ArticlePDF Available

Abstract and Figures

Creatine supplementation improves glucose tolerance in healthy subjects. The aim was to investigate whether creatine supplementation has a beneficial effect on glycemic control of type 2 diabetic patients undergoing exercise training. A 12-wk randomized, double-blind, placebo-controlled trial was performed. The patients were allocated to receive either creatine (CR) (5 g·d) or placebo (PL) and were enrolled in an exercise training program. The primary outcome was glycosylated hemoglobin (HbA1c). Secondary outcomes included the area under the curve of glucose, insulin, and C-peptide and insulin sensitivity indexes. Physical capacity, lipid profile, and GLUT-4 protein expression and translocation were also assessed. Twenty-five subjects were analyzed (CR: n=13; PL: n=12). HbA1c was significantly reduced in the creatine group when compared with the placebo group (CR: PRE=7.4 ± 0.7, POST=6.4 ± 0.4; PL: PRE=7.5 ± 0.6, POST=7.6 ± 0.7; P=0.004; difference=-1.1%, 95% confidence interval=-1.9% to -0.4%). The delta area under the curve of glucose concentration was significantly lower in the CR group than in the PL group (CR=-7790 ± 4600, PL=2008 ± 7614; P=0.05). The CR group also presented decreased glycemia at times 0, 30, and 60 min during a meal tolerance test and increased GLUT-4 translocation. Insulin and C-peptide concentrations, surrogates of insulin sensitivity, physical capacity, lipid profile, and adverse effects were comparable between the groups. Creatine supplementation combined with an exercise program improves glycemic control in type 2 diabetic patients. The underlying mechanism seems to be related to an increase in GLUT-4 recruitment to the sarcolemma.
Content may be subject to copyright.
Creatine in Type 2 Diabetes: A Randomized,
Double-Blind, Placebo-Controlled Trial
BRUNO GUALANO
1,2
, VITOR DE SALLES PAINNELI
1,2
, HAMILTON ROSCHEL
1,2
,
GUILHERME GIANNINI ARTIOLI
1,2
, MANOEL NEVES JR
2
, ANA LU
´CIA DE SA
´PINTO
2
,
MARIA ELIZABETH ROSSI DA SILVA
3
,MARIAROSA
´RIA CUNHA
3
, MARIA CONCEPCIO
´NGARCI
´AOTADUY
4
,
CLAUDIA DA COSTA LEITE
4
,JU
´LIO CE
´SAR FERREIRA
1
, ROSA MARIA PEREIRA
2
, PATRI
´CIA CHAKUR BRUM
1
,
ELOISA BONFA
´
2
, and ANTONIO HERBERT LANCHA JR
1
1
Laboratory of Applied Nutrition and Metabolism, School of Physical Education and Sports, University of Sa˜o Paulo,
Sa˜o Paulo, BRAZIL;
2
Division of Rheumatology, School of Medicine, University of Sa˜ o Paulo, Sa˜ o Paulo, BRAZIL;
3
Division of Endocrinology Laboratory of Medical Investigation, School of Medicine, University of Sa˜o Paulo, Sa˜o Paulo,
BRAZIL; and
4
Division of Radiology, School of Medicine, University of Sa˜ o Paulo, Sa˜ o Paulo, BRAZIL
ABSTRACT
GUALANO, B., V. DE. SALLES PAINNELI, H. ROSCHEL, G. G. ARTIOLI, M. NEVES JR, A. L. DE SA
´PINTO, M. E. DA SILVA,
M. R. CUNHA, M. C. G. OTADUY, C. DA COSTA LEITE, J. C. FERREIRA, R. M. PEREIRA, P. C. BRUM, E. BO NFA
´,and
A. H. LANCHA JR. Creatine in Type 2 Diabetes: A Randomized, Double-Blind, Placebo-Controlled Trial. Med. Sci. Sports Exerc.,
Vol. 43, No. 5, pp. 770–778, 2011. Creatine supplementation improves glucose tolerance in healthy subjects. Purposes:Theaimwas
to investigate whether creatine supplementation has a beneficial effect on glycemic control of type 2 diabetic patients undergoing
exercise training. Methods: A 12-wk randomized, double-blind, placebo-controlled trial was performed. The patients were allocated
to receive either creatine (CR) (5 gId
j1
) or placebo (PL) and were enrolled in an exercise training program. The primary outcome
was glycosylated hemoglobin (Hb
A1c
). Secondary outcomes included the area under the curve of glucose, insulin, and C-peptide and
insulin sensitivity indexes. Physical capacity, lipid profile, and GLUT-4 protein expression and translocation were also assessed.
Results: Twenty-five subjects were analyzed (CR: n= 13; PL: n=12).Hb
A1c
was significantly reduced in the creatine group when
compared with the placebo group (CR: PRE = 7.4 T0.7, POST = 6.4 T0.4; PL: PRE = 7.5 T0.6, POST = 7.6 T0.7; P= 0.004;
difference = j1.1%, 95% confidence interval = j1.9% to j0.4%). The delta area under the curve of glucose concentration was
significantly lower in the CR group than in the PL group (CR = j7790 T4600, PL = 2008 T7614; P= 0.05). The CR group also
presented decreased glycemia at times 0, 30, and 60 min during a meal tolerance test and increased GLUT-4 translocation. Insulin
and C-peptide concentrations, surrogates of insulin sensitivity, physical capacity, lipid profile, and adverse effects were compa-
rable between the groups. Conclusions: Creatine supplementation combined with an exercise program improves glycemic control
in type 2 diabetic patients. The underlying mechanism seems to be related to an increase in GLUT-4 recruitment to the sarcolemma.
Key Words: CREATINE SUPPLEMENTATION, EXERCISE TRAINING, TYPE 2 DIABETES, THERAPEUTIC EFFECTS
For decades, physical activity has been considered the
major cornerstone of type 2 diabetes management,
along with diet and medication (24). Indeed, strate-
gies capable of mimicking and/or enhancing the effects of
exercise are potentially therapeutic. In this context, creatine
supplementation has emerged as a novel putative candidate
for treating diabetes (8,16).
Creatine, a natural amine in the human body, is partly
synthesized by kidneys, pancreas, and liver (approximately
1–2 gId
j1
), as well as ingested from food (approximately
1–5 gId
j1
), especially meat and fish, and thereafter mainly
transported to the skeletal muscles, brain, and testes. Cre-
atine has rapidly become one of the most consumed nu-
tritional supplements worldwide owing to its efficacy to
increase muscle phosphorylcreatine content, thereby en-
hancing athletic performance and, consequently, lean mass.
Although the literature supports the role of creatine supple-
mentation in improving acute work capacity during inter-
mittent short-duration high-intensity exercise, the effect of
this supplement on skeletal muscle protein synthesis is less
clear (3,17,27).
A growing body of evidence has now revealed a number
of therapeutic potential applications of this supplement in
a broad range of diseases, notably muscle disorders, neu-
rodegenerative conditions, and metabolic dysfunctions (7).
Interestingly, some studies have also suggested that creatine
supplementation may improve glucose metabolism, partic-
ularly when combined with exercise training (16).
Address for correspondence: Bruno Gualano, Ph.D., Av Mello de Moraes,
65–Butanta
˜, 05508-030, Sa
˜o Paulo, SP, Brazil; E-mail: gualano@usp.br.
Submitted for publication July 2010.
Accepted for publication September 2010.
Supplemental digital content is available for this article. Direct URL citations
appear in the printed text and are provided in the HTML and PDF versions of
this article on the journal’s Web site (www.acsm-msse.org).
0195-9131/11/4305-0770/0
MEDICINE & SCIENCE IN SPORTS & EXERCISE
Ò
Copyright Ó2011 by the American College of Sports Medicine
DOI: 10.1249/MSS.0b013e3181fcee7d
770
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
It has been consistently demonstrated that creatine sup-
plementation along with muscle contraction can augment
muscle glycogen accumulation in humans (6,20,26). In ad-
dition, it has been reported (16) that creatine supplementa-
tion offsets the decline in muscle GLUT-4 expression after
2 wk of immobilization and increases GLUT-4 content
during subsequent rehabilitation training in healthy males.
Furthermore, creatine intake has been suggested (5) to
ameliorate hyperglycemia, typical of Huntington transgenic
mice, delaying the onset of diabetes. Supporting these
findings, a study (15) verified that creatine ingestion can
reduce the insulinogenic index in an animal model of in-
herited type 2 diabetes. Accordingly, we demonstrated that
creatine supplementation combined with aerobic training
promoted greater improvement on glucose tolerance than
aerobic training alone in physically inactive males (8). The
observations from the aforementioned studies suggest that
this nutritional intervention merits randomized controlled
trials to fully appreciate the possible therapeutic role of this
supplement in diabetes.
Thus, the aim of this study was to investigate the effects
of creatine supplementation combined with exercise train-
ing on glycemic control in type 2 diabetic patients.
METHODS
Subjects. Men and women (945 yr) prediagnosed with
type 2 diabetes, physically inactive for at least 1 yr, and
with BMI Q30 kgIm
j2
were eligible. The exclusion criteria
included use of exogenous insulin, uncontrolled hyperten-
sion (Q140/90 mm Hg), cardiovascular diseases, and/or
muscle skeletal disturbances that precluded exercise partici-
pation, vegetarian diet, previous use of creatine supplements,
glomerular filtration rate G40 mLIkg
j1
Imin
j1
, glycosylated
hemoglobin (Hb
A1c
)99%, and dyslipidemia. Patients’ charac-
teristics are presented in Table 1.
The study was approved by the local ethical committee,
and all subjects signed the written informed consent. This
trial was registered at ClinicalTrials.gov as NCT00992043.
Experimental protocol. A 12-wk, double-blind, ran-
domized, parallel-group, placebo-controlled trial was con-
ducted between October 2009 and January 2010 in Sao
Paulo (Brazil), according to the guidelines of the CONSORT
Statement.
Thepatientswererandomlyassigned (1:1) to receive
either creatine (CR) or placebo (PL) in a double-blind fash-
ion. We assigned patients to treatment sequence by using a
computer-generated randomization code with a block of eight
and stratified by gender. All the patients undertook a pro-
gram of moderate intensity aerobic training combined with
strengthening exercises for 3 months. The patients were
assessed at baseline (PRE) and after 12 wk (POST). The pri-
mary outcome was the glycemic control, as assessed through
Hb
A1c
concentrations. Secondary outcomes included the area
under the curve (AUC) of glucose, insulin, and C-peptide,
obtained from Meal Tolerance Tests (MTT), and insulin
sensitivity indexes. Muscle function and strength, aerobic
conditioning, body composition, and lipid profile were also
measured. Adverse events were recorded throughout the
trial. Possible differences in dietary intake were assessed by
means of three 24-h dietary recalls. In a subgroup of patients
randomly selected (n= 6 per group), muscle phosphoryl-
creatine content was measured through phosphorus magnetic
resonance spectroscopy (
31
P MRS), and muscle biopsies
were performed to assess GLUT-4 protein expression and
translocation.
Creatine supplementation protocol and blinding
procedure. The CR group received 5 gId
j1
of creatine
monohydrate throughout the trial. The PL group was given
the same dose of dextrose. The individuals consumed the
supplement as a single dose during their lunch. The sup-
plement packages were coded so that neither the inves-
tigators nor the participants were aware of the contents
until completion of the analyses. The compliance with the
supplementation was monitored weekly by asking the
patients personally. To verify the purity of the creatine used,
a sample was analyzed by high-performance liquid chro-
matography, and purity was established as 99.9%. The
supplementation was interrupted 72 h before the posttest
evaluation.
Exercise training program
The exercise program consisted of 12 wk of supervised
training. Exercise sessions occurred three times a week.
Training sessions consisted of a 5-min treadmill warm-up
followed by 25 min of resistance training, 30 min of treadmill
aerobic training, and 5 min of stretching exercises. All ses-
sions were monitored by at least one fitness professional. The
exercise program was performed in an intrahospital gymna-
sium (Laboratory of Assessment and Conditioning in Rheu-
matology, School of Medicine, University of Sao Paulo).
Resistance training included five exercises for the main
muscle groups: bench press, leg press, lat pulldown, leg ex-
tension, and seated row. Patients were required to perform four
sets of 8–12 repetitions maximum (RM), except during the first
TABLE 1. Patients’ characteristics.
Creatine
(n= 13)
Placebo
(n= 12)
P
(CR vs PL)
Gender (F/M) 8/5 8/4 0.56
Disease duration (years since diagnosis),
mean TSD
7T37T3 0.91
Age (yr), mean TSD 57.5 T5 56.4 T8.23 0.68
Systolic blood pressure
(mm Hg), mean TSD
125.0 T5.0 125.0 T5.0 0.92
Diastolic blood pressure
(mm Hg), mean TSD
85 T2.0 85 T1.0 0.88
Drugs, n(%)
Metformin 13 (100) 12 (100) 0.61
Sulfonylurea 7 (53.8) 6 (50) 0.58
A-blocker 2 (15.4) 2 (16.7) 0.67
ACE inhibitor 3 (23.1) 3 (25.0) 0.63
Angiotensin receptor antagonist 13 (100) 12 (100) 0.61
Thiazide 4 (30.8) 4 (33.3) 0.61
Statin 11 (84.6) 10 (83.3) 0.50
Fibrate 2 (15.4) 2 (16.7) 0.67
No significant differences were found (nonpaired t-test or Fisher exact test).
CREATINE AND DIABETES Medicine & Science in Sports & Exercise
d
771
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
week, when a reduced volume of two sets of 15–20 RM for
each exercise was performed (as an adaptation period to re-
sistance training). Overload progression was implemented
when the subject could perform Q12 repetitions on the last
training set for two consecutive workouts. Aerobic training
intensity was set at the corresponding heart rate of approxi-
mately 70% of the V
˙O
2peak
.
Food intake assessment. Food intake was assessed
by means of three 24-h dietary recalls undertaken on sepa-
rate days (two weekdays and one weekend day) using a vi-
sual aid photo album of real foods. The 24-h dietary recall
consists of listing the foods and the beverages consumed
during 24 h before the recall. Energy and macronutrient in-
takes were analyzed by the Brazilian software Virtual Nutri
Ò
.
Glycosylated hemoglobin (Hb
A1c
). Hb
A1c
was mea-
sured using the BioRad Variant II automated analyzer
(BioRad, Irvine, CA).
MTT. The subjects were requested to refrain from inten-
sive physical activity 72 h before the MTT. After an over-
night fasting, the patients were given a 4-h meal challenge.
The mixed meal (500 kcal, 60% CHO, 20% fat, and 20%
protein) contained approximately 72 g of CHO. Blood
samples were collected at 0, 30, 60, 120, 180, and 240 min
for plasma glucose, insulin, and C-peptide for plasma glu-
cose, insulin, and C-peptide measurements. The former was
assessed through a colorimetric enzymatic assay (Bioclin,
Brazil), and the two latter were assessed using human-
specific radioimmunoassay techniques (Diagnostic Products
Corporation, CA, EUA). Fasting plasma insulin and glucose
concentrations were used to perform the G/I and the Homeo-
stasis Model Assessment (HOMAIR and HOMAB) indexes.
Strength and functional muscle assessments.
The patients underwent three familiarization sessions, sepa-
rated for at least 72 h, for all strength and functional tests.
Before the 1-RM test, two light warm-up sets interspaced
for 2 min were performed. Then, the patients had up to five
attempts to achieve the 1-RM load (e.g., maximum weight
that could be lifted once with proper technique), with a
3-min interval between attempts. One-RM tests were con-
ducted for bench press and leg press. Moreover, upper- and
lower-limb isometric strength was determined with a hand-
grip (dominant arm) and lower-back extension dynamometer,
respectively. Finally, we assessed balance, mobility, and
muscle function (timed-up-and-go and timed-stands tests),
according to previous descriptions (13). Training volume
(defined as total amount of weight lifted in each training
session) was calculated by multiplying the total number of
sets and repetitions by the amount of weight lifted in each set.
Body composition. Body composition and bone min-
eral density (BMD) were measured by dual x-ray absorp-
tiometry (DXA), using Hologic densitometry equipment
(Discovery model; Hologic, Inc., Bedford, MA) at the fol-
lowing regions: lumbar spine, femoral neck, total femur,
and whole body. All measurements were carried out by
the same trained technologist. Precision error for BMD
measurements was determined according to standard Inter-
national Society for Clinical Densitometry protocols. Least
significant changes with 95% confidence were 0.033 gIcm
j2
at the lumbar spine, 0.047 gIcm
j2
at the femoral neck,
0.039 gIcm
j2
at the total femur, and 0.020 gIcm
j2
at the
whole body.
Maximal oxygen consumption (V
˙O
2max
) tests.
All the subjects underwent a treadmill cardiopulmonary
test, before and after the intervention, according to the
conventional Bruce protocol. Attainment of V
˙O
2max
was
accepted when two of three criteria were met: a plateau
in V
˙O
2
, a respiratory exchange ratio 91.1, and/or volitional
exhaustion. The ventilatory anaerobic threshold (VAT) was
determined to occur at the break point between the increase
of carbon dioxide output (V
˙CO
2
) and V
˙O
2
. The respiratory
compensation point was determined to occur where the
ventilatory equivalent for carbon dioxide (V
˙
E
/V
˙CO
2
ratio)
was the lowest before a systematic increase.
Lipid profile. Serum concentrations of blood cholesterol,
HDL-cholesterol, and triglycerides were assessed by means
of colorimetric enzymatic methods (CELM, Brazil). From
these, VLDL-cholesterol (VLDL-cholesterol = triglycerides/5)
and LDL-cholesterol (LDL-cholesterol = total cholesterol j
[HDL-cholesterol + VLDL-cholesterol]) concentrations were
calculated. Serum apolipoproteins A1 and B and lipoprotein(a)
concentrations were determined by using immunoturbidimetric
assays (Roche Diagnostics, Germany). Serum apolipoproteins
A2 and E were measured through nephelometric assays
(Behring Diagnostics, Germany).
Muscle phosphorylcreatine content. In a subsam-
ple of patients (n= 6 per group), muscle phosphorylcre-
atine content was assessed in vivo by
31
P MRS using a
whole-body 3.0-T magnetic resonance imaging scanner
(Achieva Intera, Philips, Best, the Netherlands) and a 14-cm-
diameter
31
P surface coil. In brief, the surface coil was placed
centered under the calf muscle of the left leg. The scanner body
coil was used to obtain conventional anatomical T1-weighted
magnetic resonance images in the three orthogonal planes.
31
P MRS was acquired using the image selected in vivo spec-
troscopy sequence with an echo time and repetition time of
0.62 and 4500 ms, respectively. Spectrum bandwidth was
3000 Hz with 2048 data points and 64 repetitions. Spectrum
raw data were analyzed with Java Magnetic Resonance User
Interface software, and processing steps included apodization
to 5 Hz, Fourier transform, and phase correction. For spectrum
quantification, the AMARES algorithm was used taking into
account the previous knowledge of inorganic phosphate, pho-
sphodiester and phosphorylcreatine singlets, >-ATP and F-
ATP doublets, and A-ATP triplets. The phosphorylcreatine
signal was quantified relative to the A-ATP signal, assuming a
constant A-ATP concentration of 5.5 mmolIkg
j1
.
Muscle biopsies. In a subsample of patients (n= 6 per
group), muscle samples were obtained from the midportion
of the vastus lateralis using the percutaneous needle biopsy
technique with suction. Thereafter, an aliquot of each mus-
cle sample was immediately freed from blood and visible
connective tissue, rapidly frozen in liquid nitrogen, and
http://www.acsm-msse.org772 Official Journal of the American College of Sports Medicine
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
stored at j80-C for subsequent analysis. The postintervention
biopsies were done through an adjacent incision to the baseline
site, 72 h after the last training session. All biopsies were car-
ried out after an 8-h overnight fast, and the last meal was a
standard dinner. Muscle biopsies were also obtained from
nondiabetic volunteers (n=6;age=60T5 yr, body mass
index = 31.6 T1.3 kgIm
j2
,Hb
A1c
=5.1%T0.3%, V
˙O
2max
=
25 T3mLIkg
j1
Imin
j1
) who were matched for age, gender,
body mass index, and V
˙O
2max
to the diabetic patients.
Cellular fractionation. Muscle samples were minced
and homogenized in ice-cold lysis buffer (2 mM EDTA,
10 mM EGTA, 0.25 M sucrose, 1:300 Sigma protease inhib-
itor cocktail, and 20 mM Tris–HCl at pH 7.5). The homoge-
nate was centrifuged at 100,000gfor 30 min (4-C). The
resulting pellet was dissolved in 1% Triton X-100 lysis buffer
and centrifuged again at 100,000gfor 30 min (4-C) to obtain
the nuclear pellet and the membrane fraction (supernatant).
Western blot analysis. GLUT-4 expression and translo-
cation were measured through Western blot analysis. Briefly,
samples were subjected to SDS-PAGE in polyacrylamide gel
(10%). After electrophoresis, proteins were electrotransferred
to the nitrocellulose membrane (BioRad Biosciences,
Piscataway, NJ). Equal loading of samples and transfer effi-
ciency were monitored with the use of 0.5% Ponceau S
staining of the blot membrane. The blotted membrane was
then blocked (5% nonfat dry milk, 10 mM Tris-HCl (pH 7.6),
150 mM NaCl, and 0.1% Tween 20) for 2 h at room temper-
ature and then incubated overnight at 4-C with specific anti-
bodies against GLUT-4 (Millipore, Bedford, MA). Binding of
the primary antibody was detected with the use of peroxidase-
conjugated secondary goat antirabbit antibody for 2 h at room
temperature and developed using enhanced chemilumines-
cence (Amersham Biosciences, Piscataway, NJ) detected by
autoradiography. Quantification analysis of blots was per-
formed with the use of Scion Image software (Scion based
on National Institutes of Health image). Total and membrane
fractions were normalized against glyceraldehyde-3-phosphate
dehydrogenase (GAPDH) and G
>o
.
Sample size. Before the commencement of the trial, we
determined that 24 patients would be needed to provide 80%
power (5% significance) to detect a difference of 0.6% be-
tween groups (similar effect expected with exercise training
alone (25)) in Hb
A1c
concentrations, which is the primary
outcome of this clinical trial. To account for midtrial with-
drawals, we enlarged our study population by approximately
15% to 28 participants.
Statistical analysis. Each comparison was by intention
to treat, irrespective of compliance with supplement intake.
Data were tested by a mixed model with repeated measures
using the software SAS version 9.1. A post hoc test adjusted
by Tukey was used for multicomparison purposes. Non-
repeated measures were compared using Student’s t-test
or Fisher exact test. Significance level was previously set at
PG0.05. Data are presented as mean, SD, estimated dif-
ference of means after intervention, and 95% confidence
interval (CI), except when otherwise stated.
RESULTS
Patients. The number of subjects recruited to the study
is shown in Supplemental Digital Content 1 (see Figure;
Fluxogram of participants; http://links.lww.com/MSS/A55).
Of the 121 people who responded to the initial request for
volunteers, 56 were screened and 28 met the inclusion cri-
teria. These patients were randomly assigned to either the
CR (n= 14) or PL (n= 14) groups. Three patients were
subsequently lost: two withdrew for personal reasons (one
from each group) and one was excluded because of an is-
chemic stroke episode in the first week of intervention (PL).
Then, 25 patients were analyzed (CR = 13, PL = 12).
Assessment of blinding, adherence to the exer-
cise program, and food intake. Five (38.4%) of the
patients correctly identified the supplement in the CR group,
whereas six (46.1%) patients were able to identify the cor-
rect supplement in the PL group (P= 0.57, Fisher exact test).
The adherence to the exercise program was 64.4% T
19.9% and 73.3% T19.8% for CR and PL groups, respec-
tively (P= 0.55, Student’s t-test).
Food intake did not significantly differ within or be-
tween groups (see Table Supplemental Digital Content 2;
Food intake at baseline and after the intervention;
http://links.lww.com/MSS/A56).
Muscle phosphorylcreatine content. There was
no significant difference between groups at baseline
(P= 0.77). After the intervention, the CR group presented
higher muscle phosphorylcreatine content when com-
pared with the PL group (CR: PRE = 44 T10 mmolIkg
j1
wet weight, POST = 70 T18; PL: PRE = 52 T13
POST = 46 T13 mmolIkg
j1
wet weight; P= 0.03; esti-
mated difference of means = 23.6 mmolIkg
j1
wet weight;
95% CI = 1.42–45.8 mmolIkg
j1
wet weight).
Primary outcome measure. Figure 1 demonstrates
Hb
A1c
concentrations. No significant difference between
groups was observed at baseline (P= 0.92). After the inter-
vention, Hb
A1c
concentrations were significantly reduced
only in the CR group compared with baseline (P=0.0001).
FIGURE 1—Effects of creatine supplementation combined with exer-
cise training in type 2 diabetic patients on Hb
A1c
concentrations. *Inter-
action effect (P= 0.004; estimated difference of means = j1.1%, 95%
CI = j1.9% to j0.4%). Data are means TSD. Mixed model for repeated
measures was used to compare placebo (n= 12) versus creatine (n=13).
CREATINE AND DIABETES Medicine & Science in Sports & Exercise
d
773
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
A significant difference between groups was observed
(P= 0.004; estimated difference of means = j1.1%, 95%
CI = j1.9% to j0.4%).
Secondary outcome measures. Figure 2 shows the
effects of creatine supplementation on serum glucose, insu-
lin, and C-peptide concentrations.
The CR group presented a significant decrease in glycemia
at 0 (P=0.001,difference=j48 mgIdL
j1
,95%CI=j75 to
j21 mgIdL
j1
), 30 (P=0.004,difference=j48 mgIdL
j1
,
95% CI = j103 to j15 mgIdL
j1
), and 60 min (P=0.003,
difference = j68 mgIdL
j1
,95%CI=j117 to j19 mgIdL
j1
)
compared with the PL group. The remaining points were
unchanged. No significant differences were observed for
insulin and C-peptide concentrations during MTT.
The delta AUC for glucose concentration was significantly
lower in the CR group compared with the PL group (Fig. 3).
No significant differences were observed in delta AUC
for insulin and C-peptide concentrations (data not shown).
FIGURE 2—Effects of creatine supplementation combined with exercise training in type 2 diabetic patients on glucose (A and B), insulin (C and D),
and C-peptide (E and F) concentrations. Left panels represent creatine data, whereas right panels represent placebo data. *Interaction effect (group
time) at time 0 (P= 0.001; difference = j48 mgIdL
j1
, 95% CI = j75 to j21 mgIdL
j1
), 30 (P= 0.004; difference = j48 mgIdL
j1
, 95% CI = j103 to
j15 mgIdL
j1
), and 60 (P= 0.003; difference = j68 mgIdL
j1
, 95% CI = j117 to j19 mgIdL
j1
). Data are means TSD. Mixed model for repeated
measures was used to compare placebo (n= 12) versus creatine (n= 13).
http://www.acsm-msse.org774 Official Journal of the American College of Sports Medicine
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
There were no significant differences in HOMAIR, HOMAB,
and G/I indexes (see Table; Supplemental Digital Content
3; Effects of creatine supplementation combined with
exercise training on the surrogates of insulin sensitivity;
http://links.lww.com/MSS/A57).
Physical capacity, lipid profile, and body com-
position. Table 2 shows the effects of creatine supple-
mentation on physical capacity. Both groups presented
increased strength (main time effects) in the 1-RM leg press
and 1-RM bench press, and low back strength. Handgrip
strength tended to increase in both groups but did not reach
statistical significance (P= 0.06). Muscle function, as
assessed through the timed-stands test, was also significantly
improved in both groups, whereas the performance in the
timed-up-and-go remained unchanged.
V
˙O
2
correspondent to the ventilatory anaerobic threshold
(V
˙O
2
–VAT) and to the respiratory compensation point
(V
˙O
2
–RCP) were significantly increased in both groups
(mean time effects). V
˙O
2max
did not significantly change.
No significant differences were observed between the
groups for any physical capacity variable.
Total training volume did not significantly differ be-
tween the groups (CR = 3,128,294 T1,175,009 kg, PL =
2,171,652 T1,396,930 kg, P=0.11).
No significant differences were observed either in blood
lipoproteins or in blood apolipoproteins (see Table; Sup-
plemental Digital Content 4; Effects of creatine supple-
mentation combined with exercise training on lipid profile;
http://links.lww.com/MSS/A58). Also, there were no signif-
icant differences for body composition variables (see Table;
Supplemental Digital Content 5; Effects of creatine supple-
mentation combined with exercise training on body composi-
tion; http://links.lww.com/MSS/A59).
GLUT-4 expression and translocation. Figure 4
expresses muscle GLUT-4 content and translocation data.
There was no significant difference in muscle GLUT-4
content between the diabetic patients and the healthy
individuals. In addition, Muscle GLUT-4 content was sig-
nificantly unchanged after the intervention (P= 0.91).
At baseline, membrane GLUT-4 content was significantly
lower in the diabetic patients compared with the healthy sub-
jects. After the intervention, membrane GLUT-4 content was
significantly raised in the CR and PL groups (P= 0.0008) so
that differences between the diabetic patients and healthy
subjects were no longer observed (P= 0.92). Following the
same trend, the membrane–total GLUT-4 content ratio was
increased in both experimental groups (P= 0.0001). However,
the CR group presented a greater increase than the PL in
FIGURE 3—Effects of creatine supplementation combined with exercise training in type 2 diabetic patients on the area under the curve of glucose
concentration. A, Individual data for AUC. B, Means TSD for the delta area under the curve of glucose concentration. *Significant treatment effect
compared with placebo (P= 0.05). Student’s t-test was used to compare placebo (n= 12) versus creatine (n= 13).
TABLE 2. Effects of creatine supplementation combined with exercise training on physical capacity in type 2 diabetic patients.
Creatine (n= 13) Placebo (n= 12)
Difference (CI 95%) P(CR vs PL)
Variable Pre Post Pre Post
1-RM bench press (kg)
a
56 T24 64 T27 53 T12 67 T93(j19 to 25) 0.52
1-RM leg press (kg)
b
54 T17 68 T26 60 T11 73 T13 j2(j25 to 21) 0.99
Handgrip (kg)
c
34 T10 36 T11 36 T937T10 5 (j3 to 13) 0.82
Low-back strength (kg)
d
95 T40 108 T26 102 T24 112 T23 10 (j21 to 43) 0.49
Timed-stands test (rep)
e
14 T218T215T217T20(j2 to 2) 0.48
Timed-up-and-go test (s) 6.8 T0.8 6.6 T0.6 6.4 T1.0 5.6 T0.8 0.8 (0.1 to 1.6) 0.21
V
˙O
2
–VAT (mLIkg
j1
Imin
j1
)
f
16 T317T217T319T3j1(j4 to 1) 0.68
V
˙O
2
–RCP (mLIkg
j1
Imin
j1
)
g
19 T322T426T630T6j8(j16 to 0) 0.69
V
˙O
2max
(mLIkg
j1
Imin
j1
)24T425T530T531T7j5(j13 to 3) 0.87
Data expressed as mean TSD, estimated mean of differences (95% CI) and level of significance (P) between CR and PL (mixed model for repeated measures).
Symbols represent main time effects (
a
P= 0.007,
b
P= 0.0018,
c
P= 0.06,
d
P= 0.03,
e
P= 0.0001,
f
P= 0.0004,
g
P= 0.08). No significant differences were found between groups at
baseline.
RM, repetition maximum; rep, repetitions; V
˙O
2
–VAT, oxygen consumption correspondent to ventilator anaerobic threshold; V
˙O
2
–RCP, oxygen consumption correspondent to respiratory
compensation point; V
˙O
2
, maximal oxygen consumption.
CREATINE AND DIABETES Medicine & Science in Sports & Exercise
d
775
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
membrane GLUT-4 content as well as in the membrane–total
GLUT-4 content ratio (P=0.05andP= 0.03, respectively).
Adverse events. No severe adverse events in the CR
group were observed. Symptoms such as nausea, diar-
rhea, and cramps were reported by some patients. How-
ever, these events did not significantly differ between
the groups (see Table; Supplemental Digital Content 6;
Adverse events in the creatine and the placebo groups;
http://links.lww.com/MSS/A60).
DISCUSSION
This is the first randomized controlled trial to describe the
beneficial effects of creatine supplementation on glycemic
control in type 2 diabetic patients who underwent exercise
training. As such, this supplement emerges as a valuable
nonpharmacological approach for treating diabetes.
There is evidence showing that creatine supplementation
improves insulin sensitivity in a rodent model of inherited
type 2 diabetes (15) and delays the onset of diabetes in
Huntington transgenic mice (5). In healthy young individu-
als, creatine supplementation has been proven to increase
muscle glycogen accretion and glucose tolerance, particularly
when combined with exercise training. Altogether, these
findings provided the rationale for investigating the thera-
peutic role of creatine supplementation in diabetic patients.
As a consequence of creatine supplementation, we ob-
served a significant increase in muscle phosphorylcreatine
content (n= 6) and subsequent improvement in glycemic
control. Importantly, the mean reduction in Hb
A1c
(j1.1%)
was superior to that commonly seen with exercise training
(25) or metformin (10) treatment alone, pointing out the
therapeutic potential of this novel nutritional intervention.
The glycemia-lowering effect of this supplement is most
likely an explanation for this metabolic response. On the
other hand, no change in insulinemia was verified, which is
apparently in contrast to observations from in vitro studies,
which indicated creatine-induced insulin secretion in incu-
bated mouse islets (1,12). Furthermore, it has been previ-
ously observed (21) that creatine supplementation induces
hyperinsulinemia in rats, which could ultimately lead to
disruption in glucose homeostasis. However, studies in
humans have recurrently refuted such creatine-induced
hyperinsulinemia (8,14,26). The current findings extrapolate
this idea to diabetic patients.
Regarding mechanisms, we showed suboptimal GLUT-4
translocation in the diabetic patients compared with their
healthy peers, corroborating early evidence that insulin re-
sistance in type 2 diabetes is not generally associated with
decreased muscle GLUT-4 content (9). In opposite, it is
speculated that insulin stimulation fails to induce normal
GLUT-4 protein translocation in skeletal muscle from these
patients (22,28). In this respect, it is worth noting that ex-
ercise training was able to resolve impaired GLUT-4 trans-
location in the diabetic patients. Interestingly, this response
was further enhanced by creatine supplementation, sug-
gesting that this supplement acts directly on type 2 diabetes
pathogenesis (i.e., suboptimal GLUT-4 translocation),
thereby ameliorating hyperglycemia and consequently gly-
cemic control.
On the basis of the current knowledge, it is difficult to
reveal the molecular basis for the increase in muscle GLUT-
4 translocation that occurred as a consequence of creatine
FIGURE 4—Effects of creatine supplementation combined with exer-
cise training in type 2 diabetic patients on muscle GLUT-4 content (B),
membrane GLUT-4 content (C), and membrane–total GLUT-4 ratio
(D). Representative immunoblots are shown in panel A, Dotted lines
represent mean data from healthy subjects (n= 6). #Mean time effects
for membrane GLUT-4 content (P= 0.0008) and membrane–total
GLUT-4 ratio (P= 0.0001). *Interaction effects for membrane GLUT-4
content (P= 0.05) and membrane–total GLUT-4 ratio (P= 0.03). Data
are means TSD and are expressed relative to the baseline value that
was set to be equal to 1. Mixed model for repeated measures was used to
compare placebo (n= 12) versus creatine (n= 13).
http://www.acsm-msse.org776 Official Journal of the American College of Sports Medicine
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
supplementation. In this regard, previous observation from
nondiabetic models indicates that creatine supplementation
may upregulate (i) protein kinase B mRNA expression,
which is suggested to promote GLUT-4 translocation to the
sarcolemma (23); (ii) insulin-like growth factor-1 mRNA
and protein content, which is believed to enhance insulin
action (4); and (iii) nuclear content and DNA binding ac-
tivity of myocyte enhancer factor-2 isoforms, which are
transcription factors that regulate GLUT-4 expression in
muscle (11). The influence of creatine supplementation on
the insulin-signaling pathway and transcription factors of
GLUT-4 in type 2 diabetes is currently under investigation
in our laboratory.
It is also interesting to note that only the CR group ex-
perienced improvements in glycemic control, although both
groups had undertaken an exercise training program. The
American Diabetic Association has recommended that ex-
ercise training should be performed at least 3 dIwk
j1
and
with no more than two consecutive days without physical
activity to improve glycemic control (24). Although our
exercise program was previously designed to achieve this
recommendation, the patients failed to properly accomplish
it (see the adherence data). Moreover, the short duration of
the training protocol can be also responsible for the lack of
improvements in glycemic control. Thus, it is possible to
assume that our exercise training program alone did not
reach sufficient frequency and/or volume levels to promote
improvements in glycemic control, although significant
changes in physical capacity (i.e., muscle function, aerobic
condition, and strength) were observed in both groups. In
this scenario, it is tempting to speculate that the addition of
creatine supplementation might have maximized the effects
of exercise on insulin sensitivity and glycemic control.
Nonetheless, it is impossible to distinguish whether the
current findings result from the creatine treatment per se or
the interaction between creatine and exercise training. On
one hand, some authors have speculated that dietary crea-
tine-associated changes in CHO metabolism are a result of
an interaction between creatine supplementation and other
mediators of muscle glucose transport, such as muscle con-
traction. On the other hand, recent evidence suggests that
creatine per se can modulate the expression of key proteins
and genes related to insulin sensitivity and glycemic control
(e.g., GLUT-4, protein kinase B, myocyte enhancer factor-2,
insulin-like growth factor-1) (23). Indeed, further inves-
tigations are needed to address this question.
Despite some evidence indicating that creatine supple-
mentation may be capable of improving lipid profile, lean
mass, and strength (2,7), we did not observe such adapta-
tions. In fact, it was expected from the outset that a lower
adaptive response for lipid profile and BMD would be
shown by our patients because their baseline values were
within the reference range. Compelling evidence has indi-
cated no direct benefit of creatine supplementation on mus-
cle protein synthesis (17), suggesting that major mechanism
by which chronic creatine supplementation exerts its effects
on body composition and athletic performance is by en-
hancing training volume at every training session. Because
we did not observe higher training volume, the lack of
changes in strength and lean mass was also expected.
Moreover, creatine supplementation seems to be highly
safe based on data from short-term and long-term human
studies and results from several (18,19) therapeutic trials. In
this trial, the adverse effects were comparable between
groups, further supporting this notion and extending it to
diabetic patients.
This study presented some limitation. First, considering
the sample’s characteristics, these findings cannot be ex-
trapolated to patients with poorer glycemic control, older
age, or at a different pharmacological treatment (e.g., ex-
ogenous insulin therapy). Second, the patients underwent
exercise training along with creatine intervention, thus we
cannot also generalize these results to physically inactive
individuals. Third, it currently is unknown how long the
effects of creatine may persist. Studies evaluating the effi-
cacy and safety of long-term creatine supplementation are
needed. Likewise, the optimal creatine protocol (i.e., high vs
low dose, short vs long term, continuous vs cycled regimen)
remains to be determined.
In summary, we reported a novel therapeutic role of cre-
atine supplementation on metabolic control in type 2 dia-
betic patients. Moreover, we provided convincing evidence
that creatine supplementation might modulate glucose up-
take in these patients mainly via an increase in GLUT-4
recruitment to the sarcolemma.
Bruno Gualano, Guilherme G. Artioli, and Eloisa Bonfa receive
support from Conselho Nacional de Desenvolvimento Cientı´ fico e
Tecnolo´ gico (CNPq).
The authors declare that they do not have conflict of interests.
The authors inform that the results of the present study do
not constitute endorsement by the American College of Sports
Medicine.
REFERENCES
1. Alsever RN, Georg RH, Sussman KE. Stimulation of insulin se-
cretion by guanidinoacetic acid and other guanidine derivatives.
Endocrinology. 1970;86(2):332–6.
2. Andres RH, Ducray AD, Schlattner U, Wallimann T, Widmer HR.
Functions and effects of creatine in the central nervous system.
Brain Res Bull. 2008;76(4):329–43.
3. Branch JD. Effect of creatine supplementation on body composi-
tion and performance: a meta-analysis. Int J Sport Nutr Exerc
Metab. 2003;13(2):198–226.
4. Deldicque L, Louis M, Theisen D, et al. Increased IGF mRNA in
human skeletal muscle after creatine supplementation. Med Sci
Sports Exerc. 2005;37(5):731–6.
CREATINE AND DIABETES Medicine & Science in Sports & Exercise
d
777
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
5. Ferrante RJ, Andreassen OA, Jenkins BG, et al. Neuroprotective
effects of creatine in a transgenic mouse model of Huntington’s
disease. J Neurosci. 2000;20(12):4389–97.
6. Green AL, Hultman E, Macdonald IA, Sewell DA, Greenhaff PL.
Carbohydrate ingestion augments skeletal muscle creatine accu-
mulation during creatine supplementation in humans. Am J Phys-
iol. 1996;271(5 Pt 1):E821–6.
7. Gualano B, Artioli GG, Poortmans JR, Lancha AH Jr. Exploring
the therapeutic role of creatine supplementation. Amino Acids.
38(1):31–44.
8. Gualano B, Novaes RB, Artioli GG, et al. Effects of creatine
supplementation on glucose tolerance and insulin sensitivity in
sedentary healthy males undergoing aerobic training. Amino Acids.
2008;34(2):245–50.
9. Henriksen EJ. Invited review: effects of acute exercise and exercise
training on insulin resistance. J Appl Physiol. 2002;93(2):788–96.
10. Johansen K. Efficacy of metformin in the treatment of NIDDM.
Meta-analysis. Diabetes Care. 1999;22(1):33–7.
11. Ju JS, Smith JL, Oppelt PJ, Fisher JS. Creatine feeding increases
GLUT4 expression in rat skeletal muscle. Am J Physiol Endocrinol
Metab. 2005;288(2):E347–52.
12. Marco J, Calle C, Hedo JA, Villanueva ML. Glucagon-releasing
activity of guanidine compounds in mouse pancreatic islets. FEBS
Lett. 1976;64(1):52–4.
13. Newcomer KL, Krug HE, Mahowald ML. Validity and reliability
of the timed-stands test for patients with rheumatoid arthritis and
other chronic diseases. J Rheumatol. 1993;20(1):21–7.
14. Newman JE, Hargreaves M, Garnham A, Snow RJ. Effect of cre-
atine ingestion on glucose tolerance and insulin sensitivity in men.
Med Sci Sports Exerc. 2003;35(1):69–74.
15. Op’t Eijnde B, Jijakli H, Hespel P, Malaisse WJ. Creatine sup-
plementation increases soleus muscle creatine content and lowers
the insulinogenic index in an animal model of inherited type 2
diabetes. Int J Mol Med. 2006;17(6):1077–84.
16. Op’t Eijnde B, Urso B, Richter EA, Greenhaff PL, Hespel P. Effect
of oral creatine supplementation on human muscle GLUT4 protein
content after immobilization. Diabetes. 2001;50(1):18–23.
17. Paddon-Jones D, Borsheim E, Wolfe RR. Potential ergogenic
effects of arginine and creatine supplementation. J Nutr. 2004;
134(10 suppl):2888–94S; discussion 95S.
18. Persky AM, Rawson ES. Safety of creatine supplementation.
Subcell Biochem. 2007;46:275–89.
19. Poortmans JR, Francaux M. Adverse effects of creatine supple-
mentation: fact or fiction? Sports Med. 2000;30(3):155–70.
20. Robinson TM, Sewell DA, Casey A, Steenge G, Greenhaff PL.
Dietary creatine supplementation does not affect some haemato-
logical indices, or indices of muscle damage and hepatic and renal
function. Br J Sports Med. 2000;34(4):284–8.
21. Rooney K, Bryson J, Phuyal J, Denyer G, Caterson I, Thompson
C. Creatine supplementation alters insulin secretion and glucose
homeostasis in vivo.Metabolism. 2002;51(4):518–22.
22. Ryder JW, Yang J, Galuska D, et al. Use of a novel impermeable
biotinylated photolabeling reagent to assess insulin- and hypoxia-
stimulated cell surface GLUT4 content in skeletal muscle from
type 2 diabetic patients. Diabetes. 2000;49(4):647–54.
23. Safdar A, Yardley NJ, Snow R, Melov S, Tarnopolsky MA. Global
and targeted gene expression and protein content in skeletal muscle
of young men following short-term creatine monohydrate supple-
mentation. Physiol Genomics. 2008;32(2):219–28.
24. Sigal RJ, Kenny GP, Wasserman DH, Castaneda-Sceppa C. Physi-
cal activity/exercise and type 2 diabetes. Diabetes Care.2004;27
(10):2518–39.
25. Thomas DE, Elliott EJ, Naughton GA. Exercise for type 2 diabetes
mellitus. Cochrane Database Syst Rev. 2006;3:CD002968.
26. van Loon LJ, Murphy R, Oosterlaar AM, et al. Creatine sup-
plementation increases glycogen storage but not GLUT-4 ex-
pression in human skeletal muscle. Clin Sci (Lond).2004;106(1):
99–106.
27. Wyss M, Kaddurah-Daouk R. Creatine and creatinine metabolism.
Physiol Rev. 2000;80(3):1107–213.
28. Zierath JR, He L, Guma A, Odegoard Wahlstrom E, Klip A,
Wallberg-Henriksson H. Insulin action on glucose transport and
plasma membrane GLUT4 content in skeletal muscle from patients
with NIDDM. Diabetologia. 1996;39(10):1180–9.
http://www.acsm-msse.org778 Official Journal of the American College of Sports Medicine
CLINICAL SCIENCES
Copyright © 2011 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
... 19,20,[28][29][30] The effects of creatine supplementation have been studied in a variety of chronic diseases in which muscle wasting is present, including diabetes, cancer, heart failure, and respiratory disease, with varying results. [31][32][33][34] In previous metaanalyses, creatine in combination with resistance training improves lean body mass and muscle strength in the older population, making it highly plausible that it may be effective in improving physical function in those at risk of functional disability. [35][36][37][38] This systematic review and meta-analysis will update and build on this previous work by including populations with chronic disease and studies that do not use resistance training. ...
... 59 Muscle function: Bench press strength Sixteen studies measured bench press strength, with data available from 15 studies for meta-analysis, including 450 participants. 31,49,50,52,53,56,[60][61][62][63][64][65][66][67][68][69] When compared with the placebo, creatine supplementation significantly increased bench press strength when analyzed by frequentist meta-analysis (n = 450; SMD: 0.25; 95% CI: 0.06-0.44; I 2 = 0%; P = 0.01; Figure 4). ...
... Fifteen studies measured leg press strength, including 463 participants. 31 Figure 4) and Bayesian methods (n = 463; SMD: 0.06; 95% CrI: −0.16 to 0.28; τ = 0.26; Figure S4). ...
Article
Full-text available
Background The efficacy of creatine replacement through supplementation for the optimization of physical function in the population at risk of functional disability is unclear. Methods We conducted a systematic literature search of MEDLINE, EMBASE, the Cochrane Library, and CINAHL from inception to November 2022. Studies included were randomized controlled trials (RCTs) comparing creatine supplementation with placebos in older adults and adults with chronic disease. The primary outcome was physical function measured by the sit‐to‐stand test after pooling data using random‐effects modeling. We also performed a Bayesian meta‐analysis to describe the treatment effect in probability terms. Secondary outcomes included other measures of physical function, muscle function, and body composition. The risk of bias was assessed using the Cochrane risk‐of‐bias tool. Results We identified 33 RCTs, comprising 1076 participants. From six trials reporting the primary outcome, the pooled standardized mean difference (SMD) was 0.51 (95% confidence interval [CI]: 0.01–1.00; I ² = 62%; P = 0.04); using weakly informative priors, the posterior probability that creatine supplementation improves physical function was 66.7%. Upper‐body muscle strength (SMD: 0.25; 95% CI: 0.06–0.44; I ² = 0%; P = 0.01), handgrip strength (SMD 0.23; 95% CI: 0.01–0.45; I ² = 0%; P = 0.04), and lean tissue mass (MD 1.08 kg; 95% CI: 0.77–1.38; I ² = 26%; P < 0.01) improved with creatine supplementation. The quality of evidence for all outcomes was low or very low because of a high risk of bias. Conclusion Creatine supplementation improves sit‐to‐stand performance, muscle function, and lean tissue mass. It is crucial to conduct high‐quality prospective RCTs to confirm these hypotheses (PROSPERO number, CRD42023354929).
... From a T2D perspective, creatine supplementation has the potential to improve glycemic control of skeletal muscle by improving GLUT-4 synthesis [128]. Furthermore, in a randomized, double-blind, placebo-controlled trial, Gualano and colleagues showed that 12 weeks of creatine supplementation (5g per day) combined with resistance exercise training reduced A1c compared to placebo (− 1.1% [CI -1.9; − 0.4%]; p = 0.004; [134]. Creatine supplementation has the ability to promote changes in glucose homeostasis that may favour a healthier metabolic profile, especially when combine with resistance exercise [135]. ...
Article
Full-text available
Aims: Sarcopenia generally refers to the age-related reduction in muscle strength, functional ability, and muscle mass. Sarcopenia is a multifactorial condition associated with poor glucose disposal, insulin resistance, and subsequently type 2 diabetes (T2D). The pathophysiological connection between sarcopenia and T2D is complex but likely involves glycemic control, inflammation, oxidative stress, and adiposity. Methods and results: Resistance exercise and aerobic training are two lifestyle interventions that may improve glycemic control in older adults with T2D and counteract sarcopenia. Further, there is evidence that dietary protein, Omega-3 fatty acids, creatine monohydrate, and Vitamin D hold potential to augment some of these benefits from exercise. Conclusions: The purpose of this narrative review is: (1) discuss the pathophysiological link between age-related sarcopenia and T2D, and (2) discuss lifestyle interventions involving physical activity and nutrition that may counteract sarcopenia and T2D.
... Creatine has also demonstrated a role in other diseases, such as diabetes, osteoporosis, cancer, and cardiovascular and neurological diseases [1,[3][4][5]. In addition to the aforementioned beneficial effects of creatine, a series of interesting direct and indirect cellular effects have also been demonstrated, such as the protection of tissues from ischemic and oxidative insults, the reduction of inflammatory markers, the antiapoptotic [48,49], antidiabetic [50,51], lipid-lowering role [52,53], and modulator of the immune and the intestinal system [54,55]. ...
Article
Full-text available
Creatine supplementation has been one of the most studied and useful ergogenic nutritional support for athletes to improve performance, strength, and muscular mass. Over time creatine has shown beneficial effects in several human disease conditions. This review aims to summarise the current evidence for creatine supplementation in advanced chronic liver disease and its complications, primarily in sarcopenic cirrhotic patients, because this condition is known to be associated with poor prognosis and outcomes. Although creatine supplementation in chronic liver disease seems to be barely investigated and not studied in human patients, its potential efficacy on chronic liver disease is indirectly highlighted in animal models of non-alcoholic fatty liver disease, bringing beneficial effects in the fatty liver. Similarly, encephalopathy and fatigue seem to have beneficial effects. Creatine supplementation has demonstrated effects in sarcopenia in the elderly with and without resistance training suggesting a potential role in improving this condition in patients with advanced chronic liver disease. Creatine supplementation could address several critical points of chronic liver disease and its complications. Further studies are needed to support the clinical burden of this hypothesis.
Preprint
Full-text available
Gut microbes play a vital role in human health and are influenced by numerous factors including diet, genetics, and environment. (Fermented) Camel milk, which is abundant in nutrients and lacks allergenic proteins, has been consumed for its edible and medicinal properties for centuries. Research on camel milk's impact on gut microbiota and host metabolism is still limited. The results found that sour camel milk contained various beneficial bacteria such as Lactobacillus helveticus, Acinetobacter lwoffii, Eubacterium coprostanoligenes group, Lachnospiraceae, which could be transported to the recipient's intestines by diet. This study specified that the transportation of microbiome happened both intra- and inter-species and played a principal role in the formation of progeny gut microflora. An investigation on type 2 diabetic rats revealed that the composition of gut microflora and serum metabolites of those fed with high-dose camel whey was closer to that of the normal. Eubacterium limnetica, which can reduce the risk of diseases by producing MtcB protein, was found in the gut microflora of the ones taking camel milk. These results evidenced the high potential of camel milk as a functional food.
Article
O Diabetes Mellitus tipo 2 é uma doença de caráter metabólico caracterizado por taxas elevadas de glicose sanguínea. Além do tratamento medicamentoso, o uso de suplementos tem sido bastante estudado para a manutenção da saúde, como a creatina, reconhecida como uma substância ergogênica, e recentemente tem sido demonstrada como opção terapêutica de diversas condições clínicas como é o caso de disfunções metabólicas. O objetivo desta pesquisa foi descrever os efeitos da suplementação da creatina em indivíduos adultos com Diabetes Mellitus Tipo 2, correlacionando com a massa muscular e o controle glicêmico associados a treinamento de força. O estudo foi realizado a partir de uma revisão de literatura, onde foram selecionados artigos nacionais e internacionais, das bases de dados e revistas indexadas na área da saúde: SciELO, Springer Link, PubMed e Portal Regional da BVS, que demonstrasse o uso de creatina em pessoas com diabetes associado ao treinamento de força, nos anos de 2001 a 2023. Foram encontrados nas bases de dados 9.661 resultados, dos quais apenas 19 foram incluídos após a aplicação dos critérios de inclusão e exclusão. Observa-se que a suplementação da creatina aliada a treinamento de força, aumenta a captação de glicose no músculo, o que leva a diminuição da hemoglobina glicada e uma expressiva queda nos níveis glicêmicos e melhora da massa muscular. Concluiu-se que a suplementação de creatina monoidratada associada a treinamento resistido é uma estratégia dietética segura e eficaz no que se refere ao controle glicêmico e massa muscular de indivíduos com diabetes tipo 2.
Article
Full-text available
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, affecting approximately 6.5 million older adults in the United States. Development of AD treatment has primarily centered on developing pharmaceuticals that target amyloid-β (Aβ) plaques in the brain, a hallmark pathological biomarker that precedes symptomatic AD. Though recent clinical trials of novel drugs that target Aβ have demonstrated promising preliminary data, these pharmaceuticals have a poor history of developing into AD treatments, leading to hypotheses that other therapeutic targets may be more suitable for AD prevention and treatment. Impaired brain energy metabolism is another pathological hallmark that precedes the onset of AD that may provide a target for intervention. The brain creatine (Cr) system plays a crucial role in maintaining bioenergetic flux and is disrupted in AD. Recent studies using AD mouse models have shown that supplementing with Cr improves brain bioenergetics, as well as AD biomarkers and cognition. Despite these promising findings, no human trials have investigated the potential benefits of Cr supplementation in AD. This narrative review discusses the link between Cr and AD and the potential for Cr supplementation as a treatment for AD.
Preprint
Background The efficacy of creatine replacement through supplementation for the optimisation of physical function in the population at risk of functional disability is unclear. Methods We conducted a systematic literature search of MEDLINE, EMBASE, Cochrane Library and CINAHL until November 2022. Studies included were randomised controlled trials comparing the use of creatine supplementation with placebo in older adults and adults with chronic disease. The primary outcome was physical function measured by the sit-to-stand test after pooling data using random effects modelling. We also performed a Bayesian meta-analysis to describe the treatment effect in probability terms. Secondary outcomes included other measures of physical function, muscle function and body composition. The risk of bias was assessed using the Cochrane risk-of-bias tool. Results We identified 33 RCTs, comprising 1076 participants. From 6 trials reporting the primary outcome, the pooled standardised mean difference was 0.51 (95% CI 0.01 to 1.00; I2=62%; p=0.04); using weakly informative priors, the posterior probability that creatine supplementation improves physical function was 66.7%. Upper body muscle strength (SMD 0.25, 95% CI 0.06 to 0.44; I2=0%; p=0.01), handgrip strength (SMD 0.23, 95% CI 0.01 to 0.45; I2=0%; p=0.04) and lean tissue mass (MD 1.08kg; 95% CI 0.77 to 1.38; I2=26%; p<0.01) improved with creatine supplementation. The quality of evidence for all outcomes was low or very low due to a high risk of bias. Conclusion Creatine supplementation improves sit-to-stand performance, muscle function and lean tissue mass. It is crucial to conduct high-quality prospective RCTs to confirm these hypotheses (Prospero number, CRD42023354929).
Article
Full-text available
Creatine has become one of the most popular dietary supplements among a wide range of healthy and clinical populations. However, its potential adverse effects on kidney health are still a matter of concern. This is a narrative review of the effects of creatine supplementation on kidney function. Despite a few case reports and animal studies suggesting that creatine may impair kidney function, clinical trials with controlled designs do not support this claim. Creatine supplementation may increase serum creatinine (Crn) concentration for some individuals, but it does not necessarily indicate kidney dysfunction, as creatine is spontaneously converted into Crn. Based on studies assessing kidney function using reliable methods, creatine supplements have been shown to be safe for human consumption. Further studies with people who have pre-existing kidney disease remain necessary.
Chapter
Nesta coletânea, buscou-se agregar uma visão geral e complementar acerca dos avanços científicos, tecnológicos e sociais da saúde, compilando as produções de estudiosos, acadêmicos e profissionais de saúde. Nesse sentido, a obra “Abordagens em medicina: avanços científicos, tecnológicos e sociais” foi proposta com o objetivo de contribuir para a atualização da literatura em saúde e fortalecer as discussões sobre avanços e aplicações de estudos na atenção à saúde em seus diferentes níveis de complexidade. A coletânea é dedicada a estudantes da área da saúde, profissionais de saúde, residentes em saúde, gestores e especialistas que desejem aprofundar conhecimentos em suas áreas e fora de sua especialidade.
Article
Full-text available
Creatine (Cr) plays a central role in energy provision through a reaction catalyzed by phosphorylcreatine kinase. Furthermore, this amine enhances both gene expression and satellite cell activation involved in hypertrophic response. Recent findings have indicated that Cr supplementation has a therapeutic role in several diseases characterized by atrophic conditions, weakness, and metabolic disturbances (i.e., in the muscle, bone, lung, and brain). Accordingly, there has been an evidence indicating that Cr supplementation is capable of attenuating the degenerative state in some muscle disorders (i.e., Duchenne and inflammatory myopathies), central nervous diseases (i.e., Parkinson’s, Huntington’s, and Alzheimer’s), and bone and metabolic disturbances (i.e., osteoporosis and type II diabetes). In light of this, Cr supplementation could be used as a therapeutic tool for the elderly. The aim of this review is to summarize the main studies conducted in this field and to highlight the scientific and clinical perspectives of this promising therapeutic supplement.
Article
Full-text available
The literature on creatine supplementation supporting its efficacy has grown rapidly and has included studies in both healthy volunteers and patient populations. However, the first rule in the development of therapeutic agents is safety. Creatine is well-tolerated in most individuals in short-term studies. However, isolated reports suggest creatine may be associated with various side effects affecting several organ systems including skeletal muscle, the kidney and the gastrointestinal tract. The majority of clinical studies fail to find an increased incidence of side effects with creatine supplementation. To date, studies have not found clinically significant deviations from normal values in renal, hepatic, cardiac or muscle function. Few data are available on the long-term consequences of creatine supplementation.
Article
Full-text available
Recent findings have indicated that creatine supplementation may affect glucose metabolism. This study aimed to examine the effects of creatine supplementation, combined with aerobic training, on glucose tolerance in sedentary healthy male. Subjects (n = 22) were randomly divided in two groups and were allocated to receive treatment with either creatine (CT) ( approximately 10 g . day over three months) or placebo (PT) (dextrose). Administration of treatments was double blind. Both groups underwent moderate aerobic training. An oral glucose tolerance test (OGTT) was performed and both fasting plasma insulin and the homeostasis model assessment (HOMA) index were assessed at the start, and after four, eight and twelve weeks. CT demonstrated significant decrease in OGTT area under the curve compared to PT (P = 0.034). There were no differences between groups or over time in fasting insulin or HOMA. The results suggest that creatine supplementation, combined with aerobic training, can improve glucose tolerance but does not affect insulin sensitivity, and may warrant further investigation with diabetic subjects.
Article
Full-text available
The timed-stands test (TST) is a simple measure of lower extremity strength which correlates with age in healthy people. We validated the TST as a functional assessment tool against other measurements of functional capacity and comorbidity in 147 patients with rheumatoid arthritis (RA) or other chronic diseases. The TST was significantly impacted by RA disease activity and by comorbidity. Age affected TST only in patients without arthritis or life threatening disease. TST is a simple, reproducible measure of lower extremity function that was valid in this patient population.
Article
Full-text available
Huntington's disease (HD) is a progressive neurodegenerative illness for which there is no effective therapy. We examined whether creatine, which may exert neuroprotective effects by increasing phosphocreatine levels or by stabilizing the mitochondrial permeability transition, has beneficial effects in a transgenic mouse model of HD (line 6/2). Dietary creatine supplementation significantly improved survival, slowed the development of brain atrophy, and delayed atrophy of striatal neurons and the formation of huntingtin-positive aggregates in R6/2 mice. Body weight and motor performance on the rotarod test were significantly improved in creatine-supplemented R6/2 mice, whereas the onset of diabetes was markedly delayed. Nuclear magnetic resonance spectroscopy showed that creatine supplementation significantly increased brain creatine concentrations and delayed decreases in N-acetylaspartate concentrations. These results support a role of metabolic dysfunction in a transgenic mouse model of HD and suggest a novel therapeutic strategy to slow the pathological process.
Article
1. Introduction Structure-function studies performed to investigate the betacytotropic effect of arginine, have suggested that the guanidino group may be involved in triggering insulin release, since several compounds that contain this radical within their molecule are capable of stimulating the secretion of insulin, both in vivo [l] and in vitro [2] . Given that arginine also behaves as a potent glucagon secretagogue, whose mechanism of action remains unknown, we have examined the effect of guanidine and two guanidine derivatives (guanidino- acetic acid and creatine) on the secretory activity of the alpha-cell in a preparation of isolated mouse pancreatic islets. 2. Methods and materials Four-week old Swiss male mice, fasted for 14 h, were used as pancreas donors. In each experiment a pool of 8 pancreases was employed. The islets were isolated by the collagenase technique [3] as modified by Coll-Garcia and Gill [4]. Batches of 10 islets were incubated for 60 min in 2 ml of a bicarbonate-buffered salt solution [S] (gas phase, 95:5, O2 t COa) contain- ing 0.2% bovine serum albumin (Cohn fraction IV), 2000 U of kallikrein-trypsin inhibitor (Trasylol, Bayer) and 3.3 mM glucose (‘basal medium’). In order to verify the releasing activity of the islets, on each day arginine hydrochloride was used as a control secretagogue. All incubations were performed in a water bath at 37°C and at 60 strokes per min. At the end of the incubation, an aliquot of each sample was 52 diluted 1:4 in 0.2 M glycine containing 0.25% human albumin and 1% normal sheep serum (pH 8.8) and immediately frozen at -20°C until the time of analysis. Full details of this method will be published elsewhere [6]. Radioimmunoassay was used to estimate glucagon [7] and insulin [8]. The values were read against beef-pork glucagon (Eli Lilly) and mouse insulin (Novo) standards. Statistical differences were calcu- lated by unpaired t-test analysis. Collagenase was purchased from Boehringer Mannheim GmbH; arginine hydrochloride, guanidine HCl, guanidinoacetic acid and bovine serum albumin were obtained from Sigma Chem. Co. Glucagon antiserum (30K) was a generous gift from Dr R. H. Unger, Dallas. 3. Results As expressed in table 1, glucagon release by mouse pancreatic islets incubated in the basal medium, was fairly constant in the three groups of experiments. The addition of 10 mM arginine induced, in all cases, a marked increase of glucagon output, the increments ranging from 140% to 410% of the basal values. When 10 mM guanidine was incorporated into the medium, glucagon release amounted to 110% above the basal value. Guanidinoacetic acid and creatin, both at 10 mM, were also effective stimuli of glucagon secretion, the increments being 50% and 140%, respectively. At the glucose concentration employed (3.3 mM), 10 mM arginine elicited significant increments of insulin release, varying from 180% to 590%. While
Article
Insulin secretion was measured in the isolated perfused rat pancreas in response to guanidinoacetic acid and other guanidine derivatives. Guanidinoacetic acid appears to be more potent than arginine, creatine and guanidine in stimulating insulin secretion. Parasympathetic mediation is unlikely because insulin secretion in response to these compounds is not inhibited by atropine. The guanidino group may be an important factor in arginine-stimulated insulin release. Glycine, which does not have a guanidino group, also increases insulin levels. It is possible that, in addition, arginine and glycine may release insulin by being converted to guanidinoacetic acid. (Endocrinology 86: 332, 1970)
Article
We investigated the response of the glucose transport system to insulin, in the presence of ambient glucose concentrations, in isolated skeletal muscle from seven patients with non-insulin-dependent diabetes mellitus (NIDDM) (age, 55 ± 3 years, BMI 27.4 ± 1.8 kg/m2) and seven healthy control subjects (age, 54 ± 3 years, BMI 26.5 ± 1.1 kg/m2). Insulin-mediated whole body glucose utilization was similar between the groups when studied in the presence of ambient glucose concentrations (approximately 10 mmol/l for the NIDDM patients and 5 mmol/l for the control subjects). Samples were obtained from the vastus lateralis muscle, by means of an open muscle biopsy procedure, before and after a 40-min insulin infusion. An increase in serum insulin levels from 54 ± 12 to 588 ± 42 pmol/l, induced a 1.6 ± 0.2-fold increase in glucose transporter protein (GLUT4) in skeletal muscle plasma membranes obtained from the control subjects (p < 0.05), whereas no significant increase was noted in plasma membrane fractions prepared from NIDDM muscles, despite a similar increase in serum insulin levels. At concentrations of 5 mmol/l 3-O-methylglucose in vitro, insulin (600 pmol/l) induced a 2.2-fold (p < 0.05) increase in glucose transport in NIDDM muscles and a 3.4-fold (P < 0.001) increase in the control muscles. Insulin-stimulated 3-O-methylglucose transport was positively correlated with whole body insulin-mediated glucose uptake in all participants (r = 0.78, p < 0.001) and negatively correlated with fasting plasma glucose levels in the NIDDM subjects (r = 0.93, p < 0.001). Muscle fibre type distribution and capillarization were similar between the groups. Our results suggest that insulin-stimulated glucose transport in skeletal muscle from patients with NIDDM is down-regulated in the presence of hyperglycaemia. The increased flux of glucose as a consequence of hyperglycaemia may result in resistance to any further insulin-induced gain of GLUT4 at the level of the plasma membrane.
Article
This study investigated the effect of carbohydrate (CHO) ingestion on skeletal muscle creatine (Cr) accumulation during Cr supplementation in humans. Muscle biopsy, urine, and plasma samples were obtained from 24 males before and after ingesting 5 g Cr in solution (group A) or 5 g Cr followed, 30 min later, by 93 g simple CHO in solution (group B) four times each day for 5 days. Supplementation resulted in an increase in muscle phosphocreatine (PCr), Cr, and total creatine (TCr; sum of PCr and Cr) concentration in groups A and B, but the increase in TCr in group B was 60% greater than in group A (P < 0.01). There was also a corresponding decrease in urinary Cr excretion in group B (P < 0.001). Creatine supplementation had no effect on serum insulin concentration, but Cr and CHO ingestion dramatically elevated insulin concentration (P < 0.001). These findings demonstrate that CHO ingestion substantially augments muscle Cr accumulation during Cr feeding in humans, which appears to be insulin mediated.
Article
The results differ concerning randomized controlled trials of the effects of metformin on blood glucose regulation and body weight. To get a systematic overview, a meta-analysis of the efficacy of metformin was performed by comparing metformin with placebo and sulfonylurea. All randomized controlled trials published since 1957 were selected by searching the Current List of Medical Literature, Cumulated Index Medicus, Medline, and Embase, Meta-analysis was performed calculating weighted mean difference (WMD) of fasting blood glucose, glycosylated hemoglobin, and body weight. Nine randomized controlled trials comparing metformin with placebo and ten comparing metformin with sulfonylurea were identified. The WMD between metformin and placebo after treatment for fasting blood glucose was -2.0 mmol/l (95% CI -2.4 to -1.7) and for glycosylated hemoglobin -0.9% (95% CI -1.1 to -0.7). Body weight WMD was not significant after treatment. Sulfonylurea and metformin lowered blood glucose and glycosylated hemoglobin equally, while there was a significant WMD of body weight (-2.9 kg [95% CI -4.4 to -1.1]) because of a 1.7-kg mean increase after sulfonylurea and a 1.2-kg mean decrease after metformin. Metformin lowers blood glucose and glycosylated hemoglobin significantly, compared with placebo. Metformin and sulfonylurea have an equal effect on fasting blood glucose and glycosylated hemoglobin, but the body weight is significantly lower after metformin compared with sulfonylurea treatment because of an increase in body weight after sulfonylurea treatment.