Take a look at the Recent articles

Alzheimer's disease as a multi‑directional spectrum: Integrating neuroinflammation, synaptopathy, paranodal dysfunction, and metabolic trace deficits

Nguyen Duc Huy Hoang

Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam

E-mail : aa

DOI: 10.15761/NNS.1000147

Article
Article Info
Author Info
Figures & Data

Abstract

Background: Alzheimer’s disease (AD) remains an enduring challenge in neurodegeneration, with decades of emphasis on amyloid and tau yielding neither unifying explanations nor effective therapeutics [1-4,5].

Objective: We synthesise evidence across neuropathology, molecular and cellular neuroscience, imaging biomarkers, epidemiology, and interventional trials to propose a calibrated “Alzheimer’s spectrum” model.

Methods: We conducted an integrative synthesis linking recurrent paradoxes in AD literature onto mechanistic explanatory nodes—amyloid/tau, neuroinflammation, synaptopathy, paranodal/myelin–axon interface dysfunction, and metabolic/trace-element (notably lithium) deficits—and derived operational criteria and a trial calibration checklist [6-8].

Results: The spectrum model explains (i) why amyloid lowering does not uniformly translate into cognitive benefit, (ii) why synaptic loss correlates more tightly with clinical status, (iii) the existence of biomarker-positive yet cognitively resilient individuals, and (iv) differential trial outcomes driven by phenotype heterogeneity and inadequate demonstration of central target engagement [9,10].

Conclusions: Reframing AD as a multi-mechanistic spectrum resolves key paradoxes, suggests phenotype-matched therapeutic strategies, and mandates trial designs that require central target engagement and mechanism-appropriate endpoints. Crucially, (i) Many neurodegenerative syndromes that resemble AD are part of a broader spectrum rather than classical AD (ii) a favorable prognosis does not imply functional recovery, since neuronal death is irreversible—instead, it should be understood as the absence of further exacerbation.

Keywords

alzheimer’s disease, spectrum, neuroinflammation, synaptopathy, node of ranvier, lithium, trial design

Introduction

Alzheimer’s disease (AD) is the leading cause of dementia worldwide and remains a major unmet medical challenge [1,11]. For decades, the amyloid cascade hypothesis has framed both basic research and drug development [2-4]. However, numerous large, randomized trials that successfully reduce amyloid plaque burden have shown only modest, inconsistent, or no meaningful cognitive benefit, and in some cases have introduced substantial safety concerns such as amyloid-related imaging abnormalities (ARIA) [12-17]. Persistent gaps between biomarker changes and clinical benefit underscore limitations of a single-pathway model and motivate a broader, calibrated framework that can account for biological heterogeneity and reconcile paradoxical findings [5,10].

Materials and methods

We performed an integrative conceptual synthesis. Sources included randomized clinical trials (phase II/III), neuropathological series, in vivo imaging studies (amyloid/tau PET, SV2A PET, TSPO PET), single-cell transcriptomic analyses, mechanistic animal experiments, epidemiology (including trace-element studies), and meta-analyses [6-8]. Recurrent paradoxes reported in the literature were mapped to one or more mechanistic nodes within the proposed spectrum, and an operational taxonomy plus a trial calibration checklist were derived to guide future research.

Results

Anti-amylis reduction without commensurate clinical benefit

Multiple monoclonal antibodies (aducanumab, lecanemab, donanemab) clearly lower amyloid PET signal, yet clinical effects on cognition and function are variable and often modest; early γ-secretase inhibitors worsened outcomes, highlighting the complexity of pathway modulation [13-19].

Synaptopathy as the principal structural correlation of cognition

Neuropathological and in vivo SV2A PET data show that synaptic density loss correlates more strongly with cognitive decline than plaque load, suggesting that interventions that preserve or restore synaptic integrity are central to modifying clinical course [20-31].

Neuroinflammation: Microglia as Context-Dependent Mediators Single-cell transcriptomics and disease-associated microglia (DAM) literature reveal distinct microglial states with both protective and deleterious functions; genetic modifiers such as TREM2 shift microglial trajectories and influence phenotype expression [3-8,9-36,].

Paranodal and node of ranvier dysfunction can impair conduction without gross demyelination

Emerging proteomic and histologic studies describe paranodal protein alterations and nodal disruption that can reduce saltatory conduction and disturb network timing, producing symptoms disproportionate to neuronal loss [29-39].

Peripheral immune drivers and microbiome contributions

Epidemiologic and mechanistic evidence links periodontal pathogens (e.g., Porphyromonas gingivalis) and gut dysbiosis to enhanced systemic and central inflammation, providing plausible pathways for peripheral immune priming to exacerbate central synaptic dysfunction [40-44].

Trace-element/metabolic modulators: Lithium as an illustrative node

Epidemiologic studies associate trace lithium levels in drinking water with reduced dementia incidence and recent translational work demonstrating brain lithium depletion and lithium-mediated rescue in animal models suggest a metabolic vulnerability axis that modulates synaptic resilience [45]. These findings warrant cautious clinical translation and rigorous PK/target engagement strategy [46-50,49-68].

A calibrated taxonomy: The alzheimer’s spectrum

We propose an operational taxonomy with five principal phenotypes: (i) Classical AD (amyloid–tau dominant), (ii) Inflammatory AD (neuroinflammation dominant), (iii) Synaptopathic AD (paranodal/myelin–axon interface dysfunction), (iv) Metabolic/Trace-element AD (e.g., lithium-deficit), and (v) Mixed AD. Each phenotype is defined by a priority set of biomarkers and mechanistic endpoints (Table 1) [10,69-71].

Table 1. Operational biomarker panel by phenotype

Phenotype

Dominant mechanism

Key biomarkers and endpoints

Potential therapeutic strategies

Classical AD

Amyloid-tau dominant

Amyloid PET, tau PET, plasma/CSF p-tau,

NfL

Amyloid-lowering antibodies (e.g.,

lecanemab)

Inflammatory AD

Neuroinflammation dominant

TSPO PET, CSF cytokines, microglial

markers (e.g., TREM2)

Immunomodulators, anti-inflammatory

agents

Synaptopathic AD

Paranodal/myelin-axon dysfunction

SV2A PET, EEG connectivity, myelin

imaging

Synaptic preservation, myelin-enhancing

therapies

Metabolic/Trace-element AD

Lithium-deficient or similar

CSF/plasma lithium, metabolic panels

Lithium formulations, metabolic

modulators

Mixed AD

Overlapping mechanisms

Comprehensive panel (all above)

Combination therapies matched to

dominant nodes

Trial and clinical implications

Adopting a spectrum view requires: (1) phenotype-stratified enrollment; (2) mandatory evidence of central target engagement for CNS-directed interventions (CSF drug levels, PET occupancy, or central biomarker modulation); (3) mechanism-appropriate primary endpoints (e.g., SV2A PET or EEG connectivity for synaptic targets; TSPO PET or CSF cytokines for immunomodulators); and (4) recognition that clinical stabilization or slowing of decline is a valid and meaningful outcome when neuronal loss is irreversible [9,20,26,51-54,72]. Many historical trial failures can be retrospectively rationalized by phenotype mixing, inadequate CNS exposure, or reliance on biomarker endpoints that were not proximal to the mechanistic node being targeted [13-19,58-61].

Practical trial calibration checklist

1. Baseline phenotype panel: amyloid PET, tau PET, TSPO PET (or CSF inflammatory panel), SV2A PET (if available), plasma/CSF NfL, APOE genotype, CSF/plasma lithium or surrogate, and cognitive reserve metrics.

2. Stratified randomization within phenotype strata.

3. Pharmacokinetic/target engagement confirmation: CSF drug level, PET occupancy, or central biomarker shift.

4. Primary endpoints aligned to mechanism + clinical co-endpoints with MCID and responder analyses.

5. Safety monitoring tailored to mechanism (e.g., ARIA monitoring for anti-amyloid antibodies; renal/thyroid for lithium formulations) [63,72].

Discussion

Reconceptualising AD as a spectrum reconciles multiple paradoxes and aligns therapeutic strategy with underlying pathobiology. For example, amyloid-lowering may be necessary but not sufficient in inflammatory or synaptopathic phenotypes. Conversely, immunomodulatory or metabolic strategies may deliver benefit only in phenotypes where central target engagement is demonstrable. Emphasis on synaptic endpoints, paranodal integrity, and central PK/target engagement will enhance the interpretability of trial outcomes and reduce false negatives driven by heterogeneity [73-80]. This framework also emphasizes early intervention: synaptic and conduction deficits precede overt neuronal loss, so interventions that restore conduction or synaptic function in prodromal stages are more likely to achieve clinical benefit than late-stage approaches [20,24,27,29,46,49].

Limitations

This manuscript is a conceptual and integrative synthesis rather than a novel primary data analysis. Some mechanistic links (notably trace-element depletion) are emergent and require confirmation in human randomized trials. Standardization of biomarker thresholds and broader access to advanced imaging (SV2A, TSPO) remain practical hurdles [52,81-85].

Conclusion

Alzheimer’s disease is better described as a spectrum of overlapping mechanistic phenotypes. A calibrated approach that matches interventions to biologic phenotype, demonstrates central target engagement, and uses mechanism-appropriate endpoints promises to transform the interpretation of trials and accelerate discovery of clinically meaningful therapies. Adopting a spectrum-based approach has the potential to transform therapeutic discovery and deliver meaningful clinical benefit.

Acknowledgements

The author thanks colleagues and interlocutors whose discussions informed this synthesis. Funding and conflicts of interest to be declared at submission.

Conflicts of interest

The author declares no conflict of interest.

References

  1. Hardy JA, Higgins GA (1992) Alzheimer's disease: The amyloid cascade hypothesis. Science 256: 184-185. [Crossref]
  2. Braak H, Braak E (1991) Neuropathological stageing of Alzheimer related changes. Acta Neuropathol 82: 239-259. [Crossref]
  3. Jack CR Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, et al. (2018) NIA-AA research framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement 14: 535-562. [Crossref]
  4. Karran E, Mercken M, De Strooper B (2011) The amyloid cascade hypothesis for Alzheimer’s disease: An appraisal for drug development therapeutics. Nat Rev Drug Discov 10: 698-712. [Crossref]
  5. Lab findings support the concept that reducing neuroinflammation could help fight Alzheimer's (2025). Sci Daily.
  6. University of Liverpool (2024) New discovery could offer significant answers on Alzheimer's disease. Sci Daily.
  7. Yi HY, Li JS, Trainer AE, Ficek‐Tani B, Constable T, et al. (2024) Functional connectivity of the affective blindsight pathway in atypical Alzheimer's disease. Alzheimers Dement 20: e093890. [Crossref]
  8. Bailey M, Ilchovska ZG, Hosseini AA, Jung JY (2024) Impact of apolipoprotein E ε4 in Alzheimer's disease a meta-analysis of voxel-based morphometry studies. J Clin Neurol 20: 1-10. [Crossref]
  9. Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, et al. (2024) Inflammatory signaling pathways in Alzheimer's disease. Ageing Res Rev 94: 102366. [Crossref]
  10. Xiao D, Zhang C (2024) Current therapeutics for Alzheimer’s disease and clinical trials. Explor Neurosci 3: 255-271.
  11. Cummings J, Zhong K, Kinney J (2025) Alzheimer's disease drug development pipeline: 2025. Alzheimers Dement (N Y) 11: 12131090.
  12. Selkoe DJ, Hardy J (2016) The amyloid hypothesis at 25 years. EMBO Mol Med 8: 595-608. [Crossref]
  13. Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, et al. (2016)The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 537: 50-56. [Crossref]
  14. van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, et al. (2023) Lecanemab in early Alzheimer’s disease. N Engl J Med 388: 9-21. [Crossref]
  15. Mintun MA, Lo AC, Duggan Evans C, Wessels AM, Ardayfio PA, et al. (2023) Donanemab in early symptomatic Alzheimer disease. N Engl J Med 388: 903-914. [Crossref]
  16. Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, et al. (2014) Phase 3 trials of solanezumab for mild-to-moderate Alzheimer's disease. N Engl J Med 370: 311-321. [Crossref]
  17. Salter MW, Stevens B (2017) Microglia emerge as central players in brain disease. Nat Med 23: 1018-1027. [Crossref]
  18. Salloway S, Sperling R, Gilman S (2014) A trial of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med 370: 322333.
  19. Mehta D, Jackson R, Paul G, Shi J, Sabbagh M, et al. (2017) Why do trials for Alzheimer's disease drugs keep failing? A discontinued drug perspective for 2010-2015. Expert Opin Investig Drugs 26: 735-739. [Crossref]
  20. Finnema SJ, Nabulsi NB, Eid T, Detyniecki k, Shu-Fei Lin, et al. (2016) Imaging synaptic density in the living human brain. Sci Transl Med 8: 348ra96. [Crossref]
  21. Chen MK, Mecca AP, Naganawa M, Finnema SJ, Toyonaga T, et al. (2018) Assessing synaptic density in Alzheimer disease with SV2A PET imaging. JAMA Neurol 75: 1215-1224. [Crossref]
  22. Palop JJ, Mucke L (2010) Amyloid-β–induced neuronal dysfunction: From synapses toward neural networks. Nat Neurosci 13: 812-818. [Crossref]
  23. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, et al. (2016) Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352: 712-716. [Crossref]
  24. Henstridge CM, Hyman BT, Spires-Jones TL (2019) Beyond the neuron—cellular interactions early in Alzheimer disease pathogenesis. Nat Rev Neurosci 20: 94-108. [Crossref]
  25. De Strooper B, Karran E (2016) The cellular phase of Alzheimer’s disease. Cell 164: 603-615. [Crossref]
  26. Zlokovic BV (2011) Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci 12: 723-738. [Crossref]
  27. Dean DC, Hurley SA, Kecskemeti SR, O'Grady JP, Canda C, et al. (2017) Association of amyloid pathology with myelin alteration in preclinical Alzheimer disease. JAMA Neurol 74: 41-49. [Crossref]
  28. Stadelmann C, Reimers C, Bruck W (2005) Myelin loss in early Alzheimer's disease and its correlation with cognitive decline. Brain 128: 285-296.
  29. Poliak S, Peles E (2003) The local differentiation of myelinated axons at nodes of ranvier. Nat Rev Neurosci 4: 968-980. [Crossref]
  30. Bradford CR, Koppel J, Chen H (2021) Node of ranvier and paranode pathology in neurodegenerative disease: Mechanisms and implications. Brain Res Bull. 171: 88-99.
  31. Kamer AR, Craig RG, Dasanayake AP, Brys M, Glodzik-Sobanska L, et al. (2008) Inflammation and Alzheimer’s disease: Possible role of periodontal pathogens. J Alzheimers Dis 13: 437-449. [Crossref]
  32. Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, et al. (2017) A unique microglia type associated with restricting development of Alzheimer's disease. Cell 169: 1276-1290.e17. [Crossref]
  33. Mathys H, Adaikkan C, Gao F, Young JZ, Manet E, et al. (2019) Temporal tracking of microglia and astrocyte transcriptomic changes in Alzheimer’s disease. Nature 573: 324-331. [Crossref]
  34. Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M, et al. (2016) TREM2 binds to apolipoproteins, facilitating uptake of amyloid-β by microglia. Neuron 91: 328340. [Crossref]
  35. Labzin LI, Heneka MT, Latz E (2018) Innate immunity and neurodegeneration. Annu Rev Neurosci 38: 157-182. [Crossref]
  36. Swanson CJ, Zhang Y, Dhadda S (2021) Safety and efficacy of donanemab in Alzheimer disease. JAMA 325: 50-60.
  37. Susuki K, Rasband MN (2008) Spectrin and ankyrin-based cytoskeleton at polarized domains in myelinated axons. Neurosci Res 61: 358-365. [Crossref]
  38. Zhang SC, Liu L, Wang Y (2022) Paranodal alterations and conduction defects in Alzheimer's disease models. Neurobiol Dis 162: 105555. [Crossref]
  39. Duggan MR, Morgan DG, Price BR, Rajbanshi B, Martin-Peña A, et al. (2025) Immune modulation to treat Alzheimer’s disease. Mol Neurodegener 20: 39. [Crossref]
  40. Dominy SS, Lynch C, Ermini F, Benedyk M, Marczyk M, et al. (2019) Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci Adv 5: 3333. [Crossref]
  41. Poole S, Singhrao SK, Kesavalu L (2013) Determining the presence of porphyromonas gingivalis DNA in brains of elderly people with and without Alzheimer’s disease. J Oral Microbiol 5: 22768.
  42. Riviere GR, Riviere KH, Smith KS (2002) Molecular and immunological evidence of oral treponema in the human brain and their association with Alzheimer's disease. Oral Microbiol Immunol 17: 113-118. [Crossref]
  43. Kessing LV, Jorgensen MB, Kessing SV (2017) Lithium in drinking water and dementia risk: A nationwide population-based study. J Alzheimers Dis 60: 301-309.
  44. Lin S , Zhan Y, Wang R, Pei J (2025) Decoding neuroinflammation in Alzheimer's disease: A multi-omics perspective. Front Immunol 16: 1616899. [Crossref]
  45. Aron L, Ngian ZK, Qiu C, Choi J, Liang M, et al. (2025) Lithium deficiency and the onset of Alzheimer’s disease. Nature 645: 712–721. [Crossref]
  46. Kessing LV, Gerds TA, Knudsen NN, Jørgensen MB, Andersen PK, et al. (2017) Association of lithium in drinking water with the incidence of dementia. JAMA Psychiatry 74: 1005–1010.[Crossref]
  47. Peterson D, Harman S (2019) Trace elements and brain aging: Lithium as a neuroprotective microelement. Neurosci Lett 700: 10-18.
  48. Mattsson N, Zetterberg H, Janelidze S, Insel SP, Andreasson U, et al. (2016) Plasma tau in Alzheimer disease. Neurology 87: 1827-1835. [Crossref]
  49. Blennow K, Zetterberg H (2018) Biomarkers for Alzheimer’s disease: Current status and prospects for the future. J Intern Med 284: 643-663. [Crossref]
  50. Palmqvist S, Janelidze S, Quiroz YT, LoperaF, Stomrud E, et al. (2020)Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 324: 772-781. [Crossref]
  51. Jack CR Jr, Knopman DS, Jagust WJ, Petersen RC, Weiner MW, et al. (2013) Tracking pathophysiological processes in Alzheimer's disease: An updated hypothetical model of dynamic biomarkers. Lancet Neurol 12: 207-216. [Crossref]
  52.  Boche D, GerhardA, Rodriguez-Vieitez E, MINC Faculty (2019) Prospects and challenges of imaging neuroinflammation beyond TSPO in Alzheimer's disease. Eur J Nucl Med Mol Imaging 46: 2831-2847. [Crossref]
  53. O'Bryant SE, Mielke MM, Rissman RA, Lista S, Vanderstichele H, et al. (2017) Blood-based biomarkers in Alzheimer disease: Current state of the science and a novel collaborative paradigm for advancing from discovery to clinic. Alzheimers Dement 13: 45-58. [Crossref]
  54. Cummings J, Lee G, Zhong K, Fonseca J, Taghva K, et al.(2021) Alzheimer's disease drug development pipeline: 2021. Alzheimers Dement 7: e12179. [Crossref]
  55. Rosenberg PB, Shulman MB, Marson D (2020) Drug trials in Alzheimer disease: Barriers to success and new directions. JAMA 323: 236-247.
  56. Wilcock DM, Colton CA (2010) Anti-inflammatory strategies for Alzheimer's disease: a translational perspective. Neurobiol Aging 3: 753-764.  
  57. Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, et al. (2013) A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N Eng J Med 369: 341-350. [Crossref]
  58. Henley DB, Sundell KL, Sethuraman G, Dowsett SA, May PC, et al. (2014) Safety profile of semagacestat, a gamma-secretase inhibitor: IDENTITY trial findings. Curr Med Res Opin 30: 2021-2032. [Crossref]
  59. Kuller LH, Lopez OL (2021) ENGAGE and EMERGE: Truth and consequences?. Alzheimers Dement 17: 692-695. [Crossref]
  60. Warlow SM, Robinson MJ, Berridge KC (2017) Optogenetic central amygdala stimulation intensifies and narrows motivation for cocaine. J Neurosci 37: 8330-8348. [Crossref]
  61. Roe CM, Xiong C, Miller JP, Morris JC (2010) Biomarkers predict progression in Alzheimer disease: Implications for design of clinical trials. Neurology 75: 1151-1157.
  62. Cordonnier C, Klijn C, Smith EE, Al-Shahi Salman R, Chwalisz BK, et al. (2025) Diagnosis and management of cerebral amyloid angiopathy: A scientific statement from the International CAA Association and the World Stroke Organization. Int J Stroke 20: 949-967. [Crossref]
  63. ARIA management guidance: Expert consensus (2022) Alzheimers Res Ther 14: 47.
  64. Clinical trials failure analysis review (2019) Nat Rev Drug Discov 18: 559-577.
  65. Svendsen CN (2018) Failures in translation: Why targets in AD succeed in animals but not humans. Trends Neurosci 41: 541-557. [Crossref]
  66. Frisoni GB, Fox NC, Jack CR Jr, Scheltens P, Thompson PM, et al. (2010) The clinical use of structural MRI in Alzheimer disease. Nat Rev Neurol 6: 67-77. [Crossref]
  67. Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, et al. (2016) Preclinical Alzheimer's disease: Definition, natural history, and diagnostic criteria. Lancet Neurol 12: 292-323. [Crossref]
  68.  Kamila P, Kar K, Chowdhury S, Chakraborty P, Dutta R, et al. (2025) Effect of neuroinflammation on the progression of Alzheimer's disease and its significant ramifications for novel anti-inflammatory treatments. J Neuroimmunol 389: 577923. [Crossref]
  69. Escamilla S, Badillos R, Comella JX, Solé M, Pérez‐Otaño I, et al. (2024) Synaptic and extrasynaptic distribution of NMDA receptors in the cortex of Alzheimer's disease patients. Alzheimers Dement 12: 8231-8245.[Crossref]
  70.  Roveta F,Bonino L, Piella EM, Rainero I, Rubino E, et al. (2024) Neuroinflammatory biomarkers in Alzheimer's disease. From pathophysiology to clinical implications. Int J Mol Sci 25: 11941. [Crossref]
  71. Sharma A, Rudrawar S, Bharate SB, Jadhav HR (2024) Recent advancements in the therapeutic approaches for Alzheimer's disease treatment: Current and future perspective RSC Med Chem 16: 652-693. [Crossref]
  72. Barrantes FJ (2024) Cognitive synaptopathy: Synaptic and dendritic spine dysfunction in Alzheimer's disease. Front Aging Neurosci 16: 1476909. [Crossref]
  73. Manap ASA, Almadodi R, Sultana S, Sebastian MG, Kavani KS, et al. (2024) Alzheimer's disease: A review on the current trends of the effective diagnosis and therapeutics. Front Aging Neurosci 16: 1429211. [Crossref]
  74.  Pan J, Hu J, Meng D, Chen L, Wei X, et al. (2024) Neuroinflammation in dementia: A meta-analysis of PET imaging studies. Medicine (Baltimore) 103: 38012. [Crossref]
  75. Heneka MT, Van der Flier WM, Jessen F, Hoozemanns J, Thal DR, et al. (2024) Neuroinflammation in Alzheimer disease. Nat Rev Immunol 25: 321-352. [Crossref]
  76. Mayo Clinic Staff (2024) Alzheimer's treatments: What's on the horizon? Mayo Clin.
  77.  Planalp EM, Langhough RE, Jonaitis EM, Hulle CAV, Wilson RE, et al. (2025) Synaptic markers are associated with cognitive decline after Alzheimer's disease diagnosis. Sci Rep 15: 4319. [Crossref]
  78. Amato LG, Vergani AA, Lassi M, Carpaneto J, Mazzeo S, et al. (2025) Personalized brain models link cognitive decline progression to underlying synaptic and connectivity degeneration. Alzheimers Res Ther 17: 74.[Crossref]
  79. Cheng Q, Fan Y, Zhang P, Liu H, Han J, et al. (2025) Biomarkers of synaptic degeneration in Alzheimer’s disease. Ageing Res Rev 104: 102642. [Crossref]
  80. Taddei RN, Duff KE (2025) Synapse vulnerability and resilience underlying Alzheimer’s disease. EBioMedicine 112: 105557.[Crossref]
  81. Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, et al. (2024) Recent advances in Alzheimer's disease mechanisms, clinical trials and new drug development strategies. Signal Transduct Target Ther 9: 211. [Crossref]
  82. Heneka MT, Wiesje Flier WMVR, Jessen F, Hoozemanns J, Thal DR, et al. (2024) Neuroinflammation in Alzheimer disease. Lancet Neurol 23: 802-813. [Crossref]
  83. Lee E, Chang Y (2025) Modulating neuroinflammation as a prospective therapeutic target in Alzheimer’s Disease. Cells 14: 168.[Crossref]
  84.  Heneka MT , Gauthier S, Chandekar SA, Hahn-Pedersen JH, Bentsen MA, et al. (2025) Neuroinflammatory fluid biomarkers in patients with Alzheimer's disease. A systematic review and meta-analysis. Mol Psychiatry 30: 1-12. [Crossref]
  85. Bartholomew SK, Winslow W, Sharma R, Pathak KV, Tallino S, et al. (2024) Glyphosate exposure exacerbates neuroinflammation and Alzheimer’s disease-like pathology despite a 6-month recovery period in mice. J Neuroinflammation 21: 316.[Crossref]

Editorial Information

Editor-in-Chief

Shangming Zhang
Penn State Hershey Medical Center, USA

Article Type

Review Article

Publication history

Received: September 17, 2025
Accepted: October 08, 2025
Published: October 15, 2025

Copyright

©2025 Hoang NDH. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Hoang NDH (2025) Alzheimer's disease as a multi‑directional spectrum: Integrating neuroinflammation, synaptopathy, paranodal dysfunction, and metabolic trace deficits. Neuro Neurosurg 8: DOI: 10.15761/NNS.1000147.

Corresponding author

Nguyen Duc Huy Hoang

Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam

Table 1. Operational biomarker panel by phenotype

Phenotype

Dominant mechanism

Key biomarkers and endpoints

Potential therapeutic strategies

Classical AD

Amyloid-tau dominant

Amyloid PET, tau PET, plasma/CSF p-tau,

NfL

Amyloid-lowering antibodies (e.g.,

lecanemab)

Inflammatory AD

Neuroinflammation dominant

TSPO PET, CSF cytokines, microglial

markers (e.g., TREM2)

Immunomodulators, anti-inflammatory

agents

Synaptopathic AD

Paranodal/myelin-axon dysfunction

SV2A PET, EEG connectivity, myelin

imaging

Synaptic preservation, myelin-enhancing

therapies

Metabolic/Trace-element AD

Lithium-deficient or similar

CSF/plasma lithium, metabolic panels

Lithium formulations, metabolic

modulators

Mixed AD

Overlapping mechanisms

Comprehensive panel (all above)

Combination therapies matched to

dominant nodes