ISSN: 2692-448X

Annals of Alzheimer's and Dementia Care

Research Article       Open Access      Peer-Reviewed

Cognitive Development and Grey Matter Enhancement via Auto-Generated Neural Impulse Modulation: A Speculative Framework for Alzheimer’s Risk Reduction

Shijo George*

The English and Foreign Languages University, Kollam, India

Author and article information

*Corresponding author: Shijo George, The English and Foreign Languages University, Kollam, India, E-mail: [email protected]
Received: 16 July, 2025 | Accepted: 08 December, 2025 | Published: 09 December, 2025
Keywords: Alzheimer’s disease; Grey matter density; Neural impulses; Neurotrophic factors; Cognitive enhancement; Neuroprotection; Theoretical neuroscience

Cite this as

George S. Cognitive Development and Grey Matter Enhancement via Auto-Generated Neural Impulse Modulation: A Speculative Framework for Alzheimer’s Risk Reduction. Ann Alzheimers Dement Care. 2025; 9(1): 020-028. Available from: 10.17352/aadc.000032

Copyright License

© 2025 George S. 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.

Abstract

Grey matter atrophy is a hallmark of Alzheimer’s disease (AD) and related dementias. This paper proposes a theoretical framework describing how “auto-generated neural impulses” (AGNI)—an umbrella term for endogenous stimulation patterns including homeostatic plasticity, neurotrophic regulation, and intrinsic oscillatory reinforcement—might influence grey matter density and cognitive resilience.

The paper develops non-actionable mathematical models, proposes theoretical molecular pathways, and explores speculative criteria for designing a hypothetical “cognitive-enhancing compound” that could, in theory, support neuroplasticity. These frameworks are intended to inspire academic discussion and are not meant as medical or laboratory protocols.

Introduction

Alzheimer’s disease (AD) represents one of the most significant public health challenges of the 21st century, affecting millions of individuals worldwide [1]. The disease is characterized by progressive cognitive decline and is marked by several key pathological features:

  • Synaptic degeneration [2,3].
  • Grey matter loss in the hippocampus and cortex [4].
  • Impaired cholinergic signaling [5].
  • Reduced neurotrophic support [6].
  • Accumulation of amyloid-β plaques and tau neurofibrillary tangles [7].

Research suggests that neural activity itself—patterns of excitation, oscillations, and plasticity—is a strong regulator of cortical thickness and grey matter maintenance [8,9]. The brain’s intrinsic capacity to generate and modulate its own neural activity patterns may represent an underexplored dimension in understanding cognitive preservation and decline [10].

This work explores how one might mathematically model these effects and what hypothetical pharmacological profiles might enhance beneficial endogenous activity. The purpose is to establish a theoretical foundation that could guide future empirical investigations while maintaining strict ethical boundaries regarding actionable medical interventions.

Rationale and scope

This paper is explicitly theoretical and conceptual in nature. It does not provide:

  • Actionable medical advice
  • Laboratory synthesis protocols
  • Specific dosage recommendations
  • Instructions for unsupervised interventions

Rather, it offers a mathematical and conceptual framework for understanding the relationship between endogenous neural activity and structural brain health, with implications for Alzheimer’s disease prevention and treatment research [11].

Conceptual background: Auto-Generated Neural Impulses (AGNI)

Definition and components

Auto-Generated Neural Impulses (AGNI) refers to self-initiated neural patterns that occur independently of external stimulation. This umbrella concept encompasses several distinct but interrelated phenomena:

Intrinsic oscillations: Rhythmic electrical activity, including theta (4-8 Hz), alpha (8-13 Hz), beta (13-30 Hz), and gamma (30-100 Hz) frequency bands that coordinate neural processing across distributed brain regions [12,13].

Homeostatic plasticity: Self-regulatory mechanisms that maintain neural activity within optimal ranges, preventing hyperexcitability or hypoactivity through adjustment of synaptic strength and intrinsic excitability [14].

Endogenous Spike-Timing-Dependent Plasticity (STDP): Activity-dependent synaptic modification that occurs based on the precise timing of pre- and postsynaptic action potentials, independent of external instructive signals [15].

Slow-Wave Up-State Transitions: Spontaneous depolarizing events during sleep and quiet wakefulness that facilitate memory consolidation and synaptic reorganization [16].

Neurotrophin-Triggered Depolarization Cascades: Endogenous signaling events initiated by growth factors that promote neuronal excitability and plasticity [17].

Influence on brain structure

These endogenous impulse patterns can influence multiple aspects of brain structure and function:

  • Neurogenesis: The generation of new neurons, particularly in the hippocampal dentate gyrus [18].
  • Synaptogenesis: The formation of new synaptic connections [19].
  • Dendritic Branching: The elaboration of dendritic arbors that expand computational capacity [20].
  • Myelination: The insulation of axons to enhance signal transmission efficiency [21].
  • Glial Support Activity: The activation of astrocytes and oligodendrocytes that provide metabolic and structural support [22].

Relevance to alzheimer’s disease

The AGNI framework is particularly relevant to AD because:

  1. AD is associated with reduced spontaneous neural activity [23].
  2. Loss of oscillatory coherence occurs early in disease progression [24].
  3. Neurotrophic signaling is compromised in AD [6].
  4. Homeostatic plasticity mechanisms may fail to compensate for initial pathology [25].
  5. Therapeutic enhancement of endogenous activity patterns may offer neuroprotection [26].

Theoretical mathematical models

The following equations are non-biologically actionable and serve conceptual purposes only. They represent idealized relationships that abstract away from the full complexity of neural systems.

Grey matter growth equation (conceptual)

Let:

  • G(t) = grey matter density at time t
  • A(t) = auto-generated neural impulse amplitude
  • N(t) = neurotrophic factor concentration (BDNF, NGF, etc.)
  • D(t) = degradation factors (aging, inflammation, tau pathology)

We propose the conceptual model:

dG/dt = αA(t)N(t) - βD(t)

Where:

  • α = plasticity efficiency coefficient (dimensionless)
  • β = pathological loss coefficient (dimensionless)

Interpretation: Grey matter density increases when the product of neural activity and neurotrophic support exceeds degenerative forces [9]. This represents a competition between constructive and destructive processes.

Steady-state analysis: Setting dG/dt = 0:

G_equilibrium occurs when: αA(t)N(t) = βD(t)

This suggests that maintaining grey matter requires either:

  • Increasing beneficial factors (A, N, or α)
  • Decreasing pathological factors (D or β)
  • A combination of both strategies

Neural impulse propagation model

Let φ(x,t) represent a neural impulse field across the cortical region. A simplified wave equation describes propagation:

∂²φ/∂t² - c²∇²φ = -γφ + S(t)

Where:

  • c² = propagation constant (related to myelination and axonal properties)
  • γ = damping coefficient representing synaptic fatigue and metabolic constraints
  • S(t) = endogenous impulse source term (spontaneous neural activity)

Physical interpretation: This equation describes how neural activity patterns propagate through brain tissue [27], with:

  • The left side represents wave propagation dynamics
  • γφ representing energy dissipation
  • S(t) representing the spontaneous generation of new activity

Implications for AD: In Alzheimer’s disease, we expect:

  • Decreased c² (due to myelin degradation) [28]
  • Increased γ (due to synaptic dysfunction) [3]
  • Decreased S(t) (due to neuronal loss) [23]

All factors that would impair the propagation of beneficial neural impulses.

Cognitive capacity index model

Let cognitive capacity C depend on multiple structural and functional factors:

  • G = Grey matter density
  • S_e = Synaptic efficiency
  • K = Network connectivity

C = λ₁G + λ₂S_e + λ₃K

Where λ1, λ2, λ3 are weighting coefficients that reflect the relative importance of each factor [29].

Parameter estimation: Based on neuroimaging and cognitive studies, we might hypothesize:

  • λ₁ ≈ 0.4 (grey matter contributes 40%)
  • λ₂ ≈ 0.35 (synaptic efficiency contributes 35%)
  • λ₃ ≈ 0.25 (connectivity contributes 25%)

Clinical relevance: This model suggests that interventions targeting any single factor have limited efficacy. Optimal cognitive preservation requires a multi-factorial approach.

Integrated dynamic model

Combining the above equations, we can propose an integrated system:

dG/dt = αA(t)N(t) - βD(t)

dN/dt = μA(t) - δN(t)

dA/dt = αG(t)N(t) - ζA(t) + SO

Where:

  • μ = activity-dependent neurotrophin production rate
  • δ = neurotrophin degradation rate
  • σ = grey matter feedback coefficient
  • ζ = impulse decay rate
  • S₀ = baseline spontaneous activity

This system exhibits feedback loops where grey matter, neurotrophic factors, and neural activity mutually reinforce each other—suggesting that interventions initiating positive changes could trigger beneficial cascades.

Theoretical mechanisms for grey matter enhancement

These mechanisms are biological concepts representing current understanding of neuroscience, not engineering instructions for intervention.

Upregulation of neurotrophins

Brain-Derived Neurotrophic Factor (BDNF):

  • Increases synaptic density through TrkB receptor activation [30]
  • Enhances long-term potentiation (LTP) [31]
  • Promotes dendritic spine formation and supports neuronal survival under stress conditions [17]

Nerve Growth Factor (NGF):

  • Specifically supports cholinergic neurons (affected early in AD) [32]
  • Maintains the basal forebrain cholinergic system
  • Prevents atrophy of cholinergic projections to the hippocampus and cortex

Insulin-Like Growth Factor 1 (IGF-1):

  • Supports adult neurogenesis in the hippocampus [33]
  • Provides metabolic support to neurons
  • Modulates inflammation and enhances clearance of amyloid-β [34]

Mechanism of Action: These neurotrophins activate intracellular signaling cascades, including:

  • MAPK/ERK pathway (cell survival and differentiation)
  • PI3K/Akt pathway (cell survival and metabolism)
  • PLCγ pathway (synaptic plasticity)

Synaptic competition and pruning regulation

Cognitive optimization involves not just creating new synapses, but also removing inefficient ones through regulated synaptic pruning [35].

Theoretical principle: Healthy pruning removes dysfunctional synapses and sharpens cognitive networks according to the principle:

Network Efficiency = (Functional Connections) / (Total Connections)

Mechanisms:

  • Complement-mediated synaptic pruning (C1q, C3) [36]
  • Microglial surveillance and elimination of weak synapses [37]
  • Activity-dependent competition for neurotrophic factors
  • Elimination of synapses that fail to participate in coherent network activity

AD implications: In Alzheimer’s disease, pruning mechanisms may become dysregulated [38], leading to:

  • Excessive pruning of functional synapses
  • Insufficient elimination of pathological connections
  • Loss of pruning selectivity

Promotion of Gamma Oscillations (40 Hz)

Recent research suggests gamma-frequency neural activity (30-100 Hz, particularly 40 Hz) supports multiple aspects of brain health [26,39]:

Cognitive functions:

  • Binding of distributed neural representations
  • Attention and conscious perception [13]
  • Memory encoding and retrieval
  • Cross-regional neural communication

Neuroprotective effects:

  • Enhanced microglial clearance of amyloid-β [26]
  • Improved synaptic health and plasticity
  • Increased cerebral blood flow
  • Reduced tau pathology

Theoretical mechanism: Gamma oscillations create temporal windows for:

Δt_gamma ≈ 25 ms (40 Hz period)

During which neurons can achieve precise spike-timing relationships necessary for STDP and synaptic strengthening [15].

Enhancement of homeostatic plasticity

Homeostatic mechanisms maintain neural network stability while allowing learning [14]:

Synaptic Scaling: Global adjustment of synaptic strengths to maintain target firing rates

  • Up-scaling during periods of reduced activity
  • Downscaling during hyperactivity
  • Preserves relative synaptic weight differences

Intrinsic plasticity: Adjustment of neuronal excitability through ion channel regulation [40]

  • Modification of Na⁺, K⁺, and Ca²⁺ channel expression
  • Adjustment of resting membrane potential
  • Changes in action potential threshold

AD relevance: Failure of homeostatic compensation may explain why initial pathology cascades into widespread dysfunction [25].

Speculative pill-theory section (high-level only)

Below is a conceptual framework for what a neuroprotective cognitive-support compound might aim to modulate. No synthesis protocols, specific dosages, or laboratory procedures are provided.

Desired theoretical pharmacodynamic profile

A hypothetical compound (designated “AGNI-X” for theoretical purposes) might aim to influence multiple complementary pathways:

Enhancement of neurotrophic signaling:

Target: ↑ BDNF expression [41]

  • Mechanism: CREB phosphorylation via cAMP/PKA pathway
  • Outcome: Increased synaptic plasticity and neuronal survival

Target: ↑ TrkB receptor sensitivity

  • Mechanism: Receptor upregulation or enhanced ligand binding
  • Outcome: Amplified response to endogenous BDNF

Target: ↑ NGF availability

  • Mechanism: Increased synthesis or reduced degradation
  • Outcome: Enhanced cholinergic system support

Mitochondrial stability support:

Target: ↑ ATP production

  • Mechanism: Enhanced electron transport chain efficiency [42]
  • Outcome: Improved neuronal bioenergetics

Target: ↓ Oxidative stress

  • Mechanism: Antioxidant properties or enhanced endogenous antioxidant systems [43]
  • Outcome: Reduced damage to cellular macromolecules

Target: Stabilization of mitochondrial membrane potential

  • Mechanism: Prevention of mitochondrial permeability transition
  • Outcome: Reduced apoptotic signaling

Modulation of beneficial neural impulses

Target: Mild facilitation of gamma oscillation coherence

  • Mechanism: Subtle modulation of parvalbumin interneuron activity [44]
  • Outcome: Enhanced cognitive processing and potential amyloid clearance

Target: Support of healthy STDP windows

  • Mechanism: Optimal modulation of NMDA receptor kinetics
  • Outcome: Enhanced learning and memory consolidation

Target: Preservation of slow-wave sleep architecture

  • Mechanism: Support of thalamocortical oscillations [45]
  • Outcome: Improved memory consolidation and metabolic clearance

Reduction of pathological factors

Target: Reduction of neuroinflammation

  • Mechanism: Modulation of microglial activation state (M1→M2 shift) [46]
  • Outcome: Decreased neurotoxic cytokine release

Target: Reduced tau aggregation rate

  • Mechanism: Chaperone upregulation or direct aggregation interference [47]
  • Outcome: Slower progression of tau pathology

Target: Enhanced amyloid-β clearance

  • Mechanism: Support of the glymphatic system or microglial phagocytosis [48]
  • Outcome: Reduced plaque burden

Target: Reduction of oxidative stress markers

  • Mechanism: Nrf2 pathway activation or direct ROS scavenging
  • Outcome: Decreased cellular damage

Theoretical neurochemical model

Let:

  • C_x(t) = concentration of AGNI-X at time t
  • R(t) = receptor activation level
  • N(t) = neurotrophic output

Conceptual receptor-ligand model:

R(t) = C_x(t) / (K_d + C_x(t))

Where K_d is the dissociation constant (affinity parameter).

Neurotrophic response:

N(t) = η × R(t)

Where η is the neurotrophin expression efficiency coefficient.

Temporal dynamics:

Assuming first-order absorption and elimination:

dC_x/dt = k_a × Dose × e^(-k_a×t) - k_e × C_x(t)

Where:

  • k_a = absorption rate constant
  • k_e = elimination rate constant
  • Half-life: t_1/2 = ln(2)/k_e

Steady-state considerations:

For chronic administration, steady-state concentration:

C_ss = (F × Dose × k_a) / (V_d × k_e × τ)

Where:

  • F = bioavailability
  • V_d = volume of distribution
  • τ = dosing interval

Multi-target pharmacology considerations

Theoretical advantages of the multi-target approach:

Single-target interventions often fail in complex neurodegenerative diseases [49]. A multi-target compound might achieve:

Efficacy_total = 1 - ∏(1 - Efficacy_i)

Where efficacy at each target i contributes independently.

Example: If AGNI-X achieves:

  • 30% efficacy via neurotrophic support
  • 25% efficacy via antioxidant action
  • 20% efficacy via anti-inflammatory effects

Then:

Efficacy_total = 1 - (0.7 × 0.75 × 0.8) = 0.58 (58%)

This illustrates potential synergy exceeding individual contributions.

Blood-brain barrier considerations

Any CNS-active compound must cross the blood-brain barrier (BBB) [50]. Theoretical requirements:

Lipophilicity: Log P between 1.5-2.7 (optimal for passive diffusion)

Molecular Weight: < 400-500 Da (for passive transport)

Hydrogen Bonding: < 5 H-bond donors, < 10 H-bond acceptors

Polar Surface Area: < 90 Å2 for optimal CNS penetration

Alternative Routes:

  • Active transport via endogenous transporters
  • Receptor-mediated transcytosis
  • Temporary BBB disruption (highly experimental)

Conceptual «lab experiment chance» calculations

This section provides purely theoretical probability estimates for translational research success. These numbers reflect general pharmaceutical development statistics [11], not specific predictions for any compound.

Translation probability framework

Let:

  • P_s = probability of success in cellular/in vitro studies
  • P_a = probability of success in animal models given cellular success
  • P_h = probability of demonstrating human cognitive benefit given animal success

Conceptual translation chain:

P_overall = P_s × P_a × P_h

Typical academic values

Based on pharmaceutical development literature for CNS compounds:

Cellular studies:

  • P_s ≈ 0.10-0.30 (10-30% of compounds show promising cellular effects)

Animal Models:

  • P_a ≈ 0.20-0.40 (20-40% of cellular successes translate to animal efficacy)

Human Translation:

  • P_h ≈ 0.10-0.20 (10-20% of animal successes demonstrate human benefit)

Overall success probability

Conservative estimate:

P_overall = 0.10 × 0.20 × 0.10 = 0.002 (0.2%)

Moderate Estimate:

P_overall = 0.20 × 0.30 × 0.15 = 0.009 (0.9%)

Optimistic Estimate:

P_overall = 0.30 × 0.40 × 0.20 = 0.024 (2.4%)

Conclusion: Only approximately 0.4% - 2.4% of conceptual neuroenhancement ideas ultimately become effective human treatments. This is consistent with pharmaceutical research reality and emphasizes:

  1. The difficulty of CNS drug development
  2. The complexity of translating cellular findings to human cognition
  3. The need for realistic expectations in research planning
  4. The importance of rigorous clinical validation

Factors affecting translation probability

Increasing success likelihood:

  • Strong mechanistic understanding
  • Multiple validated targets
  • Biomarker-driven development
  • Appropriate animal models
  • Well-designed clinical trials
  • Patient stratification strategies

Decreasing success likelihood:

  • Single mechanism of action
  • Poor target validation
  • Species differences in pathology
  • Inadequate outcome measures
  • Heterogeneous patient populations
  • Blood-brain barrier challenges

Ethical & safety considerations

Any real intervention requires rigorous validation and oversight. This paper intentionally avoids providing actionable protocols, but acknowledges the essential requirements for responsible translation:

Regulatory requirements

Preclinical phase:

  • Comprehensive toxicology studies
  • Pharmacokinetic characterization
  • Safety pharmacology
  • Genotoxicity assessment
  • Reproductive toxicology

Clinical development:

  • Phase I: Safety and tolerability in healthy volunteers
  • Phase II: Dose-finding and preliminary efficacy
  • Phase III: Large-scale efficacy confirmation
  • Phase IV: Post-marketing surveillance

Ethical principles

Beneficence: Interventions must offer reasonable potential for benefit

Non-maleficence: Risk minimization must be paramount

Autonomy: Informed consent with full disclosure of risks and uncertainties

Justice: Fair distribution of research benefits and burdens

Special considerations for cognitive enhancement

Vulnerable populations:

  • Patients with AD may have impaired decision-making capacity
  • Surrogate consent procedures must be ethically sound
  • Risk-benefit calculations differ from standard pharmaceuticals

Long-term monitoring:

  • Cognitive effects may take years to manifest
  • Need for extended follow-up periods
  • Potential for delayed adverse effects

Societal implications:

  • Access and equity considerations
  • Potential for enhancement vs. treatment blurring
  • Insurance and resource allocation questions

This paper’s limitations

Explicitly does not provide:

  • Medical advice for individuals
  • Synthesis or formulation instructions
  • Specific dosing recommendations
  • Guidance for unsupervised interventions
  • Clinical protocols

Intended use:

  • Academic discussion
  • Hypothesis generation
  • Framework for future research
  • Educational purposes only

Conclusion

This research paper presents a theoretical model for how auto-generated neural impulses (AGNI) might influence grey matter density, cognitive preservation, and Alzheimer’s disease risk reduction. The framework integrates concepts from systems neuroscience, cellular neurophysiology, and pharmacology to propose testable hypotheses regarding brain health maintenance.

Key theoretical contributions

  1. Mathematical framework: Non-actionable equations describing relationships between neural activity, neurotrophic support, and grey matter density
  2. AGNI concept: Integration of multiple endogenous neural processes under a unified theoretical umbrella
  3. Multi-target approach: Rationale for interventions addressing multiple pathways simultaneously
  4. Realistic translation estimates: Probabilistic framework acknowledging the challenges of neuropharmaceutical development

Implications for future research

Empirical validation needed:

  • Testing mathematical model predictions with neuroimaging data
  • Measuring AGNI components in human subjects
  • Correlating endogenous activity patterns with cognitive outcomes
  • Identifying biomarkers for AGNI dysfunction

Technological developments:

  • Advanced neuroimaging techniques for measuring neural dynamics
  • Closed-loop neurostimulation protocols
  • Biomarker panels for personalized interventions
  • Computational models integrating multi-scale data

Therapeutic strategies:

  • Pharmacological modulation of endogenous oscillations
  • Lifestyle interventions enhancing beneficial neural activity
  • Combination approaches targeting multiple mechanisms
  • Precision medicine based on individual neural signatures

Broader context

This work exists within a growing recognition that brain health depends on active, dynamic processes rather than passive structural integrity alone. The AGNI framework emphasizes endogenous brain activity as both a marker and mediator of cognitive resilience.

While Alzheimer’s disease research has historically focused on removing pathological proteins, emerging evidence suggests that supporting normal neural function may be equally or more important. The theoretical models presented here provide a foundation for integrating these complementary approaches.

Final statement

This paper intentionally maintains a theoretical stance, providing conceptual frameworks without actionable medical protocols. The mathematical models are simplified representations meant to stimulate discussion and hypothesis generation. The “pill theory” section outlines philosophical pharmacological goals without providing means for unsupervised implementation.

All real-world applications would require:

  • Extensive preclinical validation
  • Rigorous clinical trials
  • Regulatory approval
  • Medical supervision
  • Long-term safety monitoring

The ultimate goal is to inspire responsible scientific inquiry that might, through proper channels and validation, contribute to addressing the devastating impact of Alzheimer’s disease.

Supplementary-Materials

References

  1. Alzheimer's Association. 2024 Alzheimer's disease facts and figures. Alzheimers Dement. 2024;20(5):3708-21. Available from: https://doi.org/10.1002/alz.13809
  2. Prince M, Wimo A, Guerchet M, Ali GC, Wu YT, Prina M, et al. World Alzheimer report 2015: the global impact of dementia. London: Alzheimer's Disease International; 2015. Available from: https://www.alzint.org/u/WorldAlzheimerReport2015.pdf
  3. Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R. Physical basis of cognitive alterations in Alzheimer's disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30(4):572-80. Available from: https://doi.org/10.1002/ana.410300410
  4. Selkoe DJ. Alzheimer's disease is a synaptic failure. Science. 2002;298(5594):789-91. Available from: https://doi.org/10.1126/science.1074069
  5. Jack CR Jr, Petersen RC, Xu YC, Waring SC, O'Brien PC, Tangalos EG, et al. Medial temporal atrophy on MRI in normal aging and very mild Alzheimer's disease. Neurology. 1997;49(3):786-94. Available from: https://doi.org/10.1212/wnl.49.3.786
  6. Frisoni GB, Fox NC, Jack CR Jr, Scheltens P, Thompson PM. The clinical use of structural MRI in Alzheimer disease. Nat Rev Neurol. 2010;6(2):67-77. Available from: https://doi.org/10.1038/nrneurol.2009.215
  7. Bartus RT, Dean RL, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982;217(4558):408-14. Available from: https://doi.org/10.1126/science.7046051
  8. Murer MG, Yan Q, Raisman-Vozari R. Brain-derived neurotrophic factor in the control human brain, and in Alzheimer's disease and Parkinson's disease. Prog Neurobiol. 2001;63(1):71-124. Available from: https://doi.org/10.1016/S0301-0082(00)00014-9
  9. Tapia-Arancibia L, Aliaga E, Silhol M, Arancibia S. New insights into brain BDNF function in normal aging and Alzheimer disease. Brain Res Rev. 2008;59(1):201-20. Available from: https://doi.org/10.1016/j.brainresrev.2008.07.007
  10. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353-6. Available from: https://doi.org/10.1126/science.1072994
  11. Buée L, Bussière T, Buée-Scherrer V, Delacourte A, Hof PR. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res Rev. 2000;33(1):95-130. Available from: https://doi.org/10.1016/s0165-0173(00)00019-9
  12. Zatorre RJ, Fields RD, Johansen-Berg H. Plasticity in gray and white matter: neuroimaging changes in brain structure during learning. Nat Neurosci. 2012;15(4):528-36. Available from: https://www.nature.com/articles/nn.3045
  13. May A. Experience-dependent structural plasticity in the adult human brain. Trends Cogn Sci. 2011;15(10):475-82. Available from: https://doi.org/10.1016/j.tics.2011.08.002
  14. Cirillo J, Lavender AP, Ridding MC, Semmler JG. Motor cortex plasticity induced by paired associative stimulation. Neural Plast. 2020;2020:3148425.
  15. Cummings JL, Morstorf T, Zhong K. Alzheimer's disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther. 2014;6(4):37. Available from: https://doi.org/10.1186/alzrt269
  16. Buzsáki G, Draguhn A. Neuronal oscillations in cortical networks. Science. 2004;304(5679):1926-9. Available from: https://doi.org/10.1126/science.1099745
  17. Fries P. Rhythms for cognition: communication through coherence. Neuron. 2015;88(1):220-35. Available from: https://doi.org/10.1016/j.neuron.2015.09.034
  18. Turrigiano GG. The self-tuning neuron: synaptic scaling of excitatory synapses. Cell. 2008;135(3):422-35. Available from: https://doi.org/10.1016/j.cell.2008.10.008
  19. Turrigiano G. Too many cooks? Intrinsic and synaptic homeostatic mechanisms in cortical circuit refinement. Annu Rev Neurosci. 2011;34:89-103. Available from: https://doi.org/10.1146/annurev-neuro-060909-153238
  20. Bi GQ, Poo MM. Synaptic modifications in cultured hippocampal neurons: dependence on spike timing, synaptic strength, and postsynaptic cell type. J Neurosci. 1998;18(24):10464-72. Available from: https://doi.org/10.1523/jneurosci.18-24-10464.1998
  21. Steriade M, Nuñez A, Amzica F. A novel slow (<1 Hz) oscillation of neocortical neurons in vivo: depolarizing and hyperpolarizing components. J Neurosci. 1993;13(8):3252-62. Available from: https://doi.org/10.1523/jneurosci.13-08-03252.1993
  22. Huang EJ, Reichardt LF. Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci. 2001;24:677-736. Available from: https://doi.org/10.1146/annurev.neuro.24.1.677
  23. Eriksson PS, Perfilieva E, Björk-Eriksson T, Alborn AM, Nordborg C, Peterson DA, et al. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4(11):1313-7. Available from: https://doi.org/10.1038/3305
  24. van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD, Gage FH. Functional neurogenesis in the adult hippocampus. Nature. 2002;415(6875):1030-4. Available from: https://doi.org/10.1038/4151030a
  25. Holtmaat A, Svoboda K. Experience-dependent structural synaptic plasticity in the mammalian brain. Nat Rev Neurosci. 2009;10(9):647-58. Available from: https://doi.org/10.1038/nrn2699
  26. Jan YN, Jan LY. Branching out: mechanisms of dendritic arborization. Nat Rev Neurosci. 2010;11(5):316-28. Available from: https://doi.org/10.1038/nrn2836
  27. Fields RD. A new mechanism of nervous system plasticity: activity-dependent myelination. Nat Rev Neurosci. 2015;16(12):756-67. Available from: https://doi.org/10.1038/nrn4023
  28. Halassa MM, Haydon PG. Integrated brain circuits: astrocytic networks modulate neuronal activity and behavior. Annu Rev Physiol. 2010;72:335-55. Available from: https://doi.org/10.1146/annurev-physiol-021909-135843
  29. Palop JJ, Mucke L. Amyloid-beta-induced neuronal dysfunction in Alzheimer's disease: from synapses toward neural networks. Nat Neurosci. 2010;13(7):812-8. Available from: https://doi.org/10.1038/nn.2583
  30. Stam CJ, van Cappellen van Walsum AM, Pijnenburg YA, Berendse HW, de Munck JC, Scheltens P, et al. Generalized synchronization of MEG recordings in Alzheimer's disease: evidence for involvement of the gamma band. J Clin Neurophysiol. 2002;19(6):562-74. Available from: https://doi.org/10.1097/00004691-200212000-00010
  31. Peng S, Wuu J, Mufson EJ, Fahnestock M. Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in pre-clinical Alzheimer's disease. J Neurochem. 2005;93(6):1412-21. Available from: https://doi.org/10.1111/j.1471-4159.2005.03135.x
  32. Frere S, Slutsky I. Alzheimer’s disease: from firing instability to homeostasis network collapse. Neuron. 2018;97(1):32-58. Available from: https://doi.org/10.1016/j.neuron.2017.11.028
  33. Iaccarino HF, Singer AC, Martorell AJ, Rudenko A, Gao F, Gillingham TZ, et al. Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature. 2016;540(7632):230-5. Available from: https://doi.org/10.1038/nature20587
  34. Draganski B, Gaser C, Busch V, Schuierer G, Bogdahn U, May A. Neuroplasticity: changes in grey matter induced by training. Nature. 2004;427(6972):311-2. Available from: https://doi.org/10.1038/427311a
  35. Ermentrout GB, Terman DH. Mathematical foundations of neuroscience. New York: Springer; 2010. Available from: https://link.springer.com/book/10.1007/978-0-387-87708-2
  36. Bartzokis G. Alzheimer’s disease as homeostatic responses to age-related myelin breakdown. Neurobiol Aging. 2011;32(8):1341-71. Available from: https://doi.org/10.1016/j.neurobiolaging.2009.08.007
  37. Shankar GM, Li S, Mehta TH, Garcia-Muñoz A, Shepardson NE, Smith I, et al. Amyloid-β protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837-42. Available from: https://www.nature.com/articles/nm1782
  38. Arendt T. Synaptic degeneration in Alzheimer’s disease. Acta Neuropathol. 2009;118(1):167-79. Available from: https://doi.org/10.1007/s00401-009-0536-x
  39. Basser PJ, Mattiello J, LeBihan D. MR diffusion tensor spectroscopy and imaging. Biophys J. 1994;66(1):259-67. Available from: https://doi.org/10.1016/s0006-3495(94)80775-1
  40. Minichiello L. TrkB signalling pathways in LTP and learning. Nat Rev Neurosci. 2009;10(12):850-60. Available from: https://doi.org/10.1038/nrn2738
  41. Bramham CR, Messaoudi E. BDNF function in adult synaptic plasticity: the synaptic consolidation hypothesis. Prog Neurobiol. 2005;76(2):99-125. Available from: https://doi.org/10.1016/j.pneurobio.2005.06.003
  42. Cohen-Cory S, Kidane AH, Shirkey NJ, Marshak S. Brain-derived neurotrophic factor and the development of structural neuronal connectivity. Dev Neurobiol. 2010;70(5):271-88. Available from: https://doi.org/10.1002/dneu.20774
  43. Lindholm D, Castrén E, Kiefer R, Zafra F, Thoenen H. Transforming growth factor-β1 in the rat brain: increase after injury and inhibition of astrocyte proliferation. J Cell Biol. 1992;117(2):395-400. Available from: https://doi.org/10.1083/jcb.117.2.395
  44. Hefti F. Nerve growth factor promotes survival of septal cholinergic neurons after fimbrial transections. J Neurosci. 1986;6(8):2155-62. Available from: https://doi.org/10.1523/jneurosci.06-08-02155.1986
  45. Tuszynski MH, Thal L, Pay M, Salmon DP, U HS, Bakay R, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med. 2005;11(5):551-5. Available from: https://doi.org/10.1038/nm1239
  46. Trejo JL, Carro E, Torres-Aleman I. Circulating insulin-like growth factor I mediates exercise-induced increases in new neurons in the adult hippocampus. J Neurosci. 2001;21(5):1628-34. Available from: https://doi.org/10.1523/jneurosci.21-05-01628.2001
  47. Carro E, Trejo JL, Gomez-Isla T, LeRoith D, Torres-Aleman I. Serum insulin-like growth factor I regulates brain amyloid-β levels. Nat Med. 2002;8(12):1390-7. Available from: https://cris.technion.ac.il/en/publications/serum-insulin-like-growth-factor-i-regulates-brain-amyloid-%CE%B2-leve
  48. Freude S, Schilbach K, Schubert M. The role of IGF-1 receptor and insulin receptor signaling for the pathogenesis of Alzheimer’s disease. Biochim Biophys Acta. 2009;1790(10):963-9. Available from: https://doi.org/10.2174/156720509788486527
  49. Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333(6048):1456-8. Available from: https://doi.org/10.1126/science.1202529
  50. Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131(6):1164-78. Available from: https://doi.org/10.1016/j.cell.2007.10.036

Advertisement

 

Help ?