CoQ10 LIPID PROFILES

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

The Journal of Clinical Endocrinology & Metabolism, 2023, 108, 232–249

https://doi.org/10.1210/clinem/dgac585
Advance access publication 7 October 2022
Meta-Analysis

Effects of Coenzyme Q10 Supplementation on Lipid Profiles


in Adults: A Meta-analysis of Randomized Controlled Trials
Zhihao Liu,1,2,3 Zezhong Tian,1,2,3 Dan Zhao,1,2,3 Ying Liang,1,2,3 Suming Dai,1,2,3 Meitong Liu,1,2,3
Shanshan Hou,1,2,3 Xiaoxi Dong,1 Zhaxinima,1 and Yan Yang1,2,3,4
1
School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, Guangdong
Province, PR China

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


2
Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Sun Yat-sen University, Guangzhou 510080, China
3
Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Guangzhou 510080, China
4
China-DRIs Expert Committee on Other Food Substances, Guangzhou 510080, China
Correspondence: Yan Yang, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107,
Guangdong Province, PR China. Email: yangyan3@mail.sysu.edu.cn.

Abstract
Context: Previous meta-analyses have suggested that the effects of coenzyme Q10 (CoQ10) on lipid profiles remain debatable. Additionally, no
meta-analysis has explored the optimal intake of CoQ10 for attenuating lipid profiles in adults.
Objective: This study conducted a meta-analysis to determine the effects of CoQ10 on lipid profiles and assess their dose–response
relationships in adults.
Methods: Databases (Web of Science, PubMed/Medline, Embase, and the Cochrane Library) were systematically searched until August 10,
2022. The random effects model was used to calculate the mean differences (MDs) and 95% CI for changes in circulating lipid profiles. The
novel single-stage restricted cubic spline regression model was applied to explore nonlinear dose–response relationships.
Results: Fifty randomized controlled trials with a total of 2794 participants were included in the qualitative synthesis. The pooled analysis
revealed that CoQ10 supplementation significantly reduced total cholesterol (TC) (MD −5.53 mg/dL; 95% CI −8.40, −2.66; I2 = 70%),
low-density lipoprotein cholesterol (LDL-C) (MD −3.03 mg/dL; 95% CI −5.25, −0.81; I2 = 54%), and triglycerides (TGs) (MD −9.06 mg/dL;
95% CI −14.04, −4.08; I2 = 65%) and increased high-density lipoprotein cholesterol (HDL-C) (MD 0.83 mg/dL; 95% CI 0.01, 1.65; I2 = 82%).
The dose–response analysis showed an inverse J-shaped nonlinear pattern between CoQ10 supplementation and TC in which 400-500 mg/day
CoQ10 largely reduced TC (χ2 = 48.54, P < .01).
Conclusion: CoQ10 supplementation decreased the TC, LDL-C, and TG levels, and increased HDL-C levels in adults, and the dosage of 400 to
500 mg/day achieved the greatest effect on TC.
Key Words: CoQ10 supplementation, lipid profiles, dyslipidemia, meta-analysis
Abbreviations: CoQ10, coenzyme Q10; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; MD, mean difference; RCT,
randomized controlled trial; TC, total cholesterol; TG, triglyceride.

Dyslipidemia is an alteration in circulating lipid profiles char­ located in the inner mitochondrial membrane, plays an im­
acterized by increased total cholesterol (TC), low-density lipo­ portant role in the electron transport chain. CoQ10 has basic
protein cholesterol (LDL-C), triglyceride (TG), and/or physiological functions, such as participating in oxidative
decreased high-density lipoprotein cholesterol (HDL-C) levels phosphorylation and the production of adenosine triphos­
(1). Poorly controlled dyslipidemia is associated with clinical phate, activating cellular metabolism and cellular respiration,
diseases, including diabetes (2), nonalcoholic steatohepatitis regulating cytoplasmic redox potential, and inhibiting super­
(3), and cancers (4), and is considered a major risk factor oxide formation (9, 10). In addition, previous in vitro and in
for atherosclerotic cardiovascular diseases (5). Moreover, ele­ vivo studies have shown that CoQ10 could promote lipolysis
vated LDL-C levels were attributable to 3.79 million deaths of TGs in the process of endothelial metabolism and facilitate
from ischemic heart diseases and 0.532 million deaths from is­ cholesterol efflux from macrophages, which might be poten­
chemic stroke in 2017 globally (6). Therefore, it is essential tial mechanisms for CoQ10 to lower circulating lipid profiles
to explore effective strategies for the regulation of lipid (11–14). In general, the human body cannot synthesize a suf­
profiles (7). ficient amount of CoQ10 in some pathological conditions,
For decades, nutritional strategies to reduce the risk of ath­ such as metabolic syndrome, hypertension, diabetes mellitus,
erosclerotic cardiovascular diseases have attracted much at­ and dyslipidemia (15). In addition, multiple studies have
tention. Coenzyme Q10 (CoQ10) is a nonessential nutrient shown that the serum CoQ10 levels in dyslipidemic patients
and vitamin-like fat-soluble benzoquinone (8). CoQ10, taking lipid-lowering drugs (statins) were low, which

Received: 30 June 2022. Editorial Decision: 4 October 2022. Corrected and Typeset: 7 November 2022
© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1 233

restricted the use of lipid-lowering drugs (16, 17). Therefore, Data Extraction
additional CoQ10 supplementation might be utilized to ad­ Eligible studies were independently reviewed by 2 investi­
dress these health concerns. gators (Z.L. and M.L.), and extracted data included the
A number of randomized controlled trials (RCTs) have name of the first author, year of publication, country of
suggested that CoQ10 supplementation has beneficial origin, type of intervention, form of intervention, sample
effects on circulating lipid profiles (18, 19). Several previous size (in intervention and control groups), participant
meta-analyses of RCTs have shown that CoQ10 supplementa­ demographics (age, gender, baseline lipid profiles, and
tion was effective in improving lipid profiles, especially body mass index), CoQ10 form, dosage of CoQ10 supple­
TC, LDL-C, HDL-C, and TGs, in various disease states mentation, duration of intervention, health status of par­
(11, 20–22). However, some previous studies failed to show ticipants, and the changes in HDL-C, LDL-C, TC, and
beneficial effects of CoQ10 supplementation on any lipid pro­ TGs from baseline to end trial in both the intervention
file (23–25). These results suggest that the effect of CoQ10 on and the control groups.
circulating lipids is still controversial. Furthermore, previous
meta-analyses only focused on populations with specific dis­
Quality Assessment and Certainty of Evidence

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


eases, such as patients with coronary artery diseases (19),
metabolic diseases (20), or diabetes (21). However, no study Study quality for this meta-analysis was independently
has explored the effect of CoQ10 on the general population, measured by 2 reviewers (Z.L. and M.L.) using the
especially the adult population. Additionally, the effective Cochrane Collaboration’s modified risk of bias tool (28).
dose of CoQ10 on the control of lipid profiles, especially the The terms “low”, “high” or “unclear” were used to rate
dose–response relationship, has not been explored. This study bias in 7 domains, including random sequence gen­
knowledge gap has restricted the application of CoQ10 in con­ eration, allocation concealment, performance bias, detec­
trolling blood lipid levels. tion bias, attrition bias, reporting bias and other bias.
We conducted this systematic review and meta-analysis to The GRADE (Grading of Recommendations Assessment,
formulate a clear role of CoQ10 supplementation on lipid Development, and Evaluation) methods were used to as­
markers in adults. Furthermore, we assessed the dose– sess the overall certainty of evidence in studies by
response relationships to determine the effective dose of GRADEpro software (version 3.6). The primary outcomes
CoQ10 supplementation on improving plasma lipid profiles were classified into 4 levels (high, moderate, low, or very
in adults. low) according to study design, risk of bias, inconsistency,
indirectness, imprecision, and other considerations (such
as publication bias) (29).
Materials and Methods
This systematic review is based on the guidelines of the Statistical Analyses
Preferred Reporting Items for Systematic Reviews and Circulating concentrations of lipid profiles were collated in
Meta-Analyses (PRISMA) statement and has been registered milligrams per deciliter (mg/dL). Inverse-variance weighting
on PROSPERO (26). The registration number is was used to calculate the mean differences (MDs) and 95%
CRD42021252933. CI for net changes in circulating lipid profiles. We calculated
the net changes by using the differences (intervention minus
control) in the changes (final values minus baseline values)
Search Strategy in mean values. SDs of MDs were available in 9 studies, and
To find relevant studies on the effects of CoQ10 supplementa­ they were directly applied in our meta-analysis. For other
tion on lipid profiles in humans, this systematic review and studies that did not report them, we calculated the SD of the
meta-analysis conducted a comprehensive literature search MD using the following formula (30):
in online databases, including Web of Science, PubMed/
SD
Medline, Embase, and the Cochrane Library, until August 􏽱�������������������������������������������������������������������
10, 2022. The following search terms were used in our search = (SDbaseline)2 + (SDfinal)2 − (2R × SDbaseline × SDfinal),
strategy: (triglyceride) OR (TG) OR (total cholesterol) OR
(TC) OR (low-density lipoprotein cholesterol) OR (LDL chol­ where R is the correlation coefficient between preinterven­
esterol) OR (LDL-C) OR (high-density lipoprotein choles­ tion and postintervention; as R was not available in those
terol) OR (HDL cholesterol) OR (HDL-C)) AND studies, the correlation coefficient was conservatively set at
((Coenzyme Q10) OR (CoQ10) OR (Ubiquinone)). The de­ 0.5 as previously reported (31, 32). The random effects
tailed search strategies are reported elsewhere (Table S1 (27)). model of DerSimonian and Laird was conducted to deter­
mine the overall effect sizes to minimize the effects of het­
erogeneity between studies (33). Subgroup analyses were
Study Selection conducted by multiple factors, including baseline circulat­
The inclusion criteria for this meta-analysis were as follows: ing concentrations on TC (<200 and ≥200 mg/dL), LDL-C
(1) RCTs with parallel or crossover designs, (2) use of (<130 and ≥130 mg/dL), HDL-C (≥50 and <50 mg/dL),
CoQ10 intervention, (3) intervention time ≥14 days, (4) meas­ and TGs (<150 and ≥150 mg/dL), health condition
urements of TGs, TC, LDL-C, or HDL-C at the beginning and (healthy, diabetes, dyslipidemia, cardiovascular diseases,
end of the trials, and (5) a placebo control or other suitable chronic kidney disease, and other diseases), CoQ10 form
forms of control. The exclusion criteria included (1) acute (ubiquinone and ubiquinol), type of intervention (CoQ10
feeding trials, (2) pregnant or lactating women as subjects, and CoQ10 + other), dosage of intervention (<200 and
and (3) trials with a multifactorial design. ≥200 mg/day), duration of study (<12 and ≥12 weeks),
234 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

and whether funded by industry (Yes and No). The P value 95% CI −26.88, 3.44), and other diseases (MD −6.51 mg/
of the Cochrane’s Q test and I2 index were used to evalu­ dL, 95% CI −13.04, 0.02). A marginally significant subgroup
ate heterogeneity between studies. Sensitivity analyses difference was observed between CoQ10 supplemental dosage
were performed using the single-study deletion (P for subgroup difference = .08): the CoQ10 intervention ef­
(leave-one-out) approach to assess the effect of each study fect on TC was −2.55 mg/dL (95% CI −5.79, 0.70) with-low
on the overall effect size. A single-stage restricted cubic dosage CoQ10 (<200 mg/day) and −7.18 mg/dL (95% CI
spline regression model was applied to explore nonlinear −11.12, −3.23) with high-dosage CoQ10 (≥200 mg/day)
dose–response relationships (34, 35). Potential publication (Table 3).
bias was indicated using Begg’s rank test, Egger’s test, and
funnel plots (36). Statistical analysis was conducted using
the meta package (version 5.1.1) (37) and the dosresmeta
The Effects of CoQ10 Supplementation on
package (version 2.0.1) (38) based on R (version 4.1.0,
Low-Density Lipoprotein Cholesterol
www.R-project.org). A 2-tailed P < .05 was considered
statistically significant. Forty-two RCTs with 1202 subjects in the CoQ10 arm and
1185 subjects in the control arm showed a significant decrease

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


in circulating LDL-C levels (MD −3.03 mg/dL; 95% CI
Results −5.25, −0.81; I2 = 54%, P < .01) (Fig. 3B) following supple­
mentation with CoQ10 regardless of intervention type.
Flow and Characteristics of Studies
Removing any of the studies did not significantly change the
After the multiple database search, 3364 articles were identi­ pooled effect on LDL-C (Fig. 4B). We did not observe signifi­
fied, of which 1023 were rejected due to duplication. After re­ cant subgroup differences, implying that there was no signifi­
viewing the titles and abstracts of the articles, 2259 articles cant source of heterogeneity (all P for heterogeneity of
were excluded for the following reasons: titles and abstracts intervention effects of CoQ10 on LDL-C between subgroups
not relevant to this study (n = 1907), animal studies > .05, Table 3).
(n = 182), and review studies (n = 170). As a result, 82 articles
were reviewed for full text, of which 30 articles were excluded
because they did not meet the inclusion and exclusion criteria,
and 2 articles were excluded because they were duplicate tri­ The Effect of CoQ10 Supplementation on
als. Finally, this study included 50 articles that met all neces­ High-Density Lipoprotein Cholesterol
sary criteria for a meta-analysis (Fig. 1). Forty-one RCTs including 2409 subjects (1214 subjects in
The characteristics of the included studies are shown in the CoQ10 arm and 1195 subjects in the control arm) indi­
Table 1. Fifty RCTs included 60 trial comparisons with a to­ cated a significant increase in circulating HDL-C concentra­
tal of 2794 participants (interventions = 1408 and controls tions (MD 0.83 mg/dL; 95% CI 0.01, 1.65; I2 = 82%, P <
= 1386). The included articles were published between .01) (Fig. 3C) following CoQ10 supplementation. The
1994 and 2022. Fifty-five studies were designed as parallel pooled effect of HDL-C was marginally weaker after re­
studies, and the remaining 5 studies had a crossover design. moving the study of Mohseni M. 2015 (67) (MD 0.40 mg/
The CoQ10 dosages ranged from 100 mg/day to 900 mg/ dL; 95% CI −0.27, 1.06) or Singh R.B. 2003 (78) (MD
day, and durations of supplementation varied from 2 to 0.73 mg/dL; 95% CI −0.09, 1.56) (Fig. 4C). The subgroup
52 weeks. The quality assessment of the included studies analysis suggested that subjects’ health conditions were a
is provided in Fig. 2. The GRADE assessment profiles are significant source of heterogeneity across studies (P for sub­
shown in Table 2. group difference = .03); the CoQ10 intervention effect on
HDL-C was (MD 3.69 mg/dL; 95% CI 0.09, 7.30) among
subjects with dyslipidemia, which was significantly stronger
The Effects of CoQ10 Supplementation on Total
than that among subjects with diabetes (MD 0.17 mg/dL;
Cholesterol
95% CI −1.37, 1.71), cardiovascular diseases (MD
Following CoQ10 supplementation, 45 RCTs with a total of 0.91 mg/dL; 95% CI −1.10, 2.91), chronic kidney disease
2616 subjects (1318 in the CoQ10 arm and 1298 in the control (MD −2.69 mg/dL; 95% CI −4.82, −0.56), other diseases
arm) reported the effects of CoQ10 supplementation on TC. (MD 0.73 mg/dL; 95% CI −0.32, 1.78), and healthy sub­
Because of the significant heterogeneity between studies, a jects (MD 0.16 mg/dL; 95% CI −2.42, 2.73) (Table 3).
random-effects model was used to calculate pooled effects.
The results revealed that CoQ10 supplementation significantly
reduced plasma TC levels (MD −5.53 mg/dL; 95% CI −8.40,
−2.66; I2 = 70%, P < .01) (Fig. 3A). Removing individual stud­ The Effect of CoQ10 Supplementation on
ies did not materially change the pooled estimation of the Triglycerides
CoQ10 intervention effect on TC (Fig. 4A). The subgroup ana­ Forty-two RCTs including 1228 subjects in the CoQ10 arm
lysis suggested that subjects’ health conditions were a major and 1215 subjects in the control arm indicated an overall
source of heterogeneity across studies (P for subgroup differ­ effect of significantly reduced circulating TG concentrations
ence < .01); CoQ10 intervention effect on TC was significant (MD −9.06 mg/dL; 95% CI −14.04, −4.08; I2 = 65%,
(MD −12.30 mg/dL; 95% CI −14.89, −9.71) among healthy P < .01) (Fig. 3D). The pooled effect of CoQ10 on TG did
subjects, which was significantly stronger than that among not change significantly after removing any of the studies
subjects with diabetes (MD −2.08 mg/dL; 95% CI −6.18, (Fig. 4D). Despite high sources of heterogeneity, the test for
2.03), dyslipidemia (MD −4.53 mg/dL; 95% CI −15.96, subgroup differences indicated no significant effect (all Ps
6.89), cardiovascular diseases (MD −0.98 mg/dL; 95% CI for heterogeneity of intervention effects of CoQ10 on TG be­
−3.66, 1.71), chronic kidney disease (MD −11.72 mg/dL; tween subgroups > .05, Table 3).
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1 235

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


Figure 1. Flow diagram of the literature search process.

Dose–Response Relationships Between CoQ10 decreasing TC, LDL-C, and TG and increasing HDL-C lev­
Supplementation and Lipid Profiles els in the adult population. The dose–response analysis re­
The dose–response analysis showed that CoQ10 supplementa­ vealed an inverse J-shaped nonlinear pattern between
tion reduced TC with an inverse J-shaped nonlinear relation­ CoQ10 supplementation and TC, in which the optimal
ship: the beneficial effects increase with the CoQ10 dose was 400 to 500 mg/day. To date, this is the largest
supplementation dosage when it was below 400 mg/day and meta-analysis (60 studies included) evaluating this associ­
then remained flat at that low level (χ2 = 48.54, P < .01) ation, while previous meta-analyses included fewer than
(Fig. 5). We did not find clear dose–response relationships of 25 studies (11, 20–22, 24, 25).
CoQ10 with LDL-C, HDL-C, and TG. This study quantified the potential effects of CoQ10 supple­
mentation on lipid profiles: TC decreased by approximately
5.53 mg/dL, LDL-C decreased by approximately 3.03 mg/
Publication Bias dL, TG decreased by approximately 9.06 mg/dL, and
Publication bias was determined by funnel plot, and Begg’s HDL-C increased by approximately 0.83 mg/dL. Based on a
rank test and Egger’s test. As shown in Fig. 6, Begg’s rank meta-analysis quantifying the association between LDL-C
test and Egger’s test indicated no significant evidence of pub­ and coronary mortality, which showed a 19% reduction in
lication bias for studies evaluating the effects of CoQ10 on TC coronary mortality per mmol/L reduction in LDL-C (86).
(Begg’s: z = 1.05, P = .29; Egger’s: t = 0.21, P = .84), LDL-C This means that reducing LDL-C by 3.03 mg/dL might lower
(Begg’s: z = 1.38, P = 0.17; Egger’s: t = −1.46, P = .15), coronary mortality by approximately 1.49%. Moreover, evi­
HDL-C (Begg’s: z = −0.32, P = 0.75; Egger’s: t = 0.50, dence has shown that TG levels and TC levels are positively
P = .62), and TG (Begg’s: z = 0.91, P = .36; Egger’s: t = −0.58, associated with the incidence and mortality of CVDs, while
P = .57). HDL-C is negatively correlated with those risk factors (87–
89). Therefore, the modest decrease in lipid profiles by
CoQ10 supplementation might be helpful for the early preven­
Discussion tion of CVDs.
Based on 50 RCTs with 2794 subjects, the results of this sys­ The results of previous meta-analyses suggested the bene­
tematic review and meta-analysis indicated that CoQ10 sup­ ficial effects of CoQ10 on some of the 4 lipid profiles but not
plementation significantly improved lipid profiles, such as all of them, as this study has shown. For example, a previous
Table 1. Characteristics of the included studies
236

Trial Country Design Dosage Duration Age (intervention, Gender (Male/ BMI Participants Intervention Control CoQ10 Sample size Presented data
comparisonsa (per day) control) Female) (Intervention, form (intervention/control)
Control)

Abdollahzad H. 2015 Iran Parallel 100 mg 2m 48.77 ± 11.58, 50.57 3/19, 3/20 30.0 ± 4.8, Rheumatoid arthritis CoQ10 Placebo Ubiquinone 45 (22/23) TG, TC
(39) ± 11.05 29.8 ± 4.7
Akbari Fakhrabadi Iran Parallel 200 mg 12 w 56.7 ± 6.4, 10/22, 6/24 28.7 ± 4.1, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 62 (32/30) TC, LDL-C,
M. 2014 (40) 54.8 ± 6.7 29.6 ± 3.1 HDL-C
Andersen C.B. 1997 Denmark Parallel 100 mg 12 w 33.5 ± 2.0, 10/7, 9/8 23.5 ± 2.89, Insulin dependent CoQ10 Placebo Ubiquinone 34 (17/17) LDL-C, HDL-C
(41) 35.3 ± 2.4 24.0 ± 2.47 diabetes mellitus
Aya N. Yasser 2021 Iraq Parallel 200 mg 12 w 59.24 ± 5.57, 58.11 ± 14/14, 13/11 32.54 ± 4.66, Dyslipidemia CoQ10 + atorvastatin Atorvastatin Ubiquinone 52 (28/24) TG, TC, LDL-C,
(42) 7.10 31.59 ± 5.08 HDL-C
Bargossi A.M. 1994 Italy Parallel 100 mg 3m 53.7 ± 10.07, 52.8 ± 10/5, 11/4 72.1 ± 13.19, Primary CoQ10 + simvastatin Simvastatin Ubiquinone 30 (15/15) TG, LDL-C,
(43) 10.84 68.8 ± 9.9 hypercholesterolemia HDL-C
Belardinelli R. 2006 I Italy Crossover 100 mg 4w 59 ± 9 20/3 NR Chronic heart failure CoQ10 + exercise Placebo + Ubiquinone 23 (23/23) TG, TC, LDL-C,
(44) exercise HDL-C
Belardinelli R. 2006 II Italy Crossover 100 mg 4w 59 ± 9 20/3 NR Chronic heart failure CoQ10 Placebo Ubiquinone 23 (23/23) TG, TC, LDL-C,
(44) HDL-C
Chew G.T. 2008 I (45) Australia Parallel 200 mg 6m 61.3 ± 4.1, 62.4 ± 8.8 13/3, 14/6 30.1 ± 4.6, Type 2 diabetes CoQ10 Placebo Ubiquinone 36 (16/20) TG, TC, LDL-C,
30.7 ± 5.0 HDL-C
Chew G.T. 2008 II (45) Australia Parallel 200 mg 6m 63.0 ± 9.4, 64.8 ± 7.3 13/6, 13/6 28.7 ± 3.4, Type 2 diabetes CoQ10 + fenofibrate Fenofibrate Ubiquinone 38 (19/19) TG, TC, LDL-C,
29.9 ± 5.6 HDL-C
Cooke M. 2008 (46) America Parallel 200 mg 14 d 25.76 ± 7.67, 25.61 ± NR NR Aerobically trained and CoQ10 Placebo Ubiquinone 41 (21/20) TG, TC, LDL-C,
8.07 untrained individuals HDL-C
Dai Y.L. 2011 (47) China Parallel 300 mg 8w 67.7 ± 9.4, 70.1 ± 9.8 27/1, 25/3 25.3 ± 3.2, Ischemic left ventricular CoQ10 Placebo Ubiquinone 56 (28/28) TG, TC, LDL-C,
24.7 ± 3.2 systolic dysfunction HDL-C
Eriksson J.G. 1999 (48) Finland Parallel 100 mg 6m 65 ± 5, 64 ± 7 NR 29.0 ± 4.2, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 23 (12/11) TG, TC, HDL-C
29.8 ± 3.4
Fallah M. 2018 (49) Iran Parallel 120 mg 12 w 59.4 ± 12.2, 64.8 ± 22/8, 18/12 26.9 ± 3.9, Diabetic hemodialysis CoQ10 Placebo Ubiquinone 60 (30/30) TG, TC, LDL-C,
11.5 26.6 ± 3.9 HDL-C
Gholami M. 2018 (50) Iran Parallel 100 mg 12 w 53.1 ± 6.23, 53.35 ± 0/68 29.44 ± 0.60, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 68 (34/34) TG, TC, LDL-C,
6.56 28.53 ± 0.53 HDL-C
Gholami M. 2019 (51) Iran Parallel 100 mg 12 w 52.97 ± 1.04, 53.68 ± 0/70 29.30 ± 0.6, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 70 (35/35) TG, TC, LDL-C,
1.14 28.51 ± 0.52 HDL-C
Gholnari T. 2018 (23) Iran Parallel 100 mg 12 w 61.1 ± 11.3, 61.6 ± 8/17, 8/17 30.4 ± 6.1, Diabetic nephropathy CoQ10 Placebo Ubiquinone 50 (25/25) TG, TC, LDL-C,
10.0 31.3 ± 4.8 HDL-C
Hamilton S.J. 2009 Australia Crossover 200 mg 12 w 68 ± 6 NR NR Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 46 (23/23) LDL-C
(52)
Hansen M. 2019 (53) Denmark Parallel 400 mg 8w 62 ± 1, 64 ± 2 14/4, 8/9 28 ± 1, 29 ± 1 Hypercholesterolemia CoQ10 Placebo Ubiquinone 35 (18/17) TG, TC, LDL-C,
HDL-C
Henriksen J.E. 1999 Denmark Parallel 100 mg 3m 35.5 ± 8.2, 35.3 ± 10/7, 9/8 23.5 ± 2.7, Type 1 diabetes mellitus CoQ10 Placebo Ubiquinone 34 (17/17) TG, TC, LDL-C,
(54) 10.0 24.0 ± 2.6 HDL-C
Hernandez-Ojeda Mexico Parallel 400 mg 12 w 55.3 ± 8.4, 57.0 ± 8.9 5/19, 6/19 29.4 ± 7.3, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 49 (24/25) TG, TC, LDL-C,
J. 2012 (55) 29.3 ± 4.3 HDL-C
Hodgson J.M. 2002 I Australia Parallel 200 mg 12 w 51.7 ± 6.97, 53.6 ± 14/5, 14/4 NR Type 2 diabetes mellitus CoQ10 + fenofibrate Fenofibrate Ubiquinone 37 (19/18) TG, TC, LDL-C,
(56) 10.18 and dyslipidemia HDL-C
Hodgson J.M. 2002 II Australia Parallel 200 mg 12 w 52.3 ± 6.1, 55.2 ± 17/2, 13/5 NR Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 37 (19/18) TG, TC, LDL-C,
(56) 9.76 and dyslipidemia HDL-C
Hosseinzadeh-Attar Iran Parallel 200 mg 12 w 45.2 ± 7.6, 47.1 ± 8.3 19/12, 18/15 29.52 ± 2.8, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 64 (31/33) TG, TC, LDL-C,
M. 2015 (57) 29.47 ± 3.24 HDL-C
Ikematsu H. 2006 I Japan Parallel 300 mg 4w 20∼60, 24∼55 11/10, 9/11 NR Healthy CoQ10 Placebo Ubiquinone 41 (21/20) TG, TC, HDL-C
(58)
Ikematsu H. 2006 II Japan Parallel 600 mg 4w 20∼60, 24∼55 11/11, 9/11 NR Healthy CoQ10 Placebo Ubiquinone 42 (22/20) TG, TC, HDL-C
(58)
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

(continued)

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


Table 1. Continued

Trial Country Design Dosage Duration Age (intervention, Gender (Male/ BMI Participants Intervention Control CoQ10 Sample size Presented data
comparisonsa (per day) control) Female) (Intervention, form (intervention/control)
Control)

Ikematsu H. 2006 III Japan Parallel 900 mg 4w 20∼60, 24∼55 9/11, 22/0 NR Healthy CoQ10 Placebo Ubiquinone 42 (22/20) TG, TC, HDL-C
(58)
Izadi A. 2019 I (59) Iran Parallel 200 mg 8w 27.18 ± 5.77, 28.33 ± 0/43 29.28 ± 3.23, Polycystic ovary syndrome CoQ10 + vitamin E Vitamin E Ubiquinone 43 (21/22) TG, TC, LDL-C,
5.52 29.28 ± 4.24 HDL-C
Izadi A. 2019 II (59) Iran Parallel 200 mg 8w 27.64 ± 5.2, 26.0 ± 0/43 28.97 ± 2.95, Polycystic ovary syndrome CoQ10 Placebo Ubiquinone 43 (22/21) TG, TC, LDL-C,
4.53 28.73 ± 3.39 HDL-C
Jafarvand E. 2016 (60) Iran Parallel 100 mg 4w 42.7 ± 10.2, 42.2 ± 15/5 16/5 30.5 ± 3.9, Nonalcoholic fatty liver CoQ10 Placebo Ubiquinone 41 (20/21) TG, TC, LDL-C,
10.8 28.7 ± 4.02 disease HDL-C
Kaikkonen J. 2000 I Finland Parallel 200 mg 3m 60.7 ± 5.7 11/29 NR Mild hypercholesterolemia CoQ10 + α- α-Tocopherol Ubiquinone 20 (10/10) TG, TC, LDL-C,
(61) tocopherol HDL-C
Kaikkonen J. 2000 II Finland Parallel 200 mg 3m 60.7 ± 5.7 11/29 NR Mild hypercholesterolemia CoQ10 Placebo Ubiquinone 20 (10/10) TG, TC, LDL-C,
(61) HDL-C
Kuhlman A.B. 2019 Denmark Parallel 400 mg 8w 62 ± 1, 64 ± 2 14/4, 8/9 27.7 ± 2.55, Patient in primary CoQ10 Placebo Ubiquinone 35 (18/17) TG, TC
(62) 28.8 ± 2.89 prevention
with simvastatin ≥40 mg/d
Lee Y.J. 2011 (63) Korea Parallel 200 mg 12 w 42.7 ± 11.3, 42.5 ± 11/15, 10/15 27.9 ± 2.3, Obesity CoQ10 Placebo Ubiquinone 36 (17/19) TG, TC, HDL-C
11.2 27.6 ± 3.8
Mabuchi H. 2007 (64) Japan Parallel 100 mg 12 w 61 ± 8, 60 ± 8 6/18, 8/17 23.3 ± 2.7, Hypercholesterolemic CoQ10 + atorvastatin Placebo + Ubiquinone 49 (24/25) TG, TC, LDL-C,
23.9 ± 3.4 atorvastatin HDL-C
Majid Iran Parallel 100 mg 12 w 19∼54 NR 28.23 ± 3.60, Nonalcoholic fatty liver CoQ10 Placebo Ubiquinone 41 (20/21) TG, TC
Mohammadshahi F.F. 29.69 ± 5.76 disease
2014 (65)
Mohammed-Jawad Iran Parallel 150 mg 8w 49.37 ± 6.65, 51.63 ± 10/9, 8/11 28.15 ± 4.08, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 38 (19/19) TC
N.K. 2014 (66) 8.13 29.5 ± 4.29
Mohseni M. 2015 (67) Iran Parallel 200 mg 12 w 60 ± 4, 61 ± 3.5 39/13 25.91 ± 2.53, Hyperlipidemic CoQ10 Placebo Ubiquinone 52 (26/26) TG, TC, LDL-C,
26.00 ± 3.34 HDL-C
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

Mori T.A. 2009 I (68) Australia Parallel 200 mg 8w 56.9 ± 16.55, 53.3 ± 17/1, 12/8 30.5 ± 2.2, Chronic kidney disease CoQ10 + omega-3 Omega-3 fatty Ubiquinone 38 (18/20) TG, TC, LDL-C,
14.31 36.4 ± 2.8 fatty acids acids HDL-C
Mori T.A. 2009 II (68) Australia Parallel 200 mg 8w 55.4 ± 12.37, 58.6 ± 17/4, 8/7 26.6 ± 4.12, Chronic kidney disease CoQ10 Placebo Ubiquinone 36 (21/15) TG, TC, LDL-C,
10.07 27.6 ± 6.58 HDL-C
Nuku K. 2007 (69) Japan Parallel 900 mg 4w 40 ± 13, 38 ± 11 12/11, 11/12 21.8 ± 2.1, Healthy CoQ10 Placebo Ubiquinone 46 (23/23) TG, TC, LDL-C,
22.0 ± 1.7 HDL-C
Pek S.L. 2016 (70) Singapore Parallel 150 mg 12 w 43.1 ± 11.3, 49.2 ± 18/2, 17/3 29.2 ± 3.8, Hypercholesterolemic CoQ10 + simvastatin Simvastatin Ubiquinol 40 (20/20) TG, TC, LDL-C,
12.2 31.2 ± 6.1 HDL-C
Playford D.A. 2003 I Australia Parallel 200 mg 12 w 52.7 ± 8.05, 53.5 ± 14/6, 14/6 30.3 ± 4.02, Type 2 diabetes mellitus CoQ10 + fenofibrate Fenofibrate Ubiquinone 40 (20/20) TG, TC, LDL-C,
(71) 9.84 30.0 ± 3.58 and dyslipidemia HDL-C
Playford D.A. 2003 II Australia Parallel 200 mg 12 w 52.7 ± 6.26, 54.7 ± 18/2, 15/5 29.9 ± 3.13, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 40 (20/20) TG, TC, LDL-C,
(71) 9.39 30.9 ± 4.47 and dyslipidemia HDL-C
Pourmoghaddas Iran Parallel 200 mg 4m 50.70 ± 12.5, 54.47 ± 23/9, 22/8 NR Congestive heart failure CoQ10 + atorvastatin Atorvastatin Ubiquinone 62 (32/30) TC
M. 2014 (72) 14.6
Raitakari O.T. 2000 Australia Crossover 150 mg 4w 34 ± 10 3/9 23 ± 4 Hypercholesterolemia CoQ10 Placebo Ubiquinone 24 (12/12) LDL-C
(73) + endothelial dysfunction
Raygan F. 2016 (74) Iran Parallel 100 mg 8w 65.9 ± 12.5, 59.9 ± NR 28.2 ± 5.2, Obese, type 2 diabetes CoQ10 Placebo Ubiquinone 60 (30/30) TG, TC, LDL-C,
13.1 30.7 ± 5.9 mellitus, HDL-C
and coronary heart disease
Rodriguez-Carrizalez Mexico Parallel 400 mg 6 m 28.2 ± 3.7, 29.3 ± 0.8 11/9, 9/11 28.2 ± 3.7, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 40 (20/20) TG, TC, LDL-C,
A.D. 2016 (75) 29.3 ± 0.8 HDL-C
Samimi M. 2017 (19) Iran Parallel 100 mg 12 w 24.5 ± 4.3, 25.3 ± 5.7 0/30, 0/30 27.1 ± 4.3, Polycystic ovary syndrome CoQ10 Placebo Ubiquinone 60 (30/30) TG, TC, LDL-C,
27.9 ± 6.0 HDL-C

(continued)
237

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


Table 1. Continued
238

Trial Country Design Dosage Duration Age (intervention, Gender (Male/ BMI Participants Intervention Control CoQ10 Sample size Presented data
comparisonsa (per day) control) Female) (Intervention, form (intervention/control)
Control)

Shahad A. Bader 2022 Iraq Parallel 200 mg 3m 25∼33 0/50, 0/50 28.84 ± 6.74, Polycystic ovary syndrome CoQ10 Placebo Ubiquinone 100 (50/50) TG, TC, LDL-C,
(76) 30.12 ± 5.46 HDL-C
Shojaei M. 2011 I (77) Iran Parallel 100 mg 3m 52.8 ± 10.4, 55.3 ± 6/8, 6/6 23.3 ± 2.3, Hemodialysis CoQ10 + carnitine Carnitine Ubiquinone 26 (14/12) TG, TC, LDL-C,
15.6 24.3 ± 2.1 HDL-C
Shojaei M. 2011 II (77) Iran Parallel 100 mg 3m 53.5 ± 11.5, 51.6 ± 7/6, 6/7 23.6 ± 2.4, Hemodialysis CoQ10 Placebo Ubiquinone 26 (13/13) TG, TC, LDL-C,
19.2 23.6 ± 2.6 HDL-C
Singh R.B. 2003 (78) India Parallel 120 mg 12 m 48.0 ± 8.6, 47.6 ± 8.2 58/15, 57/14 23.8 ± 1.6, Acute myocardial infarction CoQ10 Placebo Ubiquinone 144 (73/71) TG, TC, LDL-C,
23.6 ± 1.5 HDL-C
Singh R.B. and M.A. India Parallel 120 mg 4w 48.4 ± 0.5, 47.6 ± 0.3 19/6, 18/4 23.6 ± 1.2, Acute myocardial CoQ10 Placebo Ubiquinone 47 (25/22) TC, LDL-C,
Niaz 1999 (79) 23.5 ± 1.2 infarction, HDL-C
unstable angina, angina
pectoris
Skarlovnik A. 2014 Slovenia Parallel 100 mg 30 d 64.5 ± 1.9, 65.6 ± 2.1 11/14, 12/13 25.3 ± 1.2, Statin myalgia CoQ10 Placebo Ubiquinone 50 (25/25) TG, TC, LDL-C
(80) 24.6 ± 1.5
Taylor B.A. 2015 (81) America Crossover 600 mg 8w 58 ± 10, 60 ± 10 27/16 29.6 ± 5.3 Statin myalgia CoQ10 + simvastatin Simvastatin Ubiquinol 76 (38/38) LDL-C
Toth S. 2017 (82) Slovakia Parallel 200 mg 3m 58.4 ± 13.8, 61.96 ± 17/18, 18/17 28.31 ± 3.81 Dyslipidemia CoQ10 + Omega-3 Omega-3 fatty Ubiquinone 70 (35/35) TG, TC, LDL-C,
12.2 fatty acids acids HDL-C
Yen C.H. 2018 (83) China Parallel 100 mg 12 w 61.5 ± 10.2, 59.6 ± 17/7, 14/9 28.0 ± 4.8, Type 2 diabetes mellitus CoQ10 Placebo Ubiquinol 47 (24/23) TG, TC, LDL-C,
11.7 27.3 ± 3.4 HDL-C
Young J.M. 2007 (84) New Parallel 200 mg 12 w 59 ± 9.4, 59 ± 9.4 12/10, 10/12 NR Myalgia CoQ10 + simvastatin Simvastatin Ubiquinone 44 (22/22) TG, TC, LDL-C,
Zealand HDL-C
Zahedi H. 2014 (85) Iran Parallel 150 mg 12 w 53.5 ± 9.7, 58.8 ± 9.6 11/9, 8/12 NR Type 2 diabetes mellitus CoQ10 Placebo Ubiquinone 40 (20/20) TG, TC, LDL-C,
HDL-C
Zhang P. 2018 (18) China Parallel 120 mg 24 w 51.78 ± 8.92, 50.02 ± 14/37, 18/32 25.23 ± 3.96, Dyslipidemia CoQ10 Placebo Ubiquinone 101 (51/50) TG, TC, LDL-C,
10.91 24.91 ± 3.32 HDL-C

Abbreviations: CoQ10, coenzyme Q10; d, days; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; m, months; NR, not reported; TC, total cholesterol; TG, triglyceride; w, weeks.
a
When 1 RCT included more than 1 dosage or intervention, we counted it as the corresponding number of trial comparisons, and named it as the RCT name followed by the number, such as Belardinelli R. 2006 I (CoQ10 + exercise vs placebo + exercise) and Belardinelli
R. 2006 II (CoQ10 vs placebo) were counted as 2 studies that were included in the same RCT (Belardinelli R. 2006).
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1 239

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


Figure 2. The quality assessment of included studies. A. Review authors’ judgements about each risk of bias item presented as percentages across all
included studies; B. Review authors’ judgements about each risk of bias item for each included study.

meta-analysis in patients with coronary artery disease dem­ Specifically, we performed a dose–response meta-analysis
onstrated that CoQ10 supplementation significantly im­ to explore the potential optimal dose of CoQ10 supplementa­
proved TC and HDL-C levels, but it did not significantly tion on improving lipid profiles by using a novel 1-stage
affect other lipid profiles (21). Another meta-analysis re­ model that allows us to include trials with only 1 interven­
ported that CoQ10 significantly decreased TG, but had tion dosage. To our knowledge, this is the first dose–
no significant effect on TC, LDL-C or HDL-C among par­ response meta-analysis of RCTs to evaluate the effect of
ticipants with metabolic diseases (11). Furthermore, a CoQ10 supplementation on lipid profiles. We found an in­
meta-analysis in diabetic patients suggested that CoQ10 verse J-shaped nonlinear relationship with the effect of
significantly reduced TC and LDL-C (90), while another CoQ10 supplementation on TC, which revealed that the
meta-analysis in patients with chronic kidney disease beneficial effects increased when CoQ10 < 400 mg/day and
showed no effects of CoQ10 on TC, LDL-C, HDL-C, and then plateaued at higher doses. However, few observational
TG (25). One major reason for the inconsistent results of studies or RCTs have supported the inverse J-shaped non­
previous meta-analyses should be the study size, as each linear dose–response relationship. Despite the lack of direct
included only part of the RCTs such that each showed a evidence, there was a similar trend in the effect of CoQ10
significant intervention effect on some lipid profiles but supplementation on lipid profiles in a meta-analysis based
not on other lipid profiles, but they were all in the right on RCTs, significantly decreasing TC at lower dosages but
direction. After we summarized all of the RCTs that met not significantly at higher dosages (21). The inverse
our criteria and updated the data with 15 more RCTs, J-shaped curves provide a reference for future RCTs to ex­
the intervention effects of CoQ10 supplementation on plore dose–response relationship between CoQ10 supplemen­
TC, TG, LDL, and HDL all reached statistically significant tation and lipid profiles.
levels. Another potential reason is the confounding effect Several potential mechanisms have been proposed by
from diseases that might conceal the true intervention ef­ which CoQ10 supplementation may improve circulating
fect. We further compared the results by subgroup analysis lipids (12, 13, 91–98). In human endothelial cells, exposure
and found that 5 studies among healthy subjects indicated to CoQ10 is associated with downregulation of the lectin-
a significant intervention effect of CoQ10 on TC without like oxidized LDL receptor, stimulation of 5’ adenosine
significant heterogeneity across studies, which was stron­ monophosphate-activated protein kinase (AMPK), and re­
ger than the intervention effect among subjects with dysli­ duction of reactive oxygen species (ROS)-induced endothe­
pidemia, diabetes, cardiovascular diseases, chronic kidney lial damage (98). In fact, the primary effect of CoQ10 on
disease, or other diseases. Regarding the intervention ef­ circulating lipid profiles appears to increase LDL resistance
fect of CoQ10 on HDL-C, 10 studies among subjects to oxidative stress (92), as demonstrated in healthy adults
with dyslipidemia indicated a significant intervention ef­ following acute strenuous physical activity (97). In vitro,
fect of CoQ10 on HDL-C, which was stronger than the it inhibits LDL oxidation better than other antioxidant
intervention effects among healthy subjects or patients molecules such as alpha-tocopherol or beta-carotene (94,
with diabetes, cardiovascular diseases, chronic kidney dis­ 95). CoQ10 can also significantly improve HDL-mediated
ease, or other diseases. cholesterol efflux capacity, and this may be related to the
240

Table 2. GRADE evidence profile of CoQ10 supplementation for lipid profiles

Quality assessment No of patients Effect Quality

No. of Design Risk of bias Inconsistency Indirectness Imprecision Other CoQ10 Control MD (95% CI)
studies considerations

TC (better indicated by lower values)


45 Randomized No serious risk Seriousa No serious No serious None 1318 1298 −5.53 (−8.40, 2.66) ⊕⊕⊕OMODERATE
trials of bias indirectness imprecision
LDL-C (better indicated by lower values)
42 Randomized No serious risk Seriousb No serious No serious None 1202 1185 −3.03 (−5.25, −0.81) ⊕⊕⊕O
trials of bias indirectness imprecision MODERATE
HDL-C (better indicated by lower values)
41 Randomized No serious risk Very seriousc No serious No serious None 1214 1195 0.83 (0.01, 1.65) ⊕⊕OO LOW
trials of bias indirectness imprecision
TG (better indicated by lower values)
42 Randomized No serious risk Seriousd No serious No serious None 1228 1215 −9.06 (−14.04, −4.08) ⊕O MODERATE
trials of bias indirectness imprecision

a
The test for heterogeneity is significant, and the I2 is moderate, 70%.
b
The test for heterogeneity is significant, and the I2 is moderate, 54%.
c
The test for heterogeneity is significant, and the I2 is high, 82%.
d
The test for heterogeneity is significant, and the I2 is moderate, 65%.
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1 241

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024

Figure 3. Forest plot detailing mean differences and 95% CIs for the effect of coenzyme Q10 supplementation on plasma lipid profiles. A. Total
cholesterol; B. Low-density lipoprotein cholesterol: C. High-density lipoprotein cholesterol; D. Triglycerides.

regulation of the activator protein-1/miR-378/adenosine tissue (91). In addition, CoQ10 increases fatty acid oxida­
triphosphate–binding cassette transporter G1-signaling tion, which reduces oxidative stress via a decrease in mito­
pathway (14, 99). Moreover, CoQ10 may activate the chondrial free fatty acids, participates in endothelial
calcium-mediated AMPK pathway and inhibit differenti­ metabolism, and increases TG lipolysis (12, 13).
ation-induced adipogenesis, inducing the gene expression Furthermore, CoQ10, in its reduced state, has been shown
of peroxisome proliferator-activated receptor-γ (PPAR-γ) to inhibit the peroxidation of cell membrane lipids and re­
(93), which is a main regulator of lipid storage in adipose duce the oxidation of circulating lipids (96).
242 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024

Figure 4. Leave-one-out sensitivity analysis for the effect of coenzyme Q10 supplementation on plasma lipid profiles. A. Total cholesterol; B. Low-
density lipoprotein cholesterol: C. High-density lipoprotein cholesterol; D. Triglycerides.
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1 243

Table 3. Overall effect and subgroup analyses of coenzyme Q10 supplementation on lipid profiles

Variable N Sample size MD (95% CI) Heterogeneity


(experimental/control)
P heterogeneity I2 % P between subgroups

TC (mg/dL)
Overall 55 1318/1298 −5.53 (−8.40, −2.66) <.01 70
Baseline TC (mg/dL)
<200 35 823/814 −6.87 (−10.14, −3.60) <.01 65 .20
≥200 20 495/484 −2.30 (−8.45, 3.84) <.01 77
Health condition
Healthy 5 109/103 −12.30 (−14.89, −9.71) .43 0 <.01
Diabetes 19 432/429 −2.08 (−6.18, 2.03) .05 37
Dyslipidemia 9 214/212 −4.53 (−15.96, 6.89) <.01 79

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


Cardiovascular diseases 7 230/223 −0.98 (−3.66, 1.71) .75 0
Chronic kidney disease 2 39/35 −11.72 (−26.88, 3.44) <.01 92
Other diseases 13 294/296 −6.51 (−13.04, 0.02) .04 44
CoQ10 form
ubiquinone 53 1274/1255 −5.68 (−8.61, −2.75) <.01 71 .41
Ubiquinol 2 44/43 0.47 (−13.81, 14.75) .94 0
Type of intervention
CoQ10 40 1004/988 −6.40 (−9.73, −3.07) <.01 76 .22
CoQ10 + other 15 314/310 −2.69 (−7.64, 2.27) .26 17
Dosage of intervention (mg/day)
<200 24 625/619 −2.55 (−5.79, 0.70) .13 25 .08
≥200 31 693/679 −7.18 (−11.12, −3.23) <.01 76
Duration of study (weeks)
<12 20 438/425 −7.95 (−11.99, −3.91) <.01 71 .14
≥12 35 880/873 −5.53 (−8.40, −2.66) <.01 67
Funded by industry
Yes 22 483/470 −5.01 (−9.19, −0.82) <.01 77 .78
No 33 835/828 −5.86 (−10.10, −1.62) <.01 63
LDL-C (mg/dL)
Overall 50 1202/1185 −3.03 (−5.25, −0.81) <.01 54
Baseline LDL-C (mg/dL)
<130 39 917/904 −3.53 (−6.09, −0.98) <.01 56 .46
≥130 11 285/281 −1.36 (−6.59, 3.87) .03 50
Health condition
Healthy 2 44/43 −5.00 (−15.55, 5.55) 1.00 0 .08
Diabetes 19 441/439 0.00 (−4.18, −4.19) <.01 63
Dyslipidemia 11 243/241 −2.78 (−9.11, 3.56) .01 56
Cardiovascular diseases 6 198/193 −1.34 (−3.50, 0.82) 0 1.00
Chronic kidney disease 2 39/35 −5.81 (−10.09, −1.53) .38 0
Other diseases 10 237/234 −9.19 (−15.14, −3.24) .14 34
CoQ10 form
Ubiquinone 47 1138/1124 −3.02 (−5.32, −0.72) <.01 57 .94
Ubiquinol 3 64/61 −3.40 (−12.91, 6.11) .88 0
Type of intervention
CoQ10 34 984/866 −2.93 (−5.59, −0.26) <.01 65 .70
CoQ10 + other 16 318/319 −3.78 (−7.28, −0.28) .67 0
Dosage of intervention (mg/day)
<200 23 598/591 −3.45 (−7.03, 0.13) <.01 58 .79
≥200 27 604/594 −2.81 (−5.90, 0.29) <.01 53
Duration of study (weeks)
<12 16 346/336 −2.40 (−4.30, −0.50) .93 0 .86
≥12 34 856/849 −2.74 (−5.91, 0.43) <.01 66
Funded by industry
Yes 20 440/434 −1.18 (−3.78, 1.41) .10 31 .10
No 30 762/751 −4.82 (−8.35, −1.30) <.01 64
HDL-C (mg/dL)

(continued)
244 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

Table 3. Continued

Variable N Sample size MD (95% CI) Heterogeneity


(experimental/control)
P heterogeneity I2 % P between subgroups

Overall 51 1214/1195 0.83 (0.01, 1.65) <.01 82


Baseline HDL-C (mg/dL)
≥50 20 425/410 −0.01 (−1.19, 1.17) <.01 73 .14
<50 31 789/785 1.30 (0.00, 2.61) .01 85
Health condition
Healthy 5 109/103 0.16 (−2.42, 2.73) <.01 70 .03
Diabetes 19 430/427 0.17 (−1.37, 1.71) <.01 59
Dyslipidemia 10 229/227 3.69 (0.09, 7.30) <.01 84
Cardiovascular diseases 6 198/193 0.91 (−1.10, 2.91) .12 42

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


Chronic kidney disease 2 39/35 −2.69 (−4.82, −0.56) .22 34
Other diseases 9 209/210 0.73 (−0.32, 1.78) <.01 83
CoQ10 form
Ubiquinone 49 1170/1152 0.77 (−0.05, 1.60) <.01 83 .24
Ubiquinol 2 44/43 4.74 (−1.81, 11.30) .64 0
Type of intervention
CoQ10 36 915/899 0.72 (−0.36, 1.79) <.01 86 .69
CoQ10 + other 15 299/296 1.05 (−0.22, 2.32) .023 45
Dosage of intervention (mg/d)
<200 22 571/563 0.87 (−0.60, 2.34) <.01 60 .96
≥200 29 643/632 0.82 (−0.21, 1.86) <.01 88
Duration of study (weeks)
<12 16 354/341 0.15 (−0.81, 1.11) <.01 69 .19
≥12 35 860/854 1.28 (−0.13, 2.68) <.01 85
Funded by industry
Yes 22 482/470 0.11 (−0.97, 1.19) <.01 76 .22
No 29 732/725 1.26 (−0.21, 2.74) <.01 85
TG (mg/dL)
Overall 52 1228/1215 −9.06 (−14.04, −4.08) <.01 65
Baseline TG (mg/dL)
<150 30 672/664 −10.11 (−17.54, −2.68) <.01 76 .22
≥150 22 556/551 −4.69 (−9.07, −0.30) .34 9
Health condition
Healthy 5 109/103 −6.47 (−29.99, 17.05) <.01 91 .99
Diabetes 17 382/381 −7.40 (−13.96, −0.84) .04 41
Dyslipidemia 10 231/229 −9.81 (−26.67, 7.06) <.01 73
Cardiovascular diseases 5 173/171 −4.76 (−13.48, 3.96) .69 0
Chronic kidney disease 2 39/35 −8.68 (−14.05, 8.68) .02 82
Other diseases 13 294/296 −8.57 (−16.47, −0.66) .93 0
CoQ10 form
Ubiquinone 50 1183/1171 −9.10 (−14.08, −4.11) <.01 65 .73
Ubiquinol 2 45/44 8.36 (−88.89, 105.61) .04 75
Type of intervention
CoQ10 37 929/918 −9.30 (−15.08, −3.53) <.01 64 .88
CoQ10 + other 15 299/297 −8.36 (−19.20, 2.49) <.01 69
Dosage of intervention (mg/d)
<200 23 599/596 −10.11 (−18.78, −1.43) <.01 63 .75
≥200 29 629/619 −8.37 (−14.79, −1.94) <.01 66
Duration of study (weeks)
<12 18 394/384 −6.36 (−15.02, 2.30) <.01 70 .45
≥12 34 834/831 −10.50 (−16.76, −4.24) <.01 60
Funded by industry
Yes 21 459/449 −4.22 (−12.72, 4.28) <.01 76 .12
No 31 769/766 −12.38 (−18.20, −6.56) <.01 43

Abbreviations: N, number of studies included; MD, mean difference; mg/d, milligrams per day.
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1 245

A major strength of this review is the large study size: data evidence of publication bias. The included studies were high-
from the 50 RCTs provided sufficient power to examine the quality randomized and double-blind trials and were robust in
effect of CoQ10 on lipid profiles. In general, there was no sensitivity analyses, minimizing the risk of confounding and
bias. Several limitations of the present study warrant consider­
ation. Most of the included studies had a small number of sub­
jects, and there was a certain degree of heterogeneity.
Sampling error might lead to substantial bias in the interven­
tion effect estimation; however, in our estimations, we
used the MD to evaluate the intervention effect. As Lin dem­
onstrated by a series of sensitivity analyses, sampling error
did not cause noticeable bias when the effect size was the
MD (100). Furthermore, in our meta-analysis, only 1 RCT
with 3 dose groups could be included in the 2-stage model.
In order to avoid the systematic exclusion of studies with few­

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


er than 3 dose groups and to allow the inclusion of RCTs with
only 1 comparison, we used the flexible 1-stage model to ex­
plore dose–response relationships as in previous meta-
analyses (101–105). In addition, the meta-analysis of CoQ10
Figure 5. Dose–response relationship between dose of CoQ10 supplementation on HDL-C showed significant heterogeneity
supplementation and mean differences in total cholesterol. The dotted in the sensitivity analysis, which warrants further replication
lines represent 95% CI. of RCTs in the future.

Figure 6. Funnel plot revealing publication bias in the studies reporting the effects of coenzyme Q10 supplementation on plasma lipid profiles. A. Total
cholesterol; B. Low-density lipoprotein cholesterol: C. High-density lipoprotein cholesterol; D. Triglycerides.
246 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

In conclusion, this comprehensive systematic review and for the Global Burden of Disease Study 2017. Lancet. 2018;392
meta-analysis demonstrated that CoQ10 supplementation (10159):1923-1994.
might have promising effects on lipid profiles by decreas­ 7. Pirillo A, Casula M, Olmastroni E, Norata GD, Catapano AL.
ing TC, TGs, and LDL-C levels and increasing HDL-C lev­ Global epidemiology of dyslipidaemias. Nat Rev Cardiol.
2021;18(10):689-700.
els. For LDL-C, the CoQ10 intervention effect is the
8. Bonakdar RA, Guarneri E. Coenzyme Q10. Am Fam Physician.
greatest when the dosage is 400 to 500 mg/day. More 2005;72(6):1065-1070.
RCTs are warranted to determine the dose–response rela­ 9. Ernster L, Dallner G. Biochemical, physiological and medical as­
tionships between CoQ10 supplementation and other lipid pects of ubiquinone function. Biochim Biophys Acta.
profiles. 1995;1271(1):195-204.
10. Turunen M, Olsson J, Dallner G. Metabolism and function of co­
enzyme Q. Biochim Biophys Acta. 2004;1660(1):171-199.
Acknowledgments 11. Sharifi N, Tabrizi R, Moosazadeh M, et al. The effects of coen­
The authors acknowledge the help of American journal ex­ zyme Q10 supplementation on lipid profiles among patients
perts in editing this manuscript. with metabolic diseases: a systematic review and meta-analysis
of randomized controlled trials. Curr Pharm Des. 2018;24(23):

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


2729-2742.
Funding 12. Marcoff L, Thompson PD. The role of coenzyme Q10 in
statin-associated myopathy: a systematic review. J Am Coll
The authors would like to acknowledge the Key Program of
Cardiol. 2007;49(23):2231-2237.
the National Natural Science Foundation of China (no. 13. Celik T, Iyisoy A. Coenzyme Q10 and coronary artery bypass sur­
82030098), the National Natural Science Foundation of gery: what we have learned from clinical trials. J Cardiothorac
China (no. 81872617), Science, Technology and Innovation Vasc Anesth. 2009;23(6):935-936.
Commission of Shenzhen Municipality (no. 14. Wang D, Yan X, Xia M, et al. Coenzyme Q10 promotes macro­
JCYJ20180307153228190), CNS Research Fund for DRI phage cholesterol efflux by regulation of the activator protein-1/
for financial support, and the National College Students miR-378/ATP-binding cassette transporter G1-signaling path­
Innovation and Entrepreneurship Training Program (no. way. Arterioscler Thromb Vasc Biol. 2014;34(9):1860-1870.
202210558159 and 202210558161). 15. Shukla S, Dubey KK. Coq10 a super-vitamin: review on applica­
tion and biosynthesis. 3 Biotech. 2018;8(5):249.
16. Gutierrez-Mariscal FM, de la Cruz-Ares S, Torres-Peña JD,
Author Contributions Alcalá-Diaz JF, Yubero-Serrano EM, López-Miranda J.
Coenzyme Q(10) and cardiovascular diseases. Antioxidants
Z.L. and M.L. had full access to all of the data and take re­ (Basel). 2021;10(6):906.
sponsibility for the integrity of the data. Concept and design: 17. Arenas-Jal M, Suñé-Negre JM, García-Montoya E. Coenzyme
Y.Y., Z.T. Acquisition, or interpretation of data: Z.L., Z.T., Q10 supplementation: efficacy, safety, and formulation chal­
D.Z., Y.L., S.D., M.L., S.H., X.D., Zhaxinima, Y.Y. lenges. Compr Rev Food Sci Food Saf. 2020;19(2):574-594.
Analysis of the data and drafting of the manuscript: Z.L. 18. Zhang P, Yang C, Guo H, et al. Treatment of coenzyme Q10 for
Critical revision of the manuscript for important intellectual 24 weeks improves lipid and glycemic profile in dyslipidemic indi­
content: Y.Y. Statistical analysis: Z.L. Obtained funding: viduals. J Clin Lipidol. 2018;12(2):417-427.e5.
Y.Y. Supervision: Y.Y., Z.T. 19. Samimi M, Zarezade Mehrizi M, Foroozanfard F, et al. The effects
of coenzyme Q10 supplementation on glucose metabolism and
lipid profiles in women with polycystic ovary syndrome: a
Disclosures randomized, double-blind, placebo-controlled trial. Clin
Endocrinol (Oxf). 2017;86(4):560-566.
The authors declare no conflict of interest.
20. Bakhshayeshkaram M, Lankarani KB, Mirhosseini N, et al. The
effects of coenzyme Q10 supplementation on metabolic profiles
Data Availability of patients with chronic kidney disease: a systematic review and
meta-analysis of randomized controlled trials. Curr Pharm Des.
Original data generated and analyzed during this study are in­ 2018;24(31):3710-3723.
cluded in the data repositories listed in reference (106). 21. Jorat MV, Tabrizi R, Mirhosseini N, et al. The effects of coenzyme
Q10 supplementation on lipid profiles among patients with coron­
ary artery disease: a systematic review and meta-analysis of
References randomized controlled trials. Lipids Health Dis. 2018;17(1):230.
1. Kopin L, Lowenstein C. Dyslipidemia. Ann Intern Med. 22. Suksomboon N, Poolsup N, Juanak N. Effects of coenzyme Q10
2017;167(11):Itc81-Itc96. supplementation on metabolic profile in diabetes: a systematic re­
2. Wu L, Parhofer KG. Diabetic dyslipidemia. Metabolism. 2014;63 view and meta-analysis. J Clin Pharm Ther. 2015;40(4):413-418.
(12):1469-1479. 23. Gholnari T, Aghadavod E, Soleimani A, Hamidi GA, Sharifi N,
3. Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty Asemi Z. The effects of coenzyme Q10 supplementation on glu­
liver disease and dyslipidemia: an update. Metabolism. cose metabolism, lipid profiles, inflammation, and oxidative stress
2016;65(8):1109-1123. in patients with diabetic nephropathy: a randomized, double-
4. Butler LM, Perone Y, Dehairs J, et al. Lipids and cancer: emerging blind, placebo-controlled trial. J Am Coll Nutr. 2018;37(3):
roles in pathogenesis, diagnosis and therapeutic intervention. Adv 188-193.
Drug Deliv Rev. 2020;159:245-293. 24. Sahebkar A, Simental-Mendía LE, Stefanutti C, Pirro M.
5. Liu HH, Li JJ. Aging and dyslipidemia: a review of potential mech­ Supplementation with coenzyme Q10 reduces plasma lipoprotei­
anisms. Ageing Res Rev. 2015;19:43-52. n(a) concentrations but not other lipid indices: a systematic review
6. Stanaway JD, Afshin A, Gakidou E, Lim SS. Global, regional, and and meta-analysis. Pharmacol Res. 2016;105:198-209.
national comparative risk assessment of 84 behavioural, environ­ 25. Xu Y, Yang G, Zuo X, et al. A systematic review for the efficacy of
mental and occupational, and metabolic risks or clusters of risks coenzyme Q10 in patients with chronic kidney disease. Int Urol
for 195 countries and territories, 1990-2017: a systematic analysis Nephrol. 2022;54(1):173-184.
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1 247

26. Shamseer L, Moher D, Clarke M, et al. Preferred reporting items 46. Cooke M, Iosia M, Buford T, et al. Effects of acute and 14-day co­
for systematic review and meta-analysis protocols (PRISMA-P) enzyme Q10 supplementation on exercise performance in both
2015: elaboration and explanation. BMJ. 2015;350:g7647. trained and untrained individuals. J Int Soc Sports Nutr.
27. Liu Z, Tian Z, Zhao D, et al. Supplementary data to the paper: ef­ 2008;5:8.
fects of coenzyme Q10 supplementation on lipid profiles. 47. Dai YL, Luk TH, Yiu KH, et al. Reversal of mitochondrial dys­
Figshare. Deposited 16 August 2022. https://figshare.com/ function by coenzyme Q10 supplement improves endothelial
articles/figure/SUMPPLEMENTAL_Table_S1_docx/20496984. function in patients with ischaemic left ventricular systolic dys­
28. Higgins JPT, Altman DG, Gøtzsche PC, et al. The cochrane function: a randomized controlled trial. Atherosclerosis.
Collaboration’s Tool for assessing risk of bias in randomised tri­ 2011;216(2):395-401.
als. BMJ. 2011;343:d5928. 48. Eriksson JG, Forsén TJ, Mortensen SA, Rohde M. The effect of co­
29. Guyatt GH, Oxman AD, Vist GE, et al. GRADE: an emerging con­ enzyme Q10 administration on metabolic control in patients with
sensus on rating quality of evidence and strength of recommenda­ type 2 diabetes mellitus. Biofactors. 1999;9(2-4):315-318.
tions. BMJ. 2008;336(7650):924-926. 49. Fallah M, Askari G, Soleimani A, Feizi A, Asemi Z. Clinical trial of
30. Borenstein M, Hedges LV, Higgins JP, Rothstein HR. the effects of coenzyme Q10 supplementation on glycemic control
Introduction to Meta-Analysis. John Wiley & Sons; 2011. and markers of lipid profiles in diabetic hemodialysis patients. Int
Urol Nephrol. 2018;50(11):2073-2079.

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


31. Fukuta H, Goto T, Wakami K, Ohte N. Effects of drug and exer­
cise intervention on functional capacity and quality of life in heart 50. Gholami M, Zarei P, Sadeghi Sedeh B, Rafiei F, Khosrowbeygi A.
failure with preserved ejection fraction: a meta-analysis of Effects of coenzyme Q10 supplementation on serum values of adi­
randomized controlled trials. Eur J Prev Cardiol. 2016;23(1): ponectin, leptin, 8-isoprostane and malondialdehyde in women
78-85. with type 2 diabetes. Gynecol Endocrinol. 2018;34(12):
32. Follmann D, Elliott P, Suh I, Cutler J. Variance imputation for 1059-1063.
overviews of clinical trials with continuous response. J Clin 51. Gholami M, Rezvanfar MR, Delavar M, Abdollahi M,
Epidemiol. 1992;45(7):769-773. Khosrowbeygi A. Effects of coenzyme Q10 supplementation on
33. DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Serum values of gamma-glutamyl transferase, pseudocholinester­
Clin Trials. 1986;7(3):177-188. ase, bilirubin, ferritin, and high-sensitivity C-reactive protein in
34. Crippa A, Orsini N. Dose–response meta-analysis of differences in women with type 2 diabetes. Exp Clin Endocrinol Diabetes.
means. BMC Med Res Methodol. 2016;16:91. 2019;127(5):311-319.
35. Crippa A, Discacciati A, Bottai M, Spiegelman D, Orsini N. 52. Hamilton SJ, Chew GT, Watts GF. Coenzyme Q10 improves
One-stage dose–response meta-analysis for aggregated data. Stat endothelial dysfunction in statin-treated type 2 diabetic patients.
Diabetes Care. 2009;32(5):810-812.
Methods Med Res. 2019;28(5):1579-1596.
53. Hansen M, Kuhlman ACB, Sahl RE, et al. Inflammatory bio­
36. Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-
markers in patients in Simvastatin treatment: no effect of co-
analysis detected by a simple, graphical test. BMJ. 1997;315
enzyme Q10 supplementation. Cytokine. 2019;113:393-399.
(7109):629-634.
54. Henriksen JE, Andersen CB, Hother-Nielsen O, Vaag A,
37. Balduzzi S, Rücker G, Schwarzer G. How to perform a meta-
Mortensen SA, Beck-Nielsen H. Impact of ubiquinone (coenzyme
analysis with R: a practical tutorial. Evid Based Ment Health.
Q10) treatment on glycaemic control, insulin requirement and
2019;22(4):153-160.
well-being in patients with type 1 diabetes mellitus. Diabet Med.
38. Crippa A, Orsini N. Multivariate dose–response meta-analysis:
1999;16(4):312-318.
the dosresmeta R package. J Stat Softw. 2016;72:1-15.
55. Hernández-Ojeda J, Cardona-Muñoz EG, Román-Pintos LM,
39. Abdollahzad H, Alipour B, Aghdashi MA, Jafarabadi MA.
et al. The effect of ubiquinone in diabetic polyneuropathy: a
Coenzyme Q10 supplementation in patients with rheumatoid
randomized double-blind placebo-controlled study. J Diabetes
arthritis: are there any effects on cardiovascular risk factors?
Complications. 2012;26(4):352-358.
Eur J Integr Med. 2015;7(5):534-539.
56. Hodgson JM, Watts GF, Playford DA, Burke V, Croft KD.
40. Fakhrabadi MA, Ghotrom AZ, Mozaffari-Khosravi H, Hadi
Coenzyme Q10 improves blood pressure and glycaemic control:
Nodoushan H, Nadjarzadeh A. Effect of coenzyme Q10 on oxida­
a controlled trial in subjects with type 2 diabetes. Eur J Clin
tive stress, glycemic control and inflammation in diabetic neur­
Nutr. 2002;56(11):1137-1142.
opathy: a double blind randomized clinical trial. Int J Vitam 57. Hosseinzadeh-Attar M, Kolahdouz Mohammadi R, Eshraghian
Nutr Res. 2014;84(5-6):252-260. M, et al. Reduction in asymmetric dimethylarginine plasma levels
41. Andersen CB, Henriksen JE, Hother-Nielsen O, Vaag A, by coenzyme Q10 supplementation in patients with type 2 dia­
Mortensen SA, Beck-Nielsen H. The effect of coenzyme Q10 on betes mellitus. Minerva Endocrinol. 2015;40(4):259-266.
blood glucose and insulin requirement in patients with insulin de­ 58. Ikematsu H, Nakamura K, Harashima S, Fujii K, Fukutomi N.
pendent diabetes mellitus. Mol Aspects Med. 1997;18(Suppl): Safety assessment of coenzyme Q10 (Kaneka Q10) in healthy sub­
S307-S309. jects: a double-blind, randomized, placebo-controlled trial. Regul
42. Yasser AN, Abdulridha MK, Shafek MA. Assessment of some Toxicol Pharmacol. 2006;44(3):212-218.
clinical and biochemical parameters after combining coenzyme 59. Izadi A, Shirazi S, Taghizadeh S, Gargari BP. Independent and
Q10 to statin in dyslipidemic patients. Int J Drug Deliv additive effects of coenzyme Q10 and vitamin E on cardiometa­
Technol. 2021;11(3):904-911. bolic outcomes and visceral adiposity in women with polycystic
43. Bargossi AM, Battino M, Gaddi A, et al. Exogenous CoQ10 pre­ ovary syndrome. Arch Med Res. 2019;50(2):1-10.
serves plasma ubiquinone levels in patients treated with 60. Jafarvand E, Abbasalizad Farhangi M, Alipoor B, Khoshbaten M.
3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors. Effects of coenzyme Q10 supplementation on the anthropometric
Int J Clin Lab Res. 1994;24(3):171-176. variables, lipid profiles and liver enzymes in patients with non-
44. Belardinelli R, Muçaj A, Lacalaprice F, et al. Coenzyme Q10 and alcoholic fatty liver disease. Bangladesh J Pharmacol.
exercise training in chronic heart failure. Eur Heart J. 2006;27 2015;11(1):35.
(22):2675-2681. 61. Kaikkonen J, Nyyssönen K, Tomasi A, et al. Antioxidative efficacy
45. Chew GT, Watts GF, Davis TM, et al. Hemodynamic effects of fe­ of parallel and combined supplementation with coenzyme Q10
nofibrate and coenzyme Q10 in type 2 diabetic subjects with left and d-alpha-tocopherol in mildly hypercholesterolemic subjects:
ventricular diastolic dysfunction. Diabetes Care. 2008;31(8): a randomized placebo-controlled clinical study. Free Radic Res.
1502-1509. 2000;33(3):329-340.
248 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1

62. Kuhlman AB, Morville T, Dohlmann TL, et al. Coenzyme Q10 lipoprotein(a) in maintenance hemodialysis patients on statin
does not improve peripheral insulin sensitivity in statin-treated therapy. Iran J Kidney Dis. 2011;5(2):114-118.
men and women: the LIFESTAT study. Appl Physiol Nutr 78. Singh RB, Neki NS, Kartikey K, et al. Effect of coenzyme Q10 on
Metab. 2019;44(5):485-492. risk of atherosclerosis in patients with recent myocardial infarc­
63. Lee Y-J, Cho W-J, Kim J-K, Lee D-C. Effects of coenzyme Q10 on tion. Mol Cell Biochem. 2003;246(1-2):75-82.
arterial stiffness, metabolic parameters, and fatigue in obese sub­ 79. Singh RB, Niaz MA. Serum concentration of lipoprotein(a) de­
jects: a double-blind randomized controlled study. J Med Food. creases on treatment with hydrosoluble coenzyme Q10 in patients
2011;14(4):386-390. with coronary artery disease: discovery of a new role. Int J
64. Mabuchi H, Nohara A, Kobayashi J, et al. Effects of CoQ10 sup­ Cardiol. 1999;68(1):23-29.
plementation on plasma lipoprotein lipid, CoQ10 and liver and 80. Skarlovnik A, Janić M, Lunder M, Turk M, Šabovič M. Coenzyme
muscle enzyme levels in hypercholesterolemic patients treated Q10 supplementation decreases statin-related mild-to-moderate
with atorvastatin: a randomized double-blind study. muscle symptoms: a randomized clinical study. Med Sci Monit.
Atherosclerosis. 2007;195(2):e182-e189. 2014;20:2183-2188.
65. Mohammadshahi M, Farsi F, Nejad PA, Hajiani E, Zarei M, 81. Taylor BA, Lorson L, White CM, Thompson PD. A randomized
Engali KA. The coenzyme Q10 supplementation effects on lipid trial of coenzyme Q10 in patients with confirmed statin myop­
profile, fasting blood sugar, blood pressure and oxidative stress athy. Atherosclerosis. 2015;238(2):329-335.

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


status among non-alcoholic fatty liver disease patients: a random­ 82. Tóth Š, Šajty M, Pekárová T, et al. Addition of omega-3 fatty acid
ized, placebo-controlled, pilot study. J Gastroenterol Hepatol Res. and coenzyme Q10 to statin therapy in patients with combined
2014;3(6):1108-1113. dyslipidemia. J Basic Clin Physiol Pharmacol. 2017;28(4):
66. Mohammed-Jawad NK, Sabbagh MA, Al-Jezaeri KA. Role of 327-336.
L-carnitine and coenzyme Q10 as adjuvant therapy in patients 83. Yen CH, Chu YJ, Lee BJ, Lin YC, Lin PT. Effect of liquid ubiqui­
with type 2 diabetes Mellitus. Am J Pharmacol Sci. 2014;2(5): nol supplementation on glucose, lipids and antioxidant capacity in
82-86. type 2 diabetes patients: a double-blind, randomised, placebo-
67. Mohseni M, Vafa M, Zarrati M, Shidfar F, Hajimiresmail SJ, controlled trial. Br J Nutr. 2018;120(1):57-63.
Rahimi Forushani A. Beneficial effects of coenzyme Q10 supple­ 84. Young JM, Florkowski CM, Molyneux SL, et al. Effect of coen­
mentation on lipid profile and intereukin-6 and intercellular adhe­ zyme Q(10) supplementation on simvastatin-induced myalgia.
Am J Cardiol. 2007;100(9):1400-1403.
sion molecule-1 reduction, preliminary results of a double-blind
85. Zahedi H, Eghtesadi S, Seifirad S, et al. Effects of CoQ10 supple­
trial in acute myocardial infarction. Int J Prev Med. 2015;6:73.
mentation on lipid profiles and glycemic control in patients with
68. Mori TA, Burke V, Puddey I, et al. The effects of [omega]3 fatty
type 2 diabetes: a randomized, double blind, placebo-controlled
acids and coenzyme Q10 on blood pressure and heart rate in
trial. J Diabetes Metab Disord. 2014;13:81.
chronic kidney disease: a randomized controlled trial. J
86. Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of
Hypertens. 2009;27(9):1863-1872.
cholesterol-lowering treatment: prospective meta-analysis of
69. Nuku K, Matsuoka Y, Yamagishi T, Miyawaki H, Sato K. Safety
data from 90,056 participants in 14 randomised trials of statins.
assessment of PureSorb-Q40 in healthy subjects and serum coen­
Lancet. 2005;366(9493):1267-1278.
zyme Q10 level in excessive dosing. J Nutr Sci Vitaminol (Tokyo).
87. Ahn N, Kim K. High-density lipoprotein cholesterol (HDL-C) in
2007;53(3):198-206.
cardiovascular disease: effect of exercise training. Integr Med
70. Pek SL, Tavintharan S, Woon K, et al. MicroRNAs as biomarkers
Res. 2016;5(3):212-215.
of hepatotoxicity in a randomized placebo-controlled study of
88. Arsenault BJ, Boekholdt SM, Kastelein JJP. Lipid parameters for
simvastatin and ubiquinol supplementation. Exp Biol Med
measuring risk of cardiovascular disease. Nat Rev Cardiol.
(Maywood). 2016;241(3):317-330. 2011;8(4):197-206.
71. Playford DA, Watts GF, Croft KD, Burke V. Combined effect of
89. Nordestgaard BG, Benn M, Schnohr P, Tybjaerg-Hansen A.
coenzyme Q10 and fenofibrate on forearm microcirculatory func­ Nonfasting triglycerides and risk of myocardial infarction, ische­
tion in type 2 diabetes. Atherosclerosis. 2003;168(1):169-179. mic heart disease, and death in men and women. JAMA.
72. Pourmoghaddas M, Rabbani M, Shahabi J, Garakyaraghi M, 2007;298(3):299-308.
Khanjani R, Hedayat P. Combination of atorvastatin/coenzyme 90. Dludla PV, Nyambuya TM, Orlando P, et al. The impact of coen­
Q10 as adjunctive treatment in congestive heart failure: a double- zyme Q(10) on metabolic and cardiovascular disease profiles in
blind randomized placebo-controlled clinical trial. ARYA diabetic patients: a systematic review and meta-analysis of
Atheroscler. 2014;10(1):1-5. randomized controlled trials. Endocrinol Diabetes Metab.
73. Raitakari OT, McCredie RJ, Witting P, et al. Coenzyme Q im­ 2020;3(2):e00118.
proves LDL resistance to ex vivo oxidation but does not enhance 91. Feige JN, Gelman L, Michalik L, Desvergne B, Wahli W. From
endothelial function in hypercholesterolemic young adults. Free molecular action to physiological outputs: peroxisome
Radic Biol Med. 2000;28(7):1100-1105. proliferator-activated receptors are nuclear receptors at the cross­
74. Raygan F, Rezavandi Z, Dadkhah Tehrani S, Farrokhian A, Asemi roads of key cellular functions. Prog Lipid Res. 2006;45(2):
Z. The effects of coenzyme Q10 administration on glucose 120-159.
homeostasis parameters, lipid profiles, biomarkers of inflamma­ 92. Kaikkonen J, Nyyssönen K, Porkkala-Sarataho E, et al. Effect of
tion and oxidative stress in patients with metabolic syndrome. oral coenzyme Q10 supplementation on the oxidation resistance
Eur J Nutr. 2016;55(8):2357-2364. of human VLDL + LDL fraction: absorption and antioxidative
75. Rodríguez-Carrizalez AD, Castellanos-González JA, Martínez- properties of oil and granule-based preparations. Free Radic
Romero EC, et al. The effect of ubiquinone and combined Biol Med. 1997;22(7):1195-1202.
antioxidant therapy on oxidative stress markers in non- 93. Lee SK, Lee JO, Kim JH, et al. Coenzyme Q10 increases the fatty
proliferative diabetic retinopathy: a phase IIa, randomized, acid oxidation through AMPK-mediated PPARα induction in
double-blind, and placebo-controlled study. Redox Rep. 3T3-L1 preadipocytes. Cell Signal. 2012;24(12):2329-2336.
2016;21(4):155-163. 94. Littarru GP, Tiano L. Bioenergetic and antioxidant properties of
76. Bader SA, Althanoon ZA, Raoof HS. The metabolic effects of coenzyme Q10: recent developments. Mol Biotechnol.
coenzyme Q10 in patients with polycystic ovary syndrome. TC 2007;37(1):31-37.
(mg\dl). 2022;176(37.56):150-193. 95. Malekmohammad K, Sewell RDE, Rafieian-Kopaei M.
77. Shojaei M, Djalali M, Khatami M, Siassi F, Eshraghian M. Effects Antioxidants and atherosclerosis: mechanistic aspects.
of carnitine and coenzyme Q10 on lipid profile and serum levels of Biomolecules. 2019;9(8):301.
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 1 249

96. Martelli A, Testai L, Colletti A, Cicero AFG. Coenzyme Q(10): 102. Sabe M, Zhao N, Crippa A, Strauss GP, Kaiser S. Intranasal oxyto­
clinical applications in cardiovascular diseases. Antioxidants cin for negative symptoms of schizophrenia: systematic review,
(Basel). 2020;9(4):341. meta-analysis, and dose–response meta-analysis of randomized con­
97. Sarmiento A, Diaz-Castro J, Pulido-Moran M, et al. Short-term trolled trials. Int J Neuropsychopharmacol. 2021;24(8):601-614.
ubiquinol supplementation reduces oxidative stress associated 103. Asoudeh F, Talebi S, Jayedi A, Marx W, Najafi MT, Mohammadi
with strenuous exercise in healthy adults: a randomized trial. H. Associations of total protein or animal protein intake and ani­
Biofactors. 2016;42(6):612-622. mal protein sources with risk of kidney stones: a systematic review
98. Tsai KL, Chen LH, Chiou SH, et al. Coenzyme Q10 suppresses and dose–response meta-analysis. Adv Nutr. 2022;13(3):
oxLDL-induced endothelial oxidative injuries by the modulation 821-832.
of LOX-1-mediated ROS generation via the AMPK/PKC/ 104. Zhang X, Ritonja JA, Zhou N, Chen BE, Li X. Omega-3 polyun­
NADPH oxidase signaling pathway. Mol Nutr Food Res. saturated fatty acids intake and blood pressure: a dose–response
2011;55(Suppl 2):S227-S240. meta-analysis of randomized controlled trials. J Am Heart
99. Zou J, Tian Z, Zhao Y, et al. Coenzyme Q10 supplementation im­ Assoc. 2022;11(11):e025071.
proves cholesterol efflux capacity and antiinflammatory proper­ 105. Filippini T, Malavolti M, Whelton PK, Naska A, Orsini N, Vinceti
ties of high-density lipoprotein in Chinese adults with M. Blood pressure effects of sodium reduction: dose–response
dyslipidemia. Nutrition. 2022;101:111703. meta-analysis of experimental studies. Circulation. 2021;143

Downloaded from https://academic.oup.com/jcem/article/108/1/232/6751027 by guest on 27 March 2024


100. Lin L. Bias caused by sampling error in meta-analysis with small (16):1542-1567.
sample sizes. PLoS One. 2018;13(9):e0204056. 106. Liu Z, Tian Z, Zhao D, et al. Original data to the paper: effects of co­
101. Jayedi A, Soltani S, Zargar MS, Khan TA, Shab-Bidar S. Central enzyme Q10 supplementation on lipid profiles. Figshare. Deposited
fatness and risk of all cause mortality: systematic review and 17 August 2022. https://figshare.com/articles/dataset/Original_
dose–response meta-analysis of 72 prospective cohort studies. data_to_the_paper_Effects_of_coenzyme_Q10_supplementation_
BMJ. 2020;370:m3324. on_lipid_profiles/20497245.

You might also like