Low plasma carnosinase activity promotes carnosinemia after carnosine ingestion in humans

Share Embed


Descripción

Articles in PresS. Am J Physiol Renal Physiol (April 11, 2012). doi:10.1152/ajprenal.00084.2012



Low plasma carnosinase activity promotes carnosinemia following



carnosine ingestion in humans

3  4 

Running title: Supplementation-induced carnosinemia in humans

5  6 

Inge Everaert1, Youri Taes2, Emile De Heer3, Hans Baelde3, Ana Zutinic3, Benito Yard4, Sibylle



Sauerhöfer4 , Lander Vanhee1, Joris Delanghe5, Giancarlo Aldini6, Wim Derave1*

8  1



Department of Movement and Sport Sciences, Ghent University, Ghent, Belgium 2

10  3

11  12 

4

Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands

Department of Medicine V, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany

13  5

14  15 

Department of Endocrinology, Ghent University Hospital, Ghent, Belgium

6

Department of Clinical Chemistry, Ghent University Hospital, Ghent, Belgium

Department of Pharmaceutical Sciences “Pietro Pratesi”, Univerity of Milan, Milan, Italy

16  17 

* Contact information: Wim Derave, Department of Movement and Sports Sciences, Ghent

18 

University, Watersportlaan 2, B-9000 Ghent, Belgium. [email protected], 0032 (09) 264 63 26

19  20 

Copyright © 2012 by the American Physiological Society.

21 

Abstract

22 

Objectives A polymorphism in the CNDP1 gene, resulting in decreased plasma carnosinase activity,

23 

is associated with a reduced risk for diabetic nephropathy. Because carnosine, a natural

24 

scavenger/suppressor of reactive oxygen species, AGE’s and reactive aldehydes, is readily degraded

25 

in blood by the highly active carnosinase enzyme, it has been postulated that low serum carnosinase

26 

activity might be advantageous to reduce diabetic complications. The aim of this study was to

27 

examine whether a low carnosinase activity promotes circulating carnosine levels following

28 

carnosine supplementation in humans.

29 

Methods Blood and urine was sampled, in 25 healthy subjects following acute supplementation

30 

with 60mg/kg BW carnosine. Pre-cooled EDTA tubes were used for blood withdrawal and plasma

31 

samples were immediately deproteinized and analyzed for carnosine and beta-alanine by HPLC.

32 

CNDP1 genotype, baseline plasma carnosinase activity and protein content were assessed.

33 

Results Upon carnosine ingestion, 8 of the 25 subjects (= responders) displayed a measurable

34 

increase in plasma carnosine up to 1h following supplementation. Subjects with no measurable

35 

increment in plasma carnosine (= non-responders) had an approximately 2-fold higher plasma

36 

carnosinase protein content and an approximately 1.5-fold higher activity compared to the

37 

responders. The urinary carnosine recovery was 2.6-fold higher in the responders versus non-

38 

responders and was negatively dependent on both the activity and protein content of the plasma

39 

carnosinase enzyme.

40 

Conclusion A low plasma carnosinase activity promotes the presence of circulating carnosine upon

41 

an oral challenge. These data may further clarify the link between CNDP1 genotype, carnosinase

42 

and diabetic nephropathy.  

43 

Keywords CNDP1, beta-alanine, diabetic nephropathy

44 

Introduction

45 

Adequate glycemic and blood pressure control are the most effective therapeutic modalities in

46 

diabetic patients to delay the onset of microvascular complications (1). Nonetheless, an increasing

47 

number of patients will continue to develop microvascular complications despite these therapeutic

48 

measures. Ample evidence indicates that susceptibility to develop diabetic nephropathy, the most

49 

common cause of renal failure in the western world, is genetically determined (23). One of the

50 

genes that have recently been linked to diabetic nephropathy is carnosinase dipeptidase-1 (CNDP1),

51 

encoding the serum carnosinase enzyme (16). Diabetic nephropathy is strongly associated with a

52 

(CTG)n polymorphism in the CNDP1 gene, affecting serum carnosinase secretion (30). Diabetic

53 

patients homozygous for (CTG)5 have a lower risk to develop diabetic nephropathy and have a

54 

lower plasma carnosinase activity (16). Although the association between the CNDP1

55 

polymorphism and diabetic nephropathy have been confirmed in an independent study in European

56 

Americans (11), other studies did not show an association in type 1 diabetic patients (37) or showed

57 

that the association in type 2 diabetic patients is sex specific (25). Inconsistent findings may be

58 

explained by differences in ethnicity (22) or alternatively, by assuming that protection from diabetic

59 

nephropathy afforded by (CTG)5 homozygosity in CNDP1 may be masked by additional risk

60 

haplotypes (9; 22).

61 

Glycation and oxidative stress (glycoxidative stress) and accelerated formation of advanced

62 

glycation end-products (AGE’s) during hyperglycemia are implicated in the development of

63 

diabetic complications (8). Carnosine (β-alanyl-L-histidine) is a versatile dipeptide, mainly present

64 

in neuronal tissue and skeletal muscle, that has the propensity to suppress several aspects of

65 

glycoxidative stress, such as inhibition of AGE formation (14), quenching of reactive aldehydes (3)

66 

and suppression of oxidative stress (18). Moreover, it has been shown that carnosine inhibits

67 

angiotensin converting enzyme (ACE) (15), albeit only at high concentrations. The influence of

68 

carnosine on blood pressure control has thusfar not been demonstrated.

69 

In humans, circulating carnosine is readily degraded by the highly active serum carnosinase

70 

enzyme, which is secreted from the liver into the plasma (30). Therefore, plasma carnosine

71 

concentrations in fasted subjects are in general below the detection limits of current quantitative

72 

carnosine assays. Also the presence of carnosine in plasma (carnosinemia) following dietary intake

73 

of pure carnosine or meat, which is a rich source of carnosine, is controversial. Both Asatoor et al.

74 

(5) and Gardner et al. (12) could not detect carnosine in plasma after administration of a high dose

75 

of carnosine (respectively ~ 60mg/kg BW and 4g). Cooling of the samples following blood

76 

withdrawal however, resulted in the detection of a small amount of plasma carnosine in one subject

77 

in the latter study of Gardner et al. (12). Despite the use of EDTA blood tubes, which have been

78 

shown to inhibit the hydrolysis of carnosine in plasma, no plasma carnosine was detected after oral

79 

supplementation with 450mg carnosine (38). In line with this, both Harris et al. (13) and Yeum et

80 

al. (38) could not detect any carnosine in plasma after the ingestion of beef, chicken breast nor

81 

chicken broth. These results are in sharp contrast to the findings of Park et al. (27) who reported a

82 

peak plasma carnosine concentration of 32.7mg/L 3.5h after ingestion of ground beef. The

83 

discrepancy between these studies could be due to differences in blood handling or due to different

84 

carnosinase expression of the volunteers as Yeum et al. (38) used female subjects (40-60 years)

85 

which are characterized by higher carnosinase activity compared to males (10).

86 

The current working hypothesis to explain the physiological mechanism for the protective effect of

87 

(CTG)5 homozygosity puts forward that 1) a CNDP1 genetic predisposition leads to low serum

88 

carnosinase activity (16) 2) low carnosinase activity promotes higher concentrations of circulating

89 

carnosine, and 3) high circulating carnosine levels protect against hyperglycemia-induced cytotoxic

90 

metabolites, resulting from oxidative stress and glycation. Evidence for the latter has been provided

91 

in a number of animal studies were carnosine supplementation could delay the development and

92 

progression of diabetes in db/db mice (32) and resulted in reduced urinary markers of oxidative

93 

stress and AGE’s in obese Zucker rats (4). There is currently however insufficient experimental

94 

evidence that supports the assumption that low carnosinase activity promotes higher concentrations

95 

of circulating carnosine. Therefore, the current study aims to explore the impact of variation

96 

between humans in carnosinase activity on circulating carnosine concentrations. We hypothesize

97 

that higher circulating carnosine concentrations can be detected in subjects with low carnosinase

98 

activity, following a single large oral dose of carnosine (60 mg/kg BW).

99 

Methods

100 

Subjects

101 

Twenty-five subjects (age: 20-31 years, body weight: 70.9 ± 9.8 kg), both male (n = 15) and female

102 

(n = 10), participated in this study. All subjects were in good health and none of the participants was

103 

vegetarian. The study protocol was approved by the local ethical committee (Ghent University

104 

Hospital, Belgium) and written informed consent was obtained from all participants prior to the

105 

study.

106  107 

Study design

108 

Heparin plasma was obtained prior to the experiments and on the morning before the

109 

supplementation to quantify plasma carnosinase protein content and activity. After an overnight fast

110 

(at least 8 h), an indwelling catheter was inserted in an antecubital vein and blood was withdrawn

111 

before and 20, 30, 40, 60 and 120 minutes following oral supplementation of 60 mg/kg BW

112 

carnosine (mean ± SD: 4209 ± 577 mg), dissolved in 330 ml water. Blood samples for

113 

determination of carnosine were collected in pre-cooled (4°C) EDTA tubes and immediately

114 

centrifuged (4°C) to separate the plasma. The anticoagulant EDTA was chosen for its ability to

115 

chelate Zn2+ ions which are essential for the catalytic activity of carnosinase (38). Plasma samples

116 

were deproteinized with SSA (35%) and stored immediately at -20 °C until further analysis. Urine

117 

was collected in EDTA-coated tubes prior and 45, 90, 135, 180 and 240 minutes after the carnosine

118 

supplementation. The subjects were allowed to drink water and received a carnosine-free meal after

119 

blood collection.

120  121 

Determination of carnosine and beta-alanine by HPLC

122 

100 µL of deproteinized EDTA plasma and urine was dried under vacuum (40 °C). Dried residues

123 

were resolved with 40 μL of coupling reagent: methanol-triethylamine-H2O-phenylisothiocyanate

124 

(PITC) (7:1:1:1) and allowed to react for 20 minutes at room temperature. The samples were dried

125 

again and resolved in 100 μL of sodium acetate buffer (10 mM, pH 6.4). The same method was

126 

applied to the standard solutions of beta-alanine (Sigma) and carnosine (Flamma, dissolved in

127 

deionized-distilled water). The derivatized samples (20 μL) were applied to a Waters HPLC system

128 

with an Hypersilica column (4.6 x 150 mm, 5 μm) and UV detector (wavelength 210 nm). The

129 

column was equilibrated with buffer A [10 mM sodium acetate adjusted to pH 6.4 with 6 % acetic

130 

acid] and buffer B [60 % acetonitrile – 40 % buffer A] at a flow rate of 0.8 ml/min at room

131 

temperature. Limit of detection and quantification were respectively 3 and 10 µM.

132  133 

Determination of carnosine by LC-ESI-MS/MS

134 

Analyses of plasma carnosine of one subject were performed by using a validated LC-ESI-MS/MS

135 

method (26). Briefly, aliquots of 100 µl of EDTA-plasma samples were spiked with H-Tyr-His-OH

136 

as internal standard (20 µM final concentration), deproteinized by perchloric acid (PCA, 700 mM

137 

final concentration) and centrifuged at 18,000 rpm for 10 min. The supernatants were then diluted

138 

1:1 with mobile phase A (CH3CN/H2O/HFBA 90/10/0.1 v:v:v), filtered through 0.2 µm filters and

139 

then injected into a ThermoFinnigan Surveyor LC system equipped with a quaternary pump and

140 

connected through an electrospray interface (ESI) to a TSQ Quantum Triple Quadrupole Mass

141 

Spectrometer (ThermoFinnigan Italia, Milan, Italy). Chromatographic separations were done by

142 

reverse phase elution with a Phenomenex Sinergy polar-RP column (150 mm x 2 mm i.d.; particle

143 

size 4 µm) (Chemtek Analytica, Anzola Emilia, Italy) protected by a polar-RP guard column (4 mm

144 

x 2 mm i.d.; 4 µm) kept at 25 °C. Separations were done by gradient elution from 100% phase A to

145 

80% phase B (CH3CN) in 12 min at a flow rate of 0.2 ml min-1 (injection volume 10 µl); the

146 

composition of the eluent was then restored to 100% A within 1 min, and the system was re-

147 

equilibrated for 6 min. Quantitations were performed in multiple reaction monitoring (MRM) mode

148 

at 2.00 kV multiplier voltage, and the following MRM transitions of [M + H]+ precursor ion 

149 

product ions were selected as follows:

150 

– H-Tyr-His-OH (IS) m/z 319.2  156.5 + 301.6 (collision energy, 25 eV);

151 

– CAR: m/z 227.0  110.6 + 156.5 (collision energy, 25 eV);

152  153 

Determination of plasma carnosinase activity and protein content

154 

At baseline, the mean plasma carnosinase activity and protein content was quantified based on the

155 

activity and protein content of two different blood collections. Plasma carnosinase activity was

156 

determined according to the method described by Teufel et al (35). Briefly, the reaction was

157 

initiated by addition of substrate (L-carnosine) to a heparin plasma sample and stopped after 10 min

158 

of incubation at 37 °C by adding 1 % SSA. Liberated histidine was derivatized with o-

159 

phtaldialdehyde (OPA) and the maximum increase was used for determining the maximum activity.

160 

Fluorescence was measured by excitation at 360 nm and emission at 460 nm. The intra- and inter-

161 

assay variations were respectively 7 % and 25 %. The lowest carnosinase activity detectable was

162 

0.117 µmol/ml/h.

163 

Plasma carnosinase protein content was measured by ELISA. In brief, a human CN1 ELISA was

164 

developed by coating high absorbant microtitre plates (Greiner BioChemia, Flacht, Germany)

165 

overnight with 100 µl of goat polyclonal anti-human CN1 (1 µg/ml) (R&D, Wiesbaden Germany).

166 

The plates were extensively washed and incubated with 5 % w/v of dry milk powder to avoid

167 

unspecific binding. For each sample and standard serial dilution were carried out. The plates were

168 

placed on a shaker for 1 hr and subsequently extensively washed with PBS/Tween. Hereafter anti-

169 

human carnosinase monoclonal antibody (clone ATLAS, Abcam) was added for 1 hr followed by

170 

extensively washing. HRP conjugated goat anti-mouse IgG was added for 1 hr and the plates were

171 

washed. After addition of peroxidase substrate (deep-Blue POD) (Roche diagnostics, Mannheim,

172 

Germany) the reaction was stopped after 15 minutes by addition of 50 µl of 1 M H2SO4 and read in

173 

an ELISA reader at 450 nm. CN1 protein concentrations were assessed in the linear part of the

174 

dilution curve. Sensitivity of the ELISA was approximately 20 ng/ml.

175  176 

CNDP1 genotyping

177 

A more detailed description of the CNDP1 genotype determination is explained in the study of

178 

Mooyaart et al. (24). In brief, a standard PCR protocol was used with primers 5-FAM-

179 

GCGGGGAGGGTGAGGAGAAC (forward) and GGTAACAGACCTTCTTGAGGAATT-TGG

180 

(reverse). The denaturing, annealing and extension temperatures were 94 °C, 60 °C and 72 °C,

181 

respectively. After PCR amplification, fragment analysis was performed on the ABI3130 analyzer

182 

(Perkin Elmer) to determine the fragment length corresponding with the different genotypes. Each

183 

peak corresponded with the number of leucine repeats on each allele. A 157, 160 and 163 base pair

184 

product corresponded with 5, 6 and 7 CTG codons encoding for 5, 6 and 7 leucine repeats,

185 

respectively.

186  187 

Statistics

188 

Data are expressed as mean ± SD. Bivariate correlations and independent sample T-tests were used

189 

for statistical analysis (SPSS 17).

190  191  192  193  194  195 

196 

Results

197 

Carnosine is detectable in human plasma by HPLC.

198 

A HPLC chromatogram of EDTA plasma derivatized with PITC (UV detection) of a subject 30

199 

minutes following 60 mg/kg BW carnosine ingestion is depicted in Figure 1A. Spiking the sample

200 

with a carnosine standard results in an identical chromatogram except for the higher peak at 15.6

201 

min, representing carnosine. If the necessary precautions to block the endogenous carnosinase

202 

activity are not taken (heparin instead of EDTA tubes; not pre-cooled; not immediately

203 

deproteinized), then the same subject at the same time point does not display a carnosine peak at

204 

15.6 min (Figure 1B) and the peak of beta-alanine (the product of the carnosinase reaction)

205 

increases, which illustrates that the peak at 15.6 min genuinely represents carnosine. Plasma

206 

samples of one subject were analysed by an independent laboratory with LC-ESI-MS/MS method

207 

(26) and carnosine was detected at 30 (11.8µM) and 45 (3.4µM) minutes following carnosine

208 

supplementation (4g) while at the other time-points no carnosine could be detected.

209  210 

Supplementation-induced carnosinemia depends on CN1 protein content and activity.

211 

The mean CN1 protein levels varied widely between subjects from 24.38 to 148.02 µg/ml (mean ±

212 

SD: 77.82 ± 30.98 µg/ml) and the CN1 activity values were situated between 2.79 and 10.90

213 

µmol/ml/h (mean ± SD: 5.95 ± 1.91 µmol/ml/h). Furthermore, the CN1 protein content is positively

214 

correlated to the activity of the enzyme (p = 0.004, r = 0.58, R² = 0.34, Figure 2), confirming that

215 

the activity level is largely determined by the amount of enzyme available in the plasma. Upon

216 

carnosine ingestion, 8 of the 25 subjects displayed an increase in plasma carnosine concentration

217 

(carnosinemia), which we termed as responders (increase of > 10 µM carnosine after

218 

supplementation). The increase in plasma carnosine reached its Cmax (mean ± SD: 73.3 ± 59.7 µM;

219 

range: 30.7 - 195 µM) at on average 30 - 40 minutes after supplementation and rapidly declined

220 

within 1 - 2 hours (Figure 3). However, the remaining 17 subjects had no measurable increment (=

221 

non-responders) in plasma carnosine after oral supplementation with a high dose of carnosine,

222 

despite the precautions that were taken to block the carnosinase activity during blood collection.

223 

Post-hoc analysis revealed that there was a marked difference in plasma carnosinase protein content

224 

(p < 0.001) and activity (p = 0.007) between the responders and non-responders (Figure 4A and B).

225 

The mean plasma carnosinase protein levels were approximately 2-fold higher in the non-

226 

responders (mean ± SD: 91.57 ± 25.37 µg/ml) compared to the responders group (mean ± SD:

227 

44.42 ± 11.22 µg/ml, p < 0.001). Moreover, it seems that there is a clear cut-off value in plasma

228 

carnosinase protein content (Figure 4A), as the highest value of the responders was approximately

229 

the same as the lowest of the non-responders (55 µg/ml). Likewise, the CN1 activity is

230 

approximately 1.5-fold higher in non-responders (mean ± SD: 6.65 ± 1.80 µmol/ml/h) compared to

231 

the responders (mean ± SD: 4.53 ± 1.26 µmol/ml/h; p = 0.007, Figure 4B). Also within the

232 

responders’ group, the amount of plasma carnosine (AUC) is negatively correlated with the plasma

233 

carnosinase protein levels (p = 0.059, r = -0.68, R² = 0.47) and carnosinase activity (p = 0.059, r = -

234 

0.73, R² = 0.52). Both women (n = 2) and men (n = 6) and subjects with 5-5 (n = 3), 5-7 (n = 1) and

235 

6-6 (n = 4) CNDP1 genotype were represented in the responders group (Table 1). The mean age of

236 

responders (mean ± SD: 21.6 ± 0.7 yrs) was significantly lower (p = 0.045) compared to the mean

237 

age of the non-responders (mean ± SD: 23.1 ± 2.6 yrs).

238  239 

Urinary carnosine is related to supplementation-induced carnosinemia.

240 

The urinary carnosine recovery (% of ingested dose) varied largely between 0.23 and 13.27 %. The

241 

responders had a significantly (p = 0.006) higher recovery (mean ± SD: 7.7 ± 3.6 %) compared to

242 

the non-responders (mean ± SD: 2.9 ± 1.3%, Figure 5A). Furthermore, a strong negative association

243 

was observed between the urinary carnosine (% of ingested dose) and the plasma carnosinase

244 

activity (p = 0.001, r = -0.64, R² = 0.41, Figure 5B) and protein content (p < 0.001, r = -0.66, R² =

245 

0.44, Figure 5C). There was a trend for a positive correlation between the areas under the curve of

246 

plasma and urinary carnosine within the responders group (n = 8, p = 0.07, r = 0.66, R² = 0.44). The

247 

kinetics of urinary carnosine 4h after carnosine supplementation were comparable between the

248 

responders and non-responders. The majority was excreted during the first hour (responders: 45.6

249 

%; non-responders: 58.6 %) and thereafter, the concentration slowly began to decline and 90 % - 95

250 

% of the increase in urinary carnosine was disappeared 4h after carnosine ingestion.

251  252 

Profiling beta-alanine in plasma after carnosine ingestion

253 

Figure 6 shows the profile of beta-alanine in plasma after 60 mg/kg carnosine supplementation

254 

separated for subjects with or without supplementation-induced carnosinemia. The total amount of

255 

beta-alanine detected in plasma (AUC) was significantly higher in the responders group (p = 0.005,

256 

+ 30%) versus the non-responders. The Cmax was reached after 30 min for non-responders (mean ±

257 

SD: 382.6 ± 122.9 µM), after 40 min for responders (mean ± SD: 468.2 ± 66.0µM) and beta-alanine

258 

was almost completely removed from the plasma 2h after supplementation. Furthermore, the plasma

259 

beta-alanine content was negatively correlated to the plasma carnosinase protein levels (p = 0.001, r

260 

= -0.61, R² = 0.37) and activity (p = 0.019, r = -0.47, R² = 0.22).

261  262 

Side effects

263 

During the first hour after supplementation with 60 mg/kg BW carnosine, a total of 6 participants

264 

transiently suffered from both headache and paraesthesia symptoms, 2 subjects only from

265 

paraesthesia and 2 subjects from headache. The presence of these symptoms was not related to

266 

supplementation-induced carnosinemia, CNDP1 genotype or gender. However, subjects

267 

complaining about paraesthesia and/or headache had a higher total amount of beta-alanine in urine

268 

(symptoms: 93.7 ± 66.8 mg, no symptoms: 48.1 ± 45.6 mg, p = 0.05), although the total amount of

269 

plasma beta-alanine was not different compared to subjects without paraesthesia and/or headache

270 

(symptoms: 1.4 ± 0.2 mM, no symptoms: 1.3 ± 0.4 mM, p > 0.5).

271 

Discussion

272 

The main finding of this study is that a high carnosinase activity counteracts the presence of

273 

circulating carnosine upon an oral challenge. This likely provides a missing link in the

274 

pathophysiological mechanism between carnosine, CNDP1 genotype and diabetic

275 

complications, as shown in Figure 7. The mechanism of the protective effect of the Mannheim

276 

allele in diabetic nephropathy has been attributed to a lower carnosinase activity (16).

277 

However, the further link with higher circulating carnosine has never been established.

278 

Carnosine has been shown to be a natural scavenger of the hyperglycemia-induced

279 

overproduction of reactive oxygen species, AGE’s and reactive aldehydes and is therefore a

280 

promising candidate therapeutic molecule in reducing the risk to develop diabetic

281 

complications. More interestingly, Riedl et al. (31) recently showed that carnosinase activity

282 

is increased by hyperglycemia through N-glycosylation, and is elevated in type-2 diabetic

283 

patients. Therefore, hyperglycemia not only directly induces glycoxidative stress, but also

284 

indirectly suppresses the endogenous protective mechanism through carnosine, which

285 

probably further speeds the development of complications (Figure 7).

286 

The findings of this study implicate that the quantification of the plasma carnosinase protein

287 

content could be a reliable tool to determine the risk for developing nephropathy in diabetic

288 

patients as there is almost no overlap between subjects with or without supplementation-

289 

induced carnosinemia with respect to plasma carnosinase protein content. Interestingly, this

290 

indirectly takes the glycemic control into account, as carnosinase protein levels are elevated in

291 

hyperglycemic conditions (31). In 2005, Janssen et al. (16) recommended to investigate the

292 

potential of the CNDP1 variants in predicting the risk for developing diabetic nephropathy.

293 

However, the potential of the CNDP1 gene may be limited as 1) not all patients with

294 

homozygosity for the Mannheim allele were protected against diabetic nephropathy (16) and

295 

2) the subjects with supplementation-induced carnosinemia in this study were characterized

296 

with both 5-5, 5-7 and 6-6 CNDP1 genotype. Therefore, it would be interesting to investigate

297 

whether diabetic patients with a low plasma carnosinase protein content will be more

298 

protected against diabetic nephropathy compared to patients with a higher carnosinase protein

299 

content (irrespective of CNDP1 genotype).

300 

The carnosinase activity and protein content was shown to be a discriminating factor for

301 

supplementation-induced carnosinemia. Consequently, a potential therapeutic strategy to

302 

reduce the risk of diabetic nephropathy could be the inactivation of the carnosinase enzyme.

303 

In addition to the (CTG)n polymorphism in the CNDP1 gene (10; 16; 24), female gender (6;

304 

10; 29) and increasing age (29) are determinants associated with higher carnosinase activity

305 

that can not be manipulated. In vitro experiments, however, revealed that the activity of

306 

carnosinase enzyme was markedly decreased in the presence of both homocarnosine (gamma-

307 

aminobutyryl-L-histidine) (28; 29) and anserine (beta-alanyl-N1-methylhistidine) (28).

308 

Though, the correlation between circulating homocarnosine or anserine levels in fasted

309 

subjects and plasma carnosinase activity in vivo is less obvious (28; 29). Importantly, a good

310 

glycemic control, either by intervention with insulin, exercise or diet, seems to be crucial for

311 

diabetic patients as this not only directly influences oxidative and glycation stress, but also

312 

indirectly affects carnosinase activity (31). Besides the inactivation of the carnosinase

313 

enzyme, the development of a carnosine-analogue resistant to the carnosinase enzyme would

314 

be a promising strategy to prevent patients from diabetic complications. In this light, Aldini et

315 

al. (4) recently showed that the enantiomer D-carnosine has the same protective effect as L-

316 

carnosine in obese Zucker rats, while it is more resistant to carnosinase activity. However, D-

317 

carnosine has a lowered bioavailability as it is less absorbed in respect to L-carnosine and

318 

therefore the relevance for human use may be limited (4).

319 

In the assumption that dietary intake of carnosine can diminish the risk for diabetic

320 

complications, the lack of carnosine in the diet, as is the case in a vegetarian diet, could be

321 

deleterious for the protection against glycation and oxidative stress. Indeed, plasma AGE

322 

content of healthy vegetarians has been reported to be higher compared to omnivores (19; 33).

323 

Although a low-fat carbohydrate-rich vegetarian diet is often recommended for diabetic

324 

patients (for its positive impact on insulin sensitivity, blood pressure, serum lipid profile, etc

325 

(21)), the dietary intake of carnosine, perhaps by supplementation, should not be neglected.

326 

Carnosine is, even after meat or carnosine supplementation, hard to detect in human plasma,

327 

as a result of the high carnosinase activity. Yet, when several precautions are taken (cooling,

328 

use of EDTA tubes and the immediate deproteinization), when pharmacological doses of

329 

carnosine (60mg/kg BW) are ingested and when subjects with low carnosinase activity are

330 

studied, plasma carnosine levels can be clearly quantified up to 1h after ingestion. In contrast

331 

to plasma, urinary carnosine is more easily detectable, even in subjects ingesting their usual

332 

diets (0.2µM - 18.6µM) (2; 36). This urinary carnosine excretion is increased after carnosine

333 

or meat supplementation with a urinary recovery up to 14% of the ingested amount (12; 38).

334 

As there is no increase in urinary carnosine after beta-alanine (2g) and histidine (2g) intake,

335 

Gardner et al. (12) hypothesized that the excreted carnosine is not arisen from resynthesis of

336 

beta-alanine and histidine after hydrolysis in intestine and/or plasma and that the amount of

337 

urinary carnosine is a reflection of plasma carnosine. This hypothesis is confirmed in the

338 

present study by the positive correlation between urinary and plasma carnosine in the

339 

responders group and by the strong negative relation between urinary carnosine and plasma

340 

carnosinase activity and protein content (Figure 5B and 5C). Following carnosine

341 

supplementation, urinary carnosine excretion seems to be a reliable and easier measurable

342 

estimation of plasma carnosine content, which is equally dependent on serum carnosinase

343 

activity and content.

344 

Since renal tubular epithelium is equipped with oligopeptide transporters with a high affinity

345 

for carnosine (17; 34), circulating carnosine can be accumulated by the kidney and may

346 

provide an additional exogenous source of protective peptides against diabetic metabolites in

347 

patients with low levels of carnosinase.

348 

One would expect that beta-alanine, the degradation product of the plasma carnosinase

349 

enzyme, would be positively related to the carnosinase activity. However, the opposite is true

350 

as the responders group showed a higher total amount of plasma beta-alanine compared to the

351 

non-responders. This may suggest that the majority of carnosine is in fact degraded in other

352 

compartments and tissues, than the circulation. Thus, low carnosinase activity favors both

353 

carnosinemia and beta-alaninemia following carnosine supplementation, although the

354 

mechanism for the latter is unclear.

355 

It can be concluded that 1) carnosine can be detected in human plasma following oral

356 

ingestion and 2) a high carnosinase activity and content potently counteracts the presence of

357 

circulating carnosine. In a diabetic environment, this could impede the ability of carnosine to

358 

exert its protective effects against cytotoxic agents leading to diabetic complications. The

359 

inhibition of the carnosinase enzyme and the development of a carnosine analogue resistant to

360 

carnosinase have to be investigated as potential therapeutic strategies for reducing the risk for

361 

diabetic complications.

362 

Acknowledgements

363 

The practical contribution of Anneke Volkaert, Sam Beelprez, Joren Biesbrouck and Fiona

364 

Albers is greatly acknowledged. We thank Flamma (Italy) for generously providing carnosine.

365 

Grants

366 

This study was financially supported by grants from the Research Foundation – Flanders

367 

(FWO 1.5.149.08 and G.0046.09).

368  369  370  371  372  373  374  375  376  377  378  379  380  381  382  383  384  385  386 

Reference List

387  388  389 

1.

UK Prospective Diabetes Study (UKPDS) Group: Intensive blood-glucose control

390 

with sulphonylureas or insulin compared with conventional treatment and risk of

391 

complications in patients with type 2 diabetes (UKPDS 33). Lancet 352: 837-853,

392 

1998.

393 

2. Abe H, Okuma E, Sekine H, Maeda A and Yoshiue S. Human urinary excretion of

394 

L-histidine - related compounds after ingestion of several meats and fish muscle. Int J

395 

Biochem 25: 1245-1249, 1993.

396 

3. Aldini G, Granata P and Carini M. Detoxification of cytotoxic alpha,beta-

397 

unsaturated aldehydes by carnosine: characterization of conjugated adducts by

398 

electrospray ionization tandem mass spectrometry and detection by liquid

399 

chromatography/mass spectrometry in rat skeletal muscle. J Mass Spectrom 37: 1219-

400 

1228, 2002.

401 

4. Aldini G, Orioli M, Rossoni G, Savi F, Braidotti P, Vistoli G, Yeum KJ, Negrisoli

402 

G and Carini M. The carbonyl scavenger carnosine ameliorates dyslipidemia and

403 

renal function in zucker obese rats. J Cell Mol Med 15:1339-1354, 2011.

404  405 

5. Asatoor AM, Bandoh JK, Lant AF, Milne MD and Navab F. Intestinal absorption of carnosine and its constituent amino acids in man. Gut 11: 250-254, 1970.

406 

6. Bando K, Shimotsuji T, oyoshima H and Miyae K. Fluoremetric assay of human

407 

serum carnosinase activity in normal children, adults and patients with myopathy. Ann

408 

Clin Biochem 21: 510-514, 1984.

409 

7. Bauchart C, Savary-Auzeloux I, Patureau MP, Thomas E, Morzel M and

410 

Remond D. Carnosine concentration of ingested meat affects carnosine net release

411 

into the portal vein of minipigs. J Nutr 137: 589-593, 2007.

412  413 

8. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes 54: 1615-1625, 2005.

414 

9. Chakkera HA, Hanson RL, Kobes S, Millis MP, Nelson RG, Knowler WC and

415 

Distefano JK. Association of variants in the carnosine peptidase 1 gene (CNDP1)

416 

with diabetic nephropathy in American Indians. Mol Genet Metab 103:185-190, 2011.

417 

10. Everaert I, Mooyaart A, Baguet A, Zutinic A, Baelde H, Achten E, Taes Y, de HE

418 

and Derave W. Vegetarianism, female gender and increasing age, but not CNDP1

419 

genotype, are associated with reduced muscle carnosine levels in humans. Amino

420 

Acids 40: 1221-1229, 2011.

421 

11. Freedman BI, Hicks PJ, Sale MM, Pierson ED, Langefeld CD, Rich SS, Xu J,

422 

McDonough C, Janssen B, Yard BA, van der Woude FJ and Bowden DW. A

423 

leucine repeat in the carnosinase gene CNDP1 is associated with diabetic end-stage

424 

renal disease in European Americans. Nephrol Dial Transplant 22: 1131-1135, 2007.

425 

12. Gardner M, Illingworth, Kelleher and Wood. Intestinal absorption of the intact

426 

peptide carnosine in man, and comparison with intestinal permeability of lactulose. J

427 

Physiol 439: 411-422, 1991.

428 

13. Harris RC, Tallon MJ, Dunnett M, Boobis L, Coakley J, Kim HJ, Fallowfield JL,

429 

Hill CA, Sale C and Wise JA. The absorption of orally supplied beta-alanine and its

430 

effect on muscle carnosine synthesis in human vastus lateralis. Amino Acids 30: 279-

431 

289, 2006.

432  433 

14. Hipkiss AR, Michaelis J and Syrris P. Non-enzymatic glycosylation of the dipeptide L-carnosine, a potential anti-protein-cross-linking agent. FEBS Lett 371: 81-85, 1995.

434 

15. Hou WC, Chen HJ and Lin YH. Antioxidant peptides with Angiotensin converting

435 

enzyme inhibitory activities and applications for Angiotensin converting enzyme

436 

purification. J Agric Food Chem 51: 1706-1709, 2003.

437 

16. Janssen B, Hohenadel D, Brinkkoetter P, Peters V, Rind N, Fischer C, Rychlik I,

438 

Cerna M, Romzova M, de HE, Baelde H, Bakker SJ, Zirie M, Rondeau E,

439 

Mathieson P, Saleem MA, Meyer J, Koppel H, Sauerhoefer S, Bartram CR,

440 

Nawroth P, Hammes HP, Yard BA, Zschocke J and van der Woude FJ. Carnosine

441 

as a protective factor in diabetic nephropathy: association with a leucine repeat of the

442 

carnosinase gene CNDP1. Diabetes 54: 2320-2327, 2005.

443 

17. Kamal MA, Jiang H, Hu Y, Keep RF and Smith DE. Influence of genetic knockout

444 

of Pept2 on the in vivo disposition of endogenous and exogenous carnosine in wild-

445 

type and Pept2 null mice. Am J Physiol Regul Integr Comp Physiol 296: R986-R991,

446 

2009.

447 

18. Kohen R, Yamamoto Y, Cundy KC and Ames BN. Antioxidant activity of

448 

carnosine, homocarnosine, and anserine present in muscle and brain. Proc Natl Acad

449 

Sci U S A 85: 3175-3179, 1988.

450  451 

452  453 

19. Krajcovicova-Kudlackova M, Sebekova K, Schinzel R and Klvanova J. Advanced glycation end products and nutrition. Physiol Res 51: 313-316, 2002.

20. Lenney JF, Peppers SC, Kucera-Orallo CM and George RP. Characterization of human tissue carnosinase. Biochem J 228: 653-660, 1985.

454 

21. McCarty MF. The low-AGE content of low-fat vegan diets could benefit diabetics -

455 

though concurrent taurine supplementation may be needed to minimize endogenous

456 

AGE production. Med Hypotheses 64: 394-398, 2005.

457 

22. McDonough CW, Hicks PJ, Lu L, Langefeld CD, Freedman BI and Bowden DW.

458 

The influence of carnosinase gene polymorphisms on diabetic nephropathy risk in

459 

African-Americans. Hum Genet 126: 265-275, 2009.

460 

23. Mooyaart AL, Valk EJ, van Es LA, Bruijn JA, de HE, Freedman BI, Dekkers

461 

OM and Baelde HJ. Genetic associations in diabetic nephropathy: a meta-analysis.

462 

diabetologia 54: 544-553, 2011.

463 

24. Mooyaart AL, van Valkengoed IG, Shaw PK, Peters V, Baelde HJ, Rabelink TJ,

464 

Bruijn JA, Stronks K and de HE. Lower frequency of the 5/5 homozygous CNDP1

465 

genotype in South Asian Surinamese. Diabetes Res Clin Pract 85: 272-278, 2009.

466 

25. Mooyaart AL, Zutinic A, Bakker SJ, Grootendorst DC, Kleefstra N, van

467 

Valkengoed IG, Bohringer S, Bilo HJ, Dekker FW, Bruijn JA, Navis G, Janssen

468 

B, Baelde HJ and de HE. Association between CNDP1 genotype and diabetic

469 

nephropathy is sex specific. Diabetes 59: 1555-1559, 2010.

470 

26. Orioli M, Aldini G, Beretta G, Facino RM and Carini M. LC-ESI-MS/MS

471 

determination of 4-hydroxy-trans-2-nonenal Michael adducts with cysteine and

472 

histidine-containing peptides as early markers of oxidative stress in excitable tissues. J

473 

Chromatogr B Analyt Technol Biomed Life Sci 827: 109-118, 2005.

474  475 

27. Park Y, Volpe SL and Decker EA. Quantification of carnosine in human plasma after dietary consumption of beef. J Agric Food Chem 53: 4736-4739, 2005.

476 

28. Peters V, Jansen EE, Jakobs C, Riedl E, Janssen B, Yard BA, Wedel J,

477 

Hoffmann GF, Zschocke J, Gotthardt D, Fischer C and Koppel H. Anserine

478 

inhibits carnosine degradation but in human serum carnosinase (CN1) is not correlated

479 

with histidine dipeptide concentration. Clin Chim Acta 412: 263-267, 2011.

480 

29. Peters V, Kebbewar M, Jansen EW, Jakobs C, Riedl E, Koeppel H, Frey D,

481 

Adelmann K, Klingbeil K, Mack M, Hoffmann GF, Janssen B, Zschocke J and

482 

Yard BA. Relevance of allosteric conformations and homocarnosine concentration on

483 

carnosinase activity. Amino Acids 38: 1607-1615, 2010.

484 

30. Riedl E, Koeppel H, Brinkkoetter P, Sternik P, Steinbeisser H, Sauerhoefer S,

485 

Janssen B, van der Woude FJ and Yard BA. A CTG polymorphism in the CNDP1

486 

gene determines the secretion of serum carnosinase in Cos-7 transfected cells.

487 

Diabetes 56: 2410-2413, 2007.

488 

31. Riedl E, Koeppel H, Pfister F, Peters V, Sauerhoefer S, Sternik P, Brinkkoetter

489 

P, Zentgraf H, Navis G, Henning RH, Van Den Born J, Bakker SJ, Janssen B,

490 

van der Woude FJ and Yard BA. N-glycosylation of carnosinase influences protein

491 

secretion and enzyme activity: implications for hyperglycemia. Diabetes 59: 1984-

492 

1990, 2010.

493 

32. Sauerhofer S, Yuan G, Braun GS, Deinzer M, Neumaier M, Gretz N, Floege J,

494 

Kriz W, van der Woude F and Moeller MJ. L-carnosine, a substrate of carnosinase-

495 

1, influences glucose metabolism. Diabetes 56: 2425-2432, 2007.

496 

33. Sebekova K, Krajcoviova-Kudlackova M, Schinzel R, Faist V, Klvanova J and

497 

Heidland A. Plasma levels of advanced glycation end products in healthy, long-term

498 

vegetarians and subjects on a western mixed diet. Eur J Nutr 40: 275-281, 2001.

499 

34. Shen H, Smith DE, Yang T, Huang YG, Schnermann JB and Brosius FC 3rd.

500 

Localization of PEPT1 and PEPT2 proton-coupled oligopeptide transporter mRNA

501 

and protein in rat kidney. Am J Physiol 276 (5 Pt 2): F658-F665, 1999.

502 

35. Teufel M, Saudek V, Ledig JP, Bernhardt A, Boularand S, Carreau A, Cairns

503 

NJ, Carter C, Cowley DJ, Duverger D, Ganzhorn AJ, Guenet C, Heintzelmann

504 

B, Laucher V, Sauvage C and Smirnova T. Sequence identification and

505 

characterization of human carnosinase and a closely related non-specific dipeptidase. J

506 

Biol Chem 278: 6521-6531, 2003.

507 

36. Tsuruta Y, Maruyama K, Inoue H, Kosha K, Date Y, Okamura N, Eto S and

508 

Kojima E. Sensitive determination of carnosine in urine by high-performance liquid

509 

chromatography using 4-(5,6-dimethoxy-2-phthalimidinyl)-2-methoxyphenylsulfonyl

510 

chloride as a fluorescent labeling reagent. J Chromatogr B Analyt Technol Biomed

511 

Life Sci 878: 327-332, 2010.

512 

37. Wanic K, Placha G, Dunn J, Smiles A, Warram JH and Krolewski AS. Exclusion

513 

of polymorphisms in carnosinase genes (CNDP1 and CNDP2) as a cause of diabetic

514 

nephropathy in type 1 diabetes: results of large case-control and follow-up studies.

515 

Diabetes 57: 2547-2551, 2008.

516 

38. Yeum KJ, Orioli M, Regazzoni L, Carini M, Rasmussen H, Russell RM and

517 

Aldini G. Profiling histidine dipeptides in plasma and urine after ingesting beef,

518 

chicken or chicken broth in humans. Amino Acids 38:847-858, 2009.

519  520 

 

Tables & figures

521 

522 

Table I

523 

CN1 activity and protein content (mean ± SD) separated for CNDP1 genotype and gender (* p <

524 

0.05 vs. male, † p < 0.1 vs. male). The subjects in the responders group were both males and

525 

females and were characterized with both 5-5, 5-7 and 6-6 CNDP1 genotype.

526 

Figure 1

527 

A. HPLC chromatogram of human plasma (subject from the responders group) withdrawn 30

528 

minutes after ingestion of 60mg/kg BW carnosine in a cooled EDTA tube which was immediately

529 

deproteinized (solid line) and the same sample spiked with standard carnosine (dotted line). The

530 

peak at 15.6 min (carnosine) increased, while all other peaks remained the same (including beta-

531 

alanine at 12.1 min).

532 

B. HPLC chromatogram from human plasma (subject from the responders group) withdrawn 40

533 

minutes after ingestion of 60mg/kg BW carnosine in 1/ a cooled EDTA tube which was

534 

immediately deproteinized (solid line) and in 2/ a non-cooled heparin tube (not deproteinized before

535 

storage, dotted line). The carnosine peak at 15.6 minutes has disappeared and beta-alanine (12.1

536 

min), taurine (13.2 min) and the peak at 14.2 min (partially histidine) were higher in heparin versus

537 

EDTA plasma. 

538 

Figure 2

539 

The activity of plasma carnosinase enzyme is dependent on the amount of plasma carnosinase

540 

protein content as there is a strong positive correlation between CN1 activity and protein content (p

541 

= 0.004, r = 0.58, R² = 0.34).

542 

Figure 3

543 

Time course of plasma carnosine of the 8 responders (individual values, mean = solid line) up to 2h

544 

after oral administration of 60mg/kg BW carnosine. The increase reached a peak (range: 30.7 –

545 

195.0 µM) at 20 - 60 minutes and rapidly declined within 1-2 hours.

546 

 

547 

Figure 4

548 

Boxplot, representing the minimum/maximum values, lower/upper quartile and median of CN1

549 

protein content (A) and CN1 activity (B) of the non-responders compared to responders. The

550 

plasma carnosinase protein content (A) and activity (B) was significantly lower in the subjects

551 

characterized with supplementation-induced carnosinemia after carnosine supplementation

552 

(60mg/kg BW) compared to the non-responders group (p < 0.001 and p = 0.007, respectively; ° =

553 

outlier).

554 

Figure 5

555 

Urinary carnosine and beta-alanine: A/ The amount of urinary carnosine (black bars, expressed as

556 

% of ingested carnosine) is 2.6-fold higher (p = 0.006) in the responders group while there is no

557 

difference in urinary beta-alanine (white bars) between the responders and non-responders

558 

(respectively 1.6 ± 0.9 vs. 1.4 ± 1.3, p > 0.05). B,C/ Relationship between CN1 activity (B, p =

559 

0.001, r = -0.64, R² = 0.41) / CN1 protein content (C, p < 0.001, r = -0.66, R² = 0.44) and urinary

560 

carnosine (% of ingested dose) during the 4h after carnosine supplementation (60mg/kg BW).

561 

Figure 6

562 

The mean (± SD) area under the curve of plasma beta-alanine (µM) is significantly higher in the

563 

responders (dotted line) versus the non-responders group (solid line, p = 0.005, +30%).

564 

Figure 7

565 

Proposed pathophysiological mechanism linking carnosine and CNDP1 genotype to diabetic

566 

complications. * denotes effects that are now supported by the findings of the current study in

567 

humans. 

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.