Skip to main content
Erschienen in: Reviews in Endocrine and Metabolic Disorders 4/2019

Open Access 13.11.2019 | Epilepsy

Keto microbiota: A powerful contributor to host disease recovery

verfasst von: Amanda Cabrera-Mulero, Alberto Tinahones, Borja Bandera, Isabel Moreno-Indias, Manuel Macías-González, Francisco J. Tinahones

Erschienen in: Reviews in Endocrine and Metabolic Disorders | Ausgabe 4/2019

Abstract

Gut microbiota (GM) is a key contributor to host metabolism and physiology. Data generated on comparing diseased and healthy subjects have reported changes in the GM profile between both health states, suggesting certain bacterial composition could be involved in pathogenesis. Moreover, studies reported that reshaping of GM could contribute actively to disease recovery. Interestingly, ketogenic diets (KD) have emerged recently as new economic dietotherapeutic strategy to combat a myriad of diseases (refractory epilepsy, obesity, cancer, neurodegenerative diseases…). KD, understood in a broad sense, refers to whatever dietetic approximation, which causes physiological ketosis. Therefore, high fat-low carbs diets, fasting periods or caloric restriction constitute different strategies to produce an increase of main ketones bodies, acetoacetate and β-hydroxybutyrate, in blood. Involved biological mechanisms in ketotherapeutic effects are still to be unravelled. However, it has been pointed out that GM remodelling by KD, from now on “keto microbiota”, may play a crucial role in patient response to KD treatment. In fact, germ-free animals were resistant to ketotherapeutic effects; reinforcing keto microbiota may be a powerful contributor to host disease recovery. In this review, we will comment the influence of gut microbiota on host, as well as, therapeutic potential of ketogenic diets and keto microbiota to restore health status. Current progress and limitations will be argued too. In spite of few studies have defined applicability and mechanisms of KD, in the light of results, keto microbiota might be a new useful therapeutic agent.
Hinweise

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

1 Gut microbiota: A prokaryotic organ with multiple functions in the human body

Gut microbiota constitute a complex and dynamic ecosystem formed by bacteria, archaea, viruses, and fungi [1]. However, bacteria are the most studied [1]. The gastrointestinal tract is one of the widest surface area in the human body [2], and probably, the most diverse microbiota within the human body [3]. Microbiota members interact between them and with the host on multiple levels.
It is known microbiota and their metabolites are able to influence our physiology, both in health and disease [3]. Microbiota participates in host digestion and nutrition by producing nutrients from non-digestible substrates, contributes to metabolic functions, protects against pathogens, modulates the immune system, synthetizes vitamins, and produces a wide variety of biochemically active compounds (including neurotransmitters and enzymatic cofactors) [2, 3], among others. Therefore, through these vital functions, gut microbiota influences our physiology.
Multiple metagenomic data generated on comparing feces from diseased and healthy subjects have reported changes in the gut microbiota profile between both health states. Increasing observations have described a significant correlation between an imbalance of the microbiota profile, or dysbiosis, and the development of certain diseases [1, 4]. In fact, gut microbiota changes have been defined during obesity, diabetes, liver diseases, cardiometabolic disorders, inflammatory bowel diseases, autoimmune conditions, cancer [1, 2], and even nervous system disorders (autism, anxiety, depression, multiple sclerosis, Parkinson’s disease, and Alzheimer’s disease) [5].
The altered microbiota profile in different diseases, as well as its putative implication in the physiopathology of these diseases, has positioned to the gut microbiota as a potential source of innovative therapeutics [1]. However, the symbiotic relationship between gut microbiota and its host has triggered in different profiles among individuals [3]. As result, there is not a consensus about the optimal microbiota profile, which has the potential of protecting against any disease. However, many efforts have been put for this purpose Fig. 1.

2 Modulation of the gut microbiota profile

Gut microbiota composition is influenced by several factors, both genetic and environmental: human genetic, mode of delivery, type of feeding, medication (laxatives, antibiotics, proton pump inhibitors, metformin [6]), stress, infections, smoking, physical inactivity, bariatric surgery, fasting … [79].
However, diet is one of the main drivers of microbiota changes [6]. The overall features of the diet (i.e., total calories, timing, variety of nutrients, vitamins and fiber ingestion, percentage of macronutrients,…) all influence the composition of the microbiota and can modify it in short time [6]. Interestingly, chrononutrition has become extremely important as modulator of microbiota, since disruption of circadian rhythms can increase the risk of disease [10]. It is known that microbiome is affected by what is eaten but also by when food is consumed [11]. Time of eating has been reported to restore circadian rhythms affecting bacterial communities and its function [1012]. In fact, time-restricted feeding (TRF), an eating pattern in which food intake is restricted to a time window which can oscillates between 8 and 12 h [13, 14], is able to restore the cycling of the microbiota [11]. Therefore, restoring circadian rhythms, including microbiota rhythm, may also contribute to optimize individual’s physiology and decrease their risk of disease [10].
Undoubtedly, microbes that reside in the human gut are key contributors to host metabolism and, consequently, they are considered potential therapeutic targets [1]. For this reason, prebiotics, probiotics and fecal microbiota transplantation have emerged as new opportunities to promote and maintain a healthy microbiota and consequently a healthy homeostasis:
  • Probiotics term refers to “administration of live microorganisms in adequate amounts able to confer a health benefit on the host”. Consumption of live microbiota in both foods and dietary supplements has been related to amelioration or prevention of intestine inflammation and other intestinal or systemic disease phenotypes [1518].
  • Prebiotics are non-digestible selectively fermented dietary fibres that specifically promote the growth of beneficial targeted bacteria in gut providing some kind of benefit to the host, like immune regulation [19].
  • Fecal microbiota transplantation (FMT) has been reported as a useful medical tool, for example, in treating Clostridium difficile infection or insulin-resistance in obese patients [2], but, it is necessary yet to optimize the process and make donor microbiota perdurable in receptor.
However, despite the fact diet is the strongest and safer influencer on gut bacterial communities, few clinical studies of different kind of dietary interventions on human gut microbiota have been reported so far [2, 20]. Therefore, in this review we discuss the effect of different diet interventions on gut microbiota, focusing on a current science hot-topic, “the ketogenic diet”.

3 Keto diet and potential benefits of “keto microbiota”

Although ketogenic diets debuted in 1920 as a medical strategy to treat refractory epilepsy, its healing properties had not been scientifically evaluated in other medical areas [21, 22]. Recently, its applicability and efficacy are being tested in the treatment of other diseases such as obesity [23], polycystic ovarian syndrome [23], cancer [24], cardiovascular problems [25] and respiratory problems [26]. Multiple worldwide open clinical trials evaluate the tolerability and efficacy of ketogenic diets in the treatment of the aforementioned pathologies as a new medical treatment (http://​ClinicalTrials.​gov).
KD, understood in a broad sense, refers to any dietetic approximation able to produce a physiological ketosis, this is, an increase of ketone bodies. Classic ketogenic diets [21], fasting periods [27], time restricted feeding [13], caloric restriction diets [28], or intense physical exercise [29] constitute different strategies to produce ketonemia (increase of main ketones bodies, acetoacetate and β-hydroxybutyrate, in blood). Moreover, supplements that mimic the ketosis state as ketone esters [30] or ketone salts [31] have been developed in an attempt to overcome disadvantages of KD without a modification of the diet.
However, little is known about the underlying mechanisms of action of KD. The most accepted hypotheses point out: metabolic changes, alteration of the signalling pathways, changes in the production of hormones and neurotransmitters, epigenetic modifications [22, 24, 27], and as would be expected, modulation of the microbiota [32].
As we mentioned above, the gut microbiota plays an intermediary role between diet and host physiology. Diet affects composition, diversity and functionality of the gut microbiota and these changes in the gut microbiota are inducible and reproducible [33].
Currently, few data is available about the effects of ketogenic diets on gut microbiota composition [34]. (In this review, we have used the term “keto microbiota” to define a profile of gut microbiota moulded by a keto diet). Most of existing data about the KD impact into microbiota comes from epilepsy studies. Recent studies have reported that gut microbiota changes induced by a KD are required to improve the symptomatology of some diseases such as autism [35], epilepsy [33], or sclerosis [36].

3.1 Ketogenic diet, keto microbiota and epilepsy

The classic ketogenic diet (CKD) is a high-fat, adequate-protein, low-carbohydrate diet [21]. The most common ratio in this diet is 3:1 or 4:1. That is, 80–90% of the energy comes from fat and 10–20% from the combination of carbohydrates and proteins [24]. The term was coined by Wilder in 1921 who found that fasting caused an improvement in their epileptic patients and tried to mimetic the ketosis state provoked by fasting with a very low carb diet [37]. Since then, CKD has been the treatment of choice in epileptic refractory patients [21].
Gut microbiota profile is significantly different between healthy and epileptic individuals. KD treatment is able to reshape gut microbiota in humans and rodents [38, 39]; and this keto microbiota is required to avoid seizures. In fact, mouse models of refractory epilepsy showed that those given antibiotics or reared in a germ-free environment were resistant to seizure protection from KD, while keto microbiota fecal transplant helped mice with seizure control. Therefore, these results support that keto microbiota is necessary to protect against seizures [32, 40].
Interestingly, after a KD intervention, patients were differentiated into responder or non-responder subjects according to their gut microbiota changes, suggesting that the effectiveness of a KD was driven by the gut microbiota [5, 32]. Moreover, responder and non- responder groups differed in gut bacteria profiles at the level of order, family and genus, but also in microbial metabolites production. Such bacterial metabolites could be act by restricting precursors availability to synthetize inhibitory neurotransmitters involved in seizure control [40].
In parallel, Hampton et al. revealed that certain combinations of bacteria are required to improve epileptic symptomatology, for instance: they exposed that “Akkermansia muciniphila and Parabacteroides sp. colonization together but not alone protected against seizures in germ-free mice fed the ketogenic diet” [40]. Taken together, these findings underlie that microbiota is a complex system, where interactions between different species enable generate determined profiles of metabolites responsible to provoke a physiological response in host.
By contrast, in spite of benefits of keto microbiota in a growing number of diseases, Tagliabue reported that prolonging the KD for 3 months could cause dysmicrobism with damage to the gut health [41]. Consequently, they recommended prebiotics or probiotics treatment to re-establish gut microbiota and intestine homeostasis [41]. However, more follow-up studies are required in order to monitor the changes of the microbiota profiles with KD, and this highlights the necessity to monitor side effects and take into account possible dysbiosis.

4 Different ketogenic diets lead to a different gut microbiota

Although more studies are required to compare microbiota profile between different KD, in the light of outcome, probably, both microbiota profile and physiological responses may be dissimilar. Currently, results regarding microbial communities profile and diversity are controversial.
High fat, adequate protein, low carbs diet (CKD) seems to be associated with a lower diversity; finding which could be justified because microbiota is responsible to degrade undigested carbohydrates [2, 3], which are diminished in this type of diet.
By contrast, intermittent or continuous reduction of the caloric intake, caloric restriction (CR), might not affect alpha-diversity [42, 43]. In spite of this, CR has been reported to produce significant changes in faecal bacteria composition and metabolite content [4, 43], thus, affecting gene expression related to metabolism and inflammation [43].
For its part, fasting periods affect clock gene transcription, [44] as well as, gut microbiota itself [45]. Disturbance of the intestinal circadian clock lead to change the uptake of nutrients, gut motility, hormones production, gut microbiota fluctuations, and ultimately, the whole body and its microbiome [43]–[45].
Main changes produced in the gut microbiota by KD are reflected in the Table 1 (below shown).
Table 1
main gut microbiota changes produced by KD treatment. Generally, CKD is related to diversity decrease [5, 33, 36], while, CR did not decrease diversity [42, 43]. CKD: Classical ketogenic diet (high fat, adecuate protein low carbs). CR: caloric restriction or energy restriction
Type of KD
Human vs animal
Disease
KD duration
Phylum
Family
Genus
Species
Ref
CKD
Human (children)
Refractory epilepsy
Responders
6 months
Increase Bacteroidetes. Decrease Firmicutes and Actinobacteria
   
[5]
CKD
Zeneca
Human (children)
Refractory epilepsy
Responder
1 week
Increase Bacteroidetes. Decrease Proteobacteria
   
[39]
CKD
Human
GLUT1 deficiency syndrome
3 months
   
Increase Desulfovibrio spp
[41]
CKD
Mouse
Refractory epilepsy
14 days
 
Increase Erysipelotrichaceae
Increase Akkermansia ParabacteroidesSutterella
Increase Akkermansia muciniphila
[33]
CKD
Human
Multiple Sclerosis
6 months
  
Increase Akkermansia
 
[36]
CKD
Mouse
Autism
14 days
Increase Bacteroidetes
  
Decrease
Akkermansia muciniphila
[35]
CKD
Mouse
Refractory-epilepsy
14 days
 
Increase Erysipelotrichaceae
Increase Parabacteroide, Sutterella
Decrease Allobaculum, Bifidobacterium, and Desulfovibrio
Increase Akkermansia muciniphila,
[40]
CR
VLCD
Human
obesity
4 weeks
Decrease Proteobacteria
 
Increase Ruminococcus and
Bifidobacterium
Increase Anaerostipes hadrus
Decrease Agathobacter rectalis
[42]
CR 25%
Mouse
 
14 days
Decrease Actinobacteria and Proteobacteria
Increase Lactobacillaceae, Lachnospiraceae, Ruminococcaceae and Erysipelotrichaceae
Decrease Clostridiales
 
[43]
Fasting periods
Human
 
1 week
   
Increase Lactobacilli, Enterobacteria, and Akkermansia.
[46]
Fasting
Hamster
 
96 h
Increase Proteobaacteria Verrucomicrobia
Increase Desulfovibrionaceae
 
Increase Akkermansia
[47]
Since Zhang et al. reported different gut microbiota profile after KD between responder and non- responder epileptic patients [5], it is necessary to understand how there are different microbiota profiles more susceptible to be changed by KD than others. In addition, we must comprehend how a KD can overcome this challenge to prescribe it as a medical treatment.

5 Benefitial effects of keto diet: Is microbiota the only responsible for?

Previously, some studies reported that microbiota by itself was sufficient to enhance insulin sensitivity, improve tolerance to glucose and cold, and reduce fat content [48].
However, surely ketotherapeutic effects are result from different mechanisms: microbiota, epigenetic, metabolic reprogramming… Those all components could act interdependent and interrelated to many others.
Importantly, Ketone bodies (KB) fulfil several functions: 1) KB are energetic substrates, which are oxidized in heart, brain, and muscle during ketotic states, therefore, participating in bioenergetic homeostasis; 2) KB are also anabolic substrates contributing to lipogenesis and sterol biosynthesis in developing brain and lactating mammary gland, among other tissues;3) KB control mitochondrial metabolism and energetics; 4) KB reduce oxidative stress by inhibiting ROS/superoxide production, preventing lipid peroxidation and protein oxidation and increasing antioxidant protein levels; 5) KB act as signalling intermediators; 6) KB modulate inflammation and immune cell function; 7) KB decrease cellular damage, injury, death and lower apoptosis in neurons, lung cells and cardiomyocytes; 8) KB activate a reprogramming of pancreatic islet cells and could be involved in improved β-cell outcomes in states of increased ketogenesis, [4953], and so on. Likewise, microbiota is involved in the regulation of multiple host metabolic pathways, giving rise to interactive host-microbiota metabolic, signalling, and immune-inflammatory axes that physiologically connect the gut, liver, muscle, heart, kidney and brain [54]. Therefore KD exert a double effect about microbiota, directly by modifying substrate availability and indirectly modifying several functions which also impacts on microbiota.
In the same line, epigenetic changes can be influenced by microbiota, but also by ketone bodies. Ketone bodies (β-hydroxybutyrate and acetate) have been confirmed to affect epigenetic mark by inhibiting histone deacetylase class I [55], modifying proteins at the post-translational level by butyrylation [56], affecting DNA methylation [57] and acetylating histone and non-histone proteins [58]. But, metabolites produced by microbiota are also substrates or cofactors of enzymes involved in epigenetic process [59]. Therefore, it is not possible to discern epigenetic changes derived from both contributors.
Likewise, microbiota is a known endocrine organ which produces hormones and bioproducts with effects on the hosts’ biology [60]. But, microbiota composition is also influenced by hormones such as estrogens [61], so, there is a mutual crosstalk. KD have shown to affect both, microbiota and several hormones levels [13, 62]. Although to date, results about importance of gender on ketotherapeutic effects are controversial [6365], due to the interplay between diet-microbiota-hormones, the sex and hormonal status of an individual may influence on the efficacy of keto treatments. Reinforcing this, KD has become as a promising treatment in those diseases where sex hormones profile is altered such as ovarian or endometrial cancer [51], obesity [65] or polycystic ovarian syndrome [51].
Similarly, metabolic reprogramming may be the result of changing the microbiota profile [66], but also as a consequence of forcing to the organism to use ketone bodies like energy substrate [67]. Inflammation decrease may be result of lower content of pro-inflammatory bacteria [1], but also, consequence of lower infiltration of macrophages in adipose tissue due to fat loss derived of energy restriction [68]… so, this linking scenario could be observed with each of hypothesized mechanisms Fig. 2.
Below is a Table 2 summarising reported effects of ketotherapeutic therapies, as well as, putative involved biological mechanisms. Casual relationships are not defined yet. Additional genomics, metabolomics, and proteomics studies are required to identify molecular mechanisms through which ketosis regulates physiology responses and nuclear signalling events in each disease.
Table 2
Beneficial therapeutic effects of KD in different diseases and defined biological mechanisms, which could be involved in advantages of KD
Type of KD tested
Disease
Effect
Involved biological mechanisms allegedly derived by KD
Fasting
Classic ketogenic diet
Refractory epilepsy
Seizure protection [22, 39, 68]
Increase gamma-aminobutyric acid (GABA)
Decrease glutamate
pH alteration affecting excitability GABA
Microbiota profile reshape
Caloric restriction
Classic ketogenic diet
Fasting periods
Obesity
Weight loss and metabolic profile improvement [48, 6873]
Appetite suppression by ketone bodies
Gut microbiota profile and its metabolites change
Beiging of white adipose tissue
Brown adipose tissue activation (increase uncoupling protein UCP1 expression)
Loss of fat (not lean mass)
Increase metabolic cost of gluconeogenesis
Increase lipogenesis and decrease lipolysis
Metabolic improvements, browning and fat loss associated with microbiota remodelling
Caloric restriction
Classic ketogenic diet
DT2
Improve insulin sensitivity and glycaemic control [68, 74, 75]
Decrease inflammation
Reduce insulin
Lipid profile improvement
Decrease body weight, glycaemia, glycosylated haemoglobin, triglyceride level and LDL cholesterol Increased HDL cholesterol levels
Greater weight loss than other diets in the short term
Remarkable metabolic improvement
Reduced or even withdrawn antidiabetic treatment
Classic ketogenic diet
Ketone supplements
Alzheimer
Improve motor performance and cognition [30, 7678]
Compensate for the deficiency in glucose metabolism
Decrease glutamate
Increase GABA
Decrease reactive oxygen species (ROS)
Decrease Inflammation
Enhancing metabolism
Metabolic state improvement
Increase neurovascular integrity
Application of theory brain-gut-microbiota axis
Classic ketogenic diet
Multiple Sclerosis
Normalized concentrations of the colonic microbiome [36]
Restore colonic microbiome
Application of theory brain-gut-microbiota axis
Classic ketogenic diet
Autism
Mitigate some
of the neurological symptoms associated with Autism Spectrum Disorder [35, 79]
Application of theory brain-gut-microbiota axis
Fasting
Cardiovascular
Improve cardiovascular health [80]
Supply of ketone bodies to the heart where KBs are efficiently oxidized
Synergic actions between butyrate production by gut bacteria and circulating high blood ketones
Application of theory gut–heart metabolic axis
Caloric Restriction
Classic ketogenic diet
Cancer
Tumor growth decrease
Increase survival patient [67, 8183]
Cancer cell starvation (glucose privation reduce energy production of cancer cells)
Repress Warburg effect
Decrease inflammation
Decrease insulin and insulin growing factor
Decrease angiogenesis
Decrease PI3K/Akt signalling (a known pathway involved in carcinogenesis)
Increase apoptosis
Affect tumor epigenetic
Avoid cachexia (inhibition of muscle and body weight loss)
Sensitize cancer cells to oncological treatment

6 Putative therapeutic scope of ketogenic diets: Keto microbiota is sought

Ketogenic diets are being tested in degenerative and non-neurodegenerative diseases with successful results, although gut microbiota profile was not assessed. Here, we make one little sweep about last therapeutic findings of KD where it would be interesting to study keto microbiota.

6.1 Ketogenic diet and cancer

KD has been linked with a decrease in tumour growth [81] and an increase of the patient survival [84]. Currently, KD has been particularly suitable for adjuvant tumour therapy able to sensitize tumor cells to conventional chemotherapeutic and radiotherapeutic treatments [24, 85]. Recent studies also showed dietary administration with keto-formula could suppress tumor progression, improve systemic inmune responses and body composition, which might help to prevent cancer cachexia [82].
Probably, keto microbiota could play an important role in some kinds of cancer due to microbiome is able to establish connecting axes with other organs.
In fact, microbiota composition differs between healthy and cancer subjects, whereby, certain microbial genes can be used as cancer diagnostic biomarkers [86]. Murine experiments confirmed that germ-free mice had significant lower risk of developing colon tumours, suggesting microbiota could participate in first steps of carcinogenesis [87].
Reinforcing this, Klemment et al. underlined that there is a complex network between cancer, diet and microbiota [88]. Dietary habits have been associated with a remarkable figure of cancers [86]. The kind of consumed diet could tip the balance towards a type of microbiota able to produce oncometabolites or tumor-suppressive metabolites [89]. Both, microbiota and KD are responsible to produce short-chain fatty acids (SCFAs), whose role on cancer prevention and treatment have been broadly demonstrated. Therefore, KD and keto microbiota could contribute synergistically to prevent tumorigenesis scenario [88, 89]. Moreover, microbiota could also affect immunotherapy response and toxicity in oncological patients since microbiota modulate host immunity [90].
As a result, KD dietary treatment and/or keto microbiota could constitute promising strategies to curb carcinogenesis and to increase effectiveness of oncological therapies.

6.2 Ketogenic diet and neurodegenerative diseases

The beneficial effects of ketogenic diets have been shown in a wide variety of these neurological disorders such as Parkinson’s, Alzheimer’s and traumatic brain damage [71, 76]. In short, any neurodegenerative disorder characterized by neuronal hypometabolism can be approached with ketosis-producer diets since ketone bodies provide an alternative source of energy for hypometabolic neurons [91]. Similarly, it has been pointed out that KD could contribute to the overcoming of the cognitive deficits that occur with aging through an increase in capillary density and levels of hypoxia inducible factors [92]. Considering, gut microbiota and neuropathology are closely interrelated [36], surely defining microbiota profile will help to define a subset of neuropathologies which can be afforded by keto diet or keto microbiota.
Several studies have announced gut dysbiosis is related to neurological disease [78, 93]. But, at the same time, gut microbiota composition can be also affected by some mental disorders [93].
At present, It is well defined the bidirectional link between neurological diseases and gut microbiota alterations [78], reinforcing the the gut–brain axis theory. Microbiota could contribute to maintain neural homeostasis by different mechanisms such as: 1) producing active biological compounds, 2) interacting with intestinal barrier, 3) connecting neuro-endocrine-immune system [78], or 4) direct neuronal communication [93]. The disruption of one of these mechanisms could increase inflammation which constitutes one of the main contributors to neurodegeneration status [93].
Therefore, it is fair to say keto microbiota could play a crucial role in the ketotherapeutic responses.
(Note: Diseases mentioned in Table 2 also require analysing gut microbiota and its metabolites to shed more light about pathways and networks, which are stablished between keto microbiota and other factors).

7 Some considerations and limitations about available literature

  • Keto microbiota resilience. So far, there is a broad lack of knowledge about remodelling capacity of microbiota after stopping KD treatment. Likewise, most studies have focused on gut microbiota. However, microbiota in other body sites could also modify by keto diet and could participate in disease recovery, so more studies are required in this field. Studying these events would contribute to improve biotherapies, even, its medical prescription to treat certain pathologies.
  • Keto microbiota study follow-up. Most studies have characterized gut microbiota prior and after KD treatment. However, considering proper treatment duration is not established yet, maybe this final point of study is not representative of the most therapeutic capacity of KD. In this sense, monitor gut microbiota periodicity could contribute to understand benefits of ketotherapy.
  • Keto microbiota analysis corrected by other confusing variables: On the other hand, classic ketogenic diet (high lipids, low carbohydrates, adequate proteins) has been reported to lead to digestive problems due to the high fat content and deficiencies of minerals, vitamins and electrolytes by restringing the consumption of fruits and vegetables [94]. For this reason, vitamin and mineral supplements ingestion are recommended to alleviate symptoms and signs associated with the deficiencies of these compounds [95, 96]. Therefore, we wonder whether studies about CKD treatment were supported with vitamins, because microbiota profile could vary based on this. Thus, microbiota changes described could not be just the result of ketosis state. Moreover, microbiota profile should be also corrected by other confusing factors (smoking, alcohol, exercise…)
  • Do KD affect keto microbiota diversity? So far, few human studies have analysed changes in microbiota and its metabolites induced by KD and data are controversial. There are how confirm KD treatment reduce diversity, while, others affirm that there are not changes in richness and variety microbial communities. Probably, these inconsistencies are result of non-standardized protocols and differences in KD treatment duration, age of study population, base disease, or type of KD, among others. Surely, gut microbiota changes induced by caloric restriction could be different from those that are produced by a high fat-low carbs diet. Since, different dietetic approximations influence on a diverse way, as we commented above. Therefore, the type of diet could modulate a keto microbiota, more than ketosis state per se.
  • Effects of ketone supplements on microbiota: It would be interesting to test whether ketone supplements can modulate microbiota profile. This approach would represent a way to understand whether ketotherapeutic effects are just the result of ketosis or whether an additional microbial pattern is required to get these health benefits.

8 Concluding remarks

  • Keto diet is the treatment of choice in refractory epilepsy but KD also seems to be a diet-therapeutic strategy with vigorous potential in a myriad of diseases such as cancer, metabolic and endocrine diseases and neurological disorders.
  • Gut microbiota is a key contributor to host metabolism and physiology. Gut microbiota profile varies among healthy and sick individuals, and certain bacterial communities have been associated with higher risk of disease.
  • KD can reshape gut microbiota profile, and this microbiota modulation is able to generate a remarkable improvement in some diseases such as epilepsy or autism.
  • Ketogenic diets modulate microbiota profile, epigenetic mark, and metabolic reprogramming, as well as many other mechanisms. Those components could act interdependent and interrelated to many others.
  • Further studies will confirm whether microbiota profile and ketophysiological responses varies between different ketotherapeutic strategies. Currently, results regarding keto microbial communities profile and diversity are controversial.
  • Ketogenic diets together with chrononutrition may play a key role in restoring circadian rhythms and microbiota cycling, protecting from potential diseases or improving disease outcome.
  • Established axis between diet-microbiota and different tissues highlights the synergic activation of different mechanisms to restore health status, reinforcing the crucial and intermediate role of microbiota for this.
In conclusion, ketogenic diets seem to be a promising new therapy, although, it is early to know its effectiveness and scope. Considering diet is the main modulator of gut microbiota, then, keto microbiota could be a key factor involved in ketotherapeutic effects since microbiome works a vital network between different organs. Nevertheless, more clinical and preclinical studies are required to define the ideal KDs exposure time, or failing that, “keto microbiota profile”, to achieve proper functioning of the body in different pathologies.

Compliance with ethical standards

Conflict of interest

None of the authors have any conflict of interest to disclose.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat P. D. Cani, Human gut microbiome: hopes, threats and promises, Gut, vol. 67, n.o 9, pp. 1716–1725, sep. 2018.PubMedCrossRef P. D. Cani, Human gut microbiome: hopes, threats and promises, Gut, vol. 67, n.o 9, pp. 1716–1725, sep. 2018.PubMedCrossRef
2.
Zurück zum Zitat E. Gianchecchi y A. Fierabracci, Recent advances on microbiota involvement in the pathogenesis of autoimmunity, Int J Mol Sci., vol. 20, no 2, p. 283, ene. 2019.PubMedCentralCrossRef E. Gianchecchi y A. Fierabracci, Recent advances on microbiota involvement in the pathogenesis of autoimmunity, Int J Mol Sci., vol. 20, no 2, p. 283, ene. 2019.PubMedCentralCrossRef
3.
Zurück zum Zitat A. B. Shreiner, J. Y. Kao, y V. B. Young, The gut microbiome in health and in disease:, Curr Opin Gastroenterol., vol. 31, n.o 1, pp. 69–75, ene. 2015.PubMedPubMedCentralCrossRef A. B. Shreiner, J. Y. Kao, y V. B. Young, The gut microbiome in health and in disease:, Curr Opin Gastroenterol., vol. 31, n.o 1, pp. 69–75, ene. 2015.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat X. Zheng, S. Wang, y W. Jia, Calorie restriction and its impact on gut microbial composition and global metabolism, Front Med., vol. 12, n.o 6, pp. 634–644, dic. 2018.PubMedCrossRef X. Zheng, S. Wang, y W. Jia, Calorie restriction and its impact on gut microbial composition and global metabolism, Front Med., vol. 12, n.o 6, pp. 634–644, dic. 2018.PubMedCrossRef
5.
Zurück zum Zitat Y. Zhang, S. Zhou, Y. Zhou, L. Yu, L. Zhang, y Y. Wang, Altered gut microbiome composition in children with refractory epilepsy after ketogenic diet, Epilepsy Res., vol. 145, pp. 163–168, sep. 2018.PubMedCrossRef Y. Zhang, S. Zhou, Y. Zhou, L. Yu, L. Zhang, y Y. Wang, Altered gut microbiome composition in children with refractory epilepsy after ketogenic diet, Epilepsy Res., vol. 145, pp. 163–168, sep. 2018.PubMedCrossRef
6.
Zurück zum Zitat E. M. M. Quigley, Microbiota-brain-gut Axis and neurodegenerative diseases, Curr Neurol Neurosci Rep., vol. 17, n.o 12, dic. 2017. E. M. M. Quigley, Microbiota-brain-gut Axis and neurodegenerative diseases, Curr Neurol Neurosci Rep., vol. 17, n.o 12, dic. 2017.
7.
Zurück zum Zitat M. C. Cenit, Y. Sanz, y P. Codoñer-Franch, Influence of gut microbiota on neuropsychiatric disorders, World J Gastroenterol., vol. 23, n.o 30, p. 5486, 2017.PubMedPubMedCentralCrossRef M. C. Cenit, Y. Sanz, y P. Codoñer-Franch, Influence of gut microbiota on neuropsychiatric disorders, World J Gastroenterol., vol. 23, n.o 30, p. 5486, 2017.PubMedPubMedCentralCrossRef
8.
9.
Zurück zum Zitat C. Fraumene et al., Caloric restriction promotes rapid expansion and long-lasting increase of Lactobacillus in the rat fecal microbiota, Gut Microbes, vol. 9, n.o 2, pp. 104–114, mar. 2018. C. Fraumene et al., Caloric restriction promotes rapid expansion and long-lasting increase of Lactobacillus in the rat fecal microbiota, Gut Microbes, vol. 9, n.o 2, pp. 104–114, mar. 2018.
10.
Zurück zum Zitat E. N. C. Manoogian y S. Panda, Circadian rhythms, time-restricted feeding, and healthy aging, Ageing Res Rev., vol. 39, pp. 59–67, oct. 2017.PubMedCrossRef E. N. C. Manoogian y S. Panda, Circadian rhythms, time-restricted feeding, and healthy aging, Ageing Res Rev., vol. 39, pp. 59–67, oct. 2017.PubMedCrossRef
11.
Zurück zum Zitat J. L. Kaczmarek, S. V. Thompson, y H. D. Holscher, Complex interactions of circadian rhythms, eating behaviors, and the gastrointestinal microbiota and their potential impact on health, Nutr Rev., vol. 75, n.o 9, pp. 673–682, sep. 2017.PubMedPubMedCentralCrossRef J. L. Kaczmarek, S. V. Thompson, y H. D. Holscher, Complex interactions of circadian rhythms, eating behaviors, and the gastrointestinal microbiota and their potential impact on health, Nutr Rev., vol. 75, n.o 9, pp. 673–682, sep. 2017.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat R. M. Voigt, C. B. Forsyth, S. J. Green, P. A. Engen, y A. Keshavarzian, Circadian rhythm and the gut microbiome, en Int Rev Neurobiol, vol. 131, Elsevier, 2016, pp. 193–205. R. M. Voigt, C. B. Forsyth, S. J. Green, P. A. Engen, y A. Keshavarzian, Circadian rhythm and the gut microbiome, en Int Rev Neurobiol, vol. 131, Elsevier, 2016, pp. 193–205.
13.
Zurück zum Zitat M. P. Mattson, V. D. Longo, y M. Harvie, Impact of intermittent fasting on health and disease processes, Ageing Res Rev., vol. 39, pp. 46–58, oct. 2017.PubMedCrossRef M. P. Mattson, V. D. Longo, y M. Harvie, Impact of intermittent fasting on health and disease processes, Ageing Res Rev., vol. 39, pp. 46–58, oct. 2017.PubMedCrossRef
14.
Zurück zum Zitat V. D. Longo y S. Panda, Fasting, circadian rhythms, and time-restricted feeding in healthy lifespan, Cell Metab., vol. 23, n.o 6, pp. 1048–1059, jun. 2016.PubMedPubMedCentralCrossRef V. D. Longo y S. Panda, Fasting, circadian rhythms, and time-restricted feeding in healthy lifespan, Cell Metab., vol. 23, n.o 6, pp. 1048–1059, jun. 2016.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat A. Ferreiro, G. Dantas, y M. A. Ciorba, Insights into how probiotics colonize the healthy human gut, Gastroenterology, ene. 2019. A. Ferreiro, G. Dantas, y M. A. Ciorba, Insights into how probiotics colonize the healthy human gut, Gastroenterology, ene. 2019.
16.
Zurück zum Zitat M. E. Sanders, A. Benson, S. Lebeer, D. J. Merenstein, y T. R. Klaenhammer, Shared mechanisms among probiotic taxa: implications for general probiotic claims, Curr Opin Biotechnol., vol. 49, pp. 207–216, feb. 2018.PubMedCrossRef M. E. Sanders, A. Benson, S. Lebeer, D. J. Merenstein, y T. R. Klaenhammer, Shared mechanisms among probiotic taxa: implications for general probiotic claims, Curr Opin Biotechnol., vol. 49, pp. 207–216, feb. 2018.PubMedCrossRef
17.
Zurück zum Zitat F. Mosca, M. L. Gianni, y M. Rescigno, Can Postbiotics represent a new strategy for NEC?, New York, NY: Springer US, 2019. F. Mosca, M. L. Gianni, y M. Rescigno, Can Postbiotics represent a new strategy for NEC?, New York, NY: Springer US, 2019.
18.
Zurück zum Zitat B. Sánchez, S. Delgado, A. Blanco-Míguez, A. Lourenço, M. Gueimonde, y A. Margolles, Probiotics, gut microbiota, and their influence on host health and disease, Mol Nutr Food Res., vol. 61, n.o 1, p. 1600240, ene. 2017.CrossRef B. Sánchez, S. Delgado, A. Blanco-Míguez, A. Lourenço, M. Gueimonde, y A. Margolles, Probiotics, gut microbiota, and their influence on host health and disease, Mol Nutr Food Res., vol. 61, n.o 1, p. 1600240, ene. 2017.CrossRef
19.
Zurück zum Zitat B. Wilson y K. Whelan, Prebiotic inulin-type fructans and galacto-oligosaccharides: definition, specificity, function, and application in gastrointestinal disorders: prebiotic fructans and GOS, J Gastroenterol Hepatol., vol. 32, pp. 64–68, mar. 2017.PubMedCrossRef B. Wilson y K. Whelan, Prebiotic inulin-type fructans and galacto-oligosaccharides: definition, specificity, function, and application in gastrointestinal disorders: prebiotic fructans and GOS, J Gastroenterol Hepatol., vol. 32, pp. 64–68, mar. 2017.PubMedCrossRef
20.
Zurück zum Zitat B. Senghor, C. Sokhna, R. Ruimy, y J.-C. Lagier, Gut microbiota diversity according to dietary habits and geographical provenance, Hum Microbiome J., vol. 7-8, pp. 1–9, abr. 2018. B. Senghor, C. Sokhna, R. Ruimy, y J.-C. Lagier, Gut microbiota diversity according to dietary habits and geographical provenance, Hum Microbiome J., vol. 7-8, pp. 1–9, abr. 2018.
21.
Zurück zum Zitat L. P. de B. Sampaio, Ketogenic diet for epilepsy treatment, Arq Neuropsiquiatr., vol. 74, n.o 10, pp. 842–848, oct. 2016.PubMedCrossRef L. P. de B. Sampaio, Ketogenic diet for epilepsy treatment, Arq Neuropsiquiatr., vol. 74, n.o 10, pp. 842–848, oct. 2016.PubMedCrossRef
22.
Zurück zum Zitat E. H. Kossoff y J. R. McGrogan, Worldwide use of the ketogenic diet, Epilepsia, vol. 46, n.o 2, pp. 280–289, feb. 2005.PubMedCrossRef E. H. Kossoff y J. R. McGrogan, Worldwide use of the ketogenic diet, Epilepsia, vol. 46, n.o 2, pp. 280–289, feb. 2005.PubMedCrossRef
23.
Zurück zum Zitat J. C. Mavropoulos, W. S. Yancy, J. Hepburn, y E. C. Westman, The effects of a low-carbohydrate, ketogenic diet on the polycystic ovary syndrome: a pilot study, Nutr Metab., vol. 2, p. 35, dic. 2005. J. C. Mavropoulos, W. S. Yancy, J. Hepburn, y E. C. Westman, The effects of a low-carbohydrate, ketogenic diet on the polycystic ovary syndrome: a pilot study, Nutr Metab., vol. 2, p. 35, dic. 2005.
25.
Zurück zum Zitat B. J. Brehm, R. J. Seeley, S. R. Daniels, y D. A. D’Alessio, A randomized trial comparing a very low carbohydrate diet and a calorie-restricted low fat diet on body weight and cardiovascular risk factors in healthy women, J Clin Endocrinol Metab., vol. 88, n.o 4, pp. 1617–1623, abr. 2003.CrossRef B. J. Brehm, R. J. Seeley, S. R. Daniels, y D. A. D’Alessio, A randomized trial comparing a very low carbohydrate diet and a calorie-restricted low fat diet on body weight and cardiovascular risk factors in healthy women, J Clin Endocrinol Metab., vol. 88, n.o 4, pp. 1617–1623, abr. 2003.CrossRef
26.
Zurück zum Zitat R. Alessandro et al., Effects of twenty days of the ketogenic diet on metabolic and respiratory parameters in healthy subjects, Lung, vol. 193, n.o 6, pp. 939–945, dic. 2015. R. Alessandro et al., Effects of twenty days of the ketogenic diet on metabolic and respiratory parameters in healthy subjects, Lung, vol. 193, n.o 6, pp. 939–945, dic. 2015.
27.
Zurück zum Zitat V. D. Longo y M. P. Mattson, Fasting: molecular mechanisms and clinical applications, Cell Metab., vol. 19, n.o 2, pp. 181–192, feb. 2014. V. D. Longo y M. P. Mattson, Fasting: molecular mechanisms and clinical applications, Cell Metab., vol. 19, n.o 2, pp. 181–192, feb. 2014.
28.
Zurück zum Zitat A. Prabhakar et al., Acetone as biomarker for ketosis buildup capability - a study in healthy individuals under combined high fat and starvation diets, Nutr J., vol. 14, n.o 1, dic. 2015. A. Prabhakar et al., Acetone as biomarker for ketosis buildup capability - a study in healthy individuals under combined high fat and starvation diets, Nutr J., vol. 14, n.o 1, dic. 2015.
29.
Zurück zum Zitat P. J. Cox y K. Clarke, Acute nutritional ketosis: implications for exercise performance and metabolism, Extreme Physiol Med., vol. 3, n.o 1, p. 17, 2014. P. J. Cox y K. Clarke, Acute nutritional ketosis: implications for exercise performance and metabolism, Extreme Physiol Med., vol. 3, n.o 1, p. 17, 2014.
30.
Zurück zum Zitat M. T. Newport, T. B. VanItallie, Y. Kashiwaya, M. T. King, y R. L. Veech, A new way to produce hyperketonemia: use of ketone ester in a case of Alzheimer’s disease, Alzheimers Dement., vol. 11, n.o 1, pp. 99–103, ene. 2015.CrossRef M. T. Newport, T. B. VanItallie, Y. Kashiwaya, M. T. King, y R. L. Veech, A new way to produce hyperketonemia: use of ketone ester in a case of Alzheimer’s disease, Alzheimers Dement., vol. 11, n.o 1, pp. 99–103, ene. 2015.CrossRef
31.
Zurück zum Zitat W. Kephart et al., The 1-week and 8-month effects of a ketogenic diet or ketone salt supplementation on multi-organ markers of oxidative stress and mitochondrial function in rats, Nutrients, vol. 9, n.o 9, p. 1019, sep. 2017. W. Kephart et al., The 1-week and 8-month effects of a ketogenic diet or ketone salt supplementation on multi-organ markers of oxidative stress and mitochondrial function in rats, Nutrients, vol. 9, n.o 9, p. 1019, sep. 2017.
32.
33.
Zurück zum Zitat C. A. Olson, H. E. Vuong, J. M. Yano, Q. Y. Liang, D. J. Nusbaum, y E. Y. Hsiao, The gut microbiota mediates the anti-seizure effects of the ketogenic diet, Cell, vol. 173, n.o 7, pp. 1728–1741.e13, jun. 2018. C. A. Olson, H. E. Vuong, J. M. Yano, Q. Y. Liang, D. J. Nusbaum, y E. Y. Hsiao, The gut microbiota mediates the anti-seizure effects of the ketogenic diet, Cell, vol. 173, n.o 7, pp. 1728–1741.e13, jun. 2018.
34.
Zurück zum Zitat A. Paoli, G. Bosco, E. M. Camporesi, y D. Mangar, Ketosis, ketogenic diet and food intake control: a complex relationship, Front Psychol., vol. 6, feb. 2015. A. Paoli, G. Bosco, E. M. Camporesi, y D. Mangar, Ketosis, ketogenic diet and food intake control: a complex relationship, Front Psychol., vol. 6, feb. 2015.
35.
Zurück zum Zitat C. Newell, M. R. Bomhof, R. A. Reimer, D. S. Hittel, J. M. Rho, y J. Shearer, Ketogenic diet modifies the gut microbiota in a murine model of autism spectrum disorder, Mol Autism, vol. 7, n.o 1, dic. 2016. C. Newell, M. R. Bomhof, R. A. Reimer, D. S. Hittel, J. M. Rho, y J. Shearer, Ketogenic diet modifies the gut microbiota in a murine model of autism spectrum disorder, Mol Autism, vol. 7, n.o 1, dic. 2016.
36.
Zurück zum Zitat A. Swidsinski et al., Reduced mass and diversity of the colonic microbiome in patients with multiple sclerosis and their improvement with ketogenic diet, Front Microbiol., vol. 8, jun. 2017. A. Swidsinski et al., Reduced mass and diversity of the colonic microbiome in patients with multiple sclerosis and their improvement with ketogenic diet, Front Microbiol., vol. 8, jun. 2017.
37.
Zurück zum Zitat J. W. Wheless, History and origin of the ketogenic diet, en Epilepsy and the ketogenic diet, C. E. Stafstrom y J. M. Rho, Eds. Totowa, NJ: Humana Press, 2004, pp. 31–50. J. W. Wheless, History and origin of the ketogenic diet, en Epilepsy and the ketogenic diet, C. E. Stafstrom y J. M. Rho, Eds. Totowa, NJ: Humana Press, 2004, pp. 31–50.
38.
Zurück zum Zitat E. G. Neal et al., The ketogenic diet for the treatment of childhood epilepsy: a randomised controlled trial, Lancet Neurol., vol. 7, n.o 6, pp. 500–506, jun. 2008.PubMedCrossRef E. G. Neal et al., The ketogenic diet for the treatment of childhood epilepsy: a randomised controlled trial, Lancet Neurol., vol. 7, n.o 6, pp. 500–506, jun. 2008.PubMedCrossRef
39.
Zurück zum Zitat G. Xie et al., Ketogenic diet poses a significant effect on imbalanced gut microbiota in infants with refractory epilepsy, World J Gastroenterol., vol. 23, n.o 33, pp. 6164–6171, sep. 2017.PubMedPubMedCentralCrossRef G. Xie et al., Ketogenic diet poses a significant effect on imbalanced gut microbiota in infants with refractory epilepsy, World J Gastroenterol., vol. 23, n.o 33, pp. 6164–6171, sep. 2017.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat T. Hampton, Gut microbes may account for the anti-seizure effects of the ketogenic diet, JAMA, vol. 320, n.o 13, p. 1307, oct. 2018. T. Hampton, Gut microbes may account for the anti-seizure effects of the ketogenic diet, JAMA, vol. 320, n.o 13, p. 1307, oct. 2018.
41.
Zurück zum Zitat A. Tagliabue et al., Short-term impact of a classical ketogenic diet on gut microbiota in GLUT1 deficiency syndrome: a 3-month prospective observational study, Clin Nutr ESPEN, vol. 17, pp. 33–37, feb. 2017.PubMedCrossRef A. Tagliabue et al., Short-term impact of a classical ketogenic diet on gut microbiota in GLUT1 deficiency syndrome: a 3-month prospective observational study, Clin Nutr ESPEN, vol. 17, pp. 33–37, feb. 2017.PubMedCrossRef
42.
Zurück zum Zitat B. Ott et al., Effect of caloric restriction on gut permeability, inflammation markers, and fecal microbiota in obese women, Sci Rep., vol. 7, n.o 1, dic. 2017. B. Ott et al., Effect of caloric restriction on gut permeability, inflammation markers, and fecal microbiota in obese women, Sci Rep., vol. 7, n.o 1, dic. 2017.
43.
Zurück zum Zitat K. Duszka et al., Complementary intestinal mucosa and microbiota responses to caloric restriction, Sci Rep., vol. 8, n.o 1, dic. 2018. K. Duszka et al., Complementary intestinal mucosa and microbiota responses to caloric restriction, Sci Rep., vol. 8, n.o 1, dic. 2018.
44.
Zurück zum Zitat A. Mukherji, A. Kobiita, T. Ye, y P. Chambon, Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs, Cell, vol. 153, n.o 4, pp. 812–827, may 2013. A. Mukherji, A. Kobiita, T. Ye, y P. Chambon, Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs, Cell, vol. 153, n.o 4, pp. 812–827, may 2013.
45.
Zurück zum Zitat R. E. Patterson y D. D. Sears, Metabolic effects of intermittent fasting, Annu Rev Nutr., vol. 37, n.o 1, pp. 371–393, ago. 2017.PubMedCrossRef R. E. Patterson y D. D. Sears, Metabolic effects of intermittent fasting, Annu Rev Nutr., vol. 37, n.o 1, pp. 371–393, ago. 2017.PubMedCrossRef
46.
Zurück zum Zitat M. Remely, B. Hippe, I. Geretschlaeger, S. Stegmayer, I. Hoefinger, y A. Haslberger, Increased gut microbiota diversity and abundance of Faecalibacterium prausnitzii and Akkermansia after fasting: a pilot study, Wien Klin Wochenschr., vol. 127, n.o 9–10, pp. 394–398, may 2015. M. Remely, B. Hippe, I. Geretschlaeger, S. Stegmayer, I. Hoefinger, y A. Haslberger, Increased gut microbiota diversity and abundance of Faecalibacterium prausnitzii and Akkermansia after fasting: a pilot study, Wien Klin Wochenschr., vol. 127, n.o 9–10, pp. 394–398, may 2015.
47.
Zurück zum Zitat K. Sonoyama et al., Response of gut microbiota to fasting and hibernation in Syrian hamsters, Appl Environ Microbiol., vol. 75, n.o 20, pp. 6451–6456, oct. 2009.PubMedPubMedCentralCrossRef K. Sonoyama et al., Response of gut microbiota to fasting and hibernation in Syrian hamsters, Appl Environ Microbiol., vol. 75, n.o 20, pp. 6451–6456, oct. 2009.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat S. Fabbiano et al., Functional gut microbiota remodeling contributes to the caloric restriction-induced metabolic improvements, Cell Metab., vol. 28, n.o 6, pp. 907–921.e7, dic. 2018.PubMedPubMedCentralCrossRef S. Fabbiano et al., Functional gut microbiota remodeling contributes to the caloric restriction-induced metabolic improvements, Cell Metab., vol. 28, n.o 6, pp. 907–921.e7, dic. 2018.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat D. G. Cotter, R. C. Schugar, y P. A. Crawford, Ketone body metabolism and cardiovascular disease, AJP Heart Circ Physiol., vol. 304, n.o 8, pp. H1060-H1076, abr. 2013.PubMedPubMedCentralCrossRef D. G. Cotter, R. C. Schugar, y P. A. Crawford, Ketone body metabolism and cardiovascular disease, AJP Heart Circ Physiol., vol. 304, n.o 8, pp. H1060-H1076, abr. 2013.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat P. Puchalska y P. A. Crawford, Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics, Cell Metab., vol. 25, n.o 2, pp. 262–284, feb. 2017. P. Puchalska y P. A. Crawford, Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics, Cell Metab., vol. 25, n.o 2, pp. 262–284, feb. 2017.
51.
Zurück zum Zitat L. Gupta, D. Khandelwal, S. Kalra, P. Gupta, D. Dutta, y S. Aggarwal, Ketogenic diet in endocrine disorders: Current perspectives, J Postgrad Med., vol. 63, n.o 4, p. 242, 2017.PubMedPubMedCentralCrossRef L. Gupta, D. Khandelwal, S. Kalra, P. Gupta, D. Dutta, y S. Aggarwal, Ketogenic diet in endocrine disorders: Current perspectives, J Postgrad Med., vol. 63, n.o 4, p. 242, 2017.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Bolla, Caretto, Laurenzi, Scavini, y Piemonti, Low-carb and ketogenic diets in type 1 and type 2 diabetes, Nutrients, vol. 11, n.o 5, p. 962, abr. 2019. Bolla, Caretto, Laurenzi, Scavini, y Piemonti, Low-carb and ketogenic diets in type 1 and type 2 diabetes, Nutrients, vol. 11, n.o 5, p. 962, abr. 2019.
53.
Zurück zum Zitat M. Sampson et al., β-Hydroxybutyrate improves β-cell mitochondrial function and survival, J Insul Resist., vol. 1, n.o 1, ago. 2017. M. Sampson et al., β-Hydroxybutyrate improves β-cell mitochondrial function and survival, J Insul Resist., vol. 1, n.o 1, ago. 2017.
54.
Zurück zum Zitat J. K. Nicholson et al., Host-gut microbiota metabolic interactions, Science, vol. 336, n.o 6086, pp. 1262–1267, jun. 2012.PubMedCrossRef J. K. Nicholson et al., Host-gut microbiota metabolic interactions, Science, vol. 336, n.o 6086, pp. 1262–1267, jun. 2012.PubMedCrossRef
55.
Zurück zum Zitat T. Shimazu et al., Suppression of oxidative stress by -Hydroxybutyrate, an endogenous histone deacetylase inhibitor, Science, vol. 339, n.o 6116, pp. 211–214, ene. 2013.PubMedPubMedCentralCrossRef T. Shimazu et al., Suppression of oxidative stress by -Hydroxybutyrate, an endogenous histone deacetylase inhibitor, Science, vol. 339, n.o 6116, pp. 211–214, ene. 2013.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat J. S. Benjamin et al., A ketogenic diet rescues hippocampal memory defects in a mouse model of kabuki syndrome, Proc Natl Acad Sci U S A., vol. 114, n.o 1, pp. 125–130, ene. 2017.CrossRef J. S. Benjamin et al., A ketogenic diet rescues hippocampal memory defects in a mouse model of kabuki syndrome, Proc Natl Acad Sci U S A., vol. 114, n.o 1, pp. 125–130, ene. 2017.CrossRef
57.
Zurück zum Zitat M. Shirahata, W.-Y. Tang, y E. W. Kostuk, A short-term fasting in neonates induces breathing instability and epigenetic modification in the carotid body, en Arterial chemoreceptors in physiology and pathophysiology, vol. 860, C. Peers, P. Kumar, C. Wyatt, E. Gauda, C. A. Nurse, y N. Prabhakar, Eds. Cham: Springer International Publishing, 2015, pp. 187–193. M. Shirahata, W.-Y. Tang, y E. W. Kostuk, A short-term fasting in neonates induces breathing instability and epigenetic modification in the carotid body, en Arterial chemoreceptors in physiology and pathophysiology, vol. 860, C. Peers, P. Kumar, C. Wyatt, E. Gauda, C. A. Nurse, y N. Prabhakar, Eds. Cham: Springer International Publishing, 2015, pp. 187–193.
58.
Zurück zum Zitat D. M. Jaworski, A. M. A. Namboodiri, y J. R. Moffett, Acetate as a metabolic and epigenetic modifier of Cancer therapy: Acetate as a modifier of cancer therapy, J Cell Biochem., vol. 117, n.o 3, pp. 574–588, mar. 2016.PubMedCrossRef D. M. Jaworski, A. M. A. Namboodiri, y J. R. Moffett, Acetate as a metabolic and epigenetic modifier of Cancer therapy: Acetate as a modifier of cancer therapy, J Cell Biochem., vol. 117, n.o 3, pp. 574–588, mar. 2016.PubMedCrossRef
59.
Zurück zum Zitat M. I. Bhat y R. Kapila, Dietary metabolites derived from gut microbiota: critical modulators of epigenetic changes in mammals, Nutr Rev., vol. 75, n.o 5, pp. 374–389, may 2017.PubMedCrossRef M. I. Bhat y R. Kapila, Dietary metabolites derived from gut microbiota: critical modulators of epigenetic changes in mammals, Nutr Rev., vol. 75, n.o 5, pp. 374–389, may 2017.PubMedCrossRef
60.
Zurück zum Zitat B. Afsar, N. D. Vaziri, G. Aslan, K. Tarim, y M. Kanbay, Gut hormones and gut microbiota: implications for kidney function and hypertension, J Am Soc Hypertens., vol. 10, n.o 12, pp. 954–961, dic. 2016.PubMedCrossRef B. Afsar, N. D. Vaziri, G. Aslan, K. Tarim, y M. Kanbay, Gut hormones and gut microbiota: implications for kidney function and hypertension, J Am Soc Hypertens., vol. 10, n.o 12, pp. 954–961, dic. 2016.PubMedCrossRef
61.
Zurück zum Zitat J. M. Baker, L. Al-Nakkash, y M. M. Herbst-Kralovetz, Estrogen–gut microbiome axis: physiological and clinical implications, Maturitas, vol. 103, pp. 45–53, sep. 2017.PubMedCrossRef J. M. Baker, L. Al-Nakkash, y M. M. Herbst-Kralovetz, Estrogen–gut microbiome axis: physiological and clinical implications, Maturitas, vol. 103, pp. 45–53, sep. 2017.PubMedCrossRef
62.
Zurück zum Zitat P. Sumithran et al., Ketosis and appetite-mediating nutrients and hormones after weight loss, Eur J Clin Nutr., vol. 67, n.o 7, pp. 759–764, jul. 2013.PubMedCrossRef P. Sumithran et al., Ketosis and appetite-mediating nutrients and hormones after weight loss, Eur J Clin Nutr., vol. 67, n.o 7, pp. 759–764, jul. 2013.PubMedCrossRef
63.
Zurück zum Zitat A. Lyngstad et al., Investigating the effect of sex and ketosis on weight-loss-induced changes in appetite, Am J Clin Nutr., vol. 109, n.o 6, pp. 1511–1518, jun. 2019.PubMedPubMedCentralCrossRef A. Lyngstad et al., Investigating the effect of sex and ketosis on weight-loss-induced changes in appetite, Am J Clin Nutr., vol. 109, n.o 6, pp. 1511–1518, jun. 2019.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat D. N. Ruskin, J. A. Fortin, S. N. Bisnauth, y S. A. Masino, Ketogenic diets improve behaviors associated with autism spectrum disorder in a sex-specific manner in the EL mouse, Physiol Behav., vol. 168, pp. 138–145, ene. 2017.CrossRef D. N. Ruskin, J. A. Fortin, S. N. Bisnauth, y S. A. Masino, Ketogenic diets improve behaviors associated with autism spectrum disorder in a sex-specific manner in the EL mouse, Physiol Behav., vol. 168, pp. 138–145, ene. 2017.CrossRef
65.
Zurück zum Zitat A. Castro et al., Effect of a very low-calorie ketogenic diet on food and alcohol cravings, physical and sexual activity, sleep disturbances, and quality of life in obese patients, Nutrients, vol. 10, n.o 10, p. 1348, sep. 2018. A. Castro et al., Effect of a very low-calorie ketogenic diet on food and alcohol cravings, physical and sexual activity, sleep disturbances, and quality of life in obese patients, Nutrients, vol. 10, n.o 10, p. 1348, sep. 2018.
66.
Zurück zum Zitat K. A. Krautkramer, R. S. Dhillon, J. M. Denu, y H. V. Carey, Metabolic programming of the epigenome: host and gut microbial metabolite interactions with host chromatin, Transl Res., vol. 189, pp. 30–50, nov. 2017.PubMedPubMedCentralCrossRef K. A. Krautkramer, R. S. Dhillon, J. M. Denu, y H. V. Carey, Metabolic programming of the epigenome: host and gut microbial metabolite interactions with host chromatin, Transl Res., vol. 189, pp. 30–50, nov. 2017.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat S. K. Shukla et al., Metabolic reprogramming induced by ketone bodies diminishes pancreatic cancer cachexia, Cancer Metab., vol. 2, n.o 1, p. 18, 2014.CrossRef S. K. Shukla et al., Metabolic reprogramming induced by ketone bodies diminishes pancreatic cancer cachexia, Cancer Metab., vol. 2, n.o 1, p. 18, 2014.CrossRef
68.
Zurück zum Zitat H. M. Dashti et al., Long-term effects of a ketogenic diet in obese patients, Exp Clin Cardiol., vol. 9, n.o 3, pp. 200–205, 2004. H. M. Dashti et al., Long-term effects of a ketogenic diet in obese patients, Exp Clin Cardiol., vol. 9, n.o 3, pp. 200–205, 2004.
69.
Zurück zum Zitat A. Persynaki, S. Karras, y C. Pichard, Unraveling the metabolic health benefits of fasting related to religious beliefs: a narrative review, Nutrition, vol. 35, pp. 14–20, mar. 2017. A. Persynaki, S. Karras, y C. Pichard, Unraveling the metabolic health benefits of fasting related to religious beliefs: a narrative review, Nutrition, vol. 35, pp. 14–20, mar. 2017.
70.
Zurück zum Zitat J. T. Haas y B. Staels, Fasting the microbiota to improve metabolism?, Cell Metab., vol. 26, n.o 4, pp. 584–585, oct. 2017.PubMedCrossRef J. T. Haas y B. Staels, Fasting the microbiota to improve metabolism?, Cell Metab., vol. 26, n.o 4, pp. 584–585, oct. 2017.PubMedCrossRef
71.
Zurück zum Zitat S. Srivastava et al., Mitochondrial biogenesis and increased uncoupling protein 1 in brown adipose tissue of mice fed a ketone ester diet, FASEB J., vol. 26, n.o 6, pp. 2351–2362, jun. 2012. S. Srivastava et al., Mitochondrial biogenesis and increased uncoupling protein 1 in brown adipose tissue of mice fed a ketone ester diet, FASEB J., vol. 26, n.o 6, pp. 2351–2362, jun. 2012.
73.
Zurück zum Zitat A. Paoli, A. Rubini, J. S. Volek, y K. A. Grimaldi, Beyond weight loss: a review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets, Eur J Clin Nutr., vol. 67, n.o 8, pp. 789–796, ago. 2013.PubMedPubMedCentralCrossRef A. Paoli, A. Rubini, J. S. Volek, y K. A. Grimaldi, Beyond weight loss: a review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets, Eur J Clin Nutr., vol. 67, n.o 8, pp. 789–796, ago. 2013.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat J. S. Volek et al., Carbohydrate restriction has a more favorable impact on the metabolic syndrome than a low fat diet, Lipids, vol. 44, n.o 4, pp. 297–309, abr. 2009. J. S. Volek et al., Carbohydrate restriction has a more favorable impact on the metabolic syndrome than a low fat diet, Lipids, vol. 44, n.o 4, pp. 297–309, abr. 2009.
75.
Zurück zum Zitat A. M. Johnstone, G. W. Horgan, S. D. Murison, D. M. Bremner, y G. E. Lobley, Effects of a high-protein ketogenic diet on hunger, appetite, and weight loss in obese men feeding ad libitum, Am J Clin Nutr., vol. 87, n.o 1, pp. 44–55, ene. 2008.PubMedCrossRef A. M. Johnstone, G. W. Horgan, S. D. Murison, D. M. Bremner, y G. E. Lobley, Effects of a high-protein ketogenic diet on hunger, appetite, and weight loss in obese men feeding ad libitum, Am J Clin Nutr., vol. 87, n.o 1, pp. 44–55, ene. 2008.PubMedCrossRef
76.
Zurück zum Zitat H. M. Francis y R. J. Stevenson, Potential for diet to prevent and remediate cognitive deficits in neurological disorders, Nutr Rev., ene. 2018. H. M. Francis y R. J. Stevenson, Potential for diet to prevent and remediate cognitive deficits in neurological disorders, Nutr Rev., ene. 2018.
77.
Zurück zum Zitat S. J. Koppel y R. H. Swerdlow, Neuroketotherapeutics: a modern review of a century-old therapy, Neurochem Int., jun. 2017. S. J. Koppel y R. H. Swerdlow, Neuroketotherapeutics: a modern review of a century-old therapy, Neurochem Int., jun. 2017.
78.
Zurück zum Zitat C. Pellegrini, L. Antonioli, R. Colucci, C. Blandizzi, y M. Fornai, Interplay among gut microbiota, intestinal mucosal barrier and enteric neuro-immune system: a common path to neurodegenerative diseases?, Acta Neuropathol. (Berl.), vol. 136, n.o 3, pp. 345–361, sep. 2018.PubMedCrossRef C. Pellegrini, L. Antonioli, R. Colucci, C. Blandizzi, y M. Fornai, Interplay among gut microbiota, intestinal mucosal barrier and enteric neuro-immune system: a common path to neurodegenerative diseases?, Acta Neuropathol. (Berl.), vol. 136, n.o 3, pp. 345–361, sep. 2018.PubMedCrossRef
79.
Zurück zum Zitat Q. Li, Y. Han, A. B. C. Dy, y R. J. Hagerman, The gut microbiota and autism Spectrum disorders, front. Cell Neurosci., vol. 11, abr. 2017. Q. Li, Y. Han, A. B. C. Dy, y R. J. Hagerman, The gut microbiota and autism Spectrum disorders, front. Cell Neurosci., vol. 11, abr. 2017.
80.
Zurück zum Zitat P. A. Crawford et al., Regulation of myocardial ketone body metabolism by the gut microbiota during nutrient deprivation, Proc Natl Acad Sci., vol. 106, n.o 27, pp. 11276–11281, jul. 2009.CrossRef P. A. Crawford et al., Regulation of myocardial ketone body metabolism by the gut microbiota during nutrient deprivation, Proc Natl Acad Sci., vol. 106, n.o 27, pp. 11276–11281, jul. 2009.CrossRef
81.
Zurück zum Zitat H.-Y. Chung y Y. K. Park, Rationale, feasibility and acceptability of ketogenic diet for Cancer treatment, J Cancer Prev., vol. 22, n.o 3, pp. 127–134, sep. 2017.PubMedPubMedCentralCrossRef H.-Y. Chung y Y. K. Park, Rationale, feasibility and acceptability of ketogenic diet for Cancer treatment, J Cancer Prev., vol. 22, n.o 3, pp. 127–134, sep. 2017.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Kentaro Nakamura, Hidekazu Tonouchi, Akina Sasayama, y Kinya Ashida, A ketogenic formula prevents tumor progression and Cancer Cachexia by attenuating systemic inflammation in Colon 26 tumor-bearing mice, Nutrients, vol. 10, n.o 2, p. 206, feb. 2018. Kentaro Nakamura, Hidekazu Tonouchi, Akina Sasayama, y Kinya Ashida, A ketogenic formula prevents tumor progression and Cancer Cachexia by attenuating systemic inflammation in Colon 26 tumor-bearing mice, Nutrients, vol. 10, n.o 2, p. 206, feb. 2018.
83.
Zurück zum Zitat C. L. P. Oliveira, S. Mattingly, R. Schirrmacher, M. B. Sawyer, E. J. Fine, y C. M. Prado, A nutritional perspective of ketogenic diet in Cancer: a narrative review, J Acad Nutr Diet., mar. 2017. C. L. P. Oliveira, S. Mattingly, R. Schirrmacher, M. B. Sawyer, E. J. Fine, y C. M. Prado, A nutritional perspective of ketogenic diet in Cancer: a narrative review, J Acad Nutr Diet., mar. 2017.
84.
Zurück zum Zitat S. Khodadadi et al., Tumor cells growth and survival time with the ketogenic diet in animal models: a systematic review, Int J Prev Med., vol. 8, n.o 1, p. 35, 2017.PubMedPubMedCentralCrossRef S. Khodadadi et al., Tumor cells growth and survival time with the ketogenic diet in animal models: a systematic review, Int J Prev Med., vol. 8, n.o 1, p. 35, 2017.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat T. N. Seyfried, M. A. Kiebish, J. Marsh, L. M. Shelton, L. C. Huysentruyt, y P. Mukherjee, Metabolic management of brain cancer, Biochim Biophys Acta BBA - Bioenerg., vol. 1807, n.o 6, pp. 577–594, jun. 2011. T. N. Seyfried, M. A. Kiebish, J. Marsh, L. M. Shelton, L. C. Huysentruyt, y P. Mukherjee, Metabolic management of brain cancer, Biochim Biophys Acta BBA - Bioenerg., vol. 1807, n.o 6, pp. 577–594, jun. 2011.
86.
Zurück zum Zitat K. Vipperla y S. J. O’Keefe, Diet, microbiota, and dysbiosis: a ‘recipe’ for colorectal cancer, Food Funct., vol. 7, n.o 4, pp. 1731–1740, 2016.CrossRef K. Vipperla y S. J. O’Keefe, Diet, microbiota, and dysbiosis: a ‘recipe’ for colorectal cancer, Food Funct., vol. 7, n.o 4, pp. 1731–1740, 2016.CrossRef
87.
Zurück zum Zitat J. Yang y J. Yu, The association of diet, gut microbiota and colorectal cancer: what we eat may imply what we get, Protein Cell, vol. 9, n.o 5, pp. 474–487, may 2018. J. Yang y J. Yu, The association of diet, gut microbiota and colorectal cancer: what we eat may imply what we get, Protein Cell, vol. 9, n.o 5, pp. 474–487, may 2018.
88.
Zurück zum Zitat R. J. Klement y V. Pazienza, Impact of different types of diet on gut microbiota profiles and Cancer prevention and treatment, Medicina (Mex), vol. 55, n.o 4, p. 84, mar. 2019.PubMedCentralCrossRef R. J. Klement y V. Pazienza, Impact of different types of diet on gut microbiota profiles and Cancer prevention and treatment, Medicina (Mex), vol. 55, n.o 4, p. 84, mar. 2019.PubMedCentralCrossRef
89.
Zurück zum Zitat S. J. Bultman, Interplay between diet, gut microbiota, epigenetic events, and colorectal cancer, Mol Nutr Food Res., vol. 61, n.o 1, p. 1500902, ene. 2017.CrossRef S. J. Bultman, Interplay between diet, gut microbiota, epigenetic events, and colorectal cancer, Mol Nutr Food Res., vol. 61, n.o 1, p. 1500902, ene. 2017.CrossRef
90.
Zurück zum Zitat V. Gopalakrishnan, B. A. Helmink, C. N. Spencer, A. Reuben, y J. A. Wargo, The influence of the gut microbiome on Cancer, immunity, and Cancer immunotherapy, Cancer Cell, vol. 33, n.o 4, pp. 570–580, abr. 2018. V. Gopalakrishnan, B. A. Helmink, C. N. Spencer, A. Reuben, y J. A. Wargo, The influence of the gut microbiome on Cancer, immunity, and Cancer immunotherapy, Cancer Cell, vol. 33, n.o 4, pp. 570–580, abr. 2018.
91.
Zurück zum Zitat S. T. Henderson, J. L. Vogel, L. J. Barr, F. Garvin, J. J. Jones, y L. C. Costantini, Study of the ketogenic agent AC-1202 in mild to moderate Alzheimer’s disease: a randomized, double-blind, placebo-controlled, multicenter trial, Nutr. Metab., vol. 6, n.o 1, p. 31, 2009.CrossRef S. T. Henderson, J. L. Vogel, L. J. Barr, F. Garvin, J. J. Jones, y L. C. Costantini, Study of the ketogenic agent AC-1202 in mild to moderate Alzheimer’s disease: a randomized, double-blind, placebo-controlled, multicenter trial, Nutr. Metab., vol. 6, n.o 1, p. 31, 2009.CrossRef
92.
Zurück zum Zitat L. C. Costantini, L. J. Barr, J. L. Vogel, y S. T. Henderson, Hypometabolism as a therapeutic target in Alzheimer’s disease, BMC Neurosci., vol. 9, n.o Suppl 2, p. S16, 2008. L. C. Costantini, L. J. Barr, J. L. Vogel, y S. T. Henderson, Hypometabolism as a therapeutic target in Alzheimer’s disease, BMC Neurosci., vol. 9, n.o Suppl 2, p. S16, 2008.
93.
Zurück zum Zitat S. Westfall, N. Lomis, I. Kahouli, S. Y. Dia, S. P. Singh, y S. Prakash, Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis, Cell Mol Life Sci., vol. 74, n.o 20, pp. 3769–3787, oct. 2017.PubMedCrossRef S. Westfall, N. Lomis, I. Kahouli, S. Y. Dia, S. P. Singh, y S. Prakash, Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis, Cell Mol Life Sci., vol. 74, n.o 20, pp. 3769–3787, oct. 2017.PubMedCrossRef
94.
Zurück zum Zitat P. R. Huttenlocher, A. J. Wilbourn, y J. M. Signore, Medium-chain triglycerides as a therapy for intractable childhood epilepsy, Neurology, vol. 21, n.o 11, pp. 1097–1103, nov. 1971.PubMedCrossRef P. R. Huttenlocher, A. J. Wilbourn, y J. M. Signore, Medium-chain triglycerides as a therapy for intractable childhood epilepsy, Neurology, vol. 21, n.o 11, pp. 1097–1103, nov. 1971.PubMedCrossRef
95.
Zurück zum Zitat J. W. Wheless, The ketogenic diet: an effective medical therapy with side effects, J Child Neurol., vol. 16, n.o 9, pp. 633–635, sep. 2001.PubMedCrossRef J. W. Wheless, The ketogenic diet: an effective medical therapy with side effects, J Child Neurol., vol. 16, n.o 9, pp. 633–635, sep. 2001.PubMedCrossRef
96.
Zurück zum Zitat A. G. C. Bergqvist, Long-term monitoring of the ketogenic diet: Do’s and Don’ts, Epilepsy Res., vol. 100, n.o 3, pp. 261–266, jul. 2012.PubMedCrossRef A. G. C. Bergqvist, Long-term monitoring of the ketogenic diet: Do’s and Don’ts, Epilepsy Res., vol. 100, n.o 3, pp. 261–266, jul. 2012.PubMedCrossRef
Metadaten
Titel
Keto microbiota: A powerful contributor to host disease recovery
verfasst von
Amanda Cabrera-Mulero
Alberto Tinahones
Borja Bandera
Isabel Moreno-Indias
Manuel Macías-González
Francisco J. Tinahones
Publikationsdatum
13.11.2019
Verlag
Springer US
Erschienen in
Reviews in Endocrine and Metabolic Disorders / Ausgabe 4/2019
Print ISSN: 1389-9155
Elektronische ISSN: 1573-2606
DOI
https://doi.org/10.1007/s11154-019-09518-8

Weitere Artikel der Ausgabe 4/2019

Reviews in Endocrine and Metabolic Disorders 4/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.