Ketone body metabolism and cardiometabolic implications for cognitive health | npj Metabolic Health and Disease
npj Metabolic Health and Disease volume 2, Article number: 29 (2024) Cite this article
894 Accesses
7 Altmetric
Metrics details
Cardiometabolic complications of obesity present a growing public health concern and are associated with poor outcomes, mediated in part by an increased risk for cardiovascular disease, metabolic dysfunction-associated fatty liver disease, and systemic insulin resistance. Recent studies support that both insulin resistance and obesity are also associated with aberrant brain metabolism and cognitive impairment similar to what is observed in neurodegenerative diseases. Central to these pathological outcomes are adverse changes in tissue glucose and ketone body metabolism, suggesting that regulation of substrate utilization could be a mechanistic link between the cardiometabolic outcomes of obesity and the progression of cognitive decline. Here, we review ketone body metabolism in physiological and pathological conditions with an emphasis on the therapeutic potential of ketone bodies in treating cardiometabolic diseases and neurodegenerative diseases that lead to cognitive decline. We highlight recent findings in the associations among cardiometabolic disease, ketone body metabolism, and cognitive health while providing a theoretical framework by which ketone bodies may promote positive health outcomes and preserve cognitive function.
The alarming rise in cardiometabolic complications of obesity among adults and adolescents places a tremendous burden on healthcare systems1. Obesity increases the risk of all-cause mortality2, mediated in part by heightened potential for cardiovascular complications3, metabolic dysfunction-associated steatotic liver disease (MASLD)4, systemic inflammation5, heart failure with preserved ejection fraction (HFpEF)6, and neurodegeneration7. Central to these pathological conditions are the cardiometabolic consequences of aberrant energy production and nutrient storage8, leading to potential disruptions in the regulation of critical cellular processes (reviewed in refs. 9,10,11,12). Specifically, glucose and ketone body metabolism are altered in cardiometabolic diseases13,14, highlighting the importance of substrate metabolism as a factor underpinning poor health outcomes.
Emerging evidence suggests a connection between cardiometabolic complications of obesity and the progression of neurodegeneration7. Supporting this notion are findings suggesting that neurodegeneration is associated with altered brain glucose metabolism15,16 and findings that ketogenic therapies have shown promise in the treatment of neurological conditions associated with neurodegeneration17,18,19. Ketogenic therapies may maintain neurological energy homeostasis, and based on findings in the nervous system and beyond, may confer benefits through mechanisms beyond canonical oxidative roles20. The emerging relationship between cardiometabolic disease and neurodegenerative disorders21 could also be linked through consequences of dysregulated substrate metabolism.
In this review, we discuss the roles of ketone body metabolism and its connectivity with cardiometabolic and neurodegenerative disorders. We highlight both canonical and noncanonical roles of ketone body metabolism in preventing these adverse health outcomes. Since ketogenic therapies have been useful in recent advancements for the preservation of cognitive function22,23, ketone body metabolism may provide valuable insight into mechanistic links that bridge cardiometabolic diseases and the risk for cognitive decline.
Specifically, we cover the following topics:
Ketone body metabolism in physiological and pathological conditions
Cardiometabolic associations with neurodegeneration and cognitive decline
Interventions affecting ketone body metabolism and the effects on complex diseases
The ketone bodies acetoacetate (AcAc), D-β-hydroxybutyrate (D-βOHB), and acetone are primarily synthesized in the mitochondrial matrix of hepatocytes proportional to rates of adipose tissue lipolysis and hepatic fat oxidation. The isoform of 3-hydroxymethylglutaryl-CoA synthase 2 (HMGCS2) in hepatic mitochondria catalyzes the commitment step of ketogenesis that condenses mitochondrial β-oxidation-derived acetoacetyl-CoA and acetyl-CoA into HMG-CoA, which is then cleaved by HMG-CoA lyase (HMGCL) to produce AcAc. AcAc can be transported into circulation through monocarboxylate transporters (SLC16A1 [MCT1] or MCT2), spontaneously decarboxylated to acetone, or reduced to D-βOHB via mitochondrial D-βOHB dehydrogenase (BDH1) in an NADH-dependent manner. D-βOHB is then transported into circulation via the same transporters (MCT1/2). Once in circulation, ketone bodies enter extrahepatic tissues via MCT1, where D-βOHB oxidation proceeds via reversal of the BDH1 reaction, regenerating AcAc and NADH. Thereafter, AcAc is converted back to AcAc-CoA by the fate-committing mitochondrial enzyme, succinyl-CoA:3-oxoacid-CoA transferase (SCOT), which is expressed in all tissues except for hepatocytes and enables extrahepatic oxidation. Subsequently, through mitochondrial thiolase activity, extrahepatic mitochondria generate acetyl-CoA for cellular utilization. In physiological conditions, hepatic ketogenesis increases in response to limitations of carbohydrate availability, such as in fasting or adherence to low-carbohydrate, high-fat diets. Ketogenesis can be further enhanced with increases in energy expenditure or following exercise. Hepatic ketogenesis also increases in pathological conditions, such as diabetic ketoacidosis in individuals with type 1 diabetes, in which unrestrained adipose tissue lipolysis generates fatty acid ketogenic substrates. Hepatic ketone body production and extrahepatic utilization are regulated by insulin and glucagon, with additional control exerted by molecular mechanisms and various transcriptional regulations (reviewed in20).
Total ketone body concentrations in circulation typically remain below 250 µM in healthy, fed adults, but can rise to 1 mM following a 24 h fast or approach 20 mM in pathological conditions like diabetic ketoacidosis20. Elevations of endogenously-produced circulating ketone bodies are most often observed in postabsorptive conditions when circulating insulin levels are low and glucagon levels are relatively high24,25 (Fig. 1A). Conversely, increased pancreatic β-cell insulin secretion suppresses hepatic ketogenesis by reducing adipose tissue lipolysis24 and inhibiting hepatic Hmgcs2 transcription through phosphatidylinositiol-3-kinase/Akt signaling26. This suppression of hepatic ketogenesis may also occur through mammalian target of rapamycin 1 (mTORC1)-mediated inhibition of peroxisome-proliferator activated receptor α (PPARα)27. In cases of insulin resistance, higher circulating insulin levels are required to lower blood glucose via tissue uptake and suppression of adipose tissue lipolysis, which triggers hepatic gluconeogenesis. Insulin resistance is the hallmark of cardiometabolic disease, with chronic hyperinsulinemia and hyperglycemia contributing to aberrant tissue glucose metabolism and altering systemic lipid metabolism (reviewed in28). Thus, in obesity and insulin resistance, which both exhibit elevated fasting and postprandial insulin production, the dynamic range of hepatic ketogenesis is also affected.
Schematic depicting key regulators of hepatic ketogenesis and extrahepatic utilization (A). Hepatic ketogenesis is regulated by levels of circulating insulin and glucagon, with overall capacity influenced by adiposity (B) and the overall rate influenced by liver disease (C) and circulating concentrations (D). HSL hormone-sensitive lipase, Sk =skeletal, MASLD metabolic dysfunction-associated steatotic liver disease, MASH metabolic dysfunction-associated steatohepatitis. Created with BioRender.com
Obesity and insulin resistance are initially associated with compensatory increases in hepatic oxidative metabolism in response to lipid oversupply to the liver29; however, these conditions eventually progress to the accumulation of excess triglycerides in the liver30 that contribute to MASLD. Accumulation of hepatic triglycerides, together with the inhibitory effects of hyperinsulinemia, lead to impaired fasting hepatic ketogenesis31,32,33. Ketosis is less dynamic in obese and insulin-resistant states, as blood ketone levels are typically higher in the fed state due to elevations in lipolysis but lower in fasted states due to hyperinsulinemia, when compared to lean individuals (reviewed in ref. 34) (Fig. 1B). In the setting of hepatic steatosis with insulin resistance, there is greater disposal of β-oxidation-derived acetyl-CoA into the TCA cycle, with augmented gluconeogenic and lipogenic rates that could contribute to reduced ketone body formation35,36. This shift in substrate shuttling not only lowers ketone body production, but it also increases liver lipid storage and glucose production that further drives insulin resistance and hyperinsulinemia. Coupled with hepatic lipid overload, these events promote oxidative stress through chronic, compensatory activation of mitochondrial metabolism31, highlighting an important mechanistic link between obesity and poor health outcomes. These derangements in substrate metabolism additionally contribute to comorbidities such as type 2 diabetes mellitus (T2DM) and downstream cardiometabolic complications (reviewed in ref. 37). Regulation of ketone body metabolism in normal and pathological conditions is summarized in Fig. 1.
Once in circulation, ketone bodies are transported down a concentration gradient into extrahepatic tissues via MCT138,39. Oxidation of ketone bodies is proportional to tissue uptake40, which is greatest in high energy-requiring tissues such as skeletal muscle, heart, and brain when carbohydrates are in short supply. In rat brain slices, high concentrations of ketone bodies reduce glycolytic flux, and ketone body oxidation becomes the primary contributor to acetyl-CoA generation41. While ketone bodies are efficient substrates for energy provision in extrahepatic tissues42, they can also serve as anabolic substrates for de novo lipogenesis and sterol biosynthesis. In rat models, knockdown of acetoacetyl-CoA synthetase (AACS) lowers blood cholesterol, and in vitro silencing of AACS impairs expression of neuronal markers43,44,45,46,47,48.
Noncanonical roles of ketone bodies were not appreciated until the mid-2000s with the finding that βOHB inhibits adipocyte lipolysis through its binding to the nicotinic acid receptor GPR109A49. Agonism of this receptor by βOHB has also been associated with reductions in the synthesis of growth hormone-releasing hormone in the hypothalamus50 and macrophage-associated neuroprotection51, which could link ketone bodies to salutary changes in brain and neuronal physiology. βOHB also inhibits signaling through GPR41, which leads to reductions in heart rate and overall sympathetic drive52, and further links βOHB to neuronal function. A key set of mechanisms through which βOHB signals is through its ability to regulate histone acylation, including (i) inhibition of histone deacetylases (HDACs) which leads to preservation of histone acetylation53, and (ii) direct covalent modification of histone lysine residues54. In addition, AcAc, but not βOHB, binds the G-protein coupled receptor GPR43 to regulate lipoprotein lipase activity55. This binding specificity of AcAc could be energetically costly by requiring the NAD+-dependent oxidation of D-βOHB, but overall, there is less known regarding the noncanonical signaling effects of AcAc. Nevertheless, the emerging roles of ketone bodies serving signaling roles in the body have sparked increasing interest in therapeutic applications.
Sustained ketosis is associated with diminished extrahepatic expression of SCOT, which catalyzes the fate-committing step in ketone body oxidation56,57,58. Indeed, a paradoxical mismatch may occur between upregulated ketogenesis and diminished ketone disposal that contributes to conditions such as diabetic ketoacidosis. Even so, the pathogenesis of diabetic ketoacidosis is more complicated than this, since patients simultaneously present with hyperglycemia and metabolic acidosis (reviewed in ref. 59). Still, coupling hepatic ketogenesis to extrahepatic ketone body oxidation is critical, because ketone bodies are preferentially oxidized over glucose as a fuel for brain metabolism in fasting patients60. Supporting this notion is the observation that germline SCOT-knockout mice do not survive more than 48 h after birth due to ketoacidosis61. Similarly, humans with bi-allelic loss-of-function mutations in the OXCT1 gene, which encodes SCOT, are susceptible to developing life-threatening bouts of severe ketosis62. Together, these observations underscore the importance of balanced ketone body production and utilization since derangements can lead to severe, pathological consequences.
While aging is one of the strongest risk factors for cognitive decline63, observational studies suggest direct associations between cardiometabolic complications of mid-life obesity and the onset of dementia64,65. This implies potential roles of altered glucose and ketone body metabolism in its progression, with the added effects of altered insulin levels likely playing a role. Glucose is the primary fuel source for the brain in the fed state and the brain is a major consumer of the body’s total glucose utilization (>20%) when at rest66,67; however, in periods of low blood glucose, cells within the brain rely on the oxidation of lactate68, ketone bodies60, and to a lesser extent, fatty acids60,69. Thus, it is tempting to speculate that conditions affecting production and/or uptake of these metabolic substrates contribute to neurodegeneration and augmented risk of dementia.
Neurodegenerative diseases share key pathological features with cardiometabolic diseases, with the most notable features being chronic inflammation and insulin resistance70. Central to this link with chronic inflammation is the finding that the adipose-derived pro-inflammatory cytokine, interleukin-1β (IL1β), promotes hippocampal neuroinflammation and cognitive impairment71 in mice. The association between adiposity and cognitive impairment might also be mediated through visceral adipose tissue (VAT) inducing the NLR family pyrin domain containing 3 (NLRP3) inflammasome72, an intracellular sensor that contributes to onset of neurodegenerative pathology, such as in Alzheimer’s disease (AD)73. Importantly, βOHB inhibits the NLRP3 inflammasome74 and could be responsible for the therapeutic potential of ketone bodies on neuroinflammation associated with cognitive impairment. Additionally, secretion of adiponectin, which is an anti-inflammatory cytokine, is inversely associated with levels of VAT75. Adiponectin is an insulin-sensitizing adipokine that can cross the blood-brain barrier to promote cerebral glucose uptake76 and neuronal plasticity in the hippocampus77. Both circulating and hippocampal levels of adiponectin are reduced in obesity78, which could exacerbate poor neurometabolic outcomes. Obesity has been shown to increase neuroinflammation, microgliosis, and amyloid-β deposition in a mouse model of AD79, further highlighting the contribution of obesity to poor brain health outcomes.
While AD is the leading cause of dementia, it remains unclear how cardiometabolic features, like insulin resistance, contribute to its progression. Patients with cognitive impairment show diminished whole-body glucose disposal quantified by a euglycemic-hyperinsulinemic clamp, and increased homeostatic model assessment of insulin resistance (HOMA-IR) after fasting80, indicating there is systemic insulin resistance occurring simultaneously with cognitive impairment. Moreover, reductions in cerebral glucose metabolism are also observed in individuals with insulin resistance and prediabetes81, linking cardiometabolic diseases to changes in brain metabolism and cognitive decline. In AD patients, there is a progressive reduction in cerebral glucose uptake and metabolism82, with significant changes in cerebral metabolism observed up to 19 years before AD symptoms manifest83. Thus, insulin resistance may play an important role in the progression of neurogenerative diseases, such as AD.
AD is characterized by progressive neurodegeneration that stems from aggregation of neuronal amyloid-β plaques and hyperphosphorylated tau that forms neurofibrillary tangles (NFT)84. This pathology particularly affects the hippocampus, which is associated with memory formation85. A positive correlation between insulin resistance and the deposition of amyloid-β has been documented in late middle-aged humans86. Insulin resistance in the brain increases phosphorylation of amyloid precursor protein (APP) at Thr668 in AD mice and in rat embryonic cortical neurons87, while diet-induced obesity increases APP levels in the hippocampus of AD mouse models88. Phosphorylation of APP-carboxy-terminal-fragments promotes APP processing and increases production of amyloid-β peptide89, which could accelerate AD pathology. Studies performed in rat hippocampal slices suggest that insulin resistance and APP-induced augmentation of amyloid-β together activate glycogen-synthase-kinase-3β via impaired Akt signaling, which induces hyperphosphorylation of Tau protein in the brain and contributes to NFT formation and memory deficits90. Brain pathology usually correlates with the progression of cognitive decline, but discrepancies between pathological severity and clinical dementia symptoms are not uncommonly reported, as the cognitive reserve hypothesis posits that lifetime experiences, such as education and socioeconomic factors, greatly influence human brain health (reviewed in refs. 91,92,93). Nevertheless, cardiometabolic diseases do reduce cerebral glucose uptake and are directly associated with both neurodegenerative pathology and cognitive decline.
Diminutions in cerebral glucose metabolism, which is observed in AD patients, predicts late progression of dementia better than amyloid-β and NFTs alone94. Overexpression of glucose transporter-1 (GLUT1) in a Drosophila model of AD increases neuronal glucose uptake and simultaneously reduces neurodegeneration95. This implies a critical role of glucose metabolism in maintaining neuronal homeostasis. Isotope tracing studies suggest that in early stages of AD, there may be a compensatory increase in glucose oxidation of cortical, but not hippocampal, neurons96; however, there is progressive reduction in glucose uptake in later stages of AD. Interestingly, the capacity for ketone body metabolism remains intact in the hippocampus of a 5xFAD model97,98, potentially rescuing energy deficits lost to reductions in glucose uptake.
Oxidation is not the only fate of glucose in the brain. Glucose-derived enrichment into the pentose phosphate pathway for de novo biosynthesis of cholesterol, lipids, and nucleotides has been observed in astrocytes from murine, homozygous carriers of E4 for human Apolipoprotein E (APOEε4), a major genetic risk allele for late-onset AD99. Genetic polymorphisms of Apolipoprotein E (APOE), a major lipid-trafficking lipoprotein, modulates the efficiency of systemic lipid transport and fatty acid mobilization100,101 while also influencing cardiometabolic102,103 and cognitive trajectories104,105,106 in humans. The mechanisms by which APOE polymorphisms contribute to cognitive decline likely include reductions in neuronal insulin sensitivity107 and associated metabolic flexibility. Since specific APOE polymorphisms negatively impact lipid metabolism, ketone body metabolism may represent a critical nexus for future studies. Hence, understanding the relationship between the glucose-derived carbon fated for oxidation and for anabolic purposes could illuminate new therapeutic targets for mitigating cognitive decline.
Neuronal energy deficits stemming from impairments of glucose uptake could be resolved with compensatory increases in the oxidation of alternative fuel sources. While ketone bodies have been emphasized in this context, additional studies have focused on the contribution of lactate to brain metabolism and cognition. Elevations in lactate have been detected in cerebrospinal fluid (CSF) of AD patients108, and lactate has been reported to support neuronal homeostasis and aid in the recovery of synaptic function following hypoxia109,110. However, increased lactate in the central nervous system (CNS) may be attributable to a relative increased ratio of glycolysis to pyruvate oxidation in the CNS, which could also be implicated in astrocytic-derived supply of lactate to neurons111. Lactate infusion therapies have so far been unsuccessful in the restoration of cognitive function in AD patients112. While increased lactate has been observed in the blood of insulin-resistant patients113,114, lactate infusions may exacerbate insulin resistance115 and may be associated with inflammatory responses in obese patients116. A current clinical trial investigates whether exercise-induced lactate production can modulate brain metabolism, measured with flurodeoxyglucose positive emission tomography (NCT04299308).
There is little known regarding the direct effects of neurodegenerative diseases like AD on hepatic ketogenesis or ketone metabolism in the brain. At rest, AD patients have lower static levels of βOHB within red blood cells and brain parenchyma18, which could imply elevated utilization of ketones, lower cellular uptake, or reduced rates of ketogenesis; however, without tracing studies that measure metabolic turnover and rigorous flux analyses, it is difficult to make meaningful conclusions. Other conditions associated with neurodegeneration, such as spinal cord injury (SCI), promote hepatic steatosis and inflammation117 which correlates with diminutions in hepatic ketogenesis118,119, suggesting that neuronal damage does influence hepatic ketogenesis. Nevertheless, more work is needed to clarify the connection(s) between cardiometabolic diseases and neurodegeneration, as increasing evidence suggests that neurodegeneration is typically a result or exacerbating factor, rather than root cause, of aberrant substrate metabolism.
Ketone bodies were initially identified and stigmatized as a pathological biomarker and mediator of type 1 diabetes mellitus120, but the therapeutic potential of ketogenic interventions was first recognized when used as a treatment for medication-resistant epilepsy17. Numerous accounts of ketogenic interventions support the beneficial effects of ketone bodies on cardiometabolic and neuronal health outcomes (summarized in Table 1). In patients with mild cognitive impairment undergoing treatment with low-carbohydrate, high-fat ketogenic diet, the concentration of circulating ketone bodies was positively correlated with memory performance121. While the mechanism(s) by which ketone bodies benefit cognitive function are still unclear, it is important to consider how the duration of ketosis and how the type of ketogenic intervention influences cognitive outcomes. The most common interventions promoting physiological ketosis are a ketogenic diet, exogenous ketone supplementation, exercise, and medications.
Ketogenic diets are high-fat, low-carbohydrate interventions with energy largely derived from fatty acid oxidation, with the spillover of excess acetyl-CoA forming ketone bodies in the liver as glucose reserves are depleted. One effect of the ketogenic diet is an energetic shift from carbohydrate to fat metabolism, which has been postulated to mediate longevity and delay age-related diseases122,123. Ketogenic diets are possibly the most popular method to promote sustained, physiological ketosis and are particularly well-studied in contexts of neurological and cardiovascular diseases. Currently, there are several clinical trials investigating the role of ketogenic diets in neurological diseases such as Parkinson’s (NCT01364545, NCT05469997) and AD (NCT04701957, NCT03690193, NCT03860792), cardiometabolic diseases such as type 2 diabetes mellitus (NCT03652649, NCT04791787) and even heart failure (NCT04235699, NCT06081543) (summarized in Table 2).
Ketogenic diets can be comprised of long-chain fatty acids (LCFAs), medium-chain fatty acids (MCFAs), or sometimes both. Ketogenic diets with MCFAs are well-tolerated in AD patients and improve cognitive function22, as do ketogenic drinks that are enriched with medium-chain triglycerides124. Higher levels of ketosis have been reported in ketogenic diets containing MCFAs than LCFAs125. Ketogenic diets increase circulating AcAc and βOHB, which are substrates for ATP synthesis in extrahepatic tissues and have important signaling roles. Acute ketogenic diet interventions (<4 wks) show anti-inflammatory effects in murine adipose tissue through reductions in Nlrp3, IL1β, and TNF-α mRNA expression, but increased inflammatory cytokine expression and accumulation of lipids in the liver126, suggesting there might be temporal or tissue-specific effects. Long-term ketogenic diet (4 mo) in mice is associated with obesity and the depletion of protective γδT cells in VAT127, again suggesting a temporal nature of benefits associated with ketogenic dietary interventions. Because of this, ketogenic diets in biomedical research are sometimes short-term or cyclical, where ketogenic diet alternates weekly with standard chow diet. One study employing a cyclical dietary strategy with ketogenic diet in middle-aged mice found improvements in survival and memory when compared to control diet, without body weight changes, and with the benefits thought to be mediated in part through intermittent upregulation of peroxisome proliferator-activated receptor α (PPARα)128.
The role of ketogenic diets in neurological diseases is well-reviewed129,130,131, but less is known regarding the utility of ketogenic diets in cardiometabolic diseases linked to cognitive outcomes. Limited studies suggest therapeutic potential in the context of MASLD132,133, while others suggest beneficial effects in obese patients through modulating appetite134, lipogenesis, and lipolysis that could result in weight loss135. Furthermore, ketogenic diets may provide protection against the neurological and cardiovascular complications typically associated with obesity. While ketogenic diets provide many benefits in the short-term, stimulating endogenous ketogenesis through high-fat dietary interventions is also associated with elevations in blood cholesterol136 and possibly poor patient compliance due to tolerance or adverse impacts associated with long-term administration137. Because of this, there is increased interest in other interventions that promote physiological ketosis, such as exercise, fasting, or exogenous supplementation.
The use of exogenous ketone supplementation and ketone body precursors, such as ketone (di)esters, has increased in popularity since they yield similar benefits as ketogenic diets in a unique physiological state where circulating insulin and glucose concentrations remain normal20. One ketone precursor, R/S-1,3-butanediol, is readily oxidized to D/L-βOHB in the liver138 and can yield physiological ketosis within 2 h of administration139. This could be important in neurodegenerative diseases, since BDH1 is stereospecific to oxidize D-βOHB20 while L-βOHB enantiomer is favored for the synthesis of fatty acids and sterols in the brain140. Interestingly, L-βOHB also binds GPR109A49 and blocks components of the NLRP3 inflammasome74, and has a longer half-life in the circulation than D-βOHB141, and thus might serve keysignaling roles that mediate neuroprotection.
The ketone monoester, R-3-hydroxybutyl R-βOHB, has been extensively studied in humans142,143,144 and rodents145,146, with findings suggesting that this ketone ester can increase circulating βOHB concentration to nearly 6 mM while concomitantly reducing caloric intake, improving insulin sensitivity, and reducing plasma cholesterol. Recently, ketone esters have been useful in lowering body weight147 and improving glucose tolerance in mouse models of obesity148, in addition to improving pathological outcomes associated with MASLD in mice fed a high-fat diet149. In humans, the metabolic effects of increasing circulating ketone body concentrations without the ketogenic diet-induced increase in fatty acids, or hormonal changes associated with these states, remains a growing area of study.
In addition to systemic metabolic benefits, ketone bodies are additionally associated with improvements in neurological and cognitive outcomes. Ketone esters have shown favorable responses in reaction times of athletes following mentally fatiguing performances150, suggesting their utility in acute neuronal processing. Unlike ketogenic diet, supplemental ketosis from exogenous sources inhibits adipose tissue lipolysis and the mobilization of fatty acids typically associated with endogenous strategies151. Administration of exogenous βOHB (sodium salt) inhibited the NLRP3 inflammasome and reduced amyloid-β plaque formation in brains of the 5XFAD mouse model of AD18. Another study used exogenous βOHB to protect hippocampal neurons from amyloid-β42 while simultaneously improving cognitive outcomes in an APP mouse model of AD19. Supplementation with ketone esters increases murine hippocampal concentration of D-βOHB and TCA cycle metabolites compared to control diet-fed mice152 and could, therefore, improve energetic efficiency in the brain. However, it remains unclear how exogenous supplementation influences neurometabolism and if there are significant differences in effectiveness comparing acute versus chronic administration.
Aerobic exercise leads to coordinated metabolic changes that maintain adequate distribution of oxygen and nutrients in tissues with augmented energy demand. The increase in ATP production is typically met through increased utilization of circulating fatty acids, lactate, and even ketone bodies153. Aerobic exercise has been shown to increase circulating fatty acids and lactate concentrations up to 2.4 mM154 and 10 mM155, respectively, while blood ketone body concentrations have been shown to increase up to 1.8 mM156 in a temporal manner referred to as post-exercise ketosis (PEK). The extent of PEK is typically regulated by training status156, nutrient intake prior to exercise157, biological sex, and exercise intensity158. Given the beneficial effects of ketone bodies on metabolic and cognitive health, a tantalizing hypothesis is that many exercise-induced health benefits could be mediated through elevations in circulating ketone bodies following bouts of aerobic exercise.
Aerobic exercise is known to improve cardiometabolic outcomes related to improving or maintaining insulin sensitivity, effectively lowering insulin levels in insulin-resistant patients and reducing risk for, or effectively treating, MASLD, reviewed in159,160, and may be in part responsible for benefits in cognitive function161,162. Due to the systemic, physiological response to exercise and transient nature of PEK, these beneficial outcomes are likely due to a combination of factors that include emerging “exerkines” such as lactate, brain-derived neurotrophic factor (BDNF), and osteocrin (reviewed in163). Exercise training increases cortical and hippocampal expression of MCTs in rats that could facilitate increased uptake and utilization of ketone bodies164, but the intermittent bouts of physiological ketosis stemming from aerobic exercise may also suggest a temporal requirement of ketosis for ketone-induced benefits. During fasted exercise, skeletal muscle has an augmented capacity for extraction and utilization of ketone bodies, increasing nearly fivefold compared to resting conditions when ketone body concentration remains below 4 mM165,166. Furthermore, while PEK is robustly observed in previously untrained individuals, PEK is attenuated in trained individuals167, likely reflecting ketogenic adaptation for greater skeletal muscle turnover. Still, ketone bodies can have effective signaling roles at concentrations observed in trained and untrained PEK (i.e. < 2.0 mM), such as increases in circulating microRNAs HAS-let7b-5p and HAS-miR-143-3p168, as well as the binding of βOHB to GPR109A49.
Key experiments are still needed to test the requirement of PEK in modulating exercise-induced neuronal benefits and the preservation of cognitive function. In addition, since PEK is transient in nature, there could be therapeutic potential in stimulating repetitive, transient bouts of ketosis on health outcomes. Nevertheless, the role of PEK in mediating metabolic and neurological benefits remains to be fully elucidated.
Certain medications promote ketosis, which might be an important component that promotes beneficial health outcomes. Emerging pharmaceutical agents that inhibit the sodium/glucose transporter 2 in the proximal renal tubule (SGLT2i), increase urinary loss of glucose, and thus reduce circulating blood glucose while increasing circulating ketone bodies in humans169 through augmented hepatic ketogenesis170. The mechanism by which this occurs is not entirely understood, but attenuated levels of insulin, or enhanced insulin sensitivity, might mediate reductions in plasma glucose and increase glucagon levels to promote mobilization of fatty acids to hepatic ketogenesis171, though this mechanism is debated172. Some of the cardiometabolic benefits associated with SGLT2i include favorable histological impacts in livers of patients with MASLD173,174,175 (comprehensively analyzed in ref. 176), favorable survival outcomes in patients with heart failure174, and reductions in hospitalization for patients with existing cardiovascular and renal disease175. However, those prescribed SGLT2i are cautioned against the risks of potential euglycemic ketoacidosis that may occur if patients concurrently use insulin, are not already significantly hyperglycemic, or have conditions that may additionally elevate ketone bodies177. Still, these agents show robustly beneficial cardiovascular outcomes that may be due, in part, to elevations in circulating ketone bodies178. There are several clinical trials employing the use of various SGLT2i in cardiometabolic conditions (NCT05782972, NCT04014192, NCT05885074), neurological conditions such as AD (NCT03801642), and even a combination of cardiometabolic and cognitive outcomes associated with T2DM (NCT04304261).
The obesity epidemic and associated cardiometabolic diseases contribute to many comorbidities that are linked through aberrant metabolic control, with increasing evidence linking obesity and other cardiometabolic diseases to the promotion of neurodegenerative diseases. Central to these conditions are insulin resistance and reductions in cellular glucose uptake, resulting in the reliance on fatty acids, lactate, and ketone bodies for energy balance. Ketone bodies are emerging as being important not only for their energetic efficiency in extrahepatic tissues, but also for non-canonical signaling roles that could mediate many of the beneficial effects of ketogenic therapies.
Future studies are needed to fully understand the connections between cardiometabolic and neurodegenerative diseases, with additional work needed to ascertain how ketone body metabolism can be leveraged via exercise, diet, or medications to promote beneficial health outcomes (summarized in Fig. 2). These strategies could begin with personalized approaches to dietary interventions, exercise protocols, and medication usage to take advantage of intermittent or chronic physiological ketosis. Furthermore, while a combination of approaches may prove to be more effective in combating or preventing adverse health outcomes, additional work is needed to ascertain metabolic, mechanistic insights regarding ketone body metabolism that govern complex diseases.
Schematic highlighting the effects of ketogenic diet, exercise, and certain medications (e.g. SGLT2i) on endogenous ketogenesis and neuronal metabolism. Exogenous ketone supplementation includes ketone esters and salts that may increase circulating ketone bodies independent of classical, hepatic ketogenesis. Neuronal uptake of ketone bodies could reduce glucose oxidation and spare glucose-derived carbon for biosynthesis. Created with BioRender.com
No datasets were generated or analysed during the current study.
Okunogbe A., Nugent R., Spencer G., Ralston J., Wilding J. Economic impacts of overweight and obesity: current and future estimates for 161 countries. BMJ Glob. Health. 2022;7: ARTN e009773. https://doi.org/10.1136/bmjgh-2022-009773.
Huai P. C., Liu J., Ye X., Li W. Q. Association of central obesity with all cause and cause-specific mortality in US adults: A prospective cohort study. Front. Cardiovasc. Medicine. 2022;9: ARTN 816144. https://doi.org/10.3389/fcvm.2022.816144.
Michalsen V. L. et al Obesity measures, metabolic health and their association with 15-year all-cause and cardiovascular mortality in the SAMINOR 1 Survey: a population-based cohort study. BMC Cardiovasc. Disord. 2021;21. ARTN 510. https://doi.org/10.1186/s12872-021-02288-9.
Fabbrini, E., Sullivan, S. & Klein, S. Obesity and nonalcoholic fatty liver disease: Biochemical, metabolic, and clinical implications. Hepatology 51, 679–689 (2010).
Article PubMed CAS Google Scholar
Ferrante, A. W. Obesity-induced inflammation: a metabolic dialogue in the language of inflammation. J. Intern. Med. 262, 408–414 (2007).
Article PubMed CAS Google Scholar
Campbell, P., Rutten, F. H., Lee, M. M., Hawkins, N. M. & Petrie, M. C. Heart failure with preserved ejection fraction: everything the clinician needs to know. Lancet 403, 1083–1092 (2024).
Article PubMed Google Scholar
de la Monte, S. M., Longato, L., Tong, M. & Wands, J. R. Insulin resistance and neurodegeneration: Roles of obesity, type 2 diabetes mellitus and non-alcoholic steatohepatitis. Curr. Opin. Invest Dr 10, 1049–1060 (2009).
Google Scholar
Dewidar, B. et al. Alterations of hepatic energy metabolism in murine models of obesity, diabetes and fatty liver diseases. EBioMedicine 94, 104714 (2023).
Article PubMed PubMed Central CAS Google Scholar
Lu, C. & Thompson, C. B. Metabolic regulation of epigenetics. Cell Metab. 16, 9–17 (2012).
Article PubMed PubMed Central CAS Google Scholar
Fan, J., Krautkramer, K. A., Feldman, J. L. & Denu, J. M. Metabolic regulation of histone post-translational modifications. ACS Chem. Biol. 10, 95–108 (2015).
Article PubMed PubMed Central CAS Google Scholar
Zhu, J. J. & Thompson, C. B. Metabolic regulation of cell growth and proliferation. Nat. Rev. Mol. Cell Biol. 20, 436–450 (2019).
Article PubMed PubMed Central CAS Google Scholar
Lee, I. H. & Finkel, T. Metabolic regulation of the cell cycle. Curr. Opin. Cell Biol. 25, 724–729 (2013).
Article PubMed CAS Google Scholar
Hall, S. E., Wastney, M. E., Bolton, T. M., Braaten, J. T. & Berman, M. Ketone body kinetics in humans: the effects of insulin-dependent diabetes, obesity, and starvation. J. Lipid Res 25, 1184–1194 (1984).
Article PubMed CAS Google Scholar
Kahn, B. B. & Flier, J. S. Obesity and insulin resistance. J. Clin. Investig. 106, 473–481 (2000).
Article PubMed PubMed Central CAS Google Scholar
Dunn, L. et al. Dysregulation of glucose metabolism is an early event in sporadic Parkinson’s disease. Neurobiol. Aging 35, 1111–1115 (2014).
Article PubMed PubMed Central CAS Google Scholar
Kim, D. Y., Park, J. & Han, I. O. Hexosamine biosynthetic pathway and -GlcNAc cycling of glucose metabolism in brain function and disease. Am. J. Physiol.-Cell Physiol. 325, C981–C998 (2023).
Article PubMed CAS Google Scholar
Wilder, R. The effect of ketonemia on the course of epilepsy. MAYO Clin. Proc. 2, 307–308 (1921).
Google Scholar
Shippy, D. C., Wilhelm, C., Viharkumar, P. A., Raife, T. J. & Ulland, T. K. beta-Hydroxybutyrate inhibits inflammasome activation to attenuate Alzheimer’s disease pathology. J. Neuroinflamm. 17, 280 (2020).
Article CAS Google Scholar
Yin, J. X. et al. Ketones block amyloid entry and improve cognition in an Alzheimer’s model. Neurobiol. Aging 39, 25–37 (2016).
Article PubMed CAS Google Scholar
Puchalska, P. & Crawford, P. A. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 25, 262–284 (2017). PMCID: PMC5313038.
Article PubMed PubMed Central CAS Google Scholar
Kivimäki M. et al. Physical inactivity, cardiometabolic disease, and risk of dementia: an individual-participant meta-analysis. BMJ-Brit Med J. 2019;365. ARTN l1495. https://doi.org/10.1136/bmj.l1495.
Taylor, M. K., Sullivan, D. K., Mahnken, J. D., Burns, J. M. & Swerdlow, R. H. Feasibility and efficacy data from a ketogenic diet intervention in Alzheimer’s disease. Alzheimers Dement (N. Y) 4, 28–36 (2017).
Article PubMed Google Scholar
Ota, M. et al. Effects of a medium-chain triglyceride-based ketogenic formula on cognitive function in patients with mild-to-moderate Alzheimer’s disease. Neurosci. Lett. 690, 232–236 (2019).
Article PubMed CAS Google Scholar
Hegardt, F. G. Mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase: a control enzyme in ketogenesis. Biochem J. 338, 569–582 (1999).
Article PubMed PubMed Central CAS Google Scholar
Quant, P. A., Tubbs, P. K. & Brand, M. D. Glucagon activates mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase in vivo by decreasing the extent of succinylation of the enzyme. Eur. J. Biochem 187, 169–174 (1990).
Article PubMed CAS Google Scholar
von Meyenn, F. et al. Glucagon-induced acetylation of Foxa2 regulates hepatic lipid metabolism. Cell Metab. 17, 436–447 (2013).
Article Google Scholar
Sengupta, S., Peterson, T. R., Laplante, M., Oh, S. & Sabatini, D. M. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing. Nature 468, 1100–1104 (2010).
Article PubMed CAS Google Scholar
Petersen, M. C. & Shulman, G. I. Mechanisms of insulin action and insulin resistance. Physiol. Rev. 98, 2133–2223 (2018).
Article PubMed PubMed Central CAS Google Scholar
Koliaki, C. et al. Adaptation of hepatic mitochondrial function in humans with non-alcoholic Fatty liver is lost in steatohepatitis. Cell Metab. 21, 739–746 (2015).
Article PubMed CAS Google Scholar
Ludwig, J., Viggiano, T. R., McGill, D. B. & Oh, B. J. Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. MAYO Clin. Proc. 55, 434–438 (1980).
PubMed CAS Google Scholar
Browning, J. D. & Horton, J. D. Molecular mediators of hepatic steatosis and liver injury. J. Clin. Investig. 114, 147–152 (2004).
Article PubMed PubMed Central CAS Google Scholar
Lee, S. et al. Impaired ketogenesis is associated with metabolic-associated fatty liver disease in subjects with type 2 diabetes. Front Endocrinol. 14, 1124576 (2023).
Article Google Scholar
Mey J. T. et al. beta-Hydroxybutyrate is reduced in humans with obesity-related NAFLD and displays a dose-dependent effect on skeletal muscle mitochondrial respiration in vitro. Am. J. Physiol. Endocrinol. Metab. https://doi.org/10.1152/ajpendo.00058.2020 (2020).
Hughey C. C., Puchalska P., Crawford P. A. Integrating the contributions of mitochondrial oxidative metabolism to lipotoxicity and inflammation in NAFLD pathogenesis. Biochim. et Biophys. Acta (BBA) - Mol. Cell Biol. Lipids. 2022:159209. https://doi.org/10.1016/j.bbalip.2022.159209.
Fletcher, J. A. et al. Impaired ketogenesis and increased acetyl-CoA oxidation promote hyperglycemia in human fatty liver. JCI Insight 5, e127737 (2019).
Article PubMed Google Scholar
Sunny, N. E., Parks, E. J., Browning, J. D. & Burgess, S. C. Excessive hepatic mitochondrial TCA Cycle and Gluconeogenesis in Humans with Nonalcoholic Fatty Liver Disease. Cell Metab. 14, 804–810 (2011).
Article PubMed PubMed Central CAS Google Scholar
Reaven, G. M. Pathophysiology of insulin-resistance in human-disease. Physiol. Rev. 75, 473–486 (1995).
Article PubMed CAS Google Scholar
Harrison, H. C. & Long, C. N. H. The distribution of ketone bodies in tissues. J. Biol. Chem. 133, 209–218 (1940).
Article CAS Google Scholar
Halestrap, A. P. The monocarboxylate transporter family–Structure and functional characterization. IUBMB Life 64, 1–9 (2012).
Article PubMed CAS Google Scholar
Balasse, E. O. & Fery, F. Ketone body production and disposal: effects of fasting, diabetes, and exercise. Diab./Metab. Rev. 5, 247–270 (1989).
Article CAS Google Scholar
Valente-Silva, P., Lemos, C., Köfalvi, A., Cunha, R. A. & Jones, J. G. Ketone bodies effectively compete with glucose for neuronal acetyl-CoA generation in rat hippocampal slices. NMR Biomed. 28, 1111–1116 (2015).
Article PubMed CAS Google Scholar
Sato, K. et al. Insulin, ketone bodies, and mitochondrial energy transduction. FASEB J. 9, 651–658 (1995).
Article PubMed CAS Google Scholar
Endemann, G., Goetz, P. G., Edmond, J. & Brunengraber, H. Lipogenesis from ketone bodies in the isolated perfused rat liver. Evidence for the cytosolic activation of acetoacetate. J. Biol. Chem. 257, 3434–3440 (1982).
Article PubMed CAS Google Scholar
Robinson, A. M. & Wlliamson, D. H. Utilization of D-3-hydroxy[3-14C]butyrate for lipogenesis in vivo in lactating rat mammary gland. Biochem. J. 176, 635–638 (1978).
Geelen, M. J., Lopes-Cardozo, M. & Edmond, J. Acetoacetate: a major substrate for the synthesis of cholesterol and fatty acids by isolated rat hepatocytes. FEBS Lett. 163, 269–273 (1983).
Article PubMed CAS Google Scholar
Hasegawa, S. et al. Acetoacetyl-CoA synthetase, a ketone body-utilizing enzyme, is controlled by SREBP-2 and affects serum cholesterol levels. Mol. Genet. Metab. 107, 553–560 (2012).
Article PubMed CAS Google Scholar
Hasegawa, S. et al. Acetoacetyl-CoA synthetase is essential for normal neuronal development. Biochem. Biophys. Res Commun. 427, 398–403 (2012).
Article PubMed CAS Google Scholar
Bergstrom, J. D. The lipogenic enzyme acetoacetyl-CoA synthetase and ketone body utilization for denovo lipid synthesis, a review. J. Lipid Res. 64, 100407 (2023).
Article PubMed PubMed Central CAS Google Scholar
Taggart, A. K. et al. (D)-beta-Hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. J. Biol. Chem. 280, 26649–26652 (2005).
Article PubMed CAS Google Scholar
Fu, S. P. et al. β-Hydroxybutyric acid inhibits growth hormone-releasing hormone synthesis and secretion through the GPR109A/extracellular signal-regulated 1/2 signalling pathway in the hypothalamus. J. Neuroendocrinol. 27, 212–222 (2015).
Article PubMed CAS Google Scholar
Rahman, M. et al. The beta-hydroxybutyrate receptor HCA2 activates a neuroprotective subset of macrophages. Nat. Commun. 5, 3944 (2014).
Article PubMed CAS Google Scholar
Kimura, I. et al. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc. Natl Acad. Sci. USA 108, 8030–8035 (2011).
Article PubMed PubMed Central CAS Google Scholar
Shimazu, T. et al. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339, 211–214 (2013).
Article PubMed CAS Google Scholar
Xie, Z. et al. Metabolic regulation of gene expression by Histone Lysine beta-Hydroxybutyrylation. Mol. Cell 62, 194–206 (2016).
Article PubMed PubMed Central CAS Google Scholar
Miyamoto, J. et al. Ketone body receptor GPR43 regulates lipid metabolism under ketogenic conditions. Proc. Natl Acad. Sci. USA 116, 23813–23821 (2019).
Article PubMed PubMed Central CAS Google Scholar
Fenselau, A. & Wallis, K. 3-oxo acid coenzyme A-transferase in normal and diabetic rat muscle. Biochem. J. 158, 509–512 (1976).
Article PubMed PubMed Central CAS Google Scholar
Grinblat, L., Pacheco Bolanos, L. F. & Stoppani, A. O. Decreased rate of ketone-body oxidation and decreased activity of D-3-hydroxybutyrate dehydrogenase and succinyl-CoA:3-oxo-acid CoA-transferase in heart mitochondria of diabetic rats. Biochem. J. 240, 49–56 (1986).
Article PubMed PubMed Central CAS Google Scholar
Turko, I. V., Marcondes, S. & Murad, F. Diabetes-associated nitration of tyrosine and inactivation of succinyl-CoA:3-oxoacid CoA-transferase. Am. J. Physiol. Heart Circ. Physiol. 281, H2289–H2294 (2001).
Article PubMed CAS Google Scholar
Nyenwe, E. A. & Kitabchi, A. E. The evolution of diabetic ketoacidosis: An update of its etiology, pathogenesis and management. Metabolism 65, 507–521 (2016).
Article PubMed CAS Google Scholar
Owen, O. E. et al. Brain metabolism during fasting. J. Clin. Invest 46, 1589–1595 (1967).
Article PubMed PubMed Central CAS Google Scholar
Cotter, D. G., d’Avignon, D. A., Wentz, A. E., Weber, M. L. & Crawford, P. A. Obligate role for ketone body oxidation in neonatal metabolic homeostasis. J. Biol. Chem. 286, 6902–6910 (2011). PMCID: PMC3044945.
Article PubMed PubMed Central CAS Google Scholar
Fukao, T. et al. A 6-bp deletion at the splice donor site of the first intron resulted in aberrant splicing using a cryptic splice site within exon 1 in a patient with succinyl-CoA: 3-Ketoacid CoA transferase (SCOT) deficiency. Mol. Genet. Metab. 89, 280–282 (2006).
Article PubMed CAS Google Scholar
Qiu, C. X., De Ronchi, D. & Fratiglioni, L. The epidemiology of the dementias: an update. Curr. Opin. Psychiatr. 20, 380–385 (2007).
Article Google Scholar
Xu, W. L. et al. Midlife overweight and obesity increase late-life dementia risk A population-based twin study. Neurology 76, 1568–1574 (2011).
Article PubMed PubMed Central CAS Google Scholar
Whitmer, R. A., Gunderson, E. P., Barrett-Connor, E., Quesenberry, C. P. & Yaffe, K. Obesity in middle age and future risk of dementia: a 27-year longitudinal population-based study. BMJ-Brit. Med. J. 330, 1360–1362b (2005).
Article Google Scholar
Schubert, D. Glucose metabolism and Alzheimer’s disease. Ageing Res. Rev. 4, 240–257 (2005).
Article PubMed CAS Google Scholar
Wang, Y., Chiu, E., Rosenberg, J. & Gambhir, S. S. Standardized uptake value atlas: characterization of physiological 2-deoxy-2-[18F]fluoro-D-glucose uptake in normal tissues. Mol. Imaging Biol. 9, 83–90 (2007).
Article PubMed Google Scholar
Boumezbeur, F. et al. The contribution of blood lactate to brain energy metabolism in humans measured by dynamic C nuclear magnetic resonance spectroscopy. J. Neurosci. 30, 13983–13991 (2010).
Article PubMed PubMed Central CAS Google Scholar
Ebert, D., Haller, R. G. & Walton, M. E. Energy contribution of octanoate to intact rat brain metabolism measured by 13C nuclear magnetic resonance spectroscopy. J. Neurosci. 23, 5928–5935 (2003).
Article PubMed PubMed Central CAS Google Scholar
Hefner, M., Baliga, V., Amphay, K., Ramos, D. & Hegde, V. Cardiometabolic modification of amyloid beta in Alzheimer’s disease pathology. Front. Aging Neurosci. 13, 721858 (2021).
Article PubMed PubMed Central CAS Google Scholar
Erion, J. R. et al. Obesity elicits Interleukin 1-mediated deficits in hippocampal synaptic plasticity. J. Neurosci. 34, 2618–2631 (2014).
Article PubMed PubMed Central CAS Google Scholar
Guo, D. H. et al. Visceral adipose NLRP3 impairs cognition in obesity via IL-1R1 on CX3CR1 cells. J. Clin. Investig. 130, 1961–1976 (2020).
Article PubMed PubMed Central CAS Google Scholar
Heneka, M. T. et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 493, 674 (2013).
Article PubMed CAS Google Scholar
Youm, Y. H. et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat. Med 21, 263–269 (2015).
Article PubMed PubMed Central CAS Google Scholar
Kadowaki, T. et al. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J. Clin. Investig. 116, 1784–1792 (2006).
Article PubMed PubMed Central CAS Google Scholar
Ng R. C. L. et al. Chronic adiponectin deficiency leads to Alzheimer’s disease-like cognitive impairments and pathologies through AMPK inactivation and cerebral insulin resistance in aged mice. Mol. Neurodegener. 2016;11. ARTN 71. https://doi.org/10.1186/s13024-016-0136-x.
Bloemer J. et al. Adiponectin knockout mice display cognitive and synaptic deficits. Front. Endocrinol. 2019;10. ARTN 819. https://doi.org/10.3389/fendo.2019.00819.
Jeon, B. T. et al. Resveratrol attenuates obesity-associated peripheral and central inflammation and improves memory deficit in mice fed a high-fat diet. Diabetes 61, 1444–1454 (2012).
Article PubMed PubMed Central CAS Google Scholar
So S. W., Fleming K. M., Nixon J. P. & Butterick T. A. Early life obesity increases neuroinflammation, amyloid beta deposition, and cognitive decline in a mouse model of Alzheimer’s disease. Nutrients. 2023;15. https://doi.org/10.3390/nu15112494.
Morris, J. K. et al. Cognitively impaired elderly exhibit insulin resistance and no memory improvement with infused insulin. Neurobiol. Aging 39, 19–24 (2016).
Article PubMed CAS Google Scholar
Baker, L. D. et al. Insulin resistance and Alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with prediabetes or early type 2 diabetes. Arch. Neurol. 68, 51–57 (2011).
Article PubMed Google Scholar
Mosconi, L., Pupi, A. & De Leon, M. J. Brain glucose hypometabolism and oxidative stress in preclinical Alzheimer’s disease. Ann. Ny. Acad. Sci. 1147, 180–195 (2008).
Article PubMed CAS Google Scholar
Gordon, B. A. et al. Spatial patterns of neuroimaging biomarker change in individuals from families with autosomal dominant Alzheimer’s disease: a longitudinal study. Lancet Neurol. 17, 241–250 (2018).
Article PubMed PubMed Central Google Scholar
Cunnane, S. et al. Brain fuel metabolism, aging, and Alzheimer’s disease. Nutrition 27, 3–20 (2011).
Article PubMed CAS Google Scholar
Mu Y. L., Gage F. H. Adult hippocampal neurogenesis and its role in Alzheimer’s disease. Mol. Neurodegener. 2011;6. Artn 85. https://doi.org/10.1186/1750-1326-6-85.
Willette, A. A. et al. Insulin resistance predicts brain amyloid deposition in late middle-aged adults. Alzheimers Dement. 11, 504–510 e501 (2015).
Article PubMed Google Scholar
Kim, B., Elzinga, S. E., Henn, R. E., McGinley, L. M. & Feldman, E. L. The effects of insulin and insulin-like growth factor I on amyloid precursor protein phosphorylation in in vitro and in vivo models of Alzheimer’s disease. Neurobiol. Dis. 132, 104541 (2019).
Article PubMed PubMed Central CAS Google Scholar
Puig, K. L., Floden, A. M., Adhikari, R., Golovko, M. Y. & Combs, C. K. Amyloid precursor protein and proinflammatory changes are regulated in brain and adipose tissue in a murine model of high fat diet-induced obesity. PLoS One 7, e30378 (2012).
Article PubMed PubMed Central CAS Google Scholar
Vingtdeux, V. et al. Phosphorylation of amyloid precursor carboxy-terminal fragments enhances their processing by a gamma-secretase-dependent mechanism. Neurobiol. Dis. 20, 625–637 (2005).
Article PubMed CAS Google Scholar
Takashima, A. GSK-3 is essential in the pathogenesis of Alzheimer’s disease. J. Alzheimers Dis. 9, 309–317 (2006).
Article PubMed CAS Google Scholar
Pettigrew, C. & Soldan, A. Defining cognitive reserve and implications for cognitive aging. Curr. Neurol. Neurosci. Rep. 19, 1 (2019).
Article PubMed PubMed Central Google Scholar
Stern, Y. Cognitive reserve in ageing and Alzheimer’s disease. Lancet Neurol. 11, 1006–1012 (2012).
Article PubMed PubMed Central Google Scholar
Whalley, L. J., Deary, I. J., Appleton, C. L. & Starr, J. M. Cognitive reserve and the neurobiology of cognitive aging. Ageing Res. Rev. 3, 369–382 (2004).
Article PubMed Google Scholar
Hammond, T. C. et al. beta-amyloid and tau drive early Alzheimer’s disease decline while glucose hypometabolism drives late decline. Commun. Biol. 3, 352 (2020).
Article PubMed PubMed Central CAS Google Scholar
Niccoli, T. et al. Increased glucose transport into neurons rescues Aβ Toxicity in (vol 26, pg 2291, 2016). Curr. Biol. 26, 2550–2550 (2016).
Article PubMed PubMed Central CAS Google Scholar
Andersen, J. V. et al. Alterations in cerebral cortical glucose and glutamine metabolism precedes amyloid plaques in the APPswe/PSEN1dE9 mouse model of Alzheimer’s disease. Neurochem Res 42, 1589–1598 (2017).
Article PubMed CAS Google Scholar
Andersen, J. V. et al. Hippocampal disruptions of synaptic and astrocyte metabolism are primary events of early amyloid pathology in the 5xFAD mouse model of Alzheimer’s disease. Cell Death Dis. 12, 954 (2021).
Article PubMed PubMed Central CAS Google Scholar
Westi, E. W., Andersen, J. V. & Aldana, B. I. Using stable isotope tracing to unravel the metabolic components of neurodegeneration: Focus on neuron-glia metabolic interactions. Neurobiol. Dis. 182, 106145 (2023).
Article PubMed CAS Google Scholar
Williams H. C. et al. alters glucose flux through central carbon pathways in astrocytes. Neurobiol. Disease. 2020;136. ARTN 104742. https://doi.org/10.1016/j.nbd.2020.104742.
Huebbe, P. et al. APOE genotype regulates body weight and fatty acid utilization-Studies in gene-targeted replacement mice. Mol. Nutr. Food Res. 59, 334–343 (2015).
Conway, V. et al. Apolipoprotein E isoforms disrupt long-chain fatty acid distribution in the plasma, the liver and the adipose tissue of mice. Prostaglandins Leukot. Ess. Fat. Acids 91, 261–267 (2014).
Article CAS Google Scholar
Arbones-Mainar, J. M. et al. Metabolic shifts toward fatty-acid usage and increased thermogenesis are associated with impaired adipogenesis in mice expressing human APOE4. Int J. Obes. (Lond.) 40, 1574–1581 (2016).
Article PubMed CAS Google Scholar
Jones N. S., Watson K. Q. & Rebeck G. W. Metabolic disturbances of a high-fat diet are dependent on APOE genotype and sex. eNeuro. 2019;6. https://doi.org/10.1523/eneuro.0267-19.2019.
Burke, J. R. & Roses, A. D. Genetics of Alzheimer’s disease. Int J. Neurol. 25-26, 41–51 (1991).
PubMed Google Scholar
Corder, E. H. et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261, 921–923 (1993).
Article PubMed CAS Google Scholar
Roses, A. D. Apolipoprotein E is a relevant susceptibility gene that affects the rate of expression of Alzheimer’s disease. Neurobiol. Aging 15, S165–S167 (1994).
Article PubMed Google Scholar
Reger, M. A. et al. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-beta in memory-impaired older adults. J. Alzheimers Dis. 13, 323–331 (2008).
Article PubMed PubMed Central CAS Google Scholar
Liguori, C. et al. Cerebrospinal fluid lactate levels and brain [18F]FDG PET hypometabolism within the default mode network in Alzheimer’s disease. Eur. J. Nucl. Med. Mol. Imaging. 43, 2040–2049 (2016).
Article PubMed CAS Google Scholar
Schurr, A., Payne, R. S., Miller, J. J. & Rigor, B. M. Brain lactate, not glucose, fuels the recovery of synaptic function from hypoxia upon reoxygenation: An in vitro study. Brain Res. 744, 105–111 (1997).
Article PubMed CAS Google Scholar
Schurr, A., West, C. A. & Rigor, B. M. Lactate-supported synaptic function in the rat Hippocampal slice preparation. Science 240, 1326–1328 (1988).
Article PubMed CAS Google Scholar
Chamaa F., Magistretti P. J., Fiumelli H. Astrocyte-derived lactate in stress disorders. Neurobiol. Dis. 2024:106417. https://doi.org/10.1016/j.nbd.2024.106417.
Kálmán, J. et al. Lactate infusion fails to improve semantic categorization in Alzheimer’s disease. Brain Res. Bull. 65, 533–539 (2005).
Article PubMed Google Scholar
Ma Y. L. et al. Blood lactate levels are associated with an increased risk of metabolic dysfunction-associated fatty liver disease in type 2 diabetes: a real-world study. Front. Endocrinol. 2023;14. ARTN 1133991 https://doi.org/10.3389/fendo.2023.1133991.
Lovejoy, J., Newby, F. D., Gebhart, S. S. P. & Digirolamo, M. Insulin resistance in obesity is associated with elevated basal lactate levels and diminished lactate appearance following intravenous glucose and insulin. Metab.-Clin. Exp. 41, 22–27 (1992).
Article PubMed CAS Google Scholar
Vettor, R. et al. Lactate infusion in anesthetized rats produces insulin resistance in heart and skeletal muscles. Metab.-Clin. Exp. 46, 684–690 (1997).
Article PubMed CAS Google Scholar
Lin, Y. J. et al. Lactate is a key mediator that links obesity to insulin resistance via modulating cytokine production from adipose tissue. Diabetes 71, 637–652 (2022).
Article PubMed CAS Google Scholar
Sauerbeck, A. D. et al. Spinal cord injury causes chronic liver pathology in rats. J. Neurotraum. 32, 159–169 (2015).
Article Google Scholar
Sun, X. F. et al. Liver-derived ketogenesis via overexpressing HMGCS2 promotes the recovery of spinal cord injury. Adv. Biol-Ger, (2023).
Eisenberg, D. et al. Interaction between increasing body mass index and spinal cord injury to the probability of developing a diagnosis of nonalcoholic fatty liver disease. Obes. Sci. Pract. 9, 253–260 (2023).
Article PubMed Google Scholar
ROSENBLOOM, J. The acetone bodies in diabetes mellitus: influence of low and high protein intake on the excretion of acetone, diacetic acid and beta-oxybutyric acid. J. Am. Med. Assoc. LXV, 1715–1717 (1915).
Article Google Scholar
Krikorian, R. et al. Dietary ketosis enhances memory in mild cognitive impairment. Neurobiol. Aging 33, 425.e419–425.e427 (2012).
Article Google Scholar
Roberts, M. N. et al. A ketogenic diet extends longevity and healthspan in adult mice. Cell Metab. 26, 539–546.e535 (2017).
Article PubMed PubMed Central CAS Google Scholar
Mujica-Parodi, L. R. et al. Diet modulates brain network stability, a biomarker for brain aging, in young adults. Proc. Natl Acad. Sci. USA 117, 6170–6177 (2020).
Article PubMed PubMed Central CAS Google Scholar
Fortier, M. et al. A ketogenic drink improves brain energy and some measures of cognition in mild cognitive impairment. Alzheimers Dement. 15, 625–634 (2019).
Article PubMed Google Scholar
Likhodii, S. S. et al. Dietary fat, ketosis, and seizure resistance in rats on the ketogenic diet. Epilepsia 41, 1400–1410 (2000).
Article PubMed CAS Google Scholar
Asrih M., Altirriba J., Rohner-Jeanrenaud F. & Jornayvaz F. R. Ketogenic diet impairs FGF21 signaling and promotes differential inflammatory responses in the liver and white adipose tissue. Plos One. 2015;10: ARTN e0126364 https://doi.org/10.1371/journal.pone.0126364.
Goldberg, E. L. et al. Ketogenesis activates metabolically protective γδ T cells in visceral adipose tissue. Nat. Metab. 2, 50–61 (2020).
Article PubMed PubMed Central CAS Google Scholar
Newman, J. C. et al. Ketogenic diet reduces midlife mortality and improves memory in aging mice. Cell Metab. 26, 547–557.e548 (2017).
Article PubMed PubMed Central CAS Google Scholar
Yang H. J., Shan W., Zhu F., Wu J. P., Wang Q. Ketone bodies in neurological diseases: focus on neuroprotection and underlying mechanisms. Front. Neurol. 2019;10. ARTN 585. https://doi.org/10.3389/fneur.2019.00585.
Koppel, S. J. & Swerdlow, R. H. Neuroketotherapeutics: A modern review of a century-old therapy. Neurochem. Int. 117, 114–125 (2018).
Article PubMed CAS Google Scholar
Murugan, M. & Boison, D. Ketogenic diet, neuroprotection, and antiepileptogenesis. Epilepsy Res. 167, 106444 (2020).
Article PubMed PubMed Central CAS Google Scholar
Browning, J. D. et al. Short-term weight loss and hepatic triglyceride reduction: evidence of a metabolic advantage with dietary carbohydrate restriction. Am. J. Clin. Nutr. 93, 1048–1052 (2011).
Article PubMed PubMed Central CAS Google Scholar
Foster, G. D. et al. Weight and metabolic outcomes after 2 years on a low-carbohydrate versus low-fat diet: a randomized trial. Ann. Intern. Med. 153, 147–157 (2010).
Article PubMed PubMed Central Google Scholar
Laeger, T., Metges, C. C. & Kuhla, B. Role of β-hydroxybutyric acid in the central regulation of energy balance. Appetite 54, 450–455 (2010).
Article PubMed CAS Google Scholar
Dashti, H. M. et al. Long-term effects of a ketogenic diet in obese patients. Exp. Clin. Cardiol. 9, 200–205 (2004).
PubMed PubMed Central CAS Google Scholar
Kwiterovich, P. O. Jr., Vining, E. P., Pyzik, P., Skolasky R, Jr. & Freeman JM. Effect of a high-fat ketogenic diet on plasma levels of lipids, lipoproteins, and apolipoproteins in children. JAMA 290, 912–920 (2003).
Nelson, A. B., Queathem, E. D., Puchalska, P. & Crawford, P. A. Metabolic messengers: ketone bodies. Nat. Metab. 5, 2062–2074 (2023).
Article PubMed CAS Google Scholar
Desrochers, S., David, F., Garneau, M., Jette, M. & Brunengraber, H. Metabolism of R- and S-1,3-butanediol in perfused livers from meal-fed and starved rats. Biochem. J. 285, 647–653 (1992).
Article PubMed PubMed Central CAS Google Scholar
D’Agostino, D. P. et al. Therapeutic ketosis with ketone ester delays central nervous system oxygen toxicity seizures in rats. Am. J. Physiol. - Regul. Integr. Comp. Physiol. 304, R829–R836 (2013).
Article PubMed Google Scholar
Webber, R. J. & Edmond, J. Utilization of L(+)-3-hydroxybutyrate, D(-)-3-hydroxybutyrate, acetoacetate, and glucose for respiration and lipid synthesis in the 18-day-old rat. J. Biol. Chem. 252, 5222–5226 (1977).
Article PubMed CAS Google Scholar
Desrochers, S. et al. Metabolism of (R,S)-1,3-butanediol acetoacetate esters, potential parenteral and enteral nutrients in conscious pigs. Am. J. Physiol. 268, E660–E667 (1995).
PubMed CAS Google Scholar
Clarke, K. et al. Kinetics, safety and tolerability of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate in healthy adult subjects. Regul. Toxicol. Pharm. 63, 401–408 (2012).
Article CAS Google Scholar
Shivva, V. et al. The population pharmacokinetics of D-β-hydroxybutyrate following administration of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate. AAPS J. 18, 678–688 (2016).
Article PubMed PubMed Central CAS Google Scholar
Monteyne A. J. et al. A ketone monoester drink reduces postprandial blood glucose concentrations in adults with type 2 diabetes: a randomised controlled trial. Diabetologia. 2024. https://doi.org/10.1007/s00125-024-06122-7.
Kashiwaya, Y. et al. A ketone ester diet increases brain malonyl-CoA and Uncoupling proteins 4 and 5 while decreasing food intake in the normal Wistar Rat. J. Biol. Chem. 285, 25950–25956 (2010).
Article PubMed PubMed Central CAS Google Scholar
Veech R. L. Ketone esters increase brown fat in mice and overcome insulin resistance in other tissues in the rat. In: Ann.N. Y. Acad. Sci. 2013:42–48.
Deemer, S. E. et al. Exogenous dietary Ketone Ester decreases body weight and adiposity in mice housed at thermoneutrality. Obesity 28, 1447–1455 (2020).
Article PubMed CAS Google Scholar
Dakhili, S. A. T. et al. Ketone ester administration improves glycemia in obese mice. Am. J. Physiol.-Cell Physiol. 325, C750–C757 (2023).
Article CAS Google Scholar
Moore, M. P. et al. A dietary ketone ester mitigates histological outcomes of NAFLD and markers of fibrosis in high-fat diet fed mice. Am. J. Physiol. Gastrointest. Liver Physiol. 320, G564–G572 (2021).
Article PubMed PubMed Central CAS Google Scholar
Quinones M. D. & Lemon P. W. R. Ketone Ester supplementation improves some aspects of cognitive function during a simulated soccer match after induced mental fatigue. Nutrients. 2022;14. https://doi.org/10.3390/nu14204376.
Margolis L. M., Pasiakos S. M. & Howard E. E. High-fat ketogenic diets and ketone monoester supplements differentially affect substrate metabolism during aerobic exercise. Am. J. Physiol. Cell Physiol. 2023. https://doi.org/10.1152/ajpcell.00359.2023.
Pawlosky, R. J. et al. Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer’s disease. J. Neurochem. 141, 195–207 (2017).
Article PubMed PubMed Central CAS Google Scholar
Fulghum, K. & Hill, B. G. Metabolic mechanisms of exercise-induced cardiac remodeling. Front. Cardiovasc. Med. 5, 127 (2018).
Article PubMed PubMed Central CAS Google Scholar
Rodahl, K., Miller, H. I. & Issekutz, B. Jr Plasma free fatty acids in exercise. J. Appl. Physiol. 19, 489–492 (1964).
Article PubMed CAS Google Scholar
Kaijser, L. & Berglund, B. Myocardial lactate extraction and release at rest and during heavy exercise in healthy men. Acta Physiol. Scand. 144, 39–45 (1992).
Article PubMed CAS Google Scholar
Johnson, R. H., Walton, J. L., Krebs, H. A. & Williamson, D. H. Metabolic fuels during and after severe exercise in athletes and non-athletes. Lancet 294, 452–455 (1969).
Article Google Scholar
Koeslag, J. H., Noakes, T. D. & Sloan, A. W. Post-exercise ketosis. J. Physiol. 301, 79–90 (1980).
Article PubMed PubMed Central CAS Google Scholar
Fulghum K., Collins H. E., Jones S. P. & Hill B. G. Influence of biological sex and exercise on murine cardiac metabolism. J Sport Health Sci. 2022: https://doi.org/10.1016/j.jshs.2022.06.001.
Thyfault, J. P. & Bergouignan, A. Exercise and metabolic health: beyond skeletal muscle. Diabetologia 63, 1464–1474 (2020).
Article PubMed PubMed Central Google Scholar
Cao, X. & Thyfault, J. P. Exercise drives metabolic integration between muscle, adipose and liver metabolism and protects against aging-related diseases. Exp. Gerontol. 176, 112178 (2023).
Article PubMed CAS Google Scholar
Morris J. K. et al. Aerobic exercise for Alzheimer’s disease: A randomized controlled pilot trial. Plos One. 2017;12. ARTN e0170547. https://doi.org/10.1371/journal.pone.0170547.
Baker, L. D. et al. Effects of aerobic exercise on mild cognitive impairment: a controlled trial. Arch. Neurol. 67, 71–79 (2010).
Article PubMed PubMed Central Google Scholar
Chow L. S. et al. Exerkines in health, resilience and disease. Nat. Rev. Endocrinol. 2022. https://doi.org/10.1038/s41574-022-00641-2.
Takimoto, M. & Hamada, T. Acute exercise increases brain region-specific expression of MCT1, MCT2, MCT4, GLUT1, and COX IV proteins. J. Appl Physiol. (1985) 116, 1238–1250 (2014).
Article PubMed CAS Google Scholar
Fery, F. & Balasse, E. O. Ketone body turnover during and after exercise in overnight-fasted and starved humans. Am. J. Physiol. 245, E318–E325 (1983).
PubMed CAS Google Scholar
Fery, F. & Balasse, E. O. Effect of exercise on the disposal of infused ketone bodies in humans. J. Clin. Endocrinol. Metab. 67, 245–250 (1988).
Article PubMed CAS Google Scholar
Johnson, R. H. & Walton, J. L. The effect of exercise upon acetoacetate metabolism in athletes and non‐athletes. Q. J. Exp. Physiol. Cogn. Med. Sci. 57, 73–79 (1972).
PubMed CAS Google Scholar
Cannataro, R. et al. Ketogenic diet acts on body remodeling and MicroRNAs expression profile. Microrna 8, 116–126 (2019).
Article PubMed CAS Google Scholar
Ferrannini, E. et al. Shift to fatty substrate utilization in response to Sodium-Glucose Cotransporter 2 inhibition in subjects without diabetes and patients with Type 2 Diabetes. Diabetes 65, 1190–1195 (2016).
Article PubMed CAS Google Scholar
Ferrannini, E. et al. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J. Clin. Invest. 124, 499–508 (2014).
Article PubMed PubMed Central CAS Google Scholar
Saucedo-Orozco, H., Voorrips, S. N., Yurista, S. R., de Boer, R. A. & Westenbrink, B. D. SGLT2 inhibitors and ketone metabolism in heart failure. J. Lipid Atheroscler. 11, 1–19 (2022).
Article PubMed PubMed Central CAS Google Scholar
Capozzi, M. E. et al. The limited role of glucagon for ketogenesis during fasting or in response to SGLT2 inhibition. Diabetes 69, 882–892 (2020).
Article PubMed PubMed Central CAS Google Scholar
Akuta N. et al. Favorable impact of long-term SGLT2 inhibitor for NAFLD complicated by diabetes mellitus: A 5-year follow-up study. Hepatol. Commun. https://doi.org/10.1002/hep4.2005 (2022).
Cai, R.-P., Xu, Y.-L. & Su, Q. Dapagliflozin in patients with chronic heart failure: a systematic review and meta-analysis. Cardiol. Res Pr. 2021, 6657380–6657380 (2021).
Google Scholar
Zelniker, T. A. et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet 393, 31–39 (2019).
Article PubMed CAS Google Scholar
Ong Lopez, A. M. C. & Pajimna, J. A. T. Efficacy of sodium glucose cotransporter 2 inhibitors on hepatic fibrosis and steatosis in non-alcoholic fatty liver disease: an updated systematic review and meta-analysis. Sci. Rep. 14, 2122 (2024).
Article PubMed PubMed Central CAS Google Scholar
Lupsa, B. C., Kibbey, R. G. & Inzucchi, S. E. Ketones: the double-edged sword of SGLT2 inhibitors. Diabetologia 66, 23–32 (2023).
Article PubMed CAS Google Scholar
Fitchett, D. et al. investigators E-ROt. Heart failure outcomes with empagliflozin in patients with type 2 diabetes at high cardiovascular risk: results of the EMPA-REG OUTCOME(R) trial. Eur. Heart J. 37, 1526–1534 (2016).
Article PubMed PubMed Central CAS Google Scholar
Download references
The authors are grateful for support from NIH (grants T32HL144472, DK091538, AG069781, DK007203, and HL166142). The authors thank the reviewers for insightful comments and regret that space limitations precluded inclusion of many impactful contributions. Figures were generated using Biorender.com.
Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
Kyle Fulghum, Patrycja Puchalska & Peter A. Crawford
Departments of Cell Biology and Physiology and Internal Medicine – Division of Endocrinology and Metabolism, Kansas University Medical Center, Kansas City, KS, USA
Sebastian F. Salathe, Xin Davis & John P. Thyfault
Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
Peter A. Crawford
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
K.F. and P.A.C. conceptualized manuscript and generated outline. K.F., S.F.S., X.D., J.P.T., P.P., and P.A.C. drafted and revised manuscript. K.F. and S.F.S. generated figures and tables.
Correspondence to Peter A. Crawford.
P.A.C. has served as an external consultant for Pfizer, Inc., Abbott Laboratories, Janssen Research & Development and Selah Therapeutics. All other authors declare no conflicts of interest.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
Reprints and permissions
Fulghum, K., Salathe, S.F., Davis, X. et al. Ketone body metabolism and cardiometabolic implications for cognitive health. npj Metab Health Dis 2, 29 (2024). https://doi.org/10.1038/s44324-024-00029-y
Download citation
Received: 13 April 2024
Accepted: 05 August 2024
Published: 11 October 2024
DOI: https://doi.org/10.1038/s44324-024-00029-y
Anyone you share the following link with will be able to read this content:
Sorry, a shareable link is not currently available for this article.
Provided by the Springer Nature SharedIt content-sharing initiative