Skip to main content
Cureus logoLink to Cureus
. 2021 Oct 6;13(10):e18521. doi: 10.7759/cureus.18521

Role of Vitamin B12 and Folate in Metabolic Syndrome

Tejaswini Ashok 1, Harivarsha Puttam 2, Victoria Clarice A Tarnate 3, Sharan Jhaveri 4, Chaithanya Avanthika 5,6,, Amanda Guadalupe Trejo Treviño 7, Sandeep SL 8, Nazia T Ahmed 9
Editors: Alexander Muacevic, John R Adler
PMCID: PMC8569690  PMID: 34754676

Abstract

Metabolic syndrome (MS) is a collection of pathological metabolic conditions that includes insulin resistance, central or abdominal obesity, dyslipidemia, and hypertension. It affects large populations worldwide, and its prevalence is rising exponentially. There is no specific mechanism that leads to the development of MS. Proposed hypotheses range from visceral adiposity being a key factor to an increase in very-low-density lipoprotein and fatty acid synthesis as the primary cause of MS. Numerous pharmaceutical therapies are widely available in the market for the treatment of the individual components of MS. The relationship between MS and vitamin B complex supplementation, specifically folic acid and vitamin B12, has been a subject of investigation worldwide, with several trials reporting a positive impact with vitamin supplementation on MS.

In this study, an all-language literature search was conducted on Medline, Cochrane, Embase, and Google Scholar till September 2021. The following search strings and Medical Subject Headings (MeSH) terms were used: “Vitamin B12,” “Folate,” “Metabolic Syndrome,” and “Insulin Resistance.” We explored the literature on MS for its epidemiology, pathophysiology, newer treatment options, with a special focus on the effectiveness of supplementation with vitamins B9 and B12.

According to the literature, vitamin B12 and folate supplementation, along with a host of novel therapies, has a considerable positive impact on MS. These findings must be kept in mind while designing newer treatment protocols in the future.

Keywords: insulin resistance, cardiovascular disease, nutrition and metabolism, obesity and diabetes, vitamin b supplementation, folic acid supplementation, vitamin b12 supplementation, diabetes and metabolic syndrome

Introduction and background

Metabolic syndrome (MS) is one of the most debilitating disorders currently affecting large populations worldwide, with its prevalence rising exponentially. MS refers to a group of pathological metabolic conditions including insulin resistance, central or abdominal obesity, atherogenic dyslipidemia, and hypertension [1]. According to the Adult Treatment Panel III (ATP III) classification, the parameters of MS include waist circumference of >102 cm for men and >88 cm for women, fasting blood glucose of >6.1 mmol/L, blood pressure of >135/85 mmHg, triglyceride level of >1.7 mmol/L, and high-density lipoprotein-cholesterol (HDL-C) level of <1.0 mmol/L [2]. The diagnosis of MS is established if an individual meets three of these five parameters [2]. MS, also known as insulin resistance syndrome, syndrome X, as well as the deadly quartet, has been recognized as an important risk factor for the development of cardiovascular disease and type 2 diabetes mellitus, with an estimated two-fold and five-fold increased incidence risk, respectively [3]. By 2030, approximately 38% of the adult population globally is predicted to be overweight and roughly 20% to be obese, which increases the prevalence of morbidity caused by chronic diseases [4]. Numerous pharmaceutical therapies are widely available in the market for the treatment of the individual components of MS, such as statins, antihypertensives, and antidiabetic drugs. Meanwhile, suggested dietary supplements are also available, such as folic acid and vitamin B12, which may be beneficial in the treatment of MS.

Vitamins B9 (folate) and B12 (cobalamin) are essential water-soluble vitamins that serve an important role in DNA methylation and the homeostasis of both amino acids and lipids through the regulation of one-carbon metabolism [5]. One-carbon metabolism is a chain of interconnected biochemical pathways mainly powered by folate and methionine to generate methyl donors utilized by several cellular processes [5]. Because vitamins B9 and B12 are crucial for cell metabolism, their deficiency can lead to alarming health consequences.

Low levels of folate and cobalamin can cause DNA synthesis interference, cellular inflammation, and elevated synthesis of fat and homocysteine [6]. Homocysteine is a non-proteinogenic amino acid produced by the breakdown of dietary proteins such as methionine [7]. Hyperhomocysteinemia can contribute to the development of cardiovascular and cerebrovascular diseases due to atherosclerotic vascular-endothelial injury [8], as well as the development of insulin resistance [9]. Studies have shown that folic acid, vitamin B12, and other B-complex supplements are effective in reducing homocysteine levels in the plasma [10], thus leading to the reduction of the prevalence of MS.

The relationship between MS and vitamin B complex, specifically folic acid and vitamin B12, has been the subject of many studies worldwide. Several studies have reported a positive association between the supplementation of folic acid and vitamin B12 and their effectiveness in the reduction of cardiovascular disease [11] and insulin resistance [12]. The scope of this literature review is to study and analyze scientific articles from PubMed and other academic journal libraries regarding the possible role and effectiveness of folic acid and vitamin B12 in MS patients. By reviewing the efficacy of folic acid and vitamin B12 supplementation in MS, we aim to enumerate potential recommendations which can be added to the current treatment regimen for MS patients. However, considerations of additional studies, such as those relating to the appropriate frequency, duration, and dosage of folic acid and vitamin B12 as an adjunct for MS treatment, will need to be evaluated to prevent any unwanted adverse effects.

Review

Sources, metabolism, and interactions

Sources

It is important to shed light on the dietary sources of folic acid and cobalamin, being vitamins of public health significance. Excellent sources of vitamin B9 (folate) are green leafy vegetables, citrus fruits, pulses, cereals, liver, and egg yolk while vitamin B12 is majorly present in meat, eggs, milk, dairy products, fish, and shellfish, with the concentration of cobalamin varying within the food of ruminant origin with the highest concentrations found in offal such as liver and kidney. Various studies have been conducted to measure the adequacy of dietary intake of folic acid and cobalamin in different populations owing to its key function in the one-carbon metabolism and its clinical significance [13,14]. In Europe, a study demonstrated subclinical deficiency of vitamin B9 and B12 to be prevalent in 20% of adolescents [15]. Correspondingly, nutritional data surveys conducted in cross-country comparative studies across the European continent showed a mean prevalence of inadequacy at or below 10% for the elderly and below 20% for the adult population [16]. Remarkable differences have been noted in vitamin B12 levels in cohort studies of meat eaters, fish eaters, vegetarians, and vegans, highlighting the inadequate dietary intake among vegans and vegetarians [17,18]. Attempts have been made to identify naturally occurring cobalamin-rich, plant-derived foods, such as dried shiitake mushroom fruiting bodies, dried green laver (Enteromorpha species) and purple laver (Porphyra species), aquatic plant Wolffia globosa (Mankai), sea buckthorn (Hippophae rhamnoides) berries and granulate products, and sidea couch grass (Elymus repens) products (dry extract and ground) [19-22]. A clinical trial conducted in India to determine improvement in vitamin B12 status among deficient vegetarians with dairy cow’s milk intake concluded that it can drastically improve blood cobalamin levels [23].

Finding new methods that increase cobalamin content in vegetarian diets has been an important field of research. Two such effective methods are the addition of cow manure to significantly increase the vitamin B12 content of spinach leaves and the fermentation of food with lactic acid or propionic bacteria [19]. Another strategy aimed to enrich animal products with natural folic acid through the addition of high doses of synthetic folic acid to animal feed. In another study, supplementation of hen’s diet with folate enriched hen’s eggs to a maximum folate content of approximately 2.5 times that of a normal egg [24]. Increasing fiber concentration of cow’s diet was positively associated with vitamin B12 concentration in milk, whereas a negative relationship was established between folate and dietary fiber concentrations [25,26]. Furthermore, intramuscular injection of vitamin B12 in early-lactation dairy cows increased vitamin B12 content in milk [27].

The association of impaired folate levels with various pathological consequences led to the initiation of exogenous supplementation and biofortification approaches. Interventional trials aimed at proving the linkage of maternal vitamin B12 consumption with the concentration of vitamin B12 in breast milk showed that vitamin B12 concentration in human milk can be modified via supplementation during periconceptional and lactational periods [28]. Implementation of mandatory fortification of cereal grains with vitamin B12 was attempted in 1998 in the United States, followed by several other countries in subsequent years. However, despite evidence of the benefits of fortification, concern has been raised regarding the detrimental effects of exposure to synthetic or crystalline folic acid, the most important being the masking of vitamin B12 deficiency and its progression leading to irreversible neurological deficits [29].

For the benefit of vulnerable populations, a holistic approach of triple fortification (iron, iodine, folic acid) and quadruple fortification (iron, iodine, folic acid, vitamin B12) has been undertaken recently [30,31]. With increasing attention on the nutritional benefits of insects, the folic acid content in two cricket species, namely, Scapsipedus icipe and Gryllus bimaculatus, was measured and found to be much higher than animal and plant-based food sources, making crickets a cheaper alternative as opposed to commercial supplements. This demonstrated the need for the inclusion of crickets into the human diet, especially in resource-poor populations [32]. In a study directed at isolating strains of lactobacilli synthesizing the highest folate concentration by milk fermentation, it was found that Lactobacillus fermentum (from kefir granules), Lactobacillus plantarum (from salted mustard), and Lactobacillus rhamnosus (from breast milk) produced the highest concentrations of folic acid in the absence of pre-existing folate in milk [33]. An interventional study conducted in India to estimate the potentiality of tea as a scalable vehicle for fortification with vitamins B9 and B12 successfully showed that the daily consumption of a cup of vitamin-fortified tea for two months could benefit women of reproductive age [34]. Investigations on the dietary value of freshwater living green unicellular algae Chlorella species proved the presence of large amounts of folate. Moreover, a meta-analysis on the effect of its supplementation on cardiovascular risk factors showed that it improves total cholesterol levels, low-density lipoprotein-cholesterol (LDL-C) levels, systolic blood pressure, diastolic blood pressure, and fasting blood glucose levels [35].

With the advent of metabolic engineering, new studies are being conducted to assist in the construction of microbial cell factories for large-scale vitamin B12 production [36]. All these studies have the common goal of finding ways to improve folate and cobalamin levels in the general population, thus emphasizing the importance of these vitamins in the prevention of avoidable hematological and neurological complications.

Metabolism

Understanding the metabolism and the effects of alterations in each of its steps is of utmost importance as it would help us interpret and prognosticate the pathological outcomes and therapeutic possibilities. The metabolic processes of vitamins B9 and B12 are complex and interrelated at the cellular level. Following the breakdown of cobalamin, its reduced form is directed toward the two enzymes, namely, cytosolic methionine synthase and mitochondrial methylmalonyl-coenzyme A mutase (MUT) [37]. MUT utilizes an adenylated form of cobalamin in the catabolism of branched-chain amino acids, odd-chain fatty acids, and the side chain of cholesterol, whereas methionine synthase requires a methylated form of cobalamin and catalyzes the remethylation of homocysteine to methionine (using 5-methyltetrahydrofolate [CH3-THF] as a methyl donor) [37]. The methionine produced is further converted to S-adenosyl methionine (AdoMet, often called SAM), whose methyl group can be further donated to essentially important methylated compounds such as creatine, epinephrine, sarcosine, methylated DNA, RNA, and proteins [37].

The linkage of cobalamin metabolism with the folate-mediated one-carbon metabolism is attributed to the utilization of CH3-THF as the methyl donor [37]. CH3-THF is the most common form of folate in blood and tissues [38]. Depending on the cellular requirements, the folate-mediated one-carbon metabolic pathway helps folate to interchange between its different forms.

This metabolic pathway is mainly compartmentalized between cytosol and mitochondria depending on the mono/polyglutamate tail of folate which serves as a localization signal within the cell [39]. Even though two separated parallel pathways exist in cytosol and mitochondria, mitochondrial one-carbon oxidation accounts for approximately 50% of the nicotinamide adenine dinucleotide phosphate produced in the cell [40]. The deficiency of vitamin B12 and/or folate leads to the disruption of the one-carbon cycle and its downstream metabolic processes. Therefore, a deeper understanding of the cause and effect of the one-carbon metabolism is essential to obtain considerable insights into its role in the development of metabolic diseases [41].

Interactions

Mammalian cells exhibit two different vitamin B12-dependent reactions: conversion of methylmalonyl coenzyme A to succinyl coenzyme A by the enzyme MUT and the methylation of homocysteine to methionine by methionine synthase. The deficiency of vitamin B12 results in elevated levels of homocysteine and methylmalonic acid in the circulation. These function as biochemical markers of vitamin B12 status. Studies have proven that excess folate resulting mostly due to high folic acid intake interferes with cobalamin metabolism and worsens conditions associated with vitamin B12 insufficiency [42].

Malabsorption of vitamin B12 among the elderly has been observed commonly due to gastric atrophy or as a result of Helicobacter pylori infection. Pernicious anemia due to lack of gastric acid and intrinsic factors is another cause of B12 malabsorption. Low gastrointestinal motility resulting from gastrointestinal surgery, including gastric bypass, jejunal diverticulosis, blind-loop syndrome after intestinal surgery, and structural abnormalities such as diverticulosis in the duodenum or jejunum lead to bacterial overgrowth in the upper intestine, which, in turn, impairs the absorption of vitamin B12 from food. The proposed mechanisms for this include competition with intestinal bacteria for the uptake of vitamin B12, bacterial conversion of the vitamin to inactive analogs, and hypochlorhydria [43,44]. Some of the infections interfering with vitamin B12 absorption are fish tapeworm infection, Giardia lamblia infection, malaria, human immunodeficiency virus infection, and acquired immunodeficiency syndrome [44]. Tropical sprue, celiac disease, Zollinger-Ellison syndrome, Imerslund-Gräsbeck syndrome, juvenile pernicious anemia, pancreatic insufficiency, and defects in haptocorrin binding proteins are some of the pathological conditions associated with reduced cobalamin absorption [44]. Chronic pancreatitis and cystic fibrosis result in impaired trypsin secretion which is required for the release of vitamin B12 from transcobalamin-1 in the intestine [44].

Drugs interacting with vitamin B12 absorption include histamine receptor antagonists such as famotidine and proton-pump inhibitors such as omeprazole and lansoprazole. Cimetidine has been shown to inhibit the secretion of gastric acid, pepsin, and intrinsic factors [45-47]. A dose-related effect of cimetidine on vitamin B12 absorption was observed with intakes >1,000 mg/day reducing food-bound vitamin B12 absorption, but not 400 mg/day [45,48]. Similarly, a 20 mg/day dose of omeprazole reduced vitamin B12 absorption from a protein-bound source by approximately 70%, while 40 mg/day reduced absorption by approximately 90% [49].

Moreover, mild transcobalamin I deficiency has been linked to the risk of low serum cobalamin concentration. A common gene polymorphism was observed in vitamin B12 carrier protein, transcobalamin II, where proline (P) is substituted for arginine (A), affecting the mean total plasma concentrations of vitamin B12 and homocysteine [50]. Unlike folic acid, dietary folates are relatively unstable to oxidation and heat; hence, large losses can occur during food preparation and cooking. Boiling has been shown to destroy up to 80% of folate in green vegetables, whereas chopping and grinding spinach has been shown to increase folate bioavailability [51,52]. Furthermore, ascorbic acid in foods was shown to improve folate stability [53].

Chronic alcoholics are prone to vitamin B9 deficiency due to low folate intake, poor folate absorption due to impaired transcription of the intestinal folate carrier, reduced liver uptake and storage, and increased urinary excretion [54]. The speed of onset of alcoholic liver disease was assessed in a micro-pig model, which demonstrated that the onset was more rapid when the animals were provided with a folate-deficient diet than when folate intake was adequate [55]. Medications that impair folate status include methotrexate (a folate antagonist used in the treatment of rheumatoid arthritis, inflammatory bowel disease, etc.), anticonvulsants (phenytoin, phenobarbital, and Dilantin), sulfasalazine (for the treatment of chronic ulcerative colitis), and pyrimethamine (for the treatment of malaria), as well as large doses of nonsteroidal anti-inflammatory drugs [44]. The 677 C→T (cytosine-thymine) gene polymorphism in methyltetrahydrofolate reductase was found to affect up to 30% of various population groups studied, mainly in developed countries, and the homozygote was associated with increased plasma homocysteine level and low serum and erythrocyte folate levels [56].

Epidemiology of insulin resistance

The worldwide incidence of MS ranges from 10% to 40% depending upon the region and the criteria used to define it [57]. The evolution of MS depends upon several factors such as obesity and insulin resistance [58]. These factors, in turn, result in an increased probability of type 2 diabetes mellitus [58,59]. Furthermore, a strong correlation exists between obesity, insulin resistance, and polycystic ovarian syndrome (PCOS) in women [60].

In the United States alone, the incidence of childhood obesity has nearly tripled from 7% to 18% between 1980 and 2013 [61]. Developing nations have also seen a 4.8% rise in obesity from 1980 to 2013 [62]. In 2014, 13% of the adult population was obese [63]. Concurrently, there were reportedly 422 million insulin-resistant diabetics in the world [64]. These numbers are only expected to escalate in the future and are predicted to increase up to 649 million by 2040 [65]. It is important to note that there has been a significant number of diabetes-related deaths globally (1.6 million) [64]. Type 2 diabetes is also a common side effect of PCOS [66]. Approximately, 60% to 80% of PCOS-affected women suffer from insulin resistance [67].

Pathophysiology of metabolic syndrome/insulin resistance

There is no specific mechanism that leads to the development of MS. Multiple studies have reported that there is an association between insulin resistance and visceral adiposity [68]. Thus, being a state that predisposes to several comorbidities, additional clinical conditions such as hypertension and dyslipidemia may emerge. In addition, it can be seen as a condition in which there is an increment in both thrombotic and inflammatory states distinguished by an elevation in the inflammatory cytokine activity [69].

Insulin resistance is a decrease in the biological function of insulin, irrespective of the serum concentration [70]. Insulin resistance can be taken as the main pathophysiology of MS. Second, the increased prevalence of insulin resistance is related to the global increase in visceral adiposity [71]. Central obesity can emerge from multiple factors, such as an inappropriate diet of consuming higher processed calories, genetic predisposition, or epigenetic factors that can relate to the environment. Recently, a correlation has been reported between the high intake of fats and an inadequate function of the intestinal barrier that can ease the movement of intestinal luminal content and bacterial lipopolysaccharide into the circulation, which throws further light on its pathophysiology [72-74].

One of the main causes that develop this resistance mechanism is the excess of free fatty acids circulating from an expanded adipose tissue mass. This additional fat decreases the insulin sensitivity in the skeletal muscle by inhibiting the glucose uptake mediated by insulin [72]. This also increases the production of endogenous glucose. With the failure of glucose transporter type 4 (GLUT4), which is mainly located in adipocytes and muscle cells, glucose intake is reduced [75-79]. This causes an increase in circulating glucose that leads to increased insulin secretion. Consequently, glucose transporter types 1-3 (GLUT1-3), which are insulin-independent and found in neurons, renal cells, and erythrocytes, are exposed to a large amount of glucose that leads to glucotoxicity [75]. This results in prediabetes, a state characterized by glucose intolerance [80,81]. Additionally, the continuation of insulin resistance can lead the beta cells of the pancreas to get to a point where they can no longer provide the high amounts of insulin secreted before, leading to a drop in insulin secretion and further failure of glucose homeostasis [80,82].

Furthermore, there is an increase in the synthesis of triglycerides and very-low-density lipoproteins (VLDL) that contribute to the failure of the GLUT4 transporter [72,80]. According to the portal theory of MS, there are free fatty acids that are released from the accumulation of visceral fat. They enter the portal circulation and are transported to the liver where they remain as triglycerides [83,84]. This induces the release of VLDL by the liver and causes a state of hypertriglyceridemia [85]. These free fatty acids act on the phosphoinositide-3-kinase activity reducing its function and worsening insulin resistance [80]. These fats also contribute to the reduced production of insulin by the beta cells of the pancreas by exerting a lipotoxic effect on them [77]. Moreover, they contribute to endothelial effect by producing reactive oxygen species, along with the hyperinsulinemia state and the cytokines produced by the adipose tissue [83,86-88].

Regarding MS and inflammation, the state of inflammation in this syndrome is associated with damage of the gut barrier activity and bacterial exposure [89,90]. This endotoxemia may activate inflammatory pathways, along with further encroachment to various organs that can lead to an invasion of macrophages that help promote insulin resistance [91-93]. Additionally, it contributes to the accumulation of adiposity and insulin resistance [94].

There are several ways by which the microbiome may contribute to the development of obesity such as encouraging the accumulation of fat deposits and change in the locomotor activity setting off systemic inflammation [95]. Recently, a correlation has been reported between the high intake of fats and an inadequate function of the intestinal barrier that can ease the movement of intestinal luminal contents and bacterial lipopolysaccharide into the circulation [94,96]. These lipopolysaccharide chain cluster of differentiation 14 (CD14) receptors promote systemic inflammation [97].

Current treatment protocols 

As mentioned above, MS is a multifaceted disease. Therefore, the management approach is targeted toward the prevention and reduction of cardiovascular risk factors such as obesity, type 2 diabetes mellitus, and hypertension [98]. Treatment goals are achieved through various modalities such as lifestyle modification and pharmaceutical management in high-risk individuals [99].

Lifestyle Modifications

To reduce the incidence of MS, early measures such as a low carbohydrate diet, increased physical activity, and controlled alcohol consumption are targeted, in particular, toward the younger generation [100,101]. Lifestyle changes have proven to be the more effective approach to reducing the incidence of MS and diabetes. In the Diabetes Prevention Study conducted over three years, a diabetic case reduction of 41% was seen with lifestyle changes compared to a 17% reduction only with metformin [102]. A diet commonly referred to as “the Mediterranean diet” is efficacious in preventing insulin resistance [103]. A Mediterranean diet consists of a wide range of fruits, vegetables, whole grains, and healthy fats such as seeds, nuts, and extra virgin olive oil. A limited quantity of fish, dairy, poultry, and red meat are also incorporated [104]. The rich plant sources consist of phenolic compounds that modify the insulin activity in the tissue and improve sensitivity [103,105]. Research-based recommendations for diet include 45% to 60% of carbohydrate intake (sourced from legumes and whole grains predominantly), 10% to 20% protein, and 10% fatty acids. [106,107]. High fiber intake (40 g/day) also improves insulin sensitivity [107]. Although regular moderate physical exercise improves MS [108], the best results are observed when exercise is implemented in conjunction with an appropriate diet. A study conducted by Anderssen et al. compared the effects of combined interventions with those without dietary restrictions. They reported a 67% and 24% drop in MS incidence in the combined and isolated groups, respectively [109]. An intervention study, the Da Qing Trial, further supported the abovementioned findings. A more significant reduction of type 2 diabetes mellitus in the combined diet and exercise groups was observed compared to the isolated diet and activity groups (46% vs. 43.8% vs. 41.1%, respectively) [110].

Pharmaceutical Management

A well-established relationship exists between MS and type 2 diabetes mellitus [111], indicating that strict glycemic control can eliminate the risk factors of MS [112]. The two classes of drugs that predominantly affect insulin resistance are biguanides (metformin) and thiazolidinediones (glitazones) [113,114]. Metformin achieves the desired results through various mechanisms such as inhibiting glucose production in the liver [115], increasing glucose reuptake via insulin receptor activation, stimulating tyrosine kinase in the peripheral tissue [113,116], and lowering free fatty acid oxidation in adipose tissues [117]. A study on the mechanism of metformin by Hundal et al. demonstrated a 25% to 30% decrease in plasma glucose because of the actions of metformin on hepatocytes [115].

Glitazones improve metabolic control by binding with nuclear receptor PPAR-γ and controlling glucose metabolism [118]. PPARs are ligand-inducible transcription factors that belong to the nuclear hormone receptors family. Three different isoforms of PPARs have been identified, namely, PPAR-α, PPAR-β/δ, and PPAR-γ. These are involved in adipogenesis, inflammation, lipid, and glucose metabolism [119,120].

Fenofibrate, the prototype of fibrate drugs, activates PPAR-α whereas bezafibrate is a pan-PPAR activator for all three PPAR isoforms. In a recent meta-analysis by Simental-Mendía et al., fibrate therapy significantly reduced fasting plasma glucose (-0.28 mmol/L), insulin levels (-3.87 pmol/L), and insulin resistance [Homeostasis Model of Assessment for Insulin Resistance (HOMA-IR) -1.09], with no effect on glycated hemoglobin (HbA1c) [121]. These effects are more pronounced with bezafibrate as it is a pan-PPAR activator.

All three drug classes have been hypothesized to have a protective effect on the cardiovascular system [122-124]. However, these results are debatable and require further investigation.

Several other targeted therapies are implemented with the predominant metabolic risk factor in mind. In severe hypertension, newly introduced angiotensin receptor blockers (telmisartan and irbesartan) have shown promising results in comparison to other antihypertensive medications. These drugs behave similarly to glitazones and stimulate PPAR-γ, resulting in a reduction of blood pressure and an increase in insulin sensitivity [125].

In severe obesity (a BMI of at least 30 kg/m2), where all other treatments have been unsatisfactory, pharmacological management can be considered [126]. The current treatment options include sibutramine and orlistat. A meta-analysis of randomized trials on sibutramine showed a 4.6% more weight loss than the placebo [127]. Increased insulin sensitivity and better glycemic control were also reported among diabetics [128]. In a diabetic study [129], a combined orlistat and diet regimen reduced MS by 35% versus 9% (diet alone). In addition to weight loss, orlistat also prevents type 2 diabetes mellitus and hyperglycemia [130].

Statins are primarily used as LDL-C-lowering agents in patients who meet the criteria for atherosclerotic cardiovascular disease (ASCVD) with MS. Its pleiotropic effects have proven to be significantly beneficial in patients with MS in reducing major coronary events [2,98]. MS has been associated with elevated C-reactive protein (CRP); statins lower CRP and reduce pro-inflammatory states, further exhibiting their prognostic value [98,99]. An interventional study in patients with MS focused on measuring effective reductions in cholesterol using rosuvastatin therapy (MERCURY Trial) found 10 mg of rosuvastatin to be more promising compared to commonly used doses of other statins in achieving the target LDL-cholesterol level [98]. A similar study conducted by Grundy et al., the Comparative study with Rosuvastatin in Subjects with Metabolic Syndrome (COMETS), also demonstrated rosuvastatin to be an effective lipid-modifying agent in patients with MS. Moreover, recent studies have established that the combination of statins and fibrates or nicotinic acid is more efficient in reducing cardiovascular outcomes in addition to minimizing the risks of myopathy [99].

Effectiveness of folate and vitamin B12 supplementation in insulin resistance and metabolic syndrome

Folate supplementation has been established with protective studies in reducing insulin and HOMA-IR; however, inconsistent results have been reported regarding the reduction of fasting blood sugar and HbA1c levels [131]. The dosage of folate supplementation plays an important role in the reduction of insulin resistance [132]. The glycemic markers were effectively reduced with a folate dose greater than 5 mg/day [132]. A systematic review published by Xie et al. in 2016 showed that folic acid supplementation in maternal and prenatal animal groups decreased both insulin resistance and obesity significantly [133]. Further, folate prevented the decline in cognitive skills associated with metformin use in diabetes [134]. To support this, we reviewed a clinical study conducted by Porter et al. (2019) in a group of community-dwelling older adults and found that using fortified vitamin B12 foods improved the patient’s cognitive status [134]. Scientific studies have evolved, paving the way for newer and promising treatments for insulin resistance. One such study used folate nanoparticles in rats to achieve an effective glycemic index [135]. It was demonstrated that using folate nanoparticles showed a five-fold enhancement in cellular uptake of insulin and that blood glucose levels were controlled effectively [135].

Furthermore, it was evident that dietary intake of folate and vitamin B12 had a positive impact on MS and reduced the risk of stroke in both men and women in the United States [136,137]. Spence et al. (2016) showed that supplementation with methylcobalamin minimized the incidence of stroke in persons with renal impairment and cyanocobalamin in patients without any renal impairment [138]. To highlight the relationship between low supplementation of folate and vitamin B12 and stroke, we reviewed a randomized controlled trial (RCT) conducted by Qin et al. (2020) in a double-blinded hypertensive population. The study revealed that people with low levels of folate and vitamin B12 were associated with an increased risk of ischemic stroke [139]. The duration of folate supplementation influenced the incidence of stroke by a reduction of 18% in people who consumed it for more than 36 months [140].

Another area of interest was to assess the effect of folate/vitamin B12 on cardiovascular diseases. Studies have highlighted plasma homocysteine levels to play an important role in the occurrence of myocardial infarction, and its levels were predominantly dependent on plasma folate values to a greater extent and vitamin B12 to a lesser extent [141]. This was further supported by a prospective study conducted by Stampfer et al. (1992) which assessed the risk of plasma homocysteine and cardiovascular diseases in U.S. physicians and found that the levels of homocysteine were higher in cases than in controls (11.1 ± 4.0 standard deviation [SD] vs. 10.5 ± 2.8 nmol/mL) who later developed myocardial infarction, irrespective of other modifiable risk factors [142]. Furthermore, low vitamin B12 levels have been associated with the development of atherosclerosis, which is an important causative factor for cardiovascular diseases [143]. Moreover, because hyperhomocysteinemia has been linked with congenital heart disease malformation in children, periconceptional folate supplementation has proven to be a key factor in reducing congenital heart disease in children [144].

Along with cardiovascular diseases and stroke, another common condition associated with low folate/vitamin B12 levels is obesity [145]. A study by Kumar et al. (2012) included female Wistar rats which were deprived of a vitamin B12 diet and allowed to mate [146]. Biochemical and body parameters were measured in maternal rats before mating and in offspring at three, six, nine, and twelve months of age. It was demonstrated that the maternal rats suffered from increased body fat and altered lipid profile and the offspring had an increase in fatty acid synthase and higher plasma cortisol levels, thus paving the way to high visceral fat at three months and dyslipidemia at 12 months [146]. Therefore, it is recommended that pregnant females remain careful about their folate/vitamin B12 supplementation to prevent dyslipidemia and neural tube defects in the offspring [147]. It has been reported that both folate and vitamin B12 have a synergistic effect on the markers of obesity and lipid profile, such as the waist to hip length ratio, triglyceride level, and HDL-C level [148-150]. However, further clinical studies are required to warrant any unwanted side effects with the use of folate and vitamin B12 for insulin resistance and MS.

Novel therapeutic approaches and targets

MS is a constellation of disorders that usually involves the treatment of the individual disorder, along with dietary intervention and physical activity [98]. Due to a better understanding of the pathophysiology of MS, novel agents acting multiple targets and mechanisms are being developed [151].

Role of Selective Peroxisome Proliferator-Activated Receptor Modulators

Novel compounds, such as selective peroxisome proliferator-activated receptor modulators (SPPARMs), have more potent PPAR-α agonist activity. These are proven to have advantages in the treatment of insulin resistance and dyslipidemia. One such agent is pemafibrate. Pemafibrate is a novel, highly selective PPAR-α agonist approved for the treatment of hyperlipidemia in Japan. The recommended dosage of oral pemafibrate is 0.1 mg twice daily, which can be adjusted to a maximum of 0.2 mg twice daily [152]. It is more robust in reducing triglycerides and elevating HDL-C, thereby enhancing reverse cholesterol transport compared to fenofibrate [153]. Based on studies in mouse models, pemafibrate is more efficient in decreasing apolipoprotein B-48 (ApoB-48)-containing chylomicron remnants than those containing ApoB-100. It is noteworthy that ApoB-48 is more atherogenic [154]. In an RCT conducted by Araki et al. on type 2 diabetes mellitus patients (HbA1c ≥ 6.2%) with fasting triglycerides of ≥150 mg/dL, not on statins, participants were randomly assigned to pemafibrate at doses of 0.2 mg/day, 0.4 mg/day, and placebo. After 24 weeks of treatment, triglycerides declined by 45% in both doses; fasting triglycerides of ≤150 mg/dL were achieved in 70.9% and 81.5% of patients on 0.4 mg/day and 0.2 mg/day, respectively. No considerable changes in LDL-C were noted. However, an increment in HDL-C and ApoA-I was observed [155]. Among glycemic parameters, a significant decrease in HOMA-IR was noticed in the 0.2 mg/day pemafibrate group, while no significant changes were noted in other parameters. The most common adverse effects reported were increased serum creatinine and liver enzymes [155].

The most recent evidence regarding pemafibrate is yet to be obtained through a Pemafibrate to Reduce Cardiovascular OutcoMes by Reducing Triglycerides IN patiENts With diabeTes (PROMINENT) study which is a multicentre, randomized, phase 3 trial. The study recruited approximately 10,000 participants from 24 countries with type 2 diabetes mellitus, mild-to-moderate hypertriglyceridemia, and low HDL-C levels to either pemafibrate (0.2 mg twice daily) or matching placebo. The trial is currently ongoing with a mean follow-up period of 3.75 years [156]. Further studies are needed to determine the effect of pemafibrate in preventing cardiovascular disease.

Farnesoid X Receptor Agonists

Farnesoid X receptor (FXR) belongs to the nuclear receptor family. Among bile acids, chenodeoxycholic acid is the most potent activator of FXR. These receptors are expressed in the liver, kidney, intestine, and adrenal glands [157]. Activation of FXR has been shown to increase insulin sensitivity and decrease triglycerides and free fatty acid levels. Recent data also suggest reduced endothelin levels, thereby playing a role in atherosclerosis [157]. Recently, obeticholic acid, an FXR agonist, has been approved for the treatment of primary biliary cholangitis. Newer FXR agonists such as nidufexor or tropifexor are under phase 2 clinical trials for the treatment of nonalcoholic fatty liver disease and non-alcoholic steatohepatitis, considered to be a hepatic manifestation of MS [158,159].

Asprosin and Asprosin Neutralizing Antibodies

Asprosin, a glucogenic adipokine, was discovered in 2016 in the study of the neonatal progeroid syndrome [160]. Sites of action include the liver and the brain. In the liver, it exerts a glucogenic effect through the cyclic adenosine monophosphate protein kinase A (cAMP-PKA)-dependent pathway [161]. It crosses the blood-brain barrier and stimulates appetite by activating orexigenic and inhibiting anorexigenic neurons in the arcuate nucleus of the hypothalamus. The levels have been found to be elevated in MS [161,162].

A preclinical study done on mice by Mishra et al. showed that a single dose of an anti-asprosin monoclonal antibody (mAb) reduced food intake by an average of 1 g/day, resulting in decreased body weight. Other parameters such as triglycerides, blood glucose, and plasma insulin levels also decreased [162]. Anti-asprosin mAbs also exert action at the target level by inhibiting hepatic cAMP signaling and orexigenic neuron activity in the hypothalamus, in addition to neutralizing asprosin [161,162]. Further studies are required in humans to delineate efficacy and safety, and these preclinical study results offer better hope for the future.

Peripheral Cannabinoid Receptor Antagonists

Selective cannabinoid (CB) receptor inhibition is a novel approach to treat MS and dyslipidemias. Cannabinoid receptors CB-1 and CB-2 are G protein-coupled receptors that activate G proteins (Gi/Go). CB-1 receptors are expressed in the brain and peripherally in the heart, liver, pancreas, and adipose tissue, whereas CB-2 receptors are found in immune cells [163]. Although the exact mechanism of CB receptor antagonists is not well understood, they have been found to modulate leptin levels. Another mechanism is through the activation of the peripheral sympathetic system [164].

The use of the CB-1 receptor antagonist dates back to 2006. Rimonabant, approved for the treatment of obesity, was withdrawn in 2008 because of an increase in depressive mood disorders associated with antagonism of central CB-1 receptors [165]. This leads to the development of compounds with limited central nervous system penetration and action on peripheral CB receptors. However, further studies are needed to demonstrate relevant metabolic endpoints [166]. Newer compounds of the CB receptor family which are in the preclinical phases of investigation are showing promising results [167,168].

Role of Fecal Microbiota Transplantation in Insulin Resistance

The human gut microbiome harbors an estimated 10-100 trillion microorganisms. Gut microbial dysbiosis has been defined as alterations in diversity with more “inflammatory” microbes (i.e., Proteobacteria) and decreased short-chain fatty acid levels [169].

A recent study among 154 human twins shed light on the link between obesity and gut microbial diversity. Excess Bacteroidetes and deficient butyrate and propionate levels were noted in the gut microbiome of twins with obesity phenotype [170,171]. Two randomized control trials conducted by Kootte et al. and Vrieze et al. showed similar effects, such as improvement in insulin sensitivity and increased microbial diversity at six weeks after allogenic fecal microbiota transplantation (FMT) compared to autologous FMT [171,172]. Further, when metabolic parameters were measured at 18 weeks in the study by Kootte et al., no changes were observed. This study showed that the short-term effects of FMT are not maintained in the long term [171].

Although the role of FMT in insulin resistance is recognized, the role of FMT in hypertension is unclear. In studies on rat models for hypertension, a decreased microbiota diversity in the form of decreased Firmicutes/Bacteroidetes ratio was observed. Interestingly, the blood pressure in these rat models could be controlled by reversing the Firmicutes/Bacteroidetes ratio by utilizing antibiotics [173,174]. More controlled trials are needed to elucidate the potential benefit and sustainability of FMT in MS.

Adenosine Triphosphate Citrate Lyase Inhibitors

Adenosine triphosphate citrate lyase (ACLY) is a cytoplasmic enzyme that mediates the production of acetyl coenzyme A (acetyl-CoA) that acts as a substrate for the de novo synthesis of fatty acids and cholesterol [175].

Bempedoic acid, an ACLY inhibitor, is approved by the U.S. Food and Drug Administration (US FDA) for the treatment of heterozygous familial hypercholesterolemia (HeFH) and established ASCVD [175]. Bempedoic acid modulates adenosine monophosphate-activated protein kinase (AMPK) activation and decreases hepatic glucose production, an action similar to metformin [176]. In a randomized, 52-week, phase 3 trial, involving adults with ASCVD, HeFH, or both, bempedoic acid at a dose of 180 mg once daily reduced the mean LDL-C level by 12.6% by week 52 [177]. The most frequent adverse effects observed included nasopharyngitis, muscle spasms, hyperuricemia, anemia, and elevated transaminases [177,178]. ACLY inhibitors can be used as the primary drug in patients who are intolerant to statins. Further studies are needed to evaluate the role of ACLY inhibitors in reducing inflammation, insulin resistance, and other components of MS.

Central Leptin Gene Therapy

The role of leptin resistance in MS has long been proven. Leptin, an adipogenic hormone, acts on the hypothalamus and increases satiety [179]. Burguera et al. postulated that “leptin resistance might be due to non-availability of leptin at hypothalamic target sites caused by either defective uptake across the blood-brain barrier, or abnormal post-receptor signal transduction” [180].

Using recombinant adeno-associated virus vector for central leptin gene therapy in animal models showed decreased weight, insulin resistance, and increased thermogenesis [181]. Further long-term studies are required in humans as leptin gene therapy can offer a permanent solution to multiple components of MS.

Conclusions

This review aimed to understand the interactions between MS and vitamins B12 and folate and summarized the most relevant literature. To date, MS remains somewhat of an enigma as there is no consensus on how or why it develops. However, progress has been made on the therapeutic end, with a whole host of novel drugs being developed to treat the condition. Moreover, the impact of folate and vitamin B12 supplementation is a new frontier that is still relatively unexplored. We recommend further observational studies and RCTs that examine exactly how much vitamin supplementation affects each component of MS. Current literature suggests that these vitamins have a significant positive impact on MS, and they must be kept in mind while designing new treatment protocols in the future.

The content published in Cureus is the result of clinical experience and/or research by independent individuals or organizations. Cureus is not responsible for the scientific accuracy or reliability of data or conclusions published herein. All content published within Cureus is intended only for educational, research and reference purposes. Additionally, articles published within Cureus should not be deemed a suitable substitute for the advice of a qualified health care professional. Do not disregard or avoid professional medical advice due to content published within Cureus.

Footnotes

The authors have declared that no competing interests exist.

References

  • 1.Clinical management of metabolic syndrome: report of the American Heart Association/National Heart, Lung, and Blood Institute/American Diabetes Association conference on scientific issues related to management. Grundy SM, Hansen B, Smith SC Jr, Cleeman JI, Kahn RA. Arterioscler Thromb Vasc Biol. 2004;24:0–24. doi: 10.1161/01.ATV.0000112379.88385.67. [DOI] [PubMed] [Google Scholar]
  • 2.Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) Expert Panel on Detection, Evaluation Evaluation, and Treatment of High Blood Cholesterol in Adults. JAMA. 2001;285:2486–2497. doi: 10.1001/jama.285.19.2486. [DOI] [PubMed] [Google Scholar]
  • 3.The metabolic syndrome. Cornier MA, Dabelea D, Hernandez TL, et al. Endocr Rev. 2008;29:777–822. doi: 10.1210/er.2008-0024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Global burden of obesity in 2005 and projections to 2030. Kelly T, Yang W, Chen CS, Reynolds K, He J. Int J Obes (Lond) 2008;32:1431–1437. doi: 10.1038/ijo.2008.102. [DOI] [PubMed] [Google Scholar]
  • 5.B vitamins and one-carbon metabolism: implications in human health and disease. Lyon P, Strippoli V, Fang B, Cimmino L. Nutrients. 2020;12:2867. doi: 10.3390/nu12092867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Implication of homocysteine in diabetes and impact of folate and vitamin B12 in diabetic population. Mursleen MT, Riaz S. Diabetes Metab Syndr. 2017;11 Suppl 1:0–6. doi: 10.1016/j.dsx.2016.12.023. [DOI] [PubMed] [Google Scholar]
  • 7.The molecular and cellular effect of homocysteine metabolism imbalance on human health. Škovierová H, Vidomanová E, Mahmood S, et al. Int J Mol Sci. 2016;17:1733. doi: 10.3390/ijms17101733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.[Association between metabolic syndrome and hyperhomocysteinemia in an Algerian population] Zendjabil M, Abbou O, Chellouai Z. Ann Pharm Fr. 2017;75:54–58. doi: 10.1016/j.pharma.2016.05.001. [DOI] [PubMed] [Google Scholar]
  • 9.Association of homocysteine with type 2 diabetes: a meta-analysis implementing Mendelian randomization approach. Huang T, Ren J, Huang J, Li D. BMC Genomics. 2013;14:867. doi: 10.1186/1471-2164-14-867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Effects of B-vitamins on plasma homocysteine concentrations and on risk of cardiovascular disease and dementia. Clarke R, Lewington S, Sherliker P, Armitage J. Curr Opin Clin Nutr Metab Care. 2007;10:32–39. doi: 10.1097/MCO.0b013e328011aa71. [DOI] [PubMed] [Google Scholar]
  • 11.Effect of vitamin B6, B9, and B12 supplementation on homocysteine level and cardiovascular outcomes in stroke patients: a meta-analysis of randomized controlled trials. Kataria N, Yadav P, Kumar R, Kumar N, Singh M, Kant R, Kalyani V. Cureus. 2021;13:0. doi: 10.7759/cureus.14958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Folic acid and vitamin B12 supplementation in subjects with type 2 diabetes mellitus: a multi-arm randomized controlled clinical trial. Satapathy S, Bandyopadhyay D, Patro BK, Khan S, Naik S. Complement Ther Med. 2020;53:102526. doi: 10.1016/j.ctim.2020.102526. [DOI] [PubMed] [Google Scholar]
  • 13.Vitamin B12 in meat and dairy products. Gille D, Schmid A. Nutr Rev. 2015;73:106–115. doi: 10.1093/nutrit/nuu011. [DOI] [PubMed] [Google Scholar]
  • 14.Dietary sources and intakes of folates and vitamin B12 in the Spanish population: findings from the ANIBES study. Partearroyo T, Samaniego-Vaesken ML, Ruiz E, et al. PLoS One. 2017;12:0. doi: 10.1371/journal.pone.0189230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gender and age influence blood folate, vitamin B12, vitamin B6, and homocysteine levels in European adolescents: the Helena Study. González-Gross M, Benser J, Breidenassel C, et al. Nutr Res. 2012;32:817–826. doi: 10.1016/j.nutres.2012.09.016. [DOI] [PubMed] [Google Scholar]
  • 16.Projected prevalence of inadequate nutrient intakes in Europe. Roman Viñas B, Ribas Barba L, Ngo J, et al. Ann Nutr Metab. 2011;59:84–95. doi: 10.1159/000332762. [DOI] [PubMed] [Google Scholar]
  • 17.High compliance with dietary recommendations in a cohort of meat eaters, fish eaters, vegetarians, and vegans: results from the European Prospective Investigation into Cancer and Nutrition-Oxford study. Sobiecki JG, Appleby PN, Bradbury KE, Key TJ. Nutr Res. 2016;36:464–477. doi: 10.1016/j.nutres.2015.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Serum concentrations of vitamin B12 and folate in British male omnivores, vegetarians and vegans: results from a cross-sectional analysis of the EPIC-Oxford cohort study. Gilsing AM, Crowe FL, Lloyd-Wright Z, Sanders TA, Appleby PN, Allen NE, Key TJ. Eur J Clin Nutr. 2010;64:933–939. doi: 10.1038/ejcn.2010.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Vitamin B₁₂-containing plant food sources for vegetarians. Watanabe F, Yabuta Y, Bito T, Teng F. Nutrients. 2014;6:1861–1873. doi: 10.3390/nu6051861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wolffia globosa-Mankai plant-based protein contains bioactive vitamin B12 and is well absorbed in humans. Sela I, Yaskolka Meir A, Brandis A, et al. Nutrients. 2020;12:3067. doi: 10.3390/nu12103067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bioactive compounds of edible purple laver Porphyra sp. (Nori) Bito T, Teng F, Watanabe F. J Agric Food Chem. 2017;65:10685–10692. doi: 10.1021/acs.jafc.7b04688. [DOI] [PubMed] [Google Scholar]
  • 22.Isolation and analysis of vitamin B12 from plant samples. Nakos M, Pepelanova I, Beutel S, Krings U, Berger RG, Scheper T. Food Chem. 2017;216:301–308. doi: 10.1016/j.foodchem.2016.08.037. [DOI] [PubMed] [Google Scholar]
  • 23.Daily milk intake improves vitamin B-12 status in young vegetarian Indians: an intervention trial. Naik S, Bhide V, Babhulkar A, Mahalle N, Parab S, Thakre R, Kulkarni M. Nutr J. 2013;12:136. doi: 10.1186/1475-2891-12-136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Validation of folate-enriched eggs as a functional food for improving folate intake in consumers. Altic L, McNulty H, Hoey L, McAnena L, Pentieva K. Nutrients. 2016;8:777. doi: 10.3390/nu8120777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Impact of diet management and composition on vitamin B12 concentration in milk of Holstein cows. Duplessis M, Pellerin D, Robichaud R, Fadul-Pacheco L, Girard CL. Animal. 2019;13:2101–2109. doi: 10.1017/S1751731119000211. [DOI] [PubMed] [Google Scholar]
  • 26.Cross-sectional study of the effect of diet composition on plasma folate and vitamin B12 concentrations in Holstein cows in the United States and Canada. Duplessis M, Ritz KE, Socha MT, Girard CL. J Dairy Sci. 2020;103:2883–2895. doi: 10.3168/jds.2019-17657. [DOI] [PubMed] [Google Scholar]
  • 27.Short communication: influence of intramuscular injection of vitamin B12 in early-lactation dairy cows on Mozzarella cheese quality and vitamin B12 stability. Xu NN, Yang DT, Zhang BX, Liu JX, Ye JA, Ren DX. J Dairy Sci. 2020;103:9835–9840. doi: 10.3168/jds.2020-18568. [DOI] [PubMed] [Google Scholar]
  • 28.Vitamin B-12 in human milk: a systematic review. Dror DK, Allen LH. Adv Nutr. 2018;9:358–366. doi: 10.1093/advances/nmx019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Recent developments in folate nutrition. Naderi N, House JD. Adv Food Nutr Res. 2018;83:195–213. doi: 10.1016/bs.afnr.2017.12.006. [DOI] [PubMed] [Google Scholar]
  • 30.Technology for triple fortification of salt with folic acid, iron, and iodine. Modupe O, Krishnaswamy K, Diosady LL. J Food Sci. 2019;84:2499–2506. doi: 10.1111/1750-3841.14730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Quadruple fortification of salt for the delivery of iron, iodine, folic acid, and vitamin B12 to vulnerable populations. Modupe O, Diosady LL. J Food Eng. 2021;300:110525. doi: 10.1016/j.jfoodeng.2021.110525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.From farm to fork: crickets as alternative source of protein, minerals, and vitamins. Murugu DK, Onyango AN, Ndiritu AK, Osuga IM, Xavier C, Nakimbugwe D, Tanga CM. Front Nutr. 2021;8:704002. doi: 10.3389/fnut.2021.704002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Folate in milk fermented by lactic acid bacteria from different food sources. Mahara FA, Nuraida L, Lioe HN. Prev Nutr Food Sci. 2021;26:230–240. doi: 10.3746/pnf.2021.26.2.230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Potential for elimination of folate and vitamin B12 deficiency in India using vitamin-fortified tea: a preliminary study. Vora RM, Alappattu MJ, Zarkar AD, Soni MS, Karmarkar SJ, Antony AC. BMJ Nutr Prev Health. 2021;4:293–306. doi: 10.1136/bmjnph-2020-000209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Potential of chlorella as a dietary supplement to promote human health. Bito T, Okumura E, Fujishima M, Watanabe F. Nutrients. 2020;12:2524. doi: 10.3390/nu12092524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Microbial production of vitamin B12: a review and future perspectives. Fang H, Kang J, Zhang D. Microb Cell Fact. 2017;16:15. doi: 10.1186/s12934-017-0631-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vitamin B12, folate, and the methionine remethylation cycle-biochemistry, pathways, and regulation. Froese DS, Fowler B, Baumgartner MR. J Inherit Metab Dis. 2019;42:673–685. doi: 10.1002/jimd.12009. [DOI] [PubMed] [Google Scholar]
  • 38.Folic acid metabolism in human subjects revisited: potential implications for proposed mandatory folic acid fortification in the UK. Wright AJ, Dainty JR, Finglas PM. Br J Nutr. 2007;98:667–675. doi: 10.1017/S0007114507777140. [DOI] [PubMed] [Google Scholar]
  • 39.The intestinal absorption of folates. Visentin M, Diop-Bove N, Zhao R, Goldman ID. Annu Rev Physiol. 2014;76:251–274. doi: 10.1146/annurev-physiol-020911-153251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Quantitative flux analysis reveals folate-dependent NADPH production. Fan J, Ye J, Kamphorst JJ, Shlomi T, Thompson CB, Rabinowitz JD. Nature. 2014;510:298–302. doi: 10.1038/nature13236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.The role of the one-carbon cycle in the developmental origins of type 2 diabetes and obesity. Finer S, Saravanan P, Hitman G, Yajnik C. Diabet Med. 2014;31:263–272. doi: 10.1111/dme.12390. [DOI] [PubMed] [Google Scholar]
  • 42.Interaction between excess folate and low vitamin B12 status. Paul L, Selhub J. Mol Aspects Med. 2017;53:43–47. doi: 10.1016/j.mam.2016.11.004. [DOI] [PubMed] [Google Scholar]
  • 43.Small bowel bacterial overgrowth. Donaldson RM Jr. https://pubmed.ncbi.nlm.nih.gov/4916360/ Adv Intern Med. 1970;16:191–212. [PubMed] [Google Scholar]
  • 44.Causes of vitamin B12 and folate deficiency. Allen LH. Food Nutr Bull. 2008;29:0–7. doi: 10.1177/15648265080292S105. [DOI] [PubMed] [Google Scholar]
  • 45.Malabsorption of protein-bound cobalamin but not unbound cobalamin during cimetidine administration. Steinberg WM, King CE, Toskes PP. Dig Dis Sci. 1980;25:188–191. doi: 10.1007/BF01308137. [DOI] [PubMed] [Google Scholar]
  • 46.Effect of cimetidine on intrinsic factor and pepsin secretion in man. Binder HJ, Donaldson RM Jr. Gastroenterology. 1978;74:371–375. [PubMed] [Google Scholar]
  • 47.Effect of cimetidine on intrinsic factor secretion stimulated by different doses of pentagastrin in patients with impaired renal function. Vatn MH, Schrumpf E, Talseth T. Scand J Gastroenterol. 1983;18:1109–1114. doi: 10.3109/00365528309181849. [DOI] [PubMed] [Google Scholar]
  • 48.Cimetidine and malabsorption of cobalamin. Streeter AM, Goulston KJ, Bathur FA, Hilmer RS, Crane GG, Pheils MT. Dig Dis Sci. 1982;27:13–16. doi: 10.1007/BF01308115. [DOI] [PubMed] [Google Scholar]
  • 49.Omeprazole therapy causes malabsorption of cyanocobalamin (vitamin B12) Marcuard SP, Albernaz L, Khazanie PG. Ann Intern Med. 1994;120:211–215. doi: 10.7326/0003-4819-120-3-199402010-00006. [DOI] [PubMed] [Google Scholar]
  • 50.Polymorphism of human transcobalamin II: substitution of proline and/or glutamine residues by arginine. Li N, Sood GK, Seetharam S, Seetharam B. Biochim Biophys Acta. 1994;1219:515–520. doi: 10.1016/0167-4781(94)90079-5. [DOI] [PubMed] [Google Scholar]
  • 51.The effect of different cooking methods on folate retention in various foods that are amongst the major contributors to folate intake in the UK diet. McKillop DJ, Pentieva K, Daly D, et al. Br J Nutr. 2002;88:681–688. doi: 10.1079/BJN2002733. [DOI] [PubMed] [Google Scholar]
  • 52.Bioavailability of folate from processed spinach in humans. Effect of food matrix and interaction with carotenoids. Castenmiller JJ, van de Poll CJ, West CE, Brouwer IA, Thomas CM, van Dusseldorp M. Ann Nutr Metab. 2000;44:163–169. doi: 10.1159/000012840. [DOI] [PubMed] [Google Scholar]
  • 53.Total folate activity in Brussels sprouts: the effects of storage, processing, cooking and ascorbic acid content. Malin JD. Int J Food Sci Technol. 2007;12:623–632. [Google Scholar]
  • 54.Increased urinary excretion and prolonged turnover time of folic acid during ethanol ingestion. Russell RM, Rosenberg IH, Wilson PD, et al. Am J Clin Nutr. 1983;38:64–70. doi: 10.1093/ajcn/38.1.64. [DOI] [PubMed] [Google Scholar]
  • 55.Folate deficiency disturbs hepatic methionine metabolism and promotes liver injury in the ethanol-fed micropig. Halsted CH, Villanueva JA, Devlin AM, et al. Proc Natl Acad Sci U S A. 2002;99:10072–10077. doi: 10.1073/pnas.112336399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Thermolabile variant of 5,10-methylenetetrahydrofolate reductase associated with low red-cell folates: implications for folate intake recommendations. Molloy AM, Daly S, Mills JL, et al. Lancet. 1997;349:1591–1593. doi: 10.1016/S0140-6736(96)12049-3. [DOI] [PubMed] [Google Scholar]
  • 57.Metabolic syndrome. Samson SL, Garber AJ. Endocrinol Metab Clin North Am. 2014;43:1–23. doi: 10.1016/j.ecl.2013.09.009. [DOI] [PubMed] [Google Scholar]
  • 58.Mechanisms linking obesity to insulin resistance and type 2 diabetes. Kahn SE, Hull RL, Utzschneider KM. Nature. 2006;444:840–846. doi: 10.1038/nature05482. [DOI] [PubMed] [Google Scholar]
  • 59.The metabolic syndrome--from insulin resistance to obesity and diabetes. Gallagher EJ, Leroith D, Karnieli E. Med Clin North Am. 2011;95:855–873. doi: 10.1016/j.mcna.2011.06.001. [DOI] [PubMed] [Google Scholar]
  • 60.Obesity and insulin resistance. Kahn BB, Flier JS. J Clin Invest. 2000;106:473–481. doi: 10.1172/JCI10842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Prevalence of obesity and trends in body mass index among US children and adolescents, 1999-2010. Ogden CL, Carroll MD, Kit BK, Flegal KM. JAMA. 2012;307:483–490. doi: 10.1001/jama.2012.40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013. Ng M, Fleming T, Robinson M, et al. Lancet. 2014;384:766–781. doi: 10.1016/S0140-6736(14)60460-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.World Health Organization. Obesity and overweight. [ Jul; 2021 ];World Health Organization. (2015. https://www.who.int/en/news-room/fact-sheets/detail/obesity-and-overweight 2015
  • 64.World Health Organization. Global report on diabetes. [ Jul; 2021 ];Organization WH. https://www.who.int/publications/i/item/9789241565257 2016
  • 65.The effect of probiotic supplementation on glycemic control and lipid profile in patients with type 2 diabetes: a randomized placebo controlled trial. Razmpoosh E, Javadi A, Ejtahed HS, Mirmiran P, Javadi M, Yousefinejad A. Diabetes Metab Syndr. 2019;13:175–182. doi: 10.1016/j.dsx.2018.08.008. [DOI] [PubMed] [Google Scholar]
  • 66.Complications and challenges associated with polycystic ovary syndrome: current perspectives. Palomba S, Santagni S, Falbo A, La Sala GB. Int J Womens Health. 2015;7:745–763. doi: 10.2147/IJWH.S70314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Prevalence of insulin resistance in the polycystic ovary syndrome using the homeostasis model assessment. DeUgarte CM, Bartolucci AA, Azziz R. Fertil Steril. 2005;83:1454–1460. doi: 10.1016/j.fertnstert.2004.11.070. [DOI] [PubMed] [Google Scholar]
  • 68.NCEP-defined metabolic syndrome, diabetes, and prevalence of coronary heart disease among NHANES III participants age 50 years and older. Alexander CM, Landsman PB, Teutsch SM, Haffner SM. Diabetes. 2003;52:1210–1214. doi: 10.2337/diabetes.52.5.1210. [DOI] [PubMed] [Google Scholar]
  • 69.Pathophysiology of the metabolic syndrome. McCracken E, Monaghan M, Sreenivasan S. Clin Dermatol. 2018;36:14–20. doi: 10.1016/j.clindermatol.2017.09.004. [DOI] [PubMed] [Google Scholar]
  • 70.The assessment of insulin resistance in man. Wallace TM, Matthews DR. Diabet Med. 2002;19:527–534. doi: 10.1046/j.1464-5491.2002.00745.x. [DOI] [PubMed] [Google Scholar]
  • 71.The metabolic syndrome. Eckel RH, Alberti KG, Grundy SM, Zimmet PZ. Lancet. 2010;375:181–183. doi: 10.1016/S0140-6736(09)61794-3. [DOI] [PubMed] [Google Scholar]
  • 72.The concept of metabolic syndrome: contribution of visceral fat accumulation and its molecular mechanism. Matsuzawa Y, Funahashi T, Nakamura T. J Atheroscler Thromb. 2011;18:629–639. doi: 10.5551/jat.7922. [DOI] [PubMed] [Google Scholar]
  • 73.Metabolic syndrome: from epidemiology to systems biology. Lusis AJ, Attie AD, Reue K. Nat Rev Genet. 2008;9:819–830. doi: 10.1038/nrg2468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Gene-diet interactions on body weight changes. Loos RJ, Rankinen T. J Am Diet Assoc. 2005;105:0–34. doi: 10.1016/j.jada.2005.02.015. [DOI] [PubMed] [Google Scholar]
  • 75.Glucose metabolism and insulin therapy. Langouche L, Van den Berghe G. Crit Care Clin. 2006;22:119-29, vii. doi: 10.1016/j.ccc.2005.09.005. [DOI] [PubMed] [Google Scholar]
  • 76.Insulin resistance: a marker of surgical stress. Thorell A, Nygren J, Ljungqvist O. Curr Opin Clin Nutr Metab Care. 1999;2:69–78. doi: 10.1097/00075197-199901000-00012. [DOI] [PubMed] [Google Scholar]
  • 77.Adverse endothelial function and the insulin resistance syndrome. Tooke JE, Hannemann MM. J Intern Med. 2000;247:425–431. doi: 10.1046/j.1365-2796.2000.00671.x. [DOI] [PubMed] [Google Scholar]
  • 78.Cellular mechanisms of insulin resistance. Shulman GI. J Clin Invest. 2000;106:171–176. doi: 10.1172/JCI10583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Surgery-induced insulin resistance in human patients: relation to glucose transport and utilization. Thorell A, Nygren J, Hirshman MF, et al. Am J Physiol. 1999;276:0–61. doi: 10.1152/ajpendo.1999.276.4.E754. [DOI] [PubMed] [Google Scholar]
  • 80.Lipid and lipoprotein dysregulation in insulin resistant states. Avramoglu RK, Basciano H, Adeli K. Clin Chim Acta. 2006;368:1–19. doi: 10.1016/j.cca.2005.12.026. [DOI] [PubMed] [Google Scholar]
  • 81.Follow-up report on the diagnosis of diabetes mellitus. Genuth S, Alberti KG, Bennett P, et al. Diabetes Care. 2003;26:3160–3167. doi: 10.2337/diacare.26.11.3160. [DOI] [PubMed] [Google Scholar]
  • 82.Barrett KE, Barman SM, Brooks HL, Yuan JX. Vol. 192. Singapore: McGraw-Hill Education; 2019. Ganong's review of medical physiology; p. 201. [Google Scholar]
  • 83.Abdominal obesity: role in the pathophysiology of metabolic disease and cardiovascular risk. Bergman RN, Kim SP, Hsu IR, et al. Am J Med. 2007;120:0–32. doi: 10.1016/j.amjmed.2006.11.012. [DOI] [PubMed] [Google Scholar]
  • 84.Dyslipidemia in obesity: mechanisms and potential targets. Klop B, Elte JW, Cabezas MC. Nutrients. 2013;5:1218–1240. doi: 10.3390/nu5041218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lipoprotein subfractions in metabolic syndrome and obesity: clinical significance and therapeutic approaches. Nikolic D, Katsiki N, Montalto G, Isenovic ER, Mikhailidis DP, Rizzo M. Nutrients. 2013;5:928–948. doi: 10.3390/nu5030928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Metabolic syndrome: what are the risks for humans? Gupta A, Gupta V. https://www.biosciencetrends.com/article/343. Biosci Trends. 2010;4:204–212. [PubMed] [Google Scholar]
  • 87.Relationships among insulin resistance, type 2 diabetes, essential hypertension, and cardiovascular disease: similarities and differences. Reaven GM. J Clin Hypertens (Greenwich) 2011;13:238–243. doi: 10.1111/j.1751-7176.2011.00439.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Metabolic syndrome, diabetes and atherosclerosis: influence of gene-environment interaction. Andreassi MG. Mutat Res. 2009;667:35–43. doi: 10.1016/j.mrfmmm.2008.10.018. [DOI] [PubMed] [Google Scholar]
  • 89.Metabolic endotoxaemia: is it more than just a gut feeling? Piya MK, Harte AL, McTernan PG. Curr Opin Lipidol. 2013;24:78–85. doi: 10.1097/MOL.0b013e32835b4431. [DOI] [PubMed] [Google Scholar]
  • 90.Viscoelastic and ultrastructural characteristics of whole blood and plasma in Alzheimer-type dementia, and the possible role of bacterial lipopolysaccharides (LPS) Bester J, Soma P, Kell DB, Pretorius E. Oncotarget. 2015;6:35284–35303. doi: 10.18632/oncotarget.6074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Inflammation and insulin resistance. Shoelson SE, Lee J, Goldfine AB. J Clin Invest. 2006;116:1793–1801. doi: 10.1172/JCI29069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Human oral, gut, and plaque microbiota in patients with atherosclerosis. Koren O, Spor A, Felin J, et al. Proc Natl Acad Sci U S A. 2011;108 Suppl 1:4592–4598. doi: 10.1073/pnas.1011383107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Differences in visceral fat and fat bacterial colonization between ulcerative colitis and Crohn's disease. An in vivo and in vitro study. Zulian A, Cancello R, Ruocco C, et al. PLoS One. 2013;8:0. doi: 10.1371/journal.pone.0078495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Metabolic endotoxemia initiates obesity and insulin resistance. Cani PD, Amar J, Iglesias MA, et al. Diabetes. 2007;56:1761–1772. doi: 10.2337/db06-1491. [DOI] [PubMed] [Google Scholar]
  • 95.The microbiome and obesity: is obesity linked to our gut flora? Tsai F, Coyle WJ. Curr Gastroenterol Rep. 2009;11:307–313. doi: 10.1007/s11894-009-0045-z. [DOI] [PubMed] [Google Scholar]
  • 96.Oligofructose promotes satiety in rats fed a high-fat diet: involvement of glucagon-like peptide-1. Cani PD, Neyrinck AM, Maton N, Delzenne NM. Obes Res. 2005;13:1000–1007. doi: 10.1038/oby.2005.117. [DOI] [PubMed] [Google Scholar]
  • 97.Gut microbiota and its possible relationship with obesity. DiBaise JK, Zhang H, Crowell MD, Krajmalnik-Brown R, Decker GA, Rittmann BE. Mayo Clin Proc. 2008;83:460–469. doi: 10.4065/83.4.460. [DOI] [PubMed] [Google Scholar]
  • 98.Treating the metabolic syndrome. Bianchi C, Penno G, Romero F, Del Prato S, Miccoli R. Expert Rev Cardiovasc Ther. 2007;5:491–506. doi: 10.1586/14779072.5.3.491. [DOI] [PubMed] [Google Scholar]
  • 99.Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Grundy SM, Cleeman JI, Daniels SR, et al. Circulation. 2005;112:2735–2752. doi: 10.1161/CIRCULATIONAHA.105.169404. [DOI] [PubMed] [Google Scholar]
  • 100.Lifestyle behaviors associated with lower risk of having the metabolic syndrome. Zhu S, St-Onge MP, Heshka S, Heymsfield SB. Metabolism. 2004;53:1503–1511. doi: 10.1016/j.metabol.2004.04.017. [DOI] [PubMed] [Google Scholar]
  • 101.Development of fatness, fitness, and lifestyle from adolescence to the age of 36 years: determinants of the metabolic syndrome in young adults: the Amsterdam growth and health longitudinal study. Ferreira I, Twisk JW, van Mechelen W, Kemper HC, Stehouwer CD. Arch Intern Med. 2005;165:42–48. doi: 10.1001/archinte.165.1.42. [DOI] [PubMed] [Google Scholar]
  • 102.The effect of metformin and intensive lifestyle intervention on the metabolic syndrome: the Diabetes Prevention Program randomized trial. Orchard TJ, Temprosa M, Goldberg R, Haffner S, Ratner R, Marcovina S, Fowler S. Ann Intern Med. 2005;142:611–619. doi: 10.7326/0003-4819-142-8-200504190-00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Association between adherence to the Mediterranean diet and physical fitness with body composition parameters in 1717 European adolescents: the AdolesHealth Study. Galan-Lopez P, Sanchez-Oliver AJ, Pihu M, Gísladóttír T, Domínguez R, Ries F. Nutrients. 2019;12:77. doi: 10.3390/nu12010077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Definition of the Mediterranean diet; a literature review. Davis C, Bryan J, Hodgson J, Murphy K. Nutrients. 2015;7:9139–9153. doi: 10.3390/nu7115459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Associations of dietary fiber with glucose metabolism in nondiabetic relatives of subjects with type 2 diabetes: the Botnia Dietary Study. Ylönen K, Saloranta C, Kronberg-Kippilä C, Groop L, Aro A, Virtanen SM. Diabetes Care. 2003;26:1979–1985. doi: 10.2337/diacare.26.7.1979. [DOI] [PubMed] [Google Scholar]
  • 106.Nutrition recommendations for the treatment and prevention of type 2 diabetes and the metabolic syndrome: an evidenced-based review. Mann JI. Nutr Rev. 2006;64:422–427. doi: 10.1111/j.1753-4887.2006.tb00227.x. [DOI] [PubMed] [Google Scholar]
  • 107.Glycemic index, glycemic load, and dietary fiber intake and incidence of type 2 diabetes in younger and middle-aged women. Schulze MB, Liu S, Rimm EB, Manson JE, Willett WC, Hu FB. Am J Clin Nutr. 2004;80:348–356. doi: 10.1093/ajcn/80.2.348. [DOI] [PubMed] [Google Scholar]
  • 108.Targeting the metabolic syndrome with exercise: evidence from the HERITAGE Family Study. Katzmarzyk PT, Leon AS, Wilmore JH, Skinner JS, Rao DC, Rankinen T, Bouchard C. Med Sci Sports Exerc. 2003;35:1703–1709. doi: 10.1249/01.MSS.0000089337.73244.9B. [DOI] [PubMed] [Google Scholar]
  • 109.Combined diet and exercise intervention reverses the metabolic syndrome in middle-aged males: results from the Oslo Diet and Exercise Study. Anderssen SA, Carroll S, Urdal P, Holme I. Scand J Med Sci Sports. 2007;17:687–695. doi: 10.1111/j.1600-0838.2006.00631.x. [DOI] [PubMed] [Google Scholar]
  • 110.Effects of diet and exercise in preventing NIDDM in people with impaired glucose tolerance. The Da Qing IGT and Diabetes Study. Pan XR, Li GW, Hu YH, et al. Diabetes Care. 1997;20:537–544. doi: 10.2337/diacare.20.4.537. [DOI] [PubMed] [Google Scholar]
  • 111.Increasing prevalence of the metabolic syndrome among U.S. Adults. Ford ES, Giles WH, Mokdad AH. Diabetes Care. 2004;27:2444–2449. doi: 10.2337/diacare.27.10.2444. [DOI] [PubMed] [Google Scholar]
  • 112.Predictors of poor glycemic control in adult with type 2 diabetes in South-Eastern Nigeria. Anioke IC, Ezedigboh AN, Dozie-Nwakile OC, Chukwu IJ, Kalu PN. Afr Health Sci. 2019;19:2819–2828. doi: 10.4314/ahs.v19i4.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Stimulation of the intracellular portion of the human insulin receptor by the antidiabetic drug metformin. Stith BJ, Woronoff K, Wiernsperger N. Biochem Pharmacol. 1998;55:533–536. doi: 10.1016/s0006-2952(97)00540-6. [DOI] [PubMed] [Google Scholar]
  • 114.Thiazolidinediones in the treatment of insulin resistance and type II diabetes. Saltiel AR, Olefsky JM. Diabetes. 1996;45:1661–1669. doi: 10.2337/diab.45.12.1661. [DOI] [PubMed] [Google Scholar]
  • 115.Mechanism by which metformin reduces glucose production in type 2 diabetes. Hundal RS, Krssak M, Dufour S, et al. Diabetes. 2000;49:2063–2069. doi: 10.2337/diabetes.49.12.2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Metformin rapidly increases insulin receptor activation in human liver and signals preferentially through insulin-receptor substrate-2. Gunton JE, Delhanty PJ, Takahashi S, Baxter RC. J Clin Endocrinol Metab. 2003;88:1323–1332. doi: 10.1210/jc.2002-021394. [DOI] [PubMed] [Google Scholar]
  • 117.Glucose and lipid metabolism in non-insulin-dependent diabetes. Effect of metformin. Riccio A, Del Prato S, Vigili de Kreutzenberg S, Tiengo A. https://pubmed.ncbi.nlm.nih.gov/1936473/ Diabete Metab. 1991;17:180–184. [PubMed] [Google Scholar]
  • 118.New oral therapies for type 2 diabetes mellitus: the glitazones or insulin sensitizers. Mudaliar S, Henry RR. Annu Rev Med. 2001;52:239–257. doi: 10.1146/annurev.med.52.1.239. [DOI] [PubMed] [Google Scholar]
  • 119.Distinct but complementary contributions of PPAR isotypes to energy homeostasis. Dubois V, Eeckhoute J, Lefebvre P, Staels B. J Clin Invest. 2017;127:1202–1214. doi: 10.1172/JCI88894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.PPAR agonists and metabolic syndrome: an established role? Botta M, Audano M, Sahebkar A, Sirtori CR, Mitro N, Ruscica M. Int J Mol Sci. 2018;19:0. doi: 10.3390/ijms19041197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Effect of fibrates on glycemic parameters: a systematic review and meta-analysis of randomized placebo-controlled trials. Simental-Mendía LE, Simental-Mendía M, Sánchez-García A, Banach M, Atkin SL, Gotto AM Jr, Sahebkar A. Pharmacol Res. 2018;132:232–241. doi: 10.1016/j.phrs.2017.12.030. [DOI] [PubMed] [Google Scholar]
  • 122.Metformin: intrinsic vasculoprotective properties. Wiernsperger NF. Diabetes Technol Ther. 2000;2:259–272. doi: 10.1089/15209150050025230. [DOI] [PubMed] [Google Scholar]
  • 123.The effects of rosiglitazone, a peroxisome proliferator-activated receptor-gamma agonist, on markers of endothelial cell activation, C-reactive protein, and fibrinogen levels in non-diabetic coronary artery disease patients. Sidhu JS, Cowan D, Kaski JC. J Am Coll Cardiol. 2003;42:1757–1763. doi: 10.1016/j.jacc.2003.04.001. [DOI] [PubMed] [Google Scholar]
  • 124.Association of fenofibrate therapy with long-term cardiovascular risk in statin-treated patients with type 2 diabetes. Elam MB, Ginsberg HN, Lovato LC, et al. JAMA Cardiol. 2017;2:370–380. doi: 10.1001/jamacardio.2016.4828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Treating the metabolic syndrome using angiotensin receptor antagonists that selectively modulate peroxisome proliferator-activated receptor-gamma. Pershadsingh HA. Int J Biochem Cell Biol. 2006;38:766–781. doi: 10.1016/j.biocel.2005.08.006. [DOI] [PubMed] [Google Scholar]
  • 126.Identification, evaluation, and treatment of overweight and obese adults. Blackwell J. J Am Acad Nurse Pract. 2002;14:196–198. doi: 10.1111/j.1745-7599.2002.tb00113.x. [DOI] [PubMed] [Google Scholar]
  • 127.Long-term pharmacotherapy for overweight and obesity: a systematic review and meta-analysis of randomized controlled trials. Padwal R, Li SK, Lau DC. Int J Obes Relat Metab Disord. 2003;27:1437–1446. doi: 10.1038/sj.ijo.0802475. [DOI] [PubMed] [Google Scholar]
  • 128.A randomized trial of sibutramine in the management of obese type 2 diabetic patients treated with metformin. McNulty SJ, Ur E, Williams G. Diabetes Care. 2003;26:125–131. doi: 10.2337/diacare.26.1.125. [DOI] [PubMed] [Google Scholar]
  • 129.The ORLIstat and CArdiovascular risk profile in patients with metabolic syndrome and type 2 DIAbetes (ORLICARDIA) Study. Didangelos TP, Thanopoulou AK, Bousboulas SH, et al. Curr Med Res Opin. 2004;20:1393–1401. doi: 10.1185/030079904125004466. [DOI] [PubMed] [Google Scholar]
  • 130.Obesity and pharmacologic therapy. Thearle M, Aronne LJ. Endocrinol Metab Clin North Am. 2003;32:1005–1024. doi: 10.1016/s0889-8529(03)00066-5. [DOI] [PubMed] [Google Scholar]
  • 131.The effects of folate supplementation on diabetes biomarkers among patients with metabolic diseases: a systematic review and meta-analysis of randomized controlled trials. Akbari M, Tabrizi R, Lankarani KB, et al. Horm Metab Res. 2018;50:93–105. doi: 10.1055/s-0043-125148. [DOI] [PubMed] [Google Scholar]
  • 132.Folic acid supplementation improves glycemic control for diabetes prevention and management: a systematic review and dose-response meta-analysis of randomized controlled trials. Asbaghi O, Ashtary-Larky D, Bagheri R, et al. Nutrients. 2021;13:2355. doi: 10.3390/nu13072355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Maternal folate status and obesity/insulin resistance in the offspring: a systematic review. Xie RH, Liu YJ, Retnakaran R, et al. Int J Obes (Lond) 2016;40:1–9. doi: 10.1038/ijo.2015.189. [DOI] [PubMed] [Google Scholar]
  • 134.Hyperglycemia and metformin use are associated with B vitamin deficiency and cognitive dysfunction in older adults. Porter KM, Ward M, Hughes CF, et al. J Clin Endocrinol Metab. 2019;104:4837–4847. doi: 10.1210/jc.2018-01791. [DOI] [PubMed] [Google Scholar]
  • 135.Folate-chitosan nanoparticles triggered insulin cellular uptake and improved in vivo hypoglycemic activity. El Leithy ES, Abdel-Bar HM, Ali RA. Int J Pharm. 2019;571:118708. doi: 10.1016/j.ijpharm.2019.118708. [DOI] [PubMed] [Google Scholar]
  • 136.Insulin resistance and endothelial function are improved after folate and vitamin B12 therapy in patients with metabolic syndrome: relationship between homocysteine levels and hyperinsulinemia. Setola E, Monti LD, Galluccio E, et al. Eur J Endocrinol. 2004;151:483–489. doi: 10.1530/eje.0.1510483. [DOI] [PubMed] [Google Scholar]
  • 137.Dietary intake of folate and risk of stroke in US men and women: NHANES I Epidemiologic Follow-up Study. National Health and Nutrition Examination Survey. Bazzano LA, He J, Ogden LG, Loria C, Vupputuri S, Myers L, Whelton PK. Stroke. 2002;33:1183–1188. doi: 10.1161/01.str.0000014607.90464.88. [DOI] [PubMed] [Google Scholar]
  • 138.Metabolic vitamin B12 deficiency: a missed opportunity to prevent dementia and stroke. Spence JD. Nutr Res. 2016;36:109–116. doi: 10.1016/j.nutres.2015.10.003. [DOI] [PubMed] [Google Scholar]
  • 139.Interaction of serum vitamin B12 and folate with MTHFR genotypes on risk of ischemic stroke. Qin X, Spence JD, Li J, et al. Neurology. 2020;94:0–36. doi: 10.1212/WNL.0000000000008932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Efficacy of folic acid supplementation in stroke prevention: a meta-analysis. Wang X, Qin X, Demirtas H, et al. Lancet. 2007;369:1876–1882. doi: 10.1016/S0140-6736(07)60854-X. [DOI] [PubMed] [Google Scholar]
  • 141.Homocysteine metabolism and risk of myocardial infarction: relation with vitamins B6, B12, and folate. Verhoef P, Stampfer MJ, Buring JE, et al. Am J Epidemiol. 1996;143:845–859. doi: 10.1093/oxfordjournals.aje.a008828. [DOI] [PubMed] [Google Scholar]
  • 142.A prospective study of plasma homocyst(e)ine and risk of myocardial infarction in US physicians. Stampfer MJ, Malinow MR, Willett WC, et al. JAMA. 1992;268:877–881. [PubMed] [Google Scholar]
  • 143.Vitamins B6, B12, and folate: association with plasma total homocysteine and risk of coronary atherosclerosis. Siri PW, Verhoef P, Kok FJ. J Am Coll Nutr. 1998;17:435–441. doi: 10.1080/07315724.1998.10718790. [DOI] [PubMed] [Google Scholar]
  • 144.Folate, vitamin B12, homocysteine and polymorphisms in folate metabolizing genes in children with congenital heart disease and their mothers. Elizabeth KE, Praveen SL, Preethi NR, Jissa VT, Pillai MR. Eur J Clin Nutr. 2017;71:1437–1441. doi: 10.1038/ejcn.2017.135. [DOI] [PubMed] [Google Scholar]
  • 145.Association between vitamin deficiency and metabolic disorders related to obesity. Thomas-Valdés S, Tostes MD, Anunciação PC, da Silva BP, Sant'Ana HM. Crit Rev Food Sci Nutr. 2017;57:3332–3343. doi: 10.1080/10408398.2015.1117413. [DOI] [PubMed] [Google Scholar]
  • 146.Maternal dietary folate and/or vitamin B12 restrictions alter body composition (adiposity) and lipid metabolism in Wistar rat offspring. Kumar KA, Lalitha A, Pavithra D, et al. J Nutr Biochem. 2013;24:25–31. doi: 10.1016/j.jnutbio.2012.01.004. [DOI] [PubMed] [Google Scholar]
  • 147.Folate and vitamin B12 levels in early pregnancy and maternal obesity. O'Malley EG, Reynolds CM, Cawley S, Woodside JV, Molloy AM, Turner MJ. Eur J Obstet Gynecol Reprod Biol. 2018;231:80–84. doi: 10.1016/j.ejogrb.2018.10.001. [DOI] [PubMed] [Google Scholar]
  • 148.Folate and vitamin B12 in morbid obesity: the influence of folate on anti-atherogenic lipid profile. Daviddi G, Ricci MA, De Vuono S, Gentili A, Boni M, Lupattelli G. Int J Vitam Nutr Res. 2020;90:295–301. doi: 10.1024/0300-9831/a000538. [DOI] [PubMed] [Google Scholar]
  • 149.Associations among serum folate, waist-to-hip ratio, lipid profile, and eating habits with homocysteine in an elderly Thai population. Paratthakonkun C, Kaewprasert S, Arthan D, et al. Int J Vitam Nutr Res. 2019;89:246–254. doi: 10.1024/0300-9831/a000402. [DOI] [PubMed] [Google Scholar]
  • 150.Effect of a diet containing folate and hazelnut oil capsule on the methylation level of the ADRB3 gene, lipid profile and oxidative stress in overweight or obese women. Lima RP, do Nascimento RA, Luna RC, et al. Clin Epigenetics. 2017;9:110. doi: 10.1186/s13148-017-0407-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Reduction of polypharmacy in the elderly: a systematic review of the role of the pharmacist. Rollason V, Vogt N. Drugs Aging. 2003;20:817–832. doi: 10.2165/00002512-200320110-00003. [DOI] [PubMed] [Google Scholar]
  • 152.Pemafibrate: first global approval. Blair HA. Drugs. 2017;77:1805–1810. doi: 10.1007/s40265-017-0818-x. [DOI] [PubMed] [Google Scholar]
  • 153.The novel selective PPARα modulator (SPPARMα) pemafibrate improves dyslipidemia, enhances reverse cholesterol transport and decreases inflammation and atherosclerosis. Hennuyer N, Duplan I, Paquet C, et al. Atherosclerosis. 2016;249:200–208. doi: 10.1016/j.atherosclerosis.2016.03.003. [DOI] [PubMed] [Google Scholar]
  • 154.A novel selective PPARα modulator (SPPARMα), K-877 (pemafibrate), attenuates postprandial hypertriglyceridemia in mice. Sairyo M, Kobayashi T, Masuda D, et al. J Atheroscler Thromb. 2018;25:142–152. doi: 10.5551/jat.39693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Effects of pemafibrate, a novel selective PPARα modulator, on lipid and glucose metabolism in patients with type 2 diabetes and hypertriglyceridemia: a randomized, double-blind, placebo-controlled, phase 3 trial. Araki E, Yamashita S, Arai H, et al. Diabetes Care. 2018;41:538–546. doi: 10.2337/dc17-1589. [DOI] [PubMed] [Google Scholar]
  • 156.Rationale and design of the Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) study. Pradhan AD, Paynter NP, Everett BM, et al. Am Heart J. 2018;206:80–93. doi: 10.1016/j.ahj.2018.09.011. [DOI] [PubMed] [Google Scholar]
  • 157.FXR: a promising target for the metabolic syndrome? Cariou B, Staels B. Trends Pharmacol Sci. 2007;28:236–243. doi: 10.1016/j.tips.2007.03.002. [DOI] [PubMed] [Google Scholar]
  • 158.Discovery of tropifexor (LJN452), a highly potent non-bile acid FXR agonist for the treatment of cholestatic liver diseases and nonalcoholic steatohepatitis (NASH) Tully DC, Rucker PV, Chianelli D, et al. J Med Chem. 2017;60:9960–9973. doi: 10.1021/acs.jmedchem.7b00907. [DOI] [PubMed] [Google Scholar]
  • 159.Nidufexor (LMB763), a novel FXR modulator for the treatment of nonalcoholic steatohepatitis. Chianelli D, Rucker PV, Roland J, et al. J Med Chem. 2020;63:3868–3880. doi: 10.1021/acs.jmedchem.9b01621. [DOI] [PubMed] [Google Scholar]
  • 160.Asprosin, a fasting-induced glucogenic protein hormone. Romere C, Duerrschmid C, Bournat J, et al. Cell. 2016;165:566–579. doi: 10.1016/j.cell.2016.02.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Asprosin is a centrally acting orexigenic hormone. Duerrschmid C, He Y, Wang C, et al. Nat Med. 2017;23:1444–1453. doi: 10.1038/nm.4432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Asprosin-neutralizing antibodies as a treatment for metabolic syndrome. Mishra I, Duerrschmid C, Ku Z, et al. Elife. 2021;10:0. doi: 10.7554/eLife.63784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.A patent update on cannabinoid receptor 1 antagonists (2015-2018) Amato G, Khan NS, Maitra R. Expert Opin Ther Pat. 2019;29:261–269. doi: 10.1080/13543776.2019.1597851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Peripherally restricted CB1 receptor blockers. Chorvat RJ. Bioorg Med Chem Lett. 2013;23:4751–4760. doi: 10.1016/j.bmcl.2013.06.066. [DOI] [PubMed] [Google Scholar]
  • 165.Efficacy and safety of the weight-loss drug rimonabant: a meta-analysis of randomised trials. Christensen R, Kristensen PK, Bartels EM, Bliddal H, Astrup A. Lancet. 2007;370:1706–1713. doi: 10.1016/S0140-6736(07)61721-8. [DOI] [PubMed] [Google Scholar]
  • 166.Peripheral selectivity of the novel cannabinoid receptor antagonist TM38837 in healthy subjects. Klumpers LE, Fridberg M, de Kam ML, et al. Br J Clin Pharmacol. 2013;76:846–857. doi: 10.1111/bcp.12141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Peripheral CB1 receptor neutral antagonist, AM6545, ameliorates hypometabolic obesity and improves adipokine secretion in monosodium glutamate induced obese mice. Ma H, Zhang G, Mou C, Fu X, Chen Y. Front Pharmacol. 2018;9:156. doi: 10.3389/fphar.2018.00156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.JD-5006 and JD-5037: peripherally restricted (PR) cannabinoid-1 receptor blockers related to SLV-319 (Ibipinabant) as metabolic disorder therapeutics devoid of CNS liabilities. Chorvat RJ, Berbaum J, Seriacki K, McElroy JF. Bioorg Med Chem Lett. 2012;22:6173–6180. doi: 10.1016/j.bmcl.2012.08.004. [DOI] [PubMed] [Google Scholar]
  • 169.Impact of fecal microbiota transplantation on obesity and metabolic syndrome-a systematic review. Zhang Z, Mocanu V, Cai C, et al. Nutrients. 2019;11:2291. doi: 10.3390/nu11102291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Gut microbiota from twins discordant for obesity modulate metabolism in mice. Ridaura VK, Faith JJ, Rey FE, et al. Science. 2013;341:1241214. doi: 10.1126/science.1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Improvement of insulin sensitivity after lean donor feces in metabolic syndrome is driven by baseline intestinal microbiota composition. Kootte RS, Levin E, Salojärvi J, et al. Cell Metab. 2017;26:611–619. doi: 10.1016/j.cmet.2017.09.008. [DOI] [PubMed] [Google Scholar]
  • 172.Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Vrieze A, Van Nood E, Holleman F, et al. Gastroenterology. 2012;143:913–916. doi: 10.1053/j.gastro.2012.06.031. [DOI] [PubMed] [Google Scholar]
  • 173.Fecal microbiota transplantation in metabolic syndrome: history, present and future. de Groot PF, Frissen MN, de Clercq NC, Nieuwdorp M. Gut Microbes. 2017;8:253–267. doi: 10.1080/19490976.2017.1293224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Gut dysbiosis is linked to hypertension. Yang T, Santisteban MM, Rodriguez V, et al. Hypertension. 2015;65:1331–1340. doi: 10.1161/HYPERTENSIONAHA.115.05315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Adenosine triphosphate citrate lyase: emerging target in the treatment of dyslipidemia. Lemus HN, Mendivil CO. J Clin Lipidol. 2015;9:384–389. doi: 10.1016/j.jacl.2015.01.002. [DOI] [PubMed] [Google Scholar]
  • 176.AMP-activated protein kinase and ATP-citrate lyase are two distinct molecular targets for ETC-1002, a novel small molecule regulator of lipid and carbohydrate metabolism. Pinkosky SL, Filippov S, Srivastava RA, et al. J Lipid Res. 2013;54:134–151. doi: 10.1194/jlr.M030528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Safety and efficacy of bempedoic acid to reduce LDL cholesterol. Ray KK, Bays HE, Catapano AL, et al. N Engl J Med. 2019;380:1022–1032. doi: 10.1056/NEJMoa1803917. [DOI] [PubMed] [Google Scholar]
  • 178.Bempedoic acid: first approval. Markham A. Drugs. 2020;80:747–753. doi: 10.1007/s40265-020-01308-w. [DOI] [PubMed] [Google Scholar]
  • 179.Leptin and the regulation of body weight in mammals. Friedman JM, Halaas JL. Nature. 1998;395:763–770. doi: 10.1038/27376. [DOI] [PubMed] [Google Scholar]
  • 180.Obesity is associated with a decreased leptin transport across the blood-brain barrier in rats. Burguera B, Couce ME, Curran GL, Jensen MD, Lloyd RV, Cleary MP, Poduslo JF. Diabetes. 2000;49:1219–1223. doi: 10.2337/diabetes.49.7.1219. [DOI] [PubMed] [Google Scholar]
  • 181.Central leptin gene therapy blocks high-fat diet-induced weight gain, hyperleptinemia, and hyperinsulinemia: increase in serum ghrelin levels. Dube MG, Beretta E, Dhillon H, Ueno N, Kalra PS, Kalra SP. Diabetes. 2002;51:1729–1736. doi: 10.2337/diabetes.51.6.1729. [DOI] [PubMed] [Google Scholar]

Articles from Cureus are provided here courtesy of Cureus Inc.

RESOURCES