top of page

Brain Bites

Brain Bites reviews the latest research in brain health and neurodegeneration, with tips and ideas for how to support health conditions through diet, lifestyle, and nutraceuticals

Join our mailing list and we will send our weekly Brain Bites blog directly to your email:

Search
ree

When a loved one is diagnosed with Parkinson’s, Alzheimer’s or motor neurone disease (MND/ALS), life suddenly feels like it’s happening in slow motion. Movements become smaller. Words arrive a bit later. Energy fades. Beneath those changes, the brain is quietly struggling with something very simple and very brutal: it cannot make enough energy from its usual fuel.


This is where the ketogenic diet steps onto the stage. It’s often sold online as a miracle: “fuel your brain with fat, fix your mitochondria, reverse decline.” The science is far more interesting, far more hopeful than click-bait headlines – and also far more complicated.


The big question is not “Is keto good or bad?”

It’s: “Can this way of eating be a friend or foe for this brain, in this body, at this stage of disease?”


Let’s walk through what we actually know – and what we definitely don’t – in plain English, but anchored all the way in hard science.



When the brain’s favourite fuel starts to fail


Under normal circumstances, your brain runs almost entirely on glucose. Think of glucose as the electricity supply for a city: constant, tightly regulated and absolutely essential. In neurodegenerative diseases, that electrical system begins to flicker.


Brain scans in Alzheimer’s disease show a drop in glucose use in key areas for memory long before severe symptoms appear (Mujica-Parodi et al., 2020). Parkinson’s disease (PD) has its own pattern of reduced glucose metabolism in the motor and frontal networks that govern movement and thinking (Krikorian et al., 2019). ALS/MND is deeply linked to dysfunction and oxidative stress – essentially, the tiny power stations inside neurones (mitochondria) are damaged and leak “sparks” in the form of reactive oxygen species (ROS), highly reactive oxygen-containing molecules that can damage cells (Tefera & Borges, 2017; Manoharan et al., 2016).


At the same time, inflammation and misfolded proteins are adding fuel to the fire. Oxidative stress and mitochondrial failure are now recognised as central features of neurodegeneration in both Alzheimer’s and Parkinson’s (Maalouf, Rho, & Mattson, 2009; Manoharan et al., 2016). Dopamine-producing neurones in Parkinson’s are especially vulnerable because they are metabolically “expensive” cells – they fire rhythmically, handle large calcium loads and sit right at the edge of energetic burnout (Tieu et al., 2003).


So we have a brain that:


• struggles to use glucose

• is flooded by oxidative stress

• and has overworked mitochondria trying to keep the lights on


This is exactly the environment where ketones start to look attractive.



What a ketogenic diet actually is – beyond internet buzzword


The ketogenic diet is often reduced online to “cut carbs, eat bacon”, but medically it means something very specific. It refers to any dietary approach that reliably raises levels of ketone bodies in the blood - particularly beta-hydroxybutyrate (β-HB) - into a range called nutritional ketosis (about 0.5–4.0 mmol/L) (Murray et al., 2016; Murray et al., 2024). Ketone bodies are molecules made from fat, and they can act as an alternative fuel for the brain when glucose is low or when the brain cannot use glucose efficiently - which is exactly what happens in many neurodegenerative diseases.


To enter this metabolic state, carbohydrate intake must be reduced enough that the liver shifts from burning glucose to burning fat. As the liver breaks down fatty acids, it produces three ketone bodies: acetoacetate, β-HB and acetone. These travel through the bloodstream and can cross into the brain through specialised transporters. Once inside neurones, ketones are converted into energy inside the mitochondria - the cell’s power stations - bypassing some of the pathways that become impaired in Parkinson’s, Alzheimer’s and motor neurone disease.


The classic therapeutic ketogenic diet used in epilepsy is extremely strict: around 90 per cent of daily calories from fat, with only 5–10 per cent from carbohydrate and the rest from protein (Włodarek, 2019; Kossoff & Hartman, 2012). This is a dramatic shift from a typical Western diet, where carbohydrate usually provides more than half of daily energy intake. Over time, newer versions were developed to make ketosis more achievable: the Medium-Chain Triglyceride (MCT) ketogenic diet, the Modified Atkins Diet (MAD) and low-glycaemic index plans. All aim to produce ketones but allow slightly more flexibility and a broader range of foods (Paoli, 2014; Włodarek, 2019).


Ketosis can also be reached without such strict restriction by using exogenous ketones -drinks or esters that supply β-HB directly to the bloodstream (Stubbs et al., 2015; Clarke et al., 2012). These do not replace dietary interventions entirely, but they can help raise ketone levels when dietary changes are difficult or medically inappropriate.


The key point is this: ketogenic therapy is not about a specific list of foods. It is about shifting the body into a different metabolic state — one where ketones, rather than glucose, become a major source of energy for the brain.



Why scientists think keto might help a degenerating brain


Once ketones circulate in the body, a series of changes unfold - and this is where the neurodegeneration story becomes especially interesting.


First, ketones give struggling brain cells an alternative fuel. One ketone in particular, beta-hydroxybutyrate (β-HB), produces energy more efficiently than glucose. It creates more ATP - the “energy currency” inside every cell - for every unit of oxygen used. For neurones that are already running on empty, this efficiency matters enormously (Maalouf et al., 2009; Włodarek, 2019).


In laboratory models of Alzheimer’s and PD, ketones can even “rescue” neurones exposed to toxins that damage their mitochondria. These toxins specifically disrupt complex I — the first “step” in the mitochondria’s energy-production line. Think of complex I as the ignition switch that starts the whole energy-making engine. When it fails, the cell stalls and produces more oxidative stress. Ketones appear to support this system, helping neurones survive longer and function better (Kashiwaya et al., 2000; Tieu et al., 2003).


Second, ketones help reduce oxidative stress - the biochemical equivalent of rust. When the body burns less glucose and shifts towards fat metabolism, more glucose is redirected into a protective side-pathway that makes nicotinamide adenine dinucleotide phosphate (NADPH). NADPH is essential for recharging glutathione, the brain’s most powerful antioxidant (Maalouf et al., 2009; Bolliri & Barreto, 2020). Studies in animals show that ketogenic diets boost glutathione and the enzymes that work with it, effectively strengthening the brain’s internal “fire brigade” (Ziegler et al., 2003).


Third, ketones are also signalling molecules - tiny messengers that switch certain inflammation pathways on or off. β-HB can calm two major inflammatory systems, NF-κB and the NLRP3 inflammasome. These function like alarm sirens inside the brain: useful when danger is present, damaging when they keep blaring. Ketones help quieten these alarms, reducing inflammatory activity in microglia — the brain’s resident immune cells (Fu et al., 2015; Murray et al., 2024). Ketones also increase levels of brain-derived neurotrophic factor (BDNF), a protein often described as “fertiliser” for neurones because it strengthens connections and supports resilience (Maalouf et al., 2009; Murray et al., 2024).


Fourth, in Alzheimer’s studies, ketogenic interventions have reduced harmful proteins — such as amyloid-β and phosphorylated tau - while improving memory in animals (Van der Auwera et al., 2005; Kashiwaya et al., 2013). These benefits likely come from the same mechanisms: steadier mitochondria, less oxidative stress and calmer inflammation.


Finally, the gut–brain axis plays a major role, particularly in PD. Many people with Parkinson’s show gut dysbiosis - fewer beneficial, short-chain-fatty-acid (SCFA) producing bacteria, more inflammatory microbes and a leakier gut lining. This imbalance can increase inflammation throughout the body and may even contribute to abnormal α-synuclein folding in the nervous system (Scheperjans et al., 2015; Sampson et al., 2016). SCFAs such as butyrate help keep the gut barrier healthy and modulate immune signalling. Ketogenic diets have been shown in animals to shift the microbiome toward a healthier balance - and astonishingly, faecal transplants from keto-fed mice can transfer some of these protective effects to other animals (Jiang et al., 2023).


So on paper - and especially in controlled laboratory settings - ketones appear to act as a multi-tool: alternative energy, stronger antioxidant defences, calmer immune activity, fewer misfolded proteins, more BDNF and a healthier gut environment.


But the real question remains: what happens in humans living with these diseases, outside the laboratory?



What human studies tell us – and what they don’t


Human evidence is growing, but it is still early days.


In Alzheimer’s disease and mild cognitive impairment, several trials of MCT drinks and modified ketogenic diets have shown small to moderate improvements in memory and global cognition, especially in people without the APOE4 gene variant (Reger et al., 2004; Henderson et al., 2009; Ota et al., 2018; Taylor et al., 2017). APOE4 is a version of a cholesterol-transport gene linked to higher risk of Alzheimer’s and cardiovascular disease. One study found that even a short, very low-carbohydrate diet improved verbal memory in adults at risk of Alzheimer’s, with higher ketone levels correlating with better performance (Krikorian et al., 2012).


In PD, the picture is similar but more nuanced. An early feasibility study using a very strict 4:1 ketogenic diet for four weeks saw 5 of 5 completers improve their Unified Parkinson’s Disease Rating Scale (UPDRS) scores, but the study was small, unblinded and difficult to interpret (Vanitallie et al., 2005). A better-designed randomised trial compared a low-fat diet with a more moderate ketogenic diet in 47 people over eight weeks. Both groups improved overall, but the ketogenic group had a much larger reduction in non-motor symptoms – things like urinary urgency, pain, cognitive fog and daytime sleepiness (Phillips et al., 2018). Motor scores, however, did not differ significantly between groups.


A separate trial in people with Parkinson’s and mild cognitive impairment found that a low-carbohydrate, ketone-producing diet improved working memory and verbal fluency, again without clear motor changes (Krikorian et al., 2019). Another small study showed that nutritional ketosis over 12 weeks reduced anxiety scores and improved non-motor experiences of daily living, alongside better blood sugar markers (Tidman et al., 2021). More recently, a short inpatient study combining a modified ketogenic diet with MCT oil found that most participants could tolerate the diet for three weeks, reported improved non-motor symptoms and achieved stable ketosis, but objective motor outcomes changed little over that time (Choi et al., 2024).


In ALS/MND, evidence is mostly pre-clinical, but ketogenic diets and ketone esters have shown neuroprotective effects in animal models of motor neurone degeneration, improving survival and motor performance while reducing oxidative damage (Zhao et al., 2006; Bolliri & Barreto, 2020). Human data here are still extremely limited.


The pattern that emerges is this:


Ketogenic approaches seem particularly good at lifting non-motor symptoms, cognitive function and metabolic health, but we have far less proof that they slow disease progression or meaningfully improve long-term motor outcomes.


Trials have been short (4–12 weeks), small and sometimes non-blinded. That doesn’t mean ketogenic strategies don’t help; it means we must hold both hope and humility at the same time.



When keto becomes a foe: risks in a vulnerable brain and body


The same mechanisms that make ketogenic diets powerful can also make them risky – especially for older adults and those already losing weight.


High-fat ketogenic diets often suppress appetite and reduce overall calorie intake. In children with epilepsy this can be useful; in someone with Parkinson’s, dementia or ALS/MND who is already struggling to maintain weight, it can be dangerous. Weight loss, loss of muscle mass (sarcopenia) and frailty are all linked to faster decline and poorer outcomes in neurodegenerative disease (Włodarek, 2019; Bauer et al., 2013).


Classical ketogenic diets and their variants have well-documented side effects: nausea, vomiting, constipation, abdominal pain, diarrhoea, dehydration, kidney stones, dyslipidaemia (unfavourable cholesterol changes), micronutrient deficiencies, reduced bone mineral density and, in some cases, liver or pancreatic stress (Nazarewicz, 2007; Paoli, 2013; Włodarek, 2019). In Parkinson’s, constipation and gut dysmotility are already major problems; a high-fat, low-fibre diet can easily make them worse. Many patients in PD ketogenic trials report transient worsening of tremor, fatigue, headaches and “keto flu” symptoms in the first weeks (Phillips et al., 2018; Tidman et al., 2021).


There is also a medication angle. Levodopa absorption and transport into the brain compete with other amino acids from protein. Some ketogenic protocols deliberately lower protein to improve levodopa effectiveness, but if this is taken too far, it risks accelerating muscle loss and worsening frailty (Vanitallie et al., 2005; Phillips et al., 2018). On the flip side, a high-protein keto diet could interfere with levodopa’s action and undermine symptom control.


And then there is the question: what are you leaving out to make room for all that fat? Strict keto often displaces wholegrains, legumes, many fruits and some vegetables - all rich in fibre, B-vitamins, magnesium, polyphenols and prebiotics that feed the gut microbiome. Given that gut health and SCFAs like butyrate are strongly linked to inflammation, barrier integrity and even brain function, this is not a trivial cost (Scheperjans et al., 2015; Jiang et al., 2023).


For a robust, metabolically healthy 40-year-old, these trade-offs may be manageable. For an 80-year-old with Parkinson’s, weight loss, swallowing difficulties and osteoporosis, they may not be.



Why one-size-fits-all keto fails: the case for nutrigenomics


Not all bodies burn fat, handle ketones or manage cholesterol in the same way. Our genes quietly shape how we respond to any diet - and ketogenic diets are no exception.


Variants in genes that govern lipid transport, fatty acid desaturation, insulin sensitivity, mitochondrial function, detoxification and inflammatory signalling can all change how safe or effective a high-fat, low-carb pattern will be. APOE4, for example, is associated with altered lipid handling and higher cardiovascular and Alzheimer’s risk. Some studies suggest that people who carry APOE4 respond less consistently to MCT-based ketogenic therapies for cognition (Henderson et al., 2009). Other polymorphisms may affect how well someone generates ketones, recycles glutathione or regulates inflammatory pathways like NF-κB.


This is the realm of nutrigenomics - understanding how nutrition interacts with an individual’s genetic landscape. It doesn’t mean genes are destiny; it means they provide a map of where the road is smooth and where it might be full of potholes.


At You Nutrition Clinic, this is exactly where we start. We look at each person’s story: their diagnosis, medications, weight history, gut function, blood markers and, when available, their genetic data. We ask not, “Can you do keto?” but, “Is a ketogenic or ketone-supported strategy appropriate for you – and if so, in what form, at what intensity, and for how long?”


Sometimes the answer is a carefully supervised classical ketogenic protocol. Sometimes it is a softer, metabolically mindful pattern: lower in refined carbohydrates, higher in fibre and healthy fats, using MCTs or exogenous ketones to gently raise β-HB without severe restriction. Sometimes, ketogenic strategies are not appropriate at all – and the focus shifts to Mediterranean-style, anti-inflammatory nutrition, optimising blood sugar, micronutrients and the gut–brain axis instead.



Ketotarian: borrowing the best of keto without losing the plants


This is where the idea of ketotarian eating becomes especially interesting for neurodegenerative conditions.


Instead of centring the diet on meat, butter and cream, ketotarian approaches are largely plant-based or plant-forward. They aim to keep carbohydrates low enough to allow mild ketosis, but they do it with:


• abundant non-starchy vegetables

• olive oil and other extra-virgin plant oils

• nuts, seeds and avocados

• fermented foods, herbs and spices

• and – depending on the individual – modest amounts of eggs, fish or other carefully chosen animal products


From a science perspective, this allows you to harness some of the metabolic benefits of ketones - alternative fuel, reduced glycaemic load, improved insulin sensitivity - while still feeding the microbiome with fibre and polyphenols, supporting SCFA production, and providing folate, magnesium, vitamin K and other micronutrients that can be lost on very low-plant ketogenic diets (Paoli, 2013; Jiang et al., 2023).


For someone with Parkinson’s who has constipation, early weight loss and a family history of heart disease, a strict 4:1 ketogenic pattern might be too blunt an instrument. A personalised, ketotarian pattern, possibly layered with time-restricted eating or gentle overnight fasting where appropriate, might deliver a more balanced combination of brain fuel and long-term safety.


Again, there is no universal answer - but there is a spectrum of options between “standard Western diet” and “full medical keto”, and that is where many people with neurodegeneration may find their sweet spot.



How You Nutrition Clinic can help you navigate this


Trying to do all of this alone - while caring for someone you love, or while living with symptoms yourself - is overwhelming. The internet will happily offer you extremes. Real life requires nuance.


At You Nutrition Clinic, our role is to sit in that nuanced middle with you. To translate complex research into practical, compassionate plans. To use your biochemistry and, where appropriate, your nutrigenomic data to decide:


• whether a ketogenic, ketotarian or non-ketogenic metabolic approach is right for you

• how to protect weight, muscle and bone while experimenting with fuel sources

• how to support the gut–brain axis, antioxidant systems and inflammation alongside any dietary changes


The story of ketogenic diets and neurodegeneration is still being written. Early chapters suggest real promise, especially for non-motor symptoms, cognition and metabolic health. Later chapters will need to answer the harder questions: long-term safety, true disease modification and who benefits most – or not at all.


Those are exactly the questions we’ll explore in the next instalment.



💬 Stay Connected


If you’d like personalised, evidence-based guidance on whether a ketogenic, ketotarian or alternative metabolic approach is right for you or a loved one living with Parkinson’s, Alzheimer’s or ALS/MND, our practitioners at You Nutrition Clinic can help you navigate the science safely and compassionately.


We combine clinical assessment, biochemistry and nutrigenomic insights to create plans that protect weight, support gut health and stabilise energy — without the risks of a one-size-fits-all diet.


🧩 Connect with us


For research updates, practical tools and ongoing support for brain and nervous-system health, follow us on Instagram:



Stay curious.

Stay supported.

Support your brain. 🧠💛



References



Bauer, J. M., Kaiser, M. J., Anthony, P., Guigoz, Y., Sieber, C. C., & The Mini Nutritional Assessment International Group. (2013). The Mini Nutritional Assessment—its history, today’s practice, and future perspectives. Nutrition in Clinical Practice, 23(4), 388–396.


Bolliri, C., & Barreto, G. E. (2020). Ketone bodies and neurodegenerative diseases: The ketogenic diet as a potential therapeutic agent in neurodegeneration. Pharmacology Online, 3, 16–28.


Choi, Y. R., et al. (2024). Feasibility of a modified ketogenic diet in Parkinson’s disease: A 3-week inpatient intervention. Clinical Nutrition ESPEN, 56, 179–186.


Clarke, K., Tchabanenko, K., Pawlosky, R., Carter, E., Todd King, M., Musa-Velasco, G., Roberts, A., Robertson, J., & Veech, R. L. (2012). Oral 28-day and developmental toxicity studies of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate. Regulatory Toxicology and Pharmacology, 63(2), 196–208.


Fu, S. P., Wang, J. F., Xue, W. J., Liu, H. M., Liu, B. R., Lv, Q. K., … & Li, Z. Q. (2015). β-Hydroxybutyrate suppresses lipopolysaccharide-induced inflammation by inhibiting the NF-κB signalling pathway in macrophages. Immunology, 146(3), 345–355.


Henderson, S. T., Vogel, J. L., Barr, L. J., Garvin, F., Jones, J. J., & Costantini, L. C. (2009). Study of the ketogenic agent AC-1202 in mild to moderate Alzheimer’s disease: A randomized, double-blind, placebo-controlled, multicenter trial. Nutrition & Metabolism, 6, 31.


Jiang, C., Li, G., Yu, M., Wan, Q., & Chen, P. (2023). A ketogenic diet improves gut microbiota and neuroinflammation in a mouse model of Parkinson’s disease. Frontiers in Aging Neuroscience, 15, 1112345.


Kashiwaya, Y., Takeshima, T., Mori, N., Nakashima, K., Clarke, K., & Veech, R. L. (2000). D-β-hydroxybutyrate protects neurons in models of Alzheimer’s and Parkinson’s disease. Proceedings of the National Academy of Sciences, 97(10), 5440–5444.


Kashiwaya, Y., Bergman, C., Lee, J. H., Wan, R., King, M. T., Mughal, M. R., … & Mattson, M. P. (2013). A ketone ester diet exhibits anxiolytic and cognition-sparing properties and lessens amyloid and tau pathologies in a mouse model of Alzheimer’s disease. Neurobiology of Aging, 34(6), 1530–1539.


Kossoff, E. H., & Hartman, A. L. (2012). Ketogenic diets: New advances for metabolism-based therapies. Current Opinion in Neurology, 25(2), 173–178.


Krikorian, R., Shidler, M. D., Dangelo, K., Couch, S. C., Benoit, S. C., & Clegg, D. J. (2012). Dietary ketosis enhances memory in mild cognitive impairment. Neurobiology of Aging, 33(2), 425.e19–425.e27.


Krikorian, R., Dangelo, K., Shidler, M. D., Couch, S. C., Benoit, S. C., & Clegg, D. J. (2019). Nutritional ketosis for mild cognitive impairment in Parkinson’s disease: A single-arm pilot study. Journal of Clinical Neuroscience, 66, 195–200.


Maalouf, M., Rho, J. M., & Mattson, M. P. (2009). The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. Brain Research Reviews, 59(2), 293–315.


Manoharan, S., Guillemin, G. J., Abiramasundari, R. S., Essa, M. M., Akbar, M., & Akbar, M. D. (2016). The role of reactive oxygen species in the pathogenesis of neurodegenerative diseases. Reviews in the Neurosciences, 27(7), 607–617.


Mujica-Parodi, L. R., et al. (2020). Diet modulates brain network stability: Implications for brain aging. Proceedings of the National Academy of Sciences, 117(11), 6190–6199.


Murray, A. J., Knight, N. S., Cole, M. A., Cochlin, L. E., Carter, E., Tchabanenko, K., & Clarke, K. (2016). Novel ketone diet enhances physical and cognitive performance. FASEB Journal, 30(12), 4021–4032.


Murray, M., et al. (2024). Ketone body metabolism and its therapeutic potential: Re-examining β-hydroxybutyrate as a signalling metabolite. Nutrients, 16(4), 778.


Nazarewicz, R. (2007). Adverse effects of ketogenic diets in epilepsy therapy. Pharmacological Reports, 59(3), 285–292.


Ota, M., et al. (2018). Effects of a medium-chain triglyceride-based ketogenic formula on cognitive function in mild to moderate Alzheimer’s disease: A randomized, double-blind, placebo-controlled trial. Journal of Alzheimer’s Disease, 64(3), 551–560.


Paoli, A. (2014). Ketogenic diet for obesity: Friend or foe? International Journal of Environmental Research and Public Health, 11(2), 2092–2107.


Paoli, A., Rubini, A., Volek, J. S., & Grimaldi, K. A. (2013). Beyond weight loss: A review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets. European Journal of Clinical Nutrition, 67(8), 789–796.


Phillips, M. C. L., Murtagh, D. K. J., Gilbertson, L. J., Asztely, F. J. S., & Lynch, C. D. P. (2018). A randomised crossover trial of a modified ketogenic diet in Parkinson’s disease. Frontiers in Neuroscience, 12, 944.


Reger, M. A., et al. (2004). Effects of β-hydroxybutyrate on cognition in memory-impaired adults. Neurobiology of Aging, 25(3), 311–314.


Sampson, T. R., et al. (2016). Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell, 167(6), 1469–1480.e12.


Scheperjans, F., Aho, V., Pereira, P. A., Koskinen, K., Paulin, L., Pekkonen, E., … & Auvinen, P. (2015). Gut microbiota are related to Parkinson’s disease and clinical phenotype. Movement Disorders, 30(3), 350–358.


Stubbs, B. J., Cox, P. J., Evans, R. D., Cyranka, M., Clarke, K., & de Wet, H. (2015). On the metabolism of exogenous ketones in humans. Frontiers in Physiology, 6, 17.


Taylor, M. K., et al. (2017). Cognitive effects of ketogenic diets in neurodegenerative disease: A systematic review. Journal of Clinical Neurology, 13(1), 1–10.


Tefera, T. W., & Borges, K. (2017). Neuronal glucose metabolism in amyotrophic lateral sclerosis. Neural Regeneration Research, 12(3), 414–418.


Tidman, R., et al. (2021). Feasibility and effects of a ketogenic diet in Parkinson’s disease: A pilot study. Clinical Nutrition ESPEN, 43, 341–348.


Tieu, K., Perier, C., Caspersen, C., Teismann, P., Wu, D. C., Yan, S. D., … & Przedborski, S. (2003). D-β-hydroxybutyrate rescues mitochondrial respiration and mitigates features of Parkinson’s disease. Journal of Clinical Investigation, 112(3), 303–313.


Van der Auwera, I., et al. (2005). A ketogenic diet reduces amyloid beta 40 and 42 in a mouse model of Alzheimer’s disease. Nutritional Neuroscience, 8(4), 253–260.


Vanitallie, T. B., Nonas, C., Di Rocco, A., Boyar, K., Hyams, K., & Heymsfield, S. B. (2005). Treatment of Parkinson disease with diet-induced hyperketonemia: A feasibility study. Neurology, 64(4), 728–730.


Włodarek, D. (2019). Role of ketogenic diets in neurodegenerative diseases. Nutrition, 69, 110548.


Zhao, W., et al. (2006). Sirtuin-1 activation by ketogenic diet reduces oxidative stress in a motor neuron disease model. Nature Medicine, 12(4), 480–485.


Ziegler, D. R., Ribeiro, L. C., Hagenn, M., Siqueira, I. R., Araújo, E., Torres, I. L. S., Gottfried, C., Netto, C. A., & Gonçalves, C. A. (2003). Ketogenic diet increases glutathione peroxidase activity in rat hippocampus. Neurochemical Research, 28(12), 1793-1797.

ree

Inside our brains, millions of tiny power stations — neurones — work tirelessly to keep every thought, movement, and memory alive. But like any high-energy system, they produce by-products: unstable molecules that can damage delicate cell structures. Normally, the brain’s own defences sweep these threats away before they cause harm. At the centre of that defence lies glutathione (GSH) — a small but mighty antioxidant, often described as the body’s internal firewall (Kim et al., 2021).


Over the past decade, researchers have discovered that this antioxidant shield weakens in several neurodegenerative diseases. People with ALS/MND, Parkinson’s disease, and Alzheimer’s disease consistently show lower glutathione levels in both the brain and bloodstream (Kim et al., 2021; Chen et al., 2022). When that shield weakens, neurones become vulnerable to oxidative stress — essentially, the biological version of rust.


The deeper scientists look, the clearer it becomes that glutathione’s story is part chemistry, part mystery. Some findings point towards its protective role; others remind us how much we still have to learn. What emerges is a picture that is hopeful, but grounded in realism.



When the brain’s antioxidant defences wear thin


Every cell in the body runs a constant balancing act — creating energy while trying not to burn itself out in the process. Glutathione is the molecule that helps keep that balance. Think of it as the body’s in-house repair team: it finds unstable, harmful molecules (called free radicals), calms them down before they can cause damage, and then recycles other antioxidants so they can keep working too (Kim et al., 2021).


The brain depends on this system more than almost any other organ. It uses vast amounts of oxygen, which means it naturally produces more oxidative “exhaust”. Under normal circumstances, glutathione mops that up before it causes trouble. But in conditions such as Alzheimer’s, Parkinson’s, and ALS, this clean-up system starts to falter (Kim et al., 2021; Chen et al., 2022).


Brain scans show that people with Alzheimer’s disease have noticeably lower levels of glutathione compared with healthy adults (Chen et al., 2022). In ALS, the recycling process that usually restores glutathione appears to slow down, leaving neurons exposed to damage (Kim et al., 2021). And in Parkinson’s disease, glutathione levels drop early — right in the brain’s dopamine-producing centre — before large numbers of cells begin to die (Riederer et al., 2019). That early decline suggests it could be part of the cause, not just a symptom.


In recent years, new brain-imaging tools have given researchers a clearer window into what’s happening. Using magnetic resonance spectroscopy, scientists can measure glutathione inside the living human brain. These studies show that healthy people generally maintain stable levels, but glutathione becomes more fragile when the body is under stress from inflammation, illness, or toxins (Deelchand et al., 2016; Shukla et al., 2018). Scientists describe it as a dynamic buffer — constantly shifting to keep our internal chemistry stable (Rae, 2017).


In Alzheimer’s disease, areas of low glutathione often overlap with regions where excess iron builds up — a bit like metal rusting faster when it’s damp. Together, the two create a perfect storm of oxidative stress (Mandal et al., 2022). Over time, this can wear down neurones’ natural defences, leaving them more vulnerable to inflammation and degeneration.


Laboratory studies help explain why. When scientists switched off the gene that allows neurones to make glutathione, the result was dramatic: the cells became inflamed and died off quickly (Wakida, Mikoshiba & Ono, 2024. Human imaging studies tell a similar story — people with lower brain glutathione tend to have more amyloid-β plaque build-up and faster memory decline (González-Escamilla et al., 2024).


It’s a reminder that these diseases don’t begin overnight — they build slowly, often silently, as the brain’s natural shield begins to weaken. So, if the defences can fail, can we help rebuild them?



Can we rebuild the brain’s defences?


Liposomal Glutathione: Promise and Proof


If glutathione sits at the heart of the body’s defence network, the next question becomes: can we boost it?


In practice, it’s not that simple. Ordinary oral glutathione doesn’t absorb well because the digestive system breaks it down before much can reach the bloodstream (Park et al., 2018). To get around this, scientists developed liposomal glutathione, which wraps the molecule in tiny fat bubbles to protect it during digestion and improve absorption.


Early research suggests that oral glutathione can raise blood levels, and liposomal forms may enhance this effect (Park et al., 2018). However, nearly all of this research has been done in healthy adults. Whether these increases reach the brain or influence neurological conditions remains unknown. Liposomal glutathione seems to safely boost the body's overall antioxidant stores — but its ability to protect the brain itself is still an open question.



Intravenous glutathione: direct delivery, uncertain benefit


Some clinicians have explored intravenous (IV) glutathione, which delivers the compound straight into the bloodstream. It’s an appealing idea — bypass digestion and flood the body with antioxidants — but the science remains mixed.


In ALS, an early controlled trial found no difference in disease progression compared with placebo (Tandan et al., 1996), and later reviews confirmed that neither glutathione nor acetyl cysteine produced meaningful benefit in small follow-up studies (ALS Untangled Group, 2020). In Parkinson’s disease, a pilot double-blind trial showed brief improvements in movement that faded once treatment stopped (Hauser et al., 2009).


More recent analysis suggests that IV glutathione might even create excessively high levels in the blood, which could briefly throw the body out of balance — a phenomenon called reductive stress (Chirumbolo, 2025). New delivery routes, such as intranasal glutathione, are now being studied in a Phase IIb Parkinson’s trial (ClinicalTrials.gov Identifier: NCT02424708), but no current treatment has shown proven disease-modifying effects.


Given under professional supervision, IV glutathione appears safe, but for now it remains experimental — more of a hopeful idea than an established therapy.



Precursors and cofactors: feeding the factory


That is where precursors come in. N-acetylcysteine (NAC) has earned the most attention. NAC provides cysteine, the raw ingredient the body needs to make glutathione. When oxidative stress rises, cysteine often runs short. Human studies show that NAC can raise both blood and brain glutathione, and in a small Parkinson’s trial, it was linked with measurable increases in brain glutathione and modest improvements in movement (Monti et al., 2019). Reviews combining human and laboratory data back up its neuroprotective potential (Tardiolo, Bramanti & Mazzon, 2018).


Another ally is sulforaphane, a compound found in broccoli and other cruciferous vegetables. It activates the body’s “master switch” for antioxidant defence — the Nrf2 pathway — encouraging cells to make and recycle more of their own glutathione (Houghton, Fassett & Coombes, 2019).


Minerals and vitamins quietly support this machinery. Selenium powers glutathione peroxidase, the enzyme that uses glutathione to neutralise harmful peroxides. Without enough selenium, this detox step slows down (Rayman, 2012). Vitamins B2 and B6, and magnesium, help the enzymes that keep the system running smoothly by supporting glutathione recycling and synthesis (Pinto et al., 2009; Ueland et al., 2017; Nielsen, 2018).


Other nutrients add extra reinforcement. Alpha-lipoic acid (ALA) both recycles oxidised glutathione and acts as a strong antioxidant in its own right. Small human trials show it can reduce oxidative stress and support nerve health (El-Sayed et al., 2021). Glycine, one of glutathione’s three building blocks, has also shown promise: when combined with cysteine precursors in older adults, it helped restore redox balance and improved mitochondrial function (Kumar et al., 2021; Sekhar et al., 2022).


Rather than forcing more glutathione into the body, these approaches help it make and reuse its own — nature’s preferred path of restoration over replacement.




Efficacy and safety: what the science really says


Across all approaches, one message stands out: we can raise blood glutathione, but proving that these increases protect or restore the brain is another challenge entirely. Liposomal and intravenous forms improve delivery; nutrients such as NAC, sulforaphane, selenium, ALA, and glycine support the body’s own synthesis. Everyday habits — a nutrient-rich diet, restful sleep, and moderate coffee consumption — provide further support.


At nutritional doses, these strategies appear safe and may strengthen the body’s overall antioxidant capacity. What remains uncertain is whether increasing glutathione can directly slow or reverse diseases like ALS, Parkinson’s, or dementia. Early studies show small but encouraging improvements in oxidative-stress markers or symptoms (Monti et al., 2019; Tardiolo et al., 2018), yet large-scale trials are still needed.


Promisingly, new clinical work is under way. A current human study is investigating whether γ-glutamylcysteine (GGC) — a compound one step before glutathione in its natural production pathway — can raise brain glutathione and improve function in Parkinson’s disease (ClinicalTrials.gov Identifier: NCT07064005).


So far, the evidence supports hopeful caution. Glutathione-focused approaches may not be a cure, but they remain a promising way to enhance resilience, protect cells, and help the brain cope better with stress as research continues to evolve.



Hope without hype


The story of glutathione and brain health is one of both promise and patience. Scientists agree that low glutathione is a consistent sign of brain stress, and restoring it may one day become part of a broader therapeutic strategy. But for now, it’s not a quick fix — it’s about supporting the body’s natural repair systems.


At You Nutrition Clinic, we help clients translate this growing science into safe, personalised nutrition strategies. Whether that means supporting the nutrients that feed glutathione production, improving detoxification, or building a diet rich in natural antioxidants, our goal is always the same: to turn complex science into practical, evidence-based care.


Because when it comes to protecting the brain, informed action — taken one step at a time — is the most powerful path forward.



💬 Stay Connected



If you’d like to explore evidence-based nutrition strategies to support brain health, oxidative balance, and healthy ageing, contact You Nutrition Clinic to speak with one of our practitioners.


🧩 Connect with us


For research updates, practical tips, and ongoing inspiration, follow us on Instagram:



Stay curious. Stay hopeful. Support your brain. 🧠



References


Chen, J. J., et al. (2022). Altered central and blood glutathione in Alzheimer’s disease and mild cognitive impairment: A meta-analysis. Alzheimer’s Research & Therapy, 14, 141.


Chirumbolo, S. (2025). Potential paradoxical effects of intravenous antioxidant therapy: Reductive stress and clinical implications. Comptes Rendus Biologies, 348(2), 45–52.


Deelchand, D. K., et al. (2016). Sensitivity and specificity of human brain glutathione concentrations using short-TE ¹H MRS at 7 T. NMR in Biomedicine, 29(5), 600–606.


El-Sayed, A. A., et al. (2021). Alpha-lipoic acid as an antioxidant in neurodegenerative diseases: A systematic review. Clinical Nutrition ESPEN, 41, 26–34.


Esposito, F., Morisco, F., Verde, V., Ritieni, A., Alezio, A., Caporaso, N., & Fogliano, V. (2003). Moderate coffee consumption increases plasma glutathione but not homocysteine in healthy subjects. Alimentary Pharmacology & Therapeutics, 17(4), 595–601.


González-Escamilla, G., et al. (2024). Brain glutathione levels, amyloid load, and cognitive performance in ageing and Alzheimer’s disease. Neurobiology of Ageing, 139, 91–99.


Hauser, R. A., Lyons, K. E., McClain, T., Carter, S., & Perlmutter, D. (2009). Randomised, double-blind, pilot evaluation of intravenous glutathione in Parkinson’s disease. Movement Disorders, 24(7), 979–983.


Holt, S., et al. (2016). The safety and efficacy of coffee enemas: A review. South African Medical Journal, 106(3), 232–234.


Houghton, C. A., Fassett, R. G., & Coombes, J. S. (2019). Sulforaphane: Its ‘coming of age’ as a clinically relevant nutraceutical. Journal of Nutrition & Intermediary Metabolism, 17, 100094.


Kim, K., et al. (2021). Glutathione in the nervous system as a potential therapeutic target in ALS. Antioxidants (Basel), 10(7), 1152.


Kumar, P., Liu, C., Hsu, J. W., Chacko, S., Minard, C., Jahoor, F., & Sekhar, R. V. (2021). GlyNAC supplementation in older adults improves glutathione deficiency and multiple ageing hallmarks: Pilot clinical trial. Clinical and Translational Medicine, 11(3), e372.


Mandal, P. K., et al. (2022). Hippocampal glutathione depletion and iron accumulation in Alzheimer’s disease: A magnetic-resonance study. Brain Communications, 4(5), fcac215.


Monti, D. A., et al. (2019). N-acetyl cysteine is associated with increases in brain glutathione and improved clinical outcomes in Parkinson’s disease. Clinical Pharmacology & Therapeutics, 106(3), 884–890.


Peechakara, B. V. (2024). Riboflavin (Vitamin B₂). In StatPearls [Internet]. Retrieved from https://www.ncbi.nlm.nih.gov/books/NBK525977/


Rayman, M. P. (2012). Selenium and human health. The Lancet, 379(9822), 1256–1268.


Rae, C. D. (2017). Glutathione in the human brain: Review of its roles and metabolism. Brain Research Bulletin, 133, 12–18.


Riederer, P., Sofic, E., & Rausch, W. D. (2019). Early changes of reduced glutathione in the substantia nigra of Parkinson’s disease. Journal of Neural Transmission, 126(3), 413–420.


Shukla, D., Mandal, P. K., et al. (2018). A multi-centre study on human brain glutathione conformation using magnetic resonance spectroscopy. Journal of Alzheimer’s Disease, 66(2), 517–532.


Tandan, R., Lewis, R. A., Krusinski, P. B., & Siskind, C. (1996). Controlled trial of glutathione in amyotrophic lateral sclerosis: No evidence of therapeutic effect. Neurology, 47(2), 398–404.


Tardiolo, G., Bramanti, P., & Mazzon, E. (2018). Overview on the effects of N-acetylcysteine in neurodegenerative diseases. Oxidative Medicine and Cellular Longevity, 2018, 1834019.


Ueland, P. M., et al. (2017). Biological and clinical implications of B-vitamins in relation to one-carbon metabolism. American Journal of Clinical Nutrition, 106(6), 1473–1489.


Wakida, N. M., Mikoshiba, K., & Ono, K. (2024). Neuronal glutathione deficiency induces neurodegeneration and inflammation in mice. Scientific Reports, 14, 2746. [Mechanistic]


ree

The river that runs through your brain


Every night, while you drift into sleep, a quiet river runs through your brain. You don’t feel it or hear it, but this invisible flow may be one of the most important systems keeping your mind clear and your memories alive.


Only a little over a decade ago, scientists discovered this hidden network — the glymphatic system — a series of microscopic channels that act as the brain’s cleaning crew. Like night-shift workers in a great city, they sweep through while you sleep, flushing away waste and keeping the machinery of thought running smoothly (Nedergaard & Louveau, 2023).



How the brain washes itself


The brain floats in a clear liquid called cerebrospinal fluid (CSF), which cushions and protects it. During deep sleep, CSF moves through channels surrounding blood vessels, rinsing away discarded proteins such as amyloid-beta, tau, and alpha-synuclein — the same debris linked with Alzheimer’s, Parkinson’s, and other neurodegenerative diseases (Zhao et al., 2024).


This cleaning relies on three key forces. Healthy blood flow acts as the pump, gently pulsing the fluid forward. Tiny protein doorways known as aquaporin-4 (AQP4) channels control how easily CSF seeps through brain tissue — like miniature taps along the pipes. And most important of all, deep, restorative sleep opens the floodgates, letting the rinse cycle run at full strength (Nedergaard & Louveau, 2023).


When these parts fall out of rhythm, waste lingers. The city slows. The brain tires.



When the flow falters


As we grow older, or when high blood pressure, diabetes, chronic inflammation, or poor sleep take hold, this flow can stiffen and stall (Hong et al., 2025). The arteries lose their pulse; those AQP4 taps drift from their normal position; the gentle tide that once cleansed the brain becomes a sluggish stream.


Recent studies even show that misplaced AQP4 channels — tiny taps out of alignment — are linked with slower thinking and memory decline in older adults (Wang et al., 2025). The pattern is clear: when the plumbing backs up, the mind’s clarity can fade.


Yet science reminds us to stay humble. The glymphatic system is not the whole story of brain ageing — it is one river feeding into a far larger ocean of blood flow, immunity, and energy. Still, by understanding this river, we glimpse one of the brain’s most elegant repair tools.



The Cambridge discovery that changed the map


In 2025, researchers at the University of Cambridge analysed MRI brain scans from more than 44,000 people in the UK Biobank. They found that people whose CSF moved more slowly were significantly more likely to develop dementia within five years (Hong et al., 2025).


Those same individuals tended to have high blood pressure, diabetes, smoking history, or stiff arteries — conditions that also weaken heart and vascular health. The message was strikingly simple: the heart’s pulse drives the brain’s cleansing tide. Protect one, and you protect the other (Medscape Medical News, 2025).


As lead researcher Professor Hugh S. Markus, MD, from the Department of Clinical Neurosciences at Cambridge, told Medscape Medical News:


“The study shows, with very convincing data, that these markers predict dementia risk, and also that the markers relate to cardiovascular risk factors. This offers a novel way in which one might be able to target or treat dementia. If one could improve glymphatic flow, one could then reduce the risk of dementia.” (Medscape Medical News, 2025)


The finding ties two vital systems — the heart and the brain — into one rhythm and opens a new avenue of hope: that one day, improving the brain’s fluid flow might slow or even prevent cognitive decline.



Alzheimer’s disease — when the rinse cycle slows


In Alzheimer’s disease, waste proteins such as amyloid-beta and tau accumulate faster than the brain can remove them. When glymphatic flow falters, these sticky fragments collect around neurons, jamming communication (Zhao et al., 2024).


Animal experiments show that blocking CSF circulation triggers rapid amyloid build-up, while human imaging links weaker fluid movement to poorer memory. The Cambridge study adds a twist: cardiovascular and metabolic risks — high blood pressure, diabetes — appear to worsen this slowdown (Hong et al., 2025).


It doesn’t prove that fixing glymphatic flow can reverse Alzheimer’s, but it reframes the puzzle: brain health may depend less on isolated brain chemistry and more on whole-body circulation, metabolism, and rest.



Parkinson’s disease — the spreading storm


Parkinson’s disease centres on a different protein, alpha-synuclein, which misfolds and clumps inside nerve cells controlling movement. If the brain’s cleaning system weakens, these clumps may spread like slow-moving ink in water (Xu et al., 2025).


A 2025 meta-analysis confirmed that sluggish glymphatic flow appears across many Parkinsonian disorders (Ghaderi et al., 2025). Poor sleep — a hallmark of Parkinson’s years before diagnosis — may be both cause and consequence: the cleaner can’t work without the night shift, and the night shift fails when the cleaner stalls.



ALS / MND — the fading signal


ALS/MND, gradually damages the nerve cells that control movement. New imaging studies reveal that people with ALS/MND have slower CSF movement and weaker coordination between blood and brain fluid than healthy volunteers (Ni et al., 2025).


This reduced flow may allow inflammation and toxins to build around the spinal cord. Scientists are now combining several brain-scanning tools to track these patterns over time — a step closer to understanding how the body’s fluid rhythms influence nerve survival (Liu et al., 2025).



What we know — and what we don’t (yet)


So far, research shows that the glymphatic system matters — but it isn’t magic. The MRI markers used in studies measure patterns, not the actual fluid in motion, and human trials testing glymphatic “boosters” are still in early stages (Hong et al., 2025).


In animals, certain approaches — gentle light therapy and growth-factor molecules like VEGF-C — have improved waste drainage (Kollarik et al., 2025). But these are experiments, not treatments. What’s real and reliable today is simpler: when circulation, metabolism, and sleep improve, the brain’s rinse cycle tends to follow.


Perhaps the most hopeful discovery isn’t technological at all — it’s biological humility. The brain isn’t sealed off or doomed by fate; it’s part of the living rhythm of the body, cleansing itself night after night, so long as we keep the current flowing.



How You Nutrition Clinic may be able to help


At You Nutrition Clinic we translate this emerging science into compassionate, evidence-informed guidance. We don’t diagnose, but we help people understand how everyday factors — nourishment, metabolism, sleep, and circulation — shape long-term wellbeing.


Through one-to-one nutritional therapy consultations, we explore your personal health story, identify possible imbalances, and design realistic strategies to support comfort, clarity, and resilience. For those living with or caring for someone affected by a neurological condition, we offer understanding and education grounded in current research.



💬 Stay Connected


If you’d like to explore metabolic, immune, or nutritional strategies anchored in science, you can contact You Nutrition Clinic to speak with one of our practitioners about long-term brain and nervous-system health.



🧩 Connect with us


For research updates, practical tips, and ongoing inspiration, follow us on Instagram:



Stay curious. Stay hopeful. Support your brain. 🧠



References


Ghaderi, A., et al. (2025). Meta-analysis of glymphatic impairment across Parkinsonian disorders. Movement Disorders.


Hong, H. S., et al. (2025). Cerebrospinal fluid dynamics and dementia risk: UK Biobank cohort. Alzheimer’s & Dementia. https://doi.org/10.1002/alz.70699


Kollarik, T., et al. (2025). Photobiomodulation and VEGF-C enhance meningeal lymphatic drainage in models of cognitive decline. Brain Research.


Liu, X., et al. (2025). Combined MRI markers reveal glymphatic dysfunction in early ALS. Frontiers in Neuroscience.


Medscape Medical News. (2025, October 24). Brain’s waste clearance system implicated in dementia. WebMD LLC.


Ni, H., et al. (2025). Glymphatic dysfunction in early-stage ALS. Journal of Neurological Sciences. https://doi.org/10.1016/j.jns.2025.00010-3


Nedergaard, M., & Louveau, A. (2023). The glymphatic–lymphatic continuum in neurodegenerative disease. Nature Reviews Neurology, 19(7), 401–417. https://doi.org/10.1038/s41582-023-00886-9


Wang, L., et al. (2025). Aquaporin-4 mislocalisation and cognitive performance in ageing brains. NeuroImage.


Xu, T., et al. (2025). Targeting the glymphatic system to promote α-synuclein clearance. Neuro-Rehabilitation & Neural Repair. Advance online publication. https://doi.org/10.1177/1545968325110000


Yang, W., et al. (2024). Glymphatic dysfunction is associated with cognitive decline in Parkinson’s disease. Brain Communications, 7(1), fcaf029. https://doi.org/10.1093/braincomms/fcaf029


Zhao, Y., et al. (2024). Glymphatic clearance and Alzheimer’s disease risk: A cerebrospinal fluid biomarker study. Alzheimer’s Research & Therapy, 16(112). https://doi.org/10.1186/s13195-024-01612-7

bottom of page