Next Article in Journal
Young Children’s Sugar-Sweetened Beverage Consumption and 5-Year Change in BMI: Lessons Learned from the Timing of Consumption
Previous Article in Journal
The Environmental Impact of the Athlete’s Plate Nutrition Education Tool
Previous Article in Special Issue
Vitamin K Nutrition and Bone Health
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Beneficial Effects of Vitamin K Status on Glycemic Regulation and Diabetes Mellitus: A Mini-Review

1
Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
2
International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
*
Author to whom correspondence should be addressed.
Submission received: 22 July 2020 / Revised: 12 August 2020 / Accepted: 13 August 2020 / Published: 18 August 2020
(This article belongs to the Special Issue Vitamin K Intake and Human Health)

Abstract

:
Type 2 diabetes mellitus is a chronic disease that is characterized by hyperglycemia, insulin resistance, and dysfunctional insulin secretion. Glycemic control remains a crucial contributor to the progression of type 2 diabetes mellitus as well as the prevention or delay in the onset of diabetes-related complications. Vitamin K is a fat-soluble vitamin that plays an important role in the regulation of the glycemic status. Supplementation of vitamin K may reduce the risk of diabetes mellitus and improve insulin sensitivity. This mini-review summarizes the recent insights into the beneficial effects of vitamin K and its possible mechanism of action on insulin sensitivity and glycemic status, thereby suppressing the progression of diabetes mellitus.

1. Introduction

Type 2 diabetes mellitus (T2DM) is a chronic health condition that occurs when insulin secretion is impaired and manifests through features associated with insulin resistance. Several pathophysiologic defects, including the disequilibrium of insulin and glucagon secretory capacities of pancreatic α- and β-cells, hepatic steatosis, insulin resistance, reduced incretin secretion in the small intestine, and impaired glucose uptake in the peripheral tissues, cause the progressive hyperglycemia of T2DM [1,2]. Glycemic control remains a crucial contributor in the progression of T2DM and the prevention or delay in the onset of diabetes-related microvascular and macrovascular complications. Glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptide-1 (GLP-1) are incretins released from gut enteroendocrine cells, which play a major role in the postprandial regulation of insulin secretion through augmentation of insulin, suppression of glucagon secretion, and decrease of endogenous glucose production [3,4,5,6,7]. Studies have highlighted the possibility of the incretin-based treatment strategies [8,9]. Recently, GLP-1 receptor agonists and sodium-glucose cotransporter-2 (SGLT2) inhibitors are considered as the glucose-lowering drugs that have moderated benefits in reducing cardiovascular risk among T2DM patients [10]. In addition, a review integrated the genome-wide association studies (GWAS) of T2DM, implicating around 250 associated genetic variations to T2DM. GWAS is a helpful approach to understand the interaction between β-cell failure, insulin sensitivity, and adipose storage in healthy subjects and T2DM patients. Genomics studies provided information of T2DM therapeutics development [11].
Vitamin K (VK) is a fat-soluble vitamin that exists in two natural forms: VK1 (phylloquinone) and VK2 (menaquinone). VK1 is the major form of dietary VK, which is abundantly present in leafy greens [12]. VK2 is present in dairy products and fermented foods [13], and a homolog of VK2, menaquinone-4 (MK-4), is the major form of VK in animal tissues and is converted from a portion of the ingested VK1 and other menaquinones [14]. The postmortem evaluation of VK status in human tissues, including brain, heart, kidney, liver, lung, and pancreas, revealed that VK1 was stored in all tissues, but with relatively high levels in the liver, heart, and pancreas, whereas VK2 was stored in most of the tissues and had relatively high distribution in the brain, kidneys, and pancreas [15]. In rodents, we previously found that both VK1 and MK-4 are present in all tissues, including the brain, heart, kidney, liver, lung, pancreas, mesenteric fat, abdominal aorta, bone, ear, testis, stomach, skin, intestine, muscle, and spleen (Figure 1). The abovementioned finding was consistent with the results reported from an early study that MK-4 is the major form of VK throughout the body [14] and is observed in high quantities in the liver, bone, brain, pancreas, and reproductive organs, even when animals are fed a low-VK diet [16,17]. The accumulation of VK in the tissues suggested that it has specific physiological roles in the human body.
Several recent studies have mentioned that VK not only plays a role in blood coagulation and bone metabolism but also has specific functions in the regulation of glycemic status; therefore, a greater status of VK may be implicated in mediating a reduced risk of DM. In this mini-review, we outline the current knowledge on the beneficial effects of VK on suppressing the progression of DM.

2. Improvement of Insulin Sensitivity and Glycemic Status

Several studies evaluated the effect of VK on insulin response and glycemic status. The evidence indicates that blood VK status is positively correlated with plasma insulin level, and the fasting plasma glucose status was not markedly changed with the intake of VK. In observational studies, at 30 min after glucose loading, the plasma glucose level tended to decrease and the insulinogenic index increased in the high VK intake group, thereby suggesting that VK intake improved the acute insulin response with regard to glucose tolerance [18]. Another study that analyzed the association between the intake of VK1 and insulin sensitivity in older adults showed that higher VK1 ingestion correlated with higher insulin sensitivity and glycemic status in the 2-h oral glucose tolerance test [19], suggesting VK1 intake may have a beneficial effect on glucose homeostasis in adult men and women. Alternatively, in the oral glucose tolerance test, men with lower VK1 intake had decreased insulin levels and an increasing glucose level than men with higher VK1 intake [20,21]. A recent study that used a Mendelian randomization approach indicated that high circulating levels of VK1 are related to a lower risk of T2DM [22]. In intervention studies, a long-term clinical trial of VK supplementation on insulin resistance in older nondiabetic men and women showed that, at the attainable doses of VK1 supplementation for 36 months, the progression of insulin resistance improved in older men but not in women [23]. More recent intervention studies also found that VK1 supplementation for four weeks improved glycemic status and insulin sensitivity in premenopausal and prediabetic women [24,25]. On the other hand, one week of VK2 intake significantly reduced the immunoreactive insulin/plasma glucose ratio following oral glucose loading [26]. Another research team also supported the similar result that four weeks of VK2 supplementation increased insulin sensitivity in healthy young men [27]. In animal studies, rats with low VK intake had poor early insulin response and late hyperinsulinemia after intravenous glucose tolerance test [28]. Furthermore, in an arteriosclerotic rat model with DM, combined administration of VK2 (MK-4) and estradiol reduced the levels of aortic calcium (Ca) and phosphorus (P) and decreased the level of serum glucose while increasing the level of serum insulin, which suppressed the progression of arteriosclerosis with DM [29]. Table 1 summarizes the evidence that shows a significant association among VK status, insulin sensitivity, and glycemic levels.

3. Possible Effect of VK Supplementation on Insulin Secretion and Glycemic Status

Based on the evidence above indicating that VK affected insulin response and improved glucose tolerance in clinical and animal studies, the possible mechanisms for the regulation of glycemic status are summarized as follows.

3.1. Insulinotropic Effect

Recent studies have highlighted the possibility that two types of agents—incretin mimetics and incretin effect amplifiers—can lower blood glucose via the incretin system. The clinical agents include glucagon-like peptide-1 agonists and glucagon-like peptide-1 analog inhibitors [8,9,30]. Incretin mimetics increase the plasma incretin concentration, thereby contributing to a reduction in the glycated hemoglobin level, fasting blood glucose level, and body weight [31]. In our recent study, we found that MK-4 amplified glucose-stimulated insulin secretion in isolated mouse islets and INS-1 rat insulinoma cells. The findings indicated that MK-4 might function as an incretin-like nutrient via elevation of cAMP levels and resultant Epac2 regulation by using INS-1 cells [32].

3.2. Modulation of VK-Dependent Proteins

VK works as a cofactor for microsomal γ-glutamyl carboxylase and has a distinct role in the posttranslational carboxylation of glutamate to γ-carboxyglutamate (Gla) residues of VK-dependent proteins (VKDPs), such as matrix Gla protein (MGP) and osteocalcin (OC), which are involved in the inhibition of vascular calcification and bone mineralization, respectively [33]. Furthermore, these proteins play several beneficial roles in the biological processes and regulate physiological functions. Several studies have found correlations between disease progression and VKDP status, suggesting that VKDPs may potentially emerge as biomarkers for various diseases and, moreover, the VK status may play a crucial role in diseases such as DM [34].
Active MGP is recognized as an inhibitor of vascular calcification, both in vitro and in vivo, and considered to be a biomarker for VK deficiency [35,36,37,38]. Inactive MGP is identified in its carboxylated or phosphorylated forms, including the uncarboxylated MGP (ucMGP), carboxylated but not phosphorylated MGP (dpcMGP), phosphorylated but uncarboxylated (pucMGP), and the fully inactive uncarboxylated, dephosphorylated MGP (dpucMGP) [39,40]. Several studies have described that medial calcification is observed in DM patients, which correlates with the presence of VKDPs [41,42,43,44,45,46,47]. An early report indicated that media calcification was higher in DM patients than in the non-diabetic population [42]. Other studies found that the accumulation of advanced glycation end products correlated with coronary artery calcification in patients with type 1 DM (T1DM) [43] and in those with severe aortic valve stenosis [40]. Furthermore, high ucMGP levels were detected in DM patients and indicated a risk of arterial calcification, which has been observed in non-diabetic subjects [44,47] and T2DM patients [44,45,46].
Another VKDP, osteoblast-specific secreted OC, has been reported to be involved in the regulation of glucose metabolism. Several studies reported a bone-pancreas endocrine loop where insulin signaling stimulates osteoblasts differentiation and OC production, which in turn regulates insulin secretion in pancreatic islet cells [48,49,50,51]. Studies revealed that carboxylated osteocalcin (cOC) modulates hydroxyapatite crystals growth, whereas the undercarboxylated osteocalcin (ucOC) acts as an endocrine hormone in glucose metabolism, energy metabolism and fertility [52,53,54,55,56]. Findings from animal studies suggested that ucOC form improves insulin sensitivity and enhances β-cell functions through the stimulation of cyclin D1 and insulin expression in β cells and adiponectin expression in adipocytes, which ameliorated glucose intolerance in mice [57,58]. However, in clinical trial, participants who received VK1 supplementation had lower serum ucOC levels than the control group, suggesting that the protective effect of VK on the progression of insulin resistance may be mediated by decreasing the ucOC levels, which does not support the findings of the animal studies [23,27]. However, the difference could be species difference between rodent and humans. It is plausible that VK may improve insulin sensitivity and regulate glucose metabolism through modulation of OC and suppression of inflammation [23,26,59,60,61]. Recent clinical studies also supported that OC plays an important role in glucose metabolism by increasing insulin secretion and adiponectin expression [21,24,25]. In addition, Varsha et al. pointed out that VK1 administration may prevent hyperglycemia by protecting pancreatic islets in a streptozotocin (STZ)-induced T1DM model in the rat [62]. A report proposed that decreased level of blood cOC may be a selective early symptom of insulin resistance in obesity, whereas the decreased level of cOC seems to be associated with the appearance of early markers for inflammation accompanying obesity [63]. The association between DM and VKDPs suggests the existence of a novel therapeutic approach for glycemic control.

3.3. Prevention of Inflammation

Previous reports have suggested that the suppression of inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-6 in adipose tissue is associated with insulin sensitivity [64,65,66]. Obesity causes a low-grade inflammation that contributes to the development of insulin resistance and T2DM, suggesting the increased proinflammatory cytokines as key mediators of innate inflammatory responses which contribute to the development of insulin resistance [67,68,69]. Several chronic diseases caused by inflammatory disorders have been associated with VK deficiency [70,71,72]. The evidence shows that VK may attenuate the insulin response and glycemic status through the inhibition of inflammation. In a study that assessed the status of fat-soluble vitamins in patients with chronic pancreatitis, the results indicate that the serum concentrations of fat-soluble vitamins and bone mineral density were decreased in those patients [72]. Similarly, other studies have reported that VK suppressed IL-6 production in lipopolysaccharide-induced inflammation models [59,60]. Moreover, high plasma VK1 concentration and VK1 intake were associated with decreased concentrations of inflammatory markers TNF-α and IL-6 [61]. All of the abovementioned evidence suggests a potential role for VK in the mediation of inflammation and insulin sensitivity.

4. Beneficial Effects of VK on Diabetes-Related Complications

The populations with DM that accompany metabolic complications continue to increase worldwide. Diabetes-related complications are generally described as microvascular and macrovascular complications, including retinopathy, kidney disease, neuropathy, and cardiovascular disease (CVD) [73,74]. In this section, we discuss the protective effects of VK on diabetes-related complications.

4.1. Cataractogenesis

A recent study reported that the VKDP, active MGP, exhibits anti-calcification and anti-stiffness properties, thereby maintaining retinal microcirculation, which might be considered to be a marker of retinal health [75]. The cataract in rats with STZ-induced diabetes was accompanied by hyperglycemia, high lens aldose reductase 2 (ALR2) activity, accumulation of sorbitol, and the formation of advanced glycation end products within the eye lens that led to diabetes-related cataractogenesis. However, in the rats that were treated with VK1, there was a decrease in the blood glucose level, ALR2 activity, and accumulation of lens sorbitol. The study indicated that VK1 is a potent inhibitor of ALR2 through the inhibition of its substrate-binding site, which suggests a possible mechanism of action of VK1 on diabetes-related cataract formation [76,77].

4.2. Diabetic Nephropathy

Increasingly, there is evidence showing that diabetic nephropathy is a serious complication of T1DM and T2DM. Several studies have demonstrated poor VK status and, subsequently, low serum VKDPs levels in patients with chronic kidney disease (CKD). With regard to the VK status, the level of the VKDP, MGP, was highly correlated with the CKD stage [78,79,80,81,82]. There was a strong inverse correlation between the circulating dpucMGP levels and CKD stages, suggesting that MGP as a predictor of mortality in patients with diabetic nephropathy [82,83]; furthermore, the plasma dpucMGP level correlated with albuminuria and proteinuria and was inversely associated with the estimated glomerular filtration rate (eGFR) [84,85]. CKD patients who were undergoing maintenance hemodialysis showed a higher plasma dpucMGP level [86,87]. Moreover, cohort studies revealed that the plasma dpucMGP levels increased with the progression of CKD, especially in patients with CKD Stages 3–5 [88,89]. In addition, other studies have evaluated the risk of increasing dpucMGP levels on renal function. An elevation of the renal resistive index (RRI) that is widely used to evaluate renal dysfunction is associated with adverse renal and cardiovascular outcomes [90]. A recent study indicated that dpucMGP levels correlated with RRI, cardiovascular risk factors, and renal function [91]. The recent Nephrotic Syndrome Study Network cohort study reported that renal MGP expression increased in 5/6 nephrectomy rats [92]. In the same study, the correlation with the MGP levels was investigated by using the information from the kidney biopsies and indicated that eGFR was inversely associated with tubulointerstitial and glomerular MGP mRNA expression in patients with nephrotic syndrome. The tubulointerstitial MGP mRNA expression was strongly correlated with renal inflammation, fibrosis, and acute tubular injury, independently of the eGFR. High MGP mRNA expression was associated with an increased risk for the composite of 40% decline in eGFR and end-stage renal disease [92]. This evidence explains the renoprotective role of MGP and further indicate that VK exerts a beneficial effect on renal function.

4.3. Diabetic Peripheral Neuropathy

Diabetic peripheral neuropathy is another frequent and severe metabolic complication of DM; poor glycemic control and dyslipidemia are known to be risk factors for diabetic neuropathy [93,94]. The evidence supports that VK status might be related to nervous system homeostasis. An early study indicated the survival-promoting role of VK on the maintenance of the survival ability of CNS neurons [95]. A report has shown that MGP is expressed by neurons and glial cells [96]. Moreover, the early differentiation and growth of neurons, dendrite formation, development of mature Schwann cells, and myelination are regulated through the interactions of the extracellular matrix and MGP [97,98,99]. Furthermore, dpucMGP plasma levels increased in patients with diabetic peripheral neuropathy and poor VK status, suggesting that MGP plays a role in the homeostasis of the nervous system [46]. Retinopathy and nephropathy are comorbidities that exist with diabetic neuropathy, and, as other reports have indicated, VK exerts a renoprotective effect, which may extend to the prevention of other diabetes-related complications.

4.4. Cardiovascular Disease

One of the commonest complications in patients with DM is CVD, including heart failure, vascular disease, and stroke [100]. As mentioned above, in an arteriosclerotic rat model with DM, MK-4 administration with estradiol reduced the levels of aortic Ca and P and suppressed the progression of arteriosclerosis with DM [29]. Moreover, poor VK status has been associated with an increasing risk of CVD in patients with DM [45].
Vascular calcification has long been considered to be a cause of cardiovascular morbidity and mortality. VK plays a role in the modulation of VKDPs that are involved in vascular cell migration, angiogenesis, and calcification [34]. Because a VK deficiency results in increased levels of ucMGP, several studies have implicated ucMGP as a risk factor for vascular calcification and CVD [38,44,45,46,101,102]. A cohort study found that the plasma level of ucMGP was associated with eGFR in patients with CVD [47].
A proof-of-concept study assigned patients with aortic valve calcification and normal renal function to either the VK1 or placebo groups for 12 months. The results indicate that VK administration decreased the serum dpucMGP levels and slowed the progression of cardiac valve calcification [103]. A multicenter family-based cross-sectional study in Switzerland showed that high plasma levels of dpucMGP were independently and positively associated with RRI, after adjustment for several common CVD risk factors [104]. As mentioned above, there are an increasing number of reports indicating that greater VK intake is associated with improved CVD risk factors [105,106], and VKDP activity is associated with CVD through the inhibition of vascular calcification [107,108,109].

4.5. Osteopenia and Osteoporosis

DM is a risk factor for osteoporotic fractures, and there is evidence of the increased incidence of osteoporosis in patients with DM. VK plays an important role in the prevention of fractures and the maintenance of bone mineral density and bone quality [110,111,112,113]. In the STZ-induced T1DM rats, the correlation between hyperglycemia and a decrease in femoral weight was confirmed. However, the oral administration of MK-4 for five days a week for 12 weeks in rats prevented the development of hyperglycemia as well as a decrease in the femoral weight, suggesting that VK has beneficial effects on cancellous bone mass in rats with STZ-induced T1DM [114]. The results of randomized controlled trials that investigated the association between osteoporosis and VK in postmenopausal women suggest that MK-4 treatment effectively prevents the occurrence of osteoporotic fractures and a decrease in the serum ucOC level. However, the effect of MK-4 might have occurred with or without an accompanying increase in the bone mineral density [115,116,117,118,119].

5. Conclusions

The potential effects of VK on DM and its complications have been demonstrated previously (Figure 2). Reports have mentioned the safety and beneficial effect of VK supplementation in humans [120,121]. However, interventional studies are required to clarify the preventive and protective effects of VK on DM and its complications.

Author Contributions

All authors contributed in writing and reviewing the article. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by a Grant-in-aid for Scientific Research from the Japan Society for the Promotion of Science (JSPS) to HS (#17H0314 and #20H02928) and by the JSPS Core-to-Core Program A (Advanced Research Networks) entitled “Establishment of international agricultural immunology research-core for a quantum improvement in food safety”.

Conflicts of Interest

The authors declare no conflict of interests.

References

  1. Chaudhury, A.; Duvoor, C.; Reddy Dendi, V.S.; Kraleti, S.; Chada, A.; Ravilla, R.; Marco, A.; Shekhawat, N.S.; Montales, M.T.; Kuriakose, K.; et al. Clinical Review of Antidiabetic Drugs: Implications for Type 2 Diabetes Mellitus Management. Front. Endocrinol. (Lausanne) 2017, 8, 6. [Google Scholar] [CrossRef] [Green Version]
  2. Defronzo, R.A. Banting Lecture. From the triumvirate to the ominous octet: A new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009, 58, 773–795. [Google Scholar] [CrossRef] [Green Version]
  3. Komatsu, R.; Matsuyama, T.; Namba, M.; Watanabe, N.; Itoh, H.; Kono, N.; Tarui, S. Glucagonostatic and insulinotropic action of glucagonlike peptide I-(7-36)-amide. Diabetes 1989, 38, 902–905. [Google Scholar] [CrossRef]
  4. Meier, J.J.; Nauck, M.A.; Schmidt, W.E.; Gallwitz, B. Gastric inhibitory polypeptide: The neglected incretin revisited. Regul. Pept. 2002, 107, 1–13. [Google Scholar] [CrossRef]
  5. Vilsbøll, T.; Krarup, T.; Madsbad, S.; Holst, J.J. Both GLP-1 and GIP are insulinotropic at basal and postprandial glucose levels and contribute nearly equally to the incretin effect of a meal in healthy subjects. Regul. Pept. 2003, 114, 115–121. [Google Scholar] [CrossRef]
  6. Prigeon, R.L.; Quddusi, S.; Paty, B.; D′Alessio, D.A. Suppression of glucose production by GLP-1 independent of islet hormones: A novel extrapancreatic effect. Am. J. Physiol. Endocrinol. Metab. 2003, 285, E701–E707. [Google Scholar] [CrossRef]
  7. Drucker, D.J. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef] [Green Version]
  8. Herzberg-Schäfer, S.; Heni, M.; Stefan, N.; Häring, H.U.; Fritsche, A. Impairment of GLP1-induced insulin secretion: Role of genetic background, insulin resistance and hyperglycaemia. Diabetes Obes. Metab. 2012, 14 (Suppl. 3), 85–90. [Google Scholar] [CrossRef]
  9. Tasyurek, H.M.; Altunbas, H.A.; Balci, M.K.; Sanlioglu, S. Incretins: Their physiology and application in the treatment of diabetes mellitus. Diabetes Metab. Res. Rev. 2014, 30, 354–371. [Google Scholar] [CrossRef]
  10. Ghosh-Swaby, O.R.; Goodman, S.G.; Leiter, L.A.; Cheng, A.; Connelly, K.A.; Fitchett, D.; Jüni, P.; Farkouh, M.E.; Udell, J.A. Glucose-lowering drugs or strategies, atherosclerotic cardiovascular events, and heart failure in people with or at risk of type 2 diabetes: An updated systematic review and meta-analysis of randomised cardiovascular outcome trials. Lancet Diabetes Endocrinol. 2020, 8, 418–435. [Google Scholar] [CrossRef]
  11. Langenberg, C.; Lotta, L.A. Genomic insights into the causes of type 2 diabetes. Lancet 2018, 391, 2463–2474. [Google Scholar] [CrossRef]
  12. Booth, S.L. Vitamin K: Food composition and dietary intakes. Food Nutr. Res. 2012, 56, 5505. [Google Scholar] [CrossRef] [Green Version]
  13. Fu, X.; Harshman, S.G.; Shen, X.; Haytowitz, D.B.; Karl, J.P.; Wolfe, B.E.; Booth, S.L. Multiple Vitamin K Forms Exist in Dairy Foods. Curr. Dev. Nutr. 2017, 1, e000638. [Google Scholar] [CrossRef] [Green Version]
  14. Martius, C.; Alvino, C. On the transformation of vitamin K-1 (phyllochinon) into vitamin K2(20) by the development of an embryo in a hen′s egg. Biochem. Z 1964, 340, 316–319. [Google Scholar]
  15. Thijssen, H.H.; Drittij-Reijnders, M.J. Vitamin K status in human tissues: Tissue-specific accumulation of phylloquinone and menaquinone-4. Br. J. Nutr. 1996, 75, 121–127. [Google Scholar] [CrossRef]
  16. 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]
  17. Shirakawa, H.; Katsurai, T.; Komai, M. Conversion of menaquinone-4 in animal organs and it functions. Jpn. Oil Chem. Soc. 2014, 14, 547–553. [Google Scholar] [CrossRef]
  18. Sakamoto, N.; Nishiike, T.; Iguchi, H.; Sakamoto, K. Relationship between acute insulin response and vitamin K intake in healthy young male volunteers. Diabetes Nutr. Metab. 1999, 12, 37–41. [Google Scholar]
  19. Yoshida, M.; Booth, S.L.; Meigs, J.B.; Saltzman, E.; Jacques, P.F. Phylloquinone intake, insulin sensitivity, and glycemic status in men and women. Am. J. Clin. Nutr. 2008, 88, 210–215. [Google Scholar] [CrossRef] [Green Version]
  20. Beulens, J.W.; van der, A.D.; 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] [Green Version]
  21. Ibarrola-Jurado, N.; Salas-Salvadó, J.; Martínez-González, M.A.; Bulló, M. Dietary phylloquinone intake and risk of type 2 diabetes in elderly subjects at high risk of cardiovascular disease. Am. J. Clin. Nutr. 2012, 96, 1113–1118. [Google Scholar] [CrossRef]
  22. Zwakenberg, S.R.; Remmelzwaal, S.; Beulens, J.W.J.; Booth, S.L.; Burgess, S.; Dashti, H.S.; Imamura, F.; Feskens, E.J.M.; van der Schouw, Y.T.; Sluijs, I. Circulating Phylloquinone Concentrations and Risk of Type 2 Diabetes: A Mendelian Randomization Study. Diabetes 2019, 68, 220–225. [Google Scholar] [CrossRef] [Green Version]
  23. Yoshida, M.; Jacques, P.F.; Meigs, J.B.; Saltzman, E.; Shea, M.K.; Gundberg, C.; Dawson-Hughes, B.; Dallal, G.; Booth, S.L. Effect of vitamin K supplementation on insulin resistance in older men and women. Diabetes Care 2008, 31, 2092–2096. [Google Scholar] [CrossRef] [Green Version]
  24. Rasekhi, H.; Karandish, M.; Jalali, M.T.; Mohammadshahi, M.; Zarei, M.; Saki, A.; Shahbazian, H. Phylloquinone supplementation improves glycemic status independent of the effects of adiponectin levels in premonopause women with prediabetes: A double-blind randomized controlled clinical trial. J. Diabetes Metab. Disord. 2015, 14, 1. [Google Scholar] [CrossRef] [Green Version]
  25. 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]
  26. Sakamoto, N.; Nishiike, T.; Iguchi, H.; Sakamoto, K. Possible effects of one week vitamin K (menaquinone-4) tablets intake on glucose tolerance in healthy young male volunteers with different descarboxy prothrombin levels. Clin. Nutr. 2000, 19, 259–263. [Google Scholar] [CrossRef]
  27. 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] [Green Version]
  28. Sakamoto, N.; Wakabayashi, I.; Sakamoto, K. Low vitamin K intake effects on glucose tolerance in rats. Int. J. Vitam. Nutr. Res. 1999, 69, 27–31. [Google Scholar] [CrossRef]
  29. Seyama, Y.; Kimoto, S.; Marukawa, Y.; Horiuchi, M.; Hayashi, M.; Usami, E. Comparative effects of vitamin K2 and estradiol on experimental arteriosclerosis with diabetes mellitus. Int. J. Vitam. Nutr. Res. 2000, 70, 301–304. [Google Scholar] [CrossRef] [Green Version]
  30. Barnett, A.H. The role of GLP-1 mimetics and basal insulin analogues in type 2 diabetes mellitus: Guidance from studies of liraglutide. Diabetes Obes. Metab. 2012, 14, 304–314. [Google Scholar] [CrossRef] [Green Version]
  31. Drucker, D.J.; Nauck, M.A. The incretin system: Glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006, 368, 1696–1705. [Google Scholar] [CrossRef]
  32. Ho, H.J.; Shirakawa, H.; Hirahara, K.; Sone, H.; Kamiyama, S.; Komai, M. Menaquinone-4 Amplified Glucose-Stimulated Insulin Secretion in Isolated Mouse Pancreatic Islets and INS-1 Rat Insulinoma Cells. Int. J. Mol. Sci. 2019, 20, 1995. [Google Scholar] [CrossRef] [Green Version]
  33. Fusaro, M.; Gallieni, M.; Rizzo, M.A.; Stucchi, A.; Delanaye, P.; Cavalier, E.; Moysés, R.M.A.; Jorgetti, V.; Iervasi, G.; Giannini, S.; et al. Vitamin K plasma levels determination in human health. Clin. Chem. Lab. Med. 2017, 55, 789–799. [Google Scholar] [CrossRef]
  34. El Asmar, M.S.; Naoum, J.J.; Arbid, E.J. Vitamin k dependent proteins and the role of vitamin k2 in the modulation of vascular calcification: A review. Oman Med. J. 2014, 29, 172–177. [Google Scholar] [CrossRef]
  35. Luo, G.; Ducy, P.; McKee, M.D.; Pinero, G.J.; Loyer, E.; Behringer, R.R.; Karsenty, G. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature 1997, 386, 78–81. [Google Scholar] [CrossRef]
  36. Teebi, A.S.; Lambert, D.M.; Kaye, G.M.; Al-Fifi, S.; Tewfik, T.L.; Azouz, E.M. Keutel syndrome: Further characterization and review. Am. J. Med. Genet. 1998, 78, 182–187. [Google Scholar]
  37. Munroe, P.B.; Olgunturk, R.O.; Fryns, J.P.; Van Maldergem, L.; Ziereisen, F.; Yuksel, B.; Gardiner, R.M.; Chung, E. Mutations in the gene encoding the human matrix Gla protein cause Keutel syndrome. Nat. Genet. 1999, 21, 142–144. [Google Scholar] [CrossRef]
  38. Cranenburg, E.C.; Koos, R.; Schurgers, L.J.; Magdeleyns, E.J.; Schoonbrood, T.H.; Landewe, R.B.; Brandenburg, V.M.; Bekers, O.; Vermeer, C. Characterisation and potential diagnostic value of circulating matrix Gla protein (MGP) species. Thromb. Haemost. 2010, 104, 811–822. [Google Scholar] [CrossRef]
  39. 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] [Green Version]
  40. Basta, G.; Corciu, A.I.; Vianello, A.; Del Turco, S.; Foffa, I.; Navarra, T.; Chiappino, D.; Berti, S.; Mazzone, A. Circulating soluble receptor for advanced glycation end-product levels are decreased in patients with calcific aortic valve stenosis. Atherosclerosis 2010, 210, 614–618. [Google Scholar] [CrossRef]
  41. Olson, J.C.; Edmundowicz, D.; Becker, D.J.; Kuller, L.H.; Orchard, T.J. Coronary calcium in adults with type 1 diabetes: A stronger correlate of clinical coronary artery disease in men than in women. Diabetes 2000, 49, 1571–1578. [Google Scholar] [CrossRef] [Green Version]
  42. Neubauer, B. A quantitative study of peripheral arterial calcification and glucose tolerance in elderly diabetics and non-diabetics. Diabetologia 1971, 7, 409–413. [Google Scholar] [CrossRef] [Green Version]
  43. Conway, B.; Edmundowicz, D.; Matter, N.; Maynard, J.; Orchard, T. Skin fluorescence correlates strongly with coronary artery calcification severity in type 1 diabetes. Diabetes Technol. 2010, 12, 339–345. [Google Scholar] [CrossRef]
  44. Thomsen, S.B.; Rathcke, C.N.; Zerahn, B.; Vestergaard, H. Increased levels of the calcification marker matrix Gla Protein and the inflammatory markers YKL-40 and CRP in patients with type 2 diabetes and ischemic heart disease. Cardiovasc. Diabetol. 2010, 9, 86. [Google Scholar] [CrossRef] [Green Version]
  45. Dalmeijer, G.W.; van der Schouw, Y.T.; Magdeleyns, E.J.; Vermeer, C.; Verschuren, W.M.; Boer, J.M.; Beulens, J.W. Matrix Gla protein species and risk of cardiovascular events in type 2 diabetic patients. Diabetes Care 2013, 36, 3766–3771. [Google Scholar] [CrossRef] [Green Version]
  46. Jeannin, A.C.; Salem, J.E.; Massy, Z.; Aubert, C.E.; Vemeer, 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]
  47. Parker, B.D.; Ix, J.H.; Cranenburg, E.C.; Vermeer, C.; Whooley, M.A.; Schurgers, L.J. Association of kidney function and uncarboxylated matrix Gla protein: Data from the Heart and Soul Study. Nephrol. Dial. Transpl. 2009, 24, 2095–2101. [Google Scholar] [CrossRef]
  48. Fulzele, K.; Riddle, R.C.; DiGirolamo, D.J.; Cao, X.; Wan, C.; Chen, D.; Faugere, M.C.; Aja, S.; Hussain, M.A.; Brüning, J.C.; et al. Insulin receptor signaling in osteoblasts regulates postnatal bone acquisition and body composition. Cell 2010, 142, 309–319. [Google Scholar] [CrossRef] [Green Version]
  49. Faienza, M.F.; Luce, V.; Ventura, A.; Colaianni, G.; Colucci, S.; Cavallo, L.; Grano, M.; Brunetti, G. Skeleton and glucose metabolism: A bone-pancreas loop. Int. J. Endocrinol. 2015, 2015, 758148. [Google Scholar] [CrossRef] [Green Version]
  50. Zoch, M.L.; Clemens, T.L.; Riddle, R.C. New insights into the biology of osteocalcin. Bone 2016, 82, 42–49. [Google Scholar] [CrossRef] [Green Version]
  51. Moser, S.C.; van der Eerden, B.C.J. Osteocalcin-A Versatile Bone-Derived Hormone. Front. Endocrinol. (Lausanne) 2018, 9, 794. [Google Scholar] [CrossRef] [Green Version]
  52. Oury, F.; Sumara, G.; Sumara, O.; Ferron, M.; Chang, H.; Smith, C.E.; Hermo, L.; Suarez, S.; Roth, B.L.; Ducy, P.; et al. Endocrine regulation of male fertility by the skeleton. Cell 2011, 144, 796–809. [Google Scholar] [CrossRef] [Green Version]
  53. Schwetz, V.; Pieber, T.; Obermayer-Pietsch, B. The endocrine role of the skeleton: Background and clinical evidence. Eur. J. Endocrinol. 2012, 166, 959–967. [Google Scholar] [CrossRef] [Green Version]
  54. Oury, F.; Ferron, M.; Huizhen, W.; Confavreux, C.; Xu, L.; Lacombe, J.; Srinivas, P.; Chamouni, A.; Lugani, F.; Lejeune, H.; et al. Osteocalcin regulates murine and human fertility through a pancreas-bone-testis axis. J. Clin. Investig. 2013, 123, 2421–2433. [Google Scholar] [CrossRef]
  55. Karsenty, G.; Oury, F. Regulation of male fertility by the bone-derived hormone osteocalcin. Mol. Cell Endocrinol. 2014, 382, 521–526. [Google Scholar] [CrossRef] [Green Version]
  56. Wei, J.; Ferron, M.; Clarke, C.J.; Hannun, Y.A.; Jiang, H.; Blaner, W.S.; Karsenty, G. Bone-specific insulin resistance disrupts whole-body glucose homeostasis via decreased osteocalcin activation. J. Clin. Investig. 2014, 124, 1781–1793. [Google Scholar] [CrossRef] [Green Version]
  57. 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] [Green Version]
  58. Ferron, M.; Hinoi, E.; Karsenty, G.; Ducy, P. Osteocalcin differentially regulates beta cell and adipocyte gene expression and affects the development of metabolic diseases in wild-type mice. Proc. Natl. Acad. Sci. USA 2008, 105, 5266–5270. [Google Scholar] [CrossRef] [Green Version]
  59. Reddi, K.; Henderson, B.; Meghji, S.; Wilson, M.; Poole, S.; Hopper, C.; Harris, M.; Hodges, S.J. Interleukin 6 production by lipopolysaccharide-stimulated human fibroblasts is potently inhibited by naphthoquinone (vitamin K) compounds. Cytokine 1995, 7, 287–290. [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] [Green Version]
  61. Shea, M.K.; Booth, S.L.; Massaro, J.M.; Jacques, P.F.; D′Agostino, R.B., Sr.; Dawson-Hughes, B.; Ordovas, J.M.; O′Donnell, C.J.; Kathiresan, S.; Keaney, J.F., Jr.; et al. Vitamin K and vitamin D status: Associations with inflammatory markers in the Framingham Offspring Study. Am. J. Epidemiol. 2008, 167, 313–320. [Google Scholar] [CrossRef] [Green Version]
  62. 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-κB activation and iNOS expression in rat pancreas. Nutrition 2015, 31, 214–222. [Google Scholar] [CrossRef]
  63. Razny, U.; Fedak, D.; Kiec-Wilk, B.; Goralska, J.; Gruca, A.; Zdzienicka, A.; Kiec-Klimczak, M.; Solnica, B.; Hubalewska-Dydejczyk, A.; Malczewska-Malec, M. Carboxylated and undercarboxylated osteocalcin in metabolic complications of human obesity and prediabetes. Diabetes Metab. Res. Rev. 2017, 33, e2862. [Google Scholar] [CrossRef]
  64. Juanola-Falgarona, M.; Salas-Salvadó, J.; Estruch, R.; Portillo, M.P.; Casas, R.; Miranda, J.; Martínez-González, M.A.; Bulló, M. Association between dietary phylloquinone intake and peripheral metabolic risk markers related to insulin resistance and diabetes in elderly subjects at high cardiovascular risk. Cardiovasc. Diabetol. 2013, 12, 7. [Google Scholar] [CrossRef] [Green Version]
  65. Wieser, V.; Moschen, A.R.; Tilg, H. Inflammation, cytokines and insulin resistance: A clinical perspective. Arch. Immunol. Exp. (Warsz) 2013, 61, 119–125. [Google Scholar] [CrossRef]
  66. Rehman, K.; Akash, M.S. Mechanisms of inflammatory responses and development of insulin resistance: How are they interlinked? J. Biomed. Sci. 2016, 23, 87. [Google Scholar] [CrossRef] [Green Version]
  67. Hotamisligil, G.S.; Murray, D.L.; Choy, L.N.; Spiegelman, B.M. Tumor necrosis factor alpha inhibits signaling from the insulin receptor. Proc. Natl. Acad. Sci. USA 1994, 91, 4854–4858. [Google Scholar] [CrossRef] [Green Version]
  68. Ballak, D.B.; Stienstra, R.; Tack, C.J.; Dinarello, C.A.; van Diepen, J.A. IL-1 family members in the pathogenesis and treatment of metabolic disease: Focus on adipose tissue inflammation and insulin resistance. Cytokine 2015, 75, 280–290. [Google Scholar] [CrossRef] [Green Version]
  69. Stefan, N. Causes, consequences, and treatment of metabolically unhealthy fat distribution. Lancet Diabetes Endocrinol. 2020, 8, 616–627. [Google Scholar] [CrossRef]
  70. Kleinman, R.E.; Fracchia, M.S. Vitamin K and cystic fibrosis: Give me a double, please. Am. J. Clin. Nutr. 2010, 92, 469–470. [Google Scholar] [CrossRef] [Green Version]
  71. Nakajima, S.; Iijima, H.; Egawa, S.; Shinzaki, S.; Kondo, J.; Inoue, T.; Hayashi, Y.; Ying, J.; Mukai, A.; Akasaka, T.; et al. Association of vitamin K deficiency with bone metabolism and clinical disease activity in inflammatory bowel disease. Nutrition 2011, 27, 1023–1028. [Google Scholar] [CrossRef]
  72. Sikkens, E.C.; Cahen, D.L.; Koch, A.D.; Braat, H.; Poley, J.W.; Kuipers, E.J.; Bruno, M.J. The prevalence of fat-soluble vitamin deficiencies and a decreased bone mass in patients with chronic pancreatitis. Pancreatology 2013, 13, 238–242. [Google Scholar] [CrossRef]
  73. Holman, R.R.; Paul, S.K.; Bethel, M.A.; Matthews, D.R.; Neil, H.A. 10-year follow-up of intensive glucose control in type 2 diabetes. N. Engl. J. Med. 2008, 359, 1577–1589. [Google Scholar] [CrossRef] [Green Version]
  74. Harding, J.L.; Pavkov, M.E.; Magliano, D.J.; Shaw, J.E.; Gregg, E.W. Global trends in diabetes complications: A review of current evidence. Diabetologia 2019, 62, 3–16. [Google Scholar] [CrossRef] [Green Version]
  75. Wei, F.F.; Huang, Q.F.; Zhang, Z.Y.; Van Keer, K.; Thijs, L.; Trenson, S.; Yang, W.Y.; Cauwenberghs, N.; Mujaj, B.; Kuznetsova, T.; et al. Inactive matrix Gla protein is a novel circulating biomarker predicting retinal arteriolar narrowing in humans. Sci. Rep. 2018, 8, 15088. [Google Scholar] [CrossRef] [Green Version]
  76. Sai Varsha, M.K.; Raman, T.; Manikandan, R. Inhibition of diabetic-cataract by vitamin K1 involves modulation of hyperglycemia-induced alterations to lens calcium homeostasis. Exp. Eye Res. 2014, 128, 73–82. [Google Scholar] [CrossRef]
  77. Thiagarajan, R.; Varsha, M.; Srinivasan, V.; Ravichandran, R.; Saraboji, K. Vitamin K1 prevents diabetic cataract by inhibiting lens aldose reductase 2 (ALR2) activity. Sci. Rep. 2019, 9, 14684. [Google Scholar] [CrossRef] [Green Version]
  78. Holden, R.M.; Morton, A.R.; Garland, J.S.; Pavlov, A.; Day, A.G.; Booth, S.L. Vitamins K and D status in stages 3–5 chronic kidney disease. Clin. J. Am. Soc. Nephrol. 2010, 5, 590–597. [Google Scholar] [CrossRef]
  79. Cranenburg, E.C.; Schurgers, L.J.; Uiterwijk, H.H.; Beulens, J.W.; Dalmeijer, G.W.; Westerhuis, R.; Magdeleyns, E.J.; Herfs, M.; Vermeer, C.; Laverman, G.D. Vitamin K intake and status are low in hemodialysis patients. Kidney Int. 2012, 82, 605–610. [Google Scholar] [CrossRef] [Green Version]
  80. 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] [Green Version]
  81. Epstein, M. Matrix Gla-Protein (MGP) Not Only Inhibits Calcification in Large Arteries but Also May Be Renoprotective: Connecting the Dots. EBioMedicine 2016, 4, 16–17. [Google Scholar] [CrossRef] [Green Version]
  82. 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]
  83. Roumeliotis, S.; Roumeliotis, A.; Panagoutsos, S.; Giannakopoulou, E.; Papanas, N.; Manolopoulos, V.G.; Passadakis, P.; Tavridou, A. Matrix Gla protein T-138C polymorphism is associated with carotid intima media thickness and predicts mortality in patients with diabetic nephropathy. J. Diabetes Complicat. 2017, 31, 1527–1532. [Google Scholar] [CrossRef]
  84. 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] [Green Version]
  85. Wei, F.F.; Trenson, S.; Thijs, L.; Huang, Q.F.; Zhang, Z.Y.; Yang, W.Y.; Moliterno, P.; Allegaert, K.; Boggia, J.; Janssens, S.; et al. Desphospho-uncarboxylated matrix Gla protein is a novel circulating biomarker predicting deterioration of renal function in the general population. Nephrol. Dial. Transpl. 2018, 33, 1122–1128. [Google Scholar] [CrossRef]
  86. 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]
  87. Aoun, M.; Makki, M.; Azar, H.; Matta, H.; Chelala, D.N. High Dephosphorylated-Uncarboxylated MGP in Hemodialysis patients: Risk factors and response to vitamin K2, a pre-post intervention clinical trial. BMC Nephrol. 2017, 18, 191. [Google Scholar] [CrossRef]
  88. Thamratnopkoon, S.; Susantitaphong, P.; Tumkosit, M.; Katavetin, P.; Tiranathanagul, K.; Praditpornsilpa, K.; Eiam-Ong, S. Correlations of Plasma Desphosphorylated Uncarboxylated Matrix Gla Protein with Vascular Calcification and Vascular Stiffness in Chronic Kidney Disease. Nephron 2017, 135, 167–172. [Google Scholar] [CrossRef]
  89. Jaminon, A.M.G.; Dai, L.; Qureshi, A.R.; Evenepoel, P.; Ripsweden, J.; Soderberg, 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] [Green Version]
  90. Doi, Y.; Iwashima, Y.; Yoshihara, F.; Kamide, K.; Hayashi, S.; Kubota, Y.; Nakamura, S.; Horio, T.; Kawano, Y. Renal resistive index and cardiovascular and renal outcomes in essential hypertension. Hypertension 2012, 60, 770–777. [Google Scholar] [CrossRef] [Green Version]
  91. Jaques, D.A.; Pivin, E.; Pruijm, M.; Ackermann, D.; Guessous, I.; Ehret, G.; Wei, F.F.; Staessen, J.A.; Pechère-Bertschi, A.; Vermeer, C.; et al. Renal Resistive Index Is Associated with Inactive Matrix Gla (γ-Carboxyglutamate) Protein in an Adult Population-Based Study. J. Am. Heart Assoc. 2019, 8, e013558. [Google Scholar] [CrossRef]
  92. 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]
  93. Dyck, P.J. Detection, characterization, and staging of polyneuropathy: Assessed in diabetics. Muscle Nerve 1988, 11, 21–32. [Google Scholar] [CrossRef]
  94. Papanas, N.; Ziegler, D. Risk Factors and Comorbidities in Diabetic Neuropathy: An Update 2015. Rev. Diabet Stud. 2015, 12, 48–62. [Google Scholar] [CrossRef] [Green Version]
  95. Nakajima, M.; Furukawa, S.; Hayashi, K.; Yamada, A.; Kawashima, T.; Hayashi, Y. Age-dependent survival-promoting activity of vitamin K on cultured CNS neurons. Brain Res. Dev. Brain Res. 1993, 73, 17–23. [Google Scholar] [CrossRef]
  96. Goritz, C.; Thiebaut, R.; Tessier, L.H.; Nieweg, K.; Moehle, C.; Buard, I.; Dupont, J.L.; Schurgers, L.J.; Schmitz, G.; Pfrieger, F.W. Glia-induced neuronal differentiation by transcriptional regulation. Glia 2007, 55, 1108–1122. [Google Scholar] [CrossRef]
  97. Nishimoto, S.K.; Nishimoto, M. Matrix Gla protein C-terminal region binds to vitronectin. Co-localization suggests binding occurs during tissue development. Matrix Biol. 2005, 24, 353–361. [Google Scholar] [CrossRef]
  98. Moon, J.I.; Birren, S.J. Target-dependent inhibition of sympathetic neuron growth via modulation of a BMP signaling pathway. Dev. Biol. 2008, 315, 404–417. [Google Scholar] [CrossRef] [Green Version]
  99. Nishimoto, S.K.; Nishimoto, M. Matrix gla protein binds to fibronectin and enhances cell attachment and spreading on fibronectin. Int. J. Cell Biol. 2014, 2014, 807013. [Google Scholar] [CrossRef] [Green Version]
  100. Kendall, D.M.; Harmel, A.P. The metabolic syndrome, type 2 diabetes, and cardiovascular disease: Understanding the role of insulin resistance. Am. J. Manag. Care 2002, 8, S635–S653. [Google Scholar]
  101. Schurgers, L.J.; Cranenburg, E.C.; Vermeer, C. Matrix Gla-protein: The calcification inhibitor in need of vitamin K. Thromb. Haemost. 2008, 100, 593–603. [Google Scholar]
  102. Shea, M.K.; Holden, R.M. Vitamin K status and vascular calcification: Evidence from observational and clinical studies. Adv. Nutr. 2012, 3, 158–165. [Google Scholar] [CrossRef] [Green Version]
  103. Brandenburg, V.M.; Reinartz, S.; Kaesler, N.; Kruger, T.; Dirrichs, T.; Kramann, R.; Peeters, F.; Floege, J.; Keszei, A.; Marx, N.; et al. Slower Progress of Aortic Valve Calcification with Vitamin K Supplementation: Results from a Prospective Interventional Proof-of-Concept Study. Circulation 2017, 135, 2081–2083. [Google Scholar] [CrossRef]
  104. Pivin, E.; Ponte, B.; Pruijm, M.; Ackermann, D.; Guessous, I.; Ehret, G.; Liu, Y.P.; Drummen, N.E.; Knapen, M.H.; Pechere-Bertschi, A.; et al. Inactive Matrix Gla-Protein Is Associated with Arterial Stiffness in an Adult Population-Based Study. Hypertension 2015, 66, 85–92. [Google Scholar] [CrossRef] [Green Version]
  105. Harshman, S.G.; Shea, M.K. The Role of Vitamin K in Chronic Aging Diseases: Inflammation, Cardiovascular Disease, and Osteoarthritis. Curr. Nutr. Rep. 2016, 5, 90–98. [Google Scholar] [CrossRef] [Green Version]
  106. Nagata, C.; Wada, K.; Tamura, T.; Konishi, K.; Goto, Y.; Koda, S.; Kawachi, T.; Tsuji, M.; Nakamura, K. Dietary soy and natto intake and cardiovascular disease mortality in Japanese adults: The Takayama study. Am. J. Clin. Nutr. 2017, 105, 426–431. [Google Scholar] [CrossRef] [Green Version]
  107. Caluwe, R.; Pyfferoen, L.; De Boeck, K.; De Vriese, A.S. The effects of vitamin K supplementation and vitamin K antagonists on progression of vascular calcification: Ongoing randomized controlled trials. Clin. Kidney J. 2016, 9, 273–279. [Google Scholar] [CrossRef]
  108. Danziger, J.; Young, R.L.; Shea, K.M.; Duprez, D.A.; Jacobs, D.R.; Tracy, R.P.; Ix, J.H.; Jenny, N.S.; Mukamal, K.J. Circulating Des-gamma-carboxy prothrombin is not associated with cardiovascular calcification or stiffness: The Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis 2016, 252, 68–74. [Google Scholar] [CrossRef]
  109. Danziger, J.; Young, R.L.; Shea, M.K.; Tracy, R.P.; Ix, J.H.; Jenny, N.S.; Mukamal, K.J. Vitamin K-Dependent Protein Activity and Incident Ischemic Cardiovascular Disease: The Multi-Ethnic Study of Atherosclerosis. Arter. Thromb. Vasc. Biol. 2016, 36, 1037–1042. [Google Scholar] [CrossRef] [Green Version]
  110. Ishida, Y.; Kawai, S. Comparative efficacy of hormone replacement therapy, etidronate, calcitonin, alfacalcidol, and vitamin K in postmenopausal women with osteoporosis: The Yamaguchi Osteoporosis Prevention Study. Am. J. Med. 2004, 117, 549–555. [Google Scholar] [CrossRef]
  111. Hofbauer, L.C.; Brueck, C.C.; Singh, S.K.; Dobnig, H. Osteoporosis in patients with diabetes mellitus. J. Bone Min. Res. 2007, 22, 1317–1328. [Google Scholar] [CrossRef]
  112. Shiraki, M.; Yamazaki, Y.; Shiraki, Y.; Hosoi, T.; Tsugawa, N.; Okano, T. High level of serum undercarboxylated osteocalcin in patients with incident fractures during bisphosphonate treatment. J. Bone Min. Metab. 2010, 28, 578–584. [Google Scholar] [CrossRef]
  113. Palermo, A.; Tuccinardi, D.; D′Onofrio, L.; Watanabe, M.; Maggi, D.; Maurizi, A.R.; Greto, V.; Buzzetti, R.; Napoli, N.; Pozzilli, P.; et al. Vitamin K and osteoporosis: Myth or reality? Metabolism 2017, 70, 57–71. [Google Scholar] [CrossRef] [Green Version]
  114. Iwamoto, J.; Seki, A.; Sato, Y.; Matsumoto, H.; Takeda, T.; Yeh, J.K. Vitamin K2 prevents hyperglycemia and cancellous osteopenia in rats with streptozotocin-induced type 1 diabetes. Calcif. Tissue Int. 2011, 88, 162–168. [Google Scholar] [CrossRef]
  115. Iwamoto, J.; Takeda, T.; Ichimura, S. Effect of combined administration of vitamin D3 and vitamin K2 on bone mineral density of the lumbar spine in postmenopausal women with osteoporosis. J. Orthop. Sci. 2000, 5, 546–551. [Google Scholar] [CrossRef]
  116. Shiraki, M.; Shiraki, Y.; Aoki, C.; Miura, M. Vitamin K2 (menatetrenone) effectively prevents fractures and sustains lumbar bone mineral density in osteoporosis. J. Bone Min. Res. 2000, 15, 515–521. [Google Scholar] [CrossRef]
  117. Iwamoto, J.; Takeda, T.; Ichimura, S. Effect of menatetrenone on bone mineral density and incidence of vertebral fractures in postmenopausal women with osteoporosis: A comparison with the effect of etidronate. J. Orthop. Sci. 2001, 6, 487–492. [Google Scholar] [CrossRef]
  118. Inoue, T.; Fujita, T.; Kishimoto, H.; Makino, T.; Nakamura, T.; Nakamura, T.; Sato, T.; Yamazaki, K. Randomized controlled study on the prevention of osteoporotic fractures (OF study): A phase IV clinical study of 15-mg menatetrenone capsules. J. Bone Min. Metab. 2009, 27, 66–75. [Google Scholar] [CrossRef]
  119. Iwamoto, J. Vitamin K2 therapy for postmenopausal osteoporosis. Nutrients 2014, 6, 1971–1980. [Google Scholar] [CrossRef] [Green Version]
  120. Beulens, J.W.; Booth, S.L.; van den Heuvel, E.G.; Stoecklin, E.; Baka, A.; Vermeer, C. The role of menaquinones (vitamin K(2)) in human health. Br. J. Nutr. 2013, 110, 1357–1368. [Google Scholar] [CrossRef] [Green Version]
  121. Vissers, L.E.; Dalmeijer, G.W.; Boer, J.M.; Monique Verschuren, W.M.; van der Schouw, Y.T.; Beulens, J.W. Intake of dietary phylloquinone and menaquinones and risk of stroke. J. Am. Heart Assoc. 2013, 2, e000455. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Vitamin K status of male Wistar rat tissues. Rats were fed a standard AIN-93G rodent diet for three weeks. The levels of vitamin K in tissues were determined using fluorescent high-performance liquid chromatography. MK-4: menaquinone-4. (Reproduced from Ref. [17]).
Figure 1. Vitamin K status of male Wistar rat tissues. Rats were fed a standard AIN-93G rodent diet for three weeks. The levels of vitamin K in tissues were determined using fluorescent high-performance liquid chromatography. MK-4: menaquinone-4. (Reproduced from Ref. [17]).
Nutrients 12 02485 g001
Figure 2. Schematic illustration of the plausible mechanisms of VK on insulin response and glycemic status. MGP: matrix Gla protein; OC: osteocalcin; TNF-α: tumor necrosis factor α, IL: interleukin.
Figure 2. Schematic illustration of the plausible mechanisms of VK on insulin response and glycemic status. MGP: matrix Gla protein; OC: osteocalcin; TNF-α: tumor necrosis factor α, IL: interleukin.
Nutrients 12 02485 g002
Table 1. Summarization of findings from studies on the effect of vitamin K on insulin sensitivity and glycemic status.
Table 1. Summarization of findings from studies on the effect of vitamin K on insulin sensitivity and glycemic status.
Subjects (N)VK dose/VK StatusPeriodOutcomeRef.
A. Human Studies
(a) Observational Studies
Healthy young men (16)Usual dietary intakeAcute insulin response
A 1-week food-frequency questionnaire to ascertain the daily VK intake
The participants with higher dietary VK intake showed a better insulin response and glucose tolerance.[18]
Framingham offspring cohort study, adult men (1247) and women (1472)Usual dietary intake12 monthsIn a cross-sectional analysis, higher dietary VK intake was associated with reduced insulin resistance in both adult men and women.[19]
Adult men (9740) and women (28,354)Usual dietary intake10.3 yearsDietary intake of both VK1 and VK2 were associated with a reduced risk of T2DM.[20]
Elderly men (861) and women (1062) with high cardiovascular riskUsual dietary intakeMedian follow-up of 5.5 yearsDietary VK1 at the baseline was significantly lower in participants who developed T2DM during the study. Increased dietary VK1 intake was associated with a reduced risk of incident T2DM.[21]
European Prospective Investigation into Cancer and Nutrition (EPIC) cohort study, Diabetes Genetics Replication and Meta-analysis (DIAGRAM), and the UK Biobank (9400 case subjects and 12,182 sub-cohort participants)Usual dietary intakeEPIC cohort: 1997–2007, DIAGRAM cohort: 2007 (included data from 23 studies), UK Biobank: 2006–2010Higher circulating VK1 may be causally related with lower risk of T2DM, highlighting the importance of sufficient phylloquinone intake in the human diet.[22]
(b) Intervention Studies
Elderly nondiabetic men (124) and women (165)With or without 500 μg/day VK1 supplementation36 monthsDietary VK1 supplementation had a protective effect on the progression of insulin resistance in older men.[23]
Prediabetic women (82)With or without 1000 μg/day VK1 supplementation4 weeksDietary VK1 supplementation had beneficial effects on glycemic status and insulin sensitivity in premenopausal and prediabetic women.[24,25]
Healthy young men (12)90 mg/day menaquinone-4 (MK-4) supplementation1 weekShort-term VK2 supplementation improved the insulin response after an oral glucose challenge in young men.[26]
Healthy young men (42)With or without 90 mg/day MK-4 supplementation4 weeksDietary VK2 supplementation improved insulin sensitivity in young men.[27]
B. Animal Studies
Rats (unknown)Low-VK diet (<20% of the required VK1)UnknownRats fed a low-VK diet had poor early insulin response and subsequently increased insulin secretion after a glucose load.[28]
Arteriosclerotic rat model with DM (unknown)100 mg/day per kilogram body weight VK23 or 6 weeksVK2 supplementation had a protective effect on arteriosclerosis, by decreasing the aortic Ca and P and the elastin fraction. Rats fed a VK2-rich diet had decreased serum glucose levels and increased serum insulin levels.[29]

Share and Cite

MDPI and ACS Style

Ho, H.-J.; Komai, M.; Shirakawa, H. Beneficial Effects of Vitamin K Status on Glycemic Regulation and Diabetes Mellitus: A Mini-Review. Nutrients 2020, 12, 2485. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12082485

AMA Style

Ho H-J, Komai M, Shirakawa H. Beneficial Effects of Vitamin K Status on Glycemic Regulation and Diabetes Mellitus: A Mini-Review. Nutrients. 2020; 12(8):2485. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12082485

Chicago/Turabian Style

Ho, Hsin-Jung, Michio Komai, and Hitoshi Shirakawa. 2020. "Beneficial Effects of Vitamin K Status on Glycemic Regulation and Diabetes Mellitus: A Mini-Review" Nutrients 12, no. 8: 2485. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12082485

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