Next Article in Journal
Pharmacokinetic Analyses of Liposomal and Non-Liposomal Multivitamin/Mineral Formulations
Previous Article in Journal
Does Native Vitamin D Supplementation Have Pleiotropic Effects in Patients with End-Stage Kidney Disease? A Systematic Review of Randomized Trials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Effect of Lactobacillus sakei OK67 in Reducing Body and Visceral Fat in Lifestyle-Modified Overweight Individuals: A 12-Week, Randomized, Double-Blind, Placebo-Controlled Clinical Trial

1
DONGWHA Pharm Research Institute, 35-71, Topsil-ro, Giheung-gu, Yongin-si 17084, Republic of Korea
2
Department of Family Medicine, Seoul Paik Hospital, College of Medicine, Inje University, 9, Mareunnae-ro, Jung-gu, Seoul 04551, Republic of Korea
3
Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(13), 3074; https://doi.org/10.3390/nu15133074
Submission received: 19 April 2023 / Revised: 28 June 2023 / Accepted: 1 July 2023 / Published: 7 July 2023
(This article belongs to the Section Prebiotics and Probiotics)

Abstract

:
Obesity is a global health problem that affects the quality of life. It is a multidimensional chronic risk factor for major medical conditions, such as cardiovascular diseases, diabetes, and cancer. This clinical trial evaluated the efficacy of Lactobacillus sakei OK67 (DW2010), a lactic acid bacterium, in reducing body and visceral fat in overweight individuals (body mass index ≥25 kg/m2 and <30 kg/m2), aged 20–60 years. A total of 100 subjects placed in a lifestyle modification program were randomly assigned to receive either DW2010 (2.0 g/day, 1.0 × 1010 CFU) or a placebo for 12 weeks. The efficacy of DW2010 was evaluated by measuring body fat mass using dual-energy X-ray absorptiometry and visceral fat area using computed tomography. After 12 weeks, the change in body fat in the DW2010 group was not markedly different from that in the placebo group. However, visceral fat area decreased more in the DW2010 group than in the placebo group (p = 0.035). During the clinical trial, no major adverse events were reported. Moreover, no statistical differences were observed in the biochemical parameters of the DW2010 and placebo groups. Overall, we concluded that the intake of DW2010 for 12 weeks is safe and potentially reduces visceral fat in lifestyle-modified overweight subjects.

1. Introduction

Excessive fat accumulation occurs when calorie consumption exceeds energy expenditure. Specifically, obesity refers to excessive fat accumulation in adipose tissue, thereby leading to low-level inflammation, which in turn induces obesity [1]. Furthermore, fat accumulation around the abdominal viscera is considered to have greater negative health implications than obesity in general [2]. Moreover, abdominal obesity is a major risk factor for systemic inflammation, hyperlipidemia, insulin resistance, and cardiovascular disease [3]. Importantly, abdominal obesity aggravates insulin resistance and metabolic syndrome [4]. Abdominal obesity does not always occur in individuals with an elevated BMI, and individuals of normal weight can be metabolically obese if they have excessive visceral fat [5].
Previous investigations on the effects of high-fat diet (HFD)-induced obesity in humans and mice have demonstrated an increase in Firmicutes and a reduction in Bacteroidetes in the intestinal microflora (gut microbiota) [6]. Upon HFD consumption, the gut microbiota increase the production of lipopolysaccharide, a potent endotoxin found in the outer membrane of Gram-negative bacteria [7,8]. Lipopolysaccharides stimulate the host immune system via the Toll-like receptor 4 and associated mitogen-activated protein kinase signaling system, thereby activating the production of inflammatory cytokines, such as tumor necrosis factor-α and interleukin-1 beta, which further induce inflammatory responses [9]. In addition, lipopolysaccharides reduce the expression of intestinal tight-junction proteins, thereby increasing the absorption of lipopolysaccharides into the bloodstream. Lipopolysaccharides in the bloodstream stimulate the secretion of cytokines in immune cells, thereby leading to lipogenesis in adipose tissue [10].
Live lactic acid bacteria, which are Gram-positive bacteria that show acid resistance, are considered as typical beneficial bacteria in the gut. They are mainly found in yogurt, kimchi, and cheese, and they inhabit the female reproductive organs and intestines of humans and animals [11,12]. Among the beneficial bacteria, several Lactobacillus species are considered safe. Moreover, Lactobacillus isolated and identified from kimchi regulate host physiology [13,14,15] and improve diabetes by lowering blood sugar levels [16,17]. Specifically, Lactobacillus sakei OK67 was found to reduce the HFD-induced lipopolysaccharide levels in the intestine of mice. Furthermore, the oral administration of L. sakei OK67 suppressed fasting blood glucose, hyperglycemia, body weight, body fat, epididymal fat, and insulin levels in HFD-treated mice [18].
Therefore, in this study, a randomized, double-blind, placebo-controlled clinical trial was conducted to assess the effect of L. sakei OK67 in regulating body and visceral fat in lifestyle-modified overweight individuals.

2. Materials and Methods

2.1. Study Product

Lactobacillus sakei OK67 was manufactured by Kebijen Co., Ltd., Jeonju, Republic of Korea. The L. sakei OK67 product used in this clinical trial (DW2010) had the appearance of a milk-white powder. The placebo used in this study was made of a dextrin mixture and had a similar appearance, flavor, and weight as DW2010. Subjects ingested DW2010 or a placebo every day from June 2018 to November 2019 (2.0 g/day, 1.0 × 1010 CFU). The product used in this clinical trial was stored at temperatures between 0–10 °C.

2.2. Subjects

The clinical trial was conducted from June 2018 to November 2019 at Inje University Seoul Paik Hospital in compliance with the Helsinki Declaration and Korean Good Clinical Practice guidelines. The clinical trial was approved by the Institutional Review Board (IRB No. PAIK 2018-04-004), and registered at the Clinical Research Information Service of South Korea (Approval Number: KCT0003222). A total of 100 subjects were recruited after screening (physical and laboratory examinations). All subjects provided informed consent prior to the initiation of the clinical trial. The following selection criteria were used: (1) male and female subjects aged ≥20 and ≤60 years old; (2) those with a BMI of ≥25 kg/m2 and <30 kg/m2; (3) those who had not consumed alcohol within a month and could abstain from alcohol during the study period; and (4) those who consented to participate in this clinical trial and signed informed consent before the study initiation.
The exclusion criteria for clinical trial subjects were: (1) those who suffered from or were diagnosed with severe cerebrovascular disease (including cerebral infarction and hemorrhage) or heart disease (angina pectoris, myocardial infarction, heart failure, and arrhythmia) within the last six months; (2) patients with uncontrolled hypertension (blood pressure ≥ 160/100 mmHg); (3) persons with a fasting blood glucose level of ≥126 mg/dL, a random blood glucose level of ≥200 mg/dL, or a diabetic patient taking oral hypoglycemic agents or insulin; (4) persons with thyroid stimulating hormone levels of ≤0.1 µU/mL or ≥10 µU/mL; (5) those whose creatinine level was more than twice the normal upper limit; (6) those whose aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were more than three times the normal upper limit; (7) those who complained of severe gastrointestinal symptoms, such as heartburn and indigestion; (8) those who had been taking drugs that affect body weight (absorption inhibitors and appetite suppressants, supplements related to obesity improvement, psychiatric drugs, beta-blockers, diuretics, contraceptives, steroids, or female hormones) within the last month; (9) those who had continuously consumed probiotics (fermented milk) within the last week; (10) those who had lost ≥3 kg within four weeks of screening; (11) those who had participated in a commercial obesity program within the last three months; (12) those who had participated or planned to participate in other clinical studies within the month before clinical screening; (13) those who had been hospitalized or were undergoing drug treatment or rehabilitation treatment for disorders induced by alcohol use, heart disease, or central nervous system disorders identified through a medical history survey; (14) those who were deemed unable to exercise due to musculoskeletal disorders; (15) those who were pregnant or planning to become pregnant during this clinical study or lactating women; and (16) those deemed ineligible by the clinical trial manager for other reasons.

2.3. Study Design

The clinical trial was a 12-week, double-blind, randomized controlled trial to assess the efficacy and safety of DW2010 in regulating body and visceral fat in overweight individuals for weight loss purposes. After receiving written consent, subjects were randomly assigned to either the DW2010 or placebo group (n = 50/group). During the intervention period, all subjects were instructed not to take any dietary supplements or traditional medicines, engage in strenuous physical activity, or consume alcohol. Compliance with these regulations were monitored at each visit. Moreover, subjects were instructed to report any adverse events (AEs) or changes to their routine exercising, lifestyle, food habits, or other relevant habits. We referred to the paper evaluating body fat mass, which detailed the same major evaluation variables in this clinical study. According to the study by Nosaka et al. [19], the fat mass changed by −4.2 ± 2.8 kg from the baseline and decreased by 1.3 ± 0.8 kg compared to the placebo group. Considering this result, we calculated a sample size of 50 subjects per group, assuming a statistical power of 0.80, a two-tailed significance level of 0.05 and a dropout rate of 30%. After randomization into the DW2010 and placebo groups in a 1:1 ratio, the clinical trial was conducted for 12 weeks on 74 subjects, excluding dropouts.

2.4. Diet and Physical Activity Counseling

During the clinical trial, all subjects from both groups were instructed to reduce energy intake by 500 kcal/day. All subjects were instructed to burn 300 kcal or more daily by working, leisure, and moving, including physical activity. Dietary intake and physical activity were evaluated at an interim and last visit by means of meal and physical activity sheets. The average daily energy intake was analyzed using CAN-Pro 5.0, which is a software developed for nutritional evaluation considering the type, amount, and number of dietary intake, and the average daily calories consumed were analyzed using the Metabolic Equivalent for Task (MET) according to the type, time, and amount of exercise recorded in the physical activity sheets for at least 3 days (including 1 weekend day).
There was no statistically significant difference in energy intake and physical activity between the DW2010 group and the placebo group after 12 weeks (Table 1).

2.5. Efficacy Measurements

The primary outcome was change in body fat (g). Body fat mass, body fat percentage, and lean body mass were measured using dual-energy X-ray absorptiometry (DEXA; Prodigy® DEXA Lunar, GE Healthcare, Madison, WI, USA) at baseline (0 weeks) and 12 weeks. Changes in visceral fat, the secondary outcome, were assessed using computed tomography (CT), which could accurately determine the area of visceral fat in the abdomen. The area of visceral fat and subcutaneous fat of the abdomen was measured by the cross section between the fourth and fifth lumbar vertebrae. It was measured at baseline (0 weeks) and 12 weeks.

2.6. Safety Measurements

At each visit, subjects underwent routine biochemical assessments and measurements, including AST, ALT, albumin, alkaline phosphatase (ALP), creatine, blood urea nitrogen (BUN), uric acid, glucose, total bilirubin, γ-glutamyl transpeptidase (GTP), systolic and diastolic blood pressure, and pulse tests. Blood parameters were measured using XN-3000, UF-5000 (Sysmex, Norderstedt, Germany), and AU5800 (Beckman Coulter, CA, USA) blood analyzers. Furthermore, AEs were recorded and listed to determine their frequency and severity.

2.7. Statistical Analysis

Statistical analysis was performed using SAS® v. 9.4 (SAS Institute, Cary, NC, USA). A two-sample t-test, Wilcoxon rank sum test, chi-square test, or Fisher’s exact test was performed for demographic, efficacy, and safety evaluation. In addition, changes from the baseline in each parameter between the two groups were analyzed using analysis of covariance. The data obtained from this clinical trial were presented by calculating the mean and standard deviation with appropriate technical statistics, and the significance of the difference was verified at a p-value of <0.05 level with two-sided tests. All p-values are reported using four digits.

3. Results

3.1. Subject Baseline Characteristics

In total, 117 volunteers were initially selected to participate in this clinical trial, of whom 17 were excluded during screening. Therefore, the DW2010 and placebo groups comprised 50 subjects each. However, during the investigation, 15 subjects from the DW2010 group who withdrew consent or met exclusion criteria, or who exhibited low trial compliance (≤65%), were excluded from the clinical trial. Similarly, 11 subjects from the placebo group were removed from the assessment. Therefore, 74 subjects (DW2010 group, n = 35; placebo group, n = 39) were included in the final analysis (Figure 1). In the DW2010 group, the proportions of males and females were 31.4% and 68.6%, respectively. In the placebo group, males and females constituted 43.6% and 56.4%, respectively. The average ages in the DW2010 and placebo groups were 39.9 ± 9.7 y and 42.1 ± 1.0 y, respectively. There were no statistically significant differences between the two groups regarding sex distribution, age, or smoking habits. The average compliance levels in the DW2010 and placebo groups were 97.9 ± 9.5% and 96.7 ± 8.2%, respectively. No significant differences were found between the DW2010 and placebo groups in the baseline measurement values for demographics, anthropometry, body composition, laboratory tests, or daily energy intake. There were also no differences in daily energy intake between the two groups at 0 weeks. The general characteristics of the subjects are shown in Table 2.

3.2. Primary Outcome Assessment

DEXA scanning revealed that the DW2010 group subjects lost on average 764.17 ± 1513.1 g of body fat mass after 12 weeks of consumption, while the placebo group subjects lost on average 898.0 ± 1 571.3 g. However, anthropometry and body composition, including body fat mass, did not differ markedly between the groups at 0 or 12 weeks (Table 3, Figure 2).

3.3. Secondary Outcome Assessment

CT scanning revealed that the DW2010 group subjects lost on average 10.27 ± 15.17 cm2 of visceral fat area after 12 weeks of consumption, while the placebo group subjects lost on average 0.94 ± 21.51 cm2. Interestingly, the difference between the groups in visceral fat area after 12 weeks of consumption was statistically significant (p = 0.035) (Table 4, Figure 2). However, the subcutaneous fat area did not differ substantially between the groups at 12 weeks.

3.4. Safety Outcome Assessment

All safety parameters, including blood pressure, liver function tests, and renal function tests, were within normal ranges from the baseline until the end of the study, with no significant differences found between the DW2010 and placebo groups. To identify potential AEs, the clinical condition of each individual was evaluated at predetermined intervals. During the clinical trial period, no major AEs were reported. There were no noticeable differences in specific parameters between the DW2010 and placebo groups. All major parameters were measured as part of the safety evaluation. Mild AEs occurred in seven cases in the DW2010 group and in four cases in the placebo group. Mild AEs included dermatitis contact, rashes, and abdominal pain in the DW2010 group. However, there was no direct association between these mild AEs and DW2010 ingestion (Table 5). There were no withdrawals from the trial due to serious AEs. The safety parameters and respective analyses are described in Table 6.

4. Discussion

This clinical trial examined changes in the body and visceral fat of overweight individuals who received DW2010 for 12 weeks. We observed no appreciable differences in body fat (primary outcome) between the groups. However, the visceral fat (secondary outcome) of the DW2010 group differed substantially from that of the placebo group at 12 weeks. Lactobacillus sakei OK67 is a typical Gram-positive, anaerobic, acid-resistant lactic acid bacterium typically found in large amounts in kimchi, which is a traditional fermented food. It inhibits gut microbiota lipopolysaccharide production, and alleviates lipopolysaccharide-induced inflammation in mice [18]. In vitro, kimchi-isolated L. sakei proBio65 exhibited immunomodulatory properties by elevating Foxp3 protein expression [20]. Similarly, in a collagen-induced arthritis mouse model, L. sakei exerted an anti-inflammatory effect by regulating Th17 and regulatory B-cell differentiation [21]. In addition, L. sakei WIKIM30 was found to alleviate atopic dermatitis by regulating intestinal flora and regulatory T cells [22]. Lactobacillus sakei ADM14 was found to exert an anti-obesity effect in an obese mouse model by causing changes in the intestinal flora [23].
In mice with HFD-induced obesity, DW2010 induced AMP-activated protein kinase activation in the intestine and liver by inhibiting the gut microbiota and endotoxin production; furthermore, it suppressed body weight, hyperglycemia, epididymal fat, and insulin levels [18]. A previous study of HFD-fed mice reported anti-obesity outcomes related to inflammatory state modulation, lipogenic gene downregulation, and adipocyte number reduction [24]. Epicardial fat and abdominal visceral fat surround the myocardium and gastrointestinal organs, respectively, and both are considered to be visceral fat [25]. These interesting observations in mice are consistent with the outcomes of this study, thereby reflecting the high degree of similarity between animal and human investigations.
Visceral fat triggers changes in the blood lipid profile, inflammation, and insulin resistance. Moreover, obesity is highly associated with visceral fat, which is recognized as the most direct risk factor for the development of obesity-related metabolic disorders [26,27]. As observed in this clinical trial, visceral fat was considerably regulated upon the ingestion of DW2010. Thus, the substantial reduction in visceral fat may have contributed positively to the regulation of obesity in participants. Dietary modifications, specific dietary supplements, and drugs can modify body fat mass in a very short time, although the effects may be temporary. However, reducing accumulated abdominal visceral fat could be challenging.

5. Conclusions

In this clinical trial, visceral fat was substantially reduced in the DW2010 group for 12 weeks, thus indicating the significance of DW2010 in visceral fat reduction. Moreover, visceral fat reduction could reduce the risk factors associated with obesity and metabolic syndrome. DW2010 is clinically safe and has therapeutic potential for treating obesity by regulating visceral fat. Subsequent studies are required to investigate the long-term visceral fat effects of DW2010 on human health.

Author Contributions

Conceptualization, Y.-G.C. and Y.-H.H.; methodology, Y.-G.C. and Y.-H.H.; investigation, Y.-H.H., Y.-G.C. and D.-H.K.; writing—original draft preparation, S.-J.O.; writing—review and editing, Y.-G.C., Y.-H.H., D.-H.K. and S.-J.O.; supervision, Y.-H.H., Y.-G.C. and D.-H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Korean Institute of Planning and Evaluation for Technology in Food, Agriculture and Forestry (IPET; grant number 816003033CG000), and it was also funded by the Ministry of Agriculture, Food, and Rural Affairs (MAFRA) of the Republic of Korea.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and the Korean Good Clinical Practice guidelines, and it was approved by the Institutional Review Board of Inje University Seoul Paik Hospital (IRB No. PAIK 2018-04-004; 2018-05-23).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study. Written informed consent has been obtained from the patients to publish this paper.

Data Availability Statement

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Acknowledgments

We would like to thank Hong-Sig Sin of CHEBIGEN Inc. for manufacturing DW2010, as well as the staff of Inje University Seoul Paik Hospital, at the Department of Family Medicine, for their help and advice.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Flanagan, A.M.; Brown, J.L.; Santiago, C.A.; Aad, P.Y.; Spicer, L.J.; Spicer, M.T. High-fat diets promote insulin resistance through cytokine gene expression in growing female rats. J. Nutr. Biochem. 2008, 19, 505–513. [Google Scholar] [CrossRef]
  2. Shuster, A.; Patlas, M.; Pinthus, J.H.; Mourtzakis, M. The clinical importance of visceral adiposity: A critical review of methods for visceral adipose tissue analysis. Br. J. Radiol. 2012, 85, 1009. [Google Scholar] [CrossRef] [Green Version]
  3. Pedersen, B.K.; Saltin, B. Exercise as medicine—Evidence for prescribing exercise as therapy in 26 different chronic diseases. Scand. J. Med. Sci. Sports 2015, 25, 1–72. [Google Scholar] [CrossRef] [Green Version]
  4. Björntorp, P. Metabolic implications of body fat distribution. Diabetes Care 1991, 14, 1132–1143. [Google Scholar] [CrossRef] [PubMed]
  5. Ruderman, N.B.; Schneider, S.H.; Berchtold, P. The “metabolically-obese,” normal-weight individual. Am. J. Clin. Nutr. 1981, 34, 1617–1621. [Google Scholar] [CrossRef] [PubMed]
  6. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef] [PubMed]
  7. Boroni Moreira, A.P.; Salles Texeira, T.F.; Barbosa Ferreira, A.; Do Carmo Gouveia Peluzio, M.; De Cássia Gonçalves Alfenas, R. Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. Br. J. Nutr. 2012, 108, 801–809. [Google Scholar] [CrossRef]
  8. Kim, K.-A.; Gu, W.; Lee, I.-A.; Joh, E.-H.; Kim, D.-H. High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS ONE 2012, 7, e47713. [Google Scholar] [CrossRef]
  9. Guha, M.; Mackman, N. LPS induction of gene expression in human monocytes. Cell Signal 2001, 13, 85–94. [Google Scholar] [CrossRef]
  10. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet–induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
  11. Behnsen, J.; Deriu, E.; Sassone-Corsi, M.; Raffatellu, M. Probiotics: Properties, examples, and specific applications. Cold Spring Harb. Perspect. Med. 2013, 3, a010074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Jang, S.-E.; Han, M.J.; Kim, S.-Y.; Kim, D.-H. Lactobacillus plantarum CLP-0611 ameliorates colitis in mice by polarizing M1 to M2-like macrophages. Int. Immunopharmacol. 2014, 21, 186–192. [Google Scholar] [CrossRef] [PubMed]
  13. Jang, S.-E.; Hyun, Y.-J.; Trinh, H.-T.; Han, M.J.; Kim, D.-H. Anti-scratching behavioral effect of Lactobacillus plantarum PM008 isolated from kimchi in mice. Immunopharmacol. Immunotoxicol. 2011, 33, 539–544. [Google Scholar] [CrossRef] [PubMed]
  14. Joo, H.-M.; Kim, K.-A.; Myoung, K.-S.; Ahn, Y.-T.; Lee, J.-H.; Huh, C.-S.; Han, M.J.; Kim, D.-H. Lactobacillus helveticus HY7801 ameliorates vulvovaginal candidiasis in mice by inhibiting fungal growth and NF-κB activation. Int. Immunopharmacol. 2012, 14, 39–46. [Google Scholar] [CrossRef]
  15. Woo, J.-Y.; Gu, W.; Kim, K.-A.; Jang, S.-E.; Han, M.J.; Kim, D.-H. Lactobacillus pentosus var. plantarum C29 ameliorates memory impairment and inflammaging in a D-galactose-induced accelerated aging mouse model. Anaerobe 2014, 27, 22–26. [Google Scholar] [CrossRef] [PubMed]
  16. Bejar, W.; Hamden, K.; Salah, R.B.; Chouayekh, H. Lactobacillus plantarum TN627 significantly reduces complications of alloxan-induced diabetes in rats. Anaerobe 2013, 24, 4–11. [Google Scholar] [CrossRef]
  17. Honda, K.; Moto, M.; Uchida, N.; He, F.; Hashizume, N. Anti-diabetic effects of lactic acid bacteria in normal and type 2 diabetic mice. J. Clin. Biochem. Nutr. 2012, 51, 96–101. [Google Scholar] [CrossRef] [Green Version]
  18. Lim, S.-M.; Jeong, J.-J.; Woo, K.H.; Han, M.J.; Kim, D.-H. Lactobacillus sakei OK67 ameliorates high-fat diet–induced blood glucose intolerance and obesity in mice by inhibiting gut microbiota lipopolysaccharide production and inducing colon tight junction protein expression. Nutr. Res. 2016, 36, 337–348. [Google Scholar] [CrossRef]
  19. Nosaka, N.; Maki, H.; Suzuki, Y.; Haruna, H.; Ohara, A.; Kasai, M.; Tsuji, H.; Aoyama, T.; Okazaki, M.; Igarashi, O.; et al. Effects of margarine containing medium-chain triacylglycerols on body fat reduction in humans. J. Atheroscler. Thromb. 2003, 10, 290–298. [Google Scholar] [CrossRef] [Green Version]
  20. Lim, J.; Seo, B.J.; Kim, J.; Chae, C.-S.; Im, S.-H.; Hahn, Y.-S.; Park, Y.H. Characteristics of immunomodulation by a Lactobacillus sakei proBio65 isolated from kimchi. Microbiol. Biotechnol. Lett. 2011, 39, 313–316. [Google Scholar]
  21. Jhun, J.; Min, H.K.; Ryu, J.; Lee, S.-Y.; Ryu, J.-G.; Choi, J.W.; Na, H.S.; Lee, S.Y.; Jung, Y.; Park, S.-J.; et al. Lactobacillus sakei suppresses collagen-induced arthritis and modulates the differentiation of T helper 17 cells and regulatory B cells. J. Transl. Med. 2020, 18, 317. [Google Scholar] [CrossRef] [PubMed]
  22. Kwon, M.-S.; Lim, S.K.; Jang, J.-Y.; Lee, J.; Park, H.K.; Kim, N.; Yun, M.; Shin, M.-Y.; Jo, H.E.; Oh, Y.J.; et al. Lactobacillus sakei WIKIM30 ameliorates atopic dermatitis-like skin lesions by inducing regulatory T cells and altering gut microbiota structure in mice. Front. Immunol. 2018, 9, 1905. [Google Scholar] [CrossRef] [Green Version]
  23. Won, S.-M.; Chen, S.; Lee, S.Y.; Lee, K.E.; Park, K.W.; Yoon, J.-H. Lactobacillus sakei ADM14 induces anti-obesity effects and changes in gut microbiome in high-fat diet-induced obese mice. Nutrients 2020, 12, 3703. [Google Scholar] [CrossRef] [PubMed]
  24. Choi, W.J.; Dong, H.J.; Jeong, H.U.; Ryu, D.W.; Song, S.M.; Kim, Y.R.; Jung, H.H.; Kim, T.H.; Kim, Y.-H. Lactobacillus plantarum LMT1-48 exerts anti-obesity effect in high-fat diet-induced obese mice by regulating expression of lipogenic genes. Sci. Rep. 2020, 10, 869. [Google Scholar] [CrossRef] [Green Version]
  25. Bertaso, A.G.; Bertol, D.; Duncan, B.B.; Foppa, M. Epicardial fat: Definition, measurements and systematic review of main outcomes. Arq. Bras. Cardiol. 2013, 101, e18–e28. [Google Scholar] [CrossRef] [PubMed]
  26. Després, J.-P.; Lemieux, I. Abdominal obesity and metabolic syndrome. Nature 2006, 444, 881–887. [Google Scholar] [CrossRef]
  27. Grundy, S.M. Obesity, metabolic syndrome, and cardiovascular disease. J. Clin. Endocrinol. Metab. 2004, 89, 2595–2600. [Google Scholar] [CrossRef]
Figure 1. Flow diagram showing the selection and distribution of subjects in the clinical trial of the effects of Lactobacillus sakei OK67 (DW2010) on body and visceral fat reduction in overweight individuals.
Figure 1. Flow diagram showing the selection and distribution of subjects in the clinical trial of the effects of Lactobacillus sakei OK67 (DW2010) on body and visceral fat reduction in overweight individuals.
Nutrients 15 03074 g001
Figure 2. Mean changes to (a) body fat mass and (b) visceral fat area caused by Lactobacillus sakei OK67 (DW2010) after 12 weeks. Negative values correspond to decreases from week 0 to week 12.
Figure 2. Mean changes to (a) body fat mass and (b) visceral fat area caused by Lactobacillus sakei OK67 (DW2010) after 12 weeks. Negative values correspond to decreases from week 0 to week 12.
Nutrients 15 03074 g002
Table 1. Changes in energy intake and physical activity during the clinical trial.
Table 1. Changes in energy intake and physical activity during the clinical trial.
DW2010 Group
(n = 35)
Placebo Group
(n = 39)
p-Value
Mean ± SDMean ± SD
Energy intake
(kcal/day)
0 weeks1824.71 ± 700.081757.08 ± 437.670.6248 *
12 weeks1583.30 ± 432.141393.53 ± 399.05
Change from baseline−241.40 ± 749.42−349.56 ± 455.110.4653 *
Physical activity (kcal/day)
0 weeks290.30 ± 192.82347.44 ± 261.720.3541 *
12 weeks286.81 ± 225.64287.45 ± 163.76
Change from baseline−3.49 ± 197.77−59.99 ± 215.940.2200 *
* Comparison between the DW2010 and placebo groups; two-sample t-test; SD, standard deviation.
Table 2. Baseline demographic characteristics and body composition of the subjects.
Table 2. Baseline demographic characteristics and body composition of the subjects.
VariablesDW2010 Group
(n = 35)
Placebo Group
(n = 39)
p-Value
Sex
No. (%)
Male11 (31.4)17 (43.6)0.4476
Female24 (68.6)22 (56.4)
Age
(years)
Mean ± SD39.9 ± 9.742.1 ± 10.00.3596 *
Min, Max21.0, 59.024.0, 59.0
Smoking status
No. (%)
Non-smoking32 (91.4)36 (92.3)0.9598
Ex-Smoker1 (2.9)0 (0.00)
Smoker2 (5.7)3 (7.7)
Smoking amountAmong smokers, number of cigarettes/day0.7671 **
Mean ± SD10.0 ± 7.17.0 ± 5.2
Min, Max5.0, 15.01.0, 10.0
Smoking periodAmong smokers, years0.8215 *
Mean ± SD15.0 ± 7.117.7 ± 13.7
Min, Max10.0, 20.03.0, 30.0
Weight (kg)Mean ± SD74.86 ± 9.3073.25 ± 10.460.7047 **
BMI (kg/m2)27.20 ± 1.5327.09 ± 1.560.7534 **
BFM (g)26,593 ± 496725,281 ± 47430.2492 *
LBM (g)44,983 ± 85144,917 ± 9630.8795 **
WC (cm)92.74 ± 5.8691.29 ± 7.100.3452 *
T-Chol (mg/dL)205.31 ± 43.25210.21 ± 36.970.6017 **
TG (mg/dL)131.77 ± 86.30108.46 ± 56.620.2511 **
HDL-C (mg/dL)50.54 ± 10.5154.08 ± 9.840.1396 **
LDL-C (mg/dL)128.40 ± 37.53134.41 ± 33.610.4697 **
AST (IU/L)25.46 ± 7.3625.74 ± 10.570.5829 **
ALT (IU/L)25.58 ± 14.6427.00 ± 22.490.5669 **
Albumin (g/dL)4.48 ± 0.294.48 ± 0.280.9439 *
ALP (IU/L)59.82 ± 13.0759.26 ± 13.740.8350 *
Creatinine (mg/dL)0.74 ± 0.160.75 ± 0.160.4626 **
BUN (mg/dL)13.02 ± 3.6612.92 ± 3.190.9778 **
Uric acid (mg/dL)5.53 ± 1.345.83 ± 1.450.3662 **
γ-GTP (IU/L)23.04 ± 15.7028.30 ± 26.830.5668 **
SBP (mmHg)126.28 ± 14.59125.88 ± 13.530.8873 *
DBP (mmHg)78.62 ± 10.2077.80 ± 8.020.7745 **
Energy intake (Kcal/day) 1824.7 ± 700.11757.1 ± 437.70.6248 *
* Two-sample t-test; ** Wilcoxon rank sum test; Chi-square test; Fisher’s exact test; SD: standard deviation; BFM: body fat mass; LBM: lean body mass; WC: waist circumference; BMI: body mass index; T-Chol: total cholesterol; TG: triglyceride; HDL-C: high-density lipoprotein cholesterol; LDL-C: low-density lipoprotein cholesterol; AST: aspartate aminotransferase; ALT: alanine aminotransferase; ALP: alkaline phosphatase; BUN: blood urea nitrogen; GTP: glutamyl transpeptidase; SBP: systolic blood pressure; DBP: diastolic blood pressure.
Table 3. Effects of Lactobacillus sakei OK67 (DW2010) on body fat mass (g) measured using dual-energy X-ray absorptiometry (DEXA).
Table 3. Effects of Lactobacillus sakei OK67 (DW2010) on body fat mass (g) measured using dual-energy X-ray absorptiometry (DEXA).
DW2010 Group (n = 35)Placebo Group (n = 39)p-Value
Mean ± SDMean ± SD
Body fat mass (g)
0 weeks26,593 ± 496725,281 ± 47430.2492 *
12 weeks25,829 ± 487624,383 ± 4661
Change from baseline−764.17 ± 1513−897.97 ± 15710.5333 $
Body fat percentage (%)
0 weeks37.39 ± 6.5436.44 ± 6.980.5523 *
12 weeks37.09 ± 6.6535.86 ± 7.06
Change from baseline−0.29 ± 1.68−0.58 ±1.450.4200 $
Lean body mass (g)
0 weeks44,983 ± 850544,917 ± 96260.8795 *
12 weeks44,318 ± 868144,402 ± 9426
Change from baseline−665.34 ± 1390−515.46 ± 11400.6153 $
Body weight (kg)
0 weeks74.86 ± 9.3073.25 ± 10.460.7047 *
12 weeks73.67 ± 9.3072.37 ± 10.09
Change from baseline−1.19 ± 1.66−0.88 ± 1.880.5333 $
BMI (kg/m2)
0 weeks27.20 ± 1.5327.09 ± 1.560.7534 **
12 weeks26.77 ± 1.6126.79 ± 1.74
Change from baseline−0.43 ± 0.61−0.30 ± 0.690.3889 $
WC (cm)
0 weeks92.74 ± 5.8691.29 ± 7.100.3452 *
12 weeks91.64 ± 5.4990.27 ± 6.97
Change from baseline−1.10 ± 2.44−1.05 ± 2.710.8860 $
* Comparing the DW2010 and placebo groups using a two-sample t-test; ** Wilcoxon rank sum test; $ Comparing the DW2010 and placebo groups using analysis of covariance with an adjusted baseline.
Table 4. Effect of Lactobacillus sakei OK67 (DW2010) on visceral fat area (cm2) measured using computed tomography (CT).
Table 4. Effect of Lactobacillus sakei OK67 (DW2010) on visceral fat area (cm2) measured using computed tomography (CT).
DW2010 Group
(n = 35)
Placebo Group
(n = 39)
p-Value
Mean ± SDMean ± SD
Visceral fat area (cm2)
0 weeks119.43 ± 42.99117.95 ± 49.700.6184 **
12 weeks109.15 ± 44.12117.01 ± 47.90
Change from baseline−10.27 ± 15.17−0.94 ± 21.510.0354 $
Subcutaneous fat area (cm2)
0 weeks234.37 ± 56.22225.05 ± 56.470.2960 **
12 weeks219.74 ± 55.54214.64 ± 53.01
Change from baseline−14.63 ± 22.68−10.41 ± 32.390.6683 $
** Comparing the DW2010 and placebo groups using Wilcoxon rank sum test; $ Comparing the DW2010 and placebo groups using analysis of covariance with an adjusted baseline.
Table 5. AE occurrences and respective analyses.
Table 5. AE occurrences and respective analyses.
DW2010 Group
(n = 50)
Placebo Group
(n = 50)
Total
(N = 100)
p-Value
NIncidence (%)NIncidence (%)NIncidence (%)
Mild714481111-
Moderate000000
Severe000000
Relationship with the test article0000000.9207
 p-value for Fisher’s exact test.
Table 6. Safety parameters in the groups ingesting Lactobacillus sakei OK67 (DW2010) or the placebo.
Table 6. Safety parameters in the groups ingesting Lactobacillus sakei OK67 (DW2010) or the placebo.
DW2010 Group
(n = 50)
Placebo Group
(n = 50)
p-Value
Mean ± SDMean ± SD
AST (GOT) (IU/L)
0 weeks25.46 ± 7.3625.74 ± 10.570.5829 **
12 weeks25.74 ± 8.0126.95 ± 13.000.1385 **
ALT (GPT) (IU/L)
0 weeks25.58 ± 14.6427.00 ± 22.490.5669 **
12 weeks23.77 ± 12.9027.81 ± 22.400.6218 **
Albumin (g/dL)
0 weeks4.48 ± 0.294.48 ± 0.280.9439 *
12 weeks4.50 ± 0.234.54 ± 0.270.8415 **
ALP (IU/L)
0 weeks59.8 ± 13.159.3 ± 13.70.8350 *
12 weeks62.4 ± 15.261.2 ± 14.40.8052 **
Creatinine (mg/dL)
0 weeks0.74 ± 0.160.75 ± 0.160.4626 **
12 weeks0.74 ± 0.190.76 ± 0.170.0768 *
BUN (mg/dL)
0 weeks13.02 ± 3.6612.92 ± 3.190.9778 **
12 weeks12.88 ± 3.7713.35 ± 3.250.9509 *
Uric acid (mg/dL)
0 weeks5.53 ± 1.345.83 ± 1.450.3662 **
12 weeks5.40 ± 1.305.69 ± 1.450.1493 *
Glucose (mg/dL)
0 weeks95.98 ± 11.6891.60 ± 9.580.2460 *
12 weeks95.98 ± 17.8592.16 ± 7.960.1900 **
Total Bilirubin (mg/dL)
0 weeks0.71 ± 0.270.82 ± 0.330.0858 **
12 weeks0.73 ± 0.340.77 ± 0.290.2027 **
γ-GTP (IU/L)
0 weeks23.04 ± 15.7028.30 ± 26.830.5668 **
12 weeks20.72 ± 14.5428.14 ± 26.350.8181 **
SBP (mmHg)
0 weeks126.28 ± 14.59125.88 ± 13.530.8873 *
12 weeks128.58 ± 12.60127.57 ± 11.530.8574 *
DBP (mmHg)
0 weeks78.62 ± 10.2077.80 ± 8.020.7745 **
12 weeks79.02 ± 9.6078.30 ± 8.330.6530 *
* Comparing the DW2010 and placebo groups using two-sample t-test. ** Comparing the DW2010 and placebo groups using Wilcoxon rank sum test. AST: aspartate aminotransferase; ALT: alanine aminotransferase; GOT: glutamic oxaloacetic transaminase; GPT: glutamic pyruvic transaminase; ALP: alkaline phosphatase; BUN: blood urea nitrogen; GTP: glutamyl transpeptidase; SBP: systolic blood pressure; DBP: diastolic blood pressure.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oh, S.-J.; Cho, Y.-G.; Kim, D.-H.; Hwang, Y.-H. Effect of Lactobacillus sakei OK67 in Reducing Body and Visceral Fat in Lifestyle-Modified Overweight Individuals: A 12-Week, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2023, 15, 3074. https://doi.org/10.3390/nu15133074

AMA Style

Oh S-J, Cho Y-G, Kim D-H, Hwang Y-H. Effect of Lactobacillus sakei OK67 in Reducing Body and Visceral Fat in Lifestyle-Modified Overweight Individuals: A 12-Week, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients. 2023; 15(13):3074. https://doi.org/10.3390/nu15133074

Chicago/Turabian Style

Oh, Seong-Jun, Young-Gyu Cho, Dong-Hyun Kim, and Yun-Ha Hwang. 2023. "Effect of Lactobacillus sakei OK67 in Reducing Body and Visceral Fat in Lifestyle-Modified Overweight Individuals: A 12-Week, Randomized, Double-Blind, Placebo-Controlled Clinical Trial" Nutrients 15, no. 13: 3074. https://doi.org/10.3390/nu15133074

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop