Hostname: page-component-848d4c4894-ndmmz Total loading time: 0 Render date: 2024-05-17T00:27:45.472Z Has data issue: false hasContentIssue false

Effects of dietary vitamins on obesity-related metabolic parameters

Published online by Cambridge University Press:  12 April 2023

Chooi Yeng Lee*
Affiliation:
School of Pharmacy, Monash University Malaysia, Subang Jaya, 47500 Selangor, Malaysia
*
Corresponding author: Chooi Yeng Lee, email chooi.yeng.lee@monash.edu

Abstract

Type 2 diabetes mellitus (T2DM) is one of the leading causes of death worldwide. Genetic factors, some underlying medical conditions, and obesity are risk factors of T2DM. Unlike other risk factors which are non-modifiable, obesity is preventable and usually treatable, and is largely contributed by lifestyle factors. Management of these lifestyle factors may curb the development of T2DM and reduces T2DM prevalence. Dietary vitamins have been recommended as a lifestyle modification intervention to support obesity treatment. Vitamins correlate negatively with body weight, body mass index and body composition. Some of the vitamins may also have anti-adipogenic, anti-inflammatory and antioxidant effects. However, results from pre-clinical and clinical studies of the effects of vitamins on obesity are inconsistent. A clear understanding of the effects of vitamins on obesity will help determine dietary intervention that is truly effective in preventing and treating obesity as well as obesity-related complications including T2DM. This article reviews existing evidences of the effects of vitamin supplementation on obesity and obesity-related metabolic status.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

The World Health Organization defines obesity as individuals having body mass index (BMI) equals to or greater than 30 kg/m2. Obesity is associated with adipose tissue (AT) dysfunction, which is contributed mainly by adipocyte hypertrophy(Reference Laforest, Labrecque and Michaud1). The adipocyte remodelling induces macrophages infiltration, inflammatory cytokine production and synthesis of collagens that limits adipogenesis, and reduces AT's storage capacity(Reference Sun, Tordjman and Cle'ment2), leading to triglyceride accumulation in the liver, heart and around blood vessels(Reference Heilbronn, Smith and Ravussin3). Weight loss improves AT dysfunction and its adipogenic effects(Reference Rossmeislová, Mališová and Kračmerová4).

Presently, the United States Food and Drug Administration (FDA)-approved pharmacotherapy for weight reduction treatment in non-diabetic patients such as liraglutide promotes weight loss, satiety and insulin secretion, the latter overcomes postprandial hyperglycaemia. Liraglutide, however, does not improve AT dysfunction in subcutaneous tissue or prevent inflammation, and does not improve adipogenesis although it has high homology to native glucagon-like peptide-1 (GLP-1), which was reported to reduce inflammation(Reference Anandhakrishnan and Korbonits5) and increase lipolysis(Reference Sharma, Verma and Vaidya6).

In a randomised controlled trial (RCT) involving type 2 diabetes (T2DM) patients, 4 months of a once-daily injection of liraglutide effectively reduced body weight, visceral AT and fasting glucose, but the treatment induced inflammation(Reference Pastel, McCulloch and Ward7). In the study(Reference Pastel, McCulloch and Ward7), the subcutaneous AT RNA and protein expression of tumour necrosis factor-α (TNF-α) and macrophage chemoattractant protein-1 (MCP-1), and the serum MCP-1 levels were significantly increased after 4 months of treatment. Besides that, the subcutaneous AT expression of peroxisome proliferator-activated receptor-γ (PPARγ) and lipoprotein lipase (LPL) were unchanged after liraglutide treatment. PPARγ is a nuclear transcription factor that up-regulates the expression of genes involved in lipid storage and adipogenesis, whereas LPL, which hydrolyses triglycerides, is a marker of adipocyte differentiation, and it increases with triglyceride accumulation. GLP-1 analogues have generally been reported to cause rebound weight gain once therapies are discontinued(Reference Bunck, Cornér and Eliasson8), and liraglutide is of no exception. Liraglutide treatment beyond 20 weeks may be associated with treatment resistance, and patients regain weight beyond 36 weeks(Reference Astrup, Rössner and Van Gaal9,Reference Astrup, Carraro and Finer10) . Another limitation of liraglutide is that it causes gastrointestinal-related side effects early in the treatment course(Reference Mehta, Marso and Neeland11).

Consider the limitation of pharmacotherapies and cost–benefit factors, combining short-term liraglutide with behavioural therapies that promote lifestyle changes was advocated(Reference Anandhakrishnan and Korbonits5). Wharton et al. (Reference Wharton, Lau and Vallis12) suggested adjunctive pharmacotherapy for weight loss and weight loss maintenance for obese individuals or individuals with BMI equals to or more than 27 kg/m2 with adiposity-related complications, to support medical nutrition therapy, physical activity and psychological interventions.

Dietary strategies have been reported for a decade or more as an effective intervention in reducing body weight, where previously, the focus was on macronutrient consumption. Now, contributed by a better understanding of the pathophysiology of obesity as well as the limitation of drug therapies, specific dietary intervention, especially diet with antioxidant and anti-inflammatory properties, is recommended for protection against obesity manifestation(Reference Vahid, Rahmani and Davoodi13). The literature on dietary intervention that is available to date is instructive to achieving positive and sustainable treatment outcomes. Accordingly, the management of obesity is ideally consisting of dietary, pharmacological, physical and psychological therapies(Reference Lee14).

This review aimed to give an overview and critical review of presently published studies on the effects of vitamins on obesity. It focuses on presenting the effects of vitamins on obese-related metabolic parameters. This enables a clear understanding of the effects of vitamins on the obese population specifically. Where studies from obese subjects are absent, studies involving patients with T2DM or metabolic syndrome, and results from pre-clinical studies are included and these are stated accordingly in the review. Studies which involved obese subjects still formed the majority of the review. Obesity is the main cause of metabolic syndrome, and both conditions increase the risk for hyperglycaemia, dyslipidemia, T2DM, cardiovascular disease and certain cancer(Reference Vahid, Rahmani and Davoodi13). The conditions are closely related where individuals who are obese, or have T2DM or metabolic syndrome could all have dysregulated adipocytokines production, increased inflammatory markers and altered blood glucose, insulin sensitivity and lipid profiles.

Dietary vitamins

Specific vitamins are deficient in obese individuals, irrespective of age groups. A negative correlation between multivitamins and the occurrence of obesity in children and adolescents was reported(Reference Tang, Zhan and Wei15). In contrast, consumption of multivitamins, which included vitamins A, B1, B2, B12 and D, reduced the risk of obesity in children and adolescents(Reference Tang, Zhan and Wei15). High prevalence of vitamin A, B1, vitamin C and vitamin D deficiency, with deficiency in vitamin D the highest among all vitamins, was found in obese adults(Reference Via16). Additionally, overweight and obese adults have baseline serum vitamin D that correlated negatively with their BMI(Reference McKay, Ho and Jane17), and they generally consume energy dense, nutrient poor diet(Reference Ledikwe, Blanck and Khan18). Based upon the above reports, the following sections, therefore, focus on vitamins that were reported to be decreased or deficient in obese children and adult, namely vitamins A, B1, C and D, highlighting evidences of the correlation between these vitamins and abdominal adiposity, lipid profile and inflammation, and providing evidences of the effects of vitamin supplementation on obesity and obesity-related metabolic status.

Vitamin C

All overweight and obese patients have vitamin C status that correlated inversely with BMI, and serum vitamin C levels were significantly lower in obese than overweight patients. Moreover, serum vitamin C levels were lower in individuals who have hypertriglyceridaemia and low levels of high-density lipoprotein-cholesterol (HDL-C) as compared with those having normal triglyceride and HDL-C levels. Low vitamin C status was the independent risk factor for obesity and lipid metabolic dysfunction(Reference Yin, Du and Sheng19). However, the effects of vitamin C on lipid metabolism, and vitamin C supplementation on lipid profile, are unclear. In rodent, vitamin C was found to reduce visceral obesity through activating peroxisome proliferator-activated receptor-α (PPARα)(Reference Lee, Ahn and Shin20). But human studies on the effects of vitamin C on adipocytes, including differentiation, triglyceride accumulation and lipolysis inhibition are conflicting(Reference Garcia-Diaz, Lopez-Legarrea and Quintero21), as detailed below.

A meta-analysis of 13 RCTs indicated that a minimum 4 weeks of at least 500 mg daily vitamin C supplementation can significantly decrease serum low-density lipoprotein-cholesterol (LDL-C) and triglyceride concentrations(Reference McRae22). However, participants in those RCTs were not obese but were either healthy, elderly, diabetic or have hyperlipidaemia. Another meta-analysis of RCTs conducted subsequently to ascertain the effects of vitamin C on blood total cholesterol, LDL-C, HDL-C and triglyceride concluded that vitamin C supplementation has no significant effect on lipid profile(Reference Ashor, Siervo and van der Velde23). Although vitamin C supplementation did not change blood lipids concentrations significantly, it has benefited specific groups such as lowering the total cholesterol of younger participants, the LDL-C of healthy participants and the triglyceride of diabetics while increasing the HDL-C in diabetics. The authors, therefore, concluded that vitamin C supplement provided more benefit to individuals with higher baseline levels of total cholesterol and triglyceride(Reference Ashor, Siervo and van der Velde23). Nevertheless, similar to the earlier meta-analysis(Reference McRae22), the adult participants in the study were non-obese.

It appears that patients who have altered metabolic parameters require high dose and chronic consumption of vitamin C if they were to gain any potential benefits of vitamin C. This is due partly to the fact that vitamin C does not accumulate in the body and the excess is eliminated immediately through urine(Reference Levine, Conry-Cantilena and Wang24). Diabetic patients administered 1000 mg daily oral vitamin C for 4 months showed improvement in whole body glucose disposal and non-oxidative glucose metabolism(Reference Paolisso, Balbi and Volpe25), but 800 mg daily intake of vitamin C for 4 weeks did not improve glucose metabolism or insulin resistance(Reference Chen, Karne and Hall26). This means that if the dose of vitamin C is insufficient to fully replenish the low baseline level of vitamin C in these patients(Reference Ford, Mokdad and Giles27), it is ineffective in improving endothelial dysfunction and insulin resistance. The same condition may apply to obesity.

In obesity, white AT overgrowth leads to increased production of pro-inflammatory cytokines including TNF-α, interleukin-6 (IL-6), MCP-1 and inducible nitric oxide synthase (iNOS), activation of inflammatory pathways of IкB kinase (IKK-β) and nuclear factor-кB (NF-кB), mitochondrial dysfunction, reactive oxygen species (ROS) overproduction and depletion of the antioxidant defense(Reference Garcia-Diaz, Campion and Milagro28). Oxidative stress has been associated with low grade chronic inflammation and insulin resistance(Reference Garcia-Diaz, Campion and Milagro28). While it might not be easy to demonstrate the causal effect of the relationship, it is believed that inflammation occurs during the early stage of obesity development, followed by an enhanced oxidative stress status(Reference Vahid, Rahmani and Davoodi13,Reference Matsuda and Shimomura29) . Therefore, dietary supplements which have anti-inflammatory and/or antioxidant properties maybe beneficial in the management of metabolic derangement in obesity.

Vitamin C is an antioxidant due to its ability to donate electron. In isolated rat adipocytes, vitamin C reduced intracellular and extracellular ROS production(Reference Garcia-Diaz, Campion and Milagro28). In vitro, vitamin C inhibited the activation of NF-кB signalling(Reference Bowie and O'Neill30) and IKK-β enzyme(Reference Carcamo, Pedraza and Borquez-Ojeda31). In vitro and in vivo animal studies showed that vitamin C increased the production of lipoxin A4, an anti-inflammatory and antioxidant(Reference Das32). Broiler chicks supplemented with a vitamin C-rich diet have decreased hepatic mRNA expressions of IL-1β, IL-6, interferon-γ, Toll-like receptor-4 and heat shock protein 70 compared with those fed with a control diet. Lipid peroxidation in the serum and liver of these animals was also significantly decreased(Reference Jang, Ko and Moon33). Data of the abovementioned effects of vitamin C supplementation on human, however, are not available, and Abdali et al. (Reference Abdali, Samson and Grover34) thought that vitamin C is of marginal benefits to obese and diabetic patients.

Vitamin B1 (thiamine)

Thiamine is a water-soluble vitamin that is not stored substantially in the body. Many morbidly obese patients are thiamine-deficient, and the prevalence of thiamine deficiency was 7–8 folds higher in African Americans and Hispanics than in Caucasian(Reference Flancbaum, Belsley and Drake35). The reason for racial differences is unknown. Thiamine catalyses several key biochemical reactions involved in glucose metabolism. This means that metabolism of a sugar-high diet requires high amount of thiamine. Obese patients who consume energy dense and high sugar content diet may, therefore, have much higher thiamine needs(Reference Kerns, Arundel and Chawla36), and accelerated thiamine depletion during glucose metabolism. Thiamine deficiency leads to impairment in insulin synthesis and secretion(Reference Mee, Sekar and Subramanian37). Severe deficiency is associated with increased endothelial nitric oxide synthase (eNOS) production, intercellular adhesion molecule-1 (ICAM-1) levels and ROS production(Reference Page, Laight and Cummings38).

Thiamine has antioxidant properties. In vitro studies showed that thiamine supplementation inhibited lipid peroxidation(Reference Lukienko, Mel'nichenko and Zverinskii39), and limited hyperglycaemia-induced von Willebrand factor secretion from bovine aortic endothelial cells thereby endothelial cell dysfunction(Reference Ascher, Gade and Hingorani40). Given the high rates as well as the consequences of thiamine deficiency in obese and diabetic patients, Via(Reference Via16) opined that thiamine supplementation may be considered in this group of people. However, to date, there is no published data on thiamine requirements in overweight and obese patients, i.e. how much thiamine should patients consume in order to tackle the consequences resulting from thiamine deficiency is unclear. Several reasons might explain this: (1) the ability of a thiamine-rich diet in causing weight loss in obese and overweight individuals has not been demonstrated(Reference Keogh, Cleanthous and Wycherley41); (2) the lack of studies and evidence of the effectiveness of long-term thiamine supplementation on combating the metabolic changes in obesity; (3) the amount of thiamine needed by obese people may simply be too high since they have high sugar level contributed by insulin resistance and/or energy dense diet, which accelerate thiamine usage, but they already have a low thiamine level in the body, therefore making the approach to increase thiamine level through diet and supplement non-feasible; (4) the beneficial effects of thiamine may be confounded by the body sugar levels; and (5) there is other dietary vitamin that is superior to thiamine in managing body weight, and glucose and lipid metabolism.

Vitamin D

The main function of vitamin D is for the maintenance of bone tissue, and homeostasis of calcium and phosphorus. Extra-skeletal functions of vitamin D include those on the muscular, insulin sensitivity and immune system, to name a few(Reference Lips42). Moderate vitamin D deficiency leads to increased bone turnover and a greater risk of bone fractures, while severe deficiency causes osteomalacia in adults and rickets in children. Vitamin D deficiency is also associated with visceral adiposity-related metabolic syndrome such as obesity, dyslipidemia, insulin resistance, diabetes, cardiovascular diseases and hypertension(Reference Vranić, Mikolašević and Milić43). Serum vitamin D status correlates negatively with the BMI of children, adolescents and adults(Reference Tang, Zhan and Wei15,Reference McKay, Ho and Jane17) .

The possible causes for the decreased vitamin D serum level in obesity have been reported in numerous review articles(Reference Vranić, Mikolašević and Milić43Reference Walsh, Bowles and Evans46), although as cited in these reviews, the studies of some of those causes were conflicting. The more probable causes of decreased vitamin D in obesity are: (1) vitamin D sequestration in AT – as the amount of AT increases, vitamin D is accumulated and retained in AT, resulting in a lower plasma concentration(Reference Wortsman, Matsuoka and Chen47); (2) low sunlight exposure(Reference Walsh, Bowles and Evans46) and (3) altered volumetric dilution of vitamin D – vitamin D is distributed into serum, muscle, fat and liver, in which all these compartments are increased in obesity(Reference Heaney, Horst and Cullen48). These mechanisms suggest that weight loss or reducing visceral adiposity by increasing physical activity for example, and exposure to sunlight will increase circulatory levels of vitamin D. If so, do obese people still need vitamin D supplementation?

A 26 weeks treatment with 7000 IU (175 μg) vitamin D daily did not change body fat accumulation of obese patients as compared to placebo(Reference Wamberg, Kampmann and Stødkilde-Jørgensen49). In a 12 months RCT, obese patients receiving 2000 IU vitamin D daily and lifestyle-based weight loss programme did not have significant reduction in body weight, body composition, BMI, insulin and C-reactive protein (CRP) levels when compared with the placebo group who received lifestyle-based weight loss programme only(Reference Mason, Xiao and Imayama50). In the present study(Reference Mason, Xiao and Imayama50) however, the placebo group also did not have significant changes in body weight and body composition between pre-treatment and at 12 months, and possibly because of this, according to the above proposed mechanisms (1) and (3), the group has no significant change in serum vitamin D at 12 months (20 ng/ml). The study group that received vitamin D supplementation has significant elevation in serum vitamin D concentration (35 ng/ml), suggesting that intake of 2000 IU once daily for 12 months is sufficient to increase serum vitamin D in obese patients. Together with the study by Wamberg et al. (Reference Wamberg, Kampmann and Stødkilde-Jørgensen49), present evidences do not support vitamin D supplementation in reducing body weight and body composition.

The effects of vitamin D on adipogenesis and lipid accumulation contradict between in vitro studies per se and between in vitro and in vivo studies(Reference Dix, Barcley and Wright44). In vitro studies using mouse cell lines showed that vitamin D inhibited adipogenesis by down-regulating CCAAT enhancer binding proteins (C/EBP) and PPARγ, and inhibited lipid accumulation by suppressing the expression of sterol regulatory element binding protein 1c (SREBP1c) and LPL. SREBP1c is a protein that promotes the expression of genes involved in glucose metabolism, lipogenesis and fatty acid production. In mouse primary cell culture, however, vitamin D was pro-adipogenic. Pro-adipogenic effects of vitamin D were also observed in in vivo animal models. To date, there is no evidence to suggest that vitamin D supplementation suppresses adipogenesis signalling pathways in human. Studies of the effects of vitamin D on lipid profiles of obese patients are available but have reported conflicting results – a placebo-controlled RCT showed that vitamin D supplement has no effect on patients’ plasma triglyceride, HDL-C and total cholesterol levels(Reference Wamberg, Kampmann and Stødkilde-Jørgensen49). But in a meta-analysis, vitamin D treatment increased HDL-C and oral glucose insulin sensitivity and decreased triglyceride but it also increased LDL-C(Reference Manousopoulou, Al-Daghri and Garbis51).

In terms of the anti-inflammatory effects of vitamin D, Wamberg et al. (Reference Wamberg, Kampmann and Stødkilde-Jørgensen49) reported that the consumption of 7000 IU vitamin D daily for 26 weeks did not significantly decrease circulatory inflammatory markers such as CRP, IL-6, MCP-1, leptin and adiponectin. In other studies, intake of 40 000 IU vitamin D weekly for one year decreased serum IL-6 but increased CRP, and demonstrated no effects on insulin resistance, TNF-α(Reference Beilfuss, Berg and Sneve52), ICAM-1, interferon-γ, MCP-1 and CRP(Reference Jorde, Sneve and Torjesen53) in overweight and obese patients. Despite these contradictory findings, since patients with BMI above 38⋅4 kg/m2 have baseline vitamin D that correlated negatively with serum levels of leptin and resistin, and positively with serum adiponectin levels, some researchers suggest consuming vitamin D to improve AT function and prevent obesity-related diseases(Reference Stokić, Kupusinac and Tomic-Naglic54), and this recommendation may be relevant to morbidly obese patients. However, if the mechanisms that cause serum vitamin D reduction in obesity are as reported in the literature, any means of reducing body weight would increase serum vitamin D. Moreover, there are very few vitamin D-rich sources, and the amount of vitamin D from the same source but of different origin could vary and could be relatively low(Reference Midtbø, Nygaard and Markhus55).

Vitamin A

Dietary vitamin A was found to be associated significantly with plasma retinol levels(Reference Wei, Peng and Cao56), while inadequate intake is the major factor contributing to vitamin A deficiency(Reference Reifen57). Overweight and obese individuals who were deficient in serum levels of vitamin A and vitamin D, were said to have high calorie malnutrition(Reference McKay, Ho and Jane17). The BMI of obese adults and children correlated negatively with serum vitamin A levels(Reference Wei, Peng and Cao56Reference Botella-Carretero, Balsa and Vázquez58). A significantly lower serum vitamin A concentration in obese children than that of normal weight children was also associated with increased waist circumference, fasting plasma glucose and triglyceride, and decreased HDL-C(Reference Wei, Peng and Cao56). Research to understand the effects of vitamin A on obesity has been done extensively on its precursor, carotenoids. This may be explained by pre-clinical and clinical studies, which indicated that carotenoids and carotenoids derivatives prevented abdominal adiposity, and have anti-inflammatory and antioxidant effects. Moreover, intact carotenoid molecules and carotenoid cleavage products may have additional biological activities whose relevance for human health are still unknown(Reference Bonet, Canas and Ribot59).

There are over 600 carotenoids present in nature, of which some 50 are found in human diet, mainly fruits and vegetables. But only about half of those found in the diet are detected in human blood and tissue(Reference Krinsky and Johnson60,Reference Moran, Mohn and Hason61) . The most abundant carotenoids in human serum are α-carotene, β-carotene, lycopene, lutein, zeaxanthin and β-cryptoxanthin(Reference Moran, Mohn and Hason61). Retinol may be generated de novo from α-carotene, β-carotene and β-cryptoxanthin(Reference Bonet, Canas and Ribot59). Carotenoids are accumulated mainly in the liver and adipose tissues(Reference Rodriguez-Concepcion, Avalos and Bonet62). The tissue distribution of carotenoids implies their potential effects on the metabolic processes within these tissues(Reference Eggersdorfer and Wyss63).

Carotenoids are lipid-soluble and widely found in plant-based sources. Besides fruits and vegetables, carotenoids are available in commonly consumed food such as bread, eggs, milk, beverages, fats and oils(Reference Rao and Rao64). Carotenoids insufficiency was reported in adults and children with obesity(Reference Yao, Yan and Guo65). A study by Harari et al. (Reference Harari, Coster and Jenkins66) showed that all the five serum carotenoids measured (α-carotene, β-carotene, lutein, lycopene and ζ-carotene) were significantly lower in adult obese patients compared with non-obese individuals. These obese patients have higher total body fat and central fat, but lower HDL-C, and reduced insulin sensitivity. Similar to adults, serum carotenoids (α-carotene and β-carotene) of obese children correlated negatively with BMI, waist circumference, fat mass and triglyceride, and positively with HDL-C(Reference Farook, Reddivari and Mummidi67). In healthy adults, serum total and specific carotenoid concentrations were associated inversely with CRP, MCP-1 and TNF-α(Reference Jing, Xiao and Dong68). In school-aged children, plasma β-carotene was associated negatively with plasma IL-6 levels(Reference Rodriguez-Rodriguez, Lopez-Sobaler and Navia69). Obese individuals who have significantly lower circulating carotenoids than healthy subjects may, therefore, be exposed to a higher risk of inflammation.

A recent meta-analysis(Reference Yao, Yan and Guo65) reported that carotenoids supplementation in overweight and obese individuals might contribute to the reduction in body weight, BMI, waist circumference and total cholesterol while increasing HDL-C. However, carotenoids have no effect on fat ratio, LDL-C and triglyceride concentrations. Among the six carotenoids present abundantly in serum, only α-carotene, β-carotene and β-cryptoxanthin have pro-vitamin A activity. The importance of the other carotenoids in human nutrition is rather limited(Reference Grune, Lietz and Palou70). Because β-carotene oxygenase 1 is the sole enzyme responsible for the conversion of carotenoids to vitamin A, significant interest, hence research has been conducted on β-carotene to delineate its mechanism of action on adipogenesis and inflammation.

In vitro and in vivo animal studies showed that β-carotene through its cleavage product, β-apo-14′-carotenal, inhibited adipogenesis through repression of PPARα, PPARγ and retinoid X receptor-α activation(Reference Ziouzenkova, Orasanu and Sukhova71). β-carotene through its metabolite retinoic acid, decreased the expression of PPARγ and C/EBP-α, and the lipid content of mature adipocytes. β-carotene administration, through increasing retinoic acid signalling, down-regulated PPARγ expression in white AT of vitamin A-deficient mice(Reference Lobo, Amengual and Li72). β-carotene exhibited anti-inflammatory effects in adipocytes by limiting TNF-α-induced ROS production as well as alterations in the expression of genes related to insulin sensitivity, including adiponectin, adipocyte lipid-binding protein, glucose transporter-4, PPARγ2 and adiponectin protein(Reference Kameji, Mochizuki and Miyoshi73). Also in adipocytes, β-carotene inhibited oxidative stress-induced adiponectin dysregulation, increased MCP-1 expression and NF-кB activation(Reference Cho, Kim and Kim74) (Fig. 1). As to whether the effects of anti-adipogenesis and anti-inflammation of β-carotene are seen in human have not been established yet.

Fig. 1. The effects of carotenoids and β-carotene on obesity. In human studies, carotenoids were reported to improve weight, body composition and HDL-C in obese subjects. In pre-clinical studies, β-carotene's cleavage product, β-apo-14′-carotenal inhibited adipogenesis through suppressing PPARα, PPARγ and RXR-α activation. The metabolite of β-carotene, retinoic acid was reported to inhibit adipogenesis and inflammation. These effects were seen in in vitro and in vivo studies. ALBP, adipocyte lipid-binding protein; C/EBP-α, CCAAT enhancer binding proteins; GLUT4, glucose transporter-4; HDL-C, high-density lipoprotein-cholesterol; MCP-1, macrophage chemoattractant protein-1; NF- кB, nuclear factor-кB; PPARα, peroxisome proliferator-activated receptor-α; PPARγ, peroxisome proliferator-activated receptor-γ; ROS, reactive oxygen species; RXR-α, retinoid X receptor-α; TNF-α, tumor necrosis factor-α; WAT, white adipose tissue.

Conclusions

A nutritional-balanced diet is especially important for obese individuals, to maintain health and support pharmacotherapies and other lifestyle modification strategies. Vitamin supplementation that takes into consideration all of the following criteria may lead to a better treatment outcome. The compound is distributed widely in food sources, cheap and easily accessible; is lipid-soluble, which will give better tissues bioavailability than water-soluble compounds; has been shown to correlate negatively with body weight and composition and has demonstrated anti-adipogenic, anti-inflammatory and antioxidant properties from RCTs. The treatment target may be on normalising the decreased serum levels of vitamins in obese patients.

There is a lack of evidence that supports any beneficial effects of vitamin C supplementation on glucose and lipid metabolism, and suppression of inflammation in obese patients. However, as an antioxidant, long-term and high dose vitamin C consumption may provide some benefits to the overall health. Thiamine, despite being an antioxidant, requires more human intervention studies to determine its benefits as well as feasibility in supporting obesity treatment. Although vitamin D may have some anti-inflammatory effects that improve AT functions, such evidence, together with evidences of its effects on body weight reduction and adipogenesis, are neither clear nor convincing. Therefore, vitamin D may not be the first line vitamin supplementation for obesity treatment. As compared with vitamins C, B1 and D, vitamin A and its precursor carotenoids, specifically β-carotene, appeared to be the more promising vitamin that can be used for the regulation of body weight, lipid metabolism and inflammatory status in obesity (Fig. 2). However, human study is warranted to confirm the anti-adipogenic, anti-inflammatory and antioxidative effects seen for β-carotene in in vitro and in vivo animal studies.

Fig. 2. Summary of the effects of vitamins A–D supplementation on metabolic changes in obesity. Dietary vitamins including vitamins C, B1 and D have either conflicting evidences or no evidence of reducing adiposity, improving lipid profile and the inflammatory status of obese subjects. The precursor of vitamin A, carotenoids, reduce adiposity and improve lipid profile. β-carotene was reported to have anti-inflammatory effects in pre-clinical studies but such evidence is lacking in human studies.

Future human intervention studies of carotenoids and β-carotene should investigate not only their effects on visceral adiposity and AT functions, but the lowest effective doses that produce health benefits. This is because dietary intervention may be implemented long-term, and as such any potential dose-dependent side effects of carotenoids should be minimised. Effective management of obesity through dietary vitamin that goes along with drug, physical and behavioural therapies, may also reduce the risk of T2DM development in obesity.

Acknowledgements

C. Y. L. is the sole author of this manuscript.

This study was not funded.

There is no conflict of interest.

References

Laforest, S, Labrecque, J, Michaud, A, et al. (2015) Adipocyte size as a determinant of metabolic disease and adipose tissue dysfunction. Crit Rev Clin Lab Sci 52, 301303.CrossRefGoogle ScholarPubMed
Sun, K, Tordjman, J, Cle'ment, K, et al. (2013) Fibrosis and adipose tissue dysfunction. Cell Metab 18, 470477.CrossRefGoogle ScholarPubMed
Heilbronn, L, Smith, SR & Ravussin, E (2004) Failure of fat cell proliferation, mitochondrial function and fat oxidation results in ectopic fat storage, insulin resistance and type II diabetes mellitus. Int J Obes Relat Metab Disord 28, S12S21.CrossRefGoogle ScholarPubMed
Rossmeislová, L, Mališová, L, Kračmerová, J, et al. (2013) Weight loss improves the adipogenic capacity of human preadipocytes and modulates their secretory profile. Diabetes 62, 19901995.CrossRefGoogle ScholarPubMed
Anandhakrishnan, A & Korbonits, M (2016) Glucagon-like peptide 1 in the pathophysiology and pharmacotherapy of clinical obesity. World J Diabetes 7, 572598.CrossRefGoogle ScholarPubMed
Sharma, D, Verma, S, Vaidya, S, et al. (2018) Recent updates on GLP-1 agonists: current advancements & challenges. Biomed Pharmacother 108, 952962.CrossRefGoogle ScholarPubMed
Pastel, E, McCulloch, LJ, Ward, R, et al. (2017) GLP-1 analogue-induced weight loss does not improve obesity-induced AT dysfunction. Clin Sci 131, 343353.CrossRefGoogle Scholar
Bunck, MC, Cornér, A, Eliasson, B, et al. (2011) Effects of exenatide on measures of β-cell function after 3 years in metformin-treated patients with type 2 diabetes. Diabetes Care 34, 20412047.CrossRefGoogle ScholarPubMed
Astrup, A, Rössner, S, Van Gaal, L, et al. (2009) Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet 374, 16061616.CrossRefGoogle ScholarPubMed
Astrup, A, Carraro, R, Finer, N, et al. (2012) Safety, tolerability and sustained weight loss over 2 years with the once-daily human GLP-1 analog, liraglutide. Int J Obes 36, 843854.CrossRefGoogle ScholarPubMed
Mehta, A, Marso, SP & Neeland, IJ (2017) Liraglutide for weight management: a critical review of the evidence. Obes Sci Pract 3, 314.CrossRefGoogle ScholarPubMed
Wharton, S, Lau, DCW, Vallis, M, et al. (2020) Obesity in adults: a clinical practice guideline. CMAJ 192, E875E891.CrossRefGoogle ScholarPubMed
Vahid, F, Rahmani, D & Davoodi, SH (2021) The correlation between serum inflammatory, antioxidant, glucose handling biomarkers, and Dietary Antioxidant Index (DAI) and the role of DAI in obesity/overweight causation: population-based case-control study. Int J Obes 45, 25912599.CrossRefGoogle ScholarPubMed
Lee, CY (2021) A combination of glucagon-like peptide-1 receptor agonist and dietary intervention could be a promising approach for obesity treatment. Front Endocrinol. doi:10.3389/fendo.2021.748477.CrossRefGoogle ScholarPubMed
Tang, W, Zhan, W, Wei, M, et al. (2022) Associations between different dietary vitamins and the risk of obesity in children and adolescents: a machine learning approach. Front Endocrinol. doi:10.3389/fendo.2021.816975.Google ScholarPubMed
Via, M (2012) The malnutrition of obesity: micronutrient deficiencies that promote diabetes. ISRN Endocrinol. doi:10.5402/2012/103472.CrossRefGoogle ScholarPubMed
McKay, J, Ho, S, Jane, M, et al. (2020) Overweight and obese Australian adults and micronutrient deficiency. BMC Nutr. doi:10.1186/s40795-020-00336-9.CrossRefGoogle ScholarPubMed
Ledikwe, JH, Blanck, HM, Khan, LK, et al. (2006) Low-energy-density diets are associated with high diet quality in adults in the United States. J Am Diet Assoc 106, 11721180.CrossRefGoogle ScholarPubMed
Yin, J, Du, L, Sheng, C, et al. (2022) Vitamin C status and its change in relation to glucose-lipid metabolism in overweight and obesity patients following laparoscopic sleeve gastrectomy. Eur J Clin Nutr 76, 13871392.CrossRefGoogle ScholarPubMed
Lee, H, Ahn, J, Shin, SS, et al. (2019) Ascorbic acid inhibits visceral obesity and nonalcoholic fatty liver disease by activating peroxisome proliferator-activated receptor α in high-fat-diet-fed C57BL/6j mice. Int J Obes 43, 16201630.CrossRefGoogle ScholarPubMed
Garcia-Diaz, DF, Lopez-Legarrea, P, Quintero, P, et al. (2014) Vitamin C in the treatment and/or prevention of obesity. J Nutr Sci Vitaminol 60, 367379.CrossRefGoogle ScholarPubMed
McRae, MP (2008) Vitamin C supplementation lowers serum low-density lipoprotein cholesterol and triglycerides; a meta-analysis of 13 randomized controlled trials. J Chiropr Med 7, 4858.CrossRefGoogle ScholarPubMed
Ashor, AW, Siervo, M, van der Velde, F, et al. (2016) Systematic review and meta-analysis of randomised controlled trials testing the effects of vitamin C supplementation on blood lipids. Clin Nutr 35, 626637.CrossRefGoogle ScholarPubMed
Levine, M, Conry-Cantilena, C, Wang, Y, et al. (1996) Vitamin C pharmacokinetics in healthy volunteers: evidence for a recommended dietary allowance. Proc Natl Acad Sci USA 93, 37043709.CrossRefGoogle ScholarPubMed
Paolisso, G, Balbi, V, Volpe, C, et al. (1995) Metabolic benefits deriving from chronic vitamin C supplementation in aged non-insulin dependent diabetics. J Am Coll Nutr 14, 387392.CrossRefGoogle ScholarPubMed
Chen, H, Karne, RJ, Hall, G, et al. (2006) High-dose oral vitamin C partially replenishes vitamin C levels in patients with Type 2 diabetes and low vitamin C levels but does not improve endothelial dysfunction or insulin resistance. Am J Physiol Heart Circ Physiol 290, H137H145.CrossRefGoogle ScholarPubMed
Ford, ES, Mokdad, AH, Giles, WH, et al. (2003) The metabolic syndrome and antioxidant concentrations: findings from the Third National Health and Nutrition Examination Survey. Diabetes 52, 23462352.CrossRefGoogle ScholarPubMed
Garcia-Diaz, DF, Campion, J, Milagro, FI, et al. (2010) Vitamin C inhibits leptin secretion and some glucose/lipid metabolic pathways in primary rat adipocytes. J Mol Endocrinol 45, 3343.CrossRefGoogle ScholarPubMed
Matsuda, M & Shimomura, L (2013) Increased oxidative stress in obesity: implications for metabolic syndrome, diabetes, hypertension, dyslipidemia, atherosclerosis, and cancer. Obes Res Clin Prac 7, e330e341.CrossRefGoogle ScholarPubMed
Bowie, AG & O'Neill, LA (2000) Vitamin C inhibits NF-кB activation by TNF via the activation of p38 mitogen-activated protein kinase. J Immunol 165, 71807188.CrossRefGoogle Scholar
Carcamo, JM, Pedraza, A, Borquez-Ojeda, O, et al. (2004) Vitamin C is a kinase inhibitor: dehydroascorbic acid inhibits IкBα kinase β. Mol Cell Biol 24, 66456652.CrossRefGoogle Scholar
Das, UN (2019) Vitamin C for type 2 diabetes mellitus and hypertension. Arch Med Res 50, 1114.CrossRefGoogle ScholarPubMed
Jang, I-S, Ko, Y-H, Moon, Y-S, et al. (2014) Effects of vitamin C or E on the pro-inflammatory cytokines, heat shock protein 70 and antioxidant status in broiler chicks under summer conditions. Asian Aust J Anim Sci 27, 749756.CrossRefGoogle ScholarPubMed
Abdali, D, Samson, SE & Grover, AK (2015) How effective are antioxidant supplements in obesity and diabetes? Med Princ Pract 24, 201215.CrossRefGoogle ScholarPubMed
Flancbaum, L, Belsley, S, Drake, V, et al. (2006) Preoperative nutritional status of patients undergoing roux-en-Y gastric bypass for morbid obesity. J Gastrointest Surg 10, 10331037.CrossRefGoogle ScholarPubMed
Kerns, JC, Arundel, C & Chawla, LS (2015) Thiamin deficiency in people with obesity. Adv Nutr 6, 147153.CrossRefGoogle ScholarPubMed
Mee, LNS, Sekar, VT, Subramanian, VS, et al. (2009) Pancreatic beta cells and islets take up thiamin by a regulated carrier-mediated process: studies using mice and human pancreatic preparations. Am J Physiol Gastrointest Liver Physiol 297, G197G206.CrossRefGoogle ScholarPubMed
Page, GLJ, Laight, D & Cummings, MH (2011) Thiamine deficiency in diabetes mellitus and the impact of thiamine replacement on glucose metabolism and vascular disease. Int J Clin Pract 65, 684690.CrossRefGoogle ScholarPubMed
Lukienko, PI, Mel'nichenko, NG, Zverinskii, IV, et al. (2000) Antioxidant properties of thiamine. Bull Exp Biol Med 130, 874876.CrossRefGoogle ScholarPubMed
Ascher, E, Gade, PV, Hingorani, A, et al. (2001) Thiamine reverses hyperglycemia-induced dysfunction in cultured endothelial cells. Surgery 130, 851858.CrossRefGoogle ScholarPubMed
Keogh, JB, Cleanthous, X, Wycherley, TP, et al. (2012) Increased thiamine intake may be required to maintain thiamine status during weight loss in patients with type 2 diabetes. Diabetes Res Clin Pract 98, e40e42.CrossRefGoogle ScholarPubMed
Lips, P (2006) Vitamin D physiology. Prog Biophys Mol Biol 92, 48.CrossRefGoogle ScholarPubMed
Vranić, L, Mikolašević, I & Milić, S (2019) Vitamin D deficiency: consequence or cause of obesity? Medicina. doi:10.3390/medicina55090541.CrossRefGoogle ScholarPubMed
Dix, CF, Barcley, JL & Wright, ORL (2017) The role of vitamin D in adipogenesis. Nutr Rev 76, 4759.CrossRefGoogle Scholar
Savastano, S, Barrea, L, Savanelli, MC, et al. (2017) Low vitamin D status and obesity: role of nutritionist. Rev Endocr Metab Disord 18, 215225.CrossRefGoogle ScholarPubMed
Walsh, JS, Bowles, S & Evans, AL (2017) Vitamin D in obesity. Curr Opin Endocrinol Diabetes Obes 24, 389394.CrossRefGoogle ScholarPubMed
Wortsman, J, Matsuoka, LY, Chen, TC, et al. (2000) Decreased bioavailability of vitamin D in obesity. Am J Clin Nutr 72, 690693.CrossRefGoogle ScholarPubMed
Heaney, RP, Horst, RL, Cullen, DM, et al. (2009) Vitamin D3 distribution and status in the body. J Am Coll Nutr 28, 252256.CrossRefGoogle ScholarPubMed
Wamberg, L, Kampmann, U, Stødkilde-Jørgensen, H, et al. (2013) Effects of vitamin D supplementation on body fat accumulation, inflammation, and metabolic risk factors in obese adults with low vitamin D levels – results from a randomized trial. Eur J Intern Med 24, 644649.CrossRefGoogle ScholarPubMed
Mason, C, Xiao, L, Imayama, I, et al. (2014) Vitamind3 supplementation during weight loss: a double-blind randomized controlled trial. Am J Clin Nutr 99, 10151025.CrossRefGoogle Scholar
Manousopoulou, A, Al-Daghri, NM, Garbis, SD, et al. (2015) Vitamin D and cardiovascular risk among adults with obesity: a systematic review and meta-analysis. Eur J Clin Invest 45, 11131126.CrossRefGoogle ScholarPubMed
Beilfuss, J, Berg, V, Sneve, M, et al. (2012) Effects of a 1-year supplementation with cholecalciferol on interleukin-6, tumor necrosis factor-alpha and insulin resistance in overweight and obese subjects. Cytokine 60, 870874.CrossRefGoogle ScholarPubMed
Jorde, R, Sneve, M, Torjesen, PA, et al. (2010) No effect of supplementation with cholecalciferol on cytokines and markers of inflammation in overweight and obese subjects. Cytokine 50, 175180.CrossRefGoogle Scholar
Stokić, E, Kupusinac, A, Tomic-Naglic, D, et al. (2015) Vitamin D and dysfunctional adipose tissue in obesity. Angiology 66, 613618.CrossRefGoogle ScholarPubMed
Midtbø, LK, Nygaard, LB, Markhus, MW, et al. (2020) Vitamin D status in preschool children and its relations to vitamin D sources and body mass index-Fish Intervention Studies-KIDS (FINS-KIDS). Nutrition. doi:10.1016/j.nut.2019.110595.CrossRefGoogle ScholarPubMed
Wei, X, Peng, R, Cao, J, et al. (2016) Serum vitamin A status is associated with obesity and the metabolic syndrome among school-age children in Chongqing, China. Asia Pac J Clin Nutr 25, 563570.Google ScholarPubMed
Reifen, R (2002) Vitamin A as an anti-inflammatory agent. Proc Nutr Soc 61, 397400.CrossRefGoogle ScholarPubMed
Botella-Carretero, JI, Balsa, JA, Vázquez, C, et al. (2010) Retinol and α-Tocopherol in morbid obesity and nonalcoholic fatty liver disease. Obes Surg 20, 6976.CrossRefGoogle ScholarPubMed
Bonet, ML, Canas, JA, Ribot, J, et al. (2011) Carotenoids and their conversion products in the control of adipocyte function, adiposity and obesity. Arch Biochem Biophys 572, 112125.CrossRefGoogle Scholar
Krinsky, NI & Johnson, EJ (2005) Carotenoid actions and their relation to health and disease. Mol Aspects Med 26, 459516.CrossRefGoogle ScholarPubMed
Moran, NE, Mohn, ES, Hason, N, et al. (2018) Intrinsic and extrinsic factors impacting absorption, metabolism, and health effects of dietary carotenoids. Adv Nutr 9, 465492.CrossRefGoogle ScholarPubMed
Rodriguez-Concepcion, M, Avalos, J, Bonet, ML, et al. (2018) A global perspective on carotenoids: metabolism, biotechnology, and benefits for nutrition and health. Prog Lipid Res 70, 6293.CrossRefGoogle ScholarPubMed
Eggersdorfer, M & Wyss, A (2018) Carotenoids in human nutrition and health. Arch Biochem Biophys 652, 1826.CrossRefGoogle ScholarPubMed
Rao, AV & Rao, LG (2007) Carotenoids and human health. Pharmacol Res 55, 207216.CrossRefGoogle ScholarPubMed
Yao, N, Yan, S, Guo, Y, et al. (2021) The association between carotenoids and subjects with overweight or obese: a systematic review and meta-analysis. Food Funct 12, 47684782.CrossRefGoogle ScholarPubMed
Harari, A, Coster, ACF, Jenkins, A, et al. (2020) Obesity and insulin resistance are inversely associated with serum and adipose tissue carotenoid concentrations in adults. J Nutr 150, 3846.CrossRefGoogle ScholarPubMed
Farook, VS, Reddivari, L, Mummidi, S, et al. (2017) Genetics of serum carotenoid concentrations and their correlation with obesity-related traits in Mexican American children. Am J Clin Nutr 106, 5258.CrossRefGoogle ScholarPubMed
Jing, L, Xiao, M, Dong, H, et al. (2018) Serum carotenoids are inversely associated with RBP4 and other inflammatory markers in middle-aged and elderly adults. Nutrients. doi:10.3390/nu10030260.CrossRefGoogle ScholarPubMed
Rodriguez-Rodriguez, E, Lopez-Sobaler, AM, Navia, B, et al. (2017) β-carotene concentration and its association with inflammatory biomarkers in Spanish schoolchildren. Ann Nutr Metab 71, 8087.CrossRefGoogle ScholarPubMed
Grune, T, Lietz, G, Palou, A, et al. (2010) β-carotene is an important vitamin A source for humans. J Nutr 140, 2268S2285S.CrossRefGoogle ScholarPubMed
Ziouzenkova, O, Orasanu, G, Sukhova, G, et al. (2007) Asymmetric cleavage of β-carotene yields a transcriptional repressor of retinoid X receptor and peroxisome proliferator-activated receptor responses. Mol Endocrinol 21, 7788.CrossRefGoogle ScholarPubMed
Lobo, GP, Amengual, J, Li, HNM, et al. (2010) Β,β-carotene decreases peroxisome proliferator receptor γ activity and reduces lipid storage capacity of adipocytes in a β,β-carotene oxygenase 1-dependent manner. J Biol Chem 285, 2789127899.CrossRefGoogle Scholar
Kameji, H, Mochizuki, K, Miyoshi, N, et al. (2010) β-Carotene accumulation in 3T3-L1 adipocytes inhibits the elevation of reactive oxygen species and the suppression of genes related to insulin sensitivity induced by tumor necrosis factor-α. Nutrition 26, 11511156.CrossRefGoogle ScholarPubMed
Cho, SO, Kim, MH & Kim, H (2018) β-Carotene inhibits activation of NF-кB, Activator Protein-1, and STAT3 and regulates abnormal expression of some adipokines in 3T3-L1 adipocytes. J Cancer Prev 23, 3743.CrossRefGoogle Scholar
Figure 0

Fig. 1. The effects of carotenoids and β-carotene on obesity. In human studies, carotenoids were reported to improve weight, body composition and HDL-C in obese subjects. In pre-clinical studies, β-carotene's cleavage product, β-apo-14′-carotenal inhibited adipogenesis through suppressing PPARα, PPARγ and RXR-α activation. The metabolite of β-carotene, retinoic acid was reported to inhibit adipogenesis and inflammation. These effects were seen in in vitro and in vivo studies. ALBP, adipocyte lipid-binding protein; C/EBP-α, CCAAT enhancer binding proteins; GLUT4, glucose transporter-4; HDL-C, high-density lipoprotein-cholesterol; MCP-1, macrophage chemoattractant protein-1; NF- кB, nuclear factor-кB; PPARα, peroxisome proliferator-activated receptor-α; PPARγ, peroxisome proliferator-activated receptor-γ; ROS, reactive oxygen species; RXR-α, retinoid X receptor-α; TNF-α, tumor necrosis factor-α; WAT, white adipose tissue.

Figure 1

Fig. 2. Summary of the effects of vitamins A–D supplementation on metabolic changes in obesity. Dietary vitamins including vitamins C, B1 and D have either conflicting evidences or no evidence of reducing adiposity, improving lipid profile and the inflammatory status of obese subjects. The precursor of vitamin A, carotenoids, reduce adiposity and improve lipid profile. β-carotene was reported to have anti-inflammatory effects in pre-clinical studies but such evidence is lacking in human studies.