Next Article in Journal
Effects of Different Na+ Concentrations on cAMP-Dependent Protein Kinase Activity in Postmortem Meat
Previous Article in Journal
Enhancement of Colorimetric pH-Sensitive Film Incorporating Amomum tsao-ko Essential Oil as Antibacterial for Mantis Shrimp Spoilage Tracking and Fresh-Keeping
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin K2 in Health and Disease: A Clinical Perspective

1
School of Food Science & Environmental Health, Technological University Dublin, Grangegorman, 7, D07 ADY7 Dublin, Ireland
2
The Trinity Centre for Natural Products Research (NatPro), School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, 2, D02 PN40 Dublin, Ireland
3
School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, 2, D02 PN40 Dublin, Ireland
4
Department of Chemistry, RCSI University of Medicine and Health Sciences, 2, D02 YN77 Dublin, Ireland
*
Authors to whom correspondence should be addressed.
Foods 2024, 13(11), 1646; https://doi.org/10.3390/foods13111646
Submission received: 30 April 2024 / Revised: 21 May 2024 / Accepted: 23 May 2024 / Published: 24 May 2024
(This article belongs to the Section Food Nutrition)

Abstract

:
Vitamins are essential organic compounds that vary widely in chemical structure and are vital in small quantities for numerous biochemical and biological functions. They are critical for metabolism, growth, development and maintaining overall health. Vitamins are categorised into two groups: hydrophilic and lipophilic. Vitamin K (VK), a lipophilic vitamin, occurs naturally in two primary forms: phylloquinone (VK1), found in green leafy vegetables and algae, and Menaquinones (VK2), present in certain fermented and animal foods and widely formulated in VK supplements. This review explores the possible factors contributing to VK deficiency, including dietary influences, and discusses the pharmacological and therapeutic potential of supplementary VK2, examining recent global clinical studies on its role in treating diseases such as osteoporosis, osteoarthritis, rheumatoid arthritis, cardiovascular disease, chronic kidney disease, diabetes, neurodegenerative disorders and cancers. The analysis includes a review of published articles from multiple databases, including Scopus, PubMed, Google Scholar, ISI Web of Science and CNKI, focusing on human studies. The findings indicate that VK2 is a versatile vitamin essential for human health and that a broadly positive correlation exists between VK2 supplementation and improved health outcomes. However, clinical data are somewhat inconsistent, highlighting the need for further detailed research into VK2′s metabolic processes, biomarker validation, dose–response relationships, bioavailability and safety. Establishing a Recommended Daily Intake for VK2 could significantly enhance global health.

Graphical Abstract

1. Introduction

Vitamin K (VK) comprises a group of menadione derivatives, first isolated from natural (plant) sources. The key role of vitamin K in blood clotting was first recognized in 1936 by Henrik Dam [1], who observed significant coagulation dysfunction in chickens fed a low-fat diet, resulting in severe bleeding. The later discovery of structurally analogous compounds in fermented foods led to the original vitamin K substance being termed phylloquinone or vitamin K1 (VK1), while those forms contained in some fermented and animal foods were collectively termed menaquinones or vitamin K2 (VK2). VK2 can also be generated by Gram-positive and Gram-negative bacteria in the human body [2]. Vitamin K3 (menadione) and vitamin K4 (menadione acetate), prepared by chemical synthesis, also have pro-coagulative effects. Figure 1 shows the structures of these substances, highlighting their shared bicyclic core. VK1 and VK2 feature a variable side chain at the 3-position of the quinone system: VK1 has a branched aliphatic side chain, featuring one double bond, while VK2 possesses a polymeric side chain with variable numbers of isoprene units [3]. The saturated alkane moiety of VK1 permits free rotation, while the repeating alkene units within VK2 confer strong rigidity and distinct cis- and trans-isomers, only the trans forms of which are biologically active [4]. As polymerised isoprene is of terpenoid origin, from a side chain perspective, VK2 may be considered a terpene, with MK-4 (4 isoprene units) and MK-7 (7 isoprene units).
VK1 is the main form of VK in the human diet, found mainly in green leafy vegetables (e.g., spinach, cabbage, kale), fruits (e.g., avocado, kiwi, grapes), as well as in some plant oils (e.g., soybean oil) [3]. VK2 is mainly synthesised by gut bacteria and exists in meat, dairy and fermented foods [5,6,7]. Natto, a traditional Japanese food, is noteworthy as the richest known source of VK2, with more than 100 times the VK2 content of other sources. Natto is produced through the fermentation of soybeans with Bacillus subtilis, primarily as MK-7 [3]. Dairy products are also rich in VK2, with hard cheeses a notable source [8]. The relevant levels of VK1 and VK2 in foods are shown in Figure 2 [9]. In addition, VK2 (as MK-4) can be synthesised from VK1 in certain animal tissues. This transformation is mediated via the UbiA prenyltransferase domain-containing 1 enzyme and involves menadione as an intermediate [10]. MK-4 derived from ingested VK1 is present at high levels in animals [11], suggesting that VK1 absorbed by herbivores from green leaves is converted into VK2 in the body and can then perform physiological functions that VK1 cannot achieve. This is of great significance in the context of species survival and evolution. It also suggests that humans, on the apex of the food chain, may need a balanced intake of animal and plant foods, which is of significance in considerations of supplementing VK1 and VK2 necessary to maintain homeostasis. In the same vein, vegetarians should consider consuming fermented foodstuffs to maintain this balance. In our previous review on VK2 [12], we demonstrated that VK2 possesses multiple biological functions through the lens of in vivo and in vitro studies and plays a wide range of roles in maintaining human health. In the present study, we reviewed different possible causes of VK2 deficiency and highlighted the clinical evidence for the beneficial roles of VK2 in diverse disease states.
The methodology used to construct this article included a review of published reports from various databases (from inception to 30 November 2022 and updated on 04 December 2023, without language restrictions): Scopus, PubMed, Google Scholar, ISI Web of Science and CNKI, to maximise the retrieval of relevant results. We considered both review and original research articles involving human studies. The search keywords utilised for the literature screening were “vitamin K2” or “menaquinone” in combination with “health” or “diseases”. The database search was supplemented by consulting the bibliography of the articles, reviews and published meta-analyses. The literature research was not limited to a time period, but a particular focus was given to studies from the past 20 years. Relevant articles were chosen after reviewing all titles and abstracts, and full texts were obtained if the information contained in the title or abstract was insufficient to exclude the study. When the form of VK used (whether VK1 or VK2) was not specified, those studies were excluded, unless the outcomes of the studies were considered to be important and significant to report.

2. Etiology of Decreased Vitamin K2 Synthesis and Absorption

At present, population-based epidemiological studies of VK2 deficiency are limited, as low levels of VK2 are typically found in normal blood, in the absence of VK2 supplementation or in cases of high intake of VK2-rich foods. There is also no harmonised detection index or cut-off value for evaluating VK2 status (measurement of coagulation factors or prothrombin time is a common surrogate in clinical practice). There is also no distinction between VK1 and VK2 in countries with established dietary recommendations for VK [9]. Despite these observations, it has been noted that VK2 intake from the food sources of today is generally lacking, especially in developing countries [13]. Possible stressors include environmental pollution, industrial farming practices, artificial feeding, residues of pesticides and fertilisers in agricultural products and the widespread use of preservatives, additives and antibiotics. All these could have an impact on VK2 intake and the synthesis of endogenous VK2. Eating foods rich in VK2 could be a good option to counter declining levels, but in practice, many people, due to dietary preferences (certain Asian diets, vegans, or vegetarians) may struggle to achieve this. Further possible causes of VK2 deficiency are an imbalance in intestinal flora, issues regarding in vivo absorption and/or metabolism and drug–drug interactions.

2.1. The Imbalance of Intestinal Flora

Although the intestinal microbiome can produce a small amount of endogenous VK2, these levels may not meet the needs of the human body, and, in addition, colonic absorption of VK2 is limited [14]. Modernisation of agricultural production, with resulting residues of pesticides and fertilizers in agricultural products and the practices of the food processing industry, including the use of preservatives and artificial food additives, have caused the dysregulation of human intestinal flora [15,16], further impacting negatively upon VK2 production. Menaquinones have been highlighted as a major class of growth factors for taxonomically diverse bacteria from the human gut microbiome, including Faecalibacterium, Bacteroides, Bilophila, Gordonibacter and Sutterella species [17]. The importance of food quality and a well-balanced diet, particularly with ageing, has been highlighted in a recent review of vitamin K in the context of diet and the gut microbiome [18]. An interesting observation is the fact that VK1 absorption from fruit and vegetables may be lower than that from processed plant oils associated with unhealthy diets; likewise, for VK2, processed meat products are a significant source of menaquinones, alongside more healthy choices such as fermented foods. Also, exogenous VK may undergo microbiome-initiated remodelling [19]. The complexities of the impact of our dietary choices on both VK bioavailability and modulating gut microbiome composition, and therefore VK status, are increasingly realised.
Antibiotics can also reduce VK2 production by destroying VK2-producing bacteria in the gut. This effect is particularly pronounced with cephalosporins such as cefoperazone [20]. In addition, proton pump inhibitors can also impact VK2 status [21], possibly through effects on the intestinal flora.

2.2. Physiological and Genetic Factors

Many objective physiological factors influence VK2 absorption and utilisation. The absorptive capacity of the human gastrointestinal tract gradually declines with age, which could cause VK2 deficiency in middle-aged and elderly people and, thus, merit dietary supplementation [22]. In addition, the decreased absorptive ability of newborns, patients with liver and gallbladder diseases and elderly patients with metabolic diseases could also lead to VK2 deficiency [22,23].
Experimental evidence in this regard was shown by Holden and colleagues [24]. In this observational and prospective study involving 167 chronic kidney disease (CKD) patients, it was observed that patients with the CG/GG genotype of vitamin K epoxide reductase complex subunit 1 (the target enzyme for warfarin) had a higher risk of coronary artery calcification (CAC) progression and poorer survival, suggesting the potential role of VK2.
Gamma-glutamyl carboxylase (GGCX) gene single nucleotide polymorphism (R325Q, 974G > A) is associated with bone mineral density (BMD). Haraikawa et al. studied the effect of GGCX gene single nucleotide polymorphism (974G > A) on vitamin K intake, serum vitamin K levels and the ratio of uncarboxylated osteocalcin (ucOC) to carboxylated osteocalcin (OC) in healthy young Japanese subjects. The results showed a significant negative correlation (p < 0.001) between the ucOC/OC ratio and vitamin K intake in homozygotes (GG type) and heterozygotes (GA type) [25]. Therefore, appropriate supplementation of vitamin K2 is warranted for people with high-risk genotypes (GG or GA).

2.3. Drug Interactions

Vitamin K interacts with certain drugs, which affect the concentration and action of VK2 in the body. Warfarin is a coumarin anticoagulant and vitamin K antagonist (VKA), blocking the circulation of VK in the liver and periphery, leading to secondary vitamin K deficiency, which has a negative impact on blood vessels, bones, kidneys, brain and other tissues and systems (e.g., inflammation, immune function and tumours). VK2 has the potential for serious drug interactions with other coumarin anticoagulants, such as phenprocoumon, acenocoumarol and tioclomarol [26].
The cholesterol-lowering drugs cholestyramine and colestipol are anion-exchange resins that reduce cholesterol levels by preventing the reabsorption of bile acids, but they also decrease the absorption of VK and other lipid-soluble vitamins [27,28]. Orlistat is a weight loss drug that reduces intestinal absorption of dietary fat but may also reduce VK absorption [29]. However, in a study of obese teenagers taking supplementary vitamins, including 25 µg VK (as synthetic VK1, phytonadione), there was a nonsignificant decrease in VK status [29].
Statins are commonly used lipid-lowering drugs, lowering cholesterol and thereby reducing atherosclerosis. However, it has been shown that statins may cause CAC. A possible mechanism may be a toxic effect on mitochondria, impairing muscle function in the heart and blood vessels by depleting Coenzyme Q10 and heme A. Statins also inhibit VK2 synthesis [30]. VK2 is a cofactor for the activation of matrix Gla protein (MGP), which protects arteries from calcification. It has been postulated that the high incidence of heart failure and atherosclerosis may thus be paradoxically exacerbated by the widespread use of statins [28,31].
Patients with CKD often experience hyperphosphatemia, and treatment with phosphorus-lowering drugs is required. Studies have shown that phosphate binders have adverse effects on the bioavailability of VK2. In vitro experiments have shown that calcium acetate/magnesium carbonate, lanthanum carbonate, calcium carbonate and sevelamer carbonate can all combine with VK2, which affects the absorption of VK2 and may counteract the ability of VK2 to inhibit vascular calcification [32]. In a study of various CKD patients taking phosphate binders, dephosphorylated-uncarboxylated MGP levels were higher in those taking sevelamer, suggesting reduced vitamin K status [33]. However, the clinical relevance of these interactions has been challenged [34].

3. Potential Therapeutic Benefits

Research conducted in recent decades has suggested the protective roles of VK2 in tissue mineralisation-, inflammation-, oxidation- and age-related conditions, which could provide new directions for VK2 in future clinical practice. The aim of this section is to summarise and analyse recent available clinical evidence on the relationship between VK2 intake and status and to describe its modulatory effects in various disease states (Figure 3).

3.1. Osteoporosis

Osteoporosis is a metabolic skeletal disorder, characterised by decreased BMD, fragile bone and increased risk of fracture. BMD typically declines with age, particularly in postmenopausal women [35], and has important consequences on general health and quality of life. Many factors are associated with osteoporosis, including genetics, age, sex, ethnicity and hormone levels, in addition to lifestyle variables such as smoking, diet and alcohol [36]. An estimated nine million fractures annually are caused by osteoporosis [37].
Long-established preventative interventions such as calcium supplementation have been challenged, due to a claimed association with an increased risk of heart disease through enhanced calcification of blood vessel walls and soft tissue [38]. More evidence-based preventative strategies are thus needed. The role of VK2 in both diet and in supplementary form has been evaluated. In Japan, researchers established a positive relationship between intake of the fermented soybean product, natto (containing VK2 380 μg/pack and VK1 20 μg/pack), and BMD among 1662 healthy men [39]. Various studies of VK, typically co-administered with vitamin D, at typical VK2 doses ranging up to 45 mg/d, have shown increases in BMD and a reduced incidence of fractures [40].
In a meta-analysis conducted by Fang et al., VK supplementation (VK1, VK2) effectively increased BMD at the lumbar spine, but not the femoral neck. The weighted mean difference in absolute BMD change was 21.60 mg/cm2 at the lumbar spine, but only 0.25 mg/cm2 at the femoral neck, while the relative change was 1.27% at the lumbar spine and 0.17% at the femoral neck. A subgroup analysis revealed a significantly favourable effect on lumbar spine BMD by VK2, rather than VK1. Moreover, ethnicity, gender and VK type were also associated with variable effects on BMD in the lumbar spine. Most of the included trials were conducted in women only [41]. In a more recently published review on VK2 and other nutrients and bone health [42], different effects of VK2 on BMD and fracture risk from several meta-analyses were reported [43,44], with many seeming to confirm the potential benefits of VK2 supplementation in osteoporotic patients, possibly via enhancement of the effects of calcium and vitamin D. However, biases in several studies were noted. As a result, the authors suggested that the efficacy of VK intake and/or supplementation to prevent or treat sarcopenia needs to be further studied, due to insufficient data [42]. Similar findings were echoed in an even more recent meta-analysis [45]. More specifically, the efficiency of MK-7 supplementation was shown, as it accumulates to 7–8-fold higher levels than VK1 upon chronic administration and significantly promotes γ-carboxylation of OC, increasing cOC levels while decreasing ucOC plasma levels, thus promoting bone metabolism [46,47]. Inaba et al. further suggested that >100 µg/d of MK-7 supplementation could be considered for enhancing bone health [46].
The synergistic effect of VK2 supplementation with antiresorptives has also been explored. Co-administration with alendronate facilitated the decrease in bone turnover markers and ucOC levels in post-menopausal women [48] and, with anabolic agents (teriparatide), increased osteoblastic lineage activation and bone healing in animals [49].
Finally, a body of evidence, from in vitro and in vivo studies, supports the pathogenic role of oxidative stress contributing to osteoporosis (especially in postmenopausal osteoporosis), most likely in synergy with inflammation [50]. Therefore, employing VK supplements including VK2 as dietary antioxidants, with the aim of decreasing levels of oxidative stress with possible beneficial effects on bone, has been suggested [3].
In Japan, VK2 is recommended as a second-line drug in guidelines on the management and treatment of glucocorticoid-induced osteoporosis by the Japanese Society for Bone and Mineral Research [51], and a 250–300 µg of daily intake of MK-7 is also recommended as a therapeutic and prophylactic medication for osteoporosis [52]. In Europe, the adequate daily intake of VK from food (as VK1) was set at 70 μg by The European Food Safety Authority for all adults, including pregnant and lactating women [53].

3.2. Osteoarthritis (OA) and Rheumatoid Arthritis (RA)

Currently, there are no effective treatments to treat OA pathogenesis, and available clinical data regarding the effects of VK on OA are also limited in providing definitive cause–effect information [22]. Certain vitamin K-dependent proteins are found in joint tissues, including cartilage, bone and periosteum, and imbalance in the action of these proteins due to genetic factors is associated with OA [54]. Evidence from both animal and human studies suggests VK status may be associated with OA risk. Specifically, a population-based cross-sectional study of 719 Japanese participants (449 females, ≥60 years) showed that an increased risk of knee OA development was associated with lower VK intake [55]. This finding was in line with the prior Framingham Offspring study, which involved 672 participants (358 females; mean age of 65.6). After adjustment for age, sex, body mass index, total energy intake, plasma vitamin D and femoral neck BMD, the data supported an inverse association of VK status with OA in the hand and knee [56]. Properly designed clinical trials of VK supplementation in OA patients are thus warranted.
Some evidence exists demonstrating a modulating effect of VK2 in inflammation-based arthritis, such as rheumatoid arthritis (RA). Ebina et al. presented both cross-sectional and longitudinal associations between VK2 supplementation (45 mg/d) and significantly reduced inflammation in RA patients [measured by marker proteins including C-reactive protein (CRP)], as compared to the VK2-naïve group [57]. In an in vitro study using mitogen-activated peripheral blood mononuclear cells of healthy subjects and RA patients, significantly enhanced immunosuppressive efficacy of methotrexate by VK2 was demonstrated [58]. Similarly, another cross-sectional study also showed that RA patients who received 100 μg/d of MK-7 for 3 months showed a significant decrease in disease activity score, along with improved biomarker levels [erythrocyte sedimentation rate (ESR), CRP and MMP-3] [59]. Human studies on MK-4 in RA patients are still lacking, but several in vitro and in vivo studies have supported the anti-inflammatory action of MK-4 [60,61].

3.3. Cardiovascular Disease (CVD)

Cardiovascular calcification, mediated via various pathophysiological processes, is a central contributor to CVDs. MGP deficiency and inactivation of MGP has been repeatedly associated with VK deficiency, resulting in stimulation of all types of calcification and contributing to the development of cardiovascular events, whereas VK status (especially VK2) appears to inversely correlate with CVD risk [62]. With the aim of examining the relationship between dietary VK intake and the incidence of coronary heart disease (CHD), Gast et al. utilised data from the Prospect—The European Prevalence of Infection in Intensive Care (EPIC) study in the Netherlands. In this study, a cohort of 16,057 women between 49 and 70 years with no previous history of CVD was followed for 8.1 years [63]. The results indicated a significant negative relationship between a high VK2 intake (especially MK-7, MK-8, MK-9) and a reduced risk of CHD, after adjustment for known risk and dietary factors. Similar results were also obtained in the Rotterdam study, following 4807 participants free from myocardial infarction at baseline over a 7-year period [64]. In both these studies, VK1 intake did not significantly impact on the outcomes. However, in a more recent prospective cohort study involving 53,372 participants with 17–22 years of follow-up, both dietary VK1 and VK2 intakes were inversely related to atherosclerotic CVD hospitalisation risk [65].
CVD risk is strongly associated with kidney dysfunction, due to enhanced vascular calcification in these patients, with even the early stages of CKD causing hypertension and potentiation of the risk of CVD [66]. More than 50% of patients undergoing dialysis develop CVD, and mortality in haemodialysis patients due to CVD is 20 times higher than in the general population [67]. Intimal atherosclerotic plaque calcification and medial calcification are commonly observed in these patients [68].
To evaluate the influence of VK supplementation on the development of cardiovascular calcification in the context of CVD, several randomised and placebo-controlled trials are currently ongoing or recently completed. The Valkyrie trial investigated the effects of VK2 supplementation alongside rivaroxaban in place of VKAs on the progression of vascular calcification among haemodialysis patients (NCT02610933), and the outcomes are pending [69]. However, in a separate clinical study, VK status improved significantly upon withdrawal of VKAs and thrice-weekly supplementation of VK2 (2 mg), changes in coronary artery, thoracic aorta and cardiac valve calcium scores and pulse wave velocity were not different among the treatment arms [70]. Importantly, the intervention did not normalise systemic dephosphorylated-uncarboxylated (dp-uc)MGP levels among patients. In another study (EudraCT Number: 2019-004906-88 and NTR number: NL7687) investigating the effect of MK-7 supplementation on serum calcification propensity and arterial stiffness in vitamin K-deficient kidney transplant recipients, supplementation did not alter serum calcification propensity but prevented progression of arterial stiffness, suggesting that vitamin K has vascular effects independent of calciprotein particles [71]. In a diabetic cohort with kidney disease, MK-7 supplementation (375 µg/d, 24 weeks) lowered the rate of progression of arterial stiffness in chronic haemodialysis patients, in an open-label, multicentre randomised controlled study (TCTR20230217001) [72], while another study of arterial calcification among diabetic patients showed that six months’ supplementation with VK (as 360 μg MK-7) did not improve serum calcification propensity time [73].
Two years’ supplementation with MK-7 (720 µg/d) alongside vitamin D (25 µg/d) did not reduce the progression rate of aortic valve calcification in patients with aortic stenosis, in a randomised, double-blind and multicentre trial study (NCT03243890) [74]. The BASIK2 trial of bicuspid aortic valve stenosis is evaluating the effect of VK2 on calcification using 18F-sodium fluoride positron emission tomography/magnetic resonance (NCT02917525) [75].The VitaK-CAC trial is evaluating MK-7 supplementation to reduce vascular calcification in patients with coronary artery disease (NCT01002157) [76], while a further trial is anticipated to start in 2024 to study the effect of VK2 supplementation on arterial micro-calcification as assessed by PET/MRI in Carotid Artery Disease (INTRICATE) (NCT04010578) [77]. The InterVitamin K trial (NCT05259046) is currently investigating the effect of daily MK-7 supplementation over three years on the progression of vascular calcification in terms of the CAC score in individuals with detectable vascular calcification [78].

3.4. Chronic Kidney Disease (CKD)

Further to the association between CKD-related calcification and CVD already discussed in Section 3.3, VK deficiency is seen across patients with diverse CKDs, including urolithiasis, nephrolithiasis and uraemia [79]. VK status becomes more significant with progression through CKD stages [80] and when undergoing maintenance haemodialysis [81]. It has been shown in both animal models and human studies that MGP is overexpressed as renal dysfunction progresses and can thus indicate deterioration of renal function [82].
A multivariate analysis involving 107 patients in all five stages of CKD showed that dp-ucMGP status was a predictor of arterial calcification, independent of age, gender, previous CVD and high total-ucMGP levels, progressively augmented with the CKD stage and positively correlated with aortic calcification [83]. In a study of 141 CKD stage-5 patients, Jaminon et al. demonstrated that plasma levels of circulating dp-ucMGP were an independent predictor of increased vascular calcification in patients with end-stage CKD and correlated with both higher coronary artery calcium scores and degree of medial calcification [84]. Moreover, etiologic in vivo evidence has shown that vascular VK deficiency in uraemia may partially result from a decrease in γ-carboxylase activity, leading to an increase in uremic vascular calcification [85] and the exposure of the vasculature to a toxic uremic milieu [86].
It has further been suggested that VK intake could contribute to CKD treatment by modulating MGP, since MGP is abundantly expressed in the kidney, 5-fold more so than in bone [87]. The effect of high-dose (100 µg/g diet) MK-4 supplementation on restoration of MGP functionality in a murine CKD model has been demonstrated, suggesting that MK-4 may offer a protective effect against cardiovascular calcification in this situation [85].
In a trial involving 50 haemodialysis patients, the average decrease in plasma dp-ucMGP levels at week 4 following MK-7 treatment (360 μg/d) was found to be 86%, with the lowest drop rate (p = 0.01) in diabetics [88]. Caluwe et al. emphasised the importance of dose and time dependency of VK2 supplementation in the carboxylation of MGP in a randomised dose-finding study of 200 haemodialysis patients. MK-7 supplementation dose-dependently reduced dp-ucMGP levels by 17, 33 and 46% after receiving 360, 720 or 1080 μg of MK-7 treatment (thrice weekly for 2 months), respectively [68]. Delanaye and coauthors showed a rapid time-effect decrease in serum dp-ucMGP levels (by at least 40%) within 5 days after terminating VK antagonist treatment in seven stable haemodialysis patients [89]. Moreover, in a 270-day trial with 38 non-dialysis CKD patients, supplementation with combined MK-7 and vitamin D resulted in a decrease in plasma dp-ucMGP level by 10.7%, whereas supplementation with vitamin D alone showed no change [90]. Nigwekar et al. showed that every 0.1-unit reduction in plasma cMGP level was associated with a more than two-fold increase in calciphylaxis risk in haemodialysis patients, suggesting that aberrant VK-mediated MGP carboxylation may have a role in the pathogenesis of calciphylaxis [91]. The use of MK-7 as the VK2 form of choice in these trials probably reflects its longer half-life (72 h) than that of MK-4 (1 h).
Additionally, Wei et al. confirmed the inverse relationship between decreased estimated glomerular filtration rate (eGFR) and risk of renal impairment with higher dp-ucMGP levels in a study involving 1166 white Flemish patients (mean age 38.2 years) and 714 South Africans (49.2% Black; 40.6 years) [92], further indicating the use of dp-ucMGP as a biomarker predicting deterioration of renal function in the general population, assessed by a decrease in eGFR and increase in albuminuria [93]. This further suggests that VK supplementation to modulate MGP not only results in inhibition of arterial calcification but also protects renal function. Despite all these observations, supplementary VK2 does not appear to translate into unambiguous cardiovascular protection in chronic CKD patients. In a trial of 159 patients (mean age 66) with CKD, VK2 supplementation did not improve vascular stiffness or other measures of vascular health [94]. Likewise, in a meta-analysis of ten trials, while both levels of relevant biomarkers of vascular calcification and elasticity improved, significant changes in calcification scores were not observed [95].

3.5. Diabetes and the Metabolic Syndrome

Studies conducted over the past two decades have lent additional support to the role of VK in diabetes, mainly Type 2-diabetes (T2-D). Dietary/supplementary VK2 intake has been associated with significant changes in insulin concentration and sensitivity in human studies, mostly focused on MK-4 and MK-7, administered at mg and µg dose levels, respectively, due to the observed difference in their bioavailability [96]. Choi et al. demonstrated that high MK-4 intake (30 mg/d) contributed to a significant increase in insulin sensitivity index in healthy young men (n = 18, 25.5–31.5 years), compared to the placebo group (n = 15, 24–31 years) over 4 weeks [97]. Similarly, a linear, inverse association between VK intake (average intakes: VK1 200 ± 98 μg/d, menaquinones 31 ± 7 μg/d) and a reduced risk of T2-D was also shown in a long and large study conducted by Beulens et al. [98].
Evidence of the beneficial effects of MK-4 and VK1 on glucose metabolism/homeostasis has also been provided by streptozotocin (STZ)-induced Type 1 diabetic rat models [99], while Khalil et al. demonstrated the protective function of MK-7 against impaired glucose homeostasis in ovariectomised exercised and nonexercised rats [100], with decreases in glucose levels and increases in insulin, lipocalcin-2 and adiponectin levels shown in both models. Similar findings were also observed in another rat study, whereby MK-4 and VK1 (600 mg/kg diet, 3-month) treatment significantly reduced fat accumulation and serum triglycerides by 48% (MK-4) and 29% (VK1), compared to the control group [101], suggesting a potential role of VK, especially VK2, in treatment of IR via its lipid-lowering effects [96].
The beneficial synergistic effects of vitamins D, K (90 µg MK-7) and Ca co-supplementation on maximum levels of left carotid intima-media thickness and metabolic status in T2-D patients with CHD was demonstrated by Asemi et al. over 12 weeks in a randomised, double-blind, placebo-controlled trial. Significant changes in insulin levels, homeostasis model assessment for insulin resistance (HOMA-IR) and HOMA-β-cell function (HOMB-B) were noted [102]. Conversely, a randomised, double-blind, placebo-controlled trial involving 68 T2-D patients (aged 30–70) did not show significant improvement in various IR-related indices following 12-week MK-7 intake (360 μg/d) [103]. In a recent case–control study (NCT04387019) involving 60 T2-D patients (30 each in controlled and uncontrolled groups) and 30 non-diabetic controls, serum VK2 level was found to be significantly lower, especially with uncontrolled hyperglycaemia, than in the control group. This indicates the possible relationship between VK2 level and glycaemic homeostasis in T2-D patients [104]. The clinical implementation of VK2 intervention for diabetes management was further suggested by another 6–month clinical trial (ChiCTR1800019663) with 60 T2-D participants and MK-7 intervention [105].
The connection between VK2 and the metabolic syndrome (MetS) has also been explored. MetS represents a combination of risk factors which increase the risk of development of diabetes and CVDs and includes abdominal obesity (waist circumference), moderate hypertension, low high-density lipoprotein (HDL) cholesterol, high glucose level and high triacylglycerol concentrations [106]. In a 10-year follow-up study including women (n = 402; 49–70 years) and men (n = 400; 40–80 years), Dam et al. showed cross-sectionally and longitudinally that high VK2 intake (210.3 ± 127.0 μg/d) was associated with a decreased occurrence of MetS (measured as lower triacylglycerol levels and waist circumference), but interestingly, no such correlation was observed in the VK1 treatment group (31.1 ± 12.5 μg/d) [107]. Conversely, a cross-sectional study with a 6-year follow-up involving 5800 adults (20–45 years) suggested that higher VK1 intake (84.8 ± 3.2 μg/d) may favourably influence the prevalence of MetS (measured as decreased high blood pressure and reduced HDL cholesterol) [108].
In the context of molecular mechanisms, studies provide insight into the involvement of OC in pancreatic β-cell proliferation, insulin expression, adiponectin expression in adipocytes, as well as glucose metabolism, suggesting beneficial effects of VK in diabetes via OC metabolism [96,109]. The association of higher cOC after VK2 intake with improved insulin sensitivity [97], lowered IR [110], decreased body mass and BMI [111] has repeatedly been established by human studies. However, other inconsistent results have been reported. Lee et al. reported a modulatory role of ucOC in regulating glucose metabolism and increasing β-cell proliferation and insulin secretion in a genetically modified mice model [112]. Iki et al. reported an inverse relationship between serum undercarboxylated OC levels and glycaemic status/IR in their study involving 2174 Japanese men [113]. Furthermore, the effect of VK in improving insulin sensitivity via inhibiting anti-inflammatory responses, through inactivation of NFκB signalling pathway and suppressions of IL-6, IL-1β, TNF-α expression, has also been reported [61,96]. More recently, SIRT1 signalling pathway in upmodulating mitochondrial function to protect against IR has been demonstrated [114].

3.6. Neurodegenerative Diseases

Neurodegenerative disorders comprise a wide spectrum of neurological disorders with variable clinical phenotypes, including behavioural and cognitive changes, and affect millions of people worldwide. Of these disorders, Alzheimer’s disease (AD) and Parkinson’s disease [86] are the two most common neurodegenerative diseases [115]. The risk of being affected by a neurodegenerative disease increases dramatically with age. Therefore, as the population ages, estimates of such diseases increase in parallel, with dementia expected to affect 81 million people by 2040 [115,116]. VK may serve to support neurological function through several mechanisms. The dependence of sphingolipid synthesis, and therefore myelination of neurons, on VK is well established. Suggested mechanisms of action of VK in preventing cognitive decline, and specifically AD, include inhibition of apoptotic signalling proteins and oxidative stress, including inhibition of p38 MAP kinase [117]. In our previous review, we discussed the potential pathways of the protective effect of VK2 on neuroprotection through modulating neurodegeneration, inflammation and oxidative stress with or without the involvement of VK-dependent proteins (Gas6, Protein S) [12].
The relationship of VK (mainly MK-4 and VK1) with cognitive and behavioural performance is supported by a body of evidence [118]. Studies on the nutritional status of community-dwelling elder patients with early-stage AD revealed that the patient group had significantly lower VK intake than those of age- and sex-matched cognitively intact control participants, even after adjusting for energy intakes [10,119]. On the other hand, in a longitudinal ageing study of 599 middle-aged adults (aged 55–65) with a 6-year follow-up, examining the association between VK status (by dp-ucMGP) and cognitive decline, no direct correlation was found [120]. Fewer studies exist in PD; one small case–control study including 93 PD patients suggested that deficiency of VK2 is associated with PD progression [121].
Some studies have focused on the impact of brain metabolism by VKAs used as anticoagulants (i.e., warfarin, acenocoumarol and fluindione). Ferland et al. demonstrated an association between VKA usage and significantly diminished performances in visual memory and verbal fluency in a large prospective study of 7133 non-demented community dwellers (aged ≥ 65), with a 10-year follow-up. However, no correlation was found between VKA treatment and MMSE scores [122]. In another longitudinal prospective study with a 24-month follow-up involving 378 geriatric outpatients (mean age 82.3 ± 5.6), the authors found those taking VKAs had more severe executive dysfunction (frontal assessment battery) at baseline and incident executive decline over 24 months. Again, no significant association with MMSE scores was seen [123]. The same research group performed another study of brain morphological changes in patients taking VKAs and observed focal atrophies in older adults exposed to VKAs (18 community-dwellers), compared to 36 matched controls [124]. Annweiler et al. further showed that fluindione treatment specifically caused more frequent cognitive impairment among 267 geriatric patients (56.9% female; mean age 83.4 ± 8.1 years) [125].
Trends have been shown between VKA usage and cognitive impairment in patients with atrial fibrillation, regardless of dependency or frailty [126], while the usage of non-VKA-based anticoagulants and a lower risk of cognitive impairment [127] has also been documented. In an attempt to define brain areas affected by VKAs, Tamadon-Nejad et al. used a rat study to show that warfarin-treated rats had a dramatic decrease in MK-4 levels in all brain regions and scored worse than controls in tests of cognition and behaviour, including longer latencies in the MWM test, lower locomotor activity and exploratory behavior, as well as having altered sphingolipid levels, notably those of the ganglioside family. These results suggested that the biosynthetic pathway of MK-4 is suppressed by warfarin, even in the presence of high local VK1 concentrations [128]. In a study involving 85 patients (≥75 years) on VKA treatment, a positive correlation was shown between VK1 concentration and cognitive status [129].
Apart from conditions of impaired cognition, other neurological disorders have been associated with VK. Multiple sclerosis [94] is a complex chronic inflammatory and degenerative disorder of the central nervous system. The immuno-regulatory role of Gas6 has been suggested to be associated with this autoimmune disease via Gas6/TAM systems [130]. In a study conducted by Lasemi et al., substantially lower VK2 levels were detected in MS patients (female: 31 and male: 14) than controls (19 female, 10 male), with VK2 levels in female patients significantly lower than those of males [131]. In an in vivo rat model of anxiety and depression, it was suggested that VK2 might have a regulatory effect on preventing their development, possibly via its effects on blood glucose [132]. Peripheral neuropathy is a frequent and severe complication in people with diabetes. In a study of 198 T2-D patients (mean age 64 ± 8.4), 15.7% of patients with peripheral neuropathy had significantly lower levels of dp-ucMGP, suggesting this fully inactive MGP form could be a biomarker of sensitive neuropathy [133]. The therapeutic effects of MK-7 in relieving peripheral neuropathy symptoms have been shown in a trial of 100 patients (aged 18–65) with peripheral neuropathy (suffering from T2-D or vitamin B12 deficiency), who received 8 weeks of treatment with 200 mg MK-7 daily. The treatment resulted in significant improvements in symptom intensity and was well tolerated by all patients [134]. Surprisingly, one study (ACTRN12615000750583 and ACTRN12617000640303) involving 1347 community-dwelling older women (≥70 years) showed that high VK1 intakes was associated with better physical function and reduced long-term risk of injurious falls, potentially due to improved neuromuscular coordination and vascular function, but no such beneficial relationship was observed with VK2 supplementation [135].

3.7. Cancer

The association of VK with cancer can be considered through the lens of chemoprevention, or as a treatment strategy, either alone or as adjuvant chemotherapy. VK has been reported to have an inhibitory role in cancer cell growth, inducing apoptosis and cell-cycle arrest in many cancer lineages, including reproductive, haematological and gastrointestinal lines. Mechanistically, besides the induction of cell-cycle arrest, cell differentiation and apoptosis, the anti-cancer effects of VK2 through autophagy and the suppression of cancer cell invasion have been proposed [136]. In cultured leukaemia cells, VK2 induced autophagy and apoptosis simultaneously [137]. While induced autophagy by VK2 was significant in cholangiocellular carcinoma cells, apoptosis/cell-cycle arrest was inconspicuous [138]. Clinically, accounts of remission in several, diverse cancers have been reported, with translation into improved prognosis in several trials [136,139]. However, the potential of VK as a possible chemopreventive is clouded by contradictory observations.
A significant inverse association between dietary VK2 intake and the risk of prostate and lung cancer incidence was identified in the Heidelberg cohort of the European Prospective Investigation into Cancer and Nutrition study, involving 24,340 participants (aged 35–64). The authors further reported that no such association was observed with VK1 intake [140]. Dasari et al. have demonstrated VK2′s effect on castration-resistant prostate cancer by inducing VCap cell death through specifically targeting ROS-mediated apoptosis and cell-cycle progression as well as metastasis-inhibiting signalling molecules [141]. In breast cancer, while some in vitro studies show promise, such as the demonstration that VK2 can induce non-apoptotic cell death and autophagy in triple negative breast cancer cell lines [142], clinical studies may show conflicting results. For example, a positive relationship between the risk of occurrence and death of breast cancer and total VK (VK1 and VK2) intake was shown in a prospective cohort (US) study involving 51,662 women [143].
Myelodysplastic syndrome (MDS) refers to a category of clonal haematological disorders characterised by dysplastic features of bone marrow cells, ineffective haematopoiesis and cytopenia and carries over 30% risk of progression to acute myeloid leukaemia (AML). A questionnaire survey of multi-centre pilot studies in Japan demonstrated haematological improvement in some MDS/post-MDS AML patients following VK2 (as MK-4, in doses ranging from 20 to 135 mg/day p.o. and 10 to 50 mg/day IV MK-4) administration over 2 years [144]. The efficacy of VK2 treatment was observed in around 30% of patients in these studies, and no adverse effects were noted. These findings correlated with previous case reports, including a female MDS patient (aged 80) treated with MK-4 at 45 mg/d [145] and a female relapsing acute promyelocytic leukaemia patient, who received combination treatment with VK2 (20 mg/d) and all-trans retinoic acid [146]. In vitro evidence suggests dichotomous effects of VK2 in leukaemia cells [147] and that it causes suppression of cMYC [148] while improving haematopoiesis and promoting anti-apoptosis of normal erythroid progenitors and differentiation of myeloid progenitors [149]. The pro-apoptotic effects of co-culture of bone marrow mesenchymal stromal/stem cells (BM-MSCs) with MDS-derived cells were enhanced by MK-4 [150]. VK2 may thus offer potential as an adjunctive anti-cancer therapy in this cohort, especially for elderly patients who cannot tolerate intensive chemotherapy and stem cell transplantation.
Other mechanisms of VK2-induced leukemic cell apoptosis and differentiation include upregulation of p27 protein expression [147], disruption of mitochondrial membrane potential by VK2-Bcl antagonist killer 1 (Bak) binding [151], cleavage of PARP and down-regulation of cyclin A2 expression [152]. VK2-initiated mitochondrial apoptosis and ROS generation pathways were also observed in cultured studies of myeloma cells [153] and human ovarian cancer TYK-nu cells [154]. Promotion of HL-60 leukaemia cell differentiation was shown with a cotylenin A/VK2 combination [148], while VK2 promoted greater induction of monocytic differentiation as compared with that by vitamin D3 alone in leukaemia cells [155].
VK2 (as MK-4) appeared to have a positive effect in preventing the development of hepatocellular carcinoma (HCC), the third leading cause of cancer mortality, in patients with type-C cirrhosis, when administered at a dose of 45 mg/d [156]. A suppressive effect of VK2 on HCC recurrence rate and a beneficial effect on survival [157], as well as HCC recurrence derived from hepatitis C viral infection [158], has also been reported, but the results from these studies were not statistically significant. In a prospective randomised controlled trial of 101 HCC post-hepatectomy patients, MK-4 treatment (45 mg/d) exhibited a moderately suppressive effect on HCC recurrence [159]. Conversely, in another double-blind, randomised, placebo-controlled trial of 548 patients, VK2, at either 45 or 90 mg/d, did not prevent HCC recurrence or death [160]. Potential limitations associated with this study, as suggested in a later review, included patient recruitment and MK-4 quality [136]. Additionally, an interactive effect of VK2 combined with vitamin E treatment was suggested by a case study of a male HCC patient (age 65), whereby observation of tumour growth suppression and intraperitoneal dissemination disappearance was noted [161]. A combination of VK2 with other therapeutic or investigational agents has been shown. Using in vitro models, enhancement of the inhibition of HCC cell proliferation with the combination of 5-fluorouracil and VK2 was shown [162], while cell-cycle arrest and apoptosis was demonstrated with sorafenib/VK2 [163]. A recent study in a clinic involving 44 HCC patients further demonstrated the efficacy and safety of VK2 and sorafenib combination treatment, where significantly higher objective response rates and extended median time of progression-free survival were observed in the combination treatment group compared to that of sorafenib alone [164].
In a prospective study of over 100,000 US adults, with a mean follow-up of over 8 years, dietary intake of VK1, but not VK2, appeared to confer a lower risk of pancreatic cancer [165]. In addition, Duan et al. studied the effect of VK2 on human bladder cancer cells, noting the induction of mitochondria-related cell apoptosis via ROS and JNK/p38 MAPK pathways [166] and promoted AMPK-dependent autophagic cell death via PI3K/AKT/HIF-1α-mediated glycolysis [167].

4. Conclusions

In summary, there is a growing body of evidence that VK2 could play an important pharmacological role in treating multiple disease conditions, including osteoporosis, osteoarthritis and rheumatoid arthritis, cardiovascular diseases, chronic kidney diseases, diabetes, neurodegenerative disorders and cancers. However, results from some clinical studies are still inconsistent. More in-depth investigations in VK2 research, including of metabolic processes, validation of biomarkers for VK2 status, dose–response and bioavailability, safety data, etc.) are still warranted. The establishment of a Recommended Daily Intake for VK2 is also needed, as this could have an important impact in improving global health.

Author Contributions

Writing—original draft, writing—review and editing, visualisation, investigation, data curation, formal analysis, conceptualisation, T.Z.; writing—review and editing, C.O.; writing—review and editing, H.S.; writing—review and editing, methodology, investigation, data curation, formal analysis, J.W.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Dam, H. The Antihaemorrhagic Vitamin of The Chick. Biochem. J. 1935, 29, 1273–1285. [Google Scholar] [CrossRef]
  2. Zhao, C.; Wan, Y.; Tang, G.; Jin, Q.; Zhang, H.; Xu, Z. Comparison of Different Fermentation Processes for the Vitamin K2 (Menaquinone-7) Production by a Novel Bacillus Velezensis ND Strain. Process Biochemi. 2021, 102, 33–41. [Google Scholar] [CrossRef]
  3. Halder, M.; Petsophonsakul, P.; Akbulut, A.C.; Pavlic, A.; Bohan, F.; Anderson, E.; Maresz, K.; Kramann, R.; Schurgers, L. Vitamin K: Double Bonds beyond Coagulation Insights into Differences between Vitamin K1 and K2 in Health and Disease. Int. J. Mol. Sci. 2019, 20, 896. [Google Scholar] [CrossRef] [PubMed]
  4. Lal, N.; Berenjian, A. Cis and Trans Isomers of The Vitamin Menaquinone-7: Which One is Biologically Significant? Appl. Microbiol. Biotechnol. 2020, 104, 2765–2776. [Google Scholar] [CrossRef]
  5. Shearer, M.J.; Newman, P. Metabolism and Cell Biology of Vitamin K. Thromb. Haemost. 2008, 100, 530–547. [Google Scholar]
  6. Booth, S.L. Vitamin K: Food Composition and Dietary Intakes. Food Nutr. Res. 2012, 56, 5505. [Google Scholar] [CrossRef] [PubMed]
  7. Marles, R.J.; Roe, A.L.; Oketch-Rabah, H.A. US Pharmacopeial Convention Safety Evaluation of Menaquinone-7, a Form of Vitamin K. Nutr. Rev. 2017, 75, 553–578. [Google Scholar] [CrossRef]
  8. Vermeer, C.; Raes, J.; van ‘t Hoofd, C.; Knapen, M.H.J.; Xanthoulea, S. Menaquinone Content of Cheese. Nutrients 2018, 10, 446. [Google Scholar] [CrossRef]
  9. Akbulut, A.C.; Pavlic, A.; Petsophonsakul, P.; Halder, M.; Maresz, K.; Kramann, R.; Schurgers, L. Vitamin K2 Needs an RDI Separate from Vitamin K1. Nutrients 2020, 12, 1852. [Google Scholar] [CrossRef]
  10. Simes, D.C.; Viegas, C.S.B.; Araújo, N.; Marreiros, C. Vitamin K as a Diet Supplement with Impact in Human Health: Current Evidence in Age-Related Diseases. Nutrients 2020, 12, 138. [Google Scholar] [CrossRef]
  11. Komai, M.; Shirakawa, H. Vitamin K Metabolism. Menaquinone-4 (MK-4) Formation from Ingested VK Analogues and Its Potent Relation to Bone Function. Clin. Calcium 2007, 17, 1663–1672. [Google Scholar]
  12. Yan, Q.; Zhang, T.; O’Connor, C.; Barlow, J.W.; Walsh, J.; Scalabrino, G.; Xu, F.; Sheridan, H. The Biological Responses of Vitamin K2: A Comprehensive Review. Food Sci. Nutr. 2023, 11, 1634–1656. [Google Scholar] [CrossRef] [PubMed]
  13. Mladěnka, P.; Macáková, K.; Kujovská Krčmová, L.; Javorská, L.; Mrštná, K.; Carazo, A.; Protti, M.; Remião, F.; Nováková, L. Vitamin K—Sources, Physiological Role, Kinetics, Deficiency, Detection, Therapeutic Use, and Toxicity. Nutr. Rev. 2022, 80, 677–698. [Google Scholar] [CrossRef]
  14. Liu, Y.; van Bennekom, E.O.; Zhang, Y.; Abee, T.; Smid, E.J. Long-Chain Vitamin K2 Production in Lactococcus Lactis Is Influenced by Temperature, Carbon Source, Aeration and Mode Of Energy Metabolism. Microb. Cell Factories 2019, 18, 129. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, X.; Qiao, K.; Wu, H.; Zhang, Y. The Impact of Food Additives on the Abundance and Composition of Gut Microbiota. Molecules 2023, 28, 631. [Google Scholar] [CrossRef] [PubMed]
  16. Gama, J.; Neves, B.; Pereira, A. Chronic Effects of Dietary Pesticides on the Gut Microbiome and Neurodevelopment. Front. Microbiol. 2022, 13, 931440. [Google Scholar] [CrossRef] [PubMed]
  17. Fenn, K.; Strandwitz, P.; Stewart, E.J.; Dimise, E.; Rubin, S.; Gurubacharya, S.; Clardy, J.; Lewis, K. Quinones are growth factors for the human gut microbiota. Microbiome 2017, 5, 161. [Google Scholar] [CrossRef] [PubMed]
  18. Dai, L.; Mafra, D.; Shiels, P.G.; Hackeng, T.M.; Stenvinkel, P.; Schurgers, L.J. Vitamin K and Hallmarks of Ageing: Focus on Diet and Gut Microbiome. Nutrients 2023, 15, 2727. [Google Scholar] [CrossRef] [PubMed]
  19. Ellis, J.L.; Karl, J.P.; Oliverio, A.M.; Fu, X.; Soares, J.W.; Wolfe, B.E.; Hernandez, C.J.; Mason, J.B.; Booth, S.L. Dietary Vitamin K is Remodeled by Gut Microbiota and Influences Community Composition. Gut Microbes 2021, 13, 1–16. [Google Scholar] [CrossRef]
  20. Ebid, A.I.; Abdeen, H.A.; Muhammed Maher, R.; Mohamed-Abdel-Motaleb, S.M. Cefoperazone-Sulbactam-Induced Coagulopathy in Critically Ill Egyptian Patients: Role of Vitamin K Prophylactic Doses. Hospital Pharmacy 2014. [Google Scholar] [CrossRef]
  21. Kaesler, N.; Schreibing, F.; Speer, T.; Puente-Secades, S.d.l.; Rapp, N.; Drechsler, C.; Kabgani, N.; Kuppe, C.; Boor, P.; Jankowski, V.; et al. Altered Vitamin K Biodistribution and Metabolism in Experimental and Human Chronic Kidney Disease. Kidney Int. 2022, 101, 338–348. [Google Scholar] [CrossRef]
  22. Simes, D.C.; Viegas, C.S.B.; Araújo, N.; Marreiros, C. Vitamin K as a Powerful Micronutrient in Aging and Age-Related Diseases: Pros and Cons from Clinical Studies. Int. J. Mol. Sci. 2019, 20, 4150. [Google Scholar] [CrossRef]
  23. Kozioł-Kozakowska, A.; Maresz, K. The Impact of Vitamin K2 (Menaquionones) in Children’s Health and Diseases: A Review of the Literature. Children 2022, 9, 78. [Google Scholar] [CrossRef]
  24. Holden, R.M.; Booth, S.L.; Tuttle, A.; James, P.D.; Morton, A.R.; Hopman, W.M.; Nolan, R.L.; Garland, J.S. Sequence Variation in Vitamin K Epoxide Reductase Gene Is Associated with Survival and Progressive Coronary Calcification In Chronic Kidney Disease. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1591–1596. [Google Scholar] [CrossRef]
  25. Haraikawa, M.; Tsugawa, N.; Sogabe, N.; Tanabe, R.; Kawamura, Y.; Okano, T.; Hosoi, T.; Goseki-Sone, M. Effects of Gamma-Glutamyl Carboxylase Gene Polymorphism (R325Q) on The association between Dietary Vitamin K Intake and Gamma-Carboxylation of Osteocalcin in Young Adults. Asia Pac. J. Clin. Nutr. 2013, 22, 646–654. [Google Scholar] [CrossRef]
  26. Theuwissen, E.; Teunissen, K.J.; Spronk, H.M.; Hamulyák, K.; Ten Cate, H.; Shearer, M.J.; Vermeer, C.; Schurgers, L.J. Effect of Low-Dose Supplements of Menaquinone-7 (vitamin K2) on The Stability of Oral Anticoagulant Treatment: Dose-Response Relationship in Healthy Volunteers. J. Thromb. Haemost. 2013, 11, 1085–1092. [Google Scholar] [CrossRef]
  27. Vroonhof, K.; van Rijn, H.J.; van Hattum, J. Vitamin K Deficiency and Bleeding after Long-Term Use of Cholestyramine. Neth. J. Med. 2003, 61, 19–21. [Google Scholar]
  28. Takagi, K.; Masuda, K.; Yamazaki, M.; Kiyohara, C.; Itoh, S.; Wasaki, M.; Inoue, H. Metal Ion and Vitamin Adsorption Profiles of Phosphate Binder Ion-Exchange Resins. Clin. Nephrol. 2010, 73, 30–35. [Google Scholar] [CrossRef]
  29. McDuffie, J.R.; Calis, K.A.; Booth, S.L.; Uwaifo, G.I.; Yanovski, J.A. Effects of Orlistat on Fat-Soluble Vitamins in Obese Adolescents. Pharmacotherapy 2002, 22, 814–822. [Google Scholar] [CrossRef]
  30. Harshman, S.G.; Shea, M.K.; Fu, X.; Grusak, M.A.; Smith, D.; Lamon-Fava, S.; Kuliopulos, A.; Greenberg, A.; Booth, S.L. Atorvastatin Decreases Renal Menaquinone-4 Formation in C57BL/6 Male Mice. J. Nutr. 2019, 149, 416–421. [Google Scholar] [CrossRef]
  31. Okuyama, H.; Langsjoen, P.H.; Hamazaki, T.; Ogushi, Y.; Hama, R.; Kobayashi, T.; Uchino, H. Statins Stimulate Atherosclerosis and Heart Failure: Pharmacological Mechanisms. Expert Rev. Clin. Pharmacol. 2015, 8, 189–199. [Google Scholar] [CrossRef] [PubMed]
  32. Neradova, A.; Schumacher, S.P.; Hubeek, I.; Lux, P.; Schurgers, L.J.; Vervloet, M.G. Phosphate Binders Affect Vitamin K Concentration by Undesired Binding, an in vitro Study. BMC Nephrol. 2017, 18, 149. [Google Scholar] [CrossRef]
  33. Jansz, T.T.; Neradova, A.; van Ballegooijen, A.J.; Verhaar, M.C.; Vervloet, M.G.; Schurgers, L.J.; van Jaarsveld, B.C. The Role of Kidney Transplantation and Phosphate Binder Use in Vitamin K Status. PLoS ONE 2018, 13, e0203157. [Google Scholar] [CrossRef]
  34. Bover, J.; Navarro-González, J.F.; daSilva, I. New Information on Phosphate Binder Interactions with Vitamin K. Nefrologia 2020, 40, 369–370. [Google Scholar] [CrossRef]
  35. Pasco, J.A.; Seeman, E.; Henry, M.J.; Merriman, E.N.; Nicholson, G.C.; Kotowicz, M.A. The Population Burden of Fractures Originates in Women with Osteopenia, not Osteoporosis. Osteoporos. Int. 2006, 17, 1404–1409. [Google Scholar] [CrossRef]
  36. Czeczuk, A.; Huk-Wieliczuk, E.; Michalska, A.; Bylina, D.; Sołtan, J.; Zofia, D. The Effect of Menopause on Bone Tissue in Former Swimmers and in Non-Athletes. Adv. Clin. Exp. Med. 2012, 21, 645–652. [Google Scholar]
  37. Ayub, N.; Faraj, M.; Ghatan, S.; Reijers, J.A.A.; Napoli, N.; Oei, L. The Treatment Gap in Osteoporosis. J. Clin. Med. 2021, 10, 3002. [Google Scholar] [CrossRef]
  38. Bolland, M.J.; Avenell, A.; Baron, J.A.; Grey, A.; MacLennan, G.S.; Gamble, G.D.; Reid, I.R. Effect of Calcium Supplements on Risk of Myocardial Infarction and Cardiovascular Events: Meta-Analysis. Bmj 2010, 341, c3691. [Google Scholar] [CrossRef]
  39. Fujita, Y.; Iki, M.; Tamaki, J.; Kouda, K.; Yura, A.; Kadowaki, E.; Sato, Y.; Moon, J.S.; Tomioka, K.; Okamoto, N.; et al. Association between Vitamin K Intake from Fermented Soybeans, Natto, and Bone Mineral Density in Elderly Japanese Men: The Fujiwara-kyo Osteoporosis Risk in Men (FORMEN) study. Osteoporos. Int. 2012, 23, 705–714. [Google Scholar] [CrossRef]
  40. van Ballegooijen, A.J.; Pilz, S.; Tomaschitz, A.; Grübler, M.R.; Verheyen, N. The Synergistic Interplay between Vitamins D and K for Bone and Cardiovascular Health: A Narrative Review. Int. J. Endocrinol. 2017, 2017, 7454376. [Google Scholar] [CrossRef] [PubMed]
  41. Fang, Y.; Hu, C.; Tao, X.; Wan, Y.; Tao, F. Effect of Vitamin K on Bone Mineral Density: A Meta-Analysis of Randomized Controlled Trials. J. Bone Miner. Metab. 2012, 30, 60–68. [Google Scholar] [CrossRef]
  42. Capozzi, A.; Scambia, G.; Lello, S. Calcium, vitamin D, vitamin K2, and magnesium supplementation and skeletal health. Maturitas 2020, 140, 55–63. [Google Scholar] [CrossRef] [PubMed]
  43. Huang, Z.B.; Wan, S.L.; Lu, Y.J.; Ning, L.; Liu, C.; Fan, S.W. Does Vitamin K2 Play a Role in The Prevention and Treatment of Osteoporosis for Postmenopausal Women: A Meta-Analysis of Randomized Controlled Trials. Osteoporos. Int. 2015, 26, 1175–1186. [Google Scholar] [CrossRef] [PubMed]
  44. Jaghsi, S.; Hammoud, T.; Haddad, S. Relation between Circulating Vitamin K1 and Osteoporosis in the Lumbar Spine in Syrian Post-Menopausal Women. Open Rheumatol. J. 2018, 12, 1–9. [Google Scholar] [CrossRef]
  45. Salma; Ahmad, S.S.; Karim, S.; Ibrahim, I.M.; Alkreathy, H.M.; Alsieni, M.; Khan, M.A. Effect of Vitamin K on Bone Mineral Density and Fracture Risk in Adults: Systematic Review and Meta-Analysis. Biomedicines 2022, 10, 1048. [Google Scholar] [CrossRef] [PubMed]
  46. Inaba, N.; Sato, T.; Yamashita, T. Low-Dose Daily Intake of Vitamin K(2) (Menaquinone-7) Improves Osteocalcin γ-Carboxylation: A Double-Blind, Randomized Controlled Trials. J. Nutr. Sci. Vitaminol. 2015, 61, 471–480. [Google Scholar] [CrossRef]
  47. Brugè, F.; Bacchetti, T.; Principi, F.; Littarru, G.P.; Tiano, L. Olive Oil Supplemented with Menaquinone-7 Significantly Affects Osteocalcin Carboxylation. Br. J. Nutr. 2011, 106, 1058–1062. [Google Scholar] [CrossRef] [PubMed]
  48. Suzuki, K.; Tsuji, S.; Fukushima, Y.; Nakase, T.; Hamada, M.; Tomita, T.; Yoshikawa, H. Clinical Results of Alendronate Monotherapy and Combined Therapy with Menatetrenone (VitK2) in Postmenopausal RA Patients. Mod. Rheumatol. 2013, 23, 450–455. [Google Scholar] [CrossRef] [PubMed]
  49. Li, H.; Zhou, Q.; Bai, B.-L.; Weng, S.-J.; Wu, Z.-Y.; Xie, Z.-J.; Feng, Z.-H.; Cheng, L.; Boodhun, V.; Yang, L. Effects of Combined Human Parathyroid Hormone (1–34) and Menaquinone-4 Treatment on The Interface of Hydroxyapatite-Coated Titanium Implants in The Femur of Osteoporotic Rats. J. Bone Miner. Metab. 2018, 36, 691–699. [Google Scholar] [CrossRef]
  50. Bonaccorsi, G.; Piva, I.; Greco, P.; Cervellati, C. Oxidative Stress as a Possible Pathogenic Cofactor of Post-Menopausal Osteoporosis: Existing Evidence in Support of The Axis Oestrogen Deficiency-Redox Imbalance-Bone Loss. Indian J. Med. Res. 2018, 147, 341–351. [Google Scholar] [CrossRef]
  51. Suzuki, Y.; Nawata, H.; Soen, S.; Fujiwara, S.; Nakayama, H.; Tanaka, I.; Ozono, K.; Sagawa, A.; Takayanagi, R.; Tanaka, H.; et al. Guidelines on The Management and Treatment of Glucocorticoid-Induced Osteoporosis of The Japanese Society for Bone and Mineral Research: 2014 Update. J. Bone Miner. Metab. 2014, 32, 337–350. [Google Scholar] [CrossRef]
  52. Orimo, H.; Nakamura, T.; Hosoi, T.; Iki, M.; Uenishi, K.; Endo, N.; Ohta, H.; Shiraki, M.; Sugimoto, T.; Suzuki, T.; et al. Japanese 2011 Guidelines for Prevention and Treatment of Osteoporosis--Executive Summary. Arch. Osteoporos. 2012, 7, 3–20. [Google Scholar] [CrossRef] [PubMed]
  53. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA); Turck, D.; Bresson, J.-L.; Burlingame, B.; Dean, T.; Fairweather-Tait, S.; Heinonen, M.; Hirsch-Ernst, K.I.; Mangelsdorf, I.; McArdle, H.J.; et al. Dietary Reference Values for Vitamin K. EFSA J. 2017, 15, e04780. [Google Scholar] [CrossRef] [PubMed]
  54. Loeser, R.F.; Berenbaum, F.; Kloppenburg, M. Vitamin K and Osteoarthritis: Is There a Link? Ann. Rheum. Dis. 2021, 80, 547–549. [Google Scholar] [CrossRef] [PubMed]
  55. Oka, H.; Akune, T.; Muraki, S.; En-yo, Y.; Yoshida, M.; Saika, A.; Sasaki, S.; Nakamura, K.; Kawaguchi, H.; Yoshimura, N. Association of Low Dietary Vitamin K Intake with Radiographic Knee Osteoarthritis in The Japanese Elderly Population: Dietary Survey in a Population-Based Cohort of the ROAD Study. J. Orthop. Sci. 2009, 14, 687–692. [Google Scholar] [CrossRef]
  56. Neogi, T.; Booth, S.L.; Zhang, Y.Q.; Jacques, P.F.; Terkeltaub, R.; Aliabadi, P.; Felson, D.T. Low Vitamin K Status Is Associated with Osteoarthritis in The Hand and Knee. Arthritis Rheum. 2006, 54, 1255–1261. [Google Scholar] [CrossRef] [PubMed]
  57. Ebina, K.; Shi, K.; Hirao, M.; Kaneshiro, S.; Morimoto, T.; Koizumi, K.; Yoshikawa, H.; Hashimoto, J. Vitamin K2 Administration Is Associated with Decreased Disease Activity in Patients with Rheumatoid Arthritis. Mod. Rheumatol. 2013, 23, 1001–1007. [Google Scholar] [CrossRef] [PubMed]
  58. Xu, W.; Wu, H.; Tahara, K.; Chen, S.; Wang, X.; Tanaka, S.; Sugiyama, K.; Sawada, T.; Hirano, T. Effects of Vitamin K(2) Combined with Methotrexate Against Mitogen-Activated Peripheral Blood Mononuclear Cells of Healthy Subjects and Rheumatoid Arthritis Patients. Fundam. Clin. Pharmacol. 2021, 35, 832–842. [Google Scholar] [CrossRef] [PubMed]
  59. Abdel-Rahman, M.S.; Alkady, E.A.M.; Ahmed, S. Menaquinone-7 as a Novel Pharmacological Therapy in The Treatment of Rheumatoid Arthritis: A Clinical Study. Eur. J. Pharmacol. 2015, 761, 273–278. [Google Scholar] [CrossRef]
  60. Ohsaki, Y.; Shirakawa, H.; Hiwatashi, K.; Furukawa, Y.; Mizutani, T.; Komai, M. Vitamin K Suppresses Lipopolysaccharide-Induced Inflammation in the Rat. Biosci. Biotechnol. Biochem. 2006, 70, 926–932. [Google Scholar] [CrossRef]
  61. Ohsaki, Y.; Shirakawa, H.; Miura, A.; Giriwono, P.; Sato, S.; Ohashi, A.; Iribe, M.; Goto, T.; Komai, M. Vitamin K Suppresses the Lipopolysaccharide-Induced Expression of Inflammatory Cytokines in Cultured Macrophage-Like Cells via the Inhibition of the Activation of Nuclear Factor αB through the Repression of IKKα/β Phosphorylation. J. Nutr. Biochem. 2010, 21, 1120–1126. [Google Scholar] [CrossRef]
  62. Schwalfenberg, G.K. Vitamins K1 and K2: The Emerging Group of Vitamins Required for Human Health. J. Nutr. Metab. 2017, 2017, 6254836. [Google Scholar] [CrossRef]
  63. Gast, G.C.M.; de Roos, N.M.; Sluijs, I.; Bots, M.L.; Beulens, J.W.J.; Geleijnse, J.M.; Witteman, J.C.; Grobbee, D.E.; Peeters, P.H.M.; van der Schouw, Y.T. A High Menaquinone Intake Reduces The Incidence of Coronary Heart Disease. Nutr. Metab. Cardiovasc. Dis. 2009, 19, 504–510. [Google Scholar] [CrossRef]
  64. Geleijnse, J.M.; Vermeer, C.; Grobbee, D.E.; Schurgers, L.J.; Knapen, M.H.; van der Meer, I.M.; Hofman, A.; Witteman, J.C. Dietary Intake of Menaquinone Is Associated with a Reduced Risk of Coronary Heart Disease: The Rotterdam Study. J. Nutr. 2004, 134, 3100–3105. [Google Scholar] [CrossRef]
  65. Bellinge, J.W.; Dalgaard, F.; Murray, K.; Connolly, E.; Blekkenhorst, L.C.; Bondonno, C.P.; Lewis, J.R.; Sim, M.; Croft, K.D.; Gislason, G.; et al. Vitamin K Intake and Atherosclerotic Cardiovascular Disease in the Danish Diet Cancer and Health Study. J. Am. Heart Assoc. 2021, 10, e020551. [Google Scholar] [CrossRef]
  66. Puzantian, H.; Akers, S.R.; Oldland, G.; Javaid, K.; Miller, R.; Ge, Y.; Ansari, B.; Lee, J.; Suri, A.; Hasmath, Z.; et al. Circulating Dephospho-Uncarboxylated Matrix Gla-Protein Is Associated with Kidney Dysfunction and Arterial Stiffness. Am. J. Hypertens. 2018, 31, 988–994. [Google Scholar] [CrossRef]
  67. Cozzolino, M.; Mangano, M.; Stucchi, A.; Ciceri, P.; Conte, F.; Galassi, A. Cardiovascular Disease in Dialysis Patients. Nephrol. Dial. Transplant. 2018, 33, iii28–iii34. [Google Scholar] [CrossRef]
  68. Caluwé, R.; Vandecasteele, S.; Van Vlem, B.; Vermeer, C.; De Vriese, A.S. Vitamin K2 Supplementation in Haemodialysis Patients: A Randomized Dose-Finding Study. Nephrol. Dial. Transplant. 2014, 29, 1385–1390. [Google Scholar] [CrossRef]
  69. Caluwé, R. Effect on Vascular Calcification of Replacing Warfarin by Rivaroxaban with or without VitK2 in Hemodialysis Patients. Available online: https://clinicaltrials.gov/study/NCT02610933#study-record-dates (accessed on 3 December 2023).
  70. De Vriese, A.S.; Caluwé, R.; Pyfferoen, L.; De Bacquer, D.; De Boeck, K.; Delanote, J.; De Surgeloose, D.; Van Hoenacker, P.; Van Vlem, B.; Verbeke, F. Multicenter Randomized Controlled Trial of Vitamin K Antagonist Replacement by Rivaroxaban with or without Vitamin K2 in Hemodialysis Patients with Atrial Fibrillation: The Valkyrie Study. J. Am. Soc. Nephrol. 2020, 31, 186–196. [Google Scholar] [CrossRef]
  71. Eelderink, C.; Kremer, D.; Riphagen, I.J.; Knobbe, T.J.; Schurgers, L.J.; Pasch, A.; Mulder, D.J.; Corpeleijn, E.; Navis, G.; Bakker, S.J.L.; et al. Effect of Vitamin K Supplementation on Serum Calcification Propensity and Arterial Stiffness In Vitamin K-Deficient Kidney Transplant Recipients: A Double-Blind, Randomized, Placebo-Controlled Clinical Trial. Am. J. Transplant 2023, 23, 520–530. [Google Scholar] [CrossRef]
  72. Naiyarakseree, N.; Phannajit, J.; Naiyarakseree, W.; Mahatanan, N.; Asavapujanamanee, P.; Lekhyananda, S.; Vanichakarn, S.; Avihingsanon, Y.; Praditpornsilpa, K.; Eiam-Ong, S.; et al. Effect of Menaquinone-7 Supplementation on Arterial Stiffness in Chronic Hemodialysis Patients: A Multicenter Randomized Controlled Trial. Nutrients 2023, 15, 2422. [Google Scholar] [CrossRef] [PubMed]
  73. Meer, R.; Romero Prats, M.L.; Vervloet, M.G.; van der Schouw, Y.T.; de Jong, P.A.; Beulens, J.W.J. The Effect of Six-Month Oral Vitamin K Supplementation on Calcification Propensity Time in Individuals with Type 2 Diabetes Mellitus: A Post HOC Analysis of A randomised, Double-Blind, Placebo-Controlled Trial. Atherosclerosis 2023, 117307. [Google Scholar] [CrossRef] [PubMed]
  74. Diederichsen, A.C.P.; Lindholt, J.S.; Möller, S.; Øvrehus, K.A.; Auscher, S.; Lambrechtsen, J.; Hosbond, S.E.; Alan, D.H.; Urbonaviciene, G.; Becker, S.W.; et al. Vitamin K2 and D in Patients with Aortic Valve Calcification: A Randomized Double-Blinded Clinical Trial. Circulation 2022, 145, 1387–1397. [Google Scholar] [CrossRef] [PubMed]
  75. Peeters, F.; van Mourik, M.J.W.; Meex, S.J.R.; Bucerius, J.; Schalla, S.M.; Gerretsen, S.C.; Mihl, C.; Dweck, M.R.; Schurgers, L.J.; Wildberger, J.E.; et al. Bicuspid Aortic Valve Stenosis and the Effect of Vitamin K2 on Calcification Using (18)F-Sodium Fluoride Positron Emission Tomography/Magnetic Resonance: The BASIK2 Rationale and Trial Design. Nutrients 2018, 10, 386. [Google Scholar] [CrossRef] [PubMed]
  76. Center, M.U.M. The Effects of Vitamin K2 Supplementation on the Progression of Coronary Artery Calcification (VitaK-CAC). Available online: https://clinicaltrials.gov/study/NCT01002157 (accessed on 3 December 2023).
  77. Maastricht, A.Z. Effects of VitamIN K2 and D3 supplementaTion on PET/MRI in Carotid Artery Disease (INTRICATE). Available online: https://clinicaltrials.gov/ct2/show/NCT04010578 (accessed on 21 January 2023).
  78. Kampmann, F.B.; Thysen, S.M.; Nielsen, C.F.B.; Kofoed, K.F.; Køber, L.; Pham, M.H.C.; Vaag, A.; Jørgensen, N.R.; Petersen, J.; Jacobsen, R.K.; et al. Study Protocol of The Intervitamink Trial: A Danish Population-Based Randomised Double-Blinded Placebo-Controlled Trial of The Effects of Vitamin K (Menaquinone-7) Supplementation on Cardiovascular, Metabolic and Bone Health. BMJ Open 2023, 13, e071885. [Google Scholar] [CrossRef] [PubMed]
  79. Boxma, P.Y.; van den Berg, E.; Geleijnse, J.M.; Laverman, G.D.; Schurgers, L.J.; Vermeer, C.; Kema, I.P.; Muskiet, F.A.; Navis, G.; Bakker, S.J.; et al. Vitamin K Intake and Plasma Desphospho-Uncarboxylated Matrix Gla-Protein Levels in Kidney Transplant Recipients. PLoS ONE 2012, 7, e47991. [Google Scholar] [CrossRef] [PubMed]
  80. Roumeliotis, S.; Dounousi, E.; Eleftheriadis, T.; Liakopoulos, V. Association of The Inactive Circulating Matrix Gla Protein with Vitamin K Intake, Calcification, Mortality, and Cardiovascular Disease: A Review. Int. J. Mol. Sci. 2019, 20, 628. [Google Scholar] [CrossRef] [PubMed]
  81. Elliott, M.J.; Booth, S.L.; Hopman, W.M.; Holden, R.M. Assessment of Potential Biomarkers of Subclinical Vitamin K Deficiency in Patients with End-Stage Kidney Disease. Can. J. Kidney Health Dis. 2014, 1, 13. [Google Scholar] [CrossRef] [PubMed]
  82. Miyata, K.N.; Nast, C.C.; Dai, T.; Dukkipati, R.; LaPage, J.A.; Troost, J.P.; Schurgers, L.J.; Kretzler, M.; Adler, S.G. Renal Matrix Gla Protein Expression Increases Progressively with CKD and Predicts Renal Outcome. Exp. Mol. Pathol. 2018, 105, 120–129. [Google Scholar] [CrossRef]
  83. Schurgers, L.J.; Barreto, D.V.; Barreto, F.C.; Liabeuf, S.; Renard, C.; Magdeleyns, E.J.; Vermeer, C.; Choukroun, G.; Massy, Z.A. The Circulating Inactive Form of Matrix Gla Protein is a Surrogate Marker for Vascular Calcification in Chronic Kidney Disease: A Preliminary Report. Clin. J. Am. Soc. Nephrol. 2010, 5, 568–575. [Google Scholar] [CrossRef]
  84. Jaminon, A.M.G.; Dai, L.; Qureshi, A.R.; Evenepoel, P.; Ripsweden, J.; Söderberg, M.; Witasp, A.; Olauson, H.; Schurgers, L.J.; Stenvinkel, P. Matrix Gla Protein Is an Independent Predictor of Both Intimal and Medial Vascular Calcification in Chronic Kidney Disease. Sci. Rep. 2020, 10, 6586. [Google Scholar] [CrossRef] [PubMed]
  85. Kaesler, N.; Magdeleyns, E.; Herfs, M.; Schettgen, T.; Brandenburg, V.; Fliser, D.; Vermeer, C.; Floege, J.; Schlieper, G.; Krüger, T. Impaired Vitamin K Recycling in Uremia Is Rescued by Vitamin K Supplementation. Kidney Int. 2014, 86, 286–293. [Google Scholar] [CrossRef] [PubMed]
  86. Opdebeeck, B.; Maudsley, S.; Azmi, A.; De Maré, A.; De Leger, W.; Meijers, B.; Verhulst, A.; Evenepoel, P.; D’Haese, P.C.; Neven, E. Indoxyl Sulfate and p-Cresyl Sulfate Promote Vascular Calcification and Associate with Glucose Intolerance. J. Am. Soc. Nephrol. 2019, 30, 751–766. [Google Scholar] [CrossRef] [PubMed]
  87. Dalmeijer, G.W.; van der Schouw, Y.T.; Vermeer, C.; Magdeleyns, E.J.; Schurgers, L.J.; Beulens, J.W. Circulating Matrix Gla Protein is Associated with Coronary Artery Calcification and Vitamin K Status in Healthy Women. J. Nutr. Biochem. 2013, 24, 624–628. [Google Scholar] [CrossRef]
  88. Aoun, M.; Makki, M.; Azar, H.; Matta, H.; Chelala, D.N. High Dephosphorylated-Uncarboxylated MGP in Hemodialysis Patients: Risk Factors and Response to Vitamin K(2), a Pre-post Intervention Clinical Trial. BMC Nephrol. 2017, 18, 191. [Google Scholar] [CrossRef] [PubMed]
  89. Delanaye, P.; Dubois, B.E.; Lukas, P.; Peters, P.; Krzesinski, J.M.; Pottel, H.; Cavalier, E. Impact of Stopping Vitamin K Antagonist Therapy on Concentrations of Dephospho-Uncarboxylated Matrix Gla Protein. Clin. Chem. Lab. Med. 2015, 53, e191–e193. [Google Scholar] [CrossRef] [PubMed]
  90. Kurnatowska, I.; Grzelak, P.; Masajtis-Zagajewska, A.; Kaczmarska, M.; Stefańczyk, L.; Vermeer, C.; Maresz, K.; Nowicki, M. Plasma Desphospho-Uncarboxylated Matrix Gla Protein as a Marker of Kidney Damage and Cardiovascular Risk in Advanced Stage of Chronic Kidney Disease. Kidney Blood Press. Res. 2016, 41, 231–239. [Google Scholar] [CrossRef] [PubMed]
  91. Nigwekar, S.U.; Bloch, D.B.; Nazarian, R.M.; Vermeer, C.; Booth, S.L.; Xu, D.; Thadhani, R.I.; Malhotra, R. Vitamin K-Dependent Carboxylation of Matrix Gla Protein Influences the Risk of Calciphylaxis. J. Am. Soc. Nephrol. 2017, 28, 1717–1722. [Google Scholar] [CrossRef]
  92. Wei, F.-F.; Drummen, N.E.A.; Schutte, A.E.; Thijs, L.; Jacobs, L.; Petit, T.; Yang, W.-Y.; Smith, W.; Zhang, Z.-Y.; Gu, Y.-M.; et al. Vitamin K Dependent Protection of Renal Function in Multi-ethnic Population Studies. EBioMedicine 2016, 4, 162–169. [Google Scholar] [CrossRef]
  93. Wei, F.-F.; Trenson, S.; Monney, P.; Yang, W.-Y.; Pruijm, M.; Zhang, Z.-Y.; Bouatou, Y.; Huang, Q.-F.; Ponte, B.; Martin, P.-Y.; et al. Epidemiological and Histological Findings Implicate Matrix Gla Protein in Diastolic Left Ventricular Dysfunction. PLoS ONE 2018, 13, e0193967. [Google Scholar] [CrossRef]
  94. Witham, M.D.; Lees, J.S.; White, M.; Band, M.; Bell, S.; Chantler, D.J.; Ford, I.; Fulton, R.L.; Kennedy, G.; Littleford, R.C.; et al. Vitamin K Supplementation to Improve Vascular Stiffness in CKD: The K4Kidneys Randomized Controlled Trial. J. Am. Soc. Nephrol. 2020, 31, 2434–2445. [Google Scholar] [CrossRef] [PubMed]
  95. Geng, C.; Huang, L.; Pu, L.; Feng, Y. Effects of Vitamin K Supplementation on Vascular Calcification in Chronic Kidney Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Nutr. 2022, 9, 1001826. [Google Scholar] [CrossRef] [PubMed]
  96. Li, Y.; Chen, J.p.; Duan, L.; Li, S. Effect of Vitamin K2 on Type 2 Diabetes Mellitus: A Review. Diabetes Res. Clin. Pract. 2018, 136, 39–51. [Google Scholar] [CrossRef] [PubMed]
  97. Choi, H.J.; Yu, J.; Choi, H.; An, J.H.; Kim, S.W.; Park, K.S.; Jang, H.C.; Kim, S.Y.; Shin, C.S. Vitamin K2 Supplementation Improves Insulin Sensitivity via Osteocalcin Metabolism: A Placebo-Controlled Trial. Diabetes Care 2011, 34, e147. [Google Scholar] [CrossRef] [PubMed]
  98. Beulens, J.W.; van der A, D.L.; Grobbee, D.E.; Sluijs, I.; Spijkerman, A.M.; van der Schouw, Y.T. Dietary Phylloquinone and Menaquinones Intakes and Risk of Type 2 Diabetes. Diabetes Care 2010, 33, 1699–1705. [Google Scholar] [CrossRef] [PubMed]
  99. Varsha, M.K.; Thiagarajan, R.; Manikandan, R.; Dhanasekaran, G. Vitamin K1 Alleviates Streptozotocin-Induced Type 1 Diabetes by Mitigating Free Radical Stress, as Well as Inhibiting NF-kB Activation and iNOS Expression in Rat Pancreas. Nutrition 2015, 31, 214–222. [Google Scholar] [CrossRef] [PubMed]
  100. Khalil, A.; Youssef, G.A.; Al-Saeed, H.F. Protective Role of Vitamin K Against Impaired Glucose Homeostasis in Ovariectomized Exercised and Nonexercised Rats. Nat. Sci. 2014, 12, 1–9. [Google Scholar] [CrossRef]
  101. Sogabe, N.; Maruyama, R.; Baba, O.; Hosoi, T.; Goseki-Sone, M. Effects of Long-Term Vitamin K1 (Phylloquinone) or Vitamin K2 (Menaquinone-4) Supplementation on Body Composition and Serum Parameters in Rats. Bone 2011, 48, 1036–1042. [Google Scholar] [CrossRef] [PubMed]
  102. Asemi, Z.; Raygan, F.; Bahmani, F.; Rezavandi, Z.; Talari, H.R.; Rafiee, M.; Poladchang, S.; Darooghegi Mofrad, M.; Taheri, S.; Mohammadi, A.A.; et al. The Effects of Vitamin D, K and Calcium Co-Supplementation on Carotid Intima-Media Thickness and Metabolic Status in Overweight Type 2 Diabetic Patients with CHD. Br. J. Nutr. 2016, 116, 286–293. [Google Scholar] [CrossRef]
  103. Rahimi Sakak, F.; Moslehi, N.; Abdi, H.; Mirmiran, P. Effects of Vitamin K2 Supplementation on Atherogenic Status of Individuals with Type 2 Diabetes: A Randomized Controlled Trial. BMC Complement. Med. Ther. 2021, 21, 134. [Google Scholar] [CrossRef]
  104. Helmy, M.Y.; Elsaid, N.H.; Gwad, M.M.A. The Association of Vitamin K2 Level with the Glycaemic Status in Type 2 Diabetic Patients: A Case-Control Study. Indian J. Endocrinol. Metab. 2022, 26, 87–92. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, Y.; Liu, L.; Wei, C.; Wang, X.; Li, R.; Xu, X.; Zhang, Y.; Geng, G.; Dang, K.; Ming, Z.; et al. Vitamin K2 Supplementation Improves Impaired Glycemic Homeostasis and Insulin Sensitivity for Type 2 Diabetes through Gut Microbiome and Fecal Metabolites. BMC Med. 2023, 21, 174. [Google Scholar] [CrossRef] [PubMed]
  106. Andersen, C.J.; Fernandez, M.L. Dietary Strategies to Reduce Metabolic Syndrome. Rev. Endocr. Metab. Disord. 2013, 14, 241–254. [Google Scholar] [CrossRef] [PubMed]
  107. Dam, V.; Dalmeijer, G.W.; Vermeer, C.; Drummen, N.E.; Knapen, M.H.; van der Schouw, Y.T.; Beulens, J.W. Association between Vitamin K and the Metabolic Syndrome: A 10-Year Follow-Up Study in Adults. J. Clin. Endocrinol. Metab. 2015, 100, 2472–2479. [Google Scholar] [CrossRef] [PubMed]
  108. Pan, Y.; Jackson, R.T. Dietary Phylloquinone Intakes and Metabolic Syndrome in US Young Adults. J. Am. Coll. Nutr. 2009, 28, 369–379. [Google Scholar] [CrossRef] [PubMed]
  109. Rasekhi, H.; Karandish, M.; Jalali, M.T.; Mohammad-shahi, M.; Zarei, M.; Saki, A.; Shahbazian, H. The Effect of Vitamin K1 Supplementation on Sensitivity and Insulin Resistance via Osteocalcin in Prediabetic Women: A Double-Blind Randomized Controlled Clinical Trial. Eur. J. Clin. Nutr. 2015, 69, 891–895. [Google Scholar] [CrossRef] [PubMed]
  110. Shea, M.K.; Gundberg, C.M.; Meigs, J.B.; Dallal, G.E.; Saltzman, E.; Yoshida, M.; Jacques, P.F.; Booth, S.L. Gamma-Carboxylation of Osteocalcin and Insulin Resistance in Older Men and Women. Am. J. Clin. Nutr. 2009, 90, 1230–1235. [Google Scholar] [CrossRef]
  111. Knapen, M.H.; Schurgers, L.J.; Shearer, M.J.; Newman, P.; Theuwissen, E.; Vermeer, C. Association of Vitamin K Status with Adiponectin and Body Composition in Healthy Subjects: Uncarboxylated Osteocalcin Is Not Associated with Fat Mass and Body Weight. Br. J. Nutr. 2012, 108, 1017–1024. [Google Scholar] [CrossRef]
  112. Lee, N.K.; Sowa, H.; Hinoi, E.; Ferron, M.; Ahn, J.D.; Confavreux, C.; Dacquin, R.; Mee, P.J.; McKee, M.D.; Jung, D.Y.; et al. Endocrine Regulation of Energy Metabolism by The Skeleton. Cell 2007, 130, 456–469. [Google Scholar] [CrossRef]
  113. Iki, M.; Tamaki, J.; Fujita, Y.; Kouda, K.; Yura, A.; Kadowaki, E.; Sato, Y.; Moon, J.S.; Tomioka, K.; Okamoto, N.; et al. Serum Undercarboxylated Osteocalcin Levels are Inversely Associated with Glycemic Status and Insulin Resistance in an Elderly Japanese Male Population: Fujiwara-kyo Osteoporosis Risk in Men (FORMEN) Study. Osteoporos. Int. 2012, 23, 761–770. [Google Scholar] [CrossRef]
  114. Su, X.; Wang, W.; Fang, C.; Ni, C.; Zhou, J.; Wang, X.; Zhang, L.; Xu, X.; Cao, R.; Lang, H.; et al. Vitamin K2 Alleviates Insulin Resistance in Skeletal Muscle by Improving Mitochondrial Function Via SIRT1 Signaling. Antioxid. Redox Signal. 2021, 34, 99–117. [Google Scholar] [CrossRef]
  115. Han, Z.; Tian, R.; Ren, P.; Zhou, W.; Wang, P.; Luo, M.; Jin, S.; Jiang, Q. Parkinson’s Disease and Alzheimer’s Disease: A Mendelian Randomization Study. BMC Med. Genet. 2018, 19, 215. [Google Scholar] [CrossRef]
  116. Alisi, L.; Cao, R.; De Angelis, C.; Cafolla, A.; Caramia, F.; Cartocci, G.; Librando, A.; Fiorelli, M. The Relationships between Vitamin K and Cognition: A Review of Current Evidence. Front. Neurol. 2019, 10, 239. [Google Scholar] [CrossRef]
  117. Emekli-Alturfan, E.; Alturfan, A.A. The Emerging Relationship between Vitamin K and Neurodegenerative Diseases: A Review of Current Evidence. Mol. Biol. Rep. 2023, 50, 815–828. [Google Scholar] [CrossRef]
  118. Ferland, G. Vitamin K and the Nervous System: An Overview of its Actions. Adv. Nutr. 2012, 3, 204–212. [Google Scholar] [CrossRef]
  119. Presse, N.; Shatenstein, B.; Kergoat, M.J.; Ferland, G. Low Vitamin K Intakes in Community-Dwelling Elders at an Early Stage of Alzheimer’s Disease. J. Am. Diet Assoc. 2008, 108, 2095–2099. [Google Scholar] [CrossRef]
  120. van den Heuvel, E.G.H.M.; van Schoor, N.M.; Vermeer, C.; Zwijsen, R.M.L.; den Heijer, M.; Comijs, H.C. Vitamin K Status Is Not Associated with Cognitive Decline in Middle Aged Adults. J. Nutr. Health Aging 2015, 19, 908–912. [Google Scholar] [CrossRef]
  121. Yu, Y.X.; Yu, X.D.; Cheng, Q.Z.; Tang, L.; Shen, M.Q. The Association of Serum Vitamin K2 Levels with Parkinson’s Disease: From Basic Case-Control Study to Big Data Mining Analysis. Aging 2020, 12, 16410–16419. [Google Scholar] [CrossRef]
  122. Ferland, G.; Feart, C.; Presse, N.; Lorrain, S.; Bazin, F.; Helmer, C.; Berr, C.; Annweiler, C.; Rouaud, O.; Dartigues, J.F.; et al. Vitamin K Antagonists and Cognitive Function in Older Adults: The Three-City Cohort Study. J. Gerontol. A Biol. Sci. Med. Sci. 2016, 71, 1356–1362. [Google Scholar] [CrossRef]
  123. Brangier, A.; Ferland, G.; Rolland, Y.; Gautier, J.; Féart, C.; Annweiler, C. Vitamin K Antagonists and Cognitive Decline in Older Adults: A 24-Month Follow-Up. Nutrients 2018, 10, 666. [Google Scholar] [CrossRef]
  124. Brangier, A.; Celle, S.; Roche, F.; Beauchet, O.; Ferland, G.; Annweiler, C. Use of Vitamin K Antagonists and Brain Morphological Changes in Older Adults: An Exposed/Unexposed Voxel-Based Morphometric Study. Dement. Geriatr. Cogn. Disord. 2018, 45, 18–26. [Google Scholar] [CrossRef]
  125. Annweiler, C.; Ferland, G.; Barberger-Gateau, P.; Brangier, A.; Rolland, Y.; Beauchet, O. Vitamin K Antagonists and Cognitive Impairment: Results from a Cross-Sectional Pilot Study among Geriatric Patients. J. Gerontol. A Biol. Sci. Med. Sci. 2015, 70, 97–101. [Google Scholar] [CrossRef]
  126. Mostaza, J.M.; Jiménez, M.J.R.; Laiglesia, F.J.R.; Peromingo, J.A.D.; Robles, M.B.; Sierra, E.G.; Bilbao, A.S.; Suárez, C. Clinical Characteristics and Type Of Antithrombotic Treatment in A Spanish Cohort of Elderly Patients with Atrial Fibrillation According to Dependency, Frailty and Cognitive Impairment. J. Geriatr. Cardiol. 2018, 15, 268–274. [Google Scholar] [CrossRef]
  127. Zhang, C.; Gu, Z.C.; Shen, L.; Pan, M.M.; Yan, Y.D.; Pu, J.; Liu, X.Y.; Lin, H.W. Non-vitamin K Antagonist Oral Anticoagulants and Cognitive Impairment in Atrial Fibrillation: Insights from the Meta-Analysis of Over 90,000 Patients of Randomized Controlled Trials and Real-World Studies. Front. Aging Neurosci. 2018, 10, 258. [Google Scholar] [CrossRef]
  128. Tamadon-Nejad, S.; Ouliass, B.; Rochford, J.; Ferland, G. Vitamin K Deficiency Induced by Warfarin Is Associated with Cognitive and Behavioral Perturbations, and Alterations in Brain Sphingolipids in Rats. Front. Aging Neurosci. 2018, 10, 213. [Google Scholar] [CrossRef]
  129. Alisi, L.; Cafolla, C.; Gentili, A.; Tartaglione, S.; Curini, R.; Cafolla, A. Vitamin K Concentration and Cognitive Status in Elderly Patients on Anticoagulant Therapy: A Pilot Study. J. Aging Res. 2020, 2020, 9695324. [Google Scholar] [CrossRef]
  130. Bellan, M.; Pirisi, M.; Sainaghi, P.P. The Gas6/TAM System and Multiple Sclerosis. Int. J. Mol. Sci. 2016, 17, 1807. [Google Scholar] [CrossRef]
  131. Lasemi, R.; Kundi, M.; Moghadam, N.B.; Moshammer, H.; Hainfellner, J.A. Vitamin K2 in Multiple Sclerosis Patients. Wien. Klin. Wochenschr. 2018, 130, 307–313. [Google Scholar] [CrossRef]
  132. Gancheva, S.M.; Zhelyazkova-Savova, M.D. Vitamin K2 Improves Anxiety and Depression but not Cognition in Rats with Metabolic Syndrome: A Role of Blood Glucose? Folia Med. 2016, 58, 264–272. [Google Scholar] [CrossRef]
  133. Jeannin, A.C.; Salem, J.E.; Massy, Z.; Aubert, C.E.; Vermeer, C.; Amouyal, C.; Phan, F.; Halbron, M.; Funck-Brentano, C.; Hartemann, A.; et al. Inactive Matrix Gla Protein Plasma Levels are Associated with Peripheral Neuropathy in Type 2 Diabetes. PLoS ONE 2020, 15, e0229145. [Google Scholar] [CrossRef]
  134. Mehta, D.; Dound, Y.; Jadhav, S.; Bhave, A.; Devale, M.; Vaidya, A. A Novel Potential Role of Vitamin K2-7 in Relieving Peripheral Neuropathy. J. Pharmacol. Pharmacother. 2018, 9, 180. [Google Scholar] [CrossRef]
  135. Sim, M.; Smith, C.; Bondonno, N.P.; Radavelli-Bagatini, S.; Blekkenhorst, L.C.; Dalla Via, J.; McCormick, R.; Zhu, K.; Hodgson, J.M.; Prince, R.L.; et al. Higher Dietary Vitamin K Intake is Associated with Better Physical Function and Lower Long-Term Injurious Falls Risk in Community-Dwelling Older Women. J. Nutr. Health Aging 2023, 27, 38–45. [Google Scholar] [CrossRef]
  136. Xv, F.; Chen, J.; Duan, L.; Li, S. Research Progress on the Anticancer Effects of Vitamin K2. Oncol. Lett. 2018, 15, 8926–8934. [Google Scholar] [CrossRef]
  137. Yokoyama, T.; Miyazawa, K.; Naito, M.; Toyotake, J.; Tauchi, T.; Itoh, M.; Yuo, A.; Hayashi, Y.; Georgescu, M.M.; Kondo, Y.; et al. Vitamin K2 Induces Autophagy and Apoptosis Simultaneously in Leukemia Cells. Autophagy 2008, 4, 629–640. [Google Scholar] [CrossRef]
  138. Enomoto, M.; Tsuchida, A.; Miyazawa, K.; Yokoyama, T.; Kawakita, H.; Tokita, H.; Naito, M.; Itoh, M.; Ohyashiki, K.; Aoki, T. Vitamin K2-Induced Cell Growth Inhibition via Autophagy Formation in Cholangiocellular Carcinoma Cell Lines. Int. J. Mol. Med. 2007, 20, 801–808. [Google Scholar] [CrossRef]
  139. Ganbat, D.; Jugder, B.E.; Ganbat, L.; Tomoeda, M.; Dungubat, E.; Takahashi, Y.; Mori, I.; Shiomi, T.; Tomita, Y. The Efficacy of Vitamin K, A Member Of Naphthoquinones in the Treatment of Cancer: A Systematic Review and Meta-Analysis. Curr. Cancer Drug Targets 2021, 21, 495–513. [Google Scholar] [CrossRef]
  140. Nimptsch, K.; Rohrmann, S.; Kaaks, R.; Linseisen, J. Dietary Vitamin K Intake in Relation to Cancer Incidence and Mortality: Results from The Heidelberg Cohort of The European Prospective Investigation into Cancer and Nutrition (EPIC-Heidelberg). Am. J. Clin. Nutr. 2010, 91, 1348–1358. [Google Scholar] [CrossRef]
  141. Dasari, S.; Samy, A.L.P.A.; Kajdacsy-Balla, A.; Bosland, M.C.; Munirathinam, G. Vitamin K2, a Menaquinone Present in Dairy Products Targets Castration-Resistant Prostate Cancer Cell-Line by Activating Apoptosis Signaling. Food Chem. Toxicol. 2018, 115, 218–227. [Google Scholar] [CrossRef]
  142. Miyazawa, S.; Moriya, S.; Kokuba, H.; Hino, H.; Takano, N.; Miyazawa, K. Vitamin K(2) Induces Non-Apoptotic Cell Death Along with Autophagosome Formation in Breast Cancer Cell Lines. Breast Cancer 2020, 27, 225–235. [Google Scholar] [CrossRef]
  143. Wang, K.; Wu, Q.; Li, Z.; Reger, M.K.; Xiong, Y.; Zhong, G.; Li, Q.; Zhang, X.; Li, H.; Foukakis, T.; et al. Vitamin K Intake and Breast Cancer Incidence and Death: Results from A Prospective Cohort Study. Clin. Nutr. 2021, 40, 3370–3378. [Google Scholar] [CrossRef]
  144. Miyazawa, K.; Nishimaki, J.; Ohyashiki, K.; Enomoto, S.; Kuriya, S.; Fukuda, R.; Hotta, T.; Teramura, M.; Mizoguchi, H.; Uchiyama, T.; et al. Vitamin K2 Therapy for Myelodysplastic Syndromes (MDS) and Post-MDS Acute Myeloid Leukemia: Information through a Questionnaire Survey of Multi-Center Pilot Studies in Japan. Leukemia 2000, 14, 1156–1157. [Google Scholar] [CrossRef]
  145. Takami, A.; Nakao, S.; Ontachi, Y.; Yamauchi, H.; Matsuda, T. Successful Therapy of Myelodysplastic Syndrome with Menatetrenone, a Vitamin K2 Analog. Int. J. Hematol. 1999, 69, 24–26. [Google Scholar]
  146. Fujita, H.; Tomiyama, J.; Tanaka, T. Vitamin K2 Combined with All-Trans Retinoic Acid Induced Complete Remission of Relapsing Acute Promyelocytic Leukaemia. Br. J. Haematol. 1998, 103, 584–585. [Google Scholar] [CrossRef]
  147. Miyazawa, K.; Yaguchi, M.; Funato, K.; Gotoh, A.; Kawanishi, Y.; Nishizawa, Y.; Yuo, A.; Ohyashiki, K. Apoptosis/Differentiation-Inducing Effects of Vitamin K2 on HL-60 Cells: Dichotomous Nature of Vitamin K2 in Leukemia Cells. Leukemia 2001, 15, 1111–1117. [Google Scholar] [CrossRef]
  148. Maniwa, Y.; Kasukabe, T.; Kumakura, S. Vitamin K2 and Cotylenin A Synergistically Induce Monocytic Differentiation and Growth arrest Along with The Suppression of c-MYC Expression and Induction of Cyclin G2 Expression in Human Leukemia HL-60 Cells. Int. J. Oncol. 2015, 47, 473–480. [Google Scholar] [CrossRef]
  149. Sada, E.; Abe, Y.; Ohba, R.; Tachikawa, Y.; Nagasawa, E.; Shiratsuchi, M.; Takayanagi, R. Vitamin K2 Modulates Differentiation and Apoptosis of Both Myeloid and Erythroid Lineages. Eur. J. Haematol. 2010, 85, 538–548. [Google Scholar] [CrossRef]
  150. Fujishiro, A.; Iwasa, M.; Fujii, S.; Maekawa, T.; Andoh, A.; Tohyama, K.; Takaori-Kondo, A.; Miura, Y. Menatetrenone Facilitates Hematopoietic Cell Generation in a Manner That is Dependent on Human Bone Marrow Mesenchymal Stromal/Stem Cells. Int. J. Hematol. 2020, 112, 316–330. [Google Scholar] [CrossRef]
  151. Karasawa, S.; Azuma, M.; Kasama, T.; Sakamoto, S.; Kabe, Y.; Imai, T.; Yamaguchi, Y.; Miyazawa, K.; Handa, H. Vitamin K2 Covalently Binds to Bak and Induces Bak-Mediated Apoptosis. Mol. Pharmacol. 2013, 83, 613–620. [Google Scholar] [CrossRef]
  152. Xu, W.; Wu, H.; Chen, S.; Wang, X.; Tanaka, S.; Sugiyama, K.; Yamada, H.; Hirano, T. Cytotoxic Effects Of Vitamins K1, K2, and K3 Against Human T Lymphoblastoid Leukemia Cells through Apoptosis Induction and Cell Cycle Arrest. Chem. Biol. Drug Des. 2020, 96, 1134–1147. [Google Scholar] [CrossRef]
  153. Tsujioka, T.; Miura, Y.; Otsuki, T.; Nishimura, Y.; Hyodoh, F.; Wada, H.; Sugihara, T. The Mechanisms of Vitamin K2-Induced Apoptosis of Myeloma Cells. Haematologica 2006, 91, 613–619. [Google Scholar]
  154. Shibayama-Imazu, T.; Sonoda, I.; Sakairi, S.; Aiuchi, T.; Ann, W.W.; Nakajo, S.; Itabe, H.; Nakaya, K. Production of Superoxide and Dissipation of Mitochondrial Transmembrane Potential by Vitamin K2 Trigger Apoptosis in Human Ovarian Cancer TYK-nu cells. Apoptosis 2006, 11, 1535–1543. [Google Scholar] [CrossRef]
  155. Iguchi, T.; Miyazawa, K.; Asada, M.; Gotoh, A.; Mizutani, S.; Ohyashiki, K. Combined Treatment of Leukemia Cells with Vitamin K2 and 1alpha,25-Dihydroxy Vitamin D3 Enhances Monocytic Differentiation Along with Becoming Resistant to Apoptosis by Induction of Cytoplasmic p21CIP1. Int. J. Oncol. 2005, 27, 893–900. [Google Scholar]
  156. Kojima, K.; Tamano, M.; Akima, T.; Hashimoto, T.; Kuniyoshi, T.; Maeda, C.; Majima, Y.; Kusano, K.; Murohisa, T.; Iijima, M.; et al. Effect of Vitamin K2 on The Development of Hepatocellular Carcinoma in Type C Cirrhosis. Hepatogastroenterology 2010, 57, 1264–1267. [Google Scholar]
  157. Mizuta, T.; Ozaki, I.; Eguchi, Y.; Yasutake, T.; Kawazoe, S.; Fujimoto, K.; Yamamoto, K. The Effect of Menatetrenone, a Vitamin K2 Analog, on Disease Recurrence and Survival In Patients with Hepatocellular Carcinoma after Curative Treatment: A Pilot Study. Cancer 2006, 106, 867–872. [Google Scholar] [CrossRef]
  158. Kakizaki, S.; Sohara, N.; Sato, K.; Suzuki, H.; Yanagisawa, M.; Nakajima, H.; Takagi, H.; Naganuma, A.; Otsuka, T.; Takahashi, H.; et al. Preventive Effects of Vitamin K on Recurrent Disease in Patients with Hepatocellular Carcinoma Arising from Hepatitis C Viral Infection. J. Gastroenterol. Hepatol. 2007, 22, 518–522. [Google Scholar] [CrossRef]
  159. Ishizuka, M.; Kubota, K.; Shimoda, M.; Kita, J.; Kato, M.; Park, K.H.; Shiraki, T. Effect of Menatetrenone, a Vitamin K2 Analog, on Recurrence of Hepatocellular Carcinoma after Surgical Resection: A Prospective Randomized Controlled Trial. Anticancer Res. 2012, 32, 5415–5420. [Google Scholar]
  160. Yoshida, H.; Shiratori, Y.; Kudo, M.; Shiina, S.; Mizuta, T.; Kojiro, M.; Yamamoto, K.; Koike, Y.; Saito, K.; Koyanagi, N.; et al. Effect of Vitamin K2 on The Recurrence of Hepatocellular Carcinoma. Hepatology 2011, 54, 532–540. [Google Scholar] [CrossRef]
  161. Otsuka, T.; Hagiwara, S.; Tojima, H.; Yoshida, H.; Takahashi, T.; Nagasaka, K.; Tomioka, S.; Ando, T.; Takeuchi, K.; Kori, T.; et al. Hepatocellular Carcinoma with Peritoneal Dissemination Which Was Regressed During Vitamin K2 and Vitamin E Administration. Intern. Med. 2007, 46, 711–715. [Google Scholar] [CrossRef]
  162. Zhang, H.; Ozaki, I.; Hamajima, H.; Iwane, S.; Takahashi, H.; Kawaguchi, Y.; Eguchi, Y.; Yamamoto, K.; Mizuta, T. Vitamin K2 Augments 5-Fluorouracil-Induced Growth Inhibition of Human Hepatocellular Carcinoma Cells by Inhibiting NF-κB Activation. Oncol. Rep. 2011, 25, 159–166. [Google Scholar]
  163. Zhang, Y.; Zhang, B.; Zhang, A.; Zhao, Y.; Zhao, J.; Liu, J.; Gao, J.; Fang, D.; Rao, Z. Synergistic Growth Inhibition by Sorafenib and Vitamin K2 in Human Hepatocellular Carcinoma Cells. Clinics 2012, 67, 1093–1099. [Google Scholar] [CrossRef]
  164. Haruna, Y.; Yakushijin, T.; Kawamoto, S. Efficacy and Safety of Sorafenib Plus Vitamin K Treatment for Hepatocellular Carcinoma: A Phase II, Randomized Study. Cancer Med. 2021, 10, 914–922. [Google Scholar] [CrossRef] [PubMed]
  165. Yu, D.W.; Li, Q.J.; Cheng, L.; Yang, P.F.; Sun, W.P.; Peng, Y.; Hu, J.J.; Wu, J.J.; Gong, J.P.; Zhong, G.C. Dietary Vitamin K Intake and the Risk of Pancreatic Cancer: A Prospective Study of 101,695 American Adults. Am. J. Epidemiol. 2021, 190, 2029–2041. [Google Scholar] [CrossRef] [PubMed]
  166. Duan, F.; Yu, Y.; Guan, R.; Xu, Z.; Liang, H.; Hong, L. Vitamin K2 Induces Mitochondria-Related Apoptosis in Human Bladder Cancer Cells via ROS and JNK/p38 MAPK Signal Pathways. PLoS ONE 2016, 11, e0161886. [Google Scholar] [CrossRef] [PubMed]
  167. Duan, F.; Mei, C.; Yang, L.; Zheng, J.; Lu, H.; Xia, Y.; Hsu, S.; Liang, H.; Hong, L. Vitamin K2 Promotes PI3K/AKT/HIF-1α-Mediated Glycolysis that Leads to AMPK-Dependent Autophagic Cell Death in Bladder Cancer Cells. Sci. Rep. 2020, 10, 7714. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of compounds in the vitamin K family.
Figure 1. Chemical structures of compounds in the vitamin K family.
Foods 13 01646 g001
Figure 2. Levels (µg per 100 g of product) of vitamin K1 (VK1) and vitamin K2 (VK2) in different foodstuffs.
Figure 2. Levels (µg per 100 g of product) of vitamin K1 (VK1) and vitamin K2 (VK2) in different foodstuffs.
Foods 13 01646 g002
Figure 3. The therapeutic potential of VK2 in various disease states.
Figure 3. The therapeutic potential of VK2 in various disease states.
Foods 13 01646 g003
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

Zhang, T.; O’Connor, C.; Sheridan, H.; Barlow, J.W. Vitamin K2 in Health and Disease: A Clinical Perspective. Foods 2024, 13, 1646. https://doi.org/10.3390/foods13111646

AMA Style

Zhang T, O’Connor C, Sheridan H, Barlow JW. Vitamin K2 in Health and Disease: A Clinical Perspective. Foods. 2024; 13(11):1646. https://doi.org/10.3390/foods13111646

Chicago/Turabian Style

Zhang, Tao, Christine O’Connor, Helen Sheridan, and James W. Barlow. 2024. "Vitamin K2 in Health and Disease: A Clinical Perspective" Foods 13, no. 11: 1646. https://doi.org/10.3390/foods13111646

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