Yang et al. and Yan & Cook discovered that PTM  
"signatures," frequently accompanied by proteolytic truncation, stimulate the production of highly pathogenic  
conformers (Yang et al., 2024; Yan & Cook, 2023). This emphasizes the significance of combined PTM  
mapping with mass spectrometry. Humanized MAPT knock-in mice (Morito et al., 2025) and MAPT mutant  
models (Wenger et al., 2023) accurately replicate sequential tau misfolding, templating, and dissemination.  
These models have helped to discover genetic drivers of tau pathology and influence the creation of biomarker  
techniques for disease progression and treatment engagement (Gonzalez-Ortiz et al., 2023). To further  
tauopathy research, human stem-cell-derived models must attain adult-like tau splicing, isoform balance, PTM  
dynamics, and neuron-glia interactions. Parra Bravo et al. and Samelson et al. found that advances in iPSC  
engineering and CRISPR-based functional genomics can bring stem-cell systems closer to physiologic adult  
tau biology (Parra Bravo et al., 2024; Samelson et al., 2024).  
CONCLUSION  
Several high-priority objectives arise based on integrated knowledge from genetic investigations, human-  
model research, and tau propagation biology. To accurately simulate adult human tauopathy, human iPSC and  
organoid models must demonstrate consistent 3R/4R tau switching, mature splicing patterns, and age-  
equivalent PTM landscapes. Given the variety of tau species shown by PTM-mapping research, targeted  
therapeutics must distinguish hazardous conformers based on phosphorylation, truncation, or acetylation  
signals. Future therapeutics should combine seeding inhibition, clearance enhancement (including glymphatic  
modulation), and intracellular proteostasis support to address the numerous mechanistic levels of tau toxicity.  
Plasma p-tau isoforms, Tau Seed Amplification Assays, and mass-spectrometry-based signatures should be  
used in the initial phases of clinical trials to allow for real-time monitoring of target engagement and disease  
change. Emerging research shows that early amyloid disease increases tau aggregation. Dual-pathway  
treatments that address amyloid triggers and tau propagation may be required.  
Page 1352  
Tauopathies are a convergence site for a variety of chemical interactions that affect proteostasis, synaptic  
integrity, and neuron survival. Advances in human stem-cell systems, long-term brain organoids, multimodal  
imaging, and high-resolution proteomics have altered our ability to simulate early tau changes and understand  
the mechanisms that lead to phosphorylation, misfolding, and disease dissemination. At the same time, cross-  
species studies involving Drosophila, zebrafish, and humanized mouse models have helped us better  
understand conserved neurotoxic pathways and cell-type vulnerabilities. Together, these tools now enable  
researchers to investigate tau biology with unparalleled precision. Despite this progress, significant gaps  
persist. Current models are still unable to properly capture adult-like tau isoform balance, late-stage  
aggregation dynamics, and the chronic neuroinflammatory environment associated with human tauopathies.  
The geographical and temporal complexities of tau propagation, particularly the interaction of synaptic  
degeneration, membrane trafficking, and glial responses, are only partially understood. To bridge these gaps,  
diverse experimental systems must be integrated, more physiologically mature human models need to be  
developed, and quantitative frameworks for tracking tau conformational states across time be improved.  
Looking ahead, the combination of stem-cell technology, nano-bioengineering, and artificial intelligence has  
the potential to produce predictive and translationally applicable platforms. These combined techniques may  
allow for earlier diagnosis of disease trajectories, more accurate testing of treatment candidates, and the  
creation of precision nanomedicines capable of modifying tau at the molecular and network levels. As the  
science works toward holistic models that account for tauopathy's molecular and systemic complexity, the  
prospects for major treatment breakthroughs improve. This review emphasizes the relevance of  
transdisciplinary, human-centered, and technology-enabled methods to understanding and, eventually, curing  
tau-driven neurodegeneration.  
Ethics Statement  
This study is a review of the literature and not an experiment involving human participants, personal data, or  
animals. Therefore, ethical approval of animal committee and informed consent were not required. Images and  
diagrams are of hypothetical nature, provided for representational and educational purposes only. All referred  
papers were analyzed and reported in compliance with established academic and ethical guidelines.  
Conflict Of Interest  
The authors declare that the research was conducted in the absence of any commercial or financial  
relationships that could be construed as a potential conflict of interest.  
Author Contributions  
AK conceptualized the review theme, created the framework, conducted the literature review, combined  
mechanistic and model-based insights, and wrote the entire manuscript. RM contributed to sections on stem  
cell technologies, organoids, and molecular biotechnology frameworks. SK contributed to the synthesis of  
pharmacological and therapeutic views, as well as the refinement of mechanistic considerations. ApK supplied  
clinical interpretation, insights into translational applicability, and the matching of preclinical models with  
human disease characteristics. SS helped to shape the public-health conceptualization, contextual significance,  
and clarity of communication for broader scientific audiences. All authors evaluated the manuscript, made  
adjustments, and approved the final version.  
ACKNOWLEDGEMENTS  
The authors are appreciative for their colleagues' valuable support and scholarly contributions throughout the  
creation of this work. We are grateful to the COST Association and the Department of Biotechnology at  
NIILM University for academic support, the Department of Pharmacology, OSGU for insights into therapeutic  
mechanisms, and the Department of Medicine, RPS Group for clinical relevance and analysis. The  
collaborative contributions improved the quality and clarity of this work.  
Page 1353  
REFERENCES  
1. Jiang, L., Roberts, R., Wong, M., Zhang, L., Webber, C. J., Libera, J., Wang, Z., Kilci, A., Jenkins, M.,  
Ortiz, A. R., Dorrian, L., Sun, J., Sun, G., Rashad, S., Kornbrek, C., Daley, S. A., Dedon, P. C.,  
Nguyen, B., Xia, W., Saito, T., et al. (2024). β-amyloid accumulation enhances microtubule associated  
protein tau pathology in an APPNL-G-F/MAPTP301S mouse model of Alzheimer's disease. Front.  
2. Wenger, K., Viode, A., Schlaffner, C. N., et al. (2023). Common mouse models of tauopathy reflect  
early but not late human disease. Mol. Neurodegener, 18(1), 10. https://doi.org/10.1186/s13024-023-  
3. Yan, Y., Cook, C. N. (2023). Using mass spectrometry to validate mouse models of tauopathy. Mol.  
4. Morito, T., Qi, M., Kamano, N., Sasaguri, H., Bez, S., Foiani, M., Duff, K., Benner, S., Endo, T.,  
Hama, H., Kurokawa, H., Miyawaki, A., Mizuma, H., Sahara, N., Shimojo, M., Higuchi, M., Saido, T.  
C., & Watamura, N. (2025). Human MAPT knockin mouse models of frontotemporal dementia for the  
neurodegenerative  
research  
community.  
Cell  
Rep.  
Methods  
5(4),  
101024.  
5. Hong, X., Huang, L., Lei, F., Li, T., Luo, Y., Zeng, M., & Wang, Z. (2025). The role and pathogenesis  
of tau protein in Alzheimer’s disease. Biomolecules, 15(6), 824. https://doi.org/10.3390/biom15060824  
6. Parra Bravo, C., Giani, A. M., Madero-Perez, J., Zhao, Z., Wan, Y., Samelson, A. J., Wong, M. Y.,  
Evangelisti, A., Cordes, E., Fan, L., Ye, P., Zhu, D., Pozner, T., Mercedes, M., Patel, T., Yarahmady,  
A., Carling, G. K., Sterky, F. H., Lee, V. M. Y., Lee, E. B., et al. (2024). Human iPSC 4R tauopathy  
model  
7. Baumann, K. (2024). Modelling  
8. Kühn, R., Mahajan, A., Canoll, P., & Hargus, G. (2021). Human induced pluripotent stem cell models  
of frontotemporal dementia with tau pathology. Front. Cell Dev. Biol, 9, 766773.  
uncovers  
modifiers  
of  
tau  
propagation.  
Cell  
187(10),  
2446-2464.e22.  
tauopathies.  
Nat.  
Rev. Mol.  
Cell  
Biol,  
25,  
338.  
9. Samelson, A. J., Ariqat, N., McKetney, J., Rohanitazangi, G., Bravo, C. P., Bose, R., Travaglini, K. J.,  
Lam, V. L., Goodness, D., Dixon, G., Marzette, E., Jin, J., Tian, R., Tse, E., Abskharon, R., Pan, H.,  
Carroll, E. C., Lawrence, R. E., Gestwicki, J. E., Eisenberg, D., et al. (2024). CRISPR screens in iPSC-  
derived  
10. Avila, J., Hernández, F., & Perry, G. (2025). Amyloid and tau pathologies cross-talk to promote  
Alzheimer's disease: novel mechanistic insights. Neuroscience, 582, 58-62.  
neurons  
reveal  
principles  
of  
tau  
proteostasis.  
bioRxiv  
2023.06.16.545386.  
11. He, Z., Guo, J. L., McBride, J. D., Narasimhan, S., Kim, H., Changolkar, L., Zhang, B., Gathagan, R.  
J., Yue, C., Dengler, C., Stieber, A., Nitla, M., Coulter, D. A., Abel, T., Brunden, K. R., Trojanowski,  
J. Q., & Lee, V. M. (2018). Amyloid-β plaques enhance Alzheimer’s brain tau-seeded pathologies by  
facilitating neuritic plaque tau aggregation. Nat. Med, 24(1), 29-38. https://doi.org/10.1038/nm.4443  
12. Song, Z., Gatch, A. J., Sun, Y., & Ding, F. (2023). Differential binding and conformational dynamics  
of tau microtubule-binding repeats with a preformed amyloid-β fibril seed. ACS Chem. Neurosci,  
13. Haut, F., Argyrousi, E. K., & Arancio, O. (2024). Re-arranging the puzzle between amyloid-beta and  
tau  
14. Mohan Kumar, D., & Talwar, P. (2025). Amyloid-β, tau, and α-synuclein protein interactomes as  
therapeutic targets in neurodegenerative diseases. Cell Mol. Neurobiol, 45, 84.  
15. Gorski, D., Evans, H., Allison, T., et al. (2025). Design and application of a Tau seed amplification  
assay for screening inhibitors of Tau seeding. Alzheimers Res. Ther, 17, 214.  
pathology:  
an  
APP-centric  
approach.  
Int.  
J.  
Mol.  
Sci,  
25(1),  
259.  
Page 1354  
16. Lopes, D. M., Wells, J. A., Ma, D., et al. (2024). Glymphatic inhibition exacerbates tau propagation in  
an Alzheimer’s disease model. Alzheimers Res. Ther, 16, 71. https://doi.org/10.1186/s13195-024-  
17. Yang, J., Shen, N., Shen, J., et al. (2024). Complicated role of post-translational modification and  
protease-cleaved fragments of tau in Alzheimer’s disease and other tauopathies. Mol. Neurobiol, 61,  
18. Gonzalez-Ortiz, F., Kac, P. R., Brum, W. S., et al. (2023). Plasma phospho-tau in Alzheimer’s disease:  
towards  
diagnostic  
and  
therapeutic  
trial  
applications.  
Mol.  
Neurodegener,  
18(1),  
18.  
19. Therriault, J., Woo, M. S., Salvadó, G., et al. (2024). Comparison of immunoassay- with mass  
spectrometry-derived p-tau quantification for the detection of Alzheimer’s disease pathology. Mol.  
20. Penny, L. K., Lofthouse, R., Arastoo, M., et al. (2024). Considerations for biomarker strategies in  
clinical trials investigating tau-targeting therapeutics for Alzheimer’s disease. Transl. Neurodegener,  
21. Mandal, P. K., Maroon, J. C., Garg, A., Arora, N. K., Bansal, R., Kaushik, A., Samkaria, A., Kumaran,  
G., & Arora, Y. (2023). Blood biomarkers in Alzheimer’s disease. ACS Chemical Neuroscience,  
Abbreviations and acronyms  
AD: Alzheimer's disease  
Aβ: Amyloid-β  
APP: Amyloid Precursor Protein  
CRISPR: Clustered Regularly Interspaced Short Palindromic Repeats  
CNS: Central nervous system  
CSF: Cerebrospinal Fluid  
DLB: Dementia with Lewy Bodies  
ER: Endoplasmic Reticulum  
FTD: Frontotemporal Dementia  
GFP: Green Fluorescent Protein  
GWAS: Genome-Wide Association Study  
iPSC: Induced Pluripotent Stem Cell  
iPSC-Ns: iPSC-derived neurons  
MAPT: Microtubule-Associated Protein Tau  
MT: Microtubules  
NGN3: Neurogenin-3  
NFTs: Neurofibrillary Tangles  
PD: Parkinson's disease  
Page 1355  
PET: Positron Emission Tomography  
PTM: Post-Translational Modification  
RNA-seq: RNA Sequencing  
SAA: Seed Amplification Assay  
SD05N10: A modified mouse genetic background utilized in tau research  
SPT: Single Particle Tracking  
TBI: Traumatic Brain Injury  
Tau-SAA: Tau Seed Amplification Assay  
3R/4R: Tau isoforms with three or four repeats  
UPR: Unfolded Protein Response  
WT: Wild Type  
Page 1356