Hostname: page-component-cd9895bd7-fscjk Total loading time: 0 Render date: 2024-12-22T23:17:45.544Z Has data issue: false hasContentIssue false

Low-carbohydrate and ketogenic diets: a scoping review of neurological and inflammatory outcomes in human studies and their relevance to chronic pain

Published online by Cambridge University Press:  19 April 2022

Rowena Field*
Affiliation:
Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
Tara Field
Affiliation:
The New South Wales Ministry of Health (NSW Health), Sydney, Australia
Fereshteh Pourkazemi
Affiliation:
Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
Kieron Rooney
Affiliation:
Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
*
*Corresponding author: Rowena Field, email [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Dietary restriction of carbohydrate has been demonstrated to be beneficial for nervous system dysfunction in animal models and may be beneficial for human chronic pain. The purpose of this review is to assess the impact of a low-carbohydrate/ketogenic diet on the adult nervous system function and inflammatory biomarkers to inform nutritional research for chronic pain. An electronic database search was carried out in May 2021. Publications were screened for prospective research with dietary carbohydrate intake <130 g per day and duration of ≥2 weeks. Studies were categorised into those reporting adult neurological outcomes to be extracted for analysis and those reporting other adult research outcomes. Both groups were screened again for reported inflammatory biomarkers. From 1548 studies, there were 847 studies included. Sixty-four reported neurological outcomes with 83% showing improvement. Five hundred and twenty-three studies had a different research focus (metabolic n = 394, sport/performance n = 51, cancer n = 33, general n = 30, neurological with non-neuro outcomes n = 12, or gastrointestinal n = 4). The second screen identified sixty-three studies reporting on inflammatory biomarkers, with 71% reporting a reduction in inflammation. The overall results suggest a favourable outcome on the nervous system and inflammatory biomarkers from a reduction in dietary carbohydrates. Both nervous system sensitisation and inflammation occur in chronic pain, and the results from this review indicate it may be improved by low-carbohydrate nutritional therapy. More clinical trials within this population are required to build on the few human trials that have been done.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2022. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

Adoption of a low-carbohydrate diet (LCD) or ketogenic diet (KD) is gaining popularity as a nutritional therapy for many dysfunctions, including those within the nervous system(Reference Christensen, Damsgaard and Fink-Jensen1Reference Wlodarek3). Both diets focus on the restriction of dietary carbohydrate. An LCD typically reduces intake to below 130 g per day, with a KD further reducing intake to below 50 g per day to induce nutritional ketosis(Reference Miller, Villamena and Volek4) not necessarily achieved with an LCD.

Ketosis is the elevation of ketones in the blood, with a therapeutic KD generally in the range of 0·5–3·0 mmol/l from a reduction in carbohydrate to <50 g per day(Reference Phinney, Horton and Sims5). The production of ketone bodies from fat oxidation can influence neurons in several ways: as an alternate fuel source with potential efficiency gains and reduced oxidative stress compared with glucose metabolism, as a regulator of neurotransmitter production and clearance, as a signalling molecule promoting neuroprotection, and as an epigenetic modulator to up-regulate gene expression of antioxidant, mitochondrial and anti-inflammatory functions(Reference Miller, Villamena and Volek4,Reference Fedorovich, Voronina and Waseem6,Reference Field, Field, Pourkazemi and Rooney7) . As such, ketones have a dual action, both as a fuel source assisting with bioenergetic stability and a signalling molecule regulating many pathways within the nervous system.

An LCD allows for up to 130 g of carbohydrate per day but does not necessarily produce the same level of fat oxidation and may not result in blood ketone elevation. Despite this, smaller reductions in carbohydrate intake still produce favourable metabolic consequences. These include reducing high blood glucose levels that can sensitise sensory neurons and stimulate pro-inflammatory pathways via receptors and channels on the neuron and microglia(Reference Ghosh, Castillo, Frias and Swanson8Reference Shen, Kapfhamer and Minnella11). Overall, whether the mechanism is glucose reduction, ketone formation or a synergistic relationship between them, the impact of an LCD or a KD on the nervous system is potentially favourable.

Chronic pain is characterised by increased sensitivity within the nervous system and an inflammatory profile(Reference Gerdle, Bäckryd, Falkenberg, Lundström and Ghafouri12,Reference Albrecht, Ahmed and Kettner13) and as such may respond to an LCD or a KD. There is limited research on how low-carbohydrate nutrition influences neurobiology in this population. We have recently reported relevant pre-clinical research which identified 170 animal model studies using a ketogenic diet to produce changes within the nervous system(Reference Field, Field, Pourkazemi and Rooney7). These studies investigated the mechanisms reported to occur with nutritional ketosis and how these alter nervous system function, with fourteen broad mechanistic themes emerging. These included: (a) metabolic, energetic and biochemical changes, (b) restoration of cortical excitability to homoeostatic levels, (c) gene regulation, (d) improved mitochondrial function and number, (e) reduced neuroinflammation, (f) adaptive neuroplasticity, (g) neurotransmitter and synaptic regulation, (h) reduced oxidative stress and (i) activation of signalling pathways. We have also more recently identified a significant and clinically meaningful reduction in reported pain of patients undergoing a whole-food ketogenic diet intervention in a pilot clinical trial(Reference Field, Pourkazemi and Rooney14).

We lacked the capacity to investigate potential neurological mechanisms in that pilot and as such conducted this scoping review. The primary aim of this scoping review was to identify any clinical trials that report changes in adult nervous system function and report outcomes that may be relevant to drivers of chronic pain. The secondary aim was to more broadly identify clinical trials across any research area that report changes in inflammation in response to an LCD or a KD. As such, this scoping review is a more comprehensive approach than other recent neurological reviews that limited their focus to particular diseases or focused on paediatric populations(Reference Christensen, Damsgaard and Fink-Jensen1,Reference Kraeuter, Phillips and Sarnyai15Reference Liu, Yang and Wang17) . It also considers LCD/KD research focused on other body systems or populations (such as metabolic studies or athletes) that report on how the diet influences inflammatory biomarkers.

Methods

Protocol

The primary research question for this review was: ‘How does an LCD or a KD influence the human adult nervous system?’, with the secondary research question being ‘Does an LCD or a KD influence inflammatory processes?’. Relevant items from the scoping review protocol and PRISMA-ScR checklist from the Joanna Briggs Institute(Reference Peters, Godfrey, McInerney, Aromataris and Munn18,Reference Tricco, Lillie and Zarin19) were used as the framework. Studies were reviewed in three phases. Phase one captured all publications reporting a human ketogenic diet that met the criteria for daily carbohydrate intake, research type and diet duration. Phase two sorted the retrieved papers into either research reporting adult neurological outcomes for the primary analysis or allocated the remaining studies into categories based on reported research focus. Research papers on paediatric populations that met the eligibility criteria were also grouped to allow for simple quantification but not further analysed. Phase three re-examined all adult papers from phase two for studies reporting common inflammatory biomarkers. The purpose of this broad approach was to avoid missing studies with a different research focus that also reported a neurological or inflammatory outcome, to establish the breadth of research in the neurological area within the context of all ketogenic diet research, and to quantify the extent of ketogenic research in human adult populations.

Eligibility criteria

For the first screen, studies were included if they met the following criteria: (a) human studies that reported a low-carbohydrate diet (LCD) (≤130 g per day and/or <26% of total energy intake (TEI)) or ketogenic diet (KD) (≤50 g per day and/or <15% of total energy intake)(Reference Volek, Phinney and Krauss20), (b) the LCD/KD was an intervention, (c) the diet lasted ≥14 days to allow for adaptation to the new diet and (d) the study design was prospective (observational prospective cohorts where the KD/LCD was a treatment, or experimental, longitudinal pre–post intervention trials including randomised controlled trials). Full texts were reviewed if the abstract did not contain this information. Studies were excluded if they: (a) were single case reports, (b) were retrospective reviews, (c) were cross-sectional or associational studies, (d) used exogenous ketones rather than diet, (e) were reviews, commentaries, letters or conference papers, or (f) were not in English where the required information was not in the abstract (Fig. 1).

Fig. 1. Inclusion/Exclusion flow chart.

For the second screen, studies were then sorted into three groups: (a) any research on adult populations reporting objective outcomes related to nervous system function, pain, cognitive functioning or psychological outcomes (including mood, anxiety, depression and cognitive processing, but not food cravings or hunger); (b) other adult studies which were categorised by research area: metabolic, cancer, sports/performance, general, gastrointestinal or neurological studies with non-neurological outcomes (for quantification, but did not have data extracted); or (c) paediatric papers (for quantification, but did not have data extracted). Many studies evaluate the effect of a low-carbohydrate or ketogenic diet on child and adolescent development. As this was not the focus of this study, paediatric studies were not included for data extraction to eliminate this confounding variable. We do, however, report the number of studies retrieved.

The third screen searched the two adult groups for any research that reported changes in common biomarkers associated with increased inflammation, including reactive oxygen species (ROS), oxidative stress, C-reactive protein (CRP), tumour necrosis factor alpha (TNF-α), leptin, interleukin (IL)1, 6 or 8, or decreased inflammation (adiponectin, fibroblast growth factor 21 (FGF-21), IL10). A concise summary of the PICOS criteria can be found in Table 1.

Table 1. PICOS criteria for inclusion and exclusion of studies

Information sources and search strategy

An electronic database search including Medline, EMBASE, Cochrane Library for controlled trials, AMED via OVID, CINAHL via EBSCO, Web of Science and PubMed was carried out on 24 May 2021 and included publication dates from database inception to search date. The search targeted any reference to an LCD or a KD intervention using the terms ‘ketogenic’, ‘Atkins’, ‘carbohydrate-restricted diet’ or ‘low-carbohydrate diet’. Additional searches included Google Scholar to check identified articles ‘cited by’ and ‘related articles’ links, and reference checks on identified articles with subsequent hand search for these and inclusion if they met the criteria. Retrieved references were downloaded into EndNote reference management software (Endnote X7·7·1, Thomson Reuters 2016) and then imported using Covidence systematic review software (Veritas Health Innovation, Melbourne, Australia).

Study selection and screening

Duplicates were removed, then titles and abstracts were assessed against the eligibility criteria in Covidence by two reviewers independently (R.F. and T.F.). Full texts of identified studies were then screened and categorised by RF. Studies combining paediatric and adult subjects were included if the data identified the adult results of interest separately or if the mean/median age was ≥18 years. Studies included in the second phase were imported into Endnote with their full reference and abstract. Then the inflammatory biomarker search terms were used to extract any study with these terms in any reference field.

Data items

The primary outcome of interest was changes in adult nervous system function (such as excitability, seizure frequency/duration/magnitude, nerve conduction, neuroinflammation, cognitive function or psychological changes). Additional data extracted included: author, study title, year of study, trial type, diet description, enrolment/completion numbers for the LCD/KD, comparator diet if applicable, age of participants and weight loss. These data items were repeated for inflammatory biomarkers.

Data charting process and synthesis of results

An excel sheet was used to compile extracted data items and generate a table of primary outcomes reported. Primary outcomes were reviewed and summarised by R.F. on the basis of whether the LCD/KD produced positive, neutral or negative outcomes, and whether the comparator diet also had significant positive outcomes.

Results

A systematic database search on 24 May 2021 retrieved 7876 publications after the removal of duplicates. Title and abstract screen extracted 1548 papers that were potentially eligible, with full text review excluding 701. The included studies were sorted into publications reporting a neurological outcome (n = 64), other adult research with a different focus (n = 524: metabolic n = 394, sport/performance n = 51, cancer n = 33, general n = 30, neurological with non-neuro outcomes n = 12, or gastrointestinal n = 4), or paediatric papers (paediatric neurological n = 227, other n = 32). A re-screen of all included papers excluding the paediatric populations (n = 588) for the key inflammatory terms retrieved sixty-three studies reporting on inflammatory outcomes (Fig. 1).

Characteristics of included studies

Neurological outcomes

The primary outcome of interest was results pertaining to adult nervous system function. Of the sixty-four publications reporting neurological outcomes (Table 2), twenty-two provided measures on cortical excitability (nineteen epilepsy studies(Reference Carrette, Vonck and de Herdt21Reference Zare, Okhovat and Esmaillzadeh39), one narcolepsy(Reference Husain, Yancy, Carwile, Miller and Westman40), one glucose transporter-1 deficiency syndrome(Reference Leen, Mewasingh and Verbeek41) and one general physiology(Reference Cantello, Varrasi and Tarletti42)). Additional categories included fifteen psychological publications (eleven mood and/or cognition(Reference Brinkworth, Buckley, Noakes, Clifton and Wilson43Reference Harvey, Schofield, Zinn and Thornley55), one schizophrenia(Reference Pacheco, Easterling and Pryer56) and one orexinergic system(Reference Valenzano, Polito and Trimigno57)), twelve neurodegenerative disease publications (three Alzheimer’s disease(Reference Neth, Mintz and Whitlow58Reference Taylor, Sullivan, Mahnken, Burns and Swerdlow60), three multiple sclerosis(Reference Bock, Karber and Kuhn61Reference Lee, Titcomb and Bisht63), three cognitive impairment(Reference Brandt, Buchholz and Henry-Barron64Reference Morrison, Fazeli and Gower66) and three Parkinson’s disease(Reference Koyuncu, Fidan, Toktas, Binay and Celik67Reference VanItallie, Nonas and Di Rocco69)), seven migraine publications(Reference Barborka70Reference Di Lorenzo, Pinto and Ienca76), two musculoskeletal studies (one hip osteoarthritis/pain(Reference Hall, Spiers and Knox77) and one knee osteoarthritis/pain(Reference Strath, Lukens and Jones78)), two autonomic nervous system studies (one sympathetic activation(Reference Fagerberg, Andersson and Nilsson79) and one heart rate variability(Reference Maunder, Dulson and Shaw80)), two nervous system bioenergetics papers (one cerebral glucose uptake(Reference Lobley, Johnstone and Fyfe81) and one cerebral blood flow(Reference Holsen, Hoge and Lennerz82)), one spinal cord injury paper(Reference Yarar-Fisher, Kulkarni and Li83) and one traumatic brain injury paper(Reference Rippee, Chen and Taylor84). Half of the included studies were randomised controlled trials (RCT) (n = 32), thirty were prospective single or two-arm studies, and two were case series(Reference Leen, Mewasingh and Verbeek41,Reference Barborka70) . Time on the diet ranged between 2 weeks(Reference Cantello, Varrasi and Tarletti42,Reference Pacheco, Easterling and Pryer56) and 112 weeks(Reference Cervenka, Henry, Felton, Patton and Kossoff23) (mean 18 weeks, standard deviation (SD) ± 19), with fifty-seven studies (90%) considered KD <50 g carbohydrate per day, four considered LCD(Reference Lee, Titcomb and Bisht63,Reference Hall, Spiers and Knox77,Reference Fagerberg, Andersson and Nilsson79,Reference Holsen, Hoge and Lennerz82) (between 50 g and 130 g), two unclear(Reference Pacheco, Easterling and Pryer56,Reference Barborka70) and one including LCD and KD(Reference Harvey, Schofield, Zinn and Thornley55). Overall, a total of 1729 participants commenced an LCD or a KD that reported a neurological outcome. Number of participants in trials ranged between 2 and 101, with an average of 26 ± 20. Adherence to the diet was reported between 22% and 100% (mean 79 ± 21%).

Table 2. Reported neurological outcomes from human studies utilising low-carbohydrate (LCD) or ketogenic (KD) diets n = 64

AED, anti-epileptic drugs; AHA, American Heart Association; ALZ, Alzheimer’s disease; BL, baseline; carb, carbohydrate; CR, calorie restriction; CVD, cardiovascular disease; DG, dietary guidelines; EEG, electroencephalogram; GLUT1DS, glucose transporter-1 deficiency syndrome; HC, high carbohydrate; kcal, kilocalorie; KD, ketogenic diet <50 g carbohydrate per day or 15% TEI; LCD, low-carbohydrate diet 50–130 g carbohydrate per day or 16–26% TEI; LCKD, low calorie ketogenic diet (typically between 1000 and 1800 kcal per day); MCI, mild cognitive impairment; MED, Mediterranean diet; MOH, medication overuse headache; MS, multiple sclerosis; N, No; NR, not reported; NS, not significant; NSPME, North Sea Progressive Myoclonus Epilepsy; NSSQ, Narcolepsy Symptom Status Questionnaire; PD, Parkinson’s disease; RCT, randomised controlled trial; rMSSD, root mean square of the sum of successive difference in R-R intervals; T2D, type 2 diabetes; TEI, total energy intake; TMS, transcranial; Uni, university; VLCKD, very-low-calorie ketogenic diet (typically below 1000 kcal per day); VLCnKD, very-low-calorie NON ketogenic diet; VLED, very-low-energy diet.

** Carbohydrate presented as either (a) ratio of fat: protein + carbohydrate, (b) grams per day or (c) a daily percentage.

# Comparator diet also showed significant positive outcome.

Most studies (n = 53, 83%) reported favourable neurological outcomes attributable to carbohydrate restriction. These included: reduction in seizure frequency, improved psychological mood states with reduced negative affect, better cognitive and memory functions, reduced narcoleptic sleepiness, reduced fatigue, depression and rate of multiple sclerosis relapse, reduced structural changes and improved cognitive function in Alzheimer’s disease, improved motor function in Parkinson’s disease, reduced migraine frequency and severity, reduced pain in osteoarthritis, reduced sympathetic nervous system activation, improved heart rate variability and improved recovery following spinal cord injury. Ten papers reported neutral results(Reference van Egmond, Weijenberg and van Rijn38,Reference D’Anci, Watts, Kanarek and Taylor46,Reference Iacovides, Goble, Paterson and Meiring48,Reference Makris, Darcey and Rosenbaum49,Reference Wing, Vazquez and Ryan52,Reference Rosen, Hunt, Sims and Bogardus53,Reference Phillips, Deprez and Mortimer59,Reference Lee, Titcomb and Bisht63,Reference Lobley, Johnstone and Fyfe81,Reference Rippee, Chen and Taylor84) and one trial reported a negative effect on mood(Reference Butki, Baumstark and Driver45). Three papers reported also similar or better improvements for the comparator diet(Reference Brinkworth, Buckley, Noakes, Clifton and Wilson43,Reference Brinkworth, Luscombe-Marsh and Thompson44,Reference Halyburton, Brinkworth and Wilson47) .

Energy intake varied between studies; most reported ad libitum food intake or intake formulated for weight maintenance (n = 43), eight had mild-to-moderate calorie restriction(Reference Klein, Janousek, Barber and Weissberger27,Reference Smith, Politzer, Macgarvie, McAndrews and Del Campo37,Reference Brinkworth, Buckley, Noakes, Clifton and Wilson43,Reference Brinkworth, Luscombe-Marsh and Thompson44,Reference Halyburton, Brinkworth and Wilson47,Reference Phillips, Murtagh, Gilbertson, Asztely and Lynch68,Reference Fagerberg, Andersson and Nilsson79,Reference Lobley, Johnstone and Fyfe81) , six were very low-calorie (<800 kcal per day)(Reference Wing, Vazquez and Ryan52,Reference Rosen, Hunt, Sims and Bogardus53,Reference Valenzano, Polito and Trimigno57,Reference Di Lorenzo, Coppola and Sirianni72,Reference Di Lorenzo, Pinto and Ienca76,Reference Hall, Spiers and Knox77) , three had combined protocols(Reference Mohorko, Cernelic-Bizjak and Poklar-Vatovec51,Reference Bongiovanni, Benedetto and Corvisieri71,Reference Di Lorenzo, Coppola and Bracaglia73) and did not specify energy intake(Reference Pacheco, Easterling and Pryer56,Reference Koyuncu, Fidan, Toktas, Binay and Celik67,Reference Barborka70) . Weight loss pre–post intervention was common (n = 45), but not always significant. Five studies reported no weight loss(Reference Kishk, Yousof and Ebraheim26,Reference McDonald, Henry-Barron and Felton32,Reference Sirven, Whedon and Caplan36,Reference van Egmond, Weijenberg and van Rijn38,Reference Iacovides, Goble, Paterson and Meiring48) , and nine did not report weight changes(Reference Green, Nguyen, Kaalund-Hansen, Rajakulendran and Murphy25,Reference Butki, Baumstark and Driver45,Reference Rosen, Hunt, Sims and Bogardus53,Reference Pacheco, Easterling and Pryer56,Reference Lee, Titcomb and Bisht63,Reference Brandt, Buchholz and Henry-Barron64,Reference Koyuncu, Fidan, Toktas, Binay and Celik67,Reference Barborka70,Reference Yarar-Fisher, Kulkarni and Li83) .

Inflammatory markers

The secondary outcome of interest was measures related to inflammation. A second screen of studies meeting the criteria retrieved sixty-three studies reporting on inflammatory biomarkers (Table 3). These were mostly metabolic studies (n = 52) investigating chronic lifestyle diseases such as diabetes, obesity and heart disease(Reference Valenzano, Polito and Trimigno57,Reference Al-Sarraj, Saadi, Calle, Volek and Fernandez85Reference Waldman, Smith and Lamberth135) . The remaining studies included three general population(Reference Mohorko, Cernelic-Bizjak and Poklar-Vatovec51,Reference Cipryan, Maffetone, Plews and Laursen136,Reference Cipryan, Dostal, Plews, Hofmann and Laursen137) , two cancer(Reference Hagihara, Kajimoto and Osaga138,Reference Khodabakhshi, Akbari and Mirzaei139) , two neurodegenerative disease(Reference Bock, Karber and Kuhn61,Reference Brenton, Banwell and Bergqvist62) , one auto-immune(Reference Schmidt, Christensen and Serifovski140), one musculoskeletal(Reference Strath, Lukens and Jones78), one sport/performance(Reference Paoli, Cenci and Pompei141) and one spinal cord injury(Reference Yarar-Fisher, Kulkarni and Li83). There was a large proportion of RCTs (n = 45), seventeen prospective studies and one case series(Reference Hagihara, Kajimoto and Osaga138). Time on the diet ranged from 2 weeks(Reference Bock, Karber and Kuhn61,Reference Boden, Sargrad, Homko, Mozzoli and Stein89) to 104 weeks(Reference Athinarayanan, Adams and Hallberg86,Reference Bluher, Rudich and Kloting88) (mean 16 ± 20 weeks) with fifty-three studies considered a KD, five mixed models(Reference Bluher, Rudich and Kloting88,Reference Buscemi, Verga, Tranchina, Cottone and Cerasola92,Reference Davis, Crandall and Gajavelli96,Reference Miller, Volpe, Coleman-Kelly, Gwazdauskas and Nickols-Richardson113,Reference O’Brien, Brehm and Seeley117) and five LCDs(Reference Jonasson, Guldbrand, Lundberg and Nystrom107,Reference Kerksick, Wismann-Bunn and Fogt109,Reference Zadeh, Kargarfard, Marandi and Habibi134,Reference Waldman, Smith and Lamberth135,Reference Schmidt, Christensen and Serifovski140) . The most reported inflammatory biomarker was CRP, followed by TNFα, then various ILs. Studies focused on weight loss commonly reported leptin and adiponectin.

Table 3. Reported inflammatory biomarkers from human studies utilising low-carbohydrate (LCD) or ketogenic (KD) diets, n = 63

ADA, American Diabetes Association diet; AI, anti-inflammatory; ALOX5, arachidonate 5-lipoxygenase; BDNF, brain-derived neurotrophic factor; BL, baseline; BMI, body mass index; COX, cyclooxygenase; CRP, C-reactive protein; CVD, cardiovascular disease; DG, dietary guidelines; DHA, docosahexaenoic acid; EE, energy expenditure; F, female; FGF21, fibroblast growth factor 21; HC, high-carbohydrate; hsCRP, high sensitivity C-reactive protein; HF, high fat; HUF, high unsaturated fat; ICAM-1, intracellular cellular adhesion molecule-1; IFN-γ, interferon gamma; IL, interleukin; kcal, kilocalorie; KD, ketogenic diet; kJ, kilojoule; LF, low-fat; M, male; MC, moderate-carbohydrate; MCP-1, monocyte chemotactic protein-1; MCT, medium-chain triacylglycerol; MD, Mediterranean diet; MetS, metabolic syndrome; NAFLD, non-alcoholic fatty liver disease; NR, not reported; O3, omega-3; PAI-1, plasminogen-activator inhibitor-1; ROS, reactive oxygen species; SF, saturated fat; T1D, type 1 diabetes; T2D, type 2 diabetes; TBARS, thiobarbituric acid reactive substances; TNF-α, tumour necrosis factor alpha; USF, unsaturated fat; VLCKD, very-low-calorie ketogenic diet; VLCnKD, very-low-calorie non-ketogenic diet.

** Carbohydrate presented as either (a) ratio of fat: protein + carbohydrate, (b) grams per day or (c) a daily percentage.

# Comparator diet showed significant positive outcome.

Forty-four studies reported positive outcomes (lowered inflammatory markers) from the diet, with fourteen of these also reporting benefits for the comparator diet(Reference Brinkworth, Noakes, Buckley, Keogh and Clifton91,Reference Dansinger, Gleason, Griffith, Selker and Schaefer95,Reference Gyorkos, Baker and Miutz103,Reference Johnstone, Lobley and Horgan106,Reference Keogh, Brinkworth and Noakes108,Reference Kerksick, Wismann-Bunn and Fogt109,Reference Miller, Volpe, Coleman-Kelly, Gwazdauskas and Nickols-Richardson113,Reference Noakes, Foster and Keogh116,Reference O’Brien, Brehm and Seeley117,Reference Ratliff, Mutungi, Puglisi, Volek and Fernandez122,Reference Sharman and Volek127,Reference Tay, Brinkworth, Noakes, Keogh and Clifton129,Reference Vetter, Wade and Womble131,Reference Zadeh, Kargarfard, Marandi and Habibi134) . Fifteen studies presented equivocal results(Reference Brenton, Banwell and Bergqvist62,Reference Ballard, Quann and Kupchak87,Reference Breukelman, Shaw and Basson90,Reference Buscemi, Verga, Tranchina, Cottone and Cerasola92,Reference Davis, Crandall and Gajavelli96,Reference Forsythe, Phinney and Feinman100,Reference Kong, Sun and Shi110,Reference Merra, Gratteri and De Lorenzo112,Reference Miller, Beach and Sorkin114,Reference Retterstol, Svendsen, Narverud and Holven123,Reference Stoernell, Tangney and Rockway128,Reference Volek, Sharman, Gomez, Scheett and Kraemer132,Reference Waldman, Smith and Lamberth135,Reference Hagihara, Kajimoto and Osaga138,Reference Schmidt, Christensen and Serifovski140) (no significant change in inflammatory markers), two reported mixed results(Reference Mohorko, Cernelic-Bizjak and Poklar-Vatovec51,Reference Rosenbaum, Hall and Guo124) and two studies reported negative results(Reference Johnston, Tjonn and Swan105,Reference Rankin and Turpyn121) (increased inflammatory markers). Total energy intake was similar to the neurological outcomes group; thirty-eight were ad libitum/weight maintenance studies, fifteen had mild-to-moderate calorie restriction(Reference Brinkworth, Noakes, Buckley, Keogh and Clifton91,Reference Buscemi, Verga, Tranchina, Cottone and Cerasola92,Reference Johnston, Tjonn and Swan105Reference Kerksick, Wismann-Bunn and Fogt109,Reference Kreider, Rasmussen and Kerksick111,Reference Noakes, Foster and Keogh116,Reference Ruth, Port and Shah125,Reference Sharman and Volek127,Reference Tay, Brinkworth, Noakes, Keogh and Clifton129,Reference Volek, Sharman, Gomez, Scheett and Kraemer132,Reference Zadeh, Kargarfard, Marandi and Habibi134) , seven were very low-calorie (<800 kcal per day) (Reference Valenzano, Polito and Trimigno57,Reference Castaldo, Pagano and Grimaldi93,Reference de Luis, Domingo and Izaola97,Reference Gu, Zhao and Huang102,Reference Merra, Gratteri and De Lorenzo112,Reference Monda, Polito and Lovino115,Reference Perticone, Maio and Sciacqua120) and three used mixed models(Reference Mohorko, Cernelic-Bizjak and Poklar-Vatovec51,Reference Castaldo, Rastrelli and Galdo94,Reference Gomez-Arbelaez, Crujeiras and Castro101) . Fifty-seven studies reported weight loss, two reported no weight loss(Reference Ebbeling, Feldman and Klein98,Reference Miller, Beach and Sorkin114) and four did not report a finding(Reference Yarar-Fisher, Kulkarni and Li83,Reference Breukelman, Shaw and Basson90,Reference Zadeh, Kargarfard, Marandi and Habibi134,Reference Khodabakhshi, Akbari and Mirzaei139) . Overall, a total of 2296 participants commenced an LCD or KD trial that reported an inflammatory outcome. Number of participants in trials ranged between 4 and 262, with an average of 35 ± 38. Adherence to the diet was reported between 38% and 100% (mean 87 ± 15%).

Other findings

There are a substantial number of interventional human KD and LCD trials in other research areas reported in the literature that meet the carbohydrate intake criteria and exceed 2 weeks in duration. In addition to the 61 neurological trials, other publications retrieved in the search included: 393 metabolic, 51 sport/performance, 33 cancer, 30 general, 12 neurological (reporting non-neurological outcomes) and 4 gastrointestinal (Supplementary Appendices 2–7).

Discussion

The primary aim of this scoping review was to review adult human studies that report outcomes related to the nervous system in response to an LCD or a KD. The secondary aim was to review the research that reports on changes in inflammatory biomarkers and evaluate the relevance of these findings to chronic pain. The overall results (83% of studies) suggest a favourable outcome on the nervous system from a reduction in dietary carbohydrates. Most studies utilised a KD below 50 g carbohydrate per day (fifty-eight out of sixty-four focused on neurological outcomes (91%), and fifty-five out of sixty-three focused on inflammatory biomarkers (87%)). The more moderate approach to carbohydrate restriction of the LCD also reported positive outcomes, with three out of four LCDs and both ‘unclear’ diets reporting improved neurological outcomes. Similarly, three out of four LCDs and four out of five mixed protocols also reported favourable reductions in inflammatory biomarkers.

The KD is reported to reduce nervous system excitability through ketone signalling. The actions include: activation of various ion channels (such as ATP-sensitive K+ channels) where ketones reduce excitability, modulation of neurotransmitter levels via the reduction of excitatory glutamate and/or increase in inhibitory GABA (or changes in clearance rates), and improved mitochondrial respiration and number with reduced reactive oxygen species formation(Reference Gano, Patel and Rho142,Reference Murano, Binda and Palestini143) . Most studies focused on cortical excitability (twenty-one of twenty-two publications) and reported improvements in seizure measures. Our recent scoping review of animal research using KDs(Reference Field, Field, Pourkazemi and Rooney7) identified fifty-three studies reporting on cortical/neuronal excitability, with forty-two reporting improvements that shifted nervous system excitability back towards homoeostatic levels. Increased neuronal excitability (sensitisation) is common in chronic pain presentations(Reference Nijs, George and Clauw144Reference Parker, Lewis, Rice and McNair146) and as such may also respond positively to the mechanisms outlined in the review. Additionally, the use of anti-seizure/anti-epileptic medication (such as pregabalin) is commonly prescribed for neuropathic pain, providing further evidence for common pathways being involved in both chronic pain and seizure disorders(Reference Masino and Ruskin147). Given that poor nutrition potentially triggers mechanisms responsible for driving increased nervous system sensitivity(Reference Nijs, Elma and Yilmaz148), nutritional strategies to address chronic pain are potential treatment options. A diet that potentially reduces inflammation is also an option.

KD has also been reported to reduce inflammatory loading, which is supported by the results from this review. Increased levels of inflammatory cytokines (such as CRP, TNF-α and ILs 1, 6 and 8) have been demonstrated in chronic pain presentations, with a potential link between an increase in low-grade inflammation and the development of persisting pain(Reference Gerdle, Bäckryd, Falkenberg, Lundström and Ghafouri12,Reference Schistad, Stubhaug, Furberg, Engdahl and Nielsen149,Reference Andrade, Cornips and Sommer150) . Within the nervous system, pro-inflammatory mediators can activate the microglia, increasing their number and size, altering morphology and increasing receptor expression. This results in further up-regulation of neuroinflammation leading to maladaptive synaptic plasticity and central sensitisation(Reference Ji, Nackley, Huh, Terrando and Maixner151Reference Vergne-Salle and Bertin154). In contrast, the presence of ketones and/or the suppression of glycolysis positively influence the microglia, shifting them towards anti-inflammatory phenotypes(Reference Morris, Puri and Maes155). In addition, ketone bodies acting as signalling molecules can mitigate NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activity in the microglia and central nervous system, and act as a ligand for G-protein receptors becoming class 1 and 2 histone deacetylase (HDAC) inhibitors within the brain up-regulating anti-inflammatory and antioxidant pathways(Reference Fedorovich, Voronina and Waseem6,Reference Morris, Puri and Maes155) . Inducing ketosis via a KD to mitigate inflammatory processes can be supported mechanistically(Reference Field, Field, Pourkazemi and Rooney7,Reference Morris, Puri and Carvalho156) as well as clinically from the human studies retrieved (Table 3).

All seven migraine studies utilised a KD and reported positive outcomes. There are several mechanisms relevant to migraine development that are influenced by a KD. In addition to cortical excitability and inflammation already discussed, an energy deficit from impaired glucose metabolism/transport, mitochondrial dysfunction and oxidative stress are also implicated in migraine as well as other neurodegenerative disorders(Reference Gross, Klement, Schoenen, D’Agostino and Fischer157,Reference Morris, Maes, Berk, Carvalho and Puri158) . In the context of glucose restriction, ketones are supplied to the neuron for fuel from the periphery through the blood–brain barrier, but also by fatty acid oxidation occurring in within the astrocyte(Reference Morris, Maes, Berk, Carvalho and Puri158). Ketones can meet the energy shortfall and provide bioenergetic stability. They also reduce oxidative stress by up-regulating antioxidant defence and improved mitochondrial respiration(Reference Miller, Villamena and Volek4,Reference Puchalska and Crawford159) . Whilst the presence of ketones on a KD provides a plausible mechanistic explanation for pain reduction, the absence of ketones on an LCD does not necessarily remove the potential benefit.

An LCD which does not achieve significant levels of blood ketones may also improve chronic pain through mechanisms related to lower blood glucose levels rather than ketone-mediated actions. Chronic hyperglycaemia results in the production of advanced glycation end products which activate receptors on the neuron, potentially damaging it and promoting inflammation (such as in diabetic neuropathy)(Reference Bestall, Hulse and Blackley10). It also results in hyper-insulinaemia where some cells fail to respond correctly to insulin signalling (insulin resistance) linked to the development of chronic pain such as fibromyalgia(Reference Pappolla, Manchikanti and Candido160). A reduction in glucose also reduces the NADH:NAD+ ratio, which signals a reduction in the transcription of pro-inflammatory genes in the microglia(Reference Shen, Kapfhamer and Minnella11). Interestingly, cross-sectional studies investigating the effect of glucose-lowering medication such as metformin report reduced musculoskeletal pain(Reference Carvalho-e-Silva, Harmer, Ferreira and Ferreira161). Overall, the beneficial outcomes on pain from an LCD or a KD may be a synergistic action of lowered blood glucose and increased blood ketones.

Defining the most beneficial diet for chronic pain has been the target of recent systematic reviews(Reference Field, Pourkazemi, Turton and Rooney162Reference Elma, Yilmaz and Deliens164); however no clear diet stands out(Reference Field, Pourkazemi, Turton and Rooney162). Only two studies using a low-carbohydrate approach specifically for pain were found during this review(Reference Hall, Spiers and Knox77,Reference Strath, Lukens and Jones78) . Both demonstrated improvements in pain outcomes, one using a KD(Reference Strath, Lukens and Jones78) and the other an LCD(Reference Hall, Spiers and Knox77); however, neither presented ketone levels or measured dietary carbohydrate data. A third study(Reference Field, Pourkazemi and Rooney14) published after the scoping review date also reported significant improvements in pain outcomes using a KD protocol with a significant but small rise in ketone levels and measured daily carbohydrate intake reduced to 70 g per day in the intervention group. The human trials presented support the use of a KD for a reduction in nervous system sensitivity; however, an LCD also reports benefits. It is unclear from the current research what level of carbohydrate restriction is required, whether the effect increases proportionately with carbohydrate reduction, or what diet duration length is required for favourable impacts on nervous system sensitisation. Further research using larger participant numbers is required to help answer these questions which are relevant for clinical application.

A limitation of this review is the confounding variable of weight loss. A large portion of included studies reported significant weight loss, which is also reduces inflammatory cytokines and may be also responsible for positive outcomes. A further limitation was the exclusion of extensive paediatric studies that may have provided further mechanistic evidence (such as Napolitano and colleagues(Reference Napolitano, Longo and Lucignani165) who were the first to show an increase in the antioxidant glutathione in human rather than animal models) (Supplementary Appendix 1). Despite this, an LCD or a KD is supported by mechanistic animal research as well as the human data presented here to provide plausible rationale as to how physiology might be influenced in a way to reduce pain perception.

Conclusion

This scoping review of LCD and KD research identified a large body of prospective adult human dietary intervention trials that reduced carbohydrate intake to below 130 g per day and exceeded 2 weeks in length. From these, there were sixty-four studies reporting neurological outcomes of which 83% showed improvement, and sixty-three studies reporting inflammatory biomarkers of which 71% improved. Both nervous system sensitisation and inflammation occur in chronic pain and as such may be improved by low-carbohydrate nutritional therapy. More clinical trials within this population are required to build on the few human trials that have been done.

Acknowledgements

This research did not receive any specific grant from funding agencies in the public, commercial or not-for-profit sector. The authors declare that there is no conflict of interest regarding the publication of this article.

Supplementary material

To view supplementary material for this article, please visit https://doi.org/10.1017/S0954422422000087

References

Christensen, MG, Damsgaard, J & Fink-Jensen, A (2021) Use of ketogenic diets in the treatment of central nervous system diseases: a systematic review. Nord J Psychiatry 75, 18.CrossRefGoogle ScholarPubMed
Camberos-Luna, L & Massieu, L (2020) Therapeutic strategies for ketosis induction and their potential efficacy for the treatment of acute brain injury and neurodegenerative diseases. Neurochem Int 133, 104614.CrossRefGoogle ScholarPubMed
Wlodarek, D (2019) Role of ketogenic diets in neurodegenerative diseases (Alzheimer’s disease and Parkinson’s disease). Nutrients 11, 15.CrossRefGoogle ScholarPubMed
Miller, VJ, Villamena, FA & Volek, JS (2018) Nutritional ketosis and mitohormesis: potential implications for mitochondrial function and human health. J Nutr Metab 2018, 5157645.10.1155/2018/5157645CrossRefGoogle ScholarPubMed
Phinney, SD, Horton, ES, Sims, EA, et al. (1980) Capacity for moderate exercise in obese subjects after adaptation to a hypocaloric, ketogenic diet. J Clin Invest 66, 11521161.10.1172/JCI109945CrossRefGoogle ScholarPubMed
Fedorovich, SV, Voronina, PP & Waseem, TV (2018) Ketogenic diet versus ketoacidosis: what determines the influence of ketone bodies on neurons? Neural Regen Res 13, 20602063.10.4103/1673-5374.241442CrossRefGoogle ScholarPubMed
Field, R, Field, T, Pourkazemi, F & Rooney, K (2021) Ketogenic diets and the nervous system: a scoping review of neurological outcomes from nutritional ketosis in animal studies. Nutr Res Rev, 139.Google ScholarPubMed
Ghosh, S, Castillo, E, Frias, ES & Swanson, RA (2018) Bioenergetic regulation of microglia. Glia 66, 12001212.10.1002/glia.23271CrossRefGoogle ScholarPubMed
Lam, D, Momeni, Z, Theaker, M, et al. (2018) RAGE-dependent potentiation of TRPV1 currents in sensory neurons exposed to high glucose. PLoS One 13, e0193312.CrossRefGoogle ScholarPubMed
Bestall, SM, Hulse, RP, Blackley, Z, et al. (2018) Sensory neuronal sensitisation occurs through HMGB-1/ RAGE and TRPV1 in high glucose conditions. J Cell Sci 131, jcs215939.CrossRefGoogle ScholarPubMed
Shen, Y, Kapfhamer, D, Minnella, A, et al. (2017) Bioenergetic state regulates innate inflammatory responses through the transcriptional co-repressor CtBP. Nat Commun 8, 624.CrossRefGoogle ScholarPubMed
Gerdle, B, Bäckryd, E, Falkenberg, T, Lundström, E & Ghafouri, B (2019) Changes in inflammatory plasma proteins from patients with chronic pain associated with treatment in an interdisciplinary multimodal rehabilitation program – an explorative multivariate pilot study. Scand J Pain 20, 125138.CrossRefGoogle Scholar
Albrecht, D, Ahmed, S, Kettner, N, et al. (2018) Neuroinflammation of the spinal cord and nerve roots in chronic radicular pain patients. Pain 159, 968977.CrossRefGoogle ScholarPubMed
Field, R, Pourkazemi, F & Rooney, K (2021) Effects of a low-carbohydrate ketogenic diet on reported pain, blood biomarkers and quality of life in patients with chronic pain: a pilot randomised clinical trial. Pain Med, pnab278.Google Scholar
Kraeuter, A-K, Phillips, R & Sarnyai, Z (2020) Ketogenic therapy in neurodegenerative and psychiatric disorders: from mice to men. Prog Neuropsychopharmacol Biol Psychiatry 101, 109913.10.1016/j.pnpbp.2020.109913CrossRefGoogle Scholar
Grammatikopoulou, MG, Goulis, DG, Gkiouras, K, et al. (2020) To keto or not to keto? A systematic review of randomized controlled trials assessing the effects of ketogenic therapy on Alzheimer disease. Adv Nutr 11, 15831602.CrossRefGoogle ScholarPubMed
Liu, H, Yang, Y, Wang, Y, et al. (2018) Ketogenic diet for treatment of intractable epilepsy in adults: a meta-analysis of observational studies. Epilepsia Open 3, 917.CrossRefGoogle ScholarPubMed
Peters, M, Godfrey, C, McInerney, P, et al. (2020) Chapter 11: scoping reviews. In: Aromataris, E, Munn, Z, eds. JBI Reviewers Manual. The Joanna Briggs Institute. https://reviewersmanual.joannabriggs.org/ Google Scholar
Tricco, A, Lillie, E, Zarin, W, et al. (2018) PRISMA extension for scoping reviews (PRISMA-ScR): checklist and explanation. Ann Intern Med 169, 467473.CrossRefGoogle ScholarPubMed
Volek, JS, Phinney, SD, Krauss, RM, et al. (2021) Alternative dietary patterns for Americans: low-carbohydrate diets. Nutrients 13, 3299.10.3390/nu13103299CrossRefGoogle ScholarPubMed
Carrette, E, Vonck, K, de Herdt, V, et al. (2008) A pilot trial with modified Atkins’ diet in adult patients with refractory epilepsy. Clin Neurol Neurosurg 110, 797803.10.1016/j.clineuro.2008.05.003CrossRefGoogle ScholarPubMed
Cervenka, MC, Terao, NN, Bosarge, JL, et al. (2012) E-mail management of the modified Atkins diet for adults with epilepsy is feasible and effective. Epilepsia 53, 728732.10.1111/j.1528-1167.2012.03406.xCrossRefGoogle ScholarPubMed
Cervenka, MC, Henry, BJ, Felton, EA, Patton, K & Kossoff, EH (2016) Establishing an adult epilepsy diet center: experience, efficacy and challenges. Epilepsy Behav 58, 6168.10.1016/j.yebeh.2016.02.038CrossRefGoogle ScholarPubMed
de Souza Neves, G, Dos Santos Lunardi, M, Papini Gabiatti, M, et al. (2020) Cardiometabolic risk and effectiveness of the modified Atkins ketogenic diet for adult patients with pharmacoresistant epilepsies in a middle-income country. Epilepsy Res 160, 106280.CrossRefGoogle Scholar
Green, SF, Nguyen, P, Kaalund-Hansen, K, Rajakulendran, S & Murphy, E (2020) Effectiveness, retention, and safety of modified ketogenic diet in adults with epilepsy at a tertiary-care centre in the UK. J Neurol 267, 11711178.10.1007/s00415-019-09658-6CrossRefGoogle Scholar
Kishk, NA, Yousof, HZ, Ebraheim, AM, et al. (2021) The effect of ketogenic diet escalation in adolescents and adults with drug-resistant epilepsy: a prospective study. Nutr Neurosci, 110.Google ScholarPubMed
Klein, P, Janousek, J, Barber, A & Weissberger, R (2010) Ketogenic diet treatment in adults with refractory epilepsy. Epilepsy Behav 19, 575579.CrossRefGoogle ScholarPubMed
Kossoff, EH, Rowley, H, Sinha, SR & Vining, EP (2008) A prospective study of the modified Atkins diet for intractable epilepsy in adults. Epilepsia 49, 316319.CrossRefGoogle ScholarPubMed
Kverneland, M, Selmer, KK, Nakken, KO, Iversen, PO & Tauboll, E (2015) A prospective study of the modified Atkins diet for adults with idiopathic generalized epilepsy. Epilepsy Behav 53, 197201.CrossRefGoogle ScholarPubMed
Kverneland, M, Molteberg, E, Iversen, PO, et al. (2018) Effect of modified Atkins diet in adults with drug-resistant focal epilepsy: a randomized clinical trial. Epilepsia 59, 15671576.CrossRefGoogle ScholarPubMed
Lambrechts, D, Wielders, LHP, Aldenkamp, AP, et al. (2012) The ketogenic diet as a treatment option in adults with chronic refractory epilepsy: efficacy and tolerability in clinical practice. Epilepsy Behav 23, 310314.10.1016/j.yebeh.2012.01.002CrossRefGoogle ScholarPubMed
McDonald, TJW, Henry-Barron, BJ, Felton, EA, et al. (2018) Improving compliance in adults with epilepsy on a modified Atkins diet: a randomized trial. Seizure 60, 132138.10.1016/j.seizure.2018.06.019CrossRefGoogle ScholarPubMed
McDonald, TJW, Diaz-Arias, L, Vizthum, D, et al. (2021) Six-month effects of modified Atkins diet implementation on indices of cardiovascular disease risk in adults with epilepsy. Nutr Neurosci, 110.Google ScholarPubMed
Mosek, A, Natour, H, Neufeld, MY, Shiff, Y & Vaisman, N (2009) Ketogenic diet treatment in adults with refractory epilepsy: a prospective pilot study. Seizure 18, 3033.CrossRefGoogle ScholarPubMed
Nei, M, Ngo, L, Sirven, JI & Sperling, MR (2014) Ketogenic diet in adolescents and adults with epilepsy. Seizure 23, 439442.CrossRefGoogle ScholarPubMed
Sirven, J, Whedon, B, Caplan, D, et al. (1999) The ketogenic diet for intractable epilepsy in adults: preliminary results. Epilepsia 40, 17211726.CrossRefGoogle ScholarPubMed
Smith, M, Politzer, N, Macgarvie, D, McAndrews, MP & Del Campo, M (2011) Efficacy and tolerability of the modified Atkins diet in adults with pharmacoresistant epilepsy: a prospective observational study. Epilepsia 52, 775780.CrossRefGoogle ScholarPubMed
van Egmond, ME, Weijenberg, A, van Rijn, ME, et al. (2017) The efficacy of the modified Atkins diet in North Sea progressive myoclonus epilepsy: an observational prospective open-label study. Orphanet J Rare Dis 12, 45.CrossRefGoogle ScholarPubMed
Zare, M, Okhovat, AA, Esmaillzadeh, A, et al. (2017) Modified Atkins diet in adult with refractory epilepsy: a controlled randomized clinical trial. Iran J Neurol 16, 7277.Google ScholarPubMed
Husain, AM, Yancy, WS Jr, Carwile, ST, Miller, PP & Westman, EC (2004) Diet therapy for narcolepsy. Neurology 62, 23002302.CrossRefGoogle ScholarPubMed
Leen, WG, Mewasingh, L, Verbeek, MM, et al. (2013) Movement disorders in GLUT1 deficiency syndrome respond to the modified Atkins diet. Mov Disord 28, 14391442.CrossRefGoogle ScholarPubMed
Cantello, R, Varrasi, C, Tarletti, R, et al. (2007) Ketogenic diet: electrophysiological effects on the normal human cortex. Epilepsia 48, 17561763.CrossRefGoogle ScholarPubMed
Brinkworth, GD, Buckley, JD, Noakes, M, Clifton, PM & Wilson, CJ (2009) Long-term effects of a very low-carbohydrate diet and a low-fat diet on mood and cognitive function. Arch Intern Med 169, 18731880.CrossRefGoogle Scholar
Brinkworth, GD, Luscombe-Marsh, ND, Thompson, CH, et al. (2016) Long-term effects of very low-carbohydrate and high-carbohydrate weight-loss diets on psychological health in obese adults with type 2 diabetes: randomized controlled trial. J Intern Med 280, 388397.10.1111/joim.12501CrossRefGoogle ScholarPubMed
Butki, BD, Baumstark, J & Driver, S (2003) Effects of a carbohydrate-restricted diet on affective responses to acute exercise among physically active participants. Percept Mot Skills 96, 607615.CrossRefGoogle ScholarPubMed
D’Anci, KE, Watts, KL, Kanarek, RB & Taylor, HA (2009) Low-carbohydrate weight-loss diets. Effects on cognition and mood. Appetite 52, 96103.CrossRefGoogle ScholarPubMed
Halyburton, AK, Brinkworth, GD, Wilson, CJ, et al. (2007) Low- and high-carbohydrate weight-loss diets have similar effects on mood but not cognitive performance. Am J Clin Nutr 86, 580587.CrossRefGoogle Scholar
Iacovides, S, Goble, D, Paterson, B & Meiring, RM (2019) Three consecutive weeks of nutritional ketosis has no effect on cognitive function, sleep, and mood compared with a high-carbohydrate, low-fat diet in healthy individuals: a randomized, crossover, controlled trial. Am J Clin Nutr 110, 349357.CrossRefGoogle ScholarPubMed
Makris, A, Darcey, VL, Rosenbaum, DL, et al. (2013) Similar effects on cognitive performance during high- and low-carbohydrate obesity treatment. Nutr Diabetes 3, e89.CrossRefGoogle ScholarPubMed
McClernon, FJ, Yancy, WS Jr, Eberstein, JA, Atkins, RC & Westman, EC (2007) The effects of a low-carbohydrate ketogenic diet and a low-fat diet on mood, hunger, and other self-reported symptoms. Obesity 15, 182.CrossRefGoogle Scholar
Mohorko, N, Cernelic-Bizjak, M, Poklar-Vatovec, T, et al. (2019) Weight loss, improved physical performance, cognitive function, eating behavior, and metabolic profile in a 12-week ketogenic diet in obese adults. Nutr Res 62, 6477.CrossRefGoogle Scholar
Wing, RR, Vazquez, JA & Ryan, CM (1995) Cognitive effects of ketogenic weight-reducing diets. Int J Obes 19, 811816.Google ScholarPubMed
Rosen, JC, Hunt, DA, Sims, E & Bogardus, C (1982) Comparison of carbohydrate-containing and carbohydrate-restricted hypocaloric diets in the treatment of obesity: effects on appetite and mood. Am J Clin Nutr 36, 463469.10.1093/ajcn/36.3.463CrossRefGoogle ScholarPubMed
Gyorkos, A, Baker, MH, Miutz, LN, et al. (2019) Carbohydrate-restricted diet and exercise increase brain-derived neurotrophic factor and cognitive function: a randomized crossover trial. Cureus 11, e5604.Google ScholarPubMed
Harvey, CJdC, Schofield, GM, Zinn, C & Thornley, S (2019) The effect of differing levels of carbohydrate restriction on the achievement of nutritional ketosis, mood, and symptoms of carbohydrate withdrawal in healthy adults: a randomised clinical trial. Nutrition X, 100005.CrossRefGoogle Scholar
Pacheco, A, Easterling, WS & Pryer, MW (1965) A pilot study of the ketogenic diet in schizophrenia. Am J Psychiatry 121, 11101111.CrossRefGoogle ScholarPubMed
Valenzano, A, Polito, R, Trimigno, V, et al. (2019) Effects of very low calorie ketogenic diet on the orexinergic system, visceral adipose tissue, and ROS production. Antioxidants 8, 13.10.3390/antiox8120643CrossRefGoogle ScholarPubMed
Neth, BJ, Mintz, A, Whitlow, C, et al. (2020) Modified ketogenic diet is associated with improved cerebrospinal fluid biomarker profile, cerebral perfusion, and cerebral ketone body uptake in older adults at risk for Alzheimer’s disease: a pilot study. Neurobiol Aging 86, 5463.CrossRefGoogle ScholarPubMed
Phillips, MCL, Deprez, LM, Mortimer, GMN, et al. (2021) Randomized crossover trial of a modified ketogenic diet in Alzheimer’s disease. Alzheimers Res Ther 13, 51.CrossRefGoogle ScholarPubMed
Taylor, M, Sullivan, D, Mahnken, J, Burns, J & Swerdlow, R (2018) Feasibility and efficacy data from a ketogenic diet intervention in Alzheimer’s disease. Alzheimers Dement 4, 2836.CrossRefGoogle ScholarPubMed
Bock, M, Karber, M & Kuhn, H (2018) Ketogenic diets attenuate cyclooxygenase and lipoxygenase gene expression in multiple sclerosis. EBioMedicine 2018, 293303.CrossRefGoogle Scholar
Brenton, JN, Banwell, B, Bergqvist, AGC, et al. (2019) Pilot study of a ketogenic diet in relapsing–remitting MS. Neurol Neuroimmunol Neuroinflammation 6, e565.CrossRefGoogle ScholarPubMed
Lee, JE, Titcomb, TJ, Bisht, B, et al. (2021) A modified MCT-based ketogenic diet increases plasma beta-hydroxybutyrate but has less effect on fatigue and quality of life in people with multiple sclerosis compared to a modified paleolithic diet: a waitlist-controlled, randomized pilot study. J Am Coll Nutr 40, 1325.CrossRefGoogle ScholarPubMed
Brandt, J, Buchholz, A, Henry-Barron, B, et al. (2019) Preliminary report on the feasibility and efficacy of the modified Atkins diet for treatment of mild cognitive impairment and early Alzheimer’s disease. J Alzheimers Dis 68, 969981.CrossRefGoogle ScholarPubMed
Krikorian, R, Shidler, MD, Dangelo, K, et al. (2012) Dietary ketosis enhances memory in mild cognitive impairment. Neurobiol Aging 33, 425 e419427.CrossRefGoogle ScholarPubMed
Morrison, SA, Fazeli, PL, Gower, B, et al. (2020) Cognitive effects of a ketogenic diet on neurocognitive impairment in adults ging with HIV: a pilot study. J Assoc Nurses AIDS Care 08, 08.Google Scholar
Koyuncu, H, Fidan, V, Toktas, H, Binay, O & Celik, H (2020) Effect of ketogenic diet versus regular diet on voice quality of patients with Parkinson’s disease. Acta Neurol Belg 06, 14.Google Scholar
Phillips, MCL, Murtagh, DKJ, Gilbertson, LJ, Asztely, FJS & Lynch, CDP (2018) Low-fat versus ketogenic diet in Parkinson’s disease: a pilot randomized controlled trial. Mov Disord 33, 13061314.CrossRefGoogle ScholarPubMed
VanItallie, TB, Nonas, C, Di Rocco, A, et al. (2005) Treatment of Parkinson disease with diet-induced hyperketonemia: a feasibility study. Neurology 64, 728730.CrossRefGoogle ScholarPubMed
Barborka, CJ (1930) Migraine – results of treatment by ketogenic diet in fifty cases. J Am Med Assoc 95, 18251828.CrossRefGoogle Scholar
Bongiovanni, D, Benedetto, C, Corvisieri, S, et al. (2021) Effectiveness of ketogenic diet in treatment of patients with refractory chronic migraine. Neurol Sci 42, 38653870.CrossRefGoogle ScholarPubMed
Di Lorenzo, C, Coppola, G, Sirianni, G, et al. (2015) Migraine improvement during short lasting ketogenesis: a proof-of-concept study. Eur J Neurol 22, 170177.CrossRefGoogle ScholarPubMed
Di Lorenzo, C, Coppola, G, Bracaglia, M, et al. (2016) Cortical functional correlates of responsiveness to short-lasting preventive intervention with ketogenic diet in migraine: a multimodal evoked potentials study. J Headache Pain 17, 58.10.1186/s10194-016-0650-9CrossRefGoogle Scholar
Di Lorenzo, C, Coppola, G, Di Lenola, D, et al. (2018) Efficacy of modified Atkins ketogenic diet in chronic cluster headache: an open-label, single-arm, clinical trial. Front Neurol 9, 64.CrossRefGoogle ScholarPubMed
Di Lorenzo, C, Coppola, G, Bracaglia, M, et al. (2019) A ketogenic diet normalizes interictal cortical but not subcortical responsivity in migraineurs. BMC Neurol 19, 136.CrossRefGoogle Scholar
Di Lorenzo, C, Pinto, A, Ienca, R, et al. (2019) A randomized double-blind, cross-over trial of very low-calorie diet in overweight migraine patients: a possible role for ketones? Nutrients 11, 28.CrossRefGoogle ScholarPubMed
Hall, M, Spiers, L, Knox, G, et al. (2021) Feasibility of exercise and weight management for people with hip osteoarthritis and overweight or obesity: a pilot study. Osteoarthr Cartil Open 3, 100174.CrossRefGoogle ScholarPubMed
Strath, L, Lukens, S, Jones, C, et al. (2019) The effect of low-carbohydrate and low-fat diets on pain in individuals with knee osteoarthritis. Pain Med 21, 150160.CrossRefGoogle Scholar
Fagerberg, B, Andersson, O, Nilsson, U, et al. (1984) Weight-reducing diets: role of carbohydrates on sympathetic nervous activity and hypotensive response. Int J Obes 8, 237243.Google ScholarPubMed
Maunder, E, Dulson, DK & Shaw, DM (2021) Autonomic and perceptual responses to induction of a ketogenic diet in free-living endurance athletes: a randomized, crossover trial. Int J Sports Physiol Perform, 17.Google ScholarPubMed
Lobley, GE, Johnstone, AM, Fyfe, C, et al. (2014) Glucose uptake by the brain on chronic high-protein weight-loss diets with either moderate or low amounts of carbohydrate. Br J Nutr 111, 586597.CrossRefGoogle ScholarPubMed
Holsen, LM, Hoge, WS, Lennerz, BS, et al. (2021) Diets varying in carbohydrate content differentially alter brain activity in homeostatic and reward regions in adults. J Nutr 151, 24652476.CrossRefGoogle ScholarPubMed
Yarar-Fisher, C, Kulkarni, A, Li, J, et al. (2018) Evaluation of a ketogenic diet for improvement of neurological recovery in individuals with acute spinal cord injury: a pilot, randomized safety and feasibility trial. Spinal Cord Series Cases 4, 88.CrossRefGoogle ScholarPubMed
Rippee, MA, Chen, J & Taylor, MK (2020) The ketogenic diet in the treatment of post-concussion syndrome – a feasibility study. Front Nutr 7, 160.CrossRefGoogle ScholarPubMed
Al-Sarraj, T, Saadi, H, Calle, MC, Volek, JS & Fernandez, ML (2009) Carbohydrate restriction, as a first-line dietary intervention, effectively reduces biomarkers of metabolic syndrome in Emirati adults. J Nutr 139, 16671676.CrossRefGoogle ScholarPubMed
Athinarayanan, S, Adams, R, Hallberg, S, et al. (2019) Long-term effects of a novel continuous remote care intervention including nutritional ketosis for the management of type 2 diabetes: a 2-year non-randomized clinical trial. Front Endocrinol (Lausanne) 10, 348.CrossRefGoogle ScholarPubMed
Ballard, KD, Quann, EE, Kupchak, BR, et al. (2013) Dietary carbohydrate restriction improves insulin sensitivity, blood pressure, microvascular function, and cellular adhesion markers in individuals taking statins. Nutr Res 33, 905912.CrossRefGoogle ScholarPubMed
Bluher, M, Rudich, A, Kloting, N, et al. (2012) Two patterns of adipokine and other biomarker dynamics in a long-term weight loss intervention. Diabetes Care 35, 342349.CrossRefGoogle Scholar
Boden, G, Sargrad, K, Homko, C, Mozzoli, M & Stein, TP (2005) Effect of a low-carbohydrate diet on appetite, blood glucose levels, and insulin resistance in obese patients with type 2 diabetes. Ann Intern Med 142, 403411.CrossRefGoogle ScholarPubMed
Breukelman, GJ, Shaw, BS, Basson, AK, et al. (2021) Immune function response following a low-carbohydrate, high-fat diet (LCHFD) in patients with type 2 diabetes. Asian J Sports Med 12, 111.CrossRefGoogle Scholar
Brinkworth, GD, Noakes, M, Buckley, JD, Keogh, JB & Clifton, PM (2009) Long-term effects of a very-low-carbohydrate weight loss diet compared with an isocaloric low-fat diet after 12 mo. Am J Clin Nutr 90, 2332.CrossRefGoogle ScholarPubMed
Buscemi, S, Verga, S, Tranchina, MR, Cottone, S & Cerasola, G (2009) Effects of hypocaloric very-low-carbohydrate diet vs. Mediterranean diet on endothelial function in obese women. Eur J Clin Invest 39, 339347.CrossRefGoogle ScholarPubMed
Castaldo, G, Pagano, I, Grimaldi, M, et al. (2021) Effect of very-low-calorie ketogenic diet on psoriasis patients: a nuclear magnetic resonance-based metabolomic study. J Proteome Res 20, 15091521.CrossRefGoogle ScholarPubMed
Castaldo, G, Rastrelli, L, Galdo, G, et al. (2020) Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: a proof-of-concept, single-arm, open-label clinical trial. Nutrition 74, 110757.CrossRefGoogle ScholarPubMed
Dansinger, ML, Gleason, JA, Griffith, JL, Selker, HP & Schaefer, EJ (2005) Comparison of the Atkins, Ornish, weight watchers, and zone diets for weight loss and heart disease risk reduction: a randomized trial. JAMA 293, 4353.CrossRefGoogle ScholarPubMed
Davis, NJ, Crandall, JP, Gajavelli, S, et al. (2011) Differential effects of low-carbohydrate and low-fat diets on inflammation and endothelial function in diabetes. J Diabetes 25, 371376.Google ScholarPubMed
de Luis, D, Domingo, JC, Izaola, O, et al. (2016) Effect of DHA supplementation in a very low-calorie ketogenic diet in the treatment of obesity: a randomized clinical trial. Endocrine 54, 111122.CrossRefGoogle Scholar
Ebbeling, CB, Feldman, HA, Klein, GL, et al. (2018) Effects of a low carbohydrate diet on energy expenditure during weight loss maintenance: randomized trial. BMJ 363, 114.Google ScholarPubMed
Forsythe, CE, Phinney, SD, Fernandez, ML, et al. (2008) Comparison of low fat and low carbohydrate diets on circulating fatty acid composition and markers of inflammation. Lipids 43, 6577.CrossRefGoogle ScholarPubMed
Forsythe, CE, Phinney, SD, Feinman, RD, et al. (2010) Limited effect of dietary saturated fat on plasma saturated fat in the context of a low carbohydrate diet. Lipids 45, 947962.CrossRefGoogle ScholarPubMed
Gomez-Arbelaez, D, Crujeiras, AB, Castro, AI, et al. (2018) Resting metabolic rate of obese patients under very low calorie ketogenic diet. Nutr Metab (Lond) 15, 18.CrossRefGoogle ScholarPubMed
Gu, Y, Zhao, A, Huang, F, et al. (2013) Very low carbohydrate diet significantly alters the serum metabolic profiles in obese subjects. J Proteome Res 12, 58015811.CrossRefGoogle ScholarPubMed
Gyorkos, A, Baker, MH, Miutz, LN, et al. (2019) Carbohydrate-restricted diet and high-intensity interval training exercise improve cardio-metabolic and inflammatory profiles in metabolic syndrome: a randomized crossover trial. Cureus 11, e5596.Google ScholarPubMed
Hu, T, Yao, L, Reynolds, K, et al. (2015) The effects of a low-carbohydrate diet vs. a low-fat diet on novel cardiovascular risk factors: a randomized controlled trial. Nutrients 7, 79787994.CrossRefGoogle Scholar
Johnston, CS, Tjonn, SL, Swan, PD, et al. (2006) Ketogenic low-carbohydrate diets have no metabolic advantage over nonketogenic low-carbohydrate diets. Am J Clin Nutr 83, 10551061.CrossRefGoogle ScholarPubMed
Johnstone, AM, Lobley, GE, Horgan, GW, et al. (2011) Effects of a high-protein, low-carbohydrate v. high-protein, moderate-carbohydrate weight-loss diet on antioxidant status, endothelial markers and plasma indices of the cardiometabolic profile. Br J Nutr 106, 282291.CrossRefGoogle ScholarPubMed
Jonasson, L, Guldbrand, H, Lundberg, AK & Nystrom, FH (2014) Advice to follow a low-carbohydrate diet has a favourable impact on low-grade inflammation in type 2 diabetes compared with advice to follow a low-fat diet. Ann Med 46, 182187.CrossRefGoogle Scholar
Keogh, JB, Brinkworth, GD, Noakes, M, et al. (2008) Effects of weight loss from a very-low-carbohydrate diet on endothelial function and markers of cardiovascular disease risk in subjects with abdominal obesity. Am J Clin Nutr 87, 567576.10.1093/ajcn/87.3.567CrossRefGoogle ScholarPubMed
Kerksick, CM, Wismann-Bunn, J, Fogt, D, et al. (2010) Changes in weight loss, body composition and cardiovascular disease risk after altering macronutrient distributions during a regular exercise program in obese women. Nutr J 9, 119.CrossRefGoogle ScholarPubMed
Kong, Z, Sun, S, Shi, Q, et al. (2020) Short-term ketogenic diet improves abdominal obesity in overweight/obese Chinese young females. Front Physiol 11, 856.CrossRefGoogle ScholarPubMed
Kreider, RB, Rasmussen, C, Kerksick, CM, et al. (2011) A carbohydrate-restricted diet during resistance training promotes more favorable changes in body composition and markers of health in obese women with and without insulin resistance. Phys Sportsmed 39, 2740.CrossRefGoogle ScholarPubMed
Merra, G, Gratteri, S, De Lorenzo, A, et al. (2017) Effects of very-low-calorie diet on body composition, metabolic state, and genes expression: a randomized double-blind placebo-controlled trial. Eur Rev Med Pharmacol Sci 21, 329345.Google ScholarPubMed
Miller, LE, Volpe, JJ, Coleman-Kelly, MD, Gwazdauskas, FC & Nickols-Richardson, SM (2009) Anthropometric and leptin changes in women following different dietary approaches to weight loss. Obesity 17, 199201.CrossRefGoogle ScholarPubMed
Miller, M, Beach, V, Sorkin, JD, et al. (2009) Comparative effects of three popular diets on lipids, endothelial function, and C-reactive protein during weight maintenance. J Am Diet Assoc 109, 713717.CrossRefGoogle ScholarPubMed
Monda, V, Polito, R, Lovino, A, et al. (2020) Short-term physiological effects of a very low-calorie ketogenic diet: effects on Adiponectin levels and inflammatory states. Int J Mol Sci 21, 02.CrossRefGoogle ScholarPubMed
Noakes, M, Foster, PR, Keogh, JB, et al. (2006) Comparison of isocaloric very low carbohydrate/high saturated fat and high carbohydrate/low saturated fat diets on body composition and cardiovascular risk. Nutr Metab (Lond) 3, 113.CrossRefGoogle ScholarPubMed
O’Brien, KD, Brehm, BJ, Seeley, RJ, et al. (2005) Diet-induced weight loss is associated with decreases in plasma serum amyloid a and C-reactive protein independent of dietary macronutrient composition in obese subjects. J Clin Endocrinol Metab 90, 22442249.10.1210/jc.2004-1011CrossRefGoogle ScholarPubMed
Paoli, A, Moro, T, Bosco, G, et al. (2015) Effects of n-3 polyunsaturated fatty acids (omega-3) supplementation on some cardiovascular risk factors with a ketogenic Mediterranean diet. 1, 9961009.Google Scholar
Bosco, G, Rizzato, A, Quartesan, S, et al. (2018) Effects of the ketogenic diet in overweight divers breathing enriched air nitrox. Sci Rep 8, 2655.CrossRefGoogle ScholarPubMed
Perticone, M, Maio, R, Sciacqua, A, et al. (2019) Ketogenic diet-induced weight loss is associated with an increase in vitamin D levels in obese adults. Molecules 24, 09.CrossRefGoogle ScholarPubMed
Rankin, JW & Turpyn, AD (2007) Low carbohydrate, high fat diet increases C-reactive protein during weight loss. J Am Coll Nutr 26, 163169.CrossRefGoogle ScholarPubMed
Ratliff, J, Mutungi, G, Puglisi, MJ, Volek, JS & Fernandez, ML (2009) Carbohydrate restriction (with or without additional dietary cholesterol provided by eggs) reduces insulin resistance and plasma leptin without modifying appetite hormones in adult men. Nutr Res 29, 262268.CrossRefGoogle ScholarPubMed
Retterstol, K, Svendsen, M, Narverud, I & Holven, KB (2018) Effect of low carbohydrate high fat diet on LDL cholesterol and gene expression in normal-weight, young adults: a randomized controlled study. Atherosclerosis 279, 5261.CrossRefGoogle ScholarPubMed
Rosenbaum, M, Hall, KD, Guo, J, et al. (2019) Glucose and lipid homeostasis and inflammation in humans following an isocaloric ketogenic diet. Obesity 27, 971981.CrossRefGoogle ScholarPubMed
Ruth, MR, Port, AM, Shah, M, et al. (2013) Consuming a hypocaloric high fat low carbohydrate diet for 12 weeks lowers C-reactive protein, and raises serum adiponectin and high density lipoprotein-cholesterol in obese subjects. Metabolism 62, 17798177.CrossRefGoogle ScholarPubMed
Seshadri, P, Iqbal, N, Stern, L, et al. (2004) A randomized study comparing the effects of a low-carbohydrate diet and a conventional diet on lipoprotein subfractions and C-reactive protein levels in patients with severe obesity. Am J Med 117, 398405.CrossRefGoogle Scholar
Sharman, MJ & Volek, JS (2004) Weight loss leads to reductions in inflammatory biomarkers after a very-low-carbohydrate diet and a low-fat diet in overweight men. Clin Sci 107, 365369.CrossRefGoogle Scholar
Stoernell, CK, Tangney, CC & Rockway, SW (2008) Short-term changes in lipoprotein subclasses and C-reactive protein levels of hypertriglyceridemic adults on low-carbohydrate and low-fat diets. Nutr Res 28, 443449.CrossRefGoogle ScholarPubMed
Tay, J, Brinkworth, GD, Noakes, M, Keogh, J & Clifton, PM (2008) Metabolic effects of weight loss on a very-low-carbohydrate diet compared with an isocaloric high-carbohydrate diet in abdominally obese subjects. J Am Coll Cardiol 51, 5967.CrossRefGoogle ScholarPubMed
Tendler, D, Lin, S, Yancy, WS Jr, et al. (2007) The effect of a low-carbohydrate, ketogenic diet on nonalcoholic fatty liver disease: a pilot study. Dig Dis Sci 52, 589593.CrossRefGoogle ScholarPubMed
Vetter, ML, Wade, A, Womble, LG, et al. (2010) Effect of a low-carbohydrate diet versus a low-fat, calorie-restricted diet on adipokine levels in obese, diabetic participants. Diabetes Metab Syndr Obes 3, 357361.CrossRefGoogle ScholarPubMed
Volek, JS, Sharman, MJ, Gomez, AL, Scheett, TP & Kraemer, WJ (2003) An isoenergetic very low carbohydrate diet improves serum HDL cholesterol and triacylglycerol concentrations, the total cholesterol to HDL cholesterol ratio and postprandial pipemic responses compared with a low fat diet in normal weight, normolipidemic women. J Nutr 133, 27562761.CrossRefGoogle ScholarPubMed
Wekesa, AL, Doyle, LM, Fitzmaurice, D, et al. (2016) Influence of a low-carbohydrate diet on endothelial microvesicles in overweight women. Appl Physiol Nutr Metab 41, 522527.CrossRefGoogle ScholarPubMed
Zadeh, MAM, Kargarfard, M, Marandi, SM & Habibi, A (2018) Diets along with interval training regimes improves inflammatory & anti-inflammatory condition in obesity with type 2 diabetes subjects. J Diabetes Metab Disorders 17, 253267.CrossRefGoogle Scholar
Waldman, HS, Smith, JW, Lamberth, J, et al. (2020) A 28-day carbohydrate-restricted diet improves markers of cardiovascular disease in professional firefighters. J Strength Cond Res 34, 27852792.CrossRefGoogle ScholarPubMed
Cipryan, L, Maffetone, PB, Plews, DJ & Laursen, PB (2020) Effects of a four-week very low-carbohydrate high-fat diet on biomarkers of inflammation: non-randomised parallel-group study. Nutr Health 26, 3542.CrossRefGoogle Scholar
Cipryan, L, Dostal, T, Plews, DJ, Hofmann, P & Laursen, PB (2021) Adiponectin/leptin ratio increases after a 12-week very low-carbohydrate, high-fat diet, and exercise training in healthy individuals: a non-randomized, parallel design study. Nutr Res 87, 2230.CrossRefGoogle ScholarPubMed
Hagihara, K, Kajimoto, K, Osaga, S, et al. (2020) Promising effect of a new ketogenic diet regimen in patients with advanced cancer. Nutrients 12, 1473.CrossRefGoogle ScholarPubMed
Khodabakhshi, A, Akbari, ME, Mirzaei, HR, et al. (2021) Effects of ketogenic metabolic therapy on patients with breast cancer: a randomized controlled clinical trial. Clin Nutr 40, 751758.CrossRefGoogle Scholar
Schmidt, S, Christensen, MB, Serifovski, N, et al. (2019) Low versus high carbohydrate diet in type 1 diabetes: a 12-week randomized open-label crossover study. Diabetes Obes Metab 21, 16801688.CrossRefGoogle ScholarPubMed
Paoli, A, Cenci, L, Pompei, P, et al. (2021) Effects of two months of very low carbohydrate ketogenic diet on body composition, muscle strength, muscle area, and blood parameters in competitive natural body builders. Nutrients 13, 26.CrossRefGoogle ScholarPubMed
Gano, LB, Patel, M & Rho, JM (2014) Ketogenic diets, mitochondria, and neurological diseases. J Lipid Res 55, 22112228.CrossRefGoogle ScholarPubMed
Murano, C, Binda, A, Palestini, P, et al. (2021) Effect of the ketogenic diet in excitable tissues. Am J Physiol Cell Physiol 320, C547C553.CrossRefGoogle ScholarPubMed
Nijs, J, George, SZ, Clauw, DJ, et al. (2021) Central sensitisation in chronic pain conditions: latest discoveries and their potential for precision medicine. Lancet Rheumatol 3, e383e392.CrossRefGoogle Scholar
Neblett, R, Cohen, H, Choi, Y, et al. (2013) The Central Sensitization Inventory (CSI): establishing clinically significant values for identifying central sensitivity syndromes in an outpatient chronic pain sample. J Pain 14, 438445.CrossRefGoogle Scholar
Parker, R, Lewis, G, Rice, D & McNair, P (2016) Is motor cortical excitability altered in people with chronic pain? A systematic review and meta-analysis. Brain Stimul 9, 488500.CrossRefGoogle ScholarPubMed
Masino, SA & Ruskin, DN (2013) Ketogenic diets and pain. J Child Neurol 28, 9931001.CrossRefGoogle ScholarPubMed
Nijs, J, Elma, Ö, Yilmaz, S, et al. (2019) Nutritional neurobiology and central nervous system sensitisation: missing link in a comprehensive treatment for chronic pain? Br J Anaesth 123, 539543.CrossRefGoogle Scholar
Schistad, EI, Stubhaug, A, Furberg, A-S, Engdahl, BL & Nielsen, CS (2017) C-reactive protein and cold-pressor tolerance in the general population: the Tromsø study. Pain 158, 12801288.CrossRefGoogle ScholarPubMed
Andrade, P, Cornips, EMJ, Sommer, C, et al. (2018) Elevated inflammatory cytokine expression in CSF from patients with symptomatic thoracic disc herniations correlates with increased pain scores. Spine J 18, 23162322.CrossRefGoogle ScholarPubMed
Ji, R, Nackley, A, Huh, Y, Terrando, N & Maixner, W (2018) Neuroinflammation and central sensitization in chronic and widespread pain. Anesthesiology 129, 343366.CrossRefGoogle ScholarPubMed
Ji, R, Chamessian, A & Zhang, Y (2016) Pain regulation by non-neuronal cells and inflammation. Science 354, 572577.CrossRefGoogle ScholarPubMed
Salter, M & Stevens, B (2017) Microglia emerge as central players in brain disease. Nat Med 23, 10181027.CrossRefGoogle ScholarPubMed
Vergne-Salle, P & Bertin, P (2021) Chronic pain and neuroinflammation. Joint Bone Spine, 88, 105222.CrossRefGoogle ScholarPubMed
Morris, G, Puri, BK, Maes, M, et al. (2020) The role of microglia in neuroprogressive disorders: mechanisms and possible neurotherapeutic effects of induced ketosis. Prog Neuropsychopharmacol Biol Psychiatry 99, 109858.CrossRefGoogle ScholarPubMed
Morris, G, Puri, B, Carvalho, A, et al. (2020) Induced ketosis as a treatment for neuroprogressive disorders: food for thought? Int J Neuropsychopharmacol 23, 366384.CrossRefGoogle ScholarPubMed
Gross, EC, Klement, RJ, Schoenen, J, D’Agostino, DP & Fischer, D (2019) Potential protective mechanisms of ketone bodies in migraine prevention. Nutrients 11, 811.CrossRefGoogle ScholarPubMed
Morris, G, Maes, M, Berk, M, Carvalho, AF & Puri, BK (2020) Nutritional ketosis as an intervention to relieve astrogliosis: possible therapeutic applications in the treatment of neurodegenerative and neuroprogressive disorders. Eur Psychiatry 63, e8.CrossRefGoogle ScholarPubMed
Puchalska, P & Crawford, PA (2017) Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab 25, 262284.CrossRefGoogle ScholarPubMed
Pappolla, MA, Manchikanti, L, Candido, KD, et al. (2021) Insulin resistance is associated with central pain in patients with fibromyalgia. Pain Physician 24, 175184.Google ScholarPubMed
Carvalho-e-Silva, AP, Harmer, AR, Ferreira, ML & Ferreira, PH (2021) The effect of the anti-diabetic drug metformin on musculoskeletal pain: a cross-sectional study with 21,889 individuals from the UK Biobank. Eur J Pain 25, 2641273.CrossRefGoogle ScholarPubMed
Field, R, Pourkazemi, F, Turton, J & Rooney, K (2020) Dietary interventions are beneficial for patients with chronic pain: a systematic review with meta-analysis. Pain Med 22, 694714.CrossRefGoogle Scholar
Brain, K, Burrows, T, Rollo, M, et al. (2018) A systematic review and meta-analysis of nutrition interventions for chronic noncancer pain. J Hum Nutr Diet 32, 198225.CrossRefGoogle ScholarPubMed
Elma, Ö, Yilmaz, S, Deliens, T, et al. (2020) Do nutritional factors interact with chronic musculoskeletal pain? A systematic review. J Clin Med 9, 702.CrossRefGoogle ScholarPubMed
Napolitano, A, Longo, D, Lucignani, M, et al. (2020) The ketogenic diet increases in vivo glutathione levels in patients with epilepsy. Metabolites 10, 10.CrossRefGoogle ScholarPubMed
Tay, J, Zajac, IT, Thompson, CH, et al. (2016) A randomised-controlled trial of the effects of very low-carbohydrate and high-carbohydrate diets on cognitive performance in patients with type 2 diabetes. Br J Nutr 116, 17451753.CrossRefGoogle Scholar
Harvey, CC, Schofield, GM, Zinn, C, et al. (2019) Low-carbohydrate diets differing in carbohydrate restriction improve cardiometabolic and anthropometric markers in healthy adults: a randomised clinical trial. PeerJ 7, e6273.CrossRefGoogle ScholarPubMed
Choi, IY, Piccio, L, Childress, P, et al. (2016) A diet mimicking fasting promotes regeneration and reduces autoimmunity and multiple sclerosis symptoms. Cell Rep 15, 21362146.CrossRefGoogle ScholarPubMed
Shaw, DM, Merien, F, Braakhuis, A, Maunder, E & Dulson, DK (2019) Effect of a ketogenic diet on submaximal exercise capacity and efficiency in runners. Med Sci Sports Exerc 51, 21352146.CrossRefGoogle ScholarPubMed
Shai, I, Schwarzfuchs, D, Henkin, Y, et al. (2008) Weight loss with a low-carbohydrate, mediterranean, or low-fat diet. N Engl J Med 359, 229241.CrossRefGoogle ScholarPubMed
Davis, NJ, Tomuta, N, Schechter, C, et al. (2009) Comparative study of the effects of a 1-year dietary intervention of a low-carbohydrate diet versus a low-fat diet on weight and glycemic control in type 2 diabetes. Diabetes Care 32, 11471152.CrossRefGoogle ScholarPubMed
Volek, JS, Phinney, SD, Forsythe, CE, et al. (2009) Carbohydrate restriction has a more favorable impact on the metabolic syndrome than a low fat diet. Lipids 44, 297309.CrossRefGoogle Scholar
Perissiou, M, Borkoles, E, Kobayashi, K & Polman, R (2020) The effect of an 8 week prescribed exercise and low-carbohydrate diet on cardiorespiratory fitness, body composition and cardiometabolic risk factors in obese individuals: a randomised controlled trial. Nutrients 12, 14.CrossRefGoogle ScholarPubMed
Ratliff, JC, Mutungi, G, Puglisi, MJ, Volek, JS & Fernandez, ML (2008) Eggs modulate the inflammatory response to carbohydrate restricted diets in overweight men. Nutr Metab (Lond) 5, 6.CrossRefGoogle ScholarPubMed
Seshadri, P, Samaha, FF, Stern, L, et al. (2005) Adipocytokine changes caused by low-carbohydrate compared to conventional diets in obesity. Metab Syndr Relat Disord 3, 6674.CrossRefGoogle ScholarPubMed
Bhanpuri, NH, Hallberg, SJ, Williams, PT, et al. (2018) Cardiovascular disease risk factor responses to t Type 2 diabetes care model including nutritional ketosis at one year: an open label, non-randomized, controlled study. Cardiovasc Diabetol 17, 116.CrossRefGoogle Scholar
Hallberg, SJ, McKenzie, AL, Williams, PT, et al. (2018) Effectiveness and safety of a novel care model for the management of type 2 diabetes at 1 year: an open-label, non-randomized, controlled study. Diabetes Ther 9, 583612.CrossRefGoogle ScholarPubMed
Cipryan, L, Plews, DJ, Ferretti, A, Maffetone, PB & Laursen, PB (2018) Effects of a 4-week very low-carbohydrate diet on high-intensity interval training responses. J Sports Sci Med 17, 259268.Google ScholarPubMed
Figure 0

Fig. 1. Inclusion/Exclusion flow chart.

Figure 1

Table 1. PICOS criteria for inclusion and exclusion of studies

Figure 2

Table 2. Reported neurological outcomes from human studies utilising low-carbohydrate (LCD) or ketogenic (KD) diets n = 64

Figure 3

Table 3. Reported inflammatory biomarkers from human studies utilising low-carbohydrate (LCD) or ketogenic (KD) diets, n = 63

Supplementary material: PDF

Field et al. supplementary material

Appendix

Download Field et al. supplementary material(PDF)
PDF 261.5 KB