Volume 8, Issue 2 (May 2023)                   JNFS 2023, 8(2): 283-297 | Back to browse issues page


XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Talenezhad N, Akhlaghi M, Raeisi‐Dehkordi H, Zarei S, Mozaffari-Khosravi H. The Effect of L-carnitine Supplementation on Liver Function, Folate and Vitamin B12 Levels in Patients with Type 2 Diabetes Mellitus: A Randomized, Double-Blind, Clinical Trial. JNFS 2023; 8 (2) :283-297
URL: http://jnfs.ssu.ac.ir/article-1-746-en.html
Department of Clinical Biochemistry,Rafsanjan University of Medical Sciences,Rafsanjan, Iran
Full-Text [PDF 598 kb]   (136 Downloads)     |   Abstract (HTML)  (624 Views)
Full-Text:   (79 Views)

The Effect of L-carnitine Supplementation on Liver Function, Folate and Vitamin B12 Levels in Patients with Type 2 Diabetes Mellitus: A Randomized, Double-Blind, Clinical Trial

Nasir Talenezhad; MSc1, 2, Milad Akhlaghi; MSc4, Hamidreza RaeisiDehkordi; MSc1, 3,
Sadegh Zarei; PhD5, Hassan Mozaffari-Khosravi; PhD*1, 6

1 Department of  Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran;2 Behbahan Faculty of Medical Sciences, Behbahan, Iran; 3 Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland; 4 Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; 5 Department of Clinical Biochemistry, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; 6 Yazd Diabetic Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
ARTICLE INFO ABSTRACT
ORIGINAL ARTICLE
Background: Carnitine is necessary for allowing the long-chain fatty acids to pass the inner mitochondrial membrane to induce β -oxidation. Lack of carnitine and abnormalities of mitochondria play an important role in forming fatty deposition in the liver, and hence, developing steatohepatitis. Carnitine and acylcarnitine identified in human erythrocytes and intra-erythrocyte acetylcarnitine have a significant relationship with the plasma levels. Methods: The present study was conducted to investigate the possible effects of L-carnitine on liver function, folate and vitamin B12 levels in patients with type 2 diabetes mellitus (T2DM). In this study, 70 patients with T2DM were randomly assigned to either a L-carnitine (CG) and a placebo group (PG). For 12 weeks, the first group received 1000 mg/d oral L-carnitine, whereas the second group received 1000 mg/day wheat starch as placebo. The alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), gamma-glutamyl transferase (GGT), folate, Vitamin B12, complete blood count (CBC) including blood cells and indicators related to anemia were assessed at baseline. Results: 64 patients managed to complete the study (32 in each group). The results indicated that consumption of L-carnitine compared with placebo had no significant effect on liver enzymes, folate, vitamin B12, and CBC with differential. Conclusions:  Daily intake of 1000 mg L-carnitine for 12 weeks had no effect on liver function and anemia indicators including CBC, folate, and vitamins B12.

Keywords: Carnitine; Diabetes mellitus; Liver; Vitamins; Clinical trial
Article history:
Received: 21 Oct 2022
Revised: 11 Dec 2022
Accepted: 11 Dec 2022
*Corresponding author
mozaffari.kh@gmail.com
Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Alem Square, Shohaday Gomname Blvd, Yazd, Iran.

Postal code: 8915173160
Tel: +98 9131531467

Introduction
Diabetes mellitus (DM) is a common chronic metabolic disorder with an increasing prevalence. It has become a major public health concern, leading to serious disorders in the body (Aekplakorn et al., 2011, Meo, 2009). In 2019, the International Diabetes Federation (IDF) estimated that approximately 463 million people aged 20-79 worldwide suffer from DM. This will increase to 578 million in 2030 and 700 in 2045 with type 2 diabetes (T2DM) accounting for about 90% of all cases. In 2019, the number of people with diabetes in Iran  at the age range of 20-79 was reported to be 5.4 million with a prevalence of 9.4%(International Diabetes Federation (IDF), 2019).
 T2DM is associated with other chronic diseases and is influenced by genetic factors, obesity, poor diet and physical inactivity. This ultimately leads to anatomical, structural and functional changes in several important organs of the body (Galicia-Garcia et al., 2020, Sami et al., 2017). Further progression of the disease is associated with cardiovascular complications (Kayama et al., 2015), nephropathy, retinopathy and neuropathy (Vinik et al., 2013). The prevalence of non-alcoholic fatty liver disease (NAFLD) in patients with T2DM was found to be high  (68.1% of general population), and diabetes, obesity, and hyperlipidemia are considered important risk factors for NAFLD (Mohamed et al., 2022). There is also evidence that liver enzymes and NAFLD act as useful biomarkers for diabetes (Atiba et al., 2013). Moreover, alanine aminotransferase/ aspartate aminotransferase (ALT/AST) ratio may be one of the best markers for insulin resistance (Zhao et al., 2017). Higher levels of AST, gamma-glutamyl transferase (GGT), and ALT have also been shown to be associated with increased HOMA-IR index and the risk of T2DM (Sheng et al., 2018, Shibabaw et al., 2019). Oxidative stress and chronic inflammation are among the disorders that can lead to progression of the disease, and higher level of serum GGT was independently associated with increased load of subclinical inflammation across metabolic states (Ali et al., 2016, Turgut and Tandogan, 2011).
Plasma levels of vitamin B12 and folate have been less reported in patients with T2DM compared to non-diabetic patients (Ebesunun and Obajobi, 2012). Vitamin B12 deficiency contributes to accumulation of methyl-malonic acid. This causes oxidative stress, impairs mitochondrial function, disrupts cellular energy metabolism, triggers cell death, and ultimately produces more inflammatory cytokines such as alpha tumor necrotizing factor (TNF-α) (Chen et al., 2008, Liu et al., 2022). In addition, these deficiencies trigger disruption of methionine synthesis and homocysteine (Hcy) ​​production as a potential risk factor for cardiovascular disease associated with oxidative stress in diabetics (Al-Maskari et al., 2012, Kolling et al., 2011). Vitamin B12 and folate deficiency cause anemia. Consequently, the serum level of these vitamins has been properly determined and defined to prevent anemia (Fenech, 2012). Evidence also suggests that deficiency of vitamin B12 is associated with acyl carnitine accumulation, indicating high intracellular levels of acyl-CoA. Moreover the deficiency leads to accumulation of propionyl carnitine (Brass and Stabler, 1988, Sarafoglou et al., 2011). There is a hypothesis that propionyl carnitine is a major product of propionate metabolism regarding vitamin B12 deficiency and the optimal rate of propionyl carnitine formation depends on availability of external carnitine. This has been investigated and proven in studies by Brass (Brass and Ruff, 1989, Brass and Stabler, 1988).
Inadequate exogenous intake, malabsorption and lack of endogenous hepatic synthesis of L-carnitine can lead to its deficiency in the body, which is accompanied by liver cirrhosis (Cave et al., 2008). L-Carnitine affects intracellular reactions by limiting the oxidative stress process, which ultimately reduces response of inflammatory mediators and improves the outcomes of nonalcoholic steatohepatitis (NASH) (Romano et al., 2008). L-carnitine supplementation improves body composition and levels of liver enzymes, having a direct effect on liver function (Pirmadah et al., 2019, Talenezhad et al., 2020b). In addition, it plays a key role in elimination of short and medium chain fatty acids, regulation of CoA/CoA acyl ratio within mitochondria, transmission and oxidation of long chain fatty acids, glucose metabolism, detoxification of toxic metabolites and cell wall stabilization (di San Filippo et al., 2008, Malaguarnera et al., 2010). The supplementary effect of L-carnitine on blood cells has been investigated in many studies (Strasser et al., 2007, Sweeney and Arduini, 2004). The role of L-carnitine in the management of anemia may improve and maintain red blood cell survival by augmenting the stability of erythrocyte membranes (Arduini et al., 1993). A study conducted by Bonomini et al. revealed that L-carnitine-enriched peritoneal dialysis solutions have demonstrated nti-anemia action due to its effects on erythropoiesis, and positive effects on the longevity and deformability of erythrocytes (Bonomini et al., 2019). Moreover, in a clinical trial study, it was identified that treatment with L-carnitine during a period of 24 months may be effective for reducing muscle cramping and improving hemoglobin levels in dialysis patients (Kuwasawa-Iwasaki et al., 2020). In some studies, the beneficial effects of L-carnitine on upgrading the survival time of red blood cells in treated patients could not be observed (Kletzmayr et al., 1999). L-Carnitine stabilizes erythrocyte membranes by increasing Hct and improving cellular sodium, potassium and adenosine triphosphate through reducing their plasma inhibition (Donatelli et al., 1987, Labonia et al., 1987). It also protects red blood cells against toxicity above the level of intracellular calcium (Agroyannis et al., 2002).
To evaluate the clinical effect of L-carnitine in the treatment of complications of T2DM, especially elevated blood Hcy levels, the researchers conducted a large randomized, double-blind, and placebo-controlled study (Talenezhad et al., 2020a, Talenezhad et al., 2020c). Considering the role of folate and vitamin B12 in Hcy ​​metabolism, their deficiency in diabetic patients can be associated with increased Hcy ​​levels (Fotiou et al., 2014), as well as the prevalence NAFLD (Raza et al., 2021). As part of a larger study, it was decided to investigate the effect of L-carnitine supplementation on patients' liver function and anemia indices including vitamins B12 and folate.
Materials and Methods
Study design and participants: This was a randomized double-blind, clinical trial. Seventy adults with T2DM in the age range of 41–75 years were recruited from Yazd Diabetic Research Center. Patients met the following inclusion criteria:  aging 30 and above, having a body mass index (BMI) of greater than 25 kg/m2, and HbA1c of lower than 10%, having no clinical diagnosis of chronic gastrointestinal, liver or  kidney disease, untreated hypothyroidism, hypertension, systemic infection, gangrene, having a history of myocardial infarction, unstable angina, severe arrhythmia, and cardiac surgery in the last 3 or 6 months,  no current use of antibiotics or other prescribed medicine, no use of L-carnitine supplements and group B vitamins over the last 3 months and antioxidant intake (vitamins A, D, C and E) within 1 month of the initiation of the experimental periods, no specific dietary practice for weight loss in the past year. Patients were excluded if they could not comply with or follow the study protocol. Those who showed no desire to continue with the trial for any reason were not included.
The patients were randomly allocated to two groups of thirty-five; the first group received L-carnitine 1000 mg/d (LG) divided into 2 equal doses of one 500 mg tablet after breakfast and during dinner for 12 weeks (L-carnitine, Karen Pharmaceutical and Nutrilife Co., Yazd, Iran). The second group received placebo (PG) with the same dose, shape, color and duration (placebo, Karen Pharmaceutical and Nutrilife Co. Yazd, Iran).
Allocation concealment and compliance with the intervention: With an allocation ratio of 1:1, patients were randomly assigned into two groups through a computer-generated randomization sequence. Researchers and participants involved in data collection and analysis were blinded to the randomization and treatment allocation until the final analyses were completed. The matching placebo capsule was identical to L-carnitine capsule in terms of appearance, taste, and smell. L-carnitine and placebo compliance was assessed by counting the number of remaining capsules at their subsequent visit.
Measurements: Once the patients had fasted for 10-12 hours, 7 to 10 ml of venous blood samples were obtained, and the serum was separated from clotted blood by centrifugation and transferred into three serum separator tubes. Serum samples were stored at −70 °C until the end of the study period. Then, ALT, AST, ALP, GGT were measured in fasting serum using biochemical kits from Pars Azmon (Tehran, Iran). In addition, folate (ng/ml) and vitamin B12 (pg/l) were determined using the ELISA kits (Monobind, USA). Erythrocyte count (RBC, 106/µl), leukocyte count (WBC, 103/µl), hemoglobin content (Hb, g/dl), Hct (%), mean corpuscular volume (MCV, fl), mean corpuscular hemoglobin (MCH, pg), mean corpuscular hemoglobin concentration (MCHC, g/dl) and platelet count (103/µl) were assessed with the routine clinical chemistry procedures using commercial kit. Patients were asked to maintain their usual diet and level of physical activity during the study period. All the subjects completed the 3-day food records and physical activity records (in a week's time) in weeks 0 and 12 of the intervention. Daily macro- and micro-nutrient dietary intakes were also analyzed by nutritionist IV software. The international physical activity questionnaire (IPAQ) was used to estimate physical activity based on the metabolic equivalent of task (MET) score.
Ethical considerations: The ethics committee affiliated with Yazd Shahid Sadoughi University
of Medical Sciences (Ethics code: IR.SSU.SPH.REC.1397.067) approved the study protocol. Moreover, this trial was registered in the Iranian Clinical Trial Registration Center (www.irct.ir) with the code IRCT2017100936681N. Informed consent was obtained from the participants before initiation of the study.

Data analysis: Data analysis was carried out using SPSS-24 (version 24; SPSS Inc., Chicago Illinois, USA). The results indicated in the manuscript and tables are reported as mean ± standard error (SE) or 95% confidence intervals (CIs). A P-value of < 0.05 was also considered statistically significant. Kolmogorov-Smirnov and Shapiro-Wilk tests followed by test graphs and numbers related to Skewness and Kurtosis were used to examine the normal distribution of the quantitative data. The paired t-test was also employed to evaluate within-group differences (before and after intervention) regarding normal data. To identify any differences in the baseline variables and at the end of the study and to compare the mean changes in the intervention group's independent samples, student t-test was deployed. Moreover, the ANCOVA test was carried out to detect any differences between the intervention groups at the end of the experiment by adjusting baseline values.
Results
Baseline characteristics of participants: 70 participants were randomly assigned to the intervention group, of whom 64 completed the trial. Six participants withdrew during the study. In the LG, two participants dropped out because of their lack of interest to continue and use of antioxidant supplementation (n=2). Moreover, one subject dropped out because of concern for gastrointestinal side effects following supplementation with L-carnitine (n=1). In the PG, one patient reported renal complications following placebo (n=1) and two patients were excluded for lumbar disc surgery and loss of interest (Figure 1).  A summary of the baseline characteristics of all the participants is provided in Table 1. The mean age of the total sample was 55.67 ± 0.99 ranging from 41 to 75 in each treatment group. 61% were women, 73% of whom were housewife. There were no significant differences between groups in terms of all baseline characteristics (P > 0.05). The patients in LG and PG were compared for other diseases, for which no significant difference were identified between the groups (P = 0.606).
Physical activity, energy and nutrient intake: Energy intake, macronutrients and some micronutrients (based on a 3-day diet) as well as the level of physical activity (in a period of one week) are presented in Table 2. After adjusting for baseline values, the comparative analysis demonstrated no statistically significant difference regarding some of the micronutrient and macronutrient intake, energy and level of physical activity between LG and PG (P > 0.05).
Effect of L-carnitine on blood and serum parameters: After 12 weeks of treatment with L-carnitine, independent samples t-test failed to detect any statistically significant difference in terms of changes in serum levels of vitamin B12, folate and liver biomarkers compared to the PG. The changes also did not appear to be considerable for CBC, which included blood cells and indicators related to anemia. In the final analysis, to achieve a more accurate and valuable P-value and identify the differences between the treatment groups, results of ANCOVA test were reported with adjustment for baseline values of the target variables. The results of ANCOVA test did not show any statistical significance for serum levels of ALT (P=0.302), AST (P=0.061), ALP (P=0.546), GGT (P=0.312), vitamin B9 (P=0.343), vitamin B12 (P=0.606) as well as RBC (P=0.050), WBC (P=0.666), Hb (P=0.355), Hct (P=0.140), MCV (P=0.523), MCH (P=0.928), MCHC (P=0.481) and platelet count (P=0.832).

Table 1. baseline characteristics of study participants.
Variables/ Groups L-carnitine group (n=35) Placebo group (n=35) P-valueb
Age (year) 56.40±1.49 54.90±1.32 0.469
Height (cm) 162.89±1.95 160.89±1.47 0.418
Weight (kg) 76.03±2.03 77.22±1.99 0.678
Body mass index (kg/m2) 28.63±0.56 29.54±0.74 0.333
Hemoglobin A1c (%) 6.90±0.14 7.00±0.16 0.642
Duration of diabetes (year) 4.91±0.64 4.48±0.80 0.678
Physical activity (Met/min/day) 1863.69±49.32 1811.66±44.14 0.435
Sex
   Male 16(45.7) 11(31.4) 0.220
   Female 19(54.3) 24(68.6)
Status of vitamin B12
   Normal 5(14.3) 2(5.7) 0.232
   Deficiency 30(85.7) 33(94.3)
Status of vitamin B9
   Normal 2(5.7) 1(2.9) 0.550
   Deficiency 33(94.3) 34(97.1)
Job
   Housewife 17(48.6) 20(57.1) 0.350
   Retired 6(17.10) 9(25.7)
   Free job 8(22.9) 3(8.6)
   Employee 4(11.4) 3(8.6)
Education
   Illiterate 2(5.7) 1(2.9) 0.661
   Elementary 11(31.4) 14(40)
   Diploma 16(46.6) 13(37.1)
   Bachelor 3(8.6) 6(17.1)
   Above bachelor 3(8.6) 1(2.9)
Drug used
   Metformin 24(72.7) 21(67.7) 0.663
   Acarbose 11(33.3) 12(40.0) 0.583
   Diabetes 9(27.3) 13(41.9) 0.217
   Ziptin 7(21.2) 8(26.7) 0.612
   Zipmet 5(16.1) 10(32.3) 0.138
   Diamicron 9(27.30) 4(12.9) 0.153
   Aspirin 8(25.0) 9(30.0) 0.659
   Anti- hypertensive 15(45.5) 14(45.2) 0.981
   Statins 18(55.5) 22(71.0) 0.175
Other disease
   Hypothyroidism 4(11.4) 2(5.7) 0.606
   Anemia 0 2(5.7)
   Waist and Neck Disc 1(2.9) 1(2.9)
   Slight fatty Liver 1(2.9) 1(2.9)
   Rheumatism 0 2(5.7)
   Pemphigus 1(2.9) 0
a:Mean ± SE; b: P-value resulted from independent t-test for quantitative and Chi-square for qualitative variables between the two groups.

Table 2.  Daily nutrient intake based on a 3-day diet  in patients with T2DM.
Variables L-carnitine  group (n=35) Placebo group (n=35) P-valuea
Energy (kcal/day)
   Before 1978.65±53.93b 1957.60±46.85 0.144
   After 1963.60±52.14 1902.45±47.67 0.122
   Change -15.05±22.93 -55.15±30.50 0.248
   P-valuec 0.516 0.080
Carbohyderate (g/day)
   Before 270.20±7.73 258.83±9.81 0.075
   After 269.28±8.49 259.92±9.10 0.165
   Change -0.92±5.57 1.08±6.39 0.926
   P-value 0.874 0.866
Protein (g/day)
   Before 79.11±2.93 76.82±2.44 0.205
   After 79.36±2.57 74.47±2.47 0.190
   Change 0.25±2.45 -2.35±2.43 0.220
   P-value 0.919 0.340
Total Fat (g/day)
   Before 67.92±2.88 70.76±2.48 0.499
   After 67.38±3.22 65.21±2.63 0.804
   Change -0.53±2.15 -5.55±2.71 0.218
   P-value 0.804 0.049
Cholestrol (mg/day)
   Before 262.04±22.07 312.15±24.49 0.147
   After 263.29±24.52 249.48±21.87 0.680
   Change 1.24±26.67 -62.67±29.13 0.395
   P-value 0.963 0.039
Total dietary fiber (g/day)
   Before 16.62±0.98 16.81±0.88 0.503
   After 15.98±0.88 17.29±0.86 0.290
   Change -0.63±1.09 0.48±0.81 0.276
   P-value 0.564 0.559
Selenium (mg/day)
   Before 0.06±0.004 0.06±0.004 0.720
   After 0.06±0.004 0.06±0.003 0.384
   Change 0.0017±0.004 0.0016±0.003 0.320
   P-value 0.700 0.691
Zinc (mg/day)
  Before 8.15±0.35 8.29±0.34 0.905
  After 7.9±0.32 7.88±0.40 0.613
  Change -0.24±0.32 -0.40±0.39 0.836
   P-value 0.505 0.309
Vitamin E (mg/day)
   Before 10.70±1.12 10.33±1.25 0.893
   After 9.77±1.47 9.56±1.11 0.718
  Change -0.93±1.48 -0.76±1.00 0.995
   P-value 0.535 0.451
Vitamin A (RE)
   Before 4.07±0.34 4.37±0.32 0.315
   After 4.32±0.32 3.87±0.22 0.220
  Change 0.25±0.40 -0.49±0.34 0.185
   P-value 0.537 0.157
Vitamin C (mg/day)
   Before 63.10±6.97 77.63±8.11 0.282
   After 62.90±6.19 67.47±7.61 0.790
  Change -0.19±8.61 -10.15±6.11 0.825
   P-value 0.982 0.106
Physical activity (Met/min/day)
   Before 1863.69±49.32 1811.66±44.14 0.511
   After 1858.77±56.11 1825.43±54.36 0.664
  Change -4.91±29.55 13.77±22.20 0.583
   P-value 0.869 0.539
a: Obtained from ANCOVA, adjusted for baseline values; b: Mean ± SE; c:  Paired t-test.
 
Table 3. Within and between comparison mean of hematologic parameters in two groups.1
Variables L-carnitine group (n=35) Placebo group (n=35) P-valuea
Alanine transaminase (u/l)
   Before 23.34±1.51b 21.36±1.25 0.322
   After 24.44±1.62 21.40±1.33 0.154
   Change 1.09±1.45 0.03±1.14 0.302
   P-valuec 0.456 0.976
Aspartate transaminase (u/l)
   Before 19.01±0.87 18.76±0.98 0.849
   After 20.15±1.13 17.87±0.69 0.100
   Change 1.13±0.92 -0.88±0.77 0.061
   P-value 0.229 0.262
Alkaline phosphatase (u/l)
   Before 205.86±8.05 202.54±9.91 0.796
   After 199.86±7.04 201.29±9.25 0.903
   Change -6±4.82 -1.25±5.26 0.546
   P-value 0.223 0.813
Gamma-glutamyltransferase (u/l)
   Before 19.07±1.38 20.93±1.66 0.395
   After 18.69±1.21 21.43±1.77 0.209
   Change -0.38±0.81 0.50±83 0.312
   P-value 0.645 0.546
Vitamin B12 (pg/l)
   Before 340.90±27.66 321.33±18.52 0.561
   After 328.89±2419 306.84±22.60 0.508
   Change -12.01±28.25 -14.49±22.96 0.643
   P-value 0.673 0.532
Vitamin B9 (ng/ml )
   Before 12.48±1.03 13.30±1.07 0.586
   After 13.86±0.84 13.08±0.96 0.544
   Change 1.38±1.26 -0.22±0.72 0.343
   P-value 0.283 0.764
Homocysteine (µmol/l)
   Before 14.43±0.79 14.81±0.96 0.764
   After 15.30±0.79 14.23±0.86 0.336
   Change 0.87±0.75 -0.57±0.75 0.170
   P-value 0.252 0.452
White blood cells (103/µl)
   Before 7.01±0.27 6.95±0.27 0.865
   After 6.93±0.20 6.79±0.24 0.680
   Change -0.08±0.18 0.15±0.17 0.666
   P-value 0.648 0.384
Red blood cells (106/µl (
   Before 4.82±0.09 4.80±0.08 0.853
   After 4.82±0.10 4.67±0.07 0.220
   Change 0.0±0.04 -0.13±0.05 0.048
   P-value 0.982 0.023
Hemoglobin(g/dl)
   Before 14.14±0.22 13.53±0.28 0.102
   After 14.34±0.23 13.63±0.28 0.058
   Change 0.20±0.13 0.10±0.15 0.355
   P-value 0.136 0.511
Hematocrit (%)
   Before 40.77±0.64 39.90±0.75 0.387
   After 39.51±0.63 38.01±0.67 0.113
   Change -1.25±0.36 -1.88±0.48 0.140
   P-value 0.002 <0.001
Mean corpuscular volume ( fl)
   Before 85.40±0.81 84.67±0.94 0.564
   After 83.99±0.81 82.97±0.88 0.408
   Change -1.41±0.53 -1.69±0.41 0.523
   P-value 0.012 <0.001
Mean corpuscular hemoglobin (pg (
   Before 29.80±0.39 29.24±0.35 0.293
   After 30.56±.35 30.08±0.40 0.380
   Change 0.75±0.29 0.84±0.20 0.928
   P-value 0.017 <0.001
Mean corpuscular hemoglobin concentration )g/dl(
   Before 34.70±0.35 34.12±0.28 0.190
   After 36.25±0.20 35.83±0.27 0.215
   Change 1.54±0.32 1.71±0.31 0.481
   P-value <0.001 <0.001
Platelet (103/µl (
   Before 225.56±8.54 252.64±9.46 0.038
   After 219.34±7.74 236.38±8.18 0.172
   Change -6.21±6.78 -16.26±4.64 0.832
   P-value 0.366 0.001
a: Obtained from ANCOVA, adjusted for baseline values; b: Mean ± SE; c:  Paired t-test.
Discussion
L-carnitine administration for 12 weeks failed to significantly affect liver enzymes (ALT, AST, ALP, and GGT), folate and vitamin B12 in patients with T2DM. In addition, the results were not significant for CBC.
To date, no clinical trial study has investigated the effect of L-carnitine on folate and vitamin B12 levels in various diseases. Moreover, few studies have considered the effect of this supplement on liver function in patients with T2DM. There is still insufficient evidence to investigate the effect of L-carnitine on anemia-related factors in animal and human models. In the current trial, the researchers did not observe any significant reduction in circulating liver enzymes' levels following supplementation with l-carnitine in patients with T2DM.
This is consistent with several clinical trials which indicated L-carnitine had no significant effect on the level of serum liver enzyme (An et al., 2016, Hassan et al., 2015, Higuchi et al., 2014). Hassan et al. found no significant changes in the level of liver enzyme among intermediate-stage hepatocellular carcinoma (HCC) patients after supplementation with 600 mg of L-carnitine over 12 weeks (Hassan et al., 2015). Another clinical trial study conducted in 2016 identified that oral treatment of L-carnitine in patients with hypothyroidism on levothyroxine treatment failed to significantly impact the serum concentration of liver enzyme (An et al., 2016). However, a number of trials provided evidence that supplementation with L-carnitine might be effective in reducing these enzymes (Alavinejad et al., 2016, Malaguarnera et al., 2011, Somi et al., 2014). Some of these studies evaluated the effect of L-carnitine on liver function and treatment of NAFLD (Alavinejad et al., 2016, Somi et al., 2014) and other liver diseases such as NASH (Malaguarnera et al., 2010) and chronic hepatitis C (Malaguarnera et al., 2011). Some desired results have been obtained from the impact of L-carnitine on elevating liver enzymes. Therefore, evidence confirms that L-carnitine can improve abnormally elevated levels of liver enzymes in these patients. However, evidence which runs counter to the study results is based on the fact that diseases that impair liver function can trigger accumulation of toxins in the body and increase production of reactive oxygen species (ROS) (Muriel and Gordillo, 2016). Then, oxidative stress produced by oxidants contributes to disorders of mitochondrial β oxidation (Santos et al., 2013). Deactivation of beta oxidation pathway or carnitine deficiency induce accumulation of fatty acids within the hepatocytes and the progression of the NAFLD (Rolo et al., 2012, Rudman et al., 1977). Due to the fact that L-carnitine is a key component in beta oxidation of long chain fatty acids in mitochondria and also bears antioxidant and anti-radical properties, it is expected to reduce liver enzymes, especially in liver diseases (Indiveri et al., 2011). This study, however, examined liver function in patients with diabetes, not liver disease. A meta-analysis focused on serum levels of enzymes mainly produced by liver, indicated that L-carnitine can have a significant effect on these enzymes, and finally, positively affect liver function especially in patients with liver diseases. Another finding of this study revealed that L-carnitine can be more effective in healthy people or patients who receive intervention doses of higher than 2000 mg/day (Pirmadah et al., 2019). Therefore, one of the reasons for not achieving a significant result in the present study could be related to the low dose of L-carnitine intervention (1000 mg/day).
The intake of 1000 mg/d of L-carnitine does not affect folate serum concentrations, vitamin B12, and CBC including blood cells and indicators related to anemia. It should be noted that the aim of investigating the effect of L-carnitine supplementation on these vitamins was to determine and control their relationship with anemia indicators in the present study and Hcy ​​research (Talenezhad et al., 2020c). There has been no clinical trial on the effect of L-carnitine supplementation regarding these vitamins. Monitoring of vitamins was of great importance in this study. This is because the plasma levels of vitamin B12 and folate was low in patients with T2DM, which was attributed to either the disease itself or anti-diabetic drugs such as metformin (Kim et al., 2019). In addition, these vitamins played an important role regarding anemia. Several studies, however, examined the effect of L-carnitine supplementation on other blood markers associated with anemia.  Kudoh et al. reported that long-term consumption of LC did not have beneficial effect on patients with renal anemia (Kudoh et al., 2014). In another RCT study by Maruyama (Maruyama et al., 2017), no significant difference was indicated between the 2 groups during the study period. Results suggested that levocarnitine consumption can decrease the dose of erythropoiesis-stimulating agent (ESA) required in patients with renal anemia regarding hemodialysis, and amend response to ESA therapy (Maruyama et al., 2017). Emami Naini reported that oral L-carnitine supplementation may insignificantly increase hemoglobin, and subsequently, reduce needed erythropoietin dose. This reveals the positive effect of oral L-carnitine on anemia. This study which is in line with ours could not detect a significant increase in hemoglobin levels; yet, it improved anemia (Emami Naini et al., 2012). Orasan et al. indicated that supplementation with 500mg/day L-carnitine during 3 months had no effect on anemia in hemodialysis (HD) patients (Orasan et al., 2011). Contrary to the results of this study, a clinical trial conducted in 2005 demonstrated that a 4-month period of L-carnitine supplementation may improve anemia with a significant increase in the Hct and Hb levels, and reduce the weekly required dose of the rHuEPO (Kadiroglu et al., 2005). Additionally, in 2002, a trial examined the effect of 2000 mg oral L-carnitine supplementation on Hct and Hb levels in patients suffering from end stage renal disease (ESRD) regarding continuous ambulatory peritoneal dialysis (CAPD). It revealed that consumption of L-carnitine can increase Hct and Hb levels and reduce the patient’s erythropoietin (rHuEpo) dose per week (Sotirakopoulos et al., 2002). The dose of L-carnitine used was twice, compared with this study. Furthermore, in another study, Trovato detected a rise in hematocrit in HD patients treated with oral L-carnitine for 12 months. Intervention in Trovato's study took much longer than this study (Trovato et al., 1982). Mechanisms which can explain the significant effects of L-carnitine on indicators associated with anemia include augmenting the survival rate of red blood cells. This was done by increasing Na–K pump activity of the erythrocyte membrane, increasing their membrane stability, effecting some enzymes and metabolic functions in the erythrocytes (Arduini et al., 1993, Labonia et al., 1987), raising red blood cell osmotic resistance (Evangeliou and Vlassopoulos, 2003), and protecting them from high toxicity of intracellular calcium (Agroyannis et al., 2002).
This study had several limitations. First, it was performed on diabetic patients without NAFLD or anemia. Moreover, the associated blood parameters were normal, and there was no deficiency. In other words, the studied indicators were not deficient or abnormal, which affected the results. Second, it related to the lack of measurement of carnitine, other parameters of NAFLD, and anemia at the baseline and at the end of the trial. And third, there was a low dose of intervention. If a higher dose was used, more favorable results might have been obtained.
Conclusion
Carnitine consumption fails to reduce the level of liver enzyme, folate, vitamin B12, and also CBC including blood cells and indicators related to anemia in T2DM. This is done without additional interventions like physical activity or low-caloric diets. Further high-quality and large RCT are needed to clarify the actual effect of L-carnitine intake on parameters of liver and anemia in the patients with T2DM.
Acknowledgments
The authors would like to thank all the participants in the study.
Conflicts of interest
The authors declared no conflict of interest.
Funding
This study was funded by School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
Authors' contributions
Mozaffari-Khosravi H involve in designing, data analyzing, supervising and writing the manuscript.  Talenezhad N involved in designing, data collecting, data analyzing and writings the manuscript. Akhlaghi M, RaeisiDehkordi H and Zarei S participated to laboratory measurements and analyzing the data. All authors read the manuscript and approved it for publishing.
References
Aekplakorn W, et al. 2011. Prevalence and management of diabetes and metabolic risk factors in Thai adults: the Thai National Health Examination Survey IV, 2009. Diabetes care. 34 (9): 1980-1985.
Agroyannis B, Paraskevopoulos A, Fourtounas C, Tzanatos H & Tsomi A 2002. Effect of L-carnitine on the calcium content of erythrocytes of uremic patients on hemodialysis. Dialysis & transplantation. 31 (2): 106-127.
Al-Maskari MY, Waly MI, Ali A, Al-Shuaibi YS & Ouhtit A 2012. Folate and vitamin B12 deficiency and hyperhomocysteinemia promote oxidative stress in adult type 2 diabetes. Nutrition. 28 (7-8): e23-e26.
Alavinejad P, et al. 2016. Evaluation of L-carnitine efficacy in the treatment of non-alcoholic fatty liver disease among diabetic patients: a randomized double blind pilot study. Journal of gastroenterology and hepatology research. 5 (5): 2191-2195.
Ali SS, et al. 2016. Elevated gamma-glutamyl transferase is associated with subclinical inflammation independent of cardiometabolic risk factors in an asymptomatic population: a cross-sectional study. Nutrition & metabolism. 13 (1): 1-7.
An JH, et al. 2016. L-carnitine supplementation for the management of fatigue in patients with hypothyroidism on levothyroxine treatment: a randomized, double-blind, placebo-controlled trial. Endocrine journal. EJ16-0109.
Arduini A, et al. 1993. Effects of L-carnitine and its acetate and propionate esters on the molecular dynamics of human erythrocyte membrane. Biochimica et biophysica acta (BBA)-biomembranes. 1146 (2): 229-235.
Atiba AS, et al. 2013. Liver enzymes and lipid profile among type 2 diabetic patients in Osogbo, Nigeria. Greener journal of medical sciences. 3 (5): 174-178.
Bonomini M, Di Liberato L, Zammit V & Arduini A 2019. Current opinion on usage of L-carnitine in end-stage renal disease patients on peritoneal dialysis. Molecules. 24 (19): 3449.
Brass EP & Ruff LJ 1989. Effect of carnitine on propionate metabolism in the vitamin B-12—deficient rat. Journal of nutrition. 119 (8): 1196-1202.
Brass EP & Stabler SP 1988. Carnitine metabolism in the vitamin B-12-deficient rat. Biochemical journal. 255 (1): 153-159.
Cave MC, et al. 2008. Obesity, inflammation, and the potential application of pharmaconutrition. Nutrition in clinical practice. 23 (1): 16-34.
Chen X, et al. 2008. Role of reactive oxygen species in tumor necrosis factor-alpha induced endothelial dysfunction. Current hypertension reviews. 4 (4): 245-255.
di San Filippo CA, Taylor MR, Mestroni L, Botto LD & Longo N 2008. Cardiomyopathy and carnitine deficiency. Molecular genetics and metabolism. 94 (2): 162-166.
Donatelli M, Terrizzi C, Zummo G, Russo V & Bucalo M 1987. Effects of L-carnitine on chronic anemia and erythrocyte adenosine triphosphate concentration in hemodialyzed patients. Current therapeutic research. 41 (5): 620-624.
Ebesunun MO & Obajobi EO 2012. Elevated plasma homocysteine in type 2 diabetes mellitus: a risk factor for cardiovascular diseases. Pan African medical journal. 12 (1).
Emami Naini A, et al. 2012. Effects of oral L-carnitine supplementation on lipid Profile, anemia, and quality of life in chronic renal disease patients under hemodialysis: a randomized, double-blinded, placebo-controlled trial. Journal of nutrition and metabolism. 2012.
Evangeliou A & Vlassopoulos D 2003. Carnitine metabolism and deficit-when supplementation is necessary? Current pharmaceutical biotechnology. 4 (3): 211-219.
Fenech M 2012. Folate (vitamin B9) and vitamin B12 and their function in the maintenance of nuclear and mitochondrial genome integrity. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis. 733 (1-2): 21-33.
Fotiou P, et al. 2014. Vitamin status as a determinant of serum homocysteine concentration in type 2 diabetic retinopathy. Journal of diabetes research. 2014.
Galicia-Garcia U, et al. 2020. Pathophysiology of type 2 diabetes mellitus. International journal of molecular sciences. 21 (17): 6275.
Hassan A, et al. 2015. Effects of oral L-carnitine on liver functions after transarterial chemoembolization in intermediate-stage HCC patients. Mediators of inflammation. 2015.
Higuchi T, et al. 2014. Effects of levocarnitine on brachial-ankle pulse wave velocity in hemodialysis patients: a randomized controlled trial. Nutrients. 6 (12): 5992-6004.
Indiveri C, et al. 2011. The mitochondrial carnitine/acylcarnitine carrier: function, structure and physiopathology. Molecular aspects of medicine. 32 (4-6): 223-233.
International Diabetes Federation (IDF) 2019. IDF diabetes atlas, https://www.diabetesatlas. org.  Kadiroglu AK, Yilmaz ME, Sit D, Kara IH & Isikoglu B 2005. The evaluation of postdialysis L-carnitine administration and its effect on weekly requiring doses of rHuEPO in hemodialysis patients. Renal failure. 27 (4): 367-372.
Kayama Y, et al. 2015. Diabetic cardiovascular disease induced by oxidative stress. International journal of molecular sciences. 16 (10): 25234-25263.
Kim J, Ahn CW, Fang S, Lee HS & Park JS 2019. Association between metformin dose and vitamin B12 deficiency in patients with type 2 diabetes. Medicine. 98 (46).
Kletzmayr J, et al. 1999. Anemia and carnitine supplementation in hemodialyzed patients. Kidney international. 55: S93-S106.
Kolling J, Scherer EB, Da Cunha AA, Da Cunha MJ & Wyse AT 2011. Homocysteine induces oxidative–nitrative stress in heart of rats: prevention by folic acid. Cardiovascular toxicology. 11 (1): 67-73.
Kudoh Y, et al. 2014. Long-term effects of oral L-carnitine supplementation on anemia in chronic hemodialysis. Cardiorenal medicine. 4 (1): 53-59.
Kuwasawa-Iwasaki M, et al. 2020. Effects of L-carnitine supplementation in patients receiving hemodialysis or peritoneal dialysis. Nutrients. 12 (11): 3371.
Labonia WD, Morelli Jr OH, Gimenez MI, Freuler PV & Morelli OH 1987. Effects of L-carnitine on sodium transport in erythrocytes from dialyzed uremic patients. Kidney international. 32 (5): 754-759.
Liu Y, et al. 2022. The Regulation and Characterization of Mitochondrial-Derived Methylmalonic Acid in Mitochondrial Dysfunction and Oxidative Stress: From Basic Research to Clinical Practice. Oxidative medicine and cellular longevity. 2022.
Malaguarnera M, et al. 2010. L-carnitine supplementation to diet: a new tool in treatment of nonalcoholic steatohepatitis—a randomized and controlled clinical trial. American journal of gastroenterology. 105 (6): 1338-1345.
Malaguarnera M, et al. 2011. L-carnitine supplementation improves hematological pattern in patients affected by HCV treated with Peg interferon-α 2b plus ribavirin. World journal of gastroenterology. 17 (39): 4414.
Maruyama T, et al. 2017. Levocarnitine injections decrease the need for erythropoiesis-stimulating agents in hemodialysis patients with renal anemia. Cardiorenal medicine. 7 (3): 188-197.Meo SA 2009. Diabetes mellitus: Health and wealth threat. International journal of diabetes mellitus. 1 (1): 42.
Mohamed AM, Isa HM, Ali MS, Dadi A & Kadhim Z 2022. Prevalence of non-alcoholic fatty liver disease among patients with diabetes mellitus attending primary health care centers in Bahrain. Oman medical journal. 37 (2): e350.
Muriel P & Gordillo KR 2016. Role of oxidative stress in liver health and disease. Hindawi.
Orasan R, et al. 2011. Efects of L-carnitine on endothelial dysfunction and , visfatin, oxidative stress, inflammation and anemia in hemodialysis patients. Acta endocrinologica. 7 (2).
Pirmadah F, et al. 2019. Does l-carnitine supplementation affect serum levels of enzymes mainly produced by liver? A systematic review and meta-analysis of randomized controlled clinical trials. European journal of nutrition. 1-17.
Raza S, Tewari A, Rajak S & Sinha RA 2021. Vitamins and non-alcoholic fatty liver disease: A molecular insight. Liver research. 5 (2): 62-71.
Rolo AP, Teodoro JS & Palmeira CM 2012. Role of oxidative stress in the pathogenesis of nonalcoholic steatohepatitis. Free radical biology and medicine. 52 (1): 59-69.
Romano M, et al. 2008. L-Carnitine treatment reduces steatosis in patients with chronic hepatitis C treated with α-interferon and ribavirin. Digestive diseases and sciences. 53 (4): 1114-1121.
Rudman D, Sewell CW & Ansley JD 1977. Deficiency of carnitine in cachectic cirrhotic patients. Journal of clinical investigation. 60 (3): 716-723.
Sami W, Ansari T, Butt NS & Ab Hamid MR 2017. Effect of diet on type 2 diabetes mellitus: A review. International journal of health sciences. 11 (2): 65.
Santos JCdF, Valentim IB, De Araújo OR, Ataide TdR & Goulart MO 2013. Development of nonalcoholic hepatopathy: contributions of oxidative stress and advanced glycation end products. International journal of molecular sciences. 14 (10): 19846-19866.
Sarafoglou K, Rodgers J, Hietala A, Matern D & Bentler K 2011. Expanded newborn screening for detection of vitamin B12 deficiency. Journal of the American medical association. 305 (12): 1198-1200.
Sheng X, et al. 2018. The relationship between liver enzymes and insulin resistance in type 2 diabetes patients with nonalcoholic fatty liver disease. Hormone and Metabolic Research. 50 (05): 397-402.
Shibabaw T, Dessie G, Molla MD, Zerihun MF & Ayelign B 2019. Assessment of liver marker enzymes and its association with type 2 diabetes mellitus in Northwest Ethiopia. BMC research notes. 12 (1): 707.
Somi MH, Fatahi E, Panahi J & Havasian MR 2014. Data from a randomized and controlled trial of LCarnitine prescription for the treatment for Non-Alcoholic Fatty Liver Disease. Bioinformation. 10 (9): 575.
Sotirakopoulos N, Athanasiou G, Tsitsios T & Mavromatidis K 2002. The influence of l-carnitine supplementation on hematocrit and hemoglobin levels in patients with end stage renal failure on CAPD. Renal failure. 24 (4): 505-510.
Strasser A, Dedoyard A, Lohninger A & Niedermüller H 2007. l-Carnitine l-tartrate (LCLT) and dehydroepiandrosterone sulfate (DHEAS) affect red and white blood cells in aged Sprague–Dawley rats. Archives of gerontology and geriatrics. 44 (3): 325-336.
Sweeney JD & Arduini A 2004. L-carnitine and its possible role in red cell and platelet storage. Transfusion medicine reviews. 18 (1): 58-65.
Talenezhad N, et al. 2020a. The Effect of L-carnitine Supplementation on Blood Pressure in Patients with Type 2 Diabetes: A Randomized, Double-blind, Placebo-controlled Trial. Obesity Medicine. 100230.
Talenezhad N, Mohammadi M, Ramezani-Jolfaie N, Mozaffari-Khosravi H & Salehi-Abargouei A 2020b. Effects of l-carnitine supplementation on weight loss and body composition: A systematic review and meta-analysis of 37 randomized controlled clinical trials with dose-response analysis. Clinical nutrition ESPEN. 37: 9-23.
Talenezhad N, et al. 2020c. Effects of L-carnitine supplementation on oxidative and metabolic status in patients with type 2 diabetes mellitus: A randomized, double-blind, clinical trial. European journal of integrative medicine. 40: 101243.
Trovato G, GM T, Di Marco V & Dell'Aira A 1982. Long-term L-carnitine treatment of chronic anaemia of patients with end-stage renal failure. Current therapeutic research. 31 (6): 1042-1049.
Turgut O & Tandogan I 2011. Gamma-glutamyltransferase to determine cardiovascular risk: shifting the paradigm forward. Journal of atherosclerosis and thrombosis. 18 (3): 177-181.
Vinik AI, Nevoret M-L, Casellini C & Parson H 2013. Diabetic neuropathy. Endocrinology and metabolism clinics. 42 (4): 747-787.
Zhao L, et al. 2017. Serum alanine aminotransferase/aspartate aminotransferase ratio is one of the best markers of insulin resistance in the Chinese population. Nutrition & metabolism. 14 (1): 64.


 
Type of article: orginal article | Subject: public specific
Received: 2022/10/21 | Published: 2023/05/20 | ePublished: 2023/05/20

Add your comments about this article : Your username or Email:
CAPTCHA

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2024 CC BY-NC 3.0 | Journal of Nutrition and Food Security

Designed & Developed by : Yektaweb