Next Article in Journal
Proteomics Analyses of Small Extracellular Vesicles of Aqueous Humor: Identification and Validation of GAS6 and SPP1 as Glaucoma Markers
Previous Article in Journal
Systemic Pharmacotherapeutic Treatment of the ACTA1-MCM/FLExDUX4 Preclinical Mouse Model of FSHD
Previous Article in Special Issue
Deflamin Attenuated Lung Tissue Damage in an Ozone-Induced COPD Murine Model by Regulating MMP-9 Catalytic Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Fenugreek in the Management of Type 2 Diabetes

Diabetes Interest Group, The Centre for Health and Life Sciences Research, London Metropolitan University, London N7 8DB, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(13), 6987; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25136987
Submission received: 30 April 2024 / Revised: 11 June 2024 / Accepted: 17 June 2024 / Published: 26 June 2024
(This article belongs to the Special Issue Natural Compounds in Health and Disease)

Abstract

:
The number of people diagnosed with type 2 diabetes is on the increase worldwide. Of growing concern, the prevalence of type 2 diabetes in children and youths is increasing rapidly and mirrors the increasing burden of childhood obesity. There are many risk factors associated with the condition; some are due to lifestyle, but many are beyond our control, such as genetics. There is an urgent need to develop better therapeutics for the prevention and management of this complex condition since current medications often cause unwanted side effects, and poorly managed diabetes can result in the onset of related comorbidities. Naturally derived compounds have gained momentum for preventing and managing several complex conditions, including type 2 diabetes. Here, we provide an update on the benefits and limitations of fenugreek and its components as a therapeutic for type 2 diabetes, including its bioavailability and interaction with the microbiome.

1. Introduction

Type 2 Diabetes Mellitus (T2DM) is a complex metabolic disorder resulting from an interplay of both modifiable and non-modifiable risk factors. Modifiable risk factors include diet, exercise and socioeconomic disparities [1,2,3,4], whilst non-modifiable risk factors include age, gender, ethnicity and genetics [5,6,7,8,9,10]. Other risk factors include having gestational diabetes [11], long-term use of certain medications, depression, alcohol intake and smoking [12]. T2DM is a complex condition, and understanding the condition at the molecular level is crucial for developing and testing new and improved therapeutics for better disease management and prevention.
Treating diabetes and diabetes-related complications has a significant financial burden on healthcare systems worldwide, and these increases have paralleled rises in obesity [1]. Increases in youth-onset T2DM have risen disproportionately in ethnic minority groups [13], highlighting the need for urgent healthcare interventions.
Diabetes mellitus is attributed to dysfunctions in both the secretion and physiological activity of insulin marked by elevated plasma glucose levels, termed hyperglycemia [14]. Pathophysiologically, T2DM is characterized by insulin resistance and impaired insulin signaling pathways, leading to an inadequate cellular response to insulin. Insulin resistance reduces glucose uptake and pancreatic beta cell dysfunction, leading to persistent hyperglycemia [15]. This prolonged hyperglycemic state contributes to diverse complications, encompassing microvascular issues such as neuropathy and nephropathy, along with macrovascular complications, predominantly affecting the cardiovascular system [16]. Prediabetes is defined as those with an impaired glucose tolerance, with higher than normal blood glucose levels, who are at a high risk of developing diabetes. In contrast, type 1 diabetes occurs due to autoimmune-mediated destruction of pancreatic beta cells, culminating in reduced insulin synthesis [14].

Current Methods for the Management of Type 2 Diabetes

It has been shown that T2DM is preventable and reversible through healthy lifestyle changes [17]. For those individuals diagnosed with T2DM and unable to reverse the condition through lifestyle changes, managing their blood glucose levels through prescribed medication is required. Commonly prescribed medications include metformin, sulfonylureas, thiazolidinediones, dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, sodium-glucose cotransporter-2 (SGLT2) inhibitors and insulin therapy. These medications primarily aim to enhance insulin sensitivity, decrease hepatic glucose production, stimulate insulin secretion, or facilitate glucose uptake by peripheral tissues [18]. However, many of the currently prescribed medications lose efficacy over time, are expensive, have side effects and can interfere with other medications [18]. Reported side effects include gastrointestinal issues [19], higher cardiovascular disease risk and weight gain [20], acute pancreatitis [21], nausea, vomiting and diarrhea [22], increased incidence of urinary tract infections and diabetic ketoacidosis [23]. Inadequate management of diabetes can result in poorly controlled blood sugar levels, predisposing individuals to a myriad of complications and a higher risk of incurable conditions such as Alzheimer’s disease [16]. There is, therefore, a need for better preventative treatments and therapeutics to manage glucose levels better.
There have been several studies demonstrating that fenugreek (Trigonella foenum-graecum) can improve glycemic control in individuals with diabetes by reducing blood glucose levels and improving insulin sensitivity in obese and overweight patients with T2DM [24,25] and non-insulin-dependent diabetics [26,27]. Fenugreek supplementation has also shown benefits in lowering cholesterol levels and reducing the risk of cardiovascular diseases [28], thus reducing the risk of T2DM onset. Hence, here we discuss the benefits and limitations of fenugreek for T2DM management.

2. The Role of Natural Compounds in Diabetes Management

2.1. Promising Naturally Derived Compounds for Diabetes Management

Natural compounds are bioactive substances derived from natural sources such as plants, animals, minerals and microorganisms. Natural products have been trusted as traditional medicines across the world for centuries, with over 80% of the world’s population relying on them for treating various conditions [29]. Their exploitation for development into therapeutics has gained momentum due to their natural abundance, efficacy and being inexpensive and accessible. Potential limitations of their widespread use by the pharmaceutical industry include challenges in identifying the bioactive compound and extracting and producing it in large quantities [30]. In the context of treating T2DM, numerous natural compounds have shown promise [31,32,33,34,35,36,37,38,39,40,41,42,43]. These compounds can vary widely in their chemical structure and biological activity and exert their effects through various mechanisms, such as improving insulin sensitivity, enhancing glucose uptake, reducing intestinal glucose absorption, and modulating glucose metabolism, showing promising results in vitro and in vivo [31]. Metformin, the most commonly prescribed medication for those with T2DM, was developed from a naturally derived compound, galegine, extracted from Galega officinalis (or ‘Goat’s rue) [44].
In T2DM research, the majority of natural products tested are polyphenols [31,32,39]. Polyphenolic compounds are 500–4000 Daltons, have several phenolic hydroxyl groups on aromatic rings in their structure and are water-soluble. Numerous studies show that the flavonoid resveratrol (3,5,4’-trihydroxystilbene, found in grapes and blueberries) can improve glycemic control [45,46]; however, these studies include small sample sizes and short intervention periods (8 weeks) limiting an assessment of their long-term use in large, diverse populations. In addition, many of these studies have been conducted using resveratrol in combination with other natural compounds, thus limiting an assessment of the efficacy of resveratrol alone [47]. The antidiabetic effects of resveratrol are thought to be attributed to its ability to reduce body weight, increase glucose metabolism, increase insulin sensitivity and inhibit phosphodiesterase thus leading to an increase in insulin secretion [47].
Bitter melon (Momordica charantia) has also shown promising pre-clinical and clinical antidiabetic effects due to its high polyphenolic content, reducing fasting blood glucose levels and improving glucose tolerance [48]. Polyphenols in black tea have been shown to target enzymes such as α-glucosidase, involved in carbohydrate metabolism and blood glucose regulation [49]. In silico studies, several more polyphenolic compounds have been identified as potential inhibitors of alpha-glucosidase and alpha-amylase, suggesting a promising direction for future drug discovery [50]. Other promising compounds for T2DM management include alkaloids such as berberine [33], saponins such as soyasaponins and ginsenosides [34], fatty acids such as Omega-3 [35], polysaccharides [36], terpenoids [37,38,39,40,41,42] and organosulfur compounds [43].

2.2. Fenugreek as a Promising Natural Treatment for the Management of Type 2 Diabetes

Fenugreek (Trigonella foenum-graecum) is a member of the Fabaceae family, native to the Mediterranean region, Western Asia and Southern Europe. Fenugreek seeds and leaves are composed of macronutrients, vitamins, minerals and are rich in bioactive compounds like alkaloids (trigonelline), flavonoids (quercetin, luteolin), saponins and steroidal sapogenins. Additionally, fenugreek seeds are palatable and nutritionally dense, containing carbohydrates, proteins, lipids and volatile oils [51], making them useful for digestive health and as a source of plant-based nutrition. Their composition is listed in Table 1.
Fenugreek seeds possess several pharmacological properties such as anti-inflammatory, antioxidant, hypoglycemic, hypocholesterolemic and antiulcerogenic effects [52]. In traditional medicine, fenugreek has been used for centuries to treat a wide range of conditions, including digestive disorders and skin inflammation [53]. Steroidal saponins such as diosgenin and gitogenin have been shown to be the principal active constituents responsible for its medicinal properties [54]. Fenugreek has also been shown to inhibit the growth of cancer cells, induce apoptosis [55], and influence gut microbiota homeostasis in chickens [56], highlighting its additional health benefits.
There have been numerous studies showing that fenugreek has antidiabetic effects in vitro [57,58,59,60,61,62,63] in animal studies [52,59,64,65,66,67,68,69,70,71,72,73,74,75] and in trials with obese and diabetic patients [76,77,78,79,80,81]. The hypoglycemic effects of fenugreek seeds in animal studies have been summarized previously [64] and shown to be largely attributed to their high soluble dietary fiber content (galactomannan), trigonelline, diosgenin, 4-hydroxyisoleucine and flavone C-glycosides. Improved glucose tolerance was observed in streptozotocin-induced diabetic rats administered fenugreek (soluble dietary fiber fraction at 0.5 g per kilogram body weight) for 28 days, in addition to increased liver glycogen content and reduced serum glucose levels [72]. Similar results were observed in another study with diabetic rats given 0.44, 0.87 and 1.74 g per kilogram per day of fenugreek extract for 6 weeks [73]. They showed decreased glycated hemoglobin levels and decreased fasting blood glucose in comparison to untreated diabetic rats. Similarly, diabetic rats orally administered fenugreek extract at 0.1, 0.25 and 0.5 g per kilogram body weight for 14 days had reported decreased serum glucose and increased insulin levels [74]. Fenugreek seed extracts administered at lower concentrations of 50, 100 and 200 milligrams per kilogram to diabetic rats for 6 weeks also reported reduced blood glucose levels compared to non-treated diabetic controls [75].
Since fenugreek has shown promising antidiabetic effects in vitro and in numerous animal studies, several clinical trials have recently been undertaken to assess its antidiabetic effects in humans. Neelakatan et al. analyzed the results of 10 separate clinical trials assessing the efficacy of fenugreek seeds on T2DM [77]. Reduced glucose levels were observed in patients after daily doses of 1–100 g of fenugreek seed (median 25 g) over 10–84 days (median 30 days). The results revealed that fenugreek notably improved glycemic control and enhanced insulin sensitivity in individuals with T2DM; however, the studies were inconclusive due to the wide range of doses, from 1 to 100 g of seed or seed extract and participants having differing diabetes status. A meta-analysis of randomly controlled clinical trials conducted with T2DM patients showed that across the studies, fenugreek seeds reduced HbA1c levels (glycated hemoglobin where glucose adheres to red blood cells); however, these studies are difficult to compare due to heterogeneity in study design and differing doses administered [78].
Another study involving 18 T2DM participants showed that fenugreek seeds reduced their fasting blood sugar and triglycerides when given 10 g of powdered fenugreek seeds for 8 weeks [79]. While promising, 18 individuals is a small test group and, therefore, would need to be repeated with a larger cohort.
More recently, Kim et al. [80] performed a systematic review and meta-analysis to assess the effectiveness of fenugreek in individuals with T2DM and prediabetes. In total, they analyzed 10 studies involving 706 participants and found that fenugreek significantly reduced fasting blood glucose levels, HbA1c and 2-h plasma glucose level (2-hPG) in the intervention groups compared to the untreated control groups. In addition, fenugreek appeared to improve lipid levels (triglyceride, total cholesterol and high-density lipoprotein), which are known risk factors for T2DM. Fenugreek did not significantly influence body mass index, reflecting similar observations in C57BL/6J mice [59]. This meta-analysis highlighted the limitations and heterogeneity in study design that influenced the results collected. There was only one study focused on prediabetes [81]; hence, the data are biased towards the efficacy of fenugreek in T2DM, not prediabetes. Non-standardized methods include differences in fenugreek extraction, preparation, study duration and dosage and number of participants. Further studies are needed using standardized methods with a larger number of participants over longer time periods to assess the effectiveness of fenugreek in T2DM. Also, further studies assessing the effects of fenugreek in prediabetes are urgently needed as a potential early intervention therapeutic to prevent the onset of T2DM.

2.3. Toxicity and Adverse Effects

Since fenugreek is consumed through diet, it is considered safe to ingest; however, this is dependent on the doses consumed, and different studies report conflicting results. Animal experiments have been used to assess the toxicity of fenugreek seed extract [65]. Doses below 1000 milligrams per kilogram weight showed no toxicity in Wistar rats or their offspring after 90 days of repeated doses. Another study with leaf glycosidic fenugreek extracts administered to mice (at 0.2–1 g per kilogram for up to 7 days) showed minimal adverse effects [82]. However, due to its estrogenic activity, fenugreek seeds are not recommended to be consumed during pregnancy. Studies with rabbits showed that fenugreek affected fetal development when pregnant rabbits were fed 30% fenugreek seeds for three months [83].
In humans, the results are conflicting [80]. No reports of severe adverse reactions such as renal or hepatic toxicity were reported across ten independent studies, although mild gastrointestinal side effects were reported in some individuals, albeit short-lived. Another study used the ‘ToxRTool’ to assess the reliability of published toxicology studies for fenugreek seeds [84]. From the initial 436 results, only 17 studies were deemed ‘reliable without restrictions’ and showed that standardized extraction of fenugreek was non-toxic, with only a few reports of gastrointestinal side effects. A study analyzing sera from 29 different patients who have IgE specific to peanuts and certain legumes revealed that sensitization to fenugreek is a consequence of cross-reactivity in patients with peanut allergy [85]; hence, it should not be given to prediabetics or T2DM patients with peanut or legume allergies. Further studies are clearly needed. These should include larger cohorts of participants from diverse backgrounds monitored over a long period of time, focusing on dose responses. However, initial studies suggest fenugreek may be a safe and effective therapeutic for those with T2DM.
Table 1. The composition of fenugreek shows the percentage present in leaves and seeds. Adapted from [86].
Table 1. The composition of fenugreek shows the percentage present in leaves and seeds. Adapted from [86].
Constituent TypeConstituentFenugreek Seeds (Approximate %)Fenugreek Leaves (Approximate %)
MacronutrientsProteins20–30%4–6%
Dietary Fibre25–30%3–5%
Carbohydrates40–60%5–10%
Fats5–10%<1%
VitaminsB Vitamins (e.g., B6, B12)0.3–0.6%0.1–0.2%
Vitamin KTrace amounts0.2–0.4%
Vitamin ATrace amounts0.02–0.05%
Vitamin C0.1–0.3%0.3–0.7%
Folate (Folic Acid)0.02–0.05%0.01–0.03%
Vitamin ETrace amountsTrace amounts
MineralsIron1–2%2–4%
Calcium0.2–0.5%1–3%
Magnesium0.1–0.3%0.2–0.5%
Phosphorus0.3–0.6%0.2–0.4%
Potassium0.4–0.8%0.5–1%
Sodium<0.1%<0.1%
Zinc0.01–0.03%0.01–0.03%
Bioactive CompoundsSaponins2–6%<1%
Alkaloids<1%Trace amounts
4-hydroxyisoleucine1–2%Trace amounts
Flavonoids0.5–1%0.2–0.5%
Phenolic Acids0.1–0.3%0.1–0.2%
CoumarinsTrace amountsTrace amounts
Lecithin0.1–0.3%Trace amounts
Other ComponentsWater Content5–10%75–85%
Essential OilsTrace to 1%Trace amounts
Mucilage1–3%<1%
Choline0.02–0.05%Trace amounts

2.4. Bioactive Components of Fenugreek

Several studies have attempted to extract and purify individual components of fenugreek to test them in relation to diabetes management. Diosgenin, a saponin (steroidal glycoalkaloid), constitutes 3–6% of the seeds and 0.6–0.8% of the leaves. Streptozotocin-induced diabetic rats fed a single dose (5 or 10 milligrams per kilogram per body weight) of diosgenin for 30 days showed increased regeneration of pancreatic beta cells and insulin granules [66].
Trigonelline, categorized as an alkaloid, comprises 0.1–0.3% and 0.6–1.2% of fenugreek seeds and leaves, respectively. Trigonelline can reduce blood glucose concentrations in rats induced with diabetes through a high-fat and low-streptozotocin diet [67]. Its ability to increase insulin sensitivity is due to its ability to improve the insulin signaling pathway by increasing insulin receptor autophosphorylation (IR-PH) and the translocation of effector molecules pT308-Akt, and glucose transporter 4 to the cell surface, thus increasing glucose uptake [68].
Galactomannan (a polysaccharide composed of galactose side groups and a mannose backbone) is a carbohydrate prevalent in fenugreek seeds, constituting 40–50% of their composition and 4–6% in leaves (Table 1). Purified galactomannan reduced fasting hyperglycemia and improved serum and hepatic lipid profiles in control and streptozotocin-induced diabetic adult male Wistar rats [69]. Clinical trials involving 24 patients with T2DM who were given 8 or 16 g of an oral galactomannan derivative showed it to be safe under the conditions tested [87], thus showing promise. However, its long-term effects need to be tested in further studies.

4-Hydroxy Isoleucine

One of the most studied fenugreek compounds is 4-Hydroxyisoleucine, (4-HIL), classified as an amino acid that constitutes 1–3% of fenugreek seeds and 0.5–1.5% of the leaves. It has been shown to have antidiabetic effects in sucrose-lipid-fed diabetic rat models, regulating pancreatic insulin secretion, stimulating insulin secretion in vivo and improving glucose tolerance [70]. It has been proposed that 4-HIL works by negatively regulating TNF-alpha production, resulting in insulin sensitivity and increasing GLUT4 (glucose transporter type 4) and p-IRS-1 (insulin receptor substrate 1) in the insulin-signaling pathway [88]. It is thought to work directly on pancreatic cells and may enhance glucose uptake in peripheral tissues such as skeletal muscle and adipose tissue, contributing to improved glycemic control.
Oxidative stress and inflammation play crucial roles in the pathogenesis of T2DM and its complications. 4-HIL exerts its antioxidant and anti-inflammatory effects by decreasing oxidative stress and the inflammatory response via activation of nuclear factor erythroid 2-related factor 2 (Nrf2 gene, a free radical scavenger and antioxidant enzyme stimulator) and the transforming growth factor β1 signaling pathway [71].

2.5. Fenugreek and the Microbiome

The role of the microbiome in maintaining health is well established, and the tools for examining the microbiome and the consequences of the metabolic activity of the microbiome, are readily available. Here, we review studies that consider the interaction of fenugreek formulations on the microbiome.
There have been a limited number of studies to date, and these have mostly utilized mouse or rat models. In one series of studies, young male C57BL/6J mice were fed control or high-fat diets with or without 2% fenugreek seed powder over 14–16 weeks. The first study indicated improved glycemic control and improved lipid profile in mice fed with a high-fat diet, including fenugreek [59]. A follow-up study with the same model analyzed changes in the microbiome and found that the inclusion of fenugreek could reverse changes in the microbiome induced by a high-fat diet [89]. Fenugreek restored the distribution of microbial taxa to one associated with a good metabolic profile. The inclusion of fenugreek in the diet did not change the variety of taxa, but only the relative proportions of species. Further studies with the same model [90] confirmed an increase in the proportion of the Verrumicrobia phylum in mice fed fenugreek seed powder due to an increase in the genus Akkermansia, which includes the mucin-degrading species Akkermansia muciniphilia. Decreased proportions of A.muciniphilia in the microbiome have been associated with adverse metabolic conditions, including obesity and diabetes, in humans and animals [91].
Other studies have examined the effect of fenugreek seed components on the microbiome. In a study on peripheral neuropathy caused as a side effect of oxaliplatin chemotherapy [92], diosgenin could relieve the symptoms of peripheral neuropathy. Diosgenin restored the variety of the microbiome in mice treated with oxaliplatin. Importantly, fecal material transferred from diosgenin-treated mice could also relieve the symptoms of peripheral neuropathy, suggesting a direct therapeutic effect of diosgenin mediated through the microbiome. In a model of osteoporosis using ovariectomized rats, treatment with diosgenin could improve bone loss [93]. The osteoporotic rats suffered a loss of microbiome composition, which was restored after treatment with diosgenin.
Trigonelline was found to inhibit the conversion of choline to the pro-atherosclerotic metabolite trimethylamine-N-oxide (TMAO) in isolated cultures of anaerobic microbes isolated from human feces [94]. Finally, the polysaccharide galactomannan, the major component of fenugreek seeds, was found to improve glycemic control in 7–8-week-old C57 BL/6 J mice, which was associated with a healthier microbiome [95].
Taken together, these studies suggest a strong, protective effect of fenugreek seed formulations on the microbiome, either as a complete formulation or as individual components.

2.6. Mechanisms Involved in the Antidiabetic Effects of Fenugreek

The mechanisms involved in the antidiabetic properties of fenugreek have been investigated [57,58,60,61,62,63,64,75,96,97,98], but more research is needed to identify all of the involved biomolecules and pathways involved. Fenugreek seeds are thought to exhibit their antidiabetic effects in a number of ways, including stimulation of insulin release from beta cells and improved insulin signaling [61], glucagon suppression and delaying gastric emptying [98]. An in vitro study using human hepatoma cells (HepG2) showed that under normal and hyperglycemic conditions, fenugreek seed extract significantly enhanced glucose uptake in comparison to metformin and insulin [58]. Additionally, fenugreek seeds have demonstrated an ability to improve insulin sensitivity in adipocytes [57], which is crucial for regulating blood sugar levels and mitigating the risk of excessive insulin secretion. Fenugreek seeds have been shown to inhibit intestinal sodium-dependent glucose uptake in vitro using rabbit intestinal brush border membrane vesicles [63]. Fenugreek seeds have also been shown to regulate glucagon-like peptide-1 (GLP-1) activity, which is important for secreting insulin after food intake [97].
Fenugreek extracts have also been shown to control blood glucose levels by targeting α-amylase and α-glucosidase enzymes, which are required for metabolizing complex carbohydrates [89]. Fenugreek exerts hypolipidemic effects [28,62,99], hence targeting molecules such as triglycerides, which are thought to contribute to its antidiabetic effects since alterations in lipoprotein levels have been associated with an increased risk of diabetes [100]. Membrane fluidity, which is impaired in those with diabetes, has been shown to improve with fenugreek, thus improving glucose transport and vesicular trafficking [60]. In addition, the antioxidant properties of fenugreek may also be instrumental in reducing oxidative stress damage that has been linked to diabetes [71,101]. Several studies have investigated or proposed the antidiabetic mechanisms of fenugreek through in vitro and in vivo animal studies [52,57,58,60,61,62,64,65,66,67,68,69,70,75,101] that we have summarized in Figure 1. Understanding the contribution of the individual bioactive ingredients found in fenugreek is needed to learn how they exert their antidiabetic effects.

3. Discussion

Fenugreek (Trigonella foenum-graecum) has received considerable scientific interest for its potential therapeutic applications, particularly in the management of diabetes mellitus. This attention is due to its rich composition of bioactive compounds, including 4-hydroxyisoleucine (4-HIL) and trigonelline, among others [86]. Research has shown that fenugreek exerts hypoglycemic effects through various mechanisms (Figure 1), such as enhancing insulin sensitivity and stimulating insulin secretion. Additionally, fenugreek exhibits antioxidant properties, which can mitigate oxidative stress and inflammation commonly associated with diabetes and its complications [102]. Fenugreek has demonstrated potential in managing lipid profiles by reducing total cholesterol, LDL cholesterol and triglycerides, addressing another critical aspect of diabetes management [27,28]. Research into the antidiabetic mechanisms of individual fenugreek components has revealed their biological targets and some of the pathways involved. Diosgenin may regenerate pancreatic beta cells and insulin granules [66]. Trigonelline increases insulin sensitivity and improves insulin signaling pathways [67,68]. Galactomannan can improve lipid profiles [69] and glycemic control [95], and 4-HIL stimulates insulin secretion [69,88] and has anti-oxidative and anti-inflammatory effects [71]. Many conventional diabetes medications, such as Metformin, primarily focus on a single target, such as insulin sensitivity or hepatic glucose production [103]. However, since fenugreek and its bioactive components have multiple biological targets and impact various biochemical pathways (such as enhancing insulin secretion and sensitivity, modulating glucose absorption and enzyme activity, providing anti-inflammatory and antioxidant effects, and improving lipid metabolism), it may offer enhanced efficacy in managing T2DM.
Additionally, in comparison to prescribed synthetic drugs, being of natural origin and having been consumed through diet for many generations with noted health benefits [51], natural products are likely to have a lower risk of unwanted side effects. Animal studies using controlled administration of doses (up to 1000 milligrams of fenugreek seed per kilogram of weight) have been shown to be non-toxic to mammals [65]. Bioactive components of fenugreek have also shown promise in regard to safety. Doses of up to 16 g of a galactomannan derivative given to human volunteers showed no side effects whilst being able to reduce glucose levels after eating [87]. However, this is an area that needs further investigation since it is known that many plants produce toxic compounds as protection mechanisms [104], so purification methods need to be optimized to ensure any toxic components are removed. The precise mode of action of fenugreek and its active components need to be fully elucidated prior to administering it as a drug to understand any potential adverse effects it may have. Several promising clinical trials with fenugreek seeds and isolated bioactive compounds with T2DM patients have been observed [26,27,76,77,78,79,80,81,86,87,105,106]; however, heterogeneity in the participant’s diabetes status, low participant numbers and the wide ranges of doses used and the duration of treatment given warrant further studies.

3.1. Bioavailability and the Microbiome

One limitation of using natural products as therapeutics is their bioavailability/bioaccessibility. This refers to the amount available for absorption in the intestines, remaining soluble and stable before cell absorption and thus being able to exert its bioactive effects. Poor intestinal absorption is one reason why several promising bioactive natural products, such as curcumin, have not yet developed into therapeutics [107]. Unlike curcumin, galactomannan isolated from fenugreek is very bioaccessible [108]. When used in combination with other bioactive compounds, galactomannan has been used as a drug-delivery molecule to enhance the bioavailability of other bioactive compounds such as curcumin as observed in experiments with rat models [109] and studies involving human participants [110].
Understanding the bioavailability of fenugreek and its interaction with the microbiome is crucial in exploiting fenugreek as a preventative and/or diabetes treatment. Limited studies have shown that fenugreek seeds have a protective effect on the microbiome [92]. The gut microbiome can influence the efficacy of drugs (and vice versa) in an individual [111] and thus influence its bioavailability and toxicity. More experiments looking at the interaction of fenugreek and its bioactive components are urgently needed to make effective therapeutics in the future.

3.2. Limitations and Future Directions

The heterogeneity and variability of clinical trial results are related to several factors, namely the diabetic state of the participant (healthy versus diabetic patients), other conditions they may have, their age, the duration of treatment, the administered dosages, and the fenugreek preparation methods used. Moving forward, having standardized methods for the preparation and isolation of bioactive compounds and being able to produce these in large quantities will facilitate testing and further clinical trials. 4-HIL has shown promise as a potential diabetes treatment; however, current methods for producing it in Escherichia coli are not economically efficient [112], and hence better, more reproducible methods are needed. Further studies also need to ascertain whether fenugreek and its active compounds interact with other prescribed medications since many people with T2DM take medications for other conditions in addition to their T2DM. One study reported that fenugreek interfered with the action of warfarin in a patient who was prescribed it for atrial fibrillation [113]. Investigating dose response is important for ascertaining safe doses for prescription. Combining fenugreek with conventional therapies such as metformin, sulfonylureas, or insulin may lead to improved glycemic control and reduced medication dosages, thereby minimizing adverse effects and enhancing patient adherence to treatment regimens. Trials combining prescribed medications such as metformin and natural products such as fenugreek have shown decreased fasting blood glucose, HbA1c levels and cholesterol when used in combination and compared to the placebo group [105].
More comprehensive clinical trials are needed to evaluate the long-term effects of fenugreek supplementation on glycemic control, insulin sensitivity and diabetes-related complications in diverse populations, including individuals with type 1 and type 2 diabetes, prediabetes and gestational diabetes, an under-researched area. More under-represented groups should be included in clinical trials since T2DM disproportionately affects non-white ethnic populations, and hence, more participants from non-white backgrounds should be included in future trials [114].
More research is also needed into the preventative effects of fenugreek. In addition to being used by those with T2DM, being able to prevent T2DM will prevent damage to the body and the onset of diabetes complications. A study by Gaddam et al. [115] showed that participants with prediabetes who were given 10 g a day over a 3-year period were four times less likely to develop T2DM compared to controls. This long-term study highlights the potential of fenugreek as a preventive measure for T2DM. Similarly, Neelakantan et al. [77] and Kim et al. [80] conducted a meta-analysis of clinical trials and found that fenugreek consistently reduced fasting blood glucose and HbA1c levels among T2DM patients, reinforcing the beneficial effects of fenugreek in glycemic control. Another study by Hadi et al. [106] revealed substantial decreases in fasting plasma glucose levels among T2DM patients supplemented with fenugreek seed powder, further substantiating its potential to enhance glycemic control. These studies collectively underscore the importance of fenugreek in the management of T2DM, suggesting that its bioactive compounds may improve insulin secretion and sensitivity, modulate glucose absorption and enzyme activity, and provide anti-inflammatory and antioxidant effects, which all contribute to better glycemic control.
The molecular mechanisms behind the mode of action of fenugreek in T2DM prevention and management are slowly being elucidated (Figure 1). The antidiabetic effects are largely due to fenugreek’s hypoglycemic effects, reducing blood glucose levels by targeting various molecules, receptors and membranes. Fenugreek also appears to decrease oxidative stress and lipoprotein levels, which also contribute to its antidiabetic effects. More mechanistic studies are required to better understand the underlying pathways through which fenugreek and its bioactive compounds affect glucose metabolism and insulin sensitivity. Technical advances being made in molecular docking [116] and artificial intelligence [117] will no doubt help advance our knowledge and understanding of fenugreek’s potential as an antidiabetic therapy by complementing current methods.

Funding

This research received no external.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wu, Y.; He, X.; Zhou, J.; Wang, Y.; Yu, L.; Li, X.; Liu, T.; Luo, J. Impact of healthy lifestyle on the risk of type 2 diabetes mellitus in southwest China: A prospective cohort study. J. Diabetes Investig. 2022, 13, 2091–2100. [Google Scholar] [CrossRef]
  2. Yaribeygi, H.; Maleki, M.; Sathyapalan, T.; Jamialahmadi, T.; Sahebkar, A. Pathophysiology of Physical Inactivity-Dependent Insulin Resistance: A Theoretical Mechanistic Review Emphasizing Clinical Evidence. J. Diabetes Res. 2021, 2021, 7796727. [Google Scholar] [CrossRef]
  3. Zheng, D.; Dou, J.; Liu, G.; Pan, Y.; Yan, Y.; Liu, F.; Gaisano, H.Y.; Lu, J.; He, Y. Association Between Triglyceride Level and Glycemic Control Among Insulin-Treated Patients With Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2019, 104, 1211–1220. [Google Scholar] [CrossRef] [PubMed]
  4. Nagar, S.D.; Nápoles, A.M.; Jordan, I.K.; Mariño-Ramírez, L. Socioeconomic deprivation and genetic ancestry interact to modify type 2 diabetes ethnic disparities in the United Kingdom. EClinicalMedicine 2021, 37, 100960. [Google Scholar] [CrossRef]
  5. Vadadokhau, U.; Varga, I.; Káplár, M.; Emri, M.; Csősz, É. Examination of the Complex Molecular Landscape in Obesity and Type 2 Diabetes. Int. J. Mol. Sci. 2024, 25, 4781. [Google Scholar] [CrossRef]
  6. Mauvais-Jarvis, F. Sex differences in metabolic homeostasis, diabetes, and obesity. Biol. Sex Differ. 2015, 6, 14. [Google Scholar] [CrossRef] [PubMed]
  7. Meigs, J.B. The Genetic Epidemiology of Type 2 Diabetes: Opportunities for Health Translation. Curr. Diabetes Rep. 2019, 19, 62. [Google Scholar] [CrossRef]
  8. Sevcuka, A.; White, K.; Terry, C. Factors That Contribute to hIAPP Amyloidosis in Type 2 Diabetes Mellitus. Life 2022, 12, 583. [Google Scholar] [CrossRef] [PubMed]
  9. Hassan, S.; White, K.; Terry, C. Linking hIAPP misfolding and aggregation with type 2 diabetes mellitus: A structural perspective. Biosci. Rep. 2022, 42, BSR20211297. [Google Scholar] [CrossRef]
  10. Juvinao-Quintero, D.L.; Sharp, G.C.; Sanderson, E.C.M.; Relton, C.L.; Elliott, H.R. Investigating causality in the association between DNA methylation and type 2 diabetes using bidirectional two-sample Mendelian randomisation. Diabetologia 2023, 66, 1247–1259. [Google Scholar] [CrossRef]
  11. Vounzoulaki, E.; Khunti, K.; Abner, S.C.; Tan, B.K.; Davies, M.J.; Gillies, C.L. Progression to type 2 diabetes in women with a known history of gestational diabetes: Systematic review and meta-analysis. BMJ Clin. Res. Ed. 2020, 369, m1361. [Google Scholar] [CrossRef]
  12. Zhang, X.; Xie, J.; You, X.; Gong, H. Risk factors and drug discovery for cognitive impairment in type 2 diabetes mellitus using artificial intelligence interpretation and graph neural networks. Front. Endocrinol. 2023, 14, 1213711. [Google Scholar] [CrossRef] [PubMed]
  13. Pinhas-Hamiel, O.; Shah, A.S.; van Raalte, D.H.; Pavkov, M.E.; Nelson, R.G. Youth-onset type 2 diabetes mellitus: An urgent challenge. Nat. Rev. Nephrol. 2023, 19, 168–184. [Google Scholar] [CrossRef]
  14. World Health Organization; International Diabetes Federation. Definition and Diagnosis of Diabetes Mellitus and Intermediate Hyperglycaemia: Report of a WHO/IDF Consultation; WHO: Geneva, Switzerland, 2006. Available online: https://www.who.int/diabetes/publications/Definition%20and%20diagnosis%20of%20diabetes_new.pdf (accessed on 10 February 2024).
  15. Wilkin, T.J. The accelerator hypothesis: A unifying explanation for type-1 and type-2 diabetes. Nestle Nutr. Workshop Ser. Clin. Perform. Programme 2006, 11, 139–153. [Google Scholar] [CrossRef] [PubMed]
  16. Tomic, D.; Shaw, J.E.; Magliano, D.J. The burden and risks of emerging complications of diabetes mellitus. Nat. Rev. Endocrinol. 2022, 18, 525–539. [Google Scholar] [CrossRef] [PubMed]
  17. Lemieux, I. Reversing Type 2 Diabetes: The Time for Lifestyle Medicine Has Come! Nutrients 2020, 12, 1974. [Google Scholar] [CrossRef] [PubMed]
  18. Galindo, R.J.; Trujillo, J.M.; Low Wang, C.C.; McCoy, R.G. Advances in the management of type 2 diabetes in adults. BMJ Med. 2023, 2, e000372. [Google Scholar] [CrossRef] [PubMed]
  19. Viollet, B.; Guigas, B.; Sanz Garcia, N.; Leclerc, J.; Foretz, M.; Andreelli, F. Cellular and molecular mechanisms of metformin: An overview. Clin. Sci. 2012, 122, 253–270. [Google Scholar] [CrossRef]
  20. Nissen, S.E.; Wolski, K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N. Engl. J. Med. 2007, 356, 2457–2471. [Google Scholar] [CrossRef]
  21. Storgaard, H.; Cold, F.; Gluud, L.L.; Vilsbøll, T.; Knop, F.K. Glucagon-like peptide-1 receptor agonists and risk of acute pancreatitis in patients with type 2 diabetes. Diabetes Obes. Metab. 2017, 19, 906–908. [Google Scholar] [CrossRef]
  22. Marso, S.P.; Daniels, G.H.; Brown-Frandsen, K.; Kristensen, P.; Mann, J.F.; Nauck, M.A.; Nissen, S.E.; Pocock, S.; Poulter, N.R.; Ravn, L.S.; et al. LEADER Steering Committee, & LEADER Trial Investigators Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 311–322. [Google Scholar] [CrossRef] [PubMed]
  23. Giorgino, F.; Vora, J.; Fenici, P.; Solini, A. Renoprotection with SGLT2 inhibitors in type 2 diabetes over a spectrum of cardiovascular and renal risk. Cardiovasc. Diabetol. 2020, 19, 196. [Google Scholar] [CrossRef] [PubMed]
  24. Mathern, J.R.; Raatz, S.K.; Thomas, W.; Slavin, J.L. Effect of fenugreek fiber on satiety, blood glucose and insulin response and energy intake in obese subjects. Phytother. Res. PTR 2009, 23, 1543–1548. [Google Scholar] [CrossRef] [PubMed]
  25. Robert, S.D.; Ismail, A.A.; Wan Rosli, W.I. Trigonella foenum-graecum seeds lowers postprandial blood glucose in overweight and obese individuals. J. Nutr. Metab. 2014, 2014, 964873. [Google Scholar] [CrossRef] [PubMed]
  26. Madar, Z.; Abel, R.; Samish, S.; Arad, J. Glucose-lowering effect of fenugreek in non-insulin dependent diabetics. Eur. J. Clin. Nutr. 1988, 42, 51–54. [Google Scholar] [PubMed]
  27. Geberemeskel, G.A.; Debebe, Y.G.; Nguse, N.A. Antidiabetic Effect of Fenugreek Seed Powder Solution (Trigonella foenum-graecum L.) on Hyperlipidemia in Diabetic Patients. J. Diabetes Res. 2019, 2019, 8507453. [Google Scholar] [CrossRef] [PubMed]
  28. Shaikh, R.F.; Ali, M.T.; Mohsin, A.A.; Hiware, S.D.; Ahmad, A.; Daimi, S.R.H.; Moizuddin, K.; Shaikh, S.A.; Siddiqui, F.B. A Comparative Study on Clinical Evaluation of the Hypolipidemic Effects of Allium sativum, Trigonella foenum-graecum, Commiphora mukul, Picrorhiza kurroa, and Piper nigrum: A Pilot Study. Cureus 2022, 14, e26597. [Google Scholar] [CrossRef] [PubMed]
  29. Wangchuk, P. Therapeutic Applications of Natural Products in Herbal Medicines, Biodiscovery Programs, and Biomedicine. J. Biol. Act. Prod. Nat. 2018, 8, 1–20. [Google Scholar] [CrossRef]
  30. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; the International Natural Product Sciences Taskforce; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug. Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  31. Terry, C. Insights from Nature: A Review of Natural Compounds That Target Protein Misfolding in Vivo. Curr. Res. Biotechnol. 2020, 2, 131–144. [Google Scholar] [CrossRef]
  32. Franko, A.; Rodriguez Camargo, D.C.; Böddrich, A.; Garg, D.; Camargo, A.R.; Rathkolb, B.; Janik, D.; Aichler, M.; Feuchtinger, A.; Neff, F.; et al. Epigallocatechin gallate (EGCG) reduces the intensity of pancreatic amyloid fibrils in human islet amyloid polypeptide (hIAPP) transgenic mice. Sci. Rep. 2018, 8, 1116. [Google Scholar] [CrossRef] [PubMed]
  33. Amssayef, A.; Eddouks, M. Alkaloids as Promising Agents for the Management of Insulin Resistance: A Review. Curr. Pharm. Des. 2023, 29, 3123–3136. [Google Scholar] [CrossRef] [PubMed]
  34. Zhou, Y.; Xu, B. New insights into anti-diabetes effects and molecular mechanisms of dietary saponins. Crit. Rev. Food Sci. Nutr. 2023, 63, 12372–12397. [Google Scholar] [CrossRef] [PubMed]
  35. Shah, M.; Adams-Huet, B.; Brinkley, L.; Grundy, S.M.; Garg, A. Lipid, glycemic, and insulin responses to meals rich in saturated, cis-monounsaturated, and polyunsaturated (n-3 and n-6) fatty acids in subjects with type 2 diabetes. Diabetes Care 2007, 30, 2993–2998. [Google Scholar] [CrossRef] [PubMed]
  36. Mitchelson, K.A.J.; Tran, T.T.T.; Dillon, E.T.; Vlckova, K.; Harrison, S.M.; Ntemiri, A.; Cunningham, K.; Gibson, I.; Finucane, F.M.; O’Connor, E.M.; et al. Yeast β-Glucan Improves Insulin Sensitivity and Hepatic Lipid Metabolism in Mice Humanized with Obese Type 2 Diabetic Gut Microbiota. Mol. Nutr. Food Res. 2022, 66, e2100819. [Google Scholar] [CrossRef] [PubMed]
  37. Pivari, F.; Mingione, A.; Brasacchio, C.; Soldati, L. Curcumin and Type 2 Diabetes Mellitus: Prevention and Treatment. Nutrients 2019, 11, 1837. [Google Scholar] [CrossRef] [PubMed]
  38. Zhong, Y.; Xiao, Y.; Gao, J.; Zheng, Z.; Zhang, Z.; Yao, L.; Li, D. Curcumin improves insulin sensitivity in high-fat diet-fed mice through gut microbiota. Nutr. Metab. 2022, 19, 76. [Google Scholar] [CrossRef]
  39. MacDonald-Ramos, K.; Michán, L.; Martínez-Ibarra, A.; Cerbón, M. Silymarin is an ally against insulin resistance: A review. Ann. Hepatol. 2021, 23, 100255. [Google Scholar] [CrossRef] [PubMed]
  40. Bacanlı, M.; Anlar, H.G.; Aydın, S.; Çal, T.; Arı, N.; Ündeğer Bucurgat, Ü.; Başaran, A.A.; Başaran, N. D-limonene ameliorates diabetes and its complications in streptozotocin-induced diabetic rats. Food Chem. Toxicol. 2017, 110, 434–442. [Google Scholar] [CrossRef]
  41. El Omari, N.; Mrabti, H.N.; Benali, T.; Ullah, R.; Alotaibi, A.; Abdullah, A.D.I.; Goh, K.W.; Bouyahya, A. Expediting Multiple Biological Properties of Limonene and α-Pinene: Main Bioactive Compounds of Pistacia lentiscus L., Essential Oils. Front. Biosci. Landmark Ed. 2023, 28, 229. [Google Scholar] [CrossRef]
  42. Liu, P.; Zhang, Z.; Cai, Y.; Yang, Y.; Yuan, J.; Chen, Q. Inhibition of the pyroptosis-associated inflammasome pathway: The important potential mechanism of ginsenosides in ameliorating diabetes and its complications. Eur. J. Med. Chem. 2023, 253, 115336. [Google Scholar] [CrossRef] [PubMed]
  43. Ruhee, R.T.; Roberts, L.A.; Ma, S.; Suzuki, K. Organosulfur Compounds: A Review of Their Anti-inflammatory Effects in Human Health. Front. Nutr. 2020, 7, 64. [Google Scholar] [CrossRef]
  44. Bailey, C.J. Metformin: Historical overview. Diabetologia 2017, 60, 1566–1576. [Google Scholar] [CrossRef]
  45. Bhatt, J.K.; Thomas, S.; Nanjan, M.J. Resveratrol supplementation improves glycemic control in type 2 diabetes mellitus. Nutr. Res. 2012, 32, 537–541. [Google Scholar] [CrossRef]
  46. García-Martínez, B.I.; Ruiz-Ramos, M.; Pedraza-Chaverri, J.; Santiago-Osorio, E.; Mendoza-Núñez, V.M. Hypoglycemic Effect of Resveratrol: A Systematic Review and Meta-Analysis. Antioxidants 2021, 10, 69. [Google Scholar] [CrossRef] [PubMed]
  47. Mongioì, L.M.; La Vignera, S.; Cannarella, R.; Cimino, L.; Compagnone, M.; Condorelli, R.A.; Calogero, A.E. The Role of Resveratrol Administration in Human Obesity. Int. J. Mol. Sci. 2021, 22, 4362. [Google Scholar] [CrossRef] [PubMed]
  48. Çiçek, S.S. Momordica charantia L.-Diabetes-Related Bioactivities, Quality Control, and Safety Considerations. Front. Pharmacol. 2022, 13, 904643. [Google Scholar] [CrossRef] [PubMed]
  49. Striegel, L.; Kang, B.; Pilkenton, S.J.; Rychlik, M.; Apostolidis, E. Effect of Black Tea and Black Tea Pomace Polyphenols on α-Glucosidase and α-Amylase Inhibition, Relevant to Type 2 Diabetes Prevention. Front. Nutr. 2015, 2, 3. [Google Scholar] [CrossRef]
  50. Riyaphan, J.; Pham, D.C.; Leong, M.K.; Weng, C.F. In Silico Approaches to Identify Polyphenol Compounds as α-Glucosidase and α-Amylase Inhibitors against Type-II Diabetes. Biomolecules 2021, 11, 1877. [Google Scholar] [CrossRef]
  51. Alu’datt, M.H.; Rababah, T.; Al-Ali, S.; Tranchant, C.C.; Gammoh, S.; Alrosan, M.; Kubow, S.; Tan, T.C.; Ghatasheh, S. Current perspectives on fenugreek bioactive compounds and their potential impact on human health: A review of recent insights into functional foods and other high value applications. J. Food Sci. 2024, 89, 1835–1864. [Google Scholar] [CrossRef]
  52. Pournamdari, M.; Mandegary, A.; Sharififar, F.; Zarei, G.; Zareshahi, R.; Asadi, A.; Mehdipour, M. Anti-Inflammatory Subfractions Separated from Acidified Chloroform Fraction of Fenugreek Seeds (Trigonella foenum-graecum L.). J. Diet. Suppl. 2018, 15, 98–107. [Google Scholar] [CrossRef] [PubMed]
  53. Idris, S.; Mishra, A.; Khushtar, M. Recent Therapeutic Interventions of Fenugreek Seed: A Mechanistic Approach. Drug Res. 2021, 71, 180–192. [Google Scholar] [CrossRef]
  54. Yang, J.; Zhang, Z.; Wu, Q.; Ding, X.; Yin, C.; Yang, E.; Sun, D.; Wang, W.; Yang, Y.; Guo, F. Multiple responses optimization of antioxidative components extracted from Fenugreek seeds using response surface methodology to identify their chemical compositions. Food Sci. Nutr. 2022, 10, 3475–3484. [Google Scholar] [CrossRef] [PubMed]
  55. Almalki, D.A.; Naguib, D.M. Anticancer Activity of Aqueous Fenugreek Seed Extract Against Pancreatic Cancer, Histological Evidence. J. Gastrointest. Cancer 2022, 53, 683–686. [Google Scholar] [CrossRef]
  56. Yang, L.; Chen, L.; Zheng, K.; Ma, Y.J.; He, R.X.; Arowolo, M.A.; Zhou, Y.J.; Xiao, D.F.; He, J.H. Effects of fenugreek seed extracts on growth performance and intestinal health of broilers. Poult. Sci. 2022, 101, 101939. [Google Scholar] [CrossRef] [PubMed]
  57. Li, G.; Luan, G.; He, Y.; Tie, F.; Wang, Z.; Suo, Y.; Ma, C.; Wang, H. Polyphenol Stilbenes from Fenugreek (Trigonella foenum-graecum L.) Seeds Improve Insulin Sensitivity and Mitochondrial Function in 3T3-L1 Adipocytes. Oxidative Med. Cell. Longev. 2018, 2018, 7634362. [Google Scholar] [CrossRef] [PubMed]
  58. Naicker, N.; Nagiah, S.; Phulukdaree, A.; Chuturgoon, A. Trigonella foenum-graecum Seed Extract, 4-Hydroxyisoleucine, and Metformin Stimulate Proximal Insulin Signaling and Increase Expression of Glycogenic Enzymes and GLUT2 in HepG2 Cells. Metab. Syndr. Relat. Disord. 2016, 14, 114–120. [Google Scholar] [CrossRef] [PubMed]
  59. Knott, E.J.; Richard, A.J.; Mynatt, R.L.; Ribnicky, D.; Stephens, J.M.; Bruce-Keller, A. Fenugreek supplementation during high-fat feeding improves specific markers of metabolic health. Sci. Rep. 2017, 7, 12770. [Google Scholar] [CrossRef]
  60. Baquer, N.Z.; Kumar, P.; Taha, A.; Kale, R.K.; Cowsik, S.M.; McLean, P. Metabolic and molecular action of Trigonella foenum-graecum (fenugreek) and trace metals in experimental diabetic tissues. J. Biosci. 2011, 36, 383–396. [Google Scholar] [CrossRef]
  61. Khound, R.; Shen, J.; Song, Y.; Santra, D.; Su, Q. Phytoceuticals in Fenugreek Ameliorate VLDL Overproduction and Insulin Resistance via the Insig Signaling Pathway. Mol. Nutr. Food Res. 2018, 62. [Google Scholar] [CrossRef]
  62. Ganeshpurkar, A.; Diwedi, V.; Bhardwaj, Y. In vitro α -amylase and α-glucosidase inhibitory potential of Trigonella foenum-graecum leaves extract. Ayu 2013, 34, 109–112. [Google Scholar] [CrossRef] [PubMed]
  63. Al-Habori, M.; Raman, A.; Lawrence, M.J.; Skett, P. In vitro effect of fenugreek extracts on intestinal sodium-dependent glucose uptake and hepatic glycogen phosphorylase A. Int. J. Exp. Diabetes Res. 2001, 2, 91–99. [Google Scholar] [CrossRef] [PubMed]
  64. Singh, S.; Chaurasia, P.K.; Bharati, S. Hypoglycemic and Hypocholesterolemic Properties of Fenugreek: A Comprehensive Assessment. Appl. Food Res. 2023, 3, 100311. [Google Scholar] [CrossRef]
  65. Deshpande, P.O.; Mohan, V.; Pore, M.P.; Gumaste, S.; Thakurdesai, P.A. Prenatal Developmental Toxicity Study of Glycosides-based Standardized Fenugreek Seed Extract in Rats. Pharmacogn. Mag. 2017, 13 (Suppl. 1), S135–S141. [Google Scholar] [CrossRef] [PubMed]
  66. Kalailingam, P.; Kannaian, B.; Tamilmani, E.; Kaliaperumal, R. Efficacy of natural diosgenin on cardiovascular risk, insulin secretion, and beta cells in streptozotocin (STZ)-induced diabetic rats. Phytomedicine Int. J. Phytother. Phytopharm. 2014, 21, 1154–1161. [Google Scholar] [CrossRef] [PubMed]
  67. Subramanian, S.P.; Prasath, G.S. Antidiabetic and antidyslipidemic nature of trigonelline, a major alkaloid of fenugreek seeds studied in high-fat-fed and low-dose streptozotocin-induced experimental diabetic rats. Biomed. Prev. Nutr. 2014, 4, 475–480. [Google Scholar] [CrossRef]
  68. Aldakinah, A.A.; Al-Shorbagy, M.Y.; Abdallah, D.M.; El-Abhar, H.S. Trigonelline and vildagliptin antidiabetic effect: Improvement of insulin signalling pathway. J. Pharm. Pharmacol. 2017, 69, 856–864. [Google Scholar] [CrossRef] [PubMed]
  69. Alsuliam, S.M.; Albadr, N.A.; Almaiman, S.A.; Al-Khalifah, A.S.; Alkhaldy, N.S.; Alshammari, G.M. Fenugreek Seed Galactomannan Aqueous and Extract Protects against Diabetic Nephropathy and Liver Damage by Targeting NF-κB and Keap1/Nrf2 Axis. Toxics 2022, 10, 362. [Google Scholar] [CrossRef] [PubMed]
  70. Broca, C.; Breil, V.; Cruciani-Guglielmacci, C.; Manteghetti, M.; Rouault, C.; Derouet, M.; Rizkalla, S.; Pau, B.; Petit, P.; Ribes, G.; et al. Insulinotropic agent ID-1101 (4-hydroxyisoleucine) activates insulin signaling in rat. Am. J. Physiol. Endocrinol. Metab. 2004, 287, E463–E471. [Google Scholar] [CrossRef]
  71. Singh, R.; Yadav, K.S.; Prajapati, R.; Sharma, S.; Rath, S.K.; Narender, T.; Mugale, M.N. 4-HIL mitigates type-2 diabetic complications through inhibiting inflammation and Nrf2 mediated oxidative stress in rats. Phytomedicine Plus 2022, 2, 100141. [Google Scholar] [CrossRef]
  72. Hannan, J.M.; Ali, L.; Rokeya, B.; Khaleque, J.; Akhter, M.; Flatt, P.R.; Abdel-Wahab, Y.H. Soluble dietary fibre fraction of Trigonella foenum-graecum (fenugreek) seed improves glucose homeostasis in animal models of type 1 and type 2 diabetes by delaying carbohydrate digestion and absorption, and enhancing insulin action. Br. J. Nutr. 2007, 97, 514–521. [Google Scholar] [CrossRef] [PubMed]
  73. Xue, W.L.; Li, X.S.; Zhang, J.; Liu, Y.H.; Wang, Z.L.; Zhang, R.J. Effect of Trigonella foenum-graecum (fenugreek) extract on blood glucose, blood lipid and hemorheological properties in streptozotocin-induced diabetic rats. Asia Pac. J. Clin. Nutr. 2007, 16 (Suppl. 1), 422–426. [Google Scholar] [PubMed]
  74. Eidi, A.; Eidi, M.; Sokteh, M. Effect of fenugreek (Trigonella foenum graecum L) seeds on serum parameters in normal and streptozotocin-induced rats. Nutr. Res. 2007, 27, 728–733. [Google Scholar] [CrossRef]
  75. Bafadam, S.; Mahmoudabady, M.; Niazmand, S.; Rezaee, S.A.; Soukhtanloo, M. Cardioprotective effects of Fenugreek (Trigonella foenum-graceum) seed extract in streptozotocin induced diabetic rats. J. Cardiovasc. Thorac. Res. 2021, 13, 28–36. [Google Scholar] [CrossRef] [PubMed]
  76. Cortez-Navarrete, M.; Pérez-Rubio, K.G.; Escobedo-Gutiérrez, M.d.J. Role of Fenugreek, Cinnamon, Curcuma longa, Berberine and Momordica charantia in Type 2 Diabetes Mellitus Treatment: A Review. Pharmaceuticals 2023, 16, 515. [Google Scholar] [CrossRef] [PubMed]
  77. Neelakantan, N.; Narayanan, M.; de Souza, R.J.; van Dam, R.M. Effect of fenugreek (Trigonella foenum-graecum L.) intake on glycemia: A meta-analysis of clinical trials. Nutr. J. 2014, 13, 7. [Google Scholar] [CrossRef] [PubMed]
  78. Willcox, M.L.; Elugbaju, C.; Al-Anbaki, M.; Lown, M.; Graz, B. Effectiveness of Medicinal Plants for Glycaemic Control in Type 2 Diabetes: An Overview of Meta-Analyses of Clinical Trials. Front. Pharmacol. 2021, 12, 777561. [Google Scholar] [CrossRef] [PubMed]
  79. Kassaian, N.; Azadbakht, L.; Forghani, B.; Amini, M. Effect of fenugreek seeds on blood glucose and lipid profiles in type 2 diabetic patients. Int. J. Vitam. Nutr. Res. 2009, 79, 34–39. [Google Scholar] [CrossRef]
  80. Kim, J.; Noh, W.; Kim, A.; Choi, Y.; Kim, Y.S. The Effect of Fenugreek in Type 2 Diabetes and Prediabetes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Int. J. Mol. Sci. 2023, 24, 13999. [Google Scholar] [CrossRef]
  81. Pickering, E.; Steels, E.; Rao, A.; Steadman, K.J. An Exploratory Study of the Safety and Efficacy of a Trigonella foenum-graecum Seed Extract in Early Glucose Dysregulation: A Double-Blind Randomized Placebo-Controlled Trial. Pharmaceutics 2022, 14, 2453. [Google Scholar] [CrossRef]
  82. Abdel-Barry, J.A.; Al-Hakiem, M.H. Acute intraperitoneal and oral toxicity of the leaf glycosidic extract of Trigonella foenum-graecum in mice. J. Ethnopharmacol. 2000, 70, 65–68. [Google Scholar] [CrossRef] [PubMed]
  83. Kassem, A.; Al-Aghbari, A.; AL-Habori, M.; Al-Mamary, M. Evaluation of the potential antifertility effect of fenugreek seeds in male and female rabbits. Contraception 2006, 73, 301–306. [Google Scholar] [CrossRef] [PubMed]
  84. Kandhare, A.D.; Thakurdesaj, P.; Wangikar, P.; Bodhankar, A. A systematic literature review of fenugreek seed toxicity by using ToxRTool: Evidence from preclinical and clinical studies. Heliyon 2019, 5, e01536. [Google Scholar] [CrossRef] [PubMed]
  85. Faeste, C.K.; Namork, E.; Lindvik, H. Allergenicity and antigenicity of fenugreek (Trigonella foenum-graecum) proteins in foods. J. Allergy Clin. Immunol. 2009, 123, 187–194. [Google Scholar] [CrossRef] [PubMed]
  86. Faisal, Z.; Irfan, R.; Akram, N.; Manzoor, H.M.I.; Aabdi, M.A.; Anwar, M.J.; Khawar, S.; Saif, A.; Shah, Y.A.; Afzaal, M. The multifaceted potential of fenugreek seeds: From health benefits to food and nanotechnology applications. Food Sci. Nutr. 2024, 12, 2294–2310. [Google Scholar] [CrossRef] [PubMed]
  87. Trask, L.E.; Chaidarun, S.S.; Platt, D.; Parkin, C.G. Treatment with novel galactomannan derivative reduces 2-hour postprandial glucose excursions in individuals with type 2 diabetes treated with oral medications and/or insulin. J. Diabetes Sci. Technol. 2014, 8, 1018–1022. [Google Scholar] [CrossRef] [PubMed]
  88. Avalos-Soriano, A.; De la Cruz-Cordero, R.; Rosado, J.L.; Garcia-Gasca, T. 4-Hydroxyisoleucine from Fenugreek (Trigonella foenum-graecum): Effects on Insulin Resistance Associated with Obesity. Molecules 2016, 21, 1596. [Google Scholar] [CrossRef] [PubMed]
  89. Bruce-Keller, A.J.; Richard, A.J.; Fernandez-Kim, S.O.; Ribnicky, D.M.; Salbaum, J.M.; Newman, S.; Carmouche, R.; Stephens, J.M. Fenugreek Counters the Effects of High Fat Diet on Gut Microbiota in Mice: Links to Metabolic Benefit. Sci. Rep. 2020, 10, 1245. [Google Scholar] [CrossRef] [PubMed]
  90. Jones, K.A.; Richard, A.J.; Salbaum, J.M.; Newman, S.; Carmouche, R.; Webb, S.; Bruce-Keller, A.J.; Stephens, J.M.; Campagna, S.R. Cross-Omics Analysis of Fenugreek Supplementation Reveals Beneficial Effects Are Caused by Gut Microbiome Changes Not Mammalian Host Physiology. Int. J. Mol. Sci. 2022, 23, 3654. [Google Scholar] [CrossRef]
  91. Zhou, K. Strategies to promote abundance of Akkermansia muciniphila, an emerging probiotics in the gut, evidence from dietary intervention studies. J. Funct. Foods 2017, 33, 194–201. [Google Scholar] [CrossRef]
  92. Man, S.; Xie, L.; Liu, X.; Wang, G.; Liu, C.; Gao, W. Diosgenin relieves oxaliplatin-induced pain by affecting TLR4/NF-κB inflammatory signaling and the gut microbiota. Food Funct. 2023, 14, 516–524. [Google Scholar] [CrossRef]
  93. Song, C.; Ma, Y.; Wang, Y.; Li, P.; Chen, Y.; Liu, H.; Zhang, Z. Diosgenin reduces bone loss through the regulation of gut microbiota in ovariectomized rats. Gene 2023, 869, 147383. [Google Scholar] [CrossRef]
  94. Anwar, S.; Bhandari, U.; Panda, B.P.; Dubey, K.; Khan, W.; Ahmad, S. Trigonelline inhibits intestinal microbial metabolism of choline and its associated cardiovascular risk. J Pharm. Biomed. Anal. 2018, 159, 100–112. [Google Scholar] [CrossRef]
  95. Shtriker, M.G.; Hahn, M.; Taieb, E.; Nyska, A.; Moallem, U.; Tirosh, O.; Madar, Z. Fenugreek galactomannan and citrus pectin improve several parameters associated with glucose metabolism and modulate gut microbiota in mice. Nutrition 2018, 46, 134–142.e3. [Google Scholar] [CrossRef] [PubMed]
  96. Ben-Nasr, H.; Bouzidi, A.; Gharbi, A.; Hammamy, S.; Zeghal, K. Some biological effects of Trigonella foenum- graecom L in healthy men. Endocrinol. Metab. Int. J. 2018, 6, 440–443. [Google Scholar] [CrossRef]
  97. King, K.; Lin, N.P.; Cheng, Y.H.; Chen, G.H.; Chein, R.J. Isolation of Positive Modulator of Glucagon-like Peptide-1 Signaling from Trigonella foenum-graecum (Fenugreek) Seed. J. Biol. Chem. 2015, 290, 26235–26248. [Google Scholar] [CrossRef]
  98. Shabil, M.; Bushi, G.; Bodige, P.K.; Maradi, P.S.; Patra, B.P.; Padhi, B.K.; Khubchandani, J. Effect of Fenugreek on Hyperglycemia: A Systematic Review and Meta-Analysis. Medicina 2023, 59, 248. [Google Scholar] [CrossRef] [PubMed]
  99. Bordia, A.; Verma, S.K.; Srivastava, K.C. Effect of ginger (Zingiber officinale Rosc.) and fenugreek (Trigonella foenumgraecum L.) on blood lipids, blood sugar and platelet aggregation in patients with coronary artery disease. Prostaglandins Leukot. Essent. Fat. Acids 1997, 56, 379–384. [Google Scholar] [CrossRef]
  100. Syvänne, M.; Taskinen, M.R. Lipids and lipoproteins as coronary risk factors in non-insulin-dependent diabetes mellitus. Lancet 1997, 350 (Suppl. 1), SI20–SI23. [Google Scholar] [CrossRef]
  101. Sheweita, S.A.; ElHady, S.A.; Hammoda, H.M. Trigonella stellata reduced the deleterious effects of diabetes mellitus through alleviation of oxidative stress, antioxidant- and drug-metabolizing enzymes activities. J. Ethnopharmacol. 2020, 256, 112821. [Google Scholar] [CrossRef]
  102. Zameer, S.; Najmi, A.K.; Vohora, D.; Akhtar, M. A review on therapeutic potentials of Trigonella foenum graecum (fenugreek) and its chemical constituents in neurological disorders: Complementary roles to its hypolipidemic, hypoglycemic, and antioxidant potential. Nutr. Neurosci. 2018, 21, 539–545. [Google Scholar] [CrossRef] [PubMed]
  103. Foretz, M.; Guigas, B.; Bertrand, L.; Pollak, M.; Viollet, B. Metformin: From mechanisms of action to therapies. Cell Metab. 2014, 20, 953–966. [Google Scholar] [CrossRef] [PubMed]
  104. Karimi, A.; Majlesi, M.; Rafieian-Kopaei, M. Herbal versus synthetic drugs; beliefs and facts. J. Nephropharmacology 2015, 4, 27–30. [Google Scholar]
  105. Mehrzadi, S.; Mirzaei, R.; Heydari, M.; Sasani, M.; Yaqoobvand, B.; Huseini, H.F. Efficacy and Safety of a Traditional Herbal Combination in Patients with Type II Diabetes Mellitus: A Randomized Controlled Trial. J. Diet. Suppl. 2021, 18, 31–43. [Google Scholar] [CrossRef] [PubMed]
  106. Hadi, A.; Arab, A.; Hajianfar, H.; Talaei, B.; Miraghajani, M.; Babajafari, S.; Marx, W.; Tavakoly, R. The effect of fenugreek seed supplementation on serum irisin levels, blood pressure, and liver and kidney function in patients with type 2 diabetes mellitus: A parallel randomized clinical trial. Complement. Ther. Med. 2020, 49, 102315. [Google Scholar] [CrossRef] [PubMed]
  107. Anand, P.; Kunnumakkara, A.B.; Newman, R.A.; Aggarwal, B.B. Bioavailability of curcumin: Problems and promises. Mol. Pharm. 2007, 4, 807–818. [Google Scholar] [CrossRef] [PubMed]
  108. Navarro del Hierro, J.; Reglero, G.; Martin, D. Chemical Characterization and Bioaccessibility of Bioactive Compounds from Saponin-Rich Extracts and Their Acid-Hydrolysates Obtained from Fenugreek and Quinoa. Foods. 2020, 9, 1159. [Google Scholar] [CrossRef] [PubMed]
  109. Sheethal, S.; Ratheesh, M.; Jose, S.P.; Asha, S.; Krishnakumar, I.M.; Sandya, S.; Girishkumar, B.; Grace, J. Anti-Ulcerative Effect of Curcumin-Galactomannoside Complex on Acetic Acid-Induced Experimental Model by Inhibiting Inflammation and Oxidative Stress. Inflammation 2020, 43, 1411–1422. [Google Scholar] [CrossRef] [PubMed]
  110. Krishnakumar, I.M.; Abhilash, R.; Dinesh, K.; Ramadasan, K.; Balu, M. An enhanced bioavailable formulation of curcumin using fenugreek-derived soluble dietary fibre. J. Funct. Foods 2012, 4, 348–357. [Google Scholar] [CrossRef]
  111. Weersma, R.K.; Zhernakova, A.; Fu, J. Interaction between drugs and the gut microbiome. Gut 2020, 69, 1510–1519. [Google Scholar] [CrossRef]
  112. Wei, M.; Li, G.; Xie, H.; Yang, W.; Xu, H.; Han, S.; Wang, J.; Meng, Y.; Xu, Q.; Li, Y.; et al. Sustainable production of 4-hydroxyisoleucine with minimised carbon loss by simultaneously utilising glucose and xylose in engineered Escherichia coli. Bioresour. Technol. 2022, 354, 127196. [Google Scholar] [CrossRef] [PubMed]
  113. Lambert, J.P.; Cormier, J. Potential interaction between warfarin and boldo-fenugreek. Pharmacotherapy 2001, 21, 509–512. [Google Scholar] [CrossRef] [PubMed]
  114. Mathur, R.; Farmer, R.E.; Eastwood, S.V.; Chaturvedi, N.; Douglas, I.; Smeeth, L. Ethnic disparities in initiation and intensification of diabetes treatment in adults with type 2 diabetes in the UK, 1990-2017: A cohort study. PLoS Med. 2020, 17, e1003106. [Google Scholar] [CrossRef] [PubMed]
  115. Gaddam, A.; Galla, C.; Thummisetti, S.; Marikanty, R.K.; Palanisamy, U.D.; Rao, P.V. Role of Fenugreek in the prevention of type 2 diabetes mellitus in prediabetes. J. Diabetes Metab. Disord. 2015, 14, 74. [Google Scholar] [CrossRef] [PubMed]
  116. Luo, W.; Deng, J.; He, J.; Yin, L.; You, R.; Zhang, L.; Shen, J.; Han, Z.; Xie, F.; He, J.; et al. Integration of molecular docking, molecular dynamics and network pharmacology to explore the multi-target pharmacology of fenugreek against diabetes. J. Cell. Mol. Med. 2023, 27, 1959–1974. [Google Scholar] [CrossRef]
  117. Tiwari, P.C.; Pal, R.; Chaudhary, M.J.; Nath, R. Artificial intelligence revolutionizing drug development: Exploring opportunities and challenges. Drug Dev. Res. 2023, 84, 1652–1663. [Google Scholar] [CrossRef]
Figure 1. Proposed biological targets of fenugreek and their molecular antidiabetic effects summarized from reviewing in vitro experiments and in vivo animal studies. Abbreviations: HbA1c—hemoglobin A1c, HDL—high-density lipoprotein, TG—triglycerides, LDL—low-density lipoproteins, SOD—superoxide dismutase, CAT—catalase, GPx—glutathione peroxidase, GR—glutathione reductase, MDA—malondialdehyde, GLUT-4—glucose transporter type 4, IRS-1—insulin receptor substrate-1, GLP-1—glucagon-like peptide-1.
Figure 1. Proposed biological targets of fenugreek and their molecular antidiabetic effects summarized from reviewing in vitro experiments and in vivo animal studies. Abbreviations: HbA1c—hemoglobin A1c, HDL—high-density lipoprotein, TG—triglycerides, LDL—low-density lipoproteins, SOD—superoxide dismutase, CAT—catalase, GPx—glutathione peroxidase, GR—glutathione reductase, MDA—malondialdehyde, GLUT-4—glucose transporter type 4, IRS-1—insulin receptor substrate-1, GLP-1—glucagon-like peptide-1.
Ijms 25 06987 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Haxhiraj, M.; White, K.; Terry, C. The Role of Fenugreek in the Management of Type 2 Diabetes. Int. J. Mol. Sci. 2024, 25, 6987. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25136987

AMA Style

Haxhiraj M, White K, Terry C. The Role of Fenugreek in the Management of Type 2 Diabetes. International Journal of Molecular Sciences. 2024; 25(13):6987. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25136987

Chicago/Turabian Style

Haxhiraj, Melina, Kenneth White, and Cassandra Terry. 2024. "The Role of Fenugreek in the Management of Type 2 Diabetes" International Journal of Molecular Sciences 25, no. 13: 6987. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25136987

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

Article Metrics

Back to TopTop