Ayuno Intermitente y Diabeticos 2

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Current Diabetes Reports (2020) 20:13

https://doi.org/10.1007/s11892-020-1295-2

LIFESTYLE MANAGEMENT TO REDUCE DIABETES/CARDIOVASCULAR RISK (B CONWAY AND H


KEENAN, SECTION EDITORS)

Intermittent Fasting as Part of the Management for T2DM:


from Animal Models to Human Clinical Studies
Liliana Muñoz-Hernández 1,2 & Ziomara Márquez-López 1 & Roopa Mehta 1 & Carlos Alberto Aguilar-Salinas 1,3,4

# Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
Purpose of Review Diet is a pillar of type 2 diabetes mellitus (T2DM) management. Intermittent fasting (IF) is postulated as a
novel approach, able to improve glucose control and potentially capable of reversing some of the pathophysiological alterations
of this condition. In this review, the molecular and clinical evidence of diets based on intermittent energy restriction (IER) in
laboratory animal models and subjects with type 2 diabetes is discussed. The mechanisms through which IF are thought to
improve glucose homeostasis and reverse β cell failure are also reviewed.
Recent Findings Studies derived from murine models suggest that IER is associated with improvements in β cell function and
insulin resistance. Two main mechanisms have been demonstrated, one derived from the autophagy-lysosome pathway and, the
other from an increase in neurogenin3 (Ngn3) levels (a marker for endocrine progenitor cells like β cells during development).
Notably, IER also promotes reconstruction of gut microbiota. In mice, all effects were independent of weight loss. By contrast, in
human studies, outcomes are widely attributable to weight loss. The more consistent results are reductions in body weight,
visceral fat, and glucose and insulin levels. Increases in HDL cholesterol levels are also frequently reported. The decrease in
insulin levels observed in humans is in opposition with the increase reported in mice, suggesting that the main mechanism in
humans is an improvement in peripheral insulin action.
Summary Recommending diets based on intermittent fasting in humans is based on the promising results found in animal models
where an improvement in β cell function has been recorded. β cell function after IF has not been assessed in human subjects with
T2DM. This review provides information regarding different protocols for the implementation of IF in diabetic persons and also
provides important safety advice in order to avoid adverse effects. Clinical studies do not show an increased risk of hypoglyce-
mia, and a recent case series reported reversal of T2DM.

Keywords Intermittent energy restriction . Intermittent fasting . Alternate-day fasting . Time-restricted feeding .
Fasting-mimicking diets . Type 2 diabetes remission

Abbreviations
This article is part of the Topical Collection on Lifestyle Management to ADF Alternate-day fasting
Reduce Diabetes/Cardiovascular Risk AL Ad libitum
ALT Alanine aminotransferase
* Carlos Alberto Aguilar-Salinas ASUI Asthma Symptom Utility Index
caguilarsalinas@yahoo.com AST Aspartate aminotransferase
1 BDNF Brain-derived neurotrophic factor
Metabolic Diseases Research Unit, Instituto Nacional de Ciencias
Medicas y Nutricion, Mexico City, Mexico BF Body fat
2 BMI Body mass index
Consejo Nacional de Ciencia y Tecnología, Mexico City, Mexico
BW Body weight
3
Escuela de Medicina y Ciencias de la Salud, Tecnológico de BUN Blood urea nitrogen
Monterrey, Monterrey, Mexico
CER Continuous energy restriction
4
Division of Nutrition, Instituto Nacional de Ciencias Medicas y CRP C-reactive protein
Nutricion, Vasco de Quiroga #15, Tlalpan, 14080 Mexico
City, Mexico CV Cardiovascular
13 Page 2 of 10 Curr Diab Rep (2020) 20:13

DBP Diastolic blood pressure benefits of dietary modifications resulting in weight loss and
FFA Free fatty acid associated positive metabolic effects are irrefutable [1, 2].
FI Food intake Recently, dietary approaches to reverse type 2 diabetes, pre-
FT Fat tissue serve B cell mass, and achieve remission of glucose alterations
FEV1 Forced expiratory volume-1 minute have gained importance [3, 4, 5••].
FGF21 Fibroblast growth factor 21 Caloric restriction in its two main modalities, continuous
FM Fat mass and intermittent, has been shown to reverse alterations in glu-
FFM Fat-free mass cose homeostasis in animal and human studies [6–8, 9•]. In
GH Growth hormone human protocols, mechanisms are tightly associated with
GLP-1 Glucose-like peptide type 1 weight loss, but animal studies suggest that the beneficial
GSK-3 Glycogen synthase kinase 3 effects of intermittent energy restriction (IER) go beyond
HDL-C High-density lipoprotein cholesterol weight loss, involving mechanisms that permit β cell
HOMA-IR Homeostasis model assessment insulin neogenesis [9•]. The objective of this review is to describe
resistance studies of intermittent fasting (IF) in both animal models and
HADS Hospital Anxiety and Depression Scale humans with diabetes. All animal studies presented here are
HbA1c Glycated hemoglobin derived from murine models where type 2 diabetes was pro-
HR Heart rate voked through either a high-fat diet or using a leptin receptor
IER Intermittent energy restriction knockout mouse model (micedb/db). In these studies, IER was
IER-PF Intermittent energy restriction plus protein and induced with alternated day fasting (ADF) or fasting-
fat ad libitum mimicking diet (FMD) [10]–[12]. In contrast, the human stud-
IF Intermittent fasting ies in subjects with type 2 diabetes (T2DM) [13]–[17] use
IFCR-F Intermittent fasting + caloric restriction + food fasting protocols such as time-restricted feeding (TRF),
based ADF, and 5:2 fasting ratio.
IFCR-L Intermittent fasting + caloric restriction + liquid These different modalities of IER are detailed in this re-
meals view. Special mention is made of a case series recently pub-
IGF Insulin-like growth factor lished where diabetes remission was achieved after an inter-
IL-6 Interleukin 6 mittent fasting period of 6 to 9 months [6••]. Finally, the po-
LBM Lean body mass tential risks of intermittent fasting are addressed, and manage-
LDL-C Low-density lipoprotein cholesterol ment issues are discussed. Figure 1 shows the outcomes re-
MiniAQLQ Mini Asthma Quality of Life Questionnaire ported from animal and human studies.
mTOR Mammalian target of rapamycin
PEF Peak expiratory flow Animal Models of Type 2 Diabetes and IER
PPAR Peroxisome proliferator-activated receptors
REE Respiratory energy expenditure We will review 3 studies with murine models of T2DM. In
SBP Systolic blood pressure one study, obesity was induced with a hypercaloric high-fat
SHBG Sex hormone–binding globulin diet, resulting in insulin resistance and T2DM In the re-
SMBG Self-monitoring blood glucose maining 2 studies, the leptin receptor knockout model was
CSN Central nervous system used. These leptin-resistant mice show obesity and insulin
T2DM Type 2 diabetes mellitus resistance early in life and subsequently develop β cell
TC Total cholesterol failure, hyperglycemia, and T2DM later in life (after 10–
TCA Tricarboxylic acid 12 weeks).
TG Triglycerides Cheng et al exposed micedb/db to FMD for 90 days. The
TNF-α Tumoral necrosis factor-alpha FMD protocol consisted of 4 days of a very low calorie diet
VLDL-C Very low-density lipoprotein cholesterol (low protein and carbohydrate content) alternating with 7 days
vs Versus of normal feed. The authors reported a temporary reduction in
WHO-5 World Health Organization 5 Well-Being Index β cell number, followed by a return to normal levels after re-
feeding. The net result was a significant improvement in β cell
proliferation and function. The investigators speculate that this
Introduction is due to in vivo lineage reprograming, through the activation
of the protein neurogenin 3 (Ngn3). Ngn3+ cells within pan-
In order to reduce cardiometabolic risk in subjects with type 2 creatic islets have been described as progenitor cells able to
diabetes (T2DM) and obesity, the promotion of moderate and generate α and β cells. With this model, a reduction in glucose
sustained weight loss is a primary goal. In this regard, the levels was observed after day 30 and was associated with an
Curr Diab Rep (2020) 20:13 Page 3 of 10 13

Animal models Human Sttudies


Intermittent Energy Restriction

Fastt-mimicking d iet: Very low


Be
enefits: prote
ein and carboh ydrate Bene
efits:
-C
Cell Neogenesiis nt based or not )
(plan HDDL-C
% -cell mass Inssulin Sensitivity
Ins
sulin secretion Quuality of life
HDDL-C Alte
ernated days: Fasting HOOMA-IR
everry other day or 5:2 ratio Faasting Insulin
-C
Cell apoptosis Weight
Fasting glucose Lip
pid profile
Ne
eutral weight Tim
me restricted feeeding:
Fasting most of da y, daily
basiis (18-20 hours )

Fig. 1 Modalities and outcomes from intermittent energy restriction protocols applied in mice and humans. HDL-C, high-density lipoprotein-cholesterol

increase in insulin secretion and HOMA-β, and a proliferation Intermittent Energy Restriction in Humans
of β cells. Between days 30 and 90, improvements in insulin
and glucose tolerance test results were documented. These As a result of experimental IER research, certain intermittent
findings suggest that the two pathophysiological mechanisms fasting methods have been translated into human clinical re-
of T2DM, insulin secretion and insulin resistance, were search and medical practice. Alternate-day fasting consists of
reverted [11••]. Liu et al. exposed a murine model of calorie restriction every other day or calorie restriction 2 days
obesity-induced diabetes (through a high-fat diet), to an IF per week (either two continuous days or two non-continuous
protocol. They reported that this regimen restored the autoph- days (5:2)). Another fasting subtype, time-restricted feeding
agic flux in islets and is associated with an improvement in (TRF), consists of daily fasting during most of the day, with
glucose tolerance by enhancing glucose-stimulated insulin se- food intake times varying from between 4 and 6 h (and fasting
cretion, β cell survival, and nuclear expression of NgN3. They times from between 18 and 20 h). Normally, the time of day
also demonstrated that mice with lysosomal dysfunction sec- for eating is chosen by the subject (this type of fasting resem-
ondary to deficiency of the lysosomal membrane protein do bles Ramadan fasting).
not exhibit these positive effects on β cells, establishing the IER is considered a weight management strategy to pre-
participation of the autophagy pathway in this process [12••]. serve metabolic health and the beneficial effects on cardiomet-
More recently, Wei et al. conducted a study with micedb/db abolic outcomes have been the object of numerous reviews
with an IF protocol based on a modified version of the previ- [18–21]. The effects on subjects with T2DM will be discussed
ously reported FMD regimen implemented by Liu et al. They later in this review.
subjected the mice to the FMD regimen (low protein and
carbohydrate content) every other week for 8 weeks. This
regimen normalized blood glucose levels, and improved insu- Prescription Modalities
lin sensitivity and β cell function. As in Liu’s study, the mice
demonstrated an increment in the expression of β cell progen- Fasting Periods
itor Ngn3. Additionally, the mice showed improvement in
hepatic steatosis. One of the main contributions of this study During these periods, intake can vary from 25% of daily en-
was to show that FMD led to an improvement in the dysbiosis ergy requirements to the consumption of only calorie-free
of T2DM mice. The diet increased the genera of beverages, coffee, and water. Other authors have shown that
Parabacteroides and Blautia while reducing Prevotellaceae, IER may be feasible with caloric restriction of about 50 to
Alistipes, and Ruminococcaceae [10••]. 90% of daily required intake during the restricted period [21,
This evidence shows that IER can effectively influence the 22]. This means an average intake of 500 kcal for women and
progression of diabetes in mice. Interestingly, in the three 600 kcal for men or between 400 and 800 kcals/day.
studies, the effects were independent of weight loss. In fact, Depending on the chosen regimen, fasting may range from
at the end of the intervention period, weight was comparable 16 to 20 h daily (time-restricted feeding) to 24–48 h per week
between groups (intervention vs control; see Table 1). (alternate fasting days).
13
Page 4 of 10

Table 1 Studies evaluating intermittent energy restriction in animal models with type 2 diabetes or insulin resistance

Date, author Mice type, age Exposure Objective Outcome

2007 Tikoo Sprague-Dawley mice, 4 groups: IF: Alternate-day fasting, Fast day: no Effect of IF in progression of diabetic No significant change in weight between groups.
[49] Diabetic IF food allowed Feeding day: AL nephropathy, through Sir2 and p53 Diabetic rats in IF: significant improvement in BUN,
Diabetic AL pathways creatinine, albumin, and HDL-C. Improvement in
Control IF onset of hypertension.
Control AL ↓ Lipid peroxidation, ↑ SOD activity, IF protected
glomerular damage and prevented an increase in
p38 protein. ↓p53 expression and ↑ Sir2 activitya
2017 Cheng Ten-week-old mice Leprdb/db IF: Fasting-mimicking diet (FMD, Effect on β cell regeneration and insulin ↓ Glucosea, ↑ Insulina, ↑ steady-state β cell function
[11••] Two groups: low-protein, low-sugar) 4 days/week resistance. β cell neogenesis through (%B)a. Improvement in insulin and glucose
IF and control alternated with 7 days of regular dog increase of neurogenin-3 (progenitor tolerance testa
chow AL pancreatic cells) Mice in FMD gained less weight than AL mice, but
Control: AL had a 22% greater weight than wild-type mice
2017 Liu Eight-week-old C57bL76J mice. Two (1) group: IF 6 week Effect of IF in glucose metabolism, B cell IF restored autophagic flux in islets and improved
[12••] groups: High-fat diet (12 week), for IF: alternate-day fasting, Fast day: no food mass, and function through glucose tolerance
induction of obesity and glucose allowed, Feeding day: AL autophagy-lysosome pathway ↑ Insulin secretion, β cell survival, islet insulin
intolerance (2) Group AL content, nuclear expression of neurogenin 3a
2018 Wei Six-week-old C57BL/ksJdb/db (1) Fasting-mimicking diet 1 week and Effect on β cell function and contribution of No differences in weight
[10••] Two groups AL 1 week microbiota ↓ Fasting glucosea, ↓glucose during GTT and ITTa, ↓
(2) Standard chow AL HOMA-IRa, ↑% Beta-cell function. Hepatic
steatosis was reduced in FMDa
↑ β cell and α cella ↑ of neurogenin 3a
Phyla: ↑Bacteroidetes and ↓ Firmicutes and
Saccharibacteriaa
Genera: ↑ Parabacteroides, Blautia and
↓Prevotellaceae, Alistipes, Ruminococcaceae
Species: ↑Bacteroidales S24-7, Parabacteroides
distasonis
Curr Diab Rep

a
p = <0.05
ADF alternate-day fasting, BUN blood urea nitrogen, FMD fasting-mimicking diet, GTT glucose tolerance test, HDL-C high-density lipoprotein cholesterol, HOMA-IR homeostatic model assessment of
insulin resistance, ITT insulin tolerance test, IER intermittent energy restriction, Leprdb/db leptin receptor–deficient mice, T2DM type 2 diabetes mellitus, IF intermittent fasting, AL ad libitum, CR calorie
restriction, SOD superoxide dismutase, Sir2 sirtuin type 2
(2020) 20:13
Curr Diab Rep (2020) 20:13 Page 5 of 10 13

Feeding Periods restricted feeding (TRF) method with a regimen of 6 meals per
day. Participants in the TRF phase took breakfast and lunch
Caloric intake during non-restricted times can range from ad (hypocaloric: resting energy expenditure REE * 1.5) while
libitum, hypocaloric (15–30% of energy restriction), those in the control phase took 6 meals per day; each crossover
normocaloric, or hypercaloric (approximately 125% of energy period lasting 12 weeks. Patients were overweight or obesity
requirements) [21]. and had an HbA1c between 6 and 11.8%. The weight de-
creased in both groups but was greater during TRF (− 2.3;
Macronutrient Distribution 95% CI − 2.7, − 2, kg vs − 3.7; 95% CI − 4.1, − 3.4 kg,
p < 0.001). The hepatic fat content (HFC) decrease in both
IER does not have a pre-defined percentage of macronutrients, groups, but again, significantly more so in the TRF group
neither in the fasting nor feeding periods. Different authors (− 0.03%; 95% CI − 0.033%, 10.027% vs − 0.04%; 95% CI
propose diverse schedules. − 0.041%, − 0.035%, p 0.009). The blood glucose and C pep-
Some studies have used the Mediterranean diets (45% en- tide concentrations decreased in both groups, again to a great-
ergy from low glycemic load carbohydrates, 30% fat; 15% er degree during TRF (p = 0.004 and 0.04, respectively).
MUFA, 8% polyunsaturated fatty acids, and 7% saturated fat- Glucagon decreased in TRF and increased during the 6 meal
ty acids) during fasting and feeding periods with periods (p 0.001). Interestingly, both regimens were isocalo-
hypoenergetic and isoenergetic intake, respectively. Other in- ric, but the consumption of calories in two meals/day instead
terventions significantly reduce the carbohydrate consump- of six meals/day was associated with better metabolic im-
tion during fasting times (less than 50 g/day) or increase pro- provement [14].
tein intake during feeding. Diet protocols are heterogeneous Carter et al. published a randomized controlled trial (RCT)
and should be individualized according to the target popula- with 63 overweight or obese T2DM patients and tested the
tion (i.e., obesity, type 2 diabetes, aging). fasting protocol 5:2 ratio (non-continuous days), described
Although information is abundant regarding obesity, insu- earlier, versus continuous hypocaloric diet. They demonstrat-
lin resistance, and other components of the metabolic syn- ed similar reductions in weight (− 5.9 ± 4%, p < 0.001),
drome associated with obesity, the next section only describes HbA1c (− 0.7 ± 0.9%, p < 0.001), and appetite over time, in
those studies where the subjects had established T2DM. favor of the non-continuous method. Furthermore, the in-
creases in the sensation of fullness and satisfaction were sim-
IER in Type 2 Diabetes Mellitus ilar [15].
Arnason et al. carried out a pilot study with 10 T2DM
Owing to the benefits in insulin response and glucose regula- patients between the ages of 40 and 60 years with a before-
tion, interest has grown for the implementation of these re- after design, including 3 phases: 2 weeks (wk) of normal con-
gimes in patients with type 2 diabetes mellitus (T2DM). All sumption, 2 wk of TRF (18–20 h) and finally 2 wk of normal
modalities of IER (previously described) have been evaluated consumption. They were interested in evaluating the clinical
in this population. tolerability and biochemical effects of TRF. They found a
Saada et al. studied a population that normally fasts for reduction in BMI (− 0.517, p = 0.013), morning fasting glu-
religious motives. They studied 66 overweight women with cose measured by self-monitoring blood glucose ((SMBG),
type 2 diabetes mellitus. The participants were studied the the increased fasting duration improved at goal, < 7 mmol/L,
week before and during the third week of Ramadan. Even morning SMBG to 34.1%, from a baseline of 13.8%) during
though no changes in body mass index (BMI) were observed the TRF phase. There was no hypoglycemia or serious side
during Ramadan or during the non-fasting days, a significant effects. Interestingly, although not a study requirement, all
decrease in glycated hemoglobin (HbA1c) was reported (9.52 participants preferentially chose eating hours starting in the
vs 8.78%, p = 0.007). The glucose levels were significantly midafternoon [16]. The last study in IF in patients with
higher during the fasting period (pre-Ramadan 1.84 ± 0.20 T2DM was published by Li et al., a RCT with 46 T2DM
vs Ramadan 1.98 ± 0.17 g/L, p < 0.05); this could be second- patients between 25 and 75 years. The regimen had duration
ary to the significant decrease (14.62 ± 2.79 vs 11.97 ± of 4 months and consisted of 1 week of fasting and 4 months
2.48 μU/mL, p < 0.05) in insulin levels. The high-density li- of follow-up. They used the Buchinger method plus 3 weeks
poprotein cholesterol (HDL-C) levels increased significantly of Mediterranean diet. This was compared with a control
(p < 0.05) demonstrating the beneficial effect of Ramadan on group assigned to 4 months of a Mediterranean diet. They
lipid profile. The augmentation in HDL-C can be explained by found a significant reduction in body weight (3.5 ± 4.5 vs 2
the improvement in insulin resistance (suggested by the reduc- ± 4.8 kg p = 0.03), waist size (− 4.4 ± 4.3 vs − 0.3 ± 2 p =
tion in insulin levels) [13]. 0.001), systolic blood pressure (SBP, − 13.9 ± 15.3 vs 0.4 ±
Kahleova et al. studied 54 patients between 30 and 70 years, 15 p = 0.02), and diastolic blood pressure (DBP, − 9 ± 12.3 vs
in a parallel group crossover study design comparing the time- 3.2 ± 11). Results also included a significant improvement in
13 Page 6 of 10 Curr Diab Rep (2020) 20:13

quality of life (WHO-5 well-being index, WHO-5 p = 0.04). should remain unchanged. Blood glucose level monitoring at
No differences in HbA1c were documented and no hypogly- least twice daily is essential during fasting. Patients should
cemia or serious adverse effects were informed. break the IER diet if dangerously low glycemic levels occur
The most consistent finding among the 5 studies was a [27, 28].
reduction in insulin levels after IF, reflected in a reduction in In addition to the hypoglycemia risk described above, there
HOMA index, suggesting an improvement in peripheral insu- is concern that there may be a loss of fat-free mass (FFM) with
lin action in and/or suppression of insulin secretion in order to the use of IER. One study reported a FFM loss of up to 27%
avoid hypoglycemia and/or activate lipolysis as a source of on IER despite an intake of 0.7 g of protein/kg [21]. In order to
energy in food deprivation conditions. Special mention must limit this loss, exercise should be included in the regimen of
be made of a recently published case series of three Canadian patients who are on an IER diet [29]. Other reports suggest
patients who achieved T2DM remission after a protocol of that increasing protein intake to 1.2 g/kg can reduce FFM loss
intermittent fasting based on ADF for 7–11 months. Two of to < 20%. Short-term studies of 3–4 months follow-up using
the patients were insulin-dependent, suggesting late-stage diets with 20–25% protein report higher satiety and a better
T2DM. The effect was widely attributable to weight loss re- preservation of FFM, compared with low-protein diets
duction [5••]. The previously discussed studies are summa- [30–32]. IER may be of greater risk in malnourished patients,
rized in Table 2. where it could lead to severe vitamin deficiencies as well as
FFM loss. This may be compensated by supplementation with
Risks of IER multivitamins and the prescription of strength exercises [32].
Heilbronn et al. suggest that IER may not be feasible as a
Careful monitoring of blood glucose levels during the fasting general public health intervention; they observed that self-
period is the most important aspect for patients taking phar- reported hunger on fasting days was considerable and did
macological agents that can cause hypoglycemia. Fasting by not decrease over time [33]. Dorighello et al. showed that
T2DM Muslim patients during Ramadan has motivated the IER is not beneficial in the context of genetic hypercholester-
development of guidelines for diabetes management during olemia [34]. In young women with normal weight, IER pre-
fasting [23, 24]. Typically, patients have been stratified ac- scribed with 70% of caloric restriction in consecutive days and
cording to hypoglycemic risk in four categories. Patients in 3 days of ad libitum eating increased feelings of hunger, wors-
very high and high risk of hypoglycemia should not follow the ened mood, heightened irritability, impaired ability to focus,
Ramadan fast, but studies demonstrate that most of the pa- increased fatigue, increased eating-related thoughts, and pro-
tients decide to take the risk [25, 26]. Therefore, careful mon- voked a fear of loss of control and overeating during non-
itoring is advised in order to avoid hypoglycemia, hypergly- restricted days [21].
cemia, or ketoacidosis. Ramadan fasting differs from clinical
fasting because Muslims must abstain from eating and drink- Differences between Animal Models and Clinical
ing during the daylight hours of dawn to sunset. Meanwhile, Studies in the Study of Glucose Homeostasis
in clinical fasting, the hour of fasting can be chosen for the
person, and liquids are permitted, thus avoiding the risk of Animal models have focused on β cell function, while clinical
dehydration. Guidelines emphasize a structured education that studies have explored peripheral insulin resistance as the main
includes risk assessment, use of SMBG, indications on when outcome to elucidate whether intermittent fasting promotes
to break the fast, when to exercise, fluid intake, and meal improvements in glucose homeostasis. Methods in mice have
planning and information on medication adjustment during included combinations of in vivo and in vitro techniques. In
fasting. In addition, other authors have discussed the manage- humans, in vitro techniques would implicate obtaining pan-
ment of T2DM during other types of fasting highlighting the creatic tissue which is not feasible from a clinical and ethical
selection and adjustment of oral hypoglycemic agents, accord- viewpoint. Although testing for β-function using in vivo ex-
ing to HbA1c levels [24]. periments is feasible in humans, to the best of our knowledge,
Short-acting secretagogues, such as nateglinide, can be this has not been done in currently published IF literature.
used safely before meals without an increased risk of hypo- There is no gold standard for measuring β cell function and
glycemia. Dosing of sulphonylureas with a longer duration of mass; in vitro approaches utilizing isolated human islets are
action (for example, glyburide) should be reduced or time- currently the method of choice for most researchers [35]. The
changed to include the largest dose before the evening meal. study of pancreatic islets can provide information regarding
In patients using insulin and sulphonylureas, the discontinua- the different stages of disease. For example, obesity and insu-
tion of the sulfonyl urea is suggested if baseline HbA1c is < lin resistance are characterized by an increased size and num-
7%. If HbA1c is between 7 and 10%, discontinuation of ber of β cells, whereas in T1DM and late-stage T2DM, an
sulphonylureas and rapid-action insulin is recommended only increase in apoptosis is seen. Β cell proliferation, apoptosis,
during the fasting periods. If HbA1c is > 10%, medications hormone secretion, signal transduction pathway, protein
Table 2 Studies evaluating intermittent energy restriction in human T2DM subjects

Date, author Population Exposure Main outcomes Effects


Curr Diab Rep

design

2009 Saada 66 overweight Ramadan fasting: Two meals between Effect of Ramadan fasting on glucose Significant ↓ HbA1c, TC, TG, and LDL-C. Significant ↑ fasting glucose and HDL-C. No
[13] women sunset and dawn, without restrictions control, lipids, and weight difference in weight
48 ± 2 years
Before and after
2014 N = 54 Time-restricted feeding: Breakfast and Effect of TRF on weight, hepatic fat BW decrease in both groups, more for TRFa
(2020) 20:13

Kahleova 30–70 years lunch (hypocaloric REE*1.5) content, insulin resistance, and β HFC decrease in both groups, more in TRFa
[14] Parallel groups Control group: 6 smaller meals function Fasting glucose and C-peptide ↓ in both groups, more in TRFa glucagon ↓ in TRFa and ↑
RCT (hypocaloric REE*1.5) in control groupa. OGIS ↑ in both, more in TRF
12 weeks
2016 Carter 63 overweight 5:2 intermittent fasting: IER vs CER in HbA1c ↓ HbA1c, BW, and appetite over time similar in both groupsa
[15] or obese IER: 2 days of 1670–2500 kj/day with ↑ Feeling of fullness and satisfaction over time similar in both groupsa
patients 5 days of AL
61 ± 9 years CER: 7 days of 5000–6500 kj/day
2 parallel group
RCT
12 weeks
2017 10 patients Time-restricted fasting: IER biochemical effects and clinical ↓ BW, BMI, SMBG in IER phasea
Arnason 53.8 ± 9.1 years 3 phases: tolerability Change in HFD correlated with SMBG morning readings (x2 likelihood ratio = 8.36)a but
[16] Before-after (1) 2 weeks normal dietary habits not for afternoon or evening SMBG. Good tolerability
design (2) 2 weeks 18–20 h fasting goal per day
6 weeks (3) 2 weeks normal dietary habits
2017 Li [17] 46 Modified intermittent fasting: 1 week fasting vs usual care in T2DM Significant ↓ in BW (3.5 ± 4.5 kg) in fasting groupa vs (2.0 ± 4.8 kg) in control group
25–75 years Two groups: Significant ↓ waist, SBP, and DBP
2 parallel group Control: 4 months of Mediterranean diet Significant ↑ WHO-5 in fasting groupa
RCT Fasting: 1 week of fasting (Buchinger ↓ BMIa, glucose, insulin, and HOMA-IR
4 months method) plus 3 weeks of No difference in HbA1c
Mediterranean diet
2018 Furmli Case series Patient 1: 3 days/week fasting for Effects of IF in control glucose Two patients with reversal of T2DM (control without insulin or medications). One patient
[5••] 3 patients 7 months with control with oral medication. Significant reduction in weight and waist
11 months Patient 2: 3 days/week fasting for circumference
11 months
Patient 3: Alternate-day fasting
11 months

a
p = <0.05
ADF alternate-day fasting, ALT alanine aminotransferase, AST aspartate aminotransferase, BDNF brain-derived neurotrophic factor, BF body fat, BMI body mass index, BW body weight, BUN blood urea
nitrogen, CER continuous energy restriction, CRP C-reactive protein, CV cardiovascular, DBP diastolic blood pressure, FFA free fatty acid, FM fat mass, FFM fat-free mass, HDL-C high-density
lipoprotein cholesterol, HFC hepatic fat content, HFD hour fasting difference, HOMA-IR homeostasis model assessment insulin resistance, HbA1c glycated hemoglobin, HR heart rate, IER intermittent
energy restriction, LDL-C low-density lipoprotein cholesterol, OGIS oral glucose insulin sensitivity, RCT randomized controlled trial, REE resting energy expenditure, SBP systolic blood pressure, SMBG
Page 7 of 10

self-monitoring blood glucose, T2DM type 2 diabetes mellitus, TC total cholesterol, TG triglycerides, TRF time-restricted feeding, WHO-5 World Health Organization 5 Well-Being Index, vs versus, IF
13

intermittent fasting, AL ad libitum, CR calorie restriction, SD standard diet, WL weight loss, BPL blood pressure levels, VLDL-C very low-density lipoprotein cholesterol
13 Page 8 of 10 Curr Diab Rep (2020) 20:13

localization (by immunohistochemical), and protein expres- These phenomena are associated with an increased expression
sion (by western blot) are some of the biological activities that of Ngn3 in islet cells. Ngn3 is a transcription factor expressed
can be investigated in in vitro models. Transcriptomics and transiently in developing pancreatic islets in utero that is re-
single-cell analysis have permitted the identification of new quired for the development of β and α cells. This benefit is
transcriptional regulators and specific cellular changes and abolished if autophagy is suppressed suggesting that fasting-
subpopulations that would be undetectable when analyzed in induced autophagy is a key mediator. There is reason to sus-
whole islets, respectively. With some of these techniques, a pect that autophagy suppresses the Nothc1 signaling pathway,
de-differentiated phenotype has been observed, supporting the and this promotes Ngn3 expression and β cell neogenesis [9•].
possible contribution of the de-differentiation of β and α cells The Notch signaling pathway plays a crucial role at different
in T2DM [36]. stages of cell development, such as proliferation, growth, dif-
In vivo, β cell function can be evaluated measuring ferentiation, and apoptosis.
fasting insulin, oral glucose tolerance test (OGTT), mixed In contrast to animal models, clinical studies have focused
meal tolerance test, intravenous glucose tolerance test on body weight and parameters associated with insulin action,
(IVGTT), and the hyperglycemic clamp. The β cell mass more than insulin secretion.
and function can also be evaluated with non-invasive imag- Clinical studies including T2DM consistently demonstrate
ing techniques; positron emission tomography (PET), single- a reduction in fasting insulin levels and HOMA index, an
photon emission computed tomography (SPECT), and mag- effect that is widely associated with weight loss, with the
netic resonance imaging (MRI) are considered the most exception of the findings of Saada et al. who reported a reduc-
promising imaging methods to achieve this purpose [35]. tion in insulin levels without loss weight [13]. We believe that
Both the evaluation of function and mass is feasible in hu- hypoinsulinemia results from weight loss consequently reduc-
man clinical studies. ing insulin resistance. Additionally, low levels of insulin are a
defense mechanism for the avoidance of hypoglycemia.
Hypoinsulinemia also activates lipolysis amid food depriva-
Discussion tion in order to utilize free fatty acids as a source of energy.
Low concentrations of insulin levels after IF can be seen in
Recently, there has been a great deal of interest in fasting insulin-resistant patients without T2DM [40]. The increase in
regimes, in particular, forms of IER. However, this is not C-HDL levels observed in studies where this parameter has
new; in the late nineteenth century, Edward Hooker Dewey, been measured also supports our theory of a reduction in pe-
the American physician who pioneered cyclic caloric restric- ripheral and hepatic insulin resistance. To our knowledge,
tion, advocated fasting every day until late noon. Similarly, in there are no clinical studies that attempt to investigate the
the early twentieth century, the French physician Guillaume possible recovery of β cell number and function. However,
Guelpa practiced continuing cycles of fasting for 2–5 days, studies including the hyperglycemic clamp, IVGTT, OGTT,
followed by vegetarian food for 12–15 days [37]. The molec- and mixed meal tolerance test could help to answer some
ular mechanisms of IER have been demonstrated in animal questions regarding β cell function. Functional studies can
models and involve adaptive cellular responses that reduce be complemented by evaluating β cell mass with non-
oxidative damage and inflammation, optimizing energy me- invasive imaging techniques like PCT, SPECT, and MRI.
tabolism and favor cellular protection. The main pathways Several physiological responses to fasting are similar to those
studied involve an increase in autophagy mediated through caused by regular aerobic exercise, which include an improve-
an increase in the sirtuin (SIRT-1), a NAD+-dependent ment in insulin sensitivity and cellular stress, and reduced
deacetylase [38]. In laboratory murine models, IER has pro- resting blood pressure and heart rate [41]. All IER modalities
found beneficial effects on many different indices of health have shown a positive impact on obesity, but few studies have
and can prevent some disease processes and improve func- evaluated the additive effects of exercise and other interven-
tional outcomes in experimental models of aging and a wide tions. Hence, more integrative studies of IER which include
range of age-related diseases such as diabetes, cardiovascular lifestyle interventions are needed. We recommend that health
disease, cancers, and neurological conditions (Alzheimer’s providers choose protocols that are individualized to patient
disease and stroke) [39]. preferences and complement the nutritional approach with
This review has aimed to describe the current evidence exercise in all cases.
regarding diets based on intermittent fasting, focusing on In T2DM, time-restricted feeding protocols are more con-
diabetes-induced animal models and human studies in persons venient and safer, since patients eat every day and the fasting
with T2DM. In the animal models, mechanisms for the rever- periods are not long enough to significantly increase hypogly-
sion of glucose alterations have been studied and authors have cemia risk. Hunger can be a barrier for therapy adherence, so
described the neogenesis of β cells, a reduction in β cell we recommend the systematic evaluation of hunger and satiety
apoptosis and improvements in insulin islet β cell content. in patients with IER diets.
Curr Diab Rep (2020) 20:13 Page 9 of 10 13

IER diets are not risk-free. Beyond hypoglycemia, the main 2. Wing R, Lang W, Wadden T, Safford M, Knowler W, Bertoni A,
et al. Benefits of modest weight loss in improving cardiovascular
adverse effects involve psychologic and anatomical/
risk factors in overweight and obese individuals with type 2 diabe-
physiological disturbances like anxiety and muscle loss. tes. Diabetes Care. 2011;34(7):1481–6.
Additionally, in women, it is important to consider the impact 3. Ajala O, English P, Pinkney J. Systematic review and meta-analysis
of IER on menstrual cycles. Multidisciplinary treatment can of different dietary approaches to the management of type 2 diabe-
mitigate most of the deleterious effects, including psychologic tes. Am J Clin Nutr. 2013;97(3):505–16.
4. N. D. Guess, “Dietary interventions for the prevention of type 2
distress imposed by diets. Approaches to manage risks depend diabetes in high-risk groups: current state of evidence and future
on diet and patient characteristics. Proteins have a positive research needs,” Nutrients, vol. 10, no. 9, 2018.
effect on satiety and muscle mass, and supplementation with 5.•• S. Furmli, R. Elmasry, M. Ramos, and J. Fung, “Therapeutic use
essential amino acids and vitamins is encouraged when low of intermittent fasting for people with type 2 diabetes as an alter-
protein diets are prescribed. native to insulin,” BMJ Case Rep., p. bcr-2017-221854, 2018.
This Canadian group, demostrated for first time, in a clinical con-
Independent to promising evidence regarding β cell func- text, the capacity of diets based in intermittent fasting to reverse
tion derived from murine models, numerous evidence support type 2 diabetes. Previous findings suggested same results for
that a weight loss of 7–10% is accompanied by improvements diets of very low calorie intake in a continuos rather an inter-
in several metabolic parameters such as insulin action, lipid mittent modality.
6. Lim EL, Hollingsworth KG, Aribisala BS, Chen MJ, Mathers JC,
profile and blood pressure, diminution of progression from Taylor R. Reversal of type 2 diabetes: normalisation of beta cell
pre-diabetes to diabetes, and diminution on diabetes progres- function in association with decreased pancreas and liver triacyl-
sion among others [1, 42–46]. Changes in lifestyle behaviors glycerol. Diabetologia. 2011;54(10):2506–14.
oriented to weigh loss are recommended for all patients with 7. Barnosky AR, Hoddy KK, Unterman TG, Varady KA. Intermittent
fasting vs daily calorie restriction for type 2 diabetes prevention: a
diabetes or at increased risk to develop it.
review of human findings. Transl Res. 2014;164(4):302–11.
In conclusion, this report supports the use of strictly super- 8. Steven S, Taylor R. Restoring normoglycaemia by use of a very low
vised IER in persons with T2DM; this technique aids in gly- calorie diet in long- and short-duration type 2 diabetes. Diabet Med.
cemic control and helps to minimize the use of insulin and oral 2015;32(9):1149–55.
hypoglycemic agents (OHA). The main concern during 9.• J. J. Dinicolantonio and M. Mccarty, “Open access autophagy-
induced degradation of Notch1, achieved through intermittent
fasting is the incidence of hypoglycemia, especially when reg-
fasting, may promote beta cell neogenesis: implications for rever-
imens include insulin or insulin secretagogues. Fortunately, sal of type 2 diabetes,” Open Hear., vol. 6, p. 1028, 2019. This
there are other OHAs with a very low risk of hypoglycemia, review higlights the mechanisms trough diets based in
such as SGLT2 inhibitors and DPP-IV inhibitors. Finally, clin- termittent fasting promotes reversion of type 2 diabetes, im-
proving beta cell function.
ical studies are needed which evaluate the functional capacity
10.•• S. Wei, R. Han, J. Zhao, S. Wang, M. Huang, Y. Wang, and Y.
and mass of β cells in order to corroborate the findings in Chen, “Intermittent administeration of a fasting mimicking diet
animal models. The techniques to evaluate in vivo function intervens in M progression and restores b cells .pdf,” pp. 1–12,
and the new methods to evaluate mass are promising in the 2018. The authors confirmed the results of Cheng 2017, dem-
quest to answer these questions. onstrating an increment in the expression of β cell progenitor
Ngn3. Additionally, the mice showed improvement in hepatic
steatosis. One of the main contributions of this study was to
Compliance with Ethical Standards show than FMD led to an improvement in the dysbiosis of
T2DM mice.
Conflict of Interest The authors declare that they have no conflicts of 11.•• C. W. Cheng, V. Villani, R. Buono, M. Wei, S. Kumar, O. H.
interest. Yilmaz, P. Cohen, J. B. Sneddon, L. Perin, and V. D. Longo,
“Fasting-mimicking diet promotes Ngn3-driven β-cell regenera-
Human and Animal Rights and Informed Consent This article does not tion to reverse diabetes,” Cell, vol. 168, no. 5, p. 775–788.e12,
contain any studies with human or animal subjects performed by any of 2017. The authors demonstrated (for the first time in an animal
the authors. model) that a diet based in intermittent fasting results in resolution
of type 2 diabetes. The net result of their experiments was a sig-
nificant improvement in β cell proliferation and function.
12.•• H. Liu, A. Javaheri, R. J. Godar, J. Murphy, X. Ma, N. Rohatgi, J.
Mahadevan, K. Hyrc, P. Saftig, C. Marshall, M. L. McDaniel, M.
References S. Remedi, B. Razani, F. Urano, and A. Diwan, “Intermittent
fasting preserves beta-cell mass in obesity-induced diabetes via
the autophagy-lysosome pathway,” Autophagy, vol. 13, no. 11,
Papers of particular interest, published recently, have been pp. 1952–1968, 2017. They reported that intermittent fasting
highlighted as: restored the autophagic flux in islets enhancing glucose-
• Of importance stimulated insulin secretion, β cell survival, and nuclear ex-
pression of NgN3. Their main contribution demonstrates that
•• Of major importance mice with lysosomal dysfunction secondary to deficiency of
the lysosomal membrane protein do not exhibit these positive
1. Liebermeister H. Effects of weight-reduction on obesity-associated effects on β cells, establishing the participation of the autoph-
diseases. Ger Med Sci. 2003;1:1–4. agy pathway in this process.
13 Page 10 of 10 Curr Diab Rep (2020) 20:13

13. A. saada, S. attou, and A. chabane, “Effect of Ramadan fasting on diabetes: one-year follow-up of a randomised trial. Diabetologia.
glucose, glycosylated haemoglobin, insulin, lipids and proteinous 2004;47(10):1677–86.
concentrations in women with non-insulin dependent diabetes 31. Larsen RN, Mann NJ, Maclean E, Shaw JE. The effect of high-protein,
mellitus,” African J Biotechnol, vol. 9, no. 1, pp. 87–94, 2010. low-carbohydrate diets in the treatment of type 2 diabetes: a 12 month
14. Kahleova H, Belinova L, Malinska H, Oliyarnyk O, Trnovska J, randomised controlled trial. Diabetologia. 2011;54(4):731–40.
Skop V, et al. Eating two larger meals a day (breakfast and lunch) is 32. Collier R. Intermittent fasting: the next big weight loss fad. CMAJ.
more effective than six smaller meals in a reduced-energy regimen 2013;185(8):321–2.
for patients with type 2 diabetes: a randomised crossover study. 33. Heilbronn LK, Smith SR, Martin CK, Anton SD, Ravussin E.
Diabetologia. 2014;57(8):1552–60. Alternate-day fasting in nonobese subjects: effects on body weight,
15. Carter S, Clifton PM, Keogh JB. The effects of intermittent com- body composition, and energy metabolism. Am J Clin Nutr.
pared to continuous energy restriction on glycaemic control in type 2005;81(1):69–73.
2 diabetes; a pragmatic pilot trial. Diabetes Res Clin Pract. 34. Dorighello GG, Rovani JC, Luhman CJF, Paim BA, Raposo HF,
2016;122:106–12. Vercesi AE, et al. Food restriction by intermittent fasting induces
16. Arnason TG, Bowen MW, Mansell KD. Effects of intermittent diabetes and obesity and aggravates spontaneous atherosclerosis
fasting on health markers in those with type 2 diabetes: a pilot study. development in hypercholesterolaemic mice. Br J Nutr.
World J Diabetes. 2017;8(4):154–64. 2014;111(6):976–86.
17. Li C, Sadraie B, Steckhan N, Kessler C, Stange R, Jeitler M, et al. 35. Chen C, Cohrs CM, Stertmann J, Bozsak R, Speier S. Human beta
Effects of a one-week fasting therapy in patients with type-2 diabetes cell mass and function in diabetes: recent advances in knowledge
mellitus and metabolic syndrome – a randomized controlled explor- and technologies to understand disease pathogenesis. Mol Metab.
ative study. Exp Clin Endocrinol Diabetes. 2017;125(9):618–24. 2017;6(9):943–57.
18. Mattson MP, Wan R. Beneficial effects of intermittent fasting and 36. M. Diedisheim, M. Oshima, O. Albagli, C. W. Huldt, I. Ahlstedt,
caloric restriction on the cardiovascular and cerebrovascular sys- M. Clausen, S. Menon, A. Aivazidis, A. C. Andreasson, W. G.
tems. J Nutr Biochem. 2005;16(3):129–37. Haynes, P. Marchetti, L. Marselli, M. Armanet, F. Chimienti, and
19. M. R. Soeters, N. M. Lammers, P. F. Dubbelhuis, T. Ackermans, C. R. Scharfmann, “Modeling human pancreatic beta cell dedifferen-
F. Jonkers-schuitema, E. Fliers, H. P. Sauerwein, J. M. Aerts, and tiation,” Mol. Metab., vol. 10, no. February, pp. 74–86, 2018.
M. J. Serlie, “Intermittent fasting does not affect whole-body glu- 37. Wilhelmi De Toledo F, Buchinger A, Burggrabe H, Hölz G, Kuhn
cose , lipid , or protein metabolism,” Am J Clin Nutr, vol. 90, pp. C, Lischka E, et al. Fasting therapy - an expert panel update of the
1244–1251, 2009. 2002 consensus guidelines. Forsch Komplementarmed.
20. N. Halberg, M. . Henriksen, N. . Söderhamn, B. . Stallknecht, T. . 2013;20(6):434–43.
Ploug, P. . Schjerling, and F. . Dela, “Effect of intermittent fasting 38. S. Golbidi, A. Daiber, B. Korac, H. Li, M. F. Essop, and I. Laher,
and refeeding on insulin action in healthy men,” J Appl Physiol, “Health benefits of fasting and caloric restriction,” Current
vol. 99, no. 6, pp. 2128–2136, 2005. Diabetes Reports. 2017.
21. M. Harvie and A. Howell, “Potential benefits and harms of intermit- 39. M. P. Mattson, V. D. Longo, and M. Harvie, “Impact of intermittent
tent energy restriction and intermittent fasting amongst obese, over- fasting on health and disease processes,” Ageing Res. Rev., 2016.
weight and normal weight subjects—a narrative review of human and
40. K. Gabel, C. M. Kroeger, J. F. Trepanowski, K. K. Hoddy, S.
animal evidence,” Behav. Sci. (Basel)., vol. 7, no. 1, p. 4, 2017.
Cienfuegos, F. Kalam, and K. A. Varady, “Differential effects of
22. M. C. Klempel, C. M. Kroeger, S. Bhutani, J. F. Trepanowski, and
alternate-day fasting versus daily calorie restriction on insulin resis-
K. a Varady, “Intermittent fasting combined with calorie restriction
tance,” Obesity, vol. 0, no. 0, p. oby.22564, 2019.
is effective for weight loss and cardio-protection in obese women.,”
41. Longo VD, Mattson MP. Fasting : molecular mechanism and clin-
Nutr. J., vol. 11, no. 1, p. 98, 2012.
ical application. Cell Metab. 2015;19(2):181–92.
23. Ali S, Davies MJ, Brady EM, Gray LJ, Khunti K, Beshyah SA,
42. Gregg EW, Shaw JE. Global health effects of overweight and obe-
et al. Guidelines for managing diabetes in Ramadan. Diabet Med.
sity. N Engl J Med. 2017;377(1):80–1.
2016;33(10):1315–29.
24. Hassanein M, Al-Arouj M, Hamdy O, Bebakar WMW, Jabbar A, 43. Diabetes Prevention Program Research Group. Reduction in the
Al-Madani A, et al. Diabetes and Ramadan: practical guidelines. incidence of type 2 diabetes with lifestyle intervention or metfor-
Diabetes Res Clin Pract. 2017;126:303–16. min. N Engl J Med. 2002;346(6):393–403.
25. Jabbar A, Hassanein M, Beshyah SA, Boye KS, Yu M, Babineaux 44. Y. M. Khazrai, G. Defeudis, and P. Pozzilli, “Effect of diet on type 2
SM. CREED study: hypoglycaemia during Ramadan in individuals diabetes mellitus: a review,” Diabetes/Metabolism Res. Rev., vol.
with type 2 diabetes mellitus from three continents. Diabetes Res 30, no. 2, p. 24–33 10p, 2014.
Clin Pract. 2017;132:19–26. 45. D. P. P. R. Diabetes Prevention Program Research Group, D. M.
26. Adnan Z. Risk stratification of patients with diabetes and the role of Nathan, E. Barrett-Connor, J. P. Crandall, S. L. Edelstein, R. B.
sodium glucose co-transporter inhibitors 2 during Ramadan fasting. Goldberg, E. S. Horton, W. C. Knowler, K. J. Mather, T. J.
Diabetes Res Clin Pract. 2017;131:217–8. Orchard, X. Pi-Sunyer, D. Schade, and M. Temprosa. Long-term
27. S. Carter, P. M. Clifton, and J. B. Keogh, “Intermittent energy effects of lifestyle intervention or metformin on diabetes develop-
restriction in type 2 diabetes: a short discussion of medication man- ment and microvascular complications over 15-year follow-up: the
agement,” World J. Diabetes, vol. 7, no. 20, p. 627, 2016. diabetes prevention program outcomes study. Lancet Diabetes
28. Olansky L. Strategies for management of intermittent fasting in Endocrinol. 2015;3(11):866–75.
patients with diabetes. Cleve Clin J Med. 2017;84(5):357–8. 46. K. E. Orchard, T. J., Temprosa, M., Barrett-Connor, E., Fowler, S.
29. Bhutani S, Klempel MC, Kroeger CM, Trepanowski JF, Varady E., Goldberg, R. B., Mather, K. J., ... & Watson, “Long-term effects
KA. Alternate day fasting and endurance exercise combine to re- of the diabetes prevention program interventions on cardiovascular
duce body weight and favorably alter plasma lipids in obese risk factors: a report from the DPP outcomes study,” Diabet Med,
humans. Obesity. 2013;21(7):1370–9. vol. 30, no. 1, pp. 46–55, 2014.
30. Brinkworth GD, Noakes M, Parker B, Foster P, Clifton PM. Long-
term effects of advice to consume a high-protein, low-fat diet, rather Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
than a conventional weight-loss diet, in obese adults with type 2 tional claims in published maps and institutional affiliations.

You might also like