/assets/images/provider/photos/2824605.jpg)
What if the common thread behind Alzheimer’s, Parkinson’s, and other neurodegenerative conditions isn’t just neuronal loss—but chronic inflammation within the brain itself? In this new chapter from the upcoming Brain Longevity eBook, Dr. Cohen explores how neuroinflammation acts as a root cause of brain aging and cognitive decline, and why emerging non-invasive therapies are reshaping the future of neurological care. Read on for a science-backed look at how targeting inflammation may be key to protecting and optimizing brain health over time.
Neuroinflammation represents a central pathophysiological mechanism in neurodegenerative diseases, functioning as both a consequence and driver of neuronal damage. Approximately 15% of the global population is affected by neurodegenerative disorders, with neuroinflammation associated with all major conditions including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), [Huntington's disease](/rare-disease/huntington-disease) (HD), and multiple sclerosis (MS). While conventional pharmacological interventions often show limited efficacy and significant adverse effects, emerging non-invasive neuromodulatory techniques—including photobiomodulation (PBM), hyperbaric oxygen therapy (HBOT), and transcranial magnetic stimulation (TMS)—offer promising alternative therapeutic strategies by targeting multiple neuroinflammatory pathways simultaneously.
Glial cell activation—particularly of microglia and astrocytes—represents the primary cellular mechanism driving neuroinflammatory responses. When damage signals or foreign pathogens enter the central nervous system (CNS), glial cells become activated, and overactivation triggers release of neuroinflammatory markers including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-10 (IL-10), nitric oxide (NO), and cyclooxygenase-2 (COX-2).
Microglia exhibit dual protective and detrimental functions, with disease-associated subtypes modulating inflammatory responses. Astrocytes polarize into A1 (neurotoxic) and A2 (neuroprotective) phenotypes, contributing differentially to protein clearance and synaptic dysfunction. Neuroinflammatory cascades involve alterations in cross-talk between glial cells and neurons, with common signaling pathways including NF-κB and MAPK activation. The NLRP3 inflammasome pathway plays a critical role, exacerbating chronic inflammation through blood-brain barrier disruption and cytokine release.
PBM uses red and near-infrared light (typically 600-1000 nm wavelengths) to stimulate cellular processes through photon absorption by chromophores, particularly cytochrome c oxidase in mitochondria.This absorption elevates ATP synthesis, reduces oxidative stress, and alleviates inflammation through activation of transcription factors and signaling mediators.
PBM exerts anti-inflammatory effects through multiple molecular pathways:
- Activation of extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase (MAPK), and protein kinase B (Akt) pathways
- Upregulation of neurotrophic factors including brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF)
- Modulation of neurotransmitter systems
- Enhanced mitochondrial biogenesis and function (increased Bcl-2, reduced Bax, enhanced ATP production)
Systematic reviews demonstrate that PBM effectively reduces glial activation, pro-inflammatory cytokine expression, and oxidative stress while increasing anti-inflammatory responses and antioxidant capacity in animal models of neurodegenerative diseases. Specifically, PBM has been shown to:
- Reduce microglial activation and astrocytic reactivity
- Decrease pro-inflammatory cytokines (TNF-α, IL-1β, IL-6)
- Increase anti-inflammatory cytokines (IL-10)
- Suppress NF-κB signaling
- Reduce oxidative stress markers (malondialdehyde) and increase antioxidant enzymes (superoxide dismutase)
Transcranial PBM with 808 nm light alters expression of genes and proteins associated with AD pathogenesis. In a 30-day course of daily 1-hour sessions in mice, PBM modulated:
- Essential genes associated with oxidative stress (NF-κBIα, JUN, JUND, PKC)
- Inflammation markers (DOCK4/6, IL-1RAPL1, TNFαIP6)
- Apoptosis regulators (CASP3, TNFαIP6, AKT3, CDKN1A)
- Main AD risk genes (BACE1, BACE2, PSEN2, APH1B) involved in amyloid precursor protein (APP) processing
APP concentration was reduced after PBM treatment, suggesting potential to inhibit development of AD pathology.
PBM has demonstrated beneficial effects in multiple neurodegenerative conditions including traumatic brain injury, ischemia, neurodegenerative diseases, aging, epilepsy, depression, and spinal cord injury.The therapy is non-invasive, broad-acting, and exhibits minimal toxicity, making it an attractive alternative to conventional pharmacological approaches.
HBOT involves breathing 100% oxygen at pressures greater than atmospheric pressure (typically 1.5-3.0 atmospheres absolute), promoting brain recovery and neuroplasticity through modulation of key cellular and molecular mechanisms.
Primary pathways affected by HBOT include:
- Mitochondrial biogenesis and function: Increased Bcl-2, reduced Bax, enhanced ATP production
- Neurogenesis: Upregulation of Wnt-3 and VEGF/ERK signaling
- Synaptogenesis: Elevated GAP43 and synaptophysin expression
- Anti-inflammatory responses: Reduced TNF-α and IL-6
HBOT significantly suppresses neuroinflammation in AD models through multiple mechanisms. In animal studies, HBOT demonstrated the following benefits:
- Promoted microglial transition to a surveillance phenotype (decreased soma area, increased branching)
- Reduced chronic neuroinflammation
- Restored mitochondrial quality control by upregulating PINK1 and parkin expression
- Enhanced autophagosome formation and modulated mitophagy-associated pathways
- Reduced amyloid-β accumulation through upregulation of LRP1 (a key Aβ clearance transporter)
Meta-analysis of randomized controlled trials demonstrates that HBOT remarkably ameliorates cognitive function in AD patients. Pooled evidence showed:
- Improved Mini-Mental State Examination scores (MD = 3.08, 95%CI [2.56, 3.61], p < 0.00001)
- Improved Alzheimer's Disease Assessment Scale-Cognitive scores (MD = -4.53, 95%CI [-5.05, -4.00], p < 0.00001)
- Improved activities of daily living (MD = 10.12, 95%CI [4.46, 15.79], p = 0.0005)
- Reduced malondialdehyde levels (oxidative stress marker)
- Increased superoxide dismutase levels (antioxidant enzyme)
- Reduced IL-1β levels
- Increased TGF-β1 levels
- No significant adverse events (OR = 1.17, 95%CI [0.68, 2.03], p = 0.58)
In elderly patients with significant memory loss, HBOT increased cerebral blood flow and improved cognitive performance. The therapy alleviates vascular dysfunction, reduces brain hypoxia, and decreases amyloid burden through persistent structural changes in blood vessels.
HBOT's ability to simultaneously act on multiple pathological cascades—combined with its noninvasive nature and favorable safety profile—makes it a uniquely promising therapeutic strategy.
Repetitive transcranial magnetic stimulation (rTMS) represents a promising non-invasive neuromodulatory intervention for neurodegenerative diseases, exerting therapeutic effects through multiple neurobiological mechanisms that directly address underlying neuroinflammatory pathways. Through electromagnetic induction, rTMS selectively modulates cortical excitability, with high-frequency stimulation (≥5 Hz) enhancing neural excitability and low-frequency stimulation (≤1 Hz) producing inhibitory effects.
At the cellular and molecular level, rTMS influences synaptic plasticity, neurotransmitter systems, and neuroinflammation. The technique modulates microglial activation, promoting neuroprotective and plasticity-enhancing processes by reducing pro-inflammatory cytokine release and promoting anti-inflammatory microglial polarization. Specifically, rTMS decreases pro-inflammatory cytokines including TNF-α, IL-1β, and IL-6 while increasing anti-inflammatory cytokines such as IL-10. Additionally, rTMS increases neurotrophic factors (particularly BDNF), counteracts amyloid and tau accumulation, and restores the excitation/inhibition balance by targeting glutamatergic and GABAergic pathways.
Meta-analyses demonstrate that rTMS produces significant cognitive improvements in patients with Alzheimer's disease and mild cognitive impairment. A comprehensive systematic review including 25 randomized controlled trials showed large effect sizes on global cognition measures: Montreal Cognitive Assessment (SMD = 0.85, 95% CI [0.26, 1.44], p = 0.005), Mini-Mental State Examination (SMD = 0.80, 95% CI [0.26, 1.33], p = 0.003), and Alzheimer's Disease Assessment Scale-Cognitive Subscale (SMD = -0.96, 95% CI [-1.32, -0.60], p < 0.001).
The pooled evidence indicates an overall medium-to-large effect size (SMD = 0.77-1.14) favoring active rTMS over sham stimulation in improving cognitive functions. Importantly, rTMS was safe and well tolerated with infrequent serious adverse events across 143 studies involving 5,800 participants.
rTMS modulates key pathological processes in Alzheimer's disease at multiple levels. The intervention:
- Reduces amyloid precursor protein (APP) concentration and modulates genes involved in APP processing (BACE1, BACE2, PSEN2, APH1B)
- Alters expression of genes associated with oxidative stress (NF-κBIα, JUN, JUND, PKC), inflammation (DOCK4/6, IL-1RAPL1, TNFαIP6), and apoptosis (CASP3, AKT3, CDKN1A)
- Enhances synaptic plasticity and promotes lasting structural changes in functional and structural connectivity
- Increases neurotrophic factors and modulates neurotransmitter circuits characteristically disrupted in AD
Using the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) criteria, high-frequency rTMS over the left DLPFC is classified as probably effective (Level B evidence) for improving cognition in Alzheimer's disease. Component network meta-analysis confirms that high-frequency rTMS demonstrates superior efficacy compared to transcranial direct current stimulation, with patients with AD showing better responses than those with MCI.
rTMS demonstrates significant efficacy in reducing motor symptoms of Parkinson's disease, with pooled evidence showing an overall medium effect size (SMD = 0.46, 95% CI [0.29-0.64], p < 0.001) favoring active rTMS over sham stimulation. A comprehensive overview of systematic reviews with meta-analysis, including 107 unique primary studies, provides low-to-moderate certainty evidence that high-frequency stimulation of the primary motor cortex (M1) and supplementary motor area (SMA) significantly improves general motor impairment, gait, functional mobility, and balance.
Conclusion:
These three non-invasive neuromodulatory techniques—photobiomodulation (PBM), transcranial magnetic stimulation (TMS), and hyperbaric oxygen therapy (HBOT)—share common anti-neuroinflammatory mechanisms while offering distinct therapeutic advantages for neurodegenerative diseases. All three modalities reduce pro-inflammatory cytokines (TNF-α, IL-1β, IL-6), modulate microglial activation, enhance mitochondrial function, increase neurotrophic factors, and promote neuroprotection through complementary pathways.
References