Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-05-14T03:47:48.948Z Has data issue: false hasContentIssue false

Effects of collagen peptide supplementation on cardiovascular markers: a systematic review and meta-analysis of randomised, placebo-controlled trials

Published online by Cambridge University Press:  06 June 2022

Zahra Jalili
Affiliation:
Imam Ali cardiovascular Centre, Kemanshah University of Medical Sciences, Kermanshah, Iran
Faramarz Jalili
Affiliation:
Graduate Studies Student, Sobey School of Business, Saint Mary’s University, Halifax, NS, Canada
Sajjad Moradi
Affiliation:
Nutritional Sciences Department, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
Reza Bagheri
Affiliation:
Department of Exercise Physiology, University of Isfahan, Isfahan, Iran
Seyedeh Parisa Moosavian
Affiliation:
Department of Community Nutrition, Vice-Chancellery for Health, Shiraz University of Medical Sciences, Shiraz, Iran
Fatemeh Naeini
Affiliation:
Department of clinical nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
Hamed Mohammadi
Affiliation:
Department of clinical nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
Seyed Mojtaba Ghoreishy
Affiliation:
Department of clinical nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
Alexei Wong
Affiliation:
Department of Health and Human Performance, Marymount University, Arlington, TX, USA
Nikolaj Travica
Affiliation:
Deakin University, IMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
Mohammad Ali Hojjati Kermani
Affiliation:
Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
Cyrus Jalili*
Affiliation:
Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah , Iran
*
*Corresponding author: Cyrus Jalili, email cjalili@kums.ac.ir
Rights & Permissions [Opens in a new window]

Abstract

Previous studies have advocated that collagen peptide supplementation (CPS) can positively affect cardiovascular health. However, the widespread impact of CPS on CVD-related markers is not fully resolved. Consequently, the current systematic review and meta-analysis aimed to assess the efficacy of CPS on CVD-related markers. A systematic search in the Scopus, PubMed and ISI Web of Science databases were completed to identify relevant randomised, placebo-controlled trials (RCT) published up to November 2021. Mean Differences were pooled using a random-effects model, while publication bias, sensitivity analyses and heterogeneity were assessed using previously validated methods. Twelve RCT, comprising of a total of eleven measured markers, were selected for the quantitative analysis. Pooled data revealed that CPS significantly decreased fat mass (–1·21 kg; 95 % CI: −2·13, −0·29; I2 = 0·0 %; P = 0·010) and increased fat-free mass, based on body mass percentage (1·49 %; 95 % CI: 0·57, 2·42; I2 = 0·0 %; P = 0·002). Moreover, collagen peptide supplementation led to a significant decrease in serum LDL (–4·09 mg/dl; 95 % CI: −8·13, −0·04; I2 = 93·4 %; P = 0·048) and systolic blood pressure (SBP) (–5·04 mmHg; 95 % CI: −9·22, −0·85; I2 = 98·9 %; P = 0·018). Our analysis also indicated that CPS did not affect glycemic markers. Our outcomes indicate that CPS reduces fat mass, LDL and SBP while increasing fat-free mass. Future investigations with longer CPS duration are needed to expand on our results.

Type
Systematic Review and Meta-Analysis
Copyright
© The Author(s), 2022. Published by Cambridge University Press on behalf of The Nutrition Society

According to the WHO’s latest report, CVD claims the lives of 17·9 million people per year, representing the leading cause of mortality(Reference Roth, Abate and Abate1,Reference Kyu, Abate and Abate2) . It is well known that major risk factors for CVD such as obesity, diabetes, dyslipidaemia, hypertension and poor nutrition lead to a rise of atherosclerosis(Reference Timmis, Townsend and Gale3Reference Aminde and Veerman8), widely considered to be the chief component in cardiovascular pathologies. Consequently, prevention and treatment strategies targeting the above-mentioned risk factors are pivotal avenues to reduce costs and mortality associated with CVD.

A growing body of evidence has suggested that dietary interventions can effectively treat and prevent CVD(Reference Mozaffarian, Appel and Van Horn9). One of these interventions is collagen, a type of extracellular protein that makes up 25–30 % of the total protein in the human body(Reference Ohara, Matsumoto and Ito10). This compound is also highly available in fish and meat and thus is broadly used as a dietary supplement. However, given that collagen is not hydrolysed, its oral absorption is minimal, requiring hydroxylation before it becomes a physiologically accessible supplement(Reference Oesser, Adam and Babel11,Reference Sibilla, Godfrey and Brewer12) . In recent years, collagen peptide has been used in the food industry as a functional component. The physiological benefits of circulating dipeptides reflect the importance of consuming collagen peptides(Reference Shigemura, Kubomura and Sato13). Collagen peptides play essential biological roles, which include inhibiting the activity of angiotensin I-converting enzyme(Reference Qiu, Li and Weeber14), acting as signal messengers in anabolic cellular processes in cartilage, tendons and ligaments(Reference Schunck and Oesser15Reference McAlindon, Nuite and Krishnan17) and activating the mechanistic target of rapamycin signalling pathway(Reference Kitakaze, Sakamoto and Kitano18). In addition, collagen peptides may improve lipid metabolism and increase insulin sensitivity(Reference Wang, Xie and Pei19), reduce Cyp450, nitric oxide and prostaglandin I 2 while increasing bradykinin(Reference Koenig, Peterson and Jones20,Reference Marion-Letellier, Dechelotte and Iacucci21) . Over the last decade, evidence from various clinical investigations focused on the effects of CPS on CVD-related markers; however, this line of research produced conflicting findings. For instance, Zhu et al. (Reference Zhu, Li and Peng22) indicated that daily supplementation of 13 g of CPS for 12 weeks led to a significant reduction in LDL, HDL, TAG, total cholesterol, systolic blood pressure (SBP), diastolic blood pressure (DBP), fasting blood sugar and HbA1c in patients with Type 2 diabetes mellitus and primary hypertension. Zdzieblik et al. (Reference Zdzieblik, Jendricke and Oesser23) also reported that 15 g/d of specific CPS for 12 weeks significantly increased fat-free mass and decreased fat mass in middle-aged men. Moreover, Kouguchi et al. (Reference Kouguchi, Ohmori and Shimizu24) demonstrated that 2·9 g/d of chicken collagen hydrolysate supplementation for 12 weeks significantly decreased SBP and DBP in patients with mild hypertension. Conversely, another randomised controlled trial showed that collagen peptide supplementation (CPS) daily supplementation of 2 g of CPS for 12 weeks did not change body mass, fat mass, LDL, HDL, TAG, total cholesterol and fasting blood sugar in healthy volunteers(Reference Tak, Kim and Lee25). Moreover, Jendricke et al. (Reference Jendricke, Kohl and Centner26) indicated that 15 g/d of specific CPS for 12 weeks significantly increased fat-free mass but did not affect body mass and fat mass in active women. These discrepancies could be attributed to differences in study design, CPS dosage and/or participant characteristics (age, sex, health status, etc.), which highlights the importance of a comprehensive review on this topic.

There are currently no comprehensive reviews to systematically assess the effect of CPS on CVD-related markers (e.g. body mass, fat mass, fat-free mass, LDL, HDL, TAG, total cholesterol, blood pressure and fasting blood sugar), which denotes a gap in knowledge. Therefore, we completed the current systematic review and meta-analysis of randomized controlled trials to determine the effects of CPS on cardiovascular markers.

Materials and methods

Systematic search and study selection

This review followed the 2020 PRISMA guidelines(Reference Page, McKenzie and Bossuyt27). Description of population, intervention, comparator and outcome is displayed in Supplementary Table 1. A systematic search was conducted using Scopus, PubMed and ISI Web of Science databases. The medical subject headings were used in the search strategy to find the relevant studies. A systematic search was carried out by applying the reported strategy and the key terms displayed in Supplementary Table 2. The search process was conducted with no date or language restrictions. The databases were searched up to 1 November 2021. Database searches were completed in conjunction with searches of reference lists derived from individual studies. Two researchers (SM and HM) independently carried out the search strategy and screening. Any discrepancies were resolved via discussion with another author (CJ).

Eligibility criteria

Two authors (SM and HM) identified the eligible records by screening the titles, abstracts and full text of eligible studies. All randomised, placebo-controlled trials in humans (either parallel or cross-over designs) that assessed the efficacy of CPS on a range of CVD-related markers were included in the review. These markers included: anthropometric and body composition (body mass, fat mass, fat-free mass and BMI), lipid profile (LDL, HDL, TAG and total cholesterol), blood pressure (both SBP and DBP) and glycaemic indices including fasting blood sugar, HbA1c and insulin resistance. The exclusion of the studies was based on the following criteria: (1) clinical trials with an intervention duration of less than two weeks, (2) clinical trials without a placebo group, (3) animal studies and (4) those with insufficient data for the outcomes of interest.

Data extraction

The extracted study characteristics are presented in Table 1. If there was no available relevant data, corresponding authors were contacted to obtain any missing data. The data extraction procedure was conducted separately by two researchers (SM and FJ) to ensure reliability. Any disagreements were resolved by consensus and discussion.

Table 1. Main characteristics of included studies

BM, body mass; NR, not reported; FM, fat mass; FFM, fat-free mass; TC, total cholesterol; SBP, systolic blood pressure; DBP, diastolic blood pressure; FBS, fasting blood sugar.

Quality assessment of studies

The Cochrane Collaboration’s tool(Reference Higgins, Altman and Gøtzsche28) was used to evaluate the quality of studies. This tool involved judging the degree of bias (either high, low or unclear) for a series of items covering different domains of potential bias. Two authors (SM and HM) assessed the quality (risk of bias) of the eligible studies individually. The quality evaluation procedure has been presented in previous works(Reference Moradi, Ziaei and Foshati29,Reference Hadi, Ghaedi and Moradi30) .

Meta-analysis of data

To evaluate the effect size of the anthropometric, lipid profile, blood pressure and glycaemic markers, the mean and standard deviation (sd) changes were extracted from the intervention and placebo groups. Sub-group analyses relating to the mean study participant age (less than 50 years or more), sex (women or men), body mass (normal, overweight or obese), dosage (less than 10 g or 10 g and over), type of intervention (with or without exercise), trial duration (6 or 12 weeks) and participant health status (healthy or unhealthy) were performed to establish possible sources of heterogeneity. For the random-effects model, the DerSimonian and Laird method was utilised(Reference DerSimonian and Kacker31). Within-group changes were calculated by subtracting the baseline mean from the final mean value in each group. The sd of the mean difference was calculated using the following formula:

sd change = $$\sqrt {\left[ {{{\left( {{\rm{sd}}\;{\rm{baseline}}} \right)}^2}\; + \;{{\left( {{\rm{sd}}\;{\rm{final}}} \right)}^2}\; - \;\left( {2\; \times \;0\cdot8\; \times \;{\rm{sd}}\;{\rm{baseline}}\; \times \;{\rm{sd}}\;{\rm{final}}} \right)} \right]} $$ (Reference Wan, Wang and Liu32). For trials that reported only the standard error of the mean, sd was calculated using the following formula: sd = $SEM \times \sqrt n $ , where ‘n’ represented the number of participants in each group. Heterogeneity between studies was assessed by Cochrane’s Q test (significance at P < 0·100) and I 2 index. Publication bias was evaluated by the visual inspection of funnel plots and formal testing by Egger’s regression asymmetry and Begg’s rank correlation tests(Reference Begg and Mazumdar33,Reference Egger, Smith and Schneider34) . All statistical analyses were conducted using STATA software, version 16 (Stata Corp LP).

Outcomes were considered significant at P < 0·05.

Quality of evidence

The general certainty of evidence across randomised-controlled trials was rated using the Grading of Recommendations Assessment, Development, and Evaluation working group guidelines. Based on the related assessment criteria, the quality of evidence was categorised into four classes: high, moderate, low and very low(Reference Guyatt, Oxman and Vist35).

Results

Selection and identification of studies

The study selection flow is displayed in Fig. 1. Our systematic database search yielded a total of 453 records, of which 239 were screened. Two hundred twenty-six studies failed to meet the inclusion criteria and were excluded from qualitative and quantitative analyses. One study was excluded from the quantitative assessment due to lacking a comparative placebo group(Reference Giglio, Schincaglia and da Silva36). Ultimately, twelve randomised-controlled trials, collectively comprising eleven outcome measures, were selected for the quantitative analysis(Reference Zdzieblik, Jendricke and Oesser23,Reference Kouguchi, Ohmori and Shimizu24,Reference Jendricke, Kohl and Centner26,Reference Han and Kang37Reference Zhu, Li and Peng45) .

Fig. 1. Study selection flow chart

Characteristics of studies

In total, the twelve eligible studies included a total of 748 participants (359 individuals in the CPS and 389 in the placebo group) (Table 1). The mean age ranged from 25·4 ± 4·2 years to 72·2 ± 4·7 years. Studies were conducted in Germany(Reference Zdzieblik, Jendricke and Oesser23,Reference Jendricke, Kohl and Centner26,Reference Jendricke, Centner and Zdzieblik40,Reference Zdzieblik, Oesser and Baumstark44) , Korea(Reference Han and Kang37,Reference Koizumi, Inoue and Shimizu41,Reference Tak, Kim and Lee43) , Japan(Reference Kouguchi, Ohmori and Shimizu24,Reference Igase, Kohara and Okada38,Reference Ishii, Okada and Matsuoka39) , China(Reference Zhu, Li and Peng45) and India(Reference Kumar, Sugihara and Suzuki42). Included studies were performed in healthy(Reference Zdzieblik, Jendricke and Oesser23,Reference Jendricke, Kohl and Centner26,Reference Han and Kang37Reference Koizumi, Inoue and Shimizu41) and overweight(Reference Tak, Kim and Lee43) participants, as well as in individuals with type 2 diabetes mellitus(Reference Zhu, Li and Peng45), mild hypertension(Reference Kouguchi, Ohmori and Shimizu24), osteoarthritis(Reference Kumar, Sugihara and Suzuki42) and age-related sarcopenia(Reference Zdzieblik, Oesser and Baumstark44). All of the trials used a parallel arms design. The twelve eligible trials were published between 2008 and 2021. The dose of CPS ranged from 900 mg/d to 15 g/d, while the duration of the clinical trials ranged from 6 to 12 weeks.

Quality assessment of studies

According to the Cochrane risk of bias assessment, ten studies were ranked as high quality (demonstrating a low risk of bias on ≥ 3 domains)(Reference Zdzieblik, Jendricke and Oesser23,Reference Kouguchi, Ohmori and Shimizu24,Reference Jendricke, Kohl and Centner26,Reference Ishii, Okada and Matsuoka39Reference Zhu, Li and Peng45) and two as low quality (low risk of bias < 3 domains)(Reference Han and Kang37,Reference Igase, Kohara and Okada38) (Table 2).

Table 2. Quality assessment

+ shows low risk of bias; - shows high risk of bias; ? shows unclear risk of bias.

Meta-analysis of data

Effects of collagen peptide supplementation on anthropometric and body composition parameters

As illustrated in Fig. 2, pooled data from six studies revealed that the CPS did not significantly change body mass (–0·54 kg; 95 % CI: −2·13, 1·06; I 2 = 65·8 %; P = 0·510; n 6) compared with the placebo group. Furthermore, results indicated that CPS significantly decreased fat mass (–1·21 kg; 95 % CI: −2·13, −0·29; I 2 = 0·0 %; P = 0·010; n 4), but not body fat percentage (–0·65 %; 95 % CI: −1·76, 0·47; I 2 = 75·5 %; P = 0·256; n 5) compared with the placebo group. Sub-group analyses suggested that body fat percentage significantly changed after CPS in men (–1·66 %; 95 % CI: −2·48, −0·47; I 2 = 0·0 %; P = 0·006; n 2), obese or overweight participants (–1·22 %; 95 % CI: −2·39, −0·06; I 2 = 0·0 %; P = 0·039; n 2) and in those supplementing in conjunction to exercise training (–1·36 %; 95 % CI: −2·60, −0·12; I 2 = 0·0 %; P = 0·032; n 3) (Table 3). Further sub-group analyses involving age demonstrated that fat mass significantly decreased after CPS for participants only aged 50 years and over (–1·50 kg; 95 % CI: −3·07, −0·18; I 2 = 0·0 %; P = 0·020; n 2) (Table 3). The observed between-study heterogeneity for anthropometric and body composition parameters was attenuated by all sub-group analysis (Table 3).

Fig. 2. (a) Forest plot displaying weighted mean difference and 95 % CI for the effects of collagen peptide supplementation v. placebo on body mass.

Table 3. Sub-group meta-analyses for the effects of collagen peptide supplementation on metabolic parameters

WMD, weighted mean differences.

In addition, CPS significantly increased free fat mass based on body mass percentage (1·49 %; 95 % CI: 0·57, 2·42; I 2 = 0·0 %; P = 0·002; n 3), but not based on kg (weighted mean differences: 0·67 kg; 95 % CI: −0·26, 1·59; I 2 = 0·0 %; P = 0·441; n 4) compared with the placebo group (Fig. 2). Due to the lack of studies measuring fat-free mass, sub-group analyses were not feasible.

Effects of collagen peptide supplementation on lipid profile

Pooled data from studies assessing lipid profile demonstrated that CPS led to a significant decrease in LDL concentrations (–4·09 mg/dl; 95 % CI: –8·13, –0·04; I 2 = 93·4 %; P = 0·048; n 5), unlike HDL (3·13 mg/dl 95 % CI: –0·18, 6·43; I 2 = 64·8 %; P = 0·064; n 3), total cholesterol (–5·28 mg/dl; 95 % CI: –14·31, 3·75; I 2 = 98·1 %; P = 0·252; n 7) and TAG (–6·51 mg/dl; 95 % CI: –13·71, 0·70; I 2 = 92·1 %; P = 0·077; n 5), which remain unchanged compared with the placebo (Fig. 3). Sub-roup analyses according to dosage of intervention suggested that CPS significantly decreased LDL (–9·08 mg/dl; 95 % CI: –10·21, –7·94; I 2 = 93·4 %; P =< 0·001; n 2), total cholesterol (–23·39, mg/dl; 95 % CI: –26·60, –19·98; I 2 = 90·7 %; P =< 0·001; n 2), TAG (–12·84, mg/dl; 95 % CI: –15·45, –10·23; I 2 = 95·3 %; P =< 0·001; n 2) and significantly increased HDL (6·76 mg/dl; 95 % CI: 2·63, 10·89; I 2 = 0·0 %; P =< 0·001; n 2) following a 10 g and over CPS intervention, in contrast to a less than 10 g intervention (Table 3). The observed extreme between-study heterogeneity for lipid profile was attenuated by sub-group analysis based on the dosage of intervention (Table 3).

Fig. 3. (a)Forest plot displaying weighted mean difference and 95 % CI for the effects of collagen peptide supplementation v. placebo on low-density lipoprotein.

Effects of collagen peptide supplementation on blood pressure

CPS significantly decreased SBP (–5·04 mmHg; 95 % CI: −9·22, −0·85; I 2 = 98·9 %; P = 0·018; n 6), but not DBP (0·71 mmHg; 95 % CI: −0·25, 1·68; I 2 = 86·7 %; P = 0·148; n 6) compared with the placebo (Fig. 4). Sub-group analyses relating to the duration of intervention indicated that CPS significantly decreased SBP following a six-week intervention (–7·00 mmHg; 95 % CI: −13·80, −0·20; I 2 = 0·0 %; P = 0·044; n 2), in contrast to a 12-week intervention duration, which failed to produce a significant results (Table 3). Sub-group analyses according to dosage of intervention suggested that CPS significantly decreased SBP (–9·35, mmHg; 95 % CI: −11·28, −7·43; I 2 = 95·3 %; P =< 0·001; n 2) and significantly increased DBP (1·15 mmHg; 95 % CI: 0·25, 1·35; I 2 = 55·1 %; P =< 0·001; n 2) following a 10 g and over CPS intervention, in contrast to a less than 10 g intervention (Table 3). The observed extreme between-study heterogeneity for DBP was attenuated by sub-group analysis based on the trial duration and dosage of intervention (Table 3).

Fig. 4. (a) Forest plot displaying weighted mean difference and 95 % CI for the effects of collagen peptide supplementation v. placebo on systolic blood pressure.

Effects of collagen peptide supplementation on glycaemic indices

Our results failed to show a significant effect of CPS on glycaemic indices which included fasting blood sugar (–11·15, mg/dl; 95 % CI: –26·15, 3·84; I 2 = 99·8 %; P = 0·145; n 6) and HbA1c (–0·26 %; 95 % CI: –0·58, 0·06; I 2 = 98·3 %; P = 0·106; n 3) compared with the placebo group (Fig. 5). Sub-group analyses according to dosage of intervention suggested that CPS significantly decreased fasting blood sugar (–29·24, mg/dl; 95 % CI: –30·19, –28·28; I 2 = 0·0 %; P =< 0·001; n 2) and HbA1c (–0·39 %; 95 % CI: –0·58, –0·20; I 2 = 88·4 %; P =< 0·001; n 2) following a 10 g and over CPS intervention, in contrast to a less than 10 g intervention (Table 3). The observed extreme between-study heterogeneity for fasting blood sugar was attenuated by sub-group analysis based on the dosage of intervention (Table 3).

Fig. 5. (a) Forest plot displaying weighted mean difference and 95 % CI for the effects of collagen peptide supplementation v. placebo on fasting blood sugar.

Sensitivity analysis

Sensitivity analysis was performed by omitting each of the included studies. The results showed that the weighted mean differences was not changed significantly by omitting each of the studies. This indicated the meta-analysis results were stable and not sensitive to any one of the twelve studies (online Supplementary Fig. 1).

Publication bias

Moreover, no evidence of publication bias was detected regarding the efficacy of CPS on fat mass (P = 0·17, Begg’s test; P = 0·30, Egger’s test), fat-free mass based on body mass percentage (P = 0·69, Begg’s test; P = 0·60, Egger’s test), kilograms (P = 0·16, Begg’s test; P = 0·33, Egger’s test), LDL (p = 0·62, Begg’s test; P = 0·14, Egger’s test), HDL (P = 0·63, Begg’s test; P = 0·95, Egger’s test), TAG (P = 0·62, Begg’s test; P = 0·77, Egger’s test), total cholesterol (P = 0·65, Begg’s test; P = 0·095, Egger’s test), SBP (P = 0·57, Begg’s test; P = 0·39, Egger’s test), DBP (P = 0·57, Begg’s test; P = 0·37, Egger’s test), fasting blood sugar (P = 0·85, Begg’s test; P = 0·82, Egger’s test) and HbA1c (P = 0·12, Begg’s test; P = 0·16, Egger’s test). However, the Egger’s test results showed publication bias for body mass (P = 0·04) and body fat percentage (P = 0·002). These results were not confirmed by the Begg’s test for body mass (P = 0·34) and body fat percentage (P = 0·06).

Quality of evidence

To evaluate the quality of evidence for results, the GRADE guideline was utilised, which demonstrated the effect of fat mass (kg) to be of high quality. The evidence about fat-free mass (%), fat-free mass (kg) and SBP were downgraded to a moderate level. Based on the GRADE framework, body mass and LDL evidence were categorised as low quality. Finally, the fat mass (%), HDL, TAG, total cholesterol, DBP, fasting blood sugar and HbA1c were established as very low quality (Table 4).

Table 4. Grade profile of collagen peptide supplementation for cardiovascular risk factors scores in adults

TC, total cholestrol; FBG, fasting blood glucose; SBP, systolic blood pressure; DBP, diastolic blood pressure.

Discussion

The present review is the first to investigate the effects of CPS on cardiovascular disease-related markers, including blood pressure, anthropometric indices, lipid profile and glycaemic parameters. The pooled data that was derived from twelve randomised, controlled trials found that CPS (900 mg/d to 15 g/d), for a duration of 6 to 12 weeks, led to the significant reduction in fat mass, SBP, serum LDL concentrations and increased fat-free mass (measured by body mass percentage). Additional sub-group analyses demonstrated that CPS significantly decreased body fat percentage in men, obese or overweight participants, and when CPS was administered in combination with exercise.

A narrative review performed by Cicero et al. (Reference Cicero, Fogacci and Colletti46) revealed that bioactive peptides play potential roles in preventing and treating chronic diseases, including cancer and cardiovascular disorders. Indeed, several beneficial effects of ingesting collagen peptides are attributed to the physiological properties of circulating dipeptides, which are derived from collagen(Reference Shigemura, Kubomura and Sato47). This includes the potential ability to alleviate a range of cardiovascular complications, including heart failure(Reference Florea and Anand48), a condition preceded by high blood pressure in 70 % of cases(Reference McKee, Castelli and McNamara49).

Our outcomes revealed that CPS significantly decreased SBP. Previous studies have documented that CPS can help improve blood pressure in various pathways(Reference Zhu, Li and Peng22,Reference Kouguchi, Ohmori and Shimizu24,Reference Igase, Kohara and Okada38) . Saiga et al. (Reference Saiga, Iwai and Hayakawa50) reported that the hydrolysed collagen peptides extracted from the drumstick significantly suppressed high blood pressure by inhibiting angiotensin I-converting enzyme in spontaneously hypertensive rats. Moreover, Kouguchi et al. (Reference Kouguchi, Ohmori and Shimizu24) suggested that CPS may have a beneficial effect on the vascular system by activating certain vasodilating agents such as nitric oxide, which decrease arterial stiffness. Zhu et al. (Reference Zhu, Li and Peng22) also showed that marine collagen peptides supplementation ameliorated hypertension by activating peroxisome proliferator-activated receptors, leading to down-regulation of nuclear factor-kB pathway and reduced production of cytochrome P450 and prostaglandin I 2. Their results also showed an upregulation of bradykinin, which confirmed the angiotensin I-converting enzyme-inhibitory effect of collagen on blood pressure regulation(Reference Zhu, Li and Peng22). Importantly, it has been established that a reduction of 2 mmHg in SBP can lower coronary heart disease and stroke mortalities by 4 % and 6 %, respectively(Reference Chobanian, Bakris and Black51). Hence, our findings indicating an SBP-lowering effect (∼5 mmHg) of CPS support its clinical significance as a nutritional strategy for blood pressure improvement.

Elevated concentrations of blood lipids are well-established risk factors for cardiovascular diseases(Reference Nelson52). Our results revealed that CPS led to a significant decrease in LDL concentrations. Several investigations have assessed the potential blood lipid modulation by CPS therapy(Reference Zhu, Li and Peng22,Reference Tak, Kim and Lee25,Reference Han and Kang37,Reference Ishii, Okada and Matsuoka39) . Lee et al. (Reference Astre, Deleruyelle and Dortignac53) indicated that fish-derived collagen peptide might improve lipid metabolism and accumulation by a reduction in genes expression of CCAAT enhancer-binding protein-α and peroxisome proliferator-activated receptor-γ and adipocyte protein 2. Furthermore, Woo et al. (Reference Woo, Song and Kang54) suggested that collagen peptide skate skin-derived with dose-dependent manner suppressed hepatic lipid accumulation and reduced the lipid droplet size in the adipose tissue. Prior investigations also reported that CPS could lower plasma and hepatic lipid concentrations by suppressing the hepatic protein expression for fatty acid cholesterol synthesis, including sterol regulatory element-binding protein-1, sterol regulatory element-binding protein-2, fatty acid synthase, acetyl-CoA carboxylase and 3-hydroxy-3-methylglutaryl-CoA reductase(Reference Woo, Song and Kang54Reference Lammi, Aiello and Bollati56). Recently, animal research by Vijayan et al. (Reference Vijayan, Raman and Dara57) showed that fish-derived collagen peptide has anti-lipidaemic properties that are mediated by a down-regulation of 3-hydroxy-3-methylglutaryl-CoA reductase expression and up-regulation Lecithin–cholesterol acyltransferase. On other hand, CPS may enhance lipid breakdown by upregulating gene expression of PPAR-α, carnitine palmitoyltransferase 1 and synthesis of bile acid cytochrome P450 family 7 subfamily A member 1(Reference Woo, Song and Kang54Reference Lammi, Aiello and Bollati56), which play a role in the process of β-oxidation.

Interestingly, the results of the current study revealed that CPS significantly increased free fat mass and decreased fat mass. A potential mechanism for the improvement of fat-free mass and decrease in fat mass is that hydroxyprolyl-glycine levels can be elevated in blood after CPS(Reference Shigemura, Akaba and Kawashima58). These dipeptides seem to have signalling effects and promote C2C12 myoblast differentiation and increase myotube hypertrophy by activating the mTOR signalling pathway(Reference Kitakaze, Sakamoto and Kitano18). The prolyl-hydroxyproline, as other collagen peptides, contain dipeptides(Reference Shigemura, Akaba and Kawashima58,Reference Jimi, Koizumi and Center59) , accelerates muscle regeneration by inducing several myogenically differentiated cells, including myogenin-positive myoblasts and myoglobin-positive myocytes(Reference Jimi, Koizumi and Center59). Furthermore, it has been revealed that specific collagen peptides could act as signal messengers in anabolic cellular activities in ligaments, tendons and cartilage(Reference Schunck and Oesser15). It also might be helpful to improve pain symptoms and performance in activity-related joint discomforts(Reference Zdzieblik, Jendricke and Oesser23,Reference Zdzieblik, Oesser and Baumstark44) . Hence, CPS could enhance body composition and increase functional performance by improving joint-related disorders and their symptoms. Other potential mechanisms for the effects of CPS on improving body composition are its anti-inflammatory and antioxidant effects. The collagen peptides’ amino acids content, such as glycine, has been found to have powerful anti-inflammatory properties which can modulate muscle wasting in various models(Reference Koopman, Caldow and Ham60,Reference Sun, Wu and Ji61) . Additionally, Vijayan et al. (Reference Vijayan, Sreerekha and Dara62) suggested that CPS can attenuate oxidative stress by improving enzymatic antioxidant defense catalase and superoxide dismutase, leading to decreased levels of membrane lipid peroxidation. Chen et al. (Reference Chen, Liang and Wu63) also indicated that CPS enhanced the anti-inflammatory process by decreasing lipoxygenase activity and nitric oxide radicals. Moreover, it is possible that the decrease in fat mass in our results might be associated with higher resting energy expenditure affected by the significant increase in fat-free mass.

Several strengths can be derived from the present review. Firstly, the reviewed studies considered a body of literature that measured several important markers associated with cardiovascular disease. A majority of the included studies appeared to be of high quality, as distinguished by a validated risk of bias assessment. In addition, there were no signs of publication bias according to Begg’s and Egger’s tests. Despite these strengths, the current review consisted of some potential limitations. Participants were supplemented with different collagen peptide doses between studies and undertook supplementation for varying timeframes. The cardiovascular markers were only considered as secondary outcomes in some of the included trials, potentially impacting the measurement of these markers. There was high heterogeneity between enrolled study participants, with studies comprising of participants displaying diverse health statuses and ages. Finally, lifestyle modifications throughout the study periods were not controlled in some of the included studies. Lifestyle modifications such as dietary intake and physical activity may be confounding factors by influencing the effects of CPS on the assessed markers.

Conclusion

Taken together, this comprehensive review of collated data suggests that CPS may significantly decrease fat mass, fat-free mass (based on body mass percentage), SBP and serum LDL levels. Additional sub-group analyses revealed an important decline in fat mass among men, obese or overweight participants and those consuming collagen peptides while exercising. Additional long-term, well-designed randomised, placebo-controlled trials are recommended further to examine the impact of CPS on CVD-related markers and expand on our findings.

Acknowledgements

None.

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

SM and CJ designed this study. SM, MHK, and HM conducted the literature search. SM and FJ performed the statistical analysis and interpretation of the data. ZJ, FN, SPM, and SMG wrote the manuscript. RB, AW, and NT critically revised the manuscript. All authors approved the final version of the manuscript.

Supplementary material

For supplementary material/s referred to in this article, please visit https://doi.org/10.1017/S0007114522001301

References

Roth, GA, Abate, D, Abate, KH, et al. (2018) Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392, 17361788.CrossRefGoogle Scholar
Kyu, HH, Abate, D, Abate, KH, et al. (2018) Global, regional, and national disability-adjusted life-years (DALYs) for 359 diseases and injuries and healthy life expectancy (HALE) for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392, 18591922.CrossRefGoogle Scholar
Timmis, A, Townsend, N, Gale, CP, et al. (2020) European Society of Cardiology: cardiovascular disease statistics 2019. Eur Heart J 41, 1285.CrossRefGoogle Scholar
Dwivedi, AK, Dubey, P, Cistola, DP, et al. (2020) Association between obesity and cardiovascular outcomes: updated evidence from meta-analysis studies. Curr Cardiol Rep 22, 119.CrossRefGoogle ScholarPubMed
Kokubo, Y & Matsumoto, C (2016) Hypertension is a risk factor for several types of heart disease: review of prospective studies. Hypertens: Basic Res Clin Pract 956, 419426.Google Scholar
Sunkara, A & Raizner, A (2019) Supplemental vitamins and minerals for cardiovascular disease prevention and treatment. Methodist Debakey Cardiovasc J 15, 179.CrossRefGoogle ScholarPubMed
Kim, J, Hoang, T, Bu, SY, et al. (2020) Associations of dietary intake with cardiovascular disease, blood pressure, and lipid profile in the Korean population: a systematic review and meta-analysis. J Lipid Atheroscler 9, 205.CrossRefGoogle ScholarPubMed
Aminde, LN & Veerman, L (2016) Interventions for the prevention of cardiovascular diseases: a protocol for a systematic review of economic evaluations in low-income and middle-income countries. BMJ Open 6, e013668.CrossRefGoogle ScholarPubMed
Mozaffarian, D, Appel, LJ & Van Horn, L (2011) Components of a cardioprotective diet: new insights. Circulation 123, 28702891.CrossRefGoogle ScholarPubMed
Ohara, H, Matsumoto, H, Ito, K, et al. (2007) Comparison of quantity and structures of hydroxyproline-containing peptides in human blood after oral ingestion of gelatin hydrolysates from different sources. J Agric Food Chem 55, 15321535.CrossRefGoogle ScholarPubMed
Oesser, S, Adam, M, Babel, W, et al. (1999) Oral administration of 14C labeled gelatin hydrolysate leads to an accumulation of radioactivity in cartilage of mice (C57/BL). J Nutr 129, 18911895.CrossRefGoogle Scholar
Sibilla, S, Godfrey, M, Brewer, S, et al. (2015) An overview of the beneficial effects of hydrolysed collagen as a nutraceutical on skin properties: scientific background and clinical studies. Open Nutraceutical J 8, 2942.CrossRefGoogle Scholar
Shigemura, Y, Kubomura, D, Sato, Y, et al. (2014) Dose-dependent changes in the levels of free and peptide forms of hydroxyproline in human plasma after collagen hydrolysate ingestion. Food Chem 159, 328332.CrossRefGoogle ScholarPubMed
Qiu, S, Li, L, Weeber, EJ, et al. (2007) Ascorbate transport by primary cultured neurons and its role in neuronal function and protection against excitotoxicity. J Neurosci Res 85, 10461056.CrossRefGoogle ScholarPubMed
Schunck, M & Oesser, S (2013) Specific collagen peptides benefit the biosynthesis of matrix molecules of tendons and ligaments. J Int Soc Sports Nutr 10, 12.CrossRefGoogle Scholar
Oesser, S & Seifert, J (2003) Stimulation of type II collagen biosynthesis and secretion in bovine chondrocytes cultured with degraded collagen. Cell Tissue Res 311, 393399.CrossRefGoogle ScholarPubMed
McAlindon, T, Nuite, M, Krishnan, N, et al. (2011) Change in knee osteoarthritis cartilage detected by delayed gadolinium enhanced magnetic resonance imaging following treatment with collagen hydrolysate: a pilot randomized controlled trial. Osteoarthritis Cartilage 19, 399405.CrossRefGoogle ScholarPubMed
Kitakaze, T, Sakamoto, T, Kitano, T, et al. (2016) The collagen derived dipeptide hydroxyprolyl-glycine promotes C2C12 myoblast differentiation and myotube hypertrophy. Biochem Biophys Res Commun 478, 12921297.CrossRefGoogle ScholarPubMed
Wang, J, Xie, Y, Pei, X, et al. (2008) The lipid-lowering and antioxidative effects of marine collagen peptides. Zhonghua yu fang yi xue za zhi (Chinese J Prev Med) 42, 226230.Google ScholarPubMed
Koenig, RJ, Peterson, CM, Jones, RL, et al. (1976) Correlation of glucose regulation and hemoglobin AIc in diabetes mellitus. N Engl J Med 295, 417420.CrossRefGoogle ScholarPubMed
Marion-Letellier, R, Dechelotte, P, Iacucci, M, et al. (2009) Dietary modulation of peroxisome proliferator-activated receptor gamma. Gut 58, 586593.CrossRefGoogle ScholarPubMed
Zhu, C-F, Li, G-Z, Peng, H-B, et al. (2010) Therapeutic effects of marine collagen peptides on Chinese patients with type 2 diabetes mellitus and primary hypertension. Am J Med Sci 340, 360366.CrossRefGoogle ScholarPubMed
Zdzieblik, D, Jendricke, P, Oesser, S, et al. (2021) The influence of specific bioactive collagen peptides on body composition and muscle strength in middle-aged, untrained men: a randomized controlled trial. Int J Environ Res Public Health 18, 4837.CrossRefGoogle ScholarPubMed
Kouguchi, T, Ohmori, T, Shimizu, M, et al. (2013) Effects of a chicken collagen hydrolysate on the circulation system in subjects with mild hypertension or high-normal blood pressure. Biosci Biotechnol, Biochem 77, 691696.CrossRefGoogle ScholarPubMed
Tak, YJ, Kim, YJ, Lee, JG, et al. (2019) Effect of oral ingestion of low-molecular collagen peptides derived from skate (Raja Kenojei) skin on body fat in overweight adults: a randomized, double-blind, placebo-controlled trial. Mar Drugs 17, 157.CrossRefGoogle ScholarPubMed
Jendricke, P, Kohl, J, Centner, C, et al. (2020) Influence of specific collagen peptides and concurrent training on cardiometabolic parameters and performance indices in women: a randomized controlled trial. Front Nutr 7, 580918.CrossRefGoogle ScholarPubMed
Page, MJ, McKenzie, JE, Bossuyt, PM, et al. (2021) The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ 372, n71.CrossRefGoogle ScholarPubMed
Higgins, JP, Altman, DG, Gøtzsche, PC, et al. (2011) The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ 343, d5928.CrossRefGoogle ScholarPubMed
Moradi, S, Ziaei, R, Foshati, S, et al. (2019) Effects of spirulina supplementation on obesity: a systematic review and meta-analysis of randomized clinical trials. Complementary Ther Med 47, 102211.CrossRefGoogle ScholarPubMed
Hadi, A, Ghaedi, E, Moradi, S, et al. (2019) Effects of melatonin supplementation on blood pressure: a systematic review and meta-analysis of randomized controlled trials. Hormone Metab Res 51, 157164.Google ScholarPubMed
DerSimonian, R & Kacker, R (2007) Random-effects model for meta-analysis of clinical trials: an update. Contemp Clin Trials 28, 105114.CrossRefGoogle ScholarPubMed
Wan, X, Wang, W, Liu, J, et al. (2014) Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Meth 14, 135.CrossRefGoogle ScholarPubMed
Begg, CB & Mazumdar, M (1994) Operating characteristics of a rank correlation test for publication bias. Biom 50, 10881101.CrossRefGoogle Scholar
Egger, M, Smith, GD, Schneider, M, et al. (1997) Bias in meta-analysis detected by a simple, graphical test. BMJ 315, 629634.CrossRefGoogle ScholarPubMed
Guyatt, GH, Oxman, AD, Vist, GE, et al. (2008) GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 336, 924926.CrossRefGoogle ScholarPubMed
Giglio, BM, Schincaglia, RM, da Silva, AS, et al. (2019) Whey protein supplementation compared to collagen increases blood Nesfatin concentrations and decreases android fat in overweight women: a randomized double-blind study. Nutrients 11, 2051.CrossRefGoogle ScholarPubMed
Han, C-J & Kang, S-M (2008) The Effect of collagen supplementation from pork skin on serum collagen, serum sex steroid hormone, serum lipid and skin crack in Korean middle-aged women. Korean J Community Nutr 13, 912921.Google Scholar
Igase, M, Kohara, K, Okada, Y, et al. (2018) A double-blind, placebo-controlled, randomised clinical study of the effect of pork collagen peptide supplementation on atherosclerosis in healthy older individuals. Biosci Biotechnol, Biochem 82, 893895.CrossRefGoogle ScholarPubMed
Ishii, Y, Okada, Y, Matsuoka, S, et al. (2016) Effect of supplement containing Silybum marianum extract, soy extract, collagen peptide, bifidobacteria and apple extract on skin: a randomized placebo-controlled, double-blind, parallel group comparative clinical study. Glycative Stress Res 3, 156171.Google Scholar
Jendricke, P, Centner, C, Zdzieblik, D, et al. (2019) Specific collagen peptides in combination with resistance training improve body composition and regional muscle strength in premenopausal women: a randomized controlled trial. Nutrients 11, 892.CrossRefGoogle ScholarPubMed
Koizumi, S, Inoue, N, Shimizu, M, et al. (2018) Effects of dietary supplementation with fish scales-derived collagen peptides on skin parameters and condition: a randomized, placebo-controlled, double-blind study. Int J Pept Res Ther 24, 397402.CrossRefGoogle Scholar
Kumar, S, Sugihara, F, Suzuki, K, et al. (2015) A double-blind, placebo-controlled, randomised, clinical study on the effectiveness of collagen peptide on osteoarthritis. J Sci Food Agric 95, 702707.CrossRefGoogle ScholarPubMed
Tak, YJ, Kim, YJ, Lee, JG, et al. (2019) Effect of oral ingestion of low-molecular collagen peptides derived from skate (Raja Kenojei) skin on body fat in overweight adults: a randomized, double-blind, placebo-controlled trial. Mar Drugs 17, 157.CrossRefGoogle ScholarPubMed
Zdzieblik, D, Oesser, S, Baumstark, MW, et al. (2015) Collagen peptide supplementation in combination with resistance training improves body composition and increases muscle strength in elderly sarcopenic men: a randomised controlled trial. Br J Nutr 114, 12371245.CrossRefGoogle Scholar
Zhu, CF, Li, GZ, Peng, HB, et al. (2010) Therapeutic effects of marine collagen peptides on chinese patients with type 2 diabetes mellitus and primary hypertension. Am J Med Sci 340, 360366.CrossRefGoogle ScholarPubMed
Cicero, AFG, Fogacci, F & Colletti, A (2017) Potential role of bioactive peptides in prevention and treatment of chronic diseases: a narrative review. Br J Pharmacol 174, 13781394.CrossRefGoogle ScholarPubMed
Shigemura, Y, Kubomura, D, Sato, Y, et al. (2014) Dose-dependent changes in the levels of free and peptide forms of hydroxyproline in human plasma after collagen hydrolysate ingestion. Food Chem 159, 328332.CrossRefGoogle ScholarPubMed
Florea, VG & Anand, IS (2012) Troponin T and plasma collagen peptides in heart failure. Circulation: Heart Failure 5, 394397.Google ScholarPubMed
McKee, PA, Castelli, WP, McNamara, PM, et al. (1971) The natural history of congestive heart failure: the Framingham study. N Engl J Med 285, 14411446.CrossRefGoogle ScholarPubMed
Saiga, A, Iwai, K, Hayakawa, T, et al. (2008) Angiotensin I-Converting enzyme-inhibitory peptides obtained from chicken collagen hydrolysate. J Agric Food Chem 56, 95869591.CrossRefGoogle ScholarPubMed
Chobanian, AV, Bakris, GL, Black, HR, et al. (2003) The seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure: the JNC 7 report. JAMA 289, 25602571.CrossRefGoogle ScholarPubMed
Nelson, RH (2013) Hyperlipidemia as a risk factor for cardiovascular disease. Primary Care: Clin Offic Pract 40, 195211.CrossRefGoogle ScholarPubMed
Astre, G, Deleruyelle, S, Dortignac, A, et al. (2018) Diet-induced obesity and associated disorders are prevented by natural bioactive type 1 fish collagen peptides (Naticol®) treatment. J Physiol Biochem 74, 647654.CrossRefGoogle ScholarPubMed
Woo, M, Song, YO, Kang, K-H, et al. (2018) Anti-obesity effects of collagen peptide derived from skate (Raja kenojei) skin through regulation of lipid metabolism. Mar Drugs 16, 306.CrossRefGoogle ScholarPubMed
Lammi, C, Zanoni, C, Scigliuolo, GM, et al. (2014) Lupin peptides lower low-density lipoprotein (LDL) cholesterol through an up-regulation of the LDL receptor/sterol regulatory element binding protein 2 (SREBP2) pathway at HepG2 cell line. J Agric Food Chem 62, 71517159.CrossRefGoogle ScholarPubMed
Lammi, C, Aiello, G, Bollati, C, et al. (2021) Trans-Epithelial transport, metabolism and biological activity assessment of the multi-target lupin peptide LILPKHSDAD (P5) and its metabolite LPKHSDAD (P5-Met). Nutrients 13, 863.CrossRefGoogle ScholarPubMed
Vijayan, DK, Raman, SP, Dara, PK, et al. (2021) In vivo anti-lipidemic and antioxidant potential of collagen peptides obtained from great hammerhead shark skin waste. J Food Sci Technol 59 , 11401151.CrossRefGoogle ScholarPubMed
Shigemura, Y, Akaba, S, Kawashima, E, et al. (2011) Identification of a novel food-derived collagen peptide, hydroxyprolyl-glycine, in human peripheral blood by pre-column derivatisation with phenyl isothiocyanate. Food Chem 129, 10191024.CrossRefGoogle ScholarPubMed
Jimi, S, Koizumi, S, Center, NGIR, et al. (2021) Collagen-derived dipeptide pro-hyp administration accelerates muscle regenerative healing accompanied by less scarring after wounding on the abdominal wall in mice. Sci Rep 11, 112.CrossRefGoogle ScholarPubMed
Koopman, R, Caldow, MK, Ham, DJ, et al. (2017) Glycine metabolism in skeletal muscle: implications for metabolic homeostasis. Curr Opin Clin Nutr Metab Care 20, 237242.CrossRefGoogle ScholarPubMed
Sun, K, Wu, Z, Ji, Y, et al. (2016) Glycine regulates protein turnover by activating protein kinase B/mammalian target of rapamycin and by inhibiting MuRF1 and atrogin-1 gene expression in C2C12 myoblasts. J Nutr 146, 24612467.CrossRefGoogle ScholarPubMed
Vijayan, DK, Sreerekha, PR, Dara, PK, et al. (2021) Antioxidant defense of fish collagen peptides attenuates oxidative stress in gastric mucosa of experimentally ulcer-induced rats. Cell Stress Chaperones 27, 4554.CrossRefGoogle ScholarPubMed
Chen, Y-P, Liang, C-H, Wu, H-T, et al. (2018) Antioxidant and anti-inflammatory capacities of collagen peptides from milkfish (Chanos chanos) scales. J Food Sci Technol 55, 23102317.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Main characteristics of included studies

Figure 1

Fig. 1. Study selection flow chart

Figure 2

Table 2. Quality assessment

Figure 3

Fig. 2. (a) Forest plot displaying weighted mean difference and 95 % CI for the effects of collagen peptide supplementation v. placebo on body mass.

Figure 4

Table 3. Sub-group meta-analyses for the effects of collagen peptide supplementation on metabolic parameters

Figure 5

Fig. 3. (a)Forest plot displaying weighted mean difference and 95 % CI for the effects of collagen peptide supplementation v. placebo on low-density lipoprotein.

Figure 6

Fig. 4. (a) Forest plot displaying weighted mean difference and 95 % CI for the effects of collagen peptide supplementation v. placebo on systolic blood pressure.

Figure 7

Fig. 5. (a) Forest plot displaying weighted mean difference and 95 % CI for the effects of collagen peptide supplementation v. placebo on fasting blood sugar.

Figure 8

Table 4. Grade profile of collagen peptide supplementation for cardiovascular risk factors scores in adults

Supplementary material: File

Jalili et al. supplementary material

Table S1
Download Jalili et al. supplementary material(File)
File 14.3 KB
Supplementary material: File

Jalili et al. supplementary material

Table S2
Download Jalili et al. supplementary material(File)
File 15.3 KB
Supplementary material: File

Jalili et al. supplementary material

Jalili et al. supplementary material 3
Download Jalili et al. supplementary material(File)
File 98.4 KB
Supplementary material: File

Jalili et al. supplementary material

Jalili et al. supplementary material 4
Download Jalili et al. supplementary material(File)
File 69.3 KB