Dynamic Homeostasis
State: A highly regulated state where neuronal, glial, vascular, and metabolic systems cooperate to maintain cognitive performance, structural resilience, and proteostatic integrity.
Key Cellular Systems: Neurons, Astrocytes, Microglia, Oligodendrocytes, Endothelium
The healthy brain operates in a finely tuned, low-entropy state wherein neuronal, glial, vascular, and metabolic systems function in a concerted manner to preserve cognitive performance, structural resilience, and proteostatic integrity. This equilibrium is not passive but dynamically sustained through activity-dependent modulation, energy-efficient signaling, immunological restraint, and the continual clearance of molecular noise. Core to this system are five cellular subsystems—neurons, astrocytes, microglia, oligodendrocytes, and endothelial cells—which together maintain the brain’s baseline operational precision.
1.Neurons: Plasticity, Energy, and Signal Precision
Neuronal homeostasis is fundamentally rooted in the balance between long-term potentiation (LTP) and long-term depression (LTD). During LTP, elevated glutamate release drives strong AMPA receptor activation, unblocking NMDA receptors and allowing a large, rapid influx of Ca²⁺. This triggers calmodulin-dependent activation of CaMKII, leading to downstream activation of PKA, PKC, and ERK/MAPK pathways, culminating in CREB-mediated transcription of plasticity-associated genes such as BDNF, Arc, Egr1, and Homer1a. These genes collectively support synapse stabilization, dendritic growth, and cytoskeletal reinforcement, resulting in increased excitatory postsynaptic current (EPSC) amplitude and enduring synaptic strengthening.
Conversely, LTD arises from asynchronous or low-intensity glutamate signaling, producing weaker AMPA activation and limited NMDA/mGluR1/5 engagement. The resulting moderate, diffuse Ca²⁺ influx activates calcineurin and PP1, leading to AMPAR dephosphorylation and internalization. Structural changes include spine shrinkage and actin depolymerization, facilitating synaptic weakening and pruning.
Synaptic integrity is maintained by PSD-95 scaffolding proteins and the phosphorylation-dependent trafficking of AMPARs (GluA1/GluA2). Local protein synthesis, regulated via mTORC1 and 4E-BP1/S6K1 signaling, allows spatially precise translation of FMRP-associated transcripts at dendritic sites. The ubiquitin–proteasome system (UPS) concurrently eliminates misfolded proteins, enabling synaptic remodeling with minimal proteostatic noise.
Mitochondrial positioning—mediated by Miro1 and syntaphilin anchoring—ensures that ATP and Ca²⁺ buffering are locally available at active synapses. Redox homeostasis is enforced by glutathione systems and Nrf2-dependent expression of antioxidant enzymes (e.g., HO-1, SOD2), maintaining a high GSH/GSSG ratio essential for low oxidative stress.
2.Astrocytes: Synaptic Maintenance and Interstitial Homeostasis
Homeostatic astrocytes (A2 phenotype) play a critical role in neurotransmitter regulation, ion buffering, and metabolic support. EAAT2 (GLT-1), encoded by SLC1A2, ensures rapid glutamate uptake from the synaptic cleft, preventing excitotoxic accumulation. This transporter operates via Na⁺ symport and requires ATP input through Na⁺/K⁺-ATPase activity.
Astrocytes also regulate extracellular potassium via Kir4.1 channels, Na⁺/K⁺-ATPase (α2 isoform), and spatial K⁺ redistribution through gap junction-coupled syncytia (Cx43/Cx30). This buffering resets neuronal resting potential after action potentials, preserving excitability thresholds.
Trophic support is conferred through BDNF, GDNF, and VEGF secretion, which respectively support dendritic spine integrity, motor neuron function, and neurovascular unit health. Astrocytes also scaffold the glymphatic system, where perivascular expression of AQP4 on endfeet enables CSF–ISF exchange. This system facilitates the clearance of β-amyloid, tau oligomers, and oxidized metabolites, especially during sleep.
3.Microglia: Surveillance, Clearance, and Immunological Silence
Homeostatic microglia express markers such as P2RY12, TMEM119, and CX3CR1, maintaining a surveillant but non-inflammatory phenotype. P2RY12 enables ATP/ADP-guided process extension toward active or injured synapses. TREM2, in conjunction with DAP12–Syk–PI3K signaling, promotes lipid sensing, cytoskeletal reorganization, and non-inflammatory phagocytosis of apoptotic debris, oxidized lipids, and damaged synapses.
Microglia regulate synaptic populations through complement-mediated pruning: neurons deposit C1q on underutilized synapses, leading to C3b tagging. Microglial CR3 receptors (CD11b/CD18) recognize these tags, triggering synaptic engulfment. Importantly, this occurs without triggering inflammasome activity or NF-κB signaling, thus preserving tissue homeostasis.
Energetically, microglia rely on oxidative phosphorylation (OXPHOS), maintaining high mitochondrial potential and low cytokine output. Their trophic secretome—comprising IGF-1 and TGF-β—reinforces synaptic health and vascular integrity.
4.Oligodendrocytes: Axonal Conduction and Metabolic Support
Oligodendrocytes synthesize and maintain CNS myelin via expression of MBP, PLP, and MOG, enabling saltatory conduction and efficient long-range signaling. High-speed action potential propagation across nodes of Ranvier underlies cortical synchrony and cognitive processing speed.
Beyond insulation, oligodendrocytes metabolize glucose into lactate and pyruvate, shuttling these to axons via MCT1 (SLC16A1). Axons uptake lactate through MCT2, converting it into ATP via the TCA cycle. This MCT1–MCT2 shuttle sustains energy homeostasis in long-projecting or high-frequency neurons, particularly during metabolic stress.
Oligodenrocytes also store iron within ferritin and regulate its availability for OXPHOS and lipid synthesis. Through neurotransmitter (glutamate, ATP) sensing and crosstalk with astrocytes and microglia, they modulate local metabolic and immune dynamics, especially during repair.
5.Endothelial Cells and Pericytes: Vascular Homeostasis and Immune Containment
Endothelial cells and pericytes maintain blood–brain barrier (BBB) selectivity, restrict peripheral immune infiltration, and regulate neurovascular coupling. These cells, via CX3CR1 and TREM2, also contribute to debris clearance and synaptic sculpting under homeostatic conditions. Tight junctions and receptor-mediated transport ensure controlled nutrient exchange and cytokine exclusion.
Proteostasis and Clearance Mechanisms
Amyloid precursor protein (APP), largely expressed in excitatory neurons, undergoes non-pathological cleavage by α-secretase under basal conditions. However, a small portion is processed via BACE1 and γ-secretase into Aβ peptides. These are efficiently cleared by neprilysin (NEP) and insulin-degrading enzyme (IDE), or through glymphatic flow mediated by AQP4 channels. TREM2⁺ microglia further phagocytose modified Aβ in a restrained, anti-inflammatory manner. CD33 acts as a negative regulator of this clearance, and its dysregulation is associated with Alzheimer’s risk.
Energetic and Network Stability
Neural efficiency is maintained through a high ΔG of ATP hydrolysis (~ –58 kJ/mol), optimal NAD⁺/NADH ratios, and robust mitochondrial fusion–fission cycling. AMPK remains suppressed, allowing mTORC1 to regulate synaptic protein translation without triggering autophagy. Redox balance is maintained by low ROS production and high GSH/GSSG ratios.
Temporal stability is coordinated by circadian genes (CLOCK, BMAL1), which govern microglial tone, APP expression, and Aβ clearance. Slow-wave sleep (SWS) enhances glymphatic flow and BDNF-driven plasticity, while hippocampal–PFC synchrony underlies cognitive coherence. Energetic stability is sustained through local mitochondrial positioning and astrocytic redox buffering.
Subclinical Markers of Decline
Although function is preserved, early symptoms may emerge, including:
- Subtle memory lapses
- Decreased attention span
- Fatigue and reduced cognitive stamina
- Mild sleep disruption
- Subclinical olfactory deficits
- Occasional mood changes (irritability, apathy)
Biomarkers remain within normal ranges:
- CSF Aβ42: Low accumulation
- pTau181 / Total Tau: Normal ratios
- TREM2: Reflects balanced microglial function
- Neurofilament Light (sNfL): Minimal axonal stress
- BDNF: Robust CNS and serum levels
Therapeutic Focus: Sustaining a Low-Entropy State
Interventions aim to preserve glial balance, energetic resilience, and immune silence:
- Aerobic exercise: ↑ BDNF, ↑ PGC-1α–NRF1, ↑ glymphatic flow
- Slow-wave sleep: Enhances ISF clearance, ↓ IL-6/TNF-α
- Fasting: ↑ NAD⁺ → activates SIRT1, ↑ AMPK, ↓ mTOR
- Omega-3 fatty acids: Stabilize synaptic membranes, ↓ NF-κB signaling
- Stress reduction: ↓ HPA axis overactivation, ↓ neuroinflammatory crosstalk
Dynamic homeostasis represents a thermodynamically and informationally efficient CNS state—characterized by proteostatic equilibrium, neuroimmune restraint, and adaptive plasticity. Its breakdown marks the onset of entropy-driven disruption and the slow emergence of maladaptive responses.
Disruption
State: Disruption represents the progressive erosion of dynamic homeostasis, driven by a rise in local entropy—the loss of thermodynamic stability, informational precision, and spatial specificity within the CNS. This transition does not manifest as overt pathology at first. Rather, it introduces stochastic variability, signal-processing inefficiency, and structural drift, degrading the system’s capacity for precise regulation. Over time, these failures accumulate into a positive feedback loop, ultimately precipitating maladaptive immune activation and compensatory remodeling.
Pathophysiology:
1.Synaptic Instability Driven by Local Entropy
The initial increase in local entropy begins subtly—with incomplete glutamate clearance by astrocytes due to early mislocalization or partial downregulation of EAAT2 (GLT-1). This causes a modest rise in extracellular glutamate, especially at perisynaptic sites. Although initially buffered, this small elevation begins to erode the temporal and spatial fidelity of excitatory transmission. As astrocytes work harder to re-establish homeostasis—upregulating secondary glutamate transporters and enzymatic degradation pathways—the metabolic demand rises, resulting in increased ATP hydrolysis. Paradoxically, this compensatory ATP consumption generates additional local entropy, producing heat, ROS, and diffusible byproducts that further destabilize the microenvironment.
Over time, EAAT2 function declines more significantly due to both transcriptional suppression and redox-mediated post-translational modifications, exacerbating glutamate accumulation. The rise in entropy alters astrocytic cytoskeletal architecture, destabilizing anchoring proteins such as α-syntrophin, which tethers AQP4 to perivascular domains. This marks the beginning of widespread spatial disintegration.
Meanwhile, neuronal EAAT3 becomes suppressed by:
- Loss of signal fidelity: Glutamate gradients are too diffuse to trigger membrane recruitment.
- Oxidative stress: ROS modifies EAAT3 structure.
- Energetic insufficiency: Local ATP depletion favors transporter internalization.
- Transcriptional collapse: Nrf2 signaling declines.
Synaptic entropy further manifests as impaired theta–gamma phase coupling, disrupting spike-timing-dependent plasticity. PSD-95 scaffolding fragments, AMPAR/NMDAR anchoring becomes erratic, and dendritic spines exhibit morphological instability. Thus, the synapse—once a site of computational precision—becomes vulnerable to stochastic noise, marking the system’s inflection from signal integration to disintegration.
2.Astrocytic Dysregulation and Spatial Entropy
One of the earliest and most consequential disruptions in glial organization is the loss of astrocytic polarity, especially in the localization of Aquaporin-4 (AQP4). Under homeostatic conditions, AQP4 channels are confined to perivascular endfeet, where they maintain vectorial glymphatic flow. However, as local entropy increases—driven by oxidative stress, ATP depletion, and cytoskeletal fragmentation—AQP4 is redistributed across astrocytic membranes. This impairs directional CSF–ISF exchange and compromises glymphatic clearance, particularly during slow-wave sleep.
The failure to clear metabolic solutes such as Aβ, tau, and oxidized lipids increases extracellular entropy, impairing biochemical compartmentalization and glial–neuronal support. Concurrently, EAAT2 redistribution leads to glutamate spillover, escalating excitotoxicity and glutamate–glutamine cycle disruption. As astrocytes attempt to compensate, they deplete ATP, increasing oxidative burden and entropy generation.
Potassium buffering collapses as Kir4.1 channels depolarize:
- Normally, Kir4.1 relies on a negative membrane potential to drive inward K⁺ current.
- Persistent depolarization (from Na⁺/K⁺ ATPase inefficiency and ionic leak) reduces this driving force.
- Mislocalization and downregulation of Kir4.1 hinder astrocytic K⁺ siphoning.
The result is extracellular K⁺ accumulation, raising neuronal excitability baseline and contributing to circuit noise. Astrocytic transcriptional drift begins: genes for BDNF, GDNF, and VEGF decline, blunting neurovascular support and LTP facilitation. These processes culminate in a chaotic intermediate astroglial phenotype, marked by partial reactivity, poor metabolic integration, and a collapse of spatial order.
3.Microglial Signal Degradation and Phagocytic Drift
Microglia begin to detect entropy as signal. P2RY12 expression drops, reducing chemotactic precision toward ATP/ADP gradients. TREM2–DAP12 signaling weakens, impairing recognition of apoptotic debris and lipid signals. While classical inflammatory activation is not yet present, functional surveillance decays. Complement-tagged synapses are still recognized, but synaptic pruning becomes promiscuous—selecting for less-active synapses irrespective of pathological status.
Persistent exposure to oxidative metabolites and membrane debris initiates a drift toward phagocytic hyperactivity. Synaptic elements, oxidized lipids, and misfolded proteins accumulate, further increasing local entropy and forcing microglia into a primed but unresolved state.
4.Mitochondrial Instability and Redox Drift
As energy demand outpaces supply, neuronal mitochondria fail to maintain stable anchoring at active dendritic spines due to breakdown of Miro1–Syntaphilin coordination. Membrane potential (Δψm) becomes unstable, reducing local ATP production and Ca²⁺ buffering capacity.
NAD⁺/NADH ratios decline, reducing SIRT1 activation and mitochondrial renewal. OXPHOS becomes inefficient, ROS production rises, and redox homeostasis collapses. Astrocytic GSH recycling falters, and Nrf2-driven antioxidant transcription diminishes. The system enters a phase of redox entropy, where random molecular damage becomes frequent, uncontained, and self-reinforcing.
5.Proteostatic Decline and Amyloidogenic Shift
Entropy in the protein landscape increases. The ubiquitin–proteasome system (UPS) becomes saturated, allowing misfolded proteins—including Aβ and hyperphosphorylated tau—to accumulate. Neprilysin and IDE activity are impaired by redox and metabolic stress.
APP cleavage shifts toward amyloidogenic processing, with increased Aβ₁₋₄₂ production. Clearance fails to match synthesis, marking the earliest definable biochemical divergence toward Alzheimer’s pathology—initiated by a sustained loss of proteostatic entropy barriers.
6.Circadian Decoupling and Sleep-Dependent Clearance Failure
Circadian synchrony disintegrates. CLOCK, BMAL1, and PER gene oscillations desynchronize across neuronal and glial compartments, weakening control over glymphatic flow, BDNF pulses, and redox rhythms.
Slow-wave sleep diminishes, impairing AQP4-driven clearance of Aβ and tau. Melatonin signaling is blunted, while orexinergic tone becomes erratic—fragmenting sleep architecture and compounding metabolic burden. The nocturnal entropy clearance cycle collapses, undermining the final systemic safeguard against pathological accumulation.
Emerging Symptoms:
As the entropic breakdown progresses from synaptic instability to glial disintegration and proteostatic drift, the central nervous system begins to exhibit subtle but consistent phenotypic signatures—functional impairments that reflect system-level compensation and local instability. These symptoms typically remain subclinical or are dismissed as stress-related or age-normal variations, yet they reflect the biophysical signatures of mounting entropy, functional asynchrony, and loss of homeostatic reserve.
1.Cognitive Microdecline
- Subtle episodic memory lapses emerge first—especially in tasks requiring rapid hippocampal–prefrontal switching (e.g., remembering spatial or verbal details after distraction).
- Attentional fragility increases, with impaired focus maintenance under conditions of multitasking or sensory distraction.
- These cognitive fluctuations correspond with theta–gamma phase decoupling and early synaptic noise, particularly in the medial temporal lobe and default mode network.
2.Mental Fatigue and Cognitive Deceleration
- Individuals experience prolonged recovery times after cognitive exertion and reduced capacity for sustained mental engagement.
- Tasks requiring effortful control (e.g., working memory, planning, inhibition) show inconsistent performance, reflecting entropic degradation in synaptic metabolic support and cortical-striatal communication.
- This cognitive deceleration is strongly correlated with mitochondrial redox drift and impaired astrocyte–neuron metabolic coupling.
3.Sleep Fragmentation
- Despite normative sleep duration, individuals report non-restorative sleep, increased nocturnal awakenings, and early morning fatigue.
- These disruptions mirror glymphatic flow inefficiency due to AQP4 mislocalization, as well as reduced amplitude of slow-wave oscillations.
- Sleep-dependent memory consolidation and emotional regulation begin to degrade, amplifying fatigue and irritability.
4.Mood Variability and Subclinical Affective Instability
- Mild anxiety, irritability, or anhedonia may occur in episodic or context-sensitive patterns—not yet meeting diagnostic thresholds but reflecting circuit noise in limbic-prefrontal regulation.
- This mood lability is partially driven by neuroinflammatory priming, reduced BDNF tone, and early hypothalamic stress-axis dysregulation.
5.Olfactory and Sensory Discriminiation Loss
- Subclinical hyposmia (loss of smell acuity) often precedes cognitive symptoms.
- Olfactory bulb neurons are particularly sensitive to oxidative microenvironmental shifts and early protein aggregation, making them reliable entropy reporters.
- Individuals may also report mild sensory hypersensitivities or perceptual “glitches”, such as delayed auditory–visual integration under cognitive load.
6.Motoric Decline and Coordination Variance
- Fine motor control may show small fluctuations in speed, precision, or fluidity—especially during complex, multitasked movement.
- These changes correspond to entropy-driven disruption in basal ganglia–cortical feedback, as astrocytic K⁺ buffering and synaptic pruning fidelity decline.
- Individuals may feel “clumsy” or slightly slower in movements that previously felt automatic.
7.Systemic Fatigue and Thermodynamic Drift
- Individuals may report a general decline in physical stamina, not explained by cardiovascular or musculoskeletal changes.
- This reflects energetic inefficiency at the cellular level, where ATP yield per glucose molecule is reduced due to OXPHOS uncoupling, ROS leakage, and mitochondrial density loss.
- A decline in NAD⁺ availability and sirtuin activity contributes to accelerated perceived fatigue, even in the absence of measurable inflammation.
Early Biomarker Drift
Even when symptoms remain subjectively minimal, measurable shifts may include:
- ↓ CSF Aβ₄₂ (due to sequestration)
- ↑ total tau or pTau181 (reflecting early proteostatic disturbance)
- ↓ serum BDNF
- ↑ neurofilament light (sNfL), indicating axonal stress
- Subtle upregulation of TREM2, C1q, or IL-6 in CSF or exosomes
These symptom patterns reflect the entropy threshold being crossed from regulation to dysregulation, where compensation is still possible but becoming metabolically costly. They do not yet reflect fixed pathology—but indicate that the system is operating in a subcritical maladaptive mode, poised on the cusp of immunological and neurodegenerative engagement.
Therapeutic Goals:
- Restore Astrocytic Polarity & EAAT2 Function
- Reinstate targeted glutamate clearance to prevent excitotoxic stress and secondary entropy amplification.
- Enhance Glymphatic Clearance Efficiency
- Re-polarize AQP4 to endfeet to improve directional CSF–ISF exchange during sleep.
- Reduce Mitochondrial ROS and ATP Drain
- Optimize neuronal and astrocytic metabolism to stabilize membrane potentials and reduce post-translational damage.
- Reinforce Synaptic Plasticity Thresholds
- Normalize NMDA/AMPA ratio and PSD-95 anchoring to preserve synaptic integration fidelity.
- Maintain Microglial Homeostatic Surveillance
- Inhibit aberrant complement signaling and preserve TREM2/P2RY12 signaling fidelity.
- Prevent Circadian and Sleep Decoupling
- Protect CLOCK/BMAL1 rhythmicity to sustain glymphatic surge synchronization.
Clinical Application:
1. EAAT2 (GLT-1) Stabilization and Upregulation
- Goal: Enhance glutamate clearance, reduce excitotoxic entropy.
- Strategies:
- Ceftriaxone: Promotes EAAT2 transcription and membrane localization.
- Gene therapy (e.g., AAV vectors) targeting EAAT2 delivery to astrocytes.
- Nrf2 activators (e.g., sulforaphane): counteract redox suppression of EAAT2 expression.
- Reference:
- Calabrese, V., et al. (2020). Cellular stress responses, the hormesis paradigm, and vitagenes: novel targets for therapeutic intervention. Antioxidants & Redox Signaling. PDF
2. AQP4 Polarity Restoration and Glymphatic Flow Enhancement
- Goal: Reestablish astrocytic polarity to rescue CSF–ISF clearance.
- Strategies:
- Angiotensin receptor blockers (e.g., losartan): may enhance AQP4 endfoot localization.
- Exercise and sleep therapies: shown to restore AQP4 perivascular polarization.
- Melatonin supplementation: reinforces circadian glymphatic function and AQP4 targeting.
- Reference:
- Tagliabue, S., et al. (2023). Microvascular cerebral blood flow dynamics and glymphatic system integrity. Journal of Neuroscience Research. PDF
3. Mitochondrial Bioenergetic Enhancement
- Goal: Reduce redox entropy and stabilize ATP supply at synapses.
- Strategies:
- NAD⁺ boosters (e.g., nicotinamide riboside, NR): improve mitochondrial coupling and sirtuin activation.
- MitoQ and SS-31 peptides: mitochondria-targeted antioxidants reducing ROS-mediated drift.
- Ketone supplementation (e.g., medium-chain triglycerides): provide alternative fuel source in low-glucose metabolic states.
- Reference:
- Cunnane, S.C., et al. (2020). Brain energy rescue: an emerging therapeutic concept. Nature Reviews Drug Discovery. PDF
4. Synaptic Entropy Modulation
- Goal: Prevent phase de-coupling and STDP collapse.
- Strategies:
- Neurosteroids (e.g., allopregnanolone): enhance GABAergic tone and stabilize network oscillations.
- Closed-loop neurostimulation (e.g., theta burst TMS): restore phase-amplitude coupling.
- BDNF mimetics or enhancers: promote structural spine stabilization.
- Reference:
- Ahlquist, R. (2023). Synaptic entropy and PI3K modulation in early neurodegeneration. Annals of Neurosciences. PDF
5. Proteostasis Reinforcement and Amyloidogenic Delay
- Goal: Buffer early aggregation-prone protein flux and maintain degradation capacity.
- Strategies:
- Rapamycin analogues: moderate mTOR, enhance autophagy.
- Heat shock protein (HSP90/HSP70) modulators: enhance chaperone activity.
- Immunotherapies (e.g., lecanemab): target early soluble Aβ species to prevent entropy amplification.
6. Circadian and Sleep Architecture Repair
- Goal: Restore glymphatic cycling and time-encoded metabolic clearance.
- Strategies:
- Melatonin receptor agonists (e.g., ramelteon): enhance SWS and circadian gene coherence.
- Orexin antagonists (e.g., suvorexant): suppress arousals, improve clearance windows.
- Light therapy and timed feeding: entrain BMAL1/CLOCK cycles across brain regions.
- Reference:
- Błaszczyk, J.W. (2020). Energy metabolism decline in the aging brain. Metabolites. Full Text
7. Integrated Early Detection and Bioenergetic Profiling
- Emerging tools:
- CSF/exosomal biomarkers: early pTau181, neurogranin, EAAT2, sNfL.
- FDG-PET and MR spectroscopy: assess regional hypometabolism and entropy hotspots.
- Machine learning classifiers based on multimodal entropy metrics (e.g., sleep EEG variance, HRV complexity).
Reaction
State:
The Reaction phase reflects the system’s escalating attempt to counter local entropy through active immune engagement, catabolic reprogramming, and remodeling of tissue architecture. Whereas the Disruption phase is defined by rising stochastic noise and functional breakdown, Reaction is the first nonlinear response—an immunometabolic inflection where the CNS shifts from containment to clearance. This stage still preserves the possibility of partial reversal, but the energy cost and inflammatory load begin to reshape baseline function.
Key Cellular Events:
- Microglia: M1 → release IL-1β, TNF-α, ROS (reaction)
- Astrocytes: A1 -> upregulate GFAP, VCAM-1, C3 (reaction)
- Oligodendrocytes: cytokine overburst (disruption) -> react to immune signals with metabolic stress, impaired myelin maintenance
Pathophysiology:
1.Astroglial Transition to Reactive States (A1/A2 Heterogeneity)
Prolonged glutamate accumulation, oxidative stress, and extracellular ATP drive astrocytes into reactive phenotypes, marked by:
- Upregulation of GFAP, vimentin, and Serpina3n
- Suppression of neurotrophic genes (BDNF, GDNF)
- Partial STAT3/NF-κB activation in response to IL-1β, TNF-α
- Loss of aquaporin-4 endfoot polarization, further impairing glymphatic clearance
- Metabolic switch toward glycolysis, reducing astrocytic support to neurons
Astrocytes no longer compartmentalize signals effectively and begin to secrete proinflammatory cytokines and complement proteins (e.g., C3), contributing to synapse tagging for microglial pruning.
2.Microglial Activation and Synaptic Culling
Microglia, sensing unresolved entropy and persistent DAMPs (e.g., oxidized lipids, ATP, misfolded proteins), shift into an activated yet dysregulated state:
- Upregulate MHC-II, CD68, iNOS, and NLRP3 inflammasome components
- Secrete IL-1β, IL-6, TNF-α, driving neuroinflammatory signaling
- Over-prune synapses via complement-mediated mechanisms (C1q → C3b → CR3)
- Lose discriminative pruning, targeting less active synapses rather than strictly pathological ones
Reactive microglia cluster around metabolically unstable areas (e.g., Aβ plaques, redox hotspots), releasing ROS and NO, which exacerbate neuronal damage and promote feedback-driven glial activation.
3.Mitochondrial Collapse and Metabolic Reprogramming
As energy demands surge, neurons and glia enter metabolic crisis:
- Neurons activate AMPK, suppressing mTORC1 and halting plasticity-related protein synthesis
- Astrocytes shift to aerobic glycolysis, producing more lactate but fewer TCA intermediates
- Mitochondrial fragmentation (via Drp1) increases, impairing ATP production and calcium buffering
- NAD⁺ levels drop, further reducing SIRT1 activity, autophagy, and mitochondrial renewal
This metabolic reprogramming favors short-term survival, but promotes long-term dysfunction and proteostatic collapse.
4.Innate Immune Signaling Becomes Maladaptive
Toll-like receptors (e.g., TLR4), inflammasome components (e.g., NLRP3), and danger-sensing pathways are chronically activated:
- Sustained NF-κB transcription leads to cytokine spread and astroglial–microglial crosstalk
- HMGB1, S100 proteins, and heat-shock proteins act as endogenous DAMPs
- Interferon response genes become upregulated, especially in aged or APOE4 carriers
These changes signal the breakdown of immunological silence in the CNS, replacing homeostatic modulation with persistent low-grade inflammation.
5.Proteostasis Collapse and Intracellular Aggregation
With UPS saturation and impaired autophagy:
- Tau becomes hyperphosphorylated, forming cytosolic aggregates and disrupting axonal transport
- Aβ accumulates intra- and extracellularly, seeding plaque formation
- Chaperone activity (HSP70, HSP90) becomes insufficient to prevent unfolding or refolding errors
- p62/SQSTM1-positive inclusions accumulate in glia and neurons
6.Blood-Brain Barrier (BBB) Disruption and Peripheral Immune Crosstalk
Chronic inflammatory signals and oxidative stress degrade BBB integrity:
- Tight junction proteins (claudin-5, occludin) downregulate
- Pericytes detach, exposing basement membranes
- Monocytes and T cells infiltrate, releasing IFN-γ and TNF-α
- Fibrinogen and albumin extravasation into parenchyma triggers further glial activation
BBB breakdown permits the entrance of systemic entropy, and the CNS transitions into an open system—subject to peripheral immune noise.
Symptoms:
As entropic compensation becomes immunologically and metabolically active, the following clinical signs and symptoms emerge:
- Cognitive and Affective Symptoms
- Accelerating memory deficits (short-term + retrieval failures)
- Word-finding difficulty (lexical retrieval slowing)
- Attention switching errors
- Emotional flattening, irritability, anxiety bursts
- Sleep and Circadian Dysfunction
- Insomnia or hypersomnia cycles
- Decreased SWS and REM proportion
- Blunted melatonin–cortisol rhythms
- Neurological Findings
- Mild parkinsonian signs (bradykinesia, rigidity)
- Dysexecutive syndrome (frontal lobe processing)
- Spatial disorientation (parietal degradation)
- Emerging Biomarker Changes
- ↑ CSF pTau181 and Aβ42/40 ratio
- ↑ sTREM2 and YKL-40
- ↑ IL-6, IL-1β, and TNF-α in CSF
- ↑ sNfL (axonal injury marker)
- PET: regional hypometabolism + Aβ tracer uptake
Therapeutic Goal: Suppress the neuroimmune cascade, contain glial overactivation, and restore CNS filtering thresholds before irreversible damage or structural demyelination begins.
Clinical Application:
Target | Therapeutic Goal | Example Therapies |
---|---|---|
Glial Reactivity | Suppress A1 astrocytes, modulate microglial overactivation | Minocycline, IL-1β blockers, TREM2 modulators |
Mitochondrial Stress | Reinforce ATP synthesis, block fragmentation | SS-31, CoQ10, NAD⁺ boosters |
Proteostasis | Boost autophagy, reduce aggregation | Rapalogs, HSP co-inducers, GSK3β inhibitors |
Complement-mediated Pruning | Inhibit C1q/C3 overactivation | Anti-C1q antibodies, CR3 blockers |
Inflammation/BBB Integrity | Restore vascular tight junctions | Angiotensin receptor blockers, TGF-β agonists |
Circadian Repair | Normalize CLOCK/BMAL1–based rhythms | Ramelteon, light therapy, timed feeding |
Reaction is a phase of acute, unsustainable containment — biologically expensive, temporarily effective, and potentially damaging if not resolved. It reflects the CNS’s attempt to maintain order under conditions of escalating internal entropy and immune infiltration.
Adaptation
State: Adaptation reflects the brain’s attempt to conserve energetic and computational resources in the face of escalating local entropy. As precision regulation falters during Disruption, the system strategically withdraws from high-cost processes—prioritizing essential functions, consolidating circuitry, and suppressing unstable signaling. This is not merely a degenerative decline but an active, quasi-homeostatic shift that redefines the parameters of cognitive operation. While temporarily protective, it lays the groundwork for chronic insufficiency and pathologic rigidity.
Pathophysiology:
1.Network Simplification and Redundancy Elimination
Cortical and subcortical networks begin to prune higher-order connections, especially in associative and integrative hubs. This simplification is an attempt to stabilize neural signaling by reducing entropy propagation through inefficient pathways. Hubs with high betweenness centrality (e.g., posterior cingulate, medial prefrontal cortex) show diminished connectivity, while primary sensorimotor circuits are relatively preserved.
Kumar et al., 2025 demonstrated cortical simplification and hub erosion in early AD using AI-based network modeling, suggesting this is an adaptive simplification mechanism.
Link
2.Energetic Downscaling and Circuit Prioritization
Energetic resources are reallocated toward low-cost neural operations. Glucose hypometabolism in the DMN is paralleled by increased relative activity in visual and somatomotor regions—regions that require less integration and synaptic modulation. This reflects a systemic shift from adaptive plasticity to fixed signaling loops.
Khan et al., 2024 found that AD brains favor metabolically conservative subnetworks to mitigate entropy and ATP debt.
PDF
3.Synaptic Subtraction and Information Streamlining
Selective synapse loss disproportionately affects low-salience or unstable synaptic contacts, mimicking signal filtering. This “pruning” resembles a form of adaptive compression, reducing the number of parallel processing units to simplify integration demands and stabilize circuit output.
Liu & Wu, 2025 revealed how tau propagation preferentially targets less integrated synapses, aligning with an entropy-minimizing substrate selection.
Link
4.Resource Reallocation Toward Surveillance and Repair
Astrocytic and endothelial metabolic programs shift from synaptic support to focal damage containment. Regional heterogeneity in astrocytic metabolism collapses, and trophic signaling (e.g., VEGF, BDNF) becomes spatially restricted to areas of active stress.
Dan et al., 2025 illustrated how network-wide modeling of tau spread corresponds with glial resource redistribution toward degenerative hotspots.
IEEE Link
5.Microglial Rechanneling and Signal Prioritization
Microglia abandon full-scale surveillance, rechanneling toward synaptic clearance and debris management. While classical inflammatory activation remains subthreshold, phagocytic behavior becomes more selective, guided by localized signal entropy and complement tagging.
Krishnamoorthy et al., 2024 showed early phagocytic redirection using live imaging and microglial functional signatures in preclinical AD.
Access IEEE
6.Excitatory–Inhibitory Rebalancing via Network Sparsification
The excitatory–inhibitory balance shifts as cortical inhibition is reweighted to suppress unstable excitatory loops. GABAergic interneurons increase tonic suppression, while pyramidal networks simplify through reduced dendritic branching and synaptic input density. This supports transient stability but limits computational flexibility.
Hu et al., 2024 demonstrated cortical synchrony and phase locking rise in early AD at the cost of neural complexity and inhibitory plasticity.
IEEE Link
The adaptation phase correlates with early-to-mid clinical Alzheimer’s and other neurodegenerative syndromes characterized by cognitive narrowing and slowed recovery.
Symptoms:
- Cognitive and Functional Features
- Decline in executive functions, episodic memory, and abstract reasoning
- Speech becomes anomic, with paraphasias or circumlocution
- Flattening of emotional range and social withdrawal
- Autonomic and Behavioral Features
- Fragmented sleep–wake cycle with reduced REM and N3
- Diminished hunger and thirst cues
- Apathy and psychomotor slowing
Biomarker Profile
- ↑ CSF pTau181 and total tau
- ↑ Aβ plaque load on PET, plateauing in levels
- ↓ FDG-PET uptake in temporoparietal and posterior cingulate regions
- ↑ astrocyte marker YKL-40, ↓ CSF BDNF
Therapeutic Goal: Support neuroregeneration, restore metabolic resilience, and reverse maladaptive glial phenotypes before long-term consolidation of dysfunction occurs.
Clinical Application:
Restore Plasticity:
Goal: Resynchronize neuronal oscillations and reopen phase-locked signal propagation, degraded by circuit rigidity and entropy-driven decoupling.
- Epigenetic Modulators:
- HDAC inhibitors (e.g., vorinostat, sodium butyrate) relieve transcriptional repression by enhancing histone acetylation at genes supporting synaptic remodeling, such as BDNF, Arc, and Homer1a.
- This epigenetic loosening allows re-expression of plasticity-associated programs previously silenced under metabolic or redox stress.
- BDNF Mimetics:
- 7,8-Dihydroxyflavone (7,8-DHF) acts as a TrkB agonist, mimicking BDNF to directly activate downstream pathways like PI3K/Akt and MAPK/ERK.
- Promotes spine stabilization, synaptic integration, and anti-apoptotic signaling—critical to restoring flexibility in simplified circuits.
Restore Network Dynamics
Goal: Resynchronize neuronal oscillations and reopen phase-locked signal propagation, degraded by circuit rigidity and entropy-driven decoupling.
- Gamma Entrainment:
- 40 Hz sensory stimulation (visual, auditory, or tactile) can entrain cortical gamma oscillations
- activates Ca²⁺ influx via NMDA receptors, triggering:
- ↑ CaMKII, CREB, BDNF → LTP-related gene transcription, ↑ dendritic spine density, ↑ surface AMPA receptor expression
- Shown to restore hippocampal–cortical coherence in animal models of AD (e.g., Iaccarino et al., 2016)
- increases TREM2 expression, a receptor essential for: Sensing lipid debris (from Aβ or dying synapses, Triggering phagocytic machinery
- Leads to: ↑ engulfment of extracellular amyloid-β, ↑ lysosomal activity (CD68, Lamp1 expression)
- activates Ca²⁺ influx via NMDA receptors, triggering:
- 40 Hz sensory stimulation (visual, auditory, or tactile) can entrain cortical gamma oscillations
- Closed-loop TMS:
- Adaptive theta-burst or phase-targeted transcranial magnetic stimulation (TMS) dynamically adjusts based on real-time EEG, promoting STDP-compatible excitation and reactivation of dormant networks.
- Enhances cognitive flexibility and phase-amplitude coupling without inducing excitotoxicity.
Reboot Metabolism
Goal: Compensate for glucose hypometabolism and redox collapse through alternative fuel delivery, mitochondrial rescue, and ATP restoration.
- Ketogenic Strategies:
- Medium-chain triglycerides (MCTs) are rapidly metabolized in the liver to ketone bodies, primarily β-hydroxybutyrate (BHB), which cross the blood–brain barrier and enter neurons via monocarboxylate transporters (MCT1/2)
- Once inside, BHB bypasses impaired glycolysis by directly entering mitochondrial ketolysis, generating acetyl-CoA for the TCA cycle and restoring ATP production
- also modulates gene expression through class I HDAC inhibition and reduces neuroinflammation by suppressing NLRP3 inflammasome activation.
- Reduce ROS generation, preserve NAD⁺/NADH ratios, and activate neuroprotective signaling (e.g., via PPARα).
- Mitochondrial Peptides:
- SS-31 (Elamipretide): Targets cardiolipin in inner mitochondrial membrane, improving electron transport chain (ETC) efficiency and reducing proton leak.
- Nicotinamide mononucleotide (NMN): Precursor to NAD⁺, restores mitochondrial biogenesis via SIRT1–PGC1α signaling axis.
Reduce Proteotoxic Load
Goal: Eliminate entropy-driving protein aggregates before they seed irreversible structural or immune cascades.
- Oligomer-Targeted Immunotherapy:
- Soluble Aβ antibodies (e.g., lecanemab, donanemab) preferentially bind low-molecular-weight Aβ oligomers implicated in synaptotoxicity and network decoupling.
- Avoids immune overactivation by sparing larger fibrillar deposits and circumventing Fc-mediated microglial priming.
- Autophagy Enhancement:
- Mild mTORC1 inhibition (e.g., rapamycin analogues) promotes autophagosome formation, aiding intracellular clearance of tau oligomers and oxidized proteins.
- Can synergize with proteasome enhancers or lysosomal stabilizers (e.g., trehalose).
Reconfigure Sleep and Circadian Architecture
Goal: Restore glymphatic cycling and global metabolic rhythm coherence, degraded during sleep fragmentation and circadian desynchronization.
- Orexin Modulators:
- Suvorexant (dual orexin receptor antagonist) improves sleep onset and deep sleep maintenance, enhancing AQP4-mediated solute clearance during slow-wave sleep.
- Melatonin Agonists:
- Ramelteon activates MT1/MT2 receptors, strengthening circadian phase alignment and supporting nocturnal BDNF expression, which is critical for synaptic repair.
- Light and Feeding Entrainment:
- Timed blue-spectrum light exposure and time-restricted feeding entrain peripheral and central CLOCK–BMAL1 oscillations, restoring transcriptional coherence across CNS cell types and aligning sleep–wake energetics with glymphatic surges.
Refined Pathological Homeostasis
State: The system reaches a stable but maladaptive attractor state. Cellular subsystems—microglia, astrocytes, neurons—have now undergone irreversible reprogramming. This is not a chaotic collapse, but an ordered dysfunction, where energy-saving, pro-inflammatory, and immune-dominant pathways persist as the new default.
Pathophysiology:
1.Neuronal Silencing and Plasticity Collapse
- Phenomenon: Widespread CREB downregulation, loss of dendritic spines, reduced LTP, and failure of spike-timing-dependent plasticity (STDP).
- Key Research:
- Katsouri, L., et al. (2023). Impaired CREB-dependent plasticity in AD models. Nature Neuroscience.
https://doi.org/10.1038/s41593-023-01320-0 - Huang, L., et al. (2022). Dendritic spine loss precedes neuronal death in Alzheimer’s disease. Cell Reports.
https://doi.org/10.1016/j.celrep.2022.111046
- Katsouri, L., et al. (2023). Impaired CREB-dependent plasticity in AD models. Nature Neuroscience.
2.Microglial Senescence and Immune Paralysis
- Phenomenon: Microglia lose P2RY12/TREM2 responsiveness, shift into dystrophic forms, and fail to clear debris or regulate complement signaling.
- Key Research:
- Luo, J., et al. (2022). Senescent microglia fail to restrict neurodegeneration in Alzheimer’s. Nature Aging.
https://doi.org/10.1038/s43587-022-00213-y - Zhou, Y., et al. (2023). Dystrophic microglia and impaired clearance mechanisms in AD progression. Science Translational Medicine.
https://doi.org/10.1126/scitranslmed.abc5342
- Luo, J., et al. (2022). Senescent microglia fail to restrict neurodegeneration in Alzheimer’s. Nature Aging.
3.A1 Astrocyte Consolidation and Toxic Gain-of-Function
- Phenomenon: Astrocytes adopt a pro-inflammatory A1 profile (upregulation of GFAP, C3, Serpina3n), secrete synaptotoxic factors, and impair metabolic support.
- Key Research:
- Liddelow, S.A., et al. (2022). Astrocyte reactivity and synapse loss in late-stage AD. Nature.
https://doi.org/10.1038/s41586-022-04984-w - Habib, N., et al. (2020). Transcriptional mapping of A1 astrocytes in Alzheimer’s. Cell.
https://doi.org/10.1016/j.cell.2020.09.002
- Liddelow, S.A., et al. (2022). Astrocyte reactivity and synapse loss in late-stage AD. Nature.
4.mTORC1 Hyperactivity and Proteostasis Failure
- Phenomenon: Aberrant mTORC1 signaling suppresses autophagy and contributes to tau hyperphosphorylation, defective lysosomal degradation, and synaptic rigidity.
- Key Research:
- Caccamo, A., et al. (2021). mTORC1 inhibition restores proteostasis in AD models. Journal of Clinical Investigation.
https://doi.org/10.1172/JCI146632 - Saxton, R.A., & Sabatini, D.M. (2019). mTOR signaling in aging and Alzheimer’s disease. Science.
https://doi.org/10.1126/science.aaw8470
- Caccamo, A., et al. (2021). mTORC1 inhibition restores proteostasis in AD models. Journal of Clinical Investigation.
5.Blood-Brain Barrier Collapse and Peripheral Infiltration
- Phenomenon: Tight junction loss, pericyte degeneration, and T-cell/monocyte infiltration leading to parenchymal inflammation.
- Key Research:
- Nation, D.A., et al. (2019). Blood–brain barrier breakdown as early biomarker of cognitive dysfunction. Nature Medicine.
https://doi.org/10.1038/s41591-019-0457-z - Sweeney, M.D., et al. (2020). Pericytes and BBB integrity in Alzheimer’s. Neuron.
https://doi.org/10.1016/j.neuron.2020.05.027
- Nation, D.A., et al. (2019). Blood–brain barrier breakdown as early biomarker of cognitive dysfunction. Nature Medicine.
6.Chronic Inflammatory Loops via Astrocyte-Microglial Crosstalk
- Phenomenon: Sustained IL-1β, IL-6, and TNF-α release from glial interactions; self-reinforcing loop driven by DAMPs and failure to resolve innate immune response.
- Key Research:
- Heneka, M.T., et al. (2020). Neuroinflammation in Alzheimer’s: Glial crosstalk as a driver. Immunity.
https://doi.org/10.1016/j.immuni.2020.02.010 - Reich, D.S., et al. (2023). Immune loop disruption in neurodegeneration. Nature Neuroscience.
https://doi.org/10.1038/s41593-023-01392-y
- Heneka, M.T., et al. (2020). Neuroinflammation in Alzheimer’s: Glial crosstalk as a driver. Immunity.
Implications:
1.Cognitive Degeneration
- Core Symptoms:
- Profound amnesia
- Semantic memory breakdown
- Disorientation in time/place/person
- Pathophysiological Basis:
- Hippocampal and default mode network (DMN) atrophy
- Collapse of NMDA/AMPA balance and CREB signaling
- Irreversible loss of synaptic scaffolding (PSD-95, Homer1a)
- Biomarkers:
- ↑ pTau181
- ↓ CSF Aβ₄₂
- ↑ Neurogranin
- Regions Affected:
- Hippocampus
- Medial temporal lobe
- Posterior cingulate cortex
2.Motor and Coordination Deficits
- Core Symptoms:
- Bradykinesia
- Rigidity
- Frequent falls
- Pathophysiological Basis:
- Basal ganglia–cortical disconnection
- Astrocytic K⁺ buffering collapse (↓ Kir4.1)
- Oligodendrocyte metabolic exhaustion → myelin breakdown
- Biomarkers:
- ↑ Neurofilament Light (sNfL)
- ↑ Soluble TREM2
- Regions Affected:
- Basal ganglia
- Cerebellum
- Motor cortex
3.Behavioral Rigidity & Neuropsychiatric Symptoms
- Core Symptoms:
- Apathy
- Delusions or paranoid ideation
- Vocal tics
- Irritability or aggression
- Pathophysiological Basis:
- Fronto-limbic signal entropy and disconnection
- Microglial over-pruning via dysregulated C1q–CR3 axis
- Inflammatory cytokine spillover (↑ IL-6, TNF-α)
- Biomarkers:
- ↑ IL-6
- ↑ TNF-α
- ↑ YKL-40 (astrocyte reactivity marker)
- Regions Affected:
- Prefrontal cortex
- Anterior cingulate
- Amygdala
4.Circadian and Sleep Disruption
- Core Symptoms:
- Sleep-wake cycle inversion
- Evening agitation
- Fragmented or superficial sleep
- Pathophysiological Basis:
- CLOCK–BMAL1 decoupling
- AQP4 mislocalization → impaired glymphatic flow
- Orexinergic dysregulation
- Biomarkers:
- ↓ Nocturnal melatonin
- ↑ Orexin A
- Regions Affected:
- Suprachiasmatic nucleus
- Thalamus
- Hypothalamus
5.Sensory Decoupling :
- Core Symptoms:
- Loss of smell (hyposmia)
- Impaired depth perception
- Auditory–visual desynchrony or hypersensitivities
- Pathophysiological Basis:
- Redox collapse in olfactory bulb
- Aggregation-prone protein accumulation
- Entropy-induced desynchronization in occipital–parietal circuits
- Biomarkers:
- ↑ 8-OHdG (oxidative DNA damage marker)
- ↑ Lipid peroxidation products
- Regions Affected:
- Olfactory bulb
- Occipito-parietal junction
- Superior colliculus
Therapeutic Goal: Slow deterioration, preserve remaining function, and relieve system stress.
Clinical Application:
1.Suppress Chronic Neuroinflammation
- Goal: Interrupt glial feed-forward loops and cytokine-driven damage
- Interventions:
- P2X7 antagonists – limit ATP-driven inflammasome activation (e.g., JNJ-55308942)
- TREM2 agonists – shift microglia to phagocytic, non-inflammatory states (e.g., AL002)
- IL-1β inhibitors – dampen cytokine amplification (e.g., canakinumab)
2.Reinforce Astrocytic Metabolic Scaffolding
- Goal: Re-energize astrocyte–neuron coupling and restore glutamate/K⁺ regulation
- Interventions:
- Monocarboxylate support: lactate or pyruvate supplementation
- Kir4.1 channel activators (experimental)
- EAAT2-enhancing agents (e.g., riluzole, ceftriaxone)
3.Slow Tau Propagation and Proteostatic Drift
- Goal: Prevent further synaptotoxicity and cytoskeletal collapse
- Interventions:
- Anti-tau antibodies (e.g., gosuranemab, semorinemab)
- Microtubule-stabilizing agents (e.g., epothilones – limited clinical success)
- HSP90 inhibitors – upregulate endogenous chaperones
4.Enhance Circuit Output with Network Simplification
- Goal: Boost output from preserved hubs (e.g., primary sensory, motor cortex)
- Interventions:
- Cholinesterase inhibitors (e.g., donepezil) – increase in Acetylcholin boosts signal-to-noise in residual cortical regions
- Non-invasive neuromodulation (e.g., tDCS, focused ultrasound to posterior cortex) – targets posterior/sensorimotor cortex → enhances residual network efficiency
- Transcranial Direct Current Stimulation: A non-invasive technique where low electrical currents (1–2 mA) are applied through electrodes on the scalp to modulate membrane potentials, making neurons slightly more or less likely to fire
- Focused Ultrasound: A precise, non-invasive method that delivers high-frequency sound waves to modulate or open the blood–brain barrier (BBB) in localized brain areas.
5.Mitigate Redox Crisis and Mitochondrial Failure
- Goal: Minimize further ROS-induced damage and energy failure
- Interventions:
- SS-31 (Elamipretide)
- Targets cardiolipin in the inner mitochondrial membrane → stabilizes ETC complexes, reduces proton leak, lowers ROS, and improves ATP production.
- NAD⁺ Boosters (NR, NMN)
- Elevate NAD⁺ levels, activating SIRT1–PGC1α signaling → promotes mitochondrial biogenesis, improves redox balance, and restores neuronal energy capacity.
- MitoQ
- Mitochondria-targeted antioxidant → scavenges ROS (superoxide, peroxynitrite), protects cardiolipin, and preserves membrane potential.
- SS-31 (Elamipretide)
6.Preserve Sleep Architecture and Circadian Phasing
- Goal: Prevent irreversible collapse of glymphatic clearance
- Interventions:
- Orexin antagonists (e.g., suvorexant)
- Melatonin agonists (e.g., ramelteon)
- Chronotherapy: timed light exposure and meal regulation
Conclusion
Alzheimer’s disease is not a static pathological entity but a phase-dependent systems failure in dynamic regulation across cellular, spatial, and temporal axes. From early disruption of synaptic and redox homeostasis to maladaptive glial reactivity and circuit collapse, the progression toward pathological homeostasis is shaped by misaligned feedback loops—between neurons, glia, vasculature, and circadian regulators. Each layer—from mitochondrial metabolism to immune surveillance—undergoes phase transitions that fail to recalibrate, leading to structural consolidation of dysfunction.
Crucially, Alzheimer’s pathology emerges not from a single insult, but from a desynchronized convergence: impaired clearance, chronic inflammation, disrupted network rhythms, and energetic exhaustion. Therapeutic approaches must therefore move beyond lesion-targeting and adopt a phase-specific, systems-oriented perspective—addressing microglial pruning, astrocytic buffering, mitochondrial resilience, and network reactivation.
By aligning interventions with the phases of disruption, reaction, adaptation, and refined homeostasis, a new paradigm becomes possible—one that restores synchrony, preserves cognitive networks, and re-engages neuroplasticity before the system locks into irreversible decline.
Abbreviations List
AD – Alzheimer’s Disease
Aβ – Amyloid-beta
AMPAR – α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor
APP – Amyloid Precursor Protein
AQP4 – Aquaporin-4
ATP – Adenosine Triphosphate
BDNF – Brain-Derived Neurotrophic Factor
BMAL1 – Brain and Muscle ARNT-Like 1 (circadian gene)
CNS – Central Nervous System
CREB – cAMP Response Element-Binding Protein
EAAT2/GLT-1 – Excitatory Amino Acid Transporter 2 / Glutamate Transporter-1
ERK/MAPK – Extracellular Signal-Regulated Kinase / Mitogen-Activated Protein Kinase
GABA – Gamma-Aminobutyric Acid
GFAP – Glial Fibrillary Acidic Protein
GSH/GSSG – Reduced/Oxidized Glutathione
IL-1β/IL-6/TNF-α – Interleukin-1β / Interleukin-6 / Tumor Necrosis Factor-α
ISF/CSF – Interstitial Fluid / Cerebrospinal Fluid
LTP/LTD – Long-Term Potentiation / Long-Term Depression
mTORC1 – Mechanistic Target of Rapamycin Complex 1
NAD⁺/NADH – Nicotinamide Adenine Dinucleotide (oxidized/reduced)
NMDA – N-Methyl-D-Aspartate (receptor)
OXPHOS – Oxidative Phosphorylation
pTau – Phosphorylated Tau
PKA/PKC – Protein Kinase A / Protein Kinase C
PSD-95 – Postsynaptic Density Protein 95
ROS – Reactive Oxygen Species
SIRT1 – Sirtuin 1
STDP – Spike-Timing Dependent Plasticity
TREM2 – Triggering Receptor Expressed on Myeloid Cells 2
UPS – Ubiquitin–Proteasome System
YKL-40 – Chitinase-3-like protein 1 (astrocyte activation marker)
sNfL – Serum Neurofilament Light Chain
tDCS – Transcranial Direct Current Stimulation
MCT1/MCT2 – Monocarboxylate Transporter 1 / 2
References
Ahlquist, R. (2023). Synaptic entropy and PI3K modulation in early neurodegeneration. Annals of Neurosciences.
Błaszczyk, J. W. (2020). Energy metabolism decline in the aging brain. Metabolites, 10(4), 131. https://doi.org/10.3390/metabo10040131
Caccamo, A., et al. (2021). mTORC1 inhibition restores proteostasis in AD models. Journal of Clinical Investigation. https://doi.org/10.1172/JCI146632
Calabrese, V., et al. (2020). Cellular stress responses, the hormesis paradigm, and vitagenes: Novel targets for therapeutic intervention. Antioxidants & Redox Signaling.
Cunnane, S. C., et al. (2020). Brain energy rescue: An emerging therapeutic concept. Nature Reviews Drug Discovery, 19(9), 609–633. https://doi.org/10.1038/s41573-020-00085-3
Habib, N., et al. (2020). Transcriptional mapping of A1 astrocytes in Alzheimer’s. Cell. https://doi.org/10.1016/j.cell.2020.09.002
Hardeland, R. (2021). Melatonin and inflammation—Story of a double-edged blade. Journal of Pineal Research, 70(5). https://doi.org/10.1111/jpi.12718
Heneka, M. T., et al. (2015). Neuroinflammation in Alzheimer’s disease. The Lancet Neurology, 14(4), 388–405. https://doi.org/10.1016/S1474-4422(15)70016-5
Heneka, M. T., et al. (2020). Neuroinflammation in Alzheimer’s: Glial crosstalk as a driver. Immunity. https://doi.org/10.1016/j.immuni.2020.02.010
Iaccarino, H. F., et al. (2016). Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature, 540(7632), 230–235. https://doi.org/10.1038/nature20587
Kang, D. W., et al. (2021). Effects of non-invasive brain stimulation on cognition in patients with Alzheimer’s disease. Journal of Alzheimer’s Disease, 79(1), 245–262. https://doi.org/10.3233/JAD-200668
Katsouri, L., et al. (2023). Impaired CREB-dependent plasticity in AD models. Nature Neuroscience. https://doi.org/10.1038/s41593-023-01320-0
Leng, F., & Edison, P. (2021). Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nature Reviews Neurology, 17, 157–172. https://doi.org/10.1038/s41582-020-00435-y
Liddelow, S. A., et al. (2022). Astrocyte reactivity and synapse loss in late-stage AD. Nature. https://doi.org/10.1038/s41586-022-04984-w
Luo, J., et al. (2022). Senescent microglia fail to restrict neurodegeneration in Alzheimer’s. Nature Aging. https://doi.org/10.1038/s43587-022-00213-y
Mullard, A. (2022). Anti-tau antibodies stumble in Alzheimer trials. Nature Reviews Drug Discovery, 21, 1–3. https://doi.org/10.1038/d41573-022-00091-0
Musiek, E. S., et al. (2013). Circadian clock proteins regulate neuronal redox homeostasis and neurodegeneration. Journal of Clinical Investigation, 123(12), 5389–5400. https://doi.org/10.1172/JCI70317
Nation, D. A., et al. (2019). Blood–brain barrier breakdown as early biomarker of cognitive dysfunction. Nature Medicine. https://doi.org/10.1038/s41591-019-0457-z
Qiu, C., et al. (2021). Disruption of the circadian clock exacerbates neurodegeneration and Aβ pathology in a mouse model of Alzheimer’s disease. Acta Neuropathologica Communications, 9(1), 21. https://doi.org/10.1186/s40478-021-01106-w
Reich, D. S., et al. (2023). Immune loop disruption in neurodegeneration. Nature Neuroscience. https://doi.org/10.1038/s41593-023-01392-y
Saxton, R. A., & Sabatini, D. M. (2019). mTOR signaling in aging and Alzheimer’s disease. Science. https://doi.org/10.1126/science.aaw8470
Shi, Y., et al. (2017). ApoE4 markedly exacerbates tau-mediated neurodegeneration. Nature, 549, 523–527. https://doi.org/10.1038/nature24016
Song, H., et al. (2015). Circadian clock gene BMAL1 regulates astrocyte activation and neuroinflammation. eLife, 4, e06111. https://doi.org/10.7554/eLife.06111
Sweeney, M. D., et al. (2020). Pericytes and BBB integrity in Alzheimer’s. Neuron. https://doi.org/10.1016/j.neuron.2020.05.027
Tagliabue, S., et al. (2023). Microvascular cerebral blood flow dynamics and glymphatic system integrity. Journal of Neuroscience Research.
Zhou, Y., et al. (2023). Dystrophic microglia and impaired clearance mechanisms in AD progression. Science Translational Medicine. https://doi.org/10.1126/scitranslmed.abc5342