1
|
Kij A, Kieronska-Rudek A, Bar A, Czyzynska-Cichon I, Strus M, Kozien L, Wiecek G, Zeber-Lubecka N, Kulecka M, Kwiatkowski G, Przyborowski K, Mohaissen T, Sternak M, Buczek E, Zakrzewska A, Proniewski B, Kus K, Franczyk-Zarow M, Kostogrys RB, Pieterman EJ, Princen HMG, Chlopicki S. Low phylloquinone intake deteriorates endothelial function in normolipidemic and dyslipidaemic mice. J Nutr Biochem 2025; 140:109867. [PMID: 39978646 DOI: 10.1016/j.jnutbio.2025.109867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 01/10/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
While the plasma phylloquinone (PK) concentration is inversely correlated with cardiovascular risk, the involvement of PK in regulating endothelial function has not been directly investigated. Therefore, in this study we assessed the effects of short-term treatment with PK-deficient diets (5-10 weeks) on endothelial function in normolipidemic 14-week-old male C57BL/6JCmd mice and age-matched dyslipidaemic male E3L.CETP mice. Our results show that in normolipidemic mice dietary PK deficiency was associated with a marked reduction of PK levels in the plasma and liver (liquid chromatography-mass spectrometry measurements) and with impaired endothelium-dependent vasodilation assessed in vivo by magnetic resonance imaging (MRI). Dietary PK deficiency-induced endothelial dysfunction was fully reversed by PK supplementation. In dyslipidaemic E3L.CETP mice, dietary PK deficiency exacerbated preexisting endothelial dysfunction. Furthermore, dietary PK deficiency decreased menaquinone-4 (MK-4) levels in the aorta but did not affect blood coagulation (calibrated automated thrombography), microbiota composition (culturing and next-generation sequencing), and gut menaquinone production. In conclusion, our study demonstrated for the first time that sufficient dietary PK intake supports endothelial function in normolipidemic and dyslipidaemic mice indicating nutritional significance of dietary PK in the maintenance of endothelial function in humans.
Collapse
Affiliation(s)
- Agnieszka Kij
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Anna Kieronska-Rudek
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland; Jagiellonian University Medical College, Krakow, Poland
| | - Anna Bar
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Izabela Czyzynska-Cichon
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Magdalena Strus
- Jagiellonian University Medical College, Department of Bacteriology, Microbial Ecology and Parasitology, Krakow, Poland
| | - Lucja Kozien
- Jagiellonian University Medical College, Department of Bacteriology, Microbial Ecology and Parasitology, Krakow, Poland
| | - Grazyna Wiecek
- Jagiellonian University Medical College, Department of Bacteriology, Microbial Ecology and Parasitology, Krakow, Poland
| | - Natalia Zeber-Lubecka
- Centre of Postgraduate Medical Education Department of Gastroenterology, Hepatology and Clinical Oncology, Warszawa, Poland; Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, Poland
| | - Maria Kulecka
- Centre of Postgraduate Medical Education Department of Gastroenterology, Hepatology and Clinical Oncology, Warszawa, Poland; Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, Poland
| | - Grzegorz Kwiatkowski
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Kamil Przyborowski
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Tasnim Mohaissen
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland; University of Copenhagen, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Magdalena Sternak
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Elzbieta Buczek
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Agnieszka Zakrzewska
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Bartosz Proniewski
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Kamil Kus
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland
| | - Magdalena Franczyk-Zarow
- University of Agriculture in Krakow, Faculty of Food Technology, Department of Human Nutrition and Dietetics, Krakow, Poland
| | - Renata B Kostogrys
- University of Agriculture in Krakow, Faculty of Food Technology, Department of Human Nutrition and Dietetics, Krakow, Poland
| | - Elsbeth J Pieterman
- The Netherlands Organization of Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Hans M G Princen
- The Netherlands Organization of Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Stefan Chlopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Krakow, Poland; Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
2
|
Turck D, Bohn T, Castenmiller J, de Henauw S, Hirsch-Ernst KI, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Traber MG, Vrolijk M, Bercovici CM, de Sesmaisons Lecarré A, Fabiani L, Karavasiloglou N, Mendes V, Valtueña Martínez S, Naska A. Scientific opinion on the tolerable upper intake level for vitamin E. EFSA J 2024; 22:e8953. [PMID: 39099617 PMCID: PMC11294871 DOI: 10.2903/j.efsa.2024.8953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the revision of the tolerable upper intake level (UL) for vitamin E. As α-tocopherol is recognised as the only essential form of vitamin E, the Panel restricted its evaluation to α-tocopherol. Systematic reviews of the literature were conducted to assess evidence on priority adverse health effects of excess intake of vitamin E, namely risk of impaired coagulation and bleeding, cardiovascular disease and prostate cancer. The effect on blood clotting and associated increased risk of bleeding is considered as the critical effect to establish an UL for vitamin E. No new evidence has been published that could improve the characterisation of a dose-response. The ULs for vitamin E from all dietary sources, which were previously established by the Scientific Committee on Food, are retained for all population groups, i.e. 300 mg/day for adults, including pregnant and lactating women, 100 mg/day for children aged 1-3 years, 120 mg/day for 4-6 years, 160 mg/day for 7-10 years, 220 mg/day for 11-14 years and 260 mg/day for 15-17 years. A UL of 50 mg/day is established for infants aged 4-6 months and a UL of 60 mg/day for infants aged 7-11 months. ULs apply to all stereoisomeric forms of α-tocopherol. ULs do not apply to individuals receiving anticoagulant or antiplatelet medications (e.g. aspirin), to patients on secondary prevention for CVD or to patients with vitamin K malabsorption syndromes. It is unlikely that the ULs for vitamin E are exceeded in European populations, except for regular users of food supplements containing high doses of vitamin E.
Collapse
|
3
|
Wei Y, He Q, Zhu H, Song Y, Liu L, Sun Y, Chen P, Wang B. A Negative Association between Plasma Phylloquinone and All-Cause Mortality in Chinese Adults with Hypertension: A Nested Case-Control Study. J Nutr 2024; 154:978-984. [PMID: 38092150 DOI: 10.1016/j.tjnut.2023.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/21/2023] [Accepted: 12/05/2023] [Indexed: 12/31/2023] Open
Abstract
BACKGROUND Previous studies have revealed that vitamin K is essential for preventing various chronic diseases. Phylloquinone is the primary dietary and circulating form of vitamin K. However, data concerning the association between plasma phylloquinone and all-cause mortality are limited. OBJECTIVES This study aimed to evaluate the association between plasma phylloquinone and risk of all-cause mortality and examine some potential confounders. METHODS This study is a post hoc analysis of the RCT and a nested, case-control design was used. Enrolled participants had to have hypertension at baseline. Study initiation was 19 May, 2008, and the median follow-up was 4.5 y. A total of 604 mortality cases and 604 controls matched for age, sex, treatment group, and study site were included in this study. Odds ratios (OR) and 95% confidence intervals (CIs) of all-cause mortality were calculated using conditional or unconditional logistic regression, without or with adjusting for pertinent covariates, respectively. The concentration of phylloquinone was measured by liquid chromatography-tandem quadrupole mass spectrometry (LC-MS/MS). RESULTS The mean and median phylloquinone levels were 1.62 nmol/L and 0.89 nmol/L, respectively. There was a significant negative association between log-transformed plasma phylloquinone and all-cause mortality after controlling for potential confounders (per 1 unit increase-OR: 0.79; 95% CI: 0.66, 0.95). Furthermore, the association of plasma phylloquinone with risk of all-cause mortality differed by body mass index (BMI) (<25 kg/m2 compared with ≥25 kg/m2, P-interaction = 0.004). A significant trend of decreasing risk with increasing concentration of phylloquinone was observed in participants with higher BMI (per 1 unit increase-OR: 0.71; 95% CI: 0.56, 0.90; P = 0.004). No significant correlation was found between phylloquinone and risk of all-cause mortality in those with BMI <25 kg/m2. CONCLUSIONS In Chinese patients with hypertension, there was a significant negative association between baseline plasma phylloquinone and all-cause mortality, especially among those with higher BMI.
Collapse
Affiliation(s)
- Yaping Wei
- College of Public Health, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Qiangqiang He
- Shenzhen International Graduate School, Tsinghua University, Shenzhen, China; Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Hehao Zhu
- School of Science, China Pharmaceutical University, Nanjing, China
| | - Yun Song
- Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Lishun Liu
- Guangdong Key Laboratory of H-type Hypertension and Stroke Precision Prevention Research and Development Enterprise, Shenzhen, China
| | - Yong Sun
- People's Hospital of Lianyungang City/The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Ping Chen
- Guangdong Key Laboratory of H-type Hypertension and Stroke Precision Prevention Research and Development Enterprise, Shenzhen, China
| | - Binyan Wang
- Shenzhen Evergreen Medical Institute, Shenzhen, China.
| |
Collapse
|
4
|
Shea MK, Booth SL. Vitamin K Status and All-Cause Mortality: Is there a link? J Nutr 2024; 154:799-800. [PMID: 38246356 DOI: 10.1016/j.tjnut.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/07/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Affiliation(s)
- M Kyla Shea
- USDA Human Nutrition Research Center on Aging at Tufts University, Boston MA, United States
| | - Sarah L Booth
- USDA Human Nutrition Research Center on Aging at Tufts University, Boston MA, United States.
| |
Collapse
|
5
|
Aaseth JO, Alehagen U, Opstad TB, Alexander J. Vitamin K and Calcium Chelation in Vascular Health. Biomedicines 2023; 11:3154. [PMID: 38137375 PMCID: PMC10740993 DOI: 10.3390/biomedicines11123154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
The observation that the extent of artery calcification correlates with the degree of atherosclerosis was the background for the alternative treatment of cardiovascular disease with chelator ethylenediamine tetraacetate (EDTA). Recent studies have indicated that such chelation treatment has only marginal impact on the course of vascular disease. In contrast, endogenous calcium chelation with removal of calcium from the cardiovascular system paralleled by improved bone mineralization exerted, i.e., by matrix Gla protein (MGP) and osteocalcin, appears to significantly delay the development of cardiovascular diseases. After post-translational vitamin-K-dependent carboxylation of glutamic acid residues, MGP and other vitamin-K-dependent proteins (VKDPs) can chelate calcium through vicinal carboxyl groups. Dietary vitamin K is mainly provided in the form of phylloquinone from green leafy vegetables and as menaquinones from fermented foods. Here, we provide a review of clinical studies, addressing the role of vitamin K in cardiovascular diseases, and an overview of vitamin K kinetics and biological actions, including vitamin-K-dependent carboxylation and calcium chelation, as compared with the action of the exogenous (therapeutic) chelator EDTA. Consumption of vitamin-K-rich foods and/or use of vitamin K supplements appear to be a better preventive strategy than EDTA chelation for maintaining vascular health.
Collapse
Affiliation(s)
- Jan O. Aaseth
- Research Department, Innlandet Hospital Trust, P.O. Box 104, N-2381 Brumunddal, Norway
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, P.O. Box 400, N-2418 Elverum, Norway
| | - Urban Alehagen
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, SE-581 85 Linköping, Sweden;
| | - Trine Baur Opstad
- Oslo Centre for Clinical Heart Research Laboratory, Department of Cardiology, Oslo University Hospital Ullevål, P.O. Box 4950, Nydalen, N-0424 Oslo, Norway;
- Faculty of Medicine, University of Oslo, N-0370 Oslo, Norway
| | - Jan Alexander
- Norwegian Institute of Public Health, P.O. Box 222, N-0213 Oslo, Norway;
| |
Collapse
|
6
|
Cozzolino M, Maffei Faccioli F, Cara A, Boni Brivio G, Rivela F, Ciceri P, Magagnoli L, Galassi A, Barbuto S, Speciale S, Minicucci C, Cianciolo G. Future treatment of vascular calcification in chronic kidney disease. Expert Opin Pharmacother 2023; 24:2041-2057. [PMID: 37776230 DOI: 10.1080/14656566.2023.2266381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/29/2023] [Indexed: 10/02/2023]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is one of the global leading causes of morbidity and mortality in chronic kidney disease (CKD) patients. Vascular calcification (VC) is a major cause of CVD in this population and is the consequence of complex interactions between inhibitor and promoter factors leading to pathological deposition of calcium and phosphate in soft tissues. Different pathological landscapes are associated with the development of VC, such as endothelial dysfunction, oxidative stress, chronic inflammation, loss of mineralization inhibitors, release of calcifying extracellular vesicles (cEVs) and circulating calcifying cells. AREAS COVERED In this review, we examined the literature and summarized the pathophysiology, biomarkers and focused on the treatments of VC. EXPERT OPINION Even though there is no consensus regarding specific treatment options, we provide the currently available treatment strategies that focus on phosphate balance, correction of vitamin D and vitamin K deficiencies, avoidance of both extremes of bone turnover, normalizing calcium levels and reduction of inflammatory response and the potential and promising therapeutic approaches liketargeting cellular mechanisms of calcification (e.g. SNF472, TNAP inhibitors).Creating novel scores to detect in advance VC and implementing targeted therapies is crucial to treat them and improve the future management of these patients.
Collapse
Affiliation(s)
- Mario Cozzolino
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Federico Maffei Faccioli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Anila Cara
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Giulia Boni Brivio
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Francesca Rivela
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Paola Ciceri
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Lorenza Magagnoli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Andrea Galassi
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Simona Barbuto
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Serena Speciale
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Carlo Minicucci
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giuseppe Cianciolo
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
7
|
Ma Y, Yu S, Mu D, Cheng J, Qiu L, Cheng X. Liquid chromatography-tandem mass spectrometry in fat-soluble vitamin deficiency. Clin Chim Acta 2023; 548:117469. [PMID: 37419302 DOI: 10.1016/j.cca.2023.117469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Fat-soluble vitamins, including vitamins A, D, E, and K, are essential for maintaining normal body function and metabolism. Fat-soluble vitamin deficiency may lead to bone diseases, anemia, bleeding, xerophthalmia, etc. Early detection and timely interventions are significant for preventing vitamin deficiency-related diseases. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is developing into a potent instrument for the precise detection of fat-soluble vitamins due to its high sensitivity, high specificity, and high resolution.
Collapse
Affiliation(s)
- Yichen Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Songlin Yu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Danni Mu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Jin Cheng
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Xinqi Cheng
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China.
| |
Collapse
|
8
|
Golüke NM, Schoffelmeer MA, De Jonghe A, Emmelot-Vonk MH, De Jong PA, Koek HL. Serum biomarkers for arterial calcification in humans: A systematic review. Bone Rep 2022; 17:101599. [PMID: 35769144 PMCID: PMC9234354 DOI: 10.1016/j.bonr.2022.101599] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2022] Open
Abstract
Aim To clarify the role of mediators of ectopic mineralization as biomarkers for arterial calcifications. Methods MEDLINE and Embase were searched for relevant literature, until January 4th 2022. The investigated biomarkers were: calcium, phosphate, parathyroid hormone, vitamin D, pyrophosphate, osteoprotegerin, receptor activator of nuclear factor-kappa B ligand (RANKL), fibroblast growth factor-23 (FGF-23), Klotho, osteopontin, osteocalcin, Matrix Gla protein (MGP) and its inactive forms and vitamin K. Studies solely performed in patients with kidney insufficiency or diabetes mellitus were excluded. Results After screening of 8985 articles, a total of 129 articles were included in this systematic review. For all biomarkers included in this review, the results were variable and more than half of the studies for each specific biomarker had a non-significant result. Also, the overall quality of the included studies was low, partly as a result of the mostly cross-sectional study designs. The largest body of evidence is available for phosphate, osteopontin and FGF-23, as a little over half of the studies showed a significant, positive association. Firm statements for these biomarkers cannot be drawn, as the number of studies was limited and hampered by residual confounding or had non-significant results. The associations of the other mediators of ectopic mineralization with arterial calcifications were not clear. Conclusion Associations between biomarkers of ectopic mineralization and arterial calcification are variable in the published literature. Future longitudinal studies differentiating medial and intimal calcification could add to the knowledge of biomarkers and mechanisms of arterial calcifications. We researched the association between biomarkers and arterial calcifications. This review focused on biomarkers of bone metabolism and Matrix Gla protein. Associations between biomarkers and arterial calcification are variable. Future studies should differentiate between medial and intimal calcifications.
Collapse
Key Words
- 1,25(OH)2D, 1,25-dihydroxyvitamin D
- 25(OH)D, 25-hydroxyvitamin D
- Arterial calcification
- Biomarkers
- CAC, coronary artery calcification
- CAD, coronary artery disease
- CVD, cardiovascular disease
- FGF-23, fibroblast growth factor-23
- GACI, generalized arterial calcification of infancy
- MGP, matrix Gla protein
- MK, menaquinone
- OPG, osteoprotegerin
- PIVKA-2, protein induced by vitamin K absence or antagonist-2
- PK, phylloquinone
- PTH, parathyroid hormone
- PXE, pseudoxanthoma elasticum
- RANKL, receptor activator of nuclear factor-kappa B ligand
- Review
- dp-cMGP, carboxylated but dephosphorylated MGP
- dp-ucMGP, uncarboxylated an dephosphorylated MGP
- uc-MGP, uncarboxylated MGP
Collapse
Affiliation(s)
- Nienke M.S. Golüke
- University Medical Center Utrecht, Department of Geriatrics, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
- Tergooi Hospitals, Department of Geriatrics, Rijksstraatweg 1, 1261 AN Blaricum, the Netherlands
- Corresponding author at: Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| | - Marit A. Schoffelmeer
- University Medical Center Utrecht, Department of Geriatrics, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Annemarieke De Jonghe
- Tergooi Hospitals, Department of Geriatrics, Rijksstraatweg 1, 1261 AN Blaricum, the Netherlands
| | - Mariëlle H. Emmelot-Vonk
- University Medical Center Utrecht, Department of Geriatrics, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Pim A. De Jong
- University Medical Center Utrecht, Department of Radiology, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Huiberdina L. Koek
- University Medical Center Utrecht, Department of Geriatrics, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| |
Collapse
|
9
|
Pinto G, Fragasso G. Aortic valve stenosis: drivers of disease progression and drug targets for therapeutic opportunities. Expert Opin Ther Targets 2022; 26:633-644. [DOI: 10.1080/14728222.2022.2118576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Giuseppe Pinto
- Departmen of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Gabriele Fragasso
- Department of Clinical Cardiology, Heart Failure Clinic, IRCCS San Raffaele Scientific Institute, Milano
| |
Collapse
|
10
|
Wei Y, Wang Z, He Q, Siddiqi SM, Zhou Z, Liu L, Song Y, Chen P, Li J, Zhang Y, Mao G, Wang B, Tang G, Qin X, Xu X, Huo Y, Guo H, Zhang H. Inverse Association between Plasma Phylloquinone and Risk of Ischemic Stroke in Chinese Adults with Hypertension and High BMI: A Nested Case-Control Study. J Nutr 2022; 152:1927-1935. [PMID: 35660920 DOI: 10.1093/jn/nxac131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Evidence on the association between phylloquinone status and cardiovascular diseases is scarce and conflicting. These inconsistencies may be due to differences in individual characteristics of the study populations, which may modify the association. OBJECTIVE This study aimed to evaluate the association between plasma phylloquinone and the risk of first total stroke and its subtypes, and to examine potential effect modifications by BMI in patients with hypertension. METHODS We performed a nested case-control study including 604 first stroke cases and 604 matched controls. The mean age was 62.2 y (range, 45 to 75). Lower BMI was defined as <25 kg/m2 and higher BMI was defined as ≥25 kg/m2. The risks of the first stroke were estimated by ORs and 95% CIs using conditional logistic regression. The primary outcome was total stroke or ischemic stroke. RESULTS The relation between log-transformed phylloquinone concentration and stroke or ischemic stroke was modified by BMI. Higher phylloquinone concentrations were associated with lower stroke risk in those with a higher BMI. When plasma phylloquinone was assessed as tertiles, the adjusted ORs of first stroke and ischemic stroke for participants with a high BMI in tertile 2-3 were 0.70 (95% CI: 0.46, 1.08) and 0.57 (95% CI: 0.35, 0.92) compared with those in tertile 1, respectively. However, there was no significant association between plasma phylloquinone and risk of first total stroke or ischemic stroke for those with a lower BMI. Patients with a higher BMI and lower phylloquinone concentrations had the highest risk of ischemic stroke and showed a statistically significant difference compared with the reference group with a lower BMI and higher phylloquinone (OR = 1.80, 95% CI: 1.06, 3.10; P-interaction: 0.017). CONCLUSIONS In Chinese patients with hypertension, there was an inverse association between baseline plasma phylloquinone and risk of first ischemic stroke among those with a higher BMI. This trial was registered at clinicaltrials.gov as NCT00794885.
Collapse
Affiliation(s)
- Yaping Wei
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zhuo Wang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qiangqiang He
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China.,Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Sultan Mehmood Siddiqi
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China.,Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Ziyi Zhou
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China.,Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Lishun Liu
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China.,Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Yun Song
- Shenzhen Evergreen Medical Institute, Shenzhen, China.,Institute for Biomedicine, Anhui Medical University, Hefei, China
| | - Ping Chen
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Guangyun Mao
- Department of Preventive Medicine, School of Public Health & Management, Wenzhou Medical University, Wenzhou, China
| | - Binyan Wang
- Institute for Biomedicine, Anhui Medical University, Hefei, China
| | - Genfu Tang
- School of Health Administration, Anhui Medical University, Hefei, China
| | - Xianhui Qin
- National Clinical Research Center for Kidney Disease, State Key Laboratory for Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiping Xu
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Huiyuan Guo
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Hao Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
11
|
Wei Y, Ma H, Xu B, Wang Z, He Q, Liu L, Zhou Z, Song Y, Chen P, Li J, Zhang Y, Mao G, Wang B, Tang G, Qin X, Zhang H, Xu X, Huo Y, Guo H. Joint Association of Low Vitamin K1 and D Status With First Stroke in General Hypertensive Adults: Results From the China Stroke Primary Prevention Trial (CSPPT). Front Neurol 2022; 13:881994. [PMID: 35645985 PMCID: PMC9135055 DOI: 10.3389/fneur.2022.881994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 11/28/2022] Open
Abstract
Background Vitamin K plays a role in preventing vascular calcification and may have a synergetic influence with vitamin D on cardiovascular health. However, whether this relationship applies to stroke, especially in a high-risk population of hypertensive individuals, remains unclear. The present study aims to study the joint association of low vitamin K1 and D status with first stroke in general hypertensive adults. Methods This study used a nested, case-control design with data from the China Stroke Primary Prevention Trial. The analysis included 604 first total stroke patients and 604 matched controls from a Chinese population with hypertension. Odds ratios (ORs) and 95% confidence intervals were calculated using conditional logistic regression. Results There was a non-linear negative association between plasma vitamin K1 and the risk of first total stroke or ischemic stroke in the enalapril-only group. Compared to participants in vitamin K1 quartile 1, a significantly lower risk of total stroke (OR = 0.58, 95% CI: 0.36, 0.91, P = 0.020) or ischemic stroke (OR = 0.34, 95% CI: 0.17, 0.63, P < 0.001) was found in participants in vitamin K1 quartile 2-4 in the enalapril-only group. When further divided into four subgroups by 25(OH)D and vitamin K1, a significantly higher risk of total stroke or ischemic stroke was observed in participants with both low vitamin K1 and 25(OH)D compared to those with both high vitamin K1 and 25(OH)D in the enalapril-only group. No increased risk was observed in the groups low in one vitamin only. Conclusion Low concentrations of both vitamin K1 and 25(OH)D were associated with increased risk of stroke.
Collapse
Affiliation(s)
- Yaping Wei
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Hai Ma
- Rongcheng Center for Disease Control and Prevention, Rongcheng, China
| | - Benjamin Xu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Zhuo Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qiangqiang He
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
- Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Lishun Liu
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
- Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Ziyi Zhou
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
- Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Yun Song
- Shenzhen Evergreen Medical Institute, Shenzhen, China
- Institute for Biomedicine, Anhui Medical University, Hefei, China
| | - Ping Chen
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Guangyun Mao
- Department of Preventive Medicine, School of Public Health & Management, Wenzhou Medical University, Wenzhou, China
| | - Binyan Wang
- Institute for Biomedicine, Anhui Medical University, Hefei, China
| | - Genfu Tang
- Institute for Biomedicine, Anhui Medical University, Hefei, China
| | - Xianhui Qin
- National Clinical Research Center for Kidney Disease, State Key Laboratory for Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiping Xu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
- National Clinical Research Center for Kidney Disease, State Key Laboratory for Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Huiyuan Guo
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
12
|
Xu C, Cunqing Y, Chun G, Min W, Jun L, Xueyun H, Jiaxin F, Li S, Cheng A, Guijian L, Fengxiang S, Bo P. The relationship between serum vitamin K concentration and coronary artery calcification in middle-aged and elderly people. Clin Chim Acta 2022; 531:325-330. [PMID: 35504322 DOI: 10.1016/j.cca.2022.04.1001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/14/2022] [Accepted: 04/27/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Vitamin K is involved in the formation of coronary artery calcification which is an independent predictor of coronary heart disease. This study aims to explore the association between coronary artery calcification score and serum concentrations of vitamin K1, menaquinone-4 (MK-4) and menaquinone-7 (MK-7) in middle-aged and elderly Chinese population. METHODS A total of 116 patients who underwent CT coronary angiography were consecutively enrolled. Serum concentrations of vitamin K1, MK-4 and MK-7 were determined by high performance liquid chromatography tandem mass spectrometry. The relationships between coronary artery calcification score and serum vitamin K concentrations were analyzed. RESULTS Significantly lower serum vitamin K1 concentration was found in the patients with CACS > 400, comparing with the other CACS categories, respectively. Log (CACS + 1) was significantly higher in MK-4 < 0.05 ng/ml group compared with MK-4 ≥ 0.05 ng/ml group [2.03(0.21, 2.58) vs 1.31(0.00, 2.19), P < 0.05]. In subjects with established coronary calcification (defined as CACS > 10), vitamin K1 was found to be an independent factor contributing to higher CACS (r = -0.288, P = 0.013). CONCLUSIONS In this retrospective analysis, serum vitamin K1 and MK-4 concentrations were significantly lower in middle-aged and elderly cohorts with increasing calcification scores. The significant effect of vitamin K1 on CACS was only found in individuals who already had calcification. Whether the detection of circulating vitamin K in patients with preexisting coronary calcification could guide vitamin K supplementation needs further exploration.
Collapse
Affiliation(s)
- Cheng Xu
- Clinical Laboratory, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Yang Cunqing
- Clinical Laboratory, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Gu Chun
- Clinical Laboratory, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Wu Min
- General Internal Department, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Li Jun
- Department of Cardiology, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Hou Xueyun
- Clinical Laboratory, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Fei Jiaxin
- Clinical Laboratory, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Sun Li
- Department of Radiology, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - An Cheng
- Clinical Laboratory, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Liu Guijian
- Clinical Laboratory, Guang' anmen Hospital, China academy of Chinese medical sciences, China
| | - Shi Fengxiang
- Department of Radiology, Guang' anmen Hospital, China academy of Chinese medical sciences, China.
| | - Pang Bo
- Clinical Laboratory, Guang' anmen Hospital, China academy of Chinese medical sciences, China.
| |
Collapse
|
13
|
Shea MK, Barger K, Booth SL, Wang J, Feldman HI, Townsend RR, Chen J, Flack J, He J, Jaar BG, Kansal M, Rosas SE, Weiner DE. Vitamin K status, all-cause mortality, and cardiovascular disease in adults with chronic kidney disease: the Chronic Renal Insufficiency Cohort. Am J Clin Nutr 2022; 115:941-948. [PMID: 34788785 PMCID: PMC8895220 DOI: 10.1093/ajcn/nqab375] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Vascular calcification contributes to cardiovascular disease (CVD) and mortality in individuals with chronic kidney disease (CKD). Vitamin K-dependent proteins function as calcification inhibitors in vascular tissue. OBJECTIVES We sought to determine the association of vitamin K status with mortality and CVD events in adults with CKD. METHODS Plasma dephospho-uncarboxylated matrix gla protein ((dp)ucMGP), which increases when vitamin K status is low, and plasma phylloquinone (vitamin K1), which decreases when vitamin K status is low, were measured in 3066 Chronic Renal Insufficiency Cohort participants (median age = 61 y, 45% female, 41% non-Hispanic black, median estimated glomerular filtration rate [eGFR] = 41 mL/min/1.73m2). The association of vitamin K status biomarkers with all-cause mortality and atherosclerotic-related CVD was determined using multivariable Cox proportional hazards regression. RESULTS There were 1122 deaths and 599 atherosclerotic CVD events over the median 12.8 follow-up years. All-cause mortality risk was 21-29% lower among participants with plasma (dp)ucMGP <450 pmol/L (n = 2361) compared with those with plasma (dp)ucMGP ≥450 pmol/L (adjusted HRs [95% CIs]: <300 pmol/L = 0.71 [0.61, 0.83], 300-449 pmol/L = 0.77 [0.66, 0.90]) and 16-19% lower among participants with plasma phylloquinone ≥0.50 nmol/L (n = 2421) compared to those with plasma phylloquinone <0.50 nmol/L (adjusted HRs: 0.50, 0.99 nmol/L = 0.84 [0.72, 0.99], ≥1.00 nmol/L = 0.81 [0.70, 0.95]). The risk of atherosclerotic CVD events did not significantly differ across plasma (dp)ucMGP or phylloquinone categories. CONCLUSIONS Two biomarkers of vitamin K status were associated with a lower all-cause mortality risk but not atherosclerotic CVD events. Additional studies are needed to clarify the mechanism underlying this association and evaluate the impact of improving vitamin K status in people with CKD.
Collapse
Affiliation(s)
- M Kyla Shea
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Kathryn Barger
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Sarah L Booth
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Jifan Wang
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Harold I Feldman
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Raymond R Townsend
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Jing Chen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - John Flack
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bernard G Jaar
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Mayank Kansal
- Department of Medicine, University of Illinois-Chicago, Chicago, IL, USA
| | - Sylvia E Rosas
- Joslin Diabetes Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Daniel E Weiner
- Division of Nephrology, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
14
|
Shea MK, Berkner KL, Ferland G, Fu X, Holden RM, Booth SL. Perspective: Evidence before Enthusiasm-A Critical Review of the Potential Cardiovascular Benefits of Vitamin K. Adv Nutr 2021; 12:632-646. [PMID: 33684212 PMCID: PMC8166540 DOI: 10.1093/advances/nmab004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022] Open
Abstract
A protective role for vitamin K in cardiovascular disease (CVD), a leading cause of morbidity and mortality, has been proposed because vitamin K-dependent proteins, such as matrix Gla (γ-carboxyglutamic acid) protein (MGP), are present in vascular tissue. MGP functions as a vascular calcification inhibitor-but only when it is carboxylated, which requires vitamin K. There is more than one naturally occurring form of vitamin K. Phylloquinone (vitamin K1) is found in plant-based foods, whereas menaquinones (vitamin K2) are a class of vitamin K compounds found in animal-based and fermented foods. Phylloquinone and menaquinones are capable of carboxylating MGP and other vitamin K-dependent proteins. In rodent models, high intakes of either phylloquinone or menaquinone reduced vascular calcification. Evidence of the relative importance of phylloquinone and menaquinone to CVD in humans is limited and controversial. In some observational studies, higher dietary menaquinone intake, but not phylloquinone intake, was associated with less coronary artery calcification (a subclinical manifestation of CVD) and a lower risk for clinical CVD events. These findings have led to claims that menaquinones have unique cardiovascular health benefits compared with phylloquinone. However, this claim is not supported by the results of the limited number of intervention trials conducted to date. The purpose of this review is to evaluate the strengths and limitations of the available evidence regarding the role of vitamin K in vascular calcification, CVD, and mortality.
Collapse
Affiliation(s)
- M Kyla Shea
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Kathleen L Berkner
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine at CWRU, Cleveland Clinic, Cleveland, OH, USA
| | - Guylaine Ferland
- Département de Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Xueyan Fu
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Rachel M Holden
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Sarah L Booth
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| |
Collapse
|
15
|
Nutrients, Genetic Factors, and Their Interaction in Non-Alcoholic Fatty Liver Disease and Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21228761. [PMID: 33228237 PMCID: PMC7699550 DOI: 10.3390/ijms21228761] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in Western countries and expose patients to increased risk of hepatic and cardiovascular (CV) morbidity and mortality. Both environmental factors and genetic predisposition contribute to the risk. An inappropriate diet, rich in refined carbohydrates, especially fructose, and saturated fats, and poor in fibers, polyunsaturated fats, and vitamins is one of the main key factors, as well as the polymorphism of patatin-like phospholipase domain containing 3 (PNPLA3 gene) for NAFLD and the apolipoproteins and the peroxisome proliferator-activated receptor (PPAR) family for the cardiovascular damage. Beyond genetic influence, also epigenetics modifications are responsible for various clinical manifestations of both hepatic and CV disease. Interestingly, data are accumulating on the interplay between diet and genetic and epigenetic modifications, modulating pathogenetic pathways in NAFLD and CV disease. We report the main evidence from literature on the influence of both macro and micronutrients in NAFLD and CV damage and the role of genetics either alone or combined with diet in increasing the risk of developing both diseases. Understanding the interaction between metabolic alterations, genetics and diet are essential to treat the diseases and tailoring nutritional therapy to control NAFLD and CV risk.
Collapse
|
16
|
Ghodsi R, Nosrati R. Effects of Minor Compounds of Edible Oils on Human Health. CURRENT NUTRITION & FOOD SCIENCE 2020. [DOI: 10.2174/1573401316666200203121034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Oils and fats are the densest sources of food energy among food groups.
Vegetable oils are constituted predominantly of triglycerides. Due to the importance of edible oils in
nutrition, food industry and human health, great attention has been paid to them in recent years.
Some minor bioactive constituents in oils include phospholipids, tocols, sterols, carotenoid, chlorophyll,
phenols, phylokynon and terpenes.
Objective:
The aim of the present study was to examine beneficial effects of minor compounds in edible oils on human health.
Results: Minor compounds of edible oils that we use daily can produce remarkable results in the prevention and treatment of various diseases like diabetes, inflammation, hypertension, cancer, allergy and central nervous system disorders due to their antimicrobial, anti-cancer, anti-viral, anti-oxidative, anti-inflammation, anti-mutagenic, hypolipidemic, and hypoglycemic properties, among others.
Conclusion:
The results of this study showed that the presence of beneficial minor compounds in oils could have significant impact on the prevention and treatment of various diseases. Therefore, the type of consumed oil can play an important role in human health.
Collapse
Affiliation(s)
- Ramin Ghodsi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rahmat Nosrati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Shea MK, Barger K, Booth SL, Matuszek G, Cushman M, Benjamin EJ, Kritchevsky SB, Weiner DE. Vitamin K status, cardiovascular disease, and all-cause mortality: a participant-level meta-analysis of 3 US cohorts. Am J Clin Nutr 2020; 111:1170-1177. [PMID: 32359159 PMCID: PMC7266692 DOI: 10.1093/ajcn/nqaa082] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/01/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Vitamin K-dependent proteins in vascular tissue affect vascular stiffness and calcification, which is associated with cardiovascular disease (CVD) and all-cause mortality. OBJECTIVE To determine the association of circulating vitamin K concentrations with CVD and all-cause mortality by conducting a participant-level meta-analysis. METHODS We obtained individual participant-level data from the Health, Aging, and Body Composition Study, the Multi-Ethnic Study of Atherosclerosis, and the Framingham Offspring Study, known cohorts with available measures of fasting circulating phylloquinone (vitamin K-1) and confirmed CVD events and mortality. Circulating phylloquinone was measured in a central laboratory from fasting blood samples and categorized as ≤0.5 nmol/L, >0.5-1.0 nmol/L, and >1.0 nmol/L. Multivariable Cox proportional hazard regression with multiple imputations was used to evaluate the association of circulating phylloquinone with incident CVD and all-cause mortality risk. RESULTS Among 3891 participants (mean age 65 ± 11 y; 55% women; 35% nonwhite), there were 858 incident CVD events and 1209 deaths over a median of 13.0 y. The risk of CVD did not significantly differ according to circulating phylloquinone [fully adjusted HR (95% CI) relative to >1.0 nmol/L: ≤0.5 nmol/L, 1.12 (0.94, 1.33); >0.5-1.0 nmol/L, 1.02 (0.86, 1.20)]. Participants with ≤0.5 nmol/L circulating phylloquinone had an adjusted 19% higher risk of all-cause mortality compared with those with >1.0 nmol/L [fully adjusted HR (95% CI): 1.19 (1.03, 1.38)]. Mortality risk was similar in participants with >0.5-1.0 nmol/L compared with >1.0 nmol/L [fully adjusted HR (95% CI): 1.04 (0.92, 1.17)]. CONCLUSIONS Low circulating phylloquinone concentrations were associated with an increased risk of all-cause mortality, but not of CVD. Additional studies are needed to clarify the mechanism underlying this association and evaluate the impact of increased phylloquinone intake on cardiovascular and other health outcomes in individuals with low vitamin K status.
Collapse
Affiliation(s)
- M Kyla Shea
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Kathryn Barger
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Sarah L Booth
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Gregory Matuszek
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Mary Cushman
- Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Emelia J Benjamin
- Boston University School of Medicine and Public Health, Boston, MA, USA
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Daniel E Weiner
- Tufts Medical Center, Division of Nephrology, Boston, MA, USA
| |
Collapse
|
18
|
Rees-Milton KJ, Norman P, Babiolakis C, Hulbert M, Turner ME, Berger C, Anastassiades TP, Hopman WM, Adams MA, Powley WL, Holden RM. Statin Use is Associated With Insulin Resistance in Participants of the Canadian Multicentre Osteoporosis Study. J Endocr Soc 2020; 4:bvaa057. [PMID: 32715271 PMCID: PMC7371386 DOI: 10.1210/jendso/bvaa057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/08/2020] [Indexed: 12/15/2022] Open
Abstract
Context Statins have been linked to the development of diabetes and atherosclerotic plaque calcification in patients with cardiac disease. Objective To determine the association between statin use and statin characteristics and insulin resistance and abdominal aortic calcification (AAC) in participants of the Canadian Multicentre Osteoporosis Study (CaMos). Design Observational study. Setting General community. Participants Nondiabetic participants of the Kingston CaMos site. Intervention Insulin resistance and AAC in statin users and nonstatin users were compared with and without the inclusion of a propensity score (PS) to be on a statin. The covariates of hypertension, sex, body mass index, smoking, kidney stones, and age that were included in the PS were selected based on clinical judgment confirmed by the statistical analysis of a difference between statin users and nonstatin users. Main Outcome Measures Insulin resistance measured by the homeostasis model assessment (HOMA-IR) and AAC assessed on lateral spine radiographs using the Framingham methodology. Results Using a general linear model, statin use was associated with higher levels of HOMA-IR after stratified PS adjustment (β = 1.52, [1.18-1.95], P < 0.01). Hydrophilic statin users (n = 9) and lipophilic statins users (n = 30) had higher HOMA-IR compared to nonstatin users (n = 125) ([β = 2.29, (1.43-3.68), P < 0.001] and [β = 1.36, (1.04-1.78), P < 0.05]), respectively, in general linear models after stratified PS adjustment. Statin use was associated with AAC without stratifying by PS in the Wilcoxon test, but was no longer significant when stratified by PS. Conclusions Statins, widely prescribed drugs to lower cholesterol, may have unintended consequences related to glucose homeostasis that could be relevant in healthy aging.
Collapse
Affiliation(s)
| | - Patrick Norman
- Kingston General Health Research Institute, Kingston, ON
| | | | - Maggie Hulbert
- Department of Medicine, Queen's University, Kingston, ON
| | - Mandy E Turner
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON
| | - Claudie Berger
- Research Institute of the McGill University Health Centre, Montreal, QC
| | - Tassos P Anastassiades
- Department of Medicine, Queen's University, Kingston, ON.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON
| | - Wilma M Hopman
- Kingston General Health Research Institute, Kingston, ON.,Department of Public Health Sciences, Queen's University, Kingston, ON
| | - Michael A Adams
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON
| | | | - Rachel M Holden
- Department of Medicine, Queen's University, Kingston, ON.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON
| |
Collapse
|
19
|
Florea A, Morgenroth A, Bucerius J, Schurgers LJ, Mottaghy FM. Locking and loading the bullet against micro-calcification. Eur J Prev Cardiol 2020; 28:1370-1375. [PMID: 33611501 DOI: 10.1177/2047487320911138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/14/2020] [Indexed: 12/24/2022]
Abstract
AIMS Despite recent medical advances, cardiovascular disease remains the leading cause of death worldwide. As (micro)-calcification is a hallmark of atherosclerosis, this review will elaborately discuss advantages of sodium fluoride positron emission tomography (PET) as a reliable cardiovascular imaging technique for identifying the early onset of vascular calcification (i.e. locking onto the target). We assess state-of-the-art meta-analysis and clinical studies of possible treatment options and evaluate the concept of vitamin K supplementation to preserve vascular health (i.e. loading the bullet). METHODS AND RESULTS After a structured PubMed search, we identified 18F-sodium fluoride (18F-NaF) PET as the most suitable technique for detecting micro-calcification. Presenting the pros and cons of available treatments, vitamin K supplementation should be considered as a possible safe and cost-effective option to inhibit vascular (micro)-calcification. CONCLUSION This review demonstrates need for more extensive research in the concept of vitamin K supplementation (i.e. loading the bullet) and recommends monitoring the effects on vascular calcification using 18F-NaF PET (i.e. locking onto the target).
Collapse
Affiliation(s)
- Alexandru Florea
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Academic Hospital Maastricht, Maastricht, Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht University, Netherlands
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Jan Bucerius
- Department of Radiology and Nuclear Medicine, Academic Hospital Maastricht, Maastricht, Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht University, Netherlands
- Department of Nuclear Medicine, University of Göttingen, Göttingen, Germany
| | - Leon J Schurgers
- School for Cardiovascular Diseases (CARIM), Maastricht University, Netherlands
- Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Academic Hospital Maastricht, Maastricht, Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht University, Netherlands
| |
Collapse
|
20
|
Zwakenberg SR, Burgess S, Sluijs I, Weiderpass E, Beulens JWJ, van der Schouw YT. Circulating phylloquinone, inactive Matrix Gla protein and coronary heart disease risk: A two-sample Mendelian Randomization study. Clin Nutr 2020; 39:1131-1136. [PMID: 31103344 PMCID: PMC7612948 DOI: 10.1016/j.clnu.2019.04.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/02/2019] [Accepted: 04/17/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Multiple observational studies and small-scale intervention studies suggest that high vitamin K intake is associated with improved markers for cardiovascular health. Circulating phylloquinone solely represents phylloquinone (vitamin K1) intake, while dephosphorylated uncarboxylated Matrix Gla Protein (dp-ucMGP) represents both phylloquinone and menaquinone (vitamin K2) intake. This study aims to investigate the causal relationship between genetically predicted vitamin K concentrations and the risk of CHD via a two-sample Mendelian Randomization approach. DESIGN We used data from three studies: the European Prospective Investigation into Cancer and Nutrition (EPIC)-CVD case-cohort study, CARDIOGRAMplusC4D and the UK Biobank, resulting in 103,097 CHD cases. Genetically predicted vitamin K concentrations were measured using SNPs related to circulating phylloquinone and dp-ucMGP. We calculated a genetic risk score (GRS) including four SNPs (rs2108622, rs2192574, rs4645543 and rs6862071) related to circulating phylloquinone levels from a genome wide association study. Rs4236 was used as an instrumental variable for dp-ucMGP. Inverse-variance weighted (IVW) analysis was used to obtain Risk Ratios (RRs) for the causal relationship between phylloquinone and dp-ucMGP concentrations and CHD risk. RESULTS Using the genetic score for circulating phylloquinone, we found that circulating phylloquinone was not causally related to CHD risk (RR 1.00 (95%-CI: 0.98; 1.04)). Lower genetically predicted dp-ucMGP concentration was associated with a lower CHD risk with a RR of 0.96 (95%-CI: 0.93; 0.99) for every 10 μg/L decrease in dp-ucMGP. CONCLUSIONS This study did not confirm a causal relationship between circulating phylloquinone and lower CHD risk. However, lower dp-ucMGP levels may be causally related with a decreased CHD risk. This inconsistent result may reflect the influence of menaquinones in the association with CHD.
Collapse
Affiliation(s)
- Sabine R Zwakenberg
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands
| | - Stephen Burgess
- Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK; MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Ivonne Sluijs
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Joline W J Beulens
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, the Netherlands
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands.
| |
Collapse
|
21
|
Bøgh Andersen I, Brasen CL, Nasimi H, Stougård M, Bliddal M, Green A, Schmedes A, Brandslund I, Madsen JS. Serum vitamin K 1 associated to microangiopathy and/or macroangiopathy in individuals with and without diabetes. BMJ Open Diabetes Res Care 2020; 8:8/1/e000961. [PMID: 32213490 PMCID: PMC7170411 DOI: 10.1136/bmjdrc-2019-000961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/02/2020] [Accepted: 02/09/2020] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Vitamin K has proposed beneficial effects on cardiovascular health. We investigated whether serum vitamin K1 was associated with prevalence of microangiopathy and/or macroangiopathy. RESEARCH DESIGN AND METHODS Serum vitamin K was quantified in 3239 individuals with and 3808 without diabetes enrolled in Vejle Diabetes Biobank (2007-2010). Each individual was assessed for microangiography and macroangiopathy at enrollment based on registered diagnoses in the Danish National Patient Registry according to the International Classification of Disease 8 (1977-1993) and 10 (since 1994). Using multinomial logistic regression, relative risk ratios (RRRs) were calculated within each group of individuals with and without diabetes. RRRs were estimated for microangiopathic/macroangiopathic status compared with individuals without complications as a function of 1 nmol/L increments in K1. Adjustment for potential confounders was also performed. RESULTS Vitamin K1 (median) varied 0.86-0.95 nmol/L depending on diabetes, microangiopathic and macroangiopathic status. In individuals with diabetes, the crude RRR for only having microangiopathy was 1.05 (95% CI 0.98 to 1.12) and was found significant when adjusting 1.10 (95% CI 1.01 to 1.19). RRR for having only macroangiopathy was 0.89 (95% CI 0.77 to 1.03) and was again significant when adjusting 0.79 (95% CI 0.66 to 0.96). In individuals without diabetes, adjustments again led to similar estimates that was not significant. The adjusted RRR for having only macroangiopathy was 1.08 (95% CI 0.98 to 1.19). CONCLUSIONS Serum vitamin K1 levels were associated with microangiopathic and macroangiopathic status in individuals with diabetes, but considered of no clinical relevance. The clinical value of other candidate markers for vitamin K status needs to be evaluated in future studies.
Collapse
Affiliation(s)
- Ida Bøgh Andersen
- Department of Clinical Biochemistry and Immunology, Lillebælt Hospital, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Claus Lohman Brasen
- Department of Clinical Biochemistry and Immunology, Lillebælt Hospital, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Hashmatullah Nasimi
- Department of Clinical Biochemistry and Immunology, Lillebælt Hospital, Vejle, Denmark
- Institute for Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Maria Stougård
- OPEN-Open Patient data Explorative Network, Department of Clinical Research, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Mette Bliddal
- OPEN-Open Patient data Explorative Network, Department of Clinical Research, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Anders Green
- OPEN-Open Patient data Explorative Network, Department of Clinical Research, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Anne Schmedes
- Department of Clinical Biochemistry and Immunology, Lillebælt Hospital, Vejle, Denmark
| | - Ivan Brandslund
- Department of Clinical Biochemistry and Immunology, Lillebælt Hospital, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Jonna Skov Madsen
- Department of Clinical Biochemistry and Immunology, Lillebælt Hospital, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
22
|
Shioi A, Morioka T, Shoji T, Emoto M. The Inhibitory Roles of Vitamin K in Progression of Vascular Calcification. Nutrients 2020; 12:nu12020583. [PMID: 32102248 PMCID: PMC7071387 DOI: 10.3390/nu12020583] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Vitamin K is a fat-soluble vitamin that is indispensable for the activation of vitamin K-dependent proteins (VKDPs) and may be implicated in cardiovascular disease (CVD). Vascular calcification is intimately associated with CV events and mortality and is a chronic inflammatory process in which activated macrophages promote osteoblastic differentiation of vascular smooth muscle cells (VSMCs) through the production of proinflammatory cytokines such as IL-1β, IL-6, TNF-α, and oncostatin M (OSM) in both intimal and medial layers of arterial walls. This process may be mainly mediated through NF-κB signaling pathway. Vitamin K has been demonstrated to exert anti-inflammatory effects through antagonizing NF-κB signaling in both in vitro and in vivo studies, suggesting that vitamin K may prevent vascular calcification via anti-inflammatory mechanisms. Matrix Gla protein (MGP) is a major inhibitor of soft tissue calcification and contributes to preventing both intimal and medial vascular calcification. Vitamin K may also inhibit progression of vascular calcification by enhancing the activity of MGP through facilitating its γ-carboxylation. In support of this hypothesis, the procalcific effects of warfarin, an antagonist of vitamin K, on arterial calcification have been demonstrated in several clinical studies. Among the inactive MGP forms, dephospho-uncarboxylated MGP (dp-ucMGP) may be regarded as the most useful biomarker of not only vitamin K deficiency, but also vascular calcification and CVD. There have been several studies showing the association of circulating levels of dp-ucMGP with vitamin K intake, vascular calcification, mortality, and CVD. However, additional larger prospective studies including randomized controlled trials are necessary to confirm the beneficial effects of vitamin K supplementation on CV health.
Collapse
Affiliation(s)
- Atsushi Shioi
- Department of Vascular Medicine and Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan;
- Correspondence: ; Tel.: +81666453931
| | - Tomoaki Morioka
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka 545-85858, Japan; (T.M.); (M.E.)
| | - Tetsuo Shoji
- Department of Vascular Medicine and Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan;
| | - Masanori Emoto
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka 545-85858, Japan; (T.M.); (M.E.)
| |
Collapse
|
23
|
Vitamin K as a Diet Supplement with Impact in Human Health: Current Evidence in Age-Related Diseases. Nutrients 2020; 12:nu12010138. [PMID: 31947821 PMCID: PMC7019739 DOI: 10.3390/nu12010138] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022] Open
Abstract
Vitamin K health benefits have been recently widely shown to extend beyond blood homeostasis and implicated in chronic low-grade inflammatory diseases such as cardiovascular disease, osteoarthritis, dementia, cognitive impairment, mobility disability, and frailty. Novel and more efficient nutritional and therapeutic options are urgently needed to lower the burden and the associated health care costs of these age-related diseases. Naturally occurring vitamin K comprise the phylloquinone (vitamin K1), and a series of menaquinones broadly designated as vitamin K2 that differ in source, absorption rates, tissue distribution, bioavailability, and target activity. Although vitamin K1 and K2 sources are mainly dietary, consumer preference for diet supplements is growing, especially when derived from marine resources. The aim of this review is to update the reader regarding the specific contribution and effect of each K1 and K2 vitamers in human health, identify potential methods for its sustainable and cost-efficient production, and novel natural sources of vitamin K and formulations to improve absorption and bioavailability. This new information will contribute to foster the use of vitamin K as a health-promoting supplement, which meets the increasing consumer demand. Simultaneously, relevant information on the clinical context and direct health consequences of vitamin K deficiency focusing in aging and age-related diseases will be discussed.
Collapse
|
24
|
Shea MK, Booth SL. Vitamin K, Vascular Calcification, and Chronic Kidney Disease: Current Evidence and Unanswered Questions. Curr Dev Nutr 2019; 3:nzz077. [PMID: 31598579 PMCID: PMC6775440 DOI: 10.1093/cdn/nzz077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/12/2019] [Accepted: 06/26/2019] [Indexed: 01/07/2023] Open
Abstract
More than 15% of the US population is currently >65 y old. As populations age there is a concomitant increase in age-related chronic diseases. One such disease is chronic kidney disease (CKD), which becomes more prevalent with age, especially over age 70 y. Individuals with CKD are at increased risk of cardiovascular disease, in part because arterial calcification increases as kidney function declines. Vitamin K is a shortfall nutrient among older adults that has been implicated in arterial calcification. Evidence suggests CKD patients have low vitamin K status, but data are equivocal because the biomarkers of vitamin K status can be influenced by CKD. Animal studies provide more compelling data on the underlying role of vitamin K in arterial calcification associated with CKD. The purpose of this review is to evaluate the strengths and limitations of the available evidence regarding the role of vitamin K in CKD.
Collapse
Affiliation(s)
- M Kyla Shea
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Sarah L Booth
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
25
|
Vitamin K as a Powerful Micronutrient in Aging and Age-Related Diseases: Pros and Cons from Clinical Studies. Int J Mol Sci 2019; 20:ijms20174150. [PMID: 31450694 PMCID: PMC6747195 DOI: 10.3390/ijms20174150] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
Vitamin K is a multifunctional micronutrient implicated in age-related diseases such as cardiovascular diseases, osteoarthritis and osteoporosis. Although vitamin K-dependent proteins (VKDPs) are described to have a crucial role in the pathogenesis of these diseases, novel roles have emerged for vitamin K, independently of its role in VKDPs carboxylation. Vitamin K has been shown to act as an anti-inflammatory by suppressing nuclear factor κB (NF-κB) signal transduction and to exert a protective effect against oxidative stress by blocking the generation of reactive oxygen species. Available clinical evidences indicate that a high vitamin K status can exert a protective role in the inflammatory and mineralization processes associated with the onset and progression of age-related diseases. Also, vitamin K involvement as a protective super-micronutrient in aging and ‘inflammaging’ is arising, highlighting its future use in clinical practice. In this review we summarize current knowledge regarding clinical data on vitamin K in skeletal and cardiovascular health, and discuss the potential of vitamin K supplementation as a health benefit. We describe the clinical evidence and explore molecular aspects of vitamin K protective role in aging and age-related diseases, and its involvement as a modulator in the interplay between pathological calcification and inflammation processes.
Collapse
|
26
|
Petsophonsakul P, Furmanik M, Forsythe R, Dweck M, Schurink GW, Natour E, Reutelingsperger C, Jacobs M, Mees B, Schurgers L. Role of Vascular Smooth Muscle Cell Phenotypic Switching and Calcification in Aortic Aneurysm Formation. Arterioscler Thromb Vasc Biol 2019; 39:1351-1368. [PMID: 31144989 DOI: 10.1161/atvbaha.119.312787] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aortic aneurysm is a vascular disease whereby the ECM (extracellular matrix) of a blood vessel degenerates, leading to dilation and eventually vessel wall rupture. Recently, it was shown that calcification of the vessel wall is involved in both the initiation and progression of aneurysms. Changes in aortic wall structure that lead to aneurysm formation and vascular calcification are actively mediated by vascular smooth muscle cells. Vascular smooth muscle cells in a healthy vessel wall are termed contractile as they maintain vascular tone and remain quiescent. However, in pathological conditions they can dedifferentiate into a synthetic phenotype, whereby they secrete extracellular vesicles, proliferate, and migrate to repair injury. This process is called phenotypic switching and is often the first step in vascular pathology. Additionally, healthy vascular smooth muscle cells synthesize VKDPs (vitamin K-dependent proteins), which are involved in inhibition of vascular calcification. The metabolism of these proteins is known to be disrupted in vascular pathologies. In this review, we summarize the current literature on vascular smooth muscle cell phenotypic switching and vascular calcification in relation to aneurysm. Moreover, we address the role of vitamin K and VKDPs that are involved in vascular calcification and aneurysm. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Ploingarm Petsophonsakul
- From the Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (P.P., M.F., C.R., L.S.)
| | - Malgorzata Furmanik
- From the Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (P.P., M.F., C.R., L.S.)
| | - Rachael Forsythe
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (R.F., M.D.)
| | - Marc Dweck
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (R.F., M.D.)
| | - Geert Willem Schurink
- Department of Vascular Surgery (G.W.S., M.J., B.M.), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Ehsan Natour
- Department of Cardiovascular Surgery (E.N.), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands.,European Vascular Center Aachen-Maastricht, Maastricht, the Netherlands (E.N., M.J., B.M.)
| | - Chris Reutelingsperger
- From the Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (P.P., M.F., C.R., L.S.)
| | - Michael Jacobs
- Department of Vascular Surgery (G.W.S., M.J., B.M.), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands.,European Vascular Center Aachen-Maastricht, Maastricht, the Netherlands (E.N., M.J., B.M.)
| | - Barend Mees
- Department of Vascular Surgery (G.W.S., M.J., B.M.), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands.,European Vascular Center Aachen-Maastricht, Maastricht, the Netherlands (E.N., M.J., B.M.)
| | - Leon Schurgers
- From the Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (P.P., M.F., C.R., L.S.)
| |
Collapse
|
27
|
Ruiz-León AM, Lapuente M, Estruch R, Casas R. Clinical Advances in Immunonutrition and Atherosclerosis: A Review. Front Immunol 2019; 10:837. [PMID: 31068933 PMCID: PMC6491827 DOI: 10.3389/fimmu.2019.00837] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/01/2019] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a chronic low-grade inflammatory disease that affects large and medium-sized arteries and is considered to be a major underlying cause of cardiovascular disease (CVD). The high risk of mortality by atherosclerosis has led to the development of new strategies for disease prevention and management, including immunonutrition. Plant-based dietary patterns, functional foods, dietary supplements, and bioactive compounds such as the Mediterranean Diet, berries, polyunsaturated fatty acids, ω-3 and ω-6, vitamins E, A, C, and D, coenzyme Q10, as well as phytochemicals including isoflavones, stilbenes, and sterols have been associated with improvement in atheroma plaque at an inflammatory level. However, many of these correlations have been obtained in vitro and in experimental animals' models. On one hand, the present review focuses on the evidence obtained from epidemiological, dietary intervention and supplementation studies in humans supporting the role of immunonutrient supplementation and its effect on anti-inflammatory response in atherosclerotic disease. On the other hand, this review also analyzes the possible molecular mechanisms underlying the protective action of these supplements, which may lead a novel therapeutic approach to prevent or attenuate diet-related disease, such as atherosclerosis.
Collapse
Affiliation(s)
- Ana María Ruiz-León
- Department of Internal Medicine, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Mediterranean Diet Foundation, Barcelona, Spain
| | - María Lapuente
- Department of Internal Medicine, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Ramon Estruch
- Department of Internal Medicine, Hospital Clinic, University of Barcelona, Barcelona, Spain.,CIBER 06/03: Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Casas
- Department of Internal Medicine, Hospital Clinic, University of Barcelona, Barcelona, Spain.,CIBER 06/03: Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
28
|
Hu T, Jacobs DR, Bazzano LA, Bertoni AG, Steffen LM. Low-carbohydrate diets and prevalence, incidence and progression of coronary artery calcium in the Multi-Ethnic Study of Atherosclerosis (MESA). Br J Nutr 2019; 121:461-468. [PMID: 30630542 PMCID: PMC7521628 DOI: 10.1017/s0007114518003513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The evidence linking low-carbohydrate diets (LCD) to CVD is controversial, and results from epidemiological studies are inconsistent. We aimed to assess the relationship between LCD patterns and coronary artery Ca (CAC) scores from computed tomography in the Multi-Ethnic Study of Atherosclerosis cohort. Our sample included 5614 men and women free of clinical CVD at baseline (2000-2002), who had a FFQ, a baseline measure and ≥1 measure of CAC during follow-up. We excluded those with implausible energy intake or daily physical activity. The overall, animal-based and plant-based LCD scores were calculated based on intakes of macronutrients. Relative risk regression and robust regression models were used to examine the cross-sectional and longitudinal relationship between LCD score quintile and CAC outcomes, after adjustment for multiple cardiovascular risk factors. The mean age of participants was 63 years. The median intakes of total carbohydrate, fat and protein were 53·7, 30·5 and 15·6 % energy/d, respectively. Among 2892 participants with zero CAC scores at baseline, 264 developed positive scores during 2·4-year follow-up (11-59 months). Among those with positive scores at baseline, the median increase in CAC was 47 units over the course of follow-up. The overall, the animal-based and the plant-based LCD scores were not associated with CAC prevalence, incidence and progression. In conclusion, diets low in carbohydrate and high in fat and/or protein, regardless of the sources of protein and fat, were not associated with higher levels of CAC, a validated predictor of cardiovascular events, in this large multi-ethnic cohort.
Collapse
Affiliation(s)
- Tian Hu
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA
| | - Lydia A Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Alain G Bertoni
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA
| |
Collapse
|
29
|
Houston M. The role of noninvasive cardiovascular testing, applied clinical nutrition and nutritional supplements in the prevention and treatment of coronary heart disease. Ther Adv Cardiovasc Dis 2018; 12:85-108. [PMID: 29316855 PMCID: PMC5933539 DOI: 10.1177/1753944717743920] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022] Open
Abstract
Numerous clinical trials suggest that we have reached a limit in our ability to decrease the incidence of coronary heart disease (CHD) and cardiovascular disease (CVD) utilizing the traditional diagnostic evaluation, prevention and treatment strategies for the top five cardiovascular risk factors of hypertension, diabetes mellitus, dyslipidemia, obesity and smoking. About 80% of heart disease (heart attacks, angina, coronary heart disease and congestive heart failure) can be prevented by optimal nutrition, optimal exercise, optimal weight and body composition, mild alcohol intake and avoiding smoking. Statistics show that approximately 50% of patients continue to have CHD or myocardial infarction (MI) despite presently defined 'normal' levels of the five risk factors listed above. This is often referred to as the 'CHD gap'. Novel and more accurate definitions and evaluations of these top five risk factors are required, such as 24 h ambulatory blood pressure (ABM) results, advanced lipid profiles, redefined fasting and 2 h dysglycemia parameters, a focus on visceral obesity and body composition and the effects of adipokines on cardiovascular risk. There are numerous traumatic insults from the environment that damage the cardiovascular system but there are only three finite vascular endothelial responses, which are inflammation, oxidative stress and immune vascular dysfunction. In addition, the concept of translational cardiovascular medicine is mandatory in order to correlate the myriad of CHD risk factors to the presence or absence of functional or structural damage to the vascular system, preclinical and clinical CHD. This can be accomplished by utilizing advanced and updated CV risk scoring systems, new and redefined CV risk factors and biomarkers, micronutrient testing, cardiovascular genetics, nutrigenomics, metabolomics, genetic expression testing and noninvasive cardiovascular testing.
Collapse
Affiliation(s)
- Mark Houston
- Vanderbilt University Medical School, Hypertension Institute and Vascular Biology, Division of Human Nutrition, Saint Thomas Medical Group, Saint Thomas Hospital, 4230 Harding Rd, Suite 400, Nashville, TN 37205, USA
| |
Collapse
|
30
|
Finnan EG, Harshman SG, Haytowitz DB, Booth SL. Mixed dishes are an unexpected source of dietary vitamin K. J Food Compost Anal 2017. [DOI: 10.1016/j.jfca.2017.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
31
|
|
32
|
van Ballegooijen AJ, Beulens JW. The Role of Vitamin K Status in Cardiovascular Health: Evidence from Observational and Clinical Studies. Curr Nutr Rep 2017; 6:197-205. [PMID: 28944098 PMCID: PMC5585988 DOI: 10.1007/s13668-017-0208-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Vitamin K is a fat-soluble vitamin required for the activation of several vitamin K-dependent proteins to confer functioning. A growing body of evidence supports that vitamin K has beneficial effects on bone and cardiovascular health. This review summarizes key evidence on vitamin K status as measured by circulating measures and cardiovascular outcomes. RECENT FINDINGS Overall, observational studies indicate that low vitamin K status as measured by high dephosphorylated uncarboxylated matrix gla protein concentrations plays a potential role in cardiovascular disease development, particularly in high-risk and chronic kidney disease populations. Very few vitamin K intervention trials have been conducted with cardiovascular-related outcomes. A couple of intervention trials studied the effect of the combination of vitamin D + K supplementation, which might have synergistic effects compared to vitamin K supplementation alone. SUMMARY Assessing vitamin K status in prospective studies and well-designed randomized trials would provide important insight whether vitamin K is causally related to vascular calcification and cardiovascular disease.
Collapse
Affiliation(s)
- A J van Ballegooijen
- Department of Health Sciences, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - J W Beulens
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, the Netherlands.,Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
33
|
Vitamins K1 and K2: The Emerging Group of Vitamins Required for Human Health. J Nutr Metab 2017; 2017:6254836. [PMID: 28698808 PMCID: PMC5494092 DOI: 10.1155/2017/6254836] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/10/2017] [Indexed: 12/21/2022] Open
Abstract
Objective To review the evidence for the use of vitamin K supplementation in clinical conditions such as osteoporosis, vascular calcification, arthritis, cancer, renal calculi, diabetes, and warfarin therapy. Quality of Evidence PubMed was searched for articles on vitamin K (K1 and K2) along with books and conference proceedings and health conditions listed above. Level I and II evidence supports the use of vitamins K1 and K2 in osteoporosis and Level II evidence supports vitamin K2 in prevention of coronary calcification and cardiovascular disease. Evidence is insufficient for use in diabetes, arthritis, renal calculi, and cancer. Main Message Vitamin K2 may be a useful adjunct for the treatment of osteoporosis, along with vitamin D and calcium, rivaling bisphosphonate therapy without toxicity. It may also significantly reduce morbidity and mortality in cardiovascular health by reducing vascular calcification. Vitamin K2 appears promising in the areas of diabetes, cancer, and osteoarthritis. Vitamin K use in warfarin therapy is safe and may improve INR control, although a dosage adjustment is required. Conclusion Vitamin K supplementation may be useful for a number of chronic conditions that are afflicting North Americans as the population ages. Supplementation may be required for bone and cardiovascular health.
Collapse
|
34
|
Shea MK, Booth SL, Weiner DE, Brinkley TE, Kanaya AM, Murphy RA, Simonsick EM, Wassel CL, Vermeer C, Kritchevsky SB. Circulating Vitamin K Is Inversely Associated with Incident Cardiovascular Disease Risk among Those Treated for Hypertension in the Health, Aging, and Body Composition Study (Health ABC). J Nutr 2017; 147:888-895. [PMID: 28356433 PMCID: PMC5404216 DOI: 10.3945/jn.117.249375] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 02/23/2017] [Accepted: 03/03/2017] [Indexed: 01/07/2023] Open
Abstract
Background: A role for vitamin K in coronary artery calcification (CAC), a subclinical manifestation of cardiovascular disease (CVD), has been proposed because vitamin K-dependent proteins, including the calcification inhibitor matrix Gla protein (MGP), are present in vascular tissue. Observational studies found that low circulating phylloquinone (vitamin K-1) was associated with increased CAC progression, especially in persons treated for hypertension. It is unknown whether hypertension treatment modifies this putative role of vitamin K in clinical CVD risk.Objective: We determined the association between vitamin K status and incident clinical CVD in older adults in the Health ABC (Health, Aging, and Body Composition Study) and whether the association differed by hypertension treatment status.Methods: Plasma phylloquinone was measured in 1061 participants free of CVD (70-79 y of age, 58% women, 39% black). Plasma uncarboxylated MGP [(dp)ucMGP] was measured in a subset of 635 participants. Multivariate Cox models estimated the HR for incident CVD over 12.1 follow-up years. Effect modification by hypertension was tested with the use of interaction terms.Results: Neither low plasma phylloquinone (<0.2 nmol/L) nor elevated (dp)ucMGP (≥574 pmol/L) was significantly associated with incident CVD [respective HRs (95% CIs): 1.27 (0.75, 2.13) and 1.02 (0.72, 1.45)]. In participants treated for hypertension (n = 489; 135 events), low plasma phylloquinone was associated with higher CVD risk overall (HR: 2.94; 95% CI: 1.41, 6.13). In those with untreated hypertension (n = 153; 48 events) and without hypertension (n = 418; 92 events), low plasma phylloquinone was not associated with incident CVD. The association between high (dp)ucMGP did not differ by hypertension treatment status (P-interaction = 0.72).Conclusions: Vitamin K status was not significantly associated with CVD risk overall, but low plasma phylloquinone was associated with a higher CVD risk in older adults treated for hypertension. Additional evidence from larger clinical studies is needed to clarify the importance of vitamin K to CVD in persons treated for hypertension, a segment of the population at high risk of clinical CVD events.
Collapse
Affiliation(s)
- M Kyla Shea
- USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA;
| | - Sarah L Booth
- USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Daniel E Weiner
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, MA
| | - Tina E Brinkley
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC
| | - Alka M Kanaya
- University of California-San Francisco, San Francisco, CA
| | - Rachel A Murphy
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Christina L Wassel
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; and
| | - Cees Vermeer
- VitaK, University of Maastricht, Maastricht, Netherlands
| | | |
Collapse
|
35
|
Danziger J, Young RL, Shea KM, Duprez DA, Jacobs DR, Tracy RP, Ix JH, Jenny NS, Mukamal KJ. 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. [PMID: 27508317 DOI: 10.1016/j.atherosclerosis.2016.07.924] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/09/2016] [Accepted: 07/27/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Vitamin K-dependent protein (VKDP) activity may have a role in preventing cardiovascular calcification, but has not previously been studied in large, generally healthy populations. METHODS Using an elevated ankle-brachial index (ABI) as a measure of medial vascular calcification, we performed a case-cohort analysis within the Multi-Ethnic Study of Atherosclerosis, measuring Des-gamma-carboxy prothrombin (DCP) to estimate VKDP activity. In secondary analyses of the weighted subcohort, we examined the cross-sectional associations between DCP and prevalent vascular calcification of the coronary vessels, aortic and mitral valves, and aortic wall, and with vascular stiffness. RESULTS In adjusted analysis, cases (n = 104) had 0.21 ng/ml (-0.94-0.52) lower DCP concentrations than the subcohort (n = 613). Furthermore, amongst the 717 participants in the weighted cohort, VKDP activity was not associated with coronary artery, mitral valve, aortic valve or aortic wall calcification, nor was it associated with vascular stiffness. CONCLUSIONS Our negative results do not support a role of circulating VKDP activity in cardiovascular calcification in community-dwelling adults.
Collapse
Affiliation(s)
- John Danziger
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States.
| | | | - Kyla M Shea
- Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Daniel A Duprez
- Division of Cardiology, University of Minnesota, United States
| | - David R Jacobs
- School of Public Health, Division of Epidemiology & Community Health, University of Minnesota, United States
| | - Russell P Tracy
- College of Medicine, University of Vermont, Burlington, VT, United States
| | - Joachim H Ix
- Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, CA, United States
| | - Nancy S Jenny
- Department of Pathology, University of Vermont, Burlington, VT, United States
| | - Kenneth J Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| |
Collapse
|
36
|
Harshman SG, Shea MK. The Role of Vitamin K in Chronic Aging Diseases: Inflammation, Cardiovascular Disease, and Osteoarthritis. Curr Nutr Rep 2016; 5:90-98. [PMID: 27648390 DOI: 10.1007/s13668-016-0162-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Vitamin K is an enzyme cofactor required for the carboxylation of vitamin K dependent proteins, several of which have been implicated in diseases of aging. Inflammation is recognized as a crucial component of many chronic aging diseases and evidence suggests vitamin K has an anti-inflammatory action that is independent of its role as an enzyme co-factor. Vitamin K-dependent proteins and inflammation have been implicated in cardiovascular disease and osteoarthritis, which are leading causes of disability and mortality in older adults. The purpose of this review is to summarize observational studies and randomized trials focused on vitamin K status and inflammation, cardiovascular disease, and osteoarthritis. Although mechanistic evidence suggests a protective role for vitamin K in these age-related conditions, the benefit of vitamin K supplementation is controversial because observational data are equivocal and the number of randomized trials is few.
Collapse
Affiliation(s)
- Stephanie G Harshman
- Jean Mayer Human Nutrition Research Center on Aging, Tufts University 711 Washington Street, Boston, MA 02111, Phone number: 617-556-3151, Fax number: 617 556 3149
| | - M Kyla Shea
- Jean Mayer Human Nutrition Research Center on Aging, Tufts University 711 Washington Street, Boston, MA 02111, Phone number: 617-556-3073, fax number: 617 556 3344
| |
Collapse
|
37
|
Shea MK, Booth SL. Concepts and Controversies in Evaluating Vitamin K Status in Population-Based Studies. Nutrients 2016; 8:E8. [PMID: 26729160 PMCID: PMC4728622 DOI: 10.3390/nu8010008] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/04/2015] [Accepted: 12/09/2015] [Indexed: 12/12/2022] Open
Abstract
A better understanding of vitamin K's role in health and disease requires the assessment of vitamin K nutritional status in population and clinical studies. This is primarily accomplished using dietary questionnaires and/or biomarkers. Because food composition databases in the US are most complete for phylloquinone (vitamin K1, the primary form in Western diets), emphasis has been on phylloquinone intakes and associations with chronic diseases. There is growing interest in menaquinone (vitamin K2) intakes for which the food composition databases need to be expanded. Phylloquinone is commonly measured in circulation, has robust quality control schemes and changes in response to phylloquinone intake. Conversely, menaquinones are generally not detected in circulation unless large quantities are consumed. The undercarboxylated fractions of three vitamin K-dependent proteins are measurable in circulation, change in response to vitamin K supplementation and are modestly correlated. Since different vitamin K dependent proteins are implicated in different diseases the appropriate vitamin K-dependent protein biomarker depends on the outcome under study. In contrast to other nutrients, there is no single biomarker that is considered a gold-standard measure of vitamin K status. Most studies have limited volume of specimens. Strategic decisions, guided by the research question, need to be made when deciding on choice of biomarkers.
Collapse
Affiliation(s)
- M Kyla Shea
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Sarah L Booth
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA.
| |
Collapse
|
38
|
Shea MK, Loeser RF, Hsu FC, Booth SL, Nevitt M, Simonsick EM, Strotmeyer ES, Vermeer C, Kritchevsky SB. Vitamin K Status and Lower Extremity Function in Older Adults: The Health Aging and Body Composition Study. J Gerontol A Biol Sci Med Sci 2015; 71:1348-55. [PMID: 26576842 DOI: 10.1093/gerona/glv209] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 10/22/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND While low vitamin K status has been associated with several chronic diseases that can lead to lower extremity disability, it is not known if low vitamin K status is associated with worse lower extremity function. METHODS Vitamin K status was measured according to plasma phylloquinone (vitamin K1) and dephosphorylated-uncarboxylated MGP (dp-ucMGP) in 1,089 community-dwelling older adults (mean ± SD age =74±3 years; 67% female). Lower extremity function was assessed using the short physical performance battery (SPPB), gait speed, and isokinetic leg strength. Linear regression and mixed models were used to determine the cross-sectional and longitudinal associations between vitamin K status and functional outcome measures. RESULTS Cross-sectionally, higher plasma phylloquinone was associated with better SPPB scores and 20-m gait speed (p ≤ .05). After 4-5 years, those with ≥1.0nM plasma phylloquinone (the concentration achieved when recommended intakes are met) had better SPPB scores (p = .03) and 20-m gait speed (p < .05). Lower plasma dp-ucMGP (reflective of better vitamin K status) was associated with better SPPB scores and leg strength cross-sectionally (p ≤ .04), but not longitudinally. Neither measure of vitamin K status was associated with walking endurance or with the rate of decline in function. CONCLUSION Older adults with higher vitamin K status had better physical performance scores at baseline, but data are less consistent longitudinally. Since lower extremity disability is a common consequence of multiple chronic diseases for which a role of vitamin K has been suggested, future studies are needed to determine if vitamin K supplementation could improve function in those with vitamin K insufficiency and clarify underlying mechanism(s).
Collapse
Affiliation(s)
- M Kyla Shea
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, Massachusetts.
| | - Richard F Loeser
- Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill
| | - Fang-Chi Hsu
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem North Carolina
| | - Sarah L Booth
- Tufts University USDA Human Nutrition Research Center on Aging, Boston, Massachusetts
| | - Michael Nevitt
- Department of Epidemiology and Biostatistics, University of California San Francisco
| | | | - Elsa S Strotmeyer
- Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| | | | - Stephen B Kritchevsky
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | |
Collapse
|
39
|
Delanaye P, Liabeuf S, Bouquegneau A, Cavalier É, Massy ZA. [The matrix-gla protein awakening may lead to the demise of vascular calcification]. Nephrol Ther 2015; 11:191-200. [PMID: 25794931 DOI: 10.1016/j.nephro.2014.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 12/03/2014] [Accepted: 12/09/2014] [Indexed: 12/12/2022]
Abstract
Matrix-gla-protein (MGP) is mainly secreted by chondrocytes and smooth vascular muscle cells. This potent inhibitor of vascular calcification need to undergo 2 post-transcriptional steps to be fully active: one phosphorylation of 3 serine residues (on 5) and a carboxylation of 5 glutamate residues (on 9). Like other "Gla" proteins, this carboxylation is vitamin K dependant. Several forms of MGP thus circulate in the plasma, some of them being totally inactive (the unphosphorylated and uncarboxylated MGP), some others being partially or fully active, according to the number of phosphorylated or carboxylated sites. A theoretical link exists between MGP, vitamin K, vascular calcifications and cardiovascular diseases. This link is even more evident in patients suffering from chronic kidney diseases (CKD), and notably hemodialysis patients. If this link has been demonstrated in different experimental studies, clinical studies are mainly observational and their results must be interpreted accordingly. MGP concentrations are definitely not yet a surrogate to estimate the levels of vascular calcification, but could allow the monitoring of vitamin K treatment. Modulation of MGP concentrations may reduce vascular calcification in hemodialyzed patients, if the large ongoing trials show an efficiency of this treatment. In this review, we will summarize the role of MGP in the vascular calcifications process, describe the problems linked to the analytical determination of MGP in plasma and finally describe the different clinical studies on MGP and vascular calcifications in the general population and in CKD patients.
Collapse
Affiliation(s)
- Pierre Delanaye
- Service de néphrologie-dialyse, université de Liège, CHU Sart Tilman, 4000 Liège, Belgique.
| | | | - Antoine Bouquegneau
- Service de néphrologie-dialyse, université de Liège, CHU Sart Tilman, 4000 Liège, Belgique
| | - Étienne Cavalier
- Service de chimie clinique, université de Liège, CHU Sart Tilman, 4000 Liège, Belgique
| | - Ziad A Massy
- Inserm U-1088, UPJV, Amiens, France; Service de néphrologie, hôpital Ambroise-Paré, UVSQ, Boulogne-Billancourt, France
| |
Collapse
|
40
|
Karl JP, Fu X, Wang X, Zhao Y, Shen J, Zhang C, Wolfe BE, Saltzman E, Zhao L, Booth SL. Fecal menaquinone profiles of overweight adults are associated with gut microbiota composition during a gut microbiota-targeted dietary intervention. Am J Clin Nutr 2015; 102:84-93. [PMID: 26016865 DOI: 10.3945/ajcn.115.109496] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/04/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Emerging evidence supports novel roles for vitamin K in cardiometabolic health, some of which may be unique to the bacterially synthesized vitamin K forms known as menaquinones. However, factors influencing menaquinone biosynthesis by the gut microbiota and associations with cardiometabolic health have not been examined. OBJECTIVE The objective of this study was to identify associations between fecal menaquinone profiles, gut microbiota composition, and biomarkers of cardiometabolic health. DESIGN The menaquinone profile and gut microbiota structure were periodically measured in fecal samples collected from 77 overweight Chinese adults who consumed a prescribed diet previously shown to alter gut microbiota composition and to improve cardiometabolic biomarkers. RESULTS Covariance among menaquinones within individual fecal samples partitioned individuals into 2 distinct groups, herein introduced as menaquinotypes of the human gut. Menaquinotypes were characterized by differences in menaquinone (MK) 5 and MK9-MK13 and differences in the relative abundance of several operational taxonomic units (OTUs) delineated at the species level, predominantly within the genera Prevotella spp. and Bacteroides spp. Fecal MK4, MK6, and MK8 decreased during the intervention (P < 0.05); and longitudinal changes in the relative abundance of >100 OTUs were associated with altered fecal content of ≥1 individual menaquinone. The strongest and most consistent relations were between Prevotella spp. and MK5 and MK11-MK13, between Bacteroides spp. and MK9 and MK10, and between Escherichia/Shigella spp. and MK8. Neither individual menaquinones nor menaquinotypes were longitudinally associated with markers of glycemia, insulin resistance, or inflammation. CONCLUSIONS These findings suggest that variability in fecal menaquinone content is predominantly determined by relatively few genera within the gut microbiota and that diet-mediated alterations in gut microbiota composition may provide a feasible approach for altering gut menaquinone content. This trial was registered at the Chinese Clinical Trials Registry as ChiCTR-TRC-09000353.
Collapse
Affiliation(s)
- J Philip Karl
- Vitamin K Laboratory and Energy Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | | | - Xiaoxin Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yufeng Zhao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Shen
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | | | - Edward Saltzman
- Energy Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China; Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China; and
| | | |
Collapse
|
41
|
A review of the effect of diet on cardiovascular calcification. Int J Mol Sci 2015; 16:8861-83. [PMID: 25906474 PMCID: PMC4425113 DOI: 10.3390/ijms16048861] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/19/2015] [Accepted: 04/07/2015] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular (CV) calcification is known as sub-clinical atherosclerosis and is recognised as a predictor of CV events and mortality. As yet there is no treatment for CV calcification and conventional CV risk factors are not consistently correlated, leaving clinicians uncertain as to optimum management for these patients. For this reason, a review of studies investigating diet and serum levels of macro- and micronutrients was carried out. Although there were few human studies of macronutrients, nevertheless transfats and simple sugars should be avoided, while long chain ω-3 fats from oily fish may be protective. Among the micronutrients, an intake of 800 μg/day calcium was beneficial in those without renal disease or hyperparathyroidism, while inorganic phosphorus from food preservatives and colas may induce calcification. A high intake of magnesium (≥380 mg/day) and phylloquinone (500 μg/day) proved protective, as did a serum 25(OH)D concentration of ≥75 nmol/L. Although oxidative damage appears to be a cause of CV calcification, the antioxidant vitamins proved to be largely ineffective, while supplementation of α-tocopherol may induce calcification. Nevertheless other antioxidant compounds (epigallocatechin gallate from green tea and resveratrol from red wine) were protective. Finally, a homocysteine concentration >12 µmol/L was predictive of CV calcification, although a plasma folate concentration of >39.4 nmol/L could both lower homocysteine and protect against calcification. In terms of a dietary programme, these recommendations indicate avoiding sugar and the transfats and preservatives found in processed foods and drinks and adopting a diet high in oily fish and vegetables. The micronutrients magnesium and vitamin K may be worthy of further investigation as a treatment option for CV calcification.
Collapse
|
42
|
Shea MK, Kritchevsky SB, Hsu FC, Nevitt M, Booth SL, Kwoh CK, McAlindon TE, Vermeer C, Drummen N, Harris TB, Womack C, Loeser RF. The association between vitamin K status and knee osteoarthritis features in older adults: the Health, Aging and Body Composition Study. Osteoarthritis Cartilage 2015; 23:370-8. [PMID: 25528106 PMCID: PMC4339507 DOI: 10.1016/j.joca.2014.12.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 12/02/2014] [Accepted: 12/10/2014] [Indexed: 02/02/2023]
Abstract
BACKGROUND Vitamin K-dependent (VKD) proteins, including the mineralization inhibitor matrix-gla protein (MGP), are found in joint tissues including cartilage and bone. Previous studies suggest low vitamin K status is associated with higher osteoarthritis (OA) prevalence and incidence. OBJECTIVE To clarify what joint tissues vitamin K is relevant to in OA, we investigated the cross-sectional and longitudinal association between vitamin K status and knee OA structural features measured using magnetic resonance imaging (MRI). METHODS Plasma phylloquinone (PK, vitamin K1) and dephosphorylated-uncarboxylated MGP ((dp)ucMGP) were measured in 791 older community-dwelling adults who had bilateral knee MRIs (mean ± SD age = 74 ± 3 y; 67% female). The adjusted odds ratios (and 95% confidence intervals) [OR (95%CI)] for presence and progression of knee OA features according to vitamin K status were calculated using marginal models with generalized estimating equations (GEEs), adjusted for age, sex, body mass index (BMI), triglycerides and other pertinent confounders. RESULTS Longitudinally, participants with very low plasma PK (<0.2 nM) were more likely to have articular cartilage and meniscus damage progression after 3 years [OR (95% CIs): 1.7(1.0-3.0), 2.6(1.3-5.2) respectively] compared to sufficient PK (≥ 1.0 nM). Higher plasma (dp)ucMGP (reflective of lower vitamin K status) was associated with higher odds of meniscus damage, osteophytes, bone marrow lesions, and subarticular cysts cross-sectionally [ORs (95% CIs) comparing highest to lowest quartile: 1.6(1.1-2.3); 1.7(1.1-2.5); 1.9(1.3-2.8); 1.5(1.0-2.1), respectively]. CONCLUSION Community-dwelling men and women with very low plasma PK were more likely to have progression of articular cartilage and meniscus damage. Plasma (dp)ucMGP was associated with presence of knee OA features but not progression. Future studies are needed to clarify mechanisms underlying vitamin Ks role in OA.
Collapse
Affiliation(s)
- M K Shea
- USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
| | - S B Kritchevsky
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - F-C Hsu
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - M Nevitt
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - S L Booth
- USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - C K Kwoh
- Division of Rheumatology, University of Arizona, Tucson, AZ, USA
| | - T E McAlindon
- Division of Rheumatology, Tufts Medical Center, Boston, MA, USA
| | | | | | - T B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, USA
| | - C Womack
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - R F Loeser
- Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
43
|
Dashti HS, Shea MK, Smith CE, Tanaka T, Hruby A, Richardson K, Wang TJ, Nalls MA, Guo X, Liu Y, Yao J, Li D, Johnson WC, Benjamin EJ, Kritchevsky SB, Siscovick DS, Ordovás JM, Booth SL. Meta-analysis of genome-wide association studies for circulating phylloquinone concentrations. Am J Clin Nutr 2014; 100:1462-9. [PMID: 25411281 PMCID: PMC4232014 DOI: 10.3945/ajcn.114.093146] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Poor vitamin K status is linked to greater risk of several chronic diseases. Age, sex, and diet are determinants of circulating vitamin K; however, there is still large unexplained interindividual variability in vitamin K status. Although a strong genetic component has been hypothesized, this has yet to be examined by a genome-wide association (GWA) study. OBJECTIVE The objective was to identify common genetic variants associated with concentrations of circulating phylloquinone, the primary circulating form of vitamin K. DESIGN We conducted a 2-stage GWA meta-analysis of circulating phylloquinone in 2 populations of European descent from the Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium Nutrition Working Group. Circulating phylloquinone was measured by using reversed-phase high-performance liquid chromatography. Results from adjusted cohort-specific discovery GWA analyses were meta-analyzed with inverse variance weights (n = 2138). Associations with circulating phylloquinone at P < 1 × 10(-6) were then evaluated in a second-stage analysis consisting of one independent cohort (n = 265). RESULTS No significant association was observed for circulating phylloquinone at the genome-wide significance level of 5 × 10(-8). However, from the discovery GWA, there were 11 single-nucleotide polymorphism (SNP) associations with circulating phylloquinone at P < 1 × 10(-6), including a functional variant previously associated with warfarin dose and altered phylloquinone metabolism. These SNPs are on 5 independent loci on 11q23.3, 8q24.3, 5q22.3, 2p12, and 19p13.12, and they fall within or near the candidate genes APOA1/C3/A4/A5 cluster (involved in lipoprotein metabolism), COL22A1, CDO1, CTNAA2, and CYP4F2 (a phylloquinone oxidase), respectively. Second-stage analysis in an independent cohort further suggests the association of the 5q22.3 locus with circulating phylloquinone (P < 0.05). CONCLUSIONS Multiple candidate genes related to lipoprotein and vitamin K metabolism were identified as potential determinants of circulating phylloquinone. Further investigation with a larger sample is warranted to verify our initial findings and identify other loci contributing to circulating phylloquinone. Trials related to this study were registered at clinicaltrials.gov as NCT00005121 (Framingham Offspring Study) and NCT00005487 (Multi-Ethnic Study of Atherosclerosis).
Collapse
Affiliation(s)
- Hassan S Dashti
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - M Kyla Shea
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Caren E Smith
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Toshiko Tanaka
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Adela Hruby
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Kris Richardson
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Thomas J Wang
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Mike A Nalls
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Xiuqing Guo
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Yongmei Liu
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Jie Yao
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Dalin Li
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - W Craig Johnson
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Emelia J Benjamin
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Stephen B Kritchevsky
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - David S Siscovick
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - José M Ordovás
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| | - Sarah L Booth
- From the Nutrition and Genomics Laboratory (HSD, CES, KR, and JMO), Vitamin K Laboratory (MKS and SLB), Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA; the Translational Gerontology Branch (TT), Laboratory of Neurogenetics (MAN), National Institute on Aging, Baltimore, MD; the Department of Nutrition, Harvard School of Public Health, Boston, MA (AH); the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (TJW); the Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA (XG and JY); the Department of Public Health Sciences (YL), Sticht Center on Aging (SBK), Wake Forest Medical Center, Winston-Salem, NC; Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA (DL); the Department of Biostatistics, University of Washington, Seattle, WA (WCJ); Boston University and National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA (EJB); the Department of Medicine, Boston University School of Medicine, Boston, MA (EJB); New York Academy of Medicine, New York, NY (DSS); the Department of Epidemiology, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain (JMO); and Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain (JMO). Any opinions, findings, conclusions, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture
| |
Collapse
|
44
|
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; 86:286-93. [PMID: 24429407 DOI: 10.1038/ki.2013.530] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 10/26/2013] [Accepted: 10/31/2013] [Indexed: 12/20/2022]
Abstract
In chronic kidney disease, vitamin K-dependent proteins, including the calcification inhibitor matrix Gla protein, are largely uncarboxylated indicating that functional vitamin K deficiency may contribute to uremic vascular calcification. Since the effects of uremia on the vitamin K cycle are unknown, we investigated the influence of uremia and vitamin K supplementation on the activity of the vitamin K cycle and extraosseous calcification. Uremia was induced in rats by an adenine-supplemented diet and vitamin K1 or K2 was administered over 4 and 7 weeks. After 4 weeks of adenine diet, the activity of the vitamin K cycle enzyme γ-carboxylase but not the activities of DT-diaphorase or vitamin K epoxide reductase were reduced. Serum levels of undercarboxylated matrix Gla protein increased, indicating functional vitamin K deficiency. There was no light microscopy-detectable calcification at this stage but chemically determined aortic and renal calcium content was increased. Vitamin K treatment reduced aortic and renal calcium content after 4 weeks. Seven weeks of uremia induced overt calcification in the aorta, heart, and kidneys; however, addition of vitamin K restored intrarenal γ-carboxylase activity and overstimulated it in the liver along with reducing heart and kidney calcification. Thus, uremic vitamin K deficiency may partially result from a reduction of the γ-carboxylase activity which possibly contributes to calcification. Pharmacological vitamin K supplementation restored the vitamin K cycle and slowed development of soft tissue calcification in experimental uremia.
Collapse
Affiliation(s)
- Nadine Kaesler
- Department of Nephrology, RWTH University Hospital Aachen, Aachen, Germany
| | - Elke Magdeleyns
- VitaK BV, University of Maastricht, Maastricht, The Netherlands
| | - Marjolein Herfs
- VitaK BV, University of Maastricht, Maastricht, The Netherlands
| | - Thomas Schettgen
- Department of Environmental and Social Medicine, University Hospital of the RWTH Aachen, Aachen, Germany
| | | | - Danilo Fliser
- Department of Internal Medicine IV, Saarland University Medical Centre, Homburg, Germany
| | - Cees Vermeer
- VitaK BV, University of Maastricht, Maastricht, The Netherlands
| | - Jürgen Floege
- Department of Nephrology, RWTH University Hospital Aachen, Aachen, Germany
| | - Georg Schlieper
- Department of Nephrology, RWTH University Hospital Aachen, Aachen, Germany
| | - Thilo Krüger
- Department of Nephrology, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
45
|
Dalmeijer GW, van der Schouw YT, Booth SL, de Jong PA, Beulens JWJ. Phylloquinone concentrations and the risk of vascular calcification in healthy women. Arterioscler Thromb Vasc Biol 2014; 34:1587-90. [PMID: 24855054 DOI: 10.1161/atvbaha.114.303853] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate the association of plasma phylloquinone concentrations with coronary artery calcification (CAC) and vascular calcification. APPROACH AND RESULTS In a prospective cohort of 508 postmenopausal women, plasma phylloquinone concentrations were measured by high-pressure liquid chromatography. Calcification was measured in the coronary arteries, aortic valve, mitral valve, and thoracic aorta by multidetector computed tomography. To combine these calcification scores, we dichotomized each of the 4 areas into present or absent. Because of the continuous measurement of CAC, we categorized this as calcification present if Agatston score was >0, and calcification score was calculated as the sum of the calcified areas. Multivariate-adjusted prevalence ratios and odds ratios were estimated using Poisson regression and multinomial logistic regression. After 8.5 years of follow-up, 22% of the women had no calcification, whereas 5% had calcification in all measured areas. Detectable phylloquinone concentrations were associated with increased CAC compared with nondetectable phylloquinone concentrations with a prevalence ratio of 1.34 (95% confidence interval, 1.01-1.77). When dividing women with detectable phylloquinone concentrations into low detectable (>0-0.70 nmol/L) and moderate to high detectable (>0.70 nmol/L) phylloquinone concentrations versus nondetectable phylloquinone concentrations, both were associated with increased CAC with a prevalence ratio of 1.32 (95% confidence interval, 0.99-1.76) and 1.36 (95% confidence interval, 1.02-1.81), respectively. Detectable phylloquinone concentrations were not associated with the number of calcified areas with an odds ratio(no versus ≥ 3 areas calcifications) of 1.60 (95% confidence interval, 0.65-3.99; P=0.31). CONCLUSIONS Detectable phylloquinone concentrations are not associated with reduced vascular calcification but seemed to be associated with an increased prevalence of CAC.
Collapse
Affiliation(s)
- Geertje W Dalmeijer
- From the Julius Center for Health Sciences and Primary Care (G.W.D., Y.T.v.d.S., J.W.J.B.) and Department of Radiology (P.A.d.J.), University Medical Center Utrecht, Utrecht, The Netherlands; and Jean Mayer US Department of Agriculture Human Nutrition Research Center of Aging at Tufts University, Boston, MA (S.L.B.).
| | - Yvonne T van der Schouw
- From the Julius Center for Health Sciences and Primary Care (G.W.D., Y.T.v.d.S., J.W.J.B.) and Department of Radiology (P.A.d.J.), University Medical Center Utrecht, Utrecht, The Netherlands; and Jean Mayer US Department of Agriculture Human Nutrition Research Center of Aging at Tufts University, Boston, MA (S.L.B.)
| | - Sarah L Booth
- From the Julius Center for Health Sciences and Primary Care (G.W.D., Y.T.v.d.S., J.W.J.B.) and Department of Radiology (P.A.d.J.), University Medical Center Utrecht, Utrecht, The Netherlands; and Jean Mayer US Department of Agriculture Human Nutrition Research Center of Aging at Tufts University, Boston, MA (S.L.B.)
| | - Pim A de Jong
- From the Julius Center for Health Sciences and Primary Care (G.W.D., Y.T.v.d.S., J.W.J.B.) and Department of Radiology (P.A.d.J.), University Medical Center Utrecht, Utrecht, The Netherlands; and Jean Mayer US Department of Agriculture Human Nutrition Research Center of Aging at Tufts University, Boston, MA (S.L.B.)
| | - Joline W J Beulens
- From the Julius Center for Health Sciences and Primary Care (G.W.D., Y.T.v.d.S., J.W.J.B.) and Department of Radiology (P.A.d.J.), University Medical Center Utrecht, Utrecht, The Netherlands; and Jean Mayer US Department of Agriculture Human Nutrition Research Center of Aging at Tufts University, Boston, MA (S.L.B.)
| |
Collapse
|
46
|
Demer LL, Tintut Y. Inflammatory, metabolic, and genetic mechanisms of vascular calcification. Arterioscler Thromb Vasc Biol 2014; 34:715-23. [PMID: 24665125 PMCID: PMC3975044 DOI: 10.1161/atvbaha.113.302070] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 01/13/2014] [Indexed: 01/06/2023]
Abstract
This review centers on updating the active research area of vascular calcification. This pathology underlies substantial cardiovascular morbidity and mortality, through adverse mechanical effects on vascular compliance, vasomotion, and, most likely, plaque stability. Biomineralization is a complex, regulated process occurring widely throughout nature. Decades ago, its presence in the vasculature was considered a mere curiosity and an unregulated, dystrophic process that does not involve biological mechanisms. Although it remains controversial whether the process has any adaptive value or past evolutionary advantage, substantial advances have been made in understanding the biological mechanisms driving the process. Different types of calcific vasculopathy, such as inflammatory versus metabolic, have parallel mechanisms in skeletal bone calcification, such as intramembranous and endochondral ossification. Recent work has identified important regulatory roles for inflammation, oxidized lipids, elastin, alkaline phosphatase, osteoprogenitor cells, matrix γ-carboxyglutamic acid protein, transglutaminase, osteoclastic regulatory factors, phosphate regulatory hormones and receptors, apoptosis, prelamin A, autophagy, and microvesicles or microparticles similar to the matrix vesicles of skeletal bone. Recent work has uncovered fascinating interactions between matrix γ-carboxyglutamic acid protein, vitamin K, warfarin, and transport proteins. And, lastly, recent breakthroughs in inherited forms of calcific vasculopathy have identified the genes responsible as well as an unexpected overlap of phenotypes. Until recently, vascular calcification was considered a purely degenerative, unregulated process. Since then, investigative groups around the world have identified a wide range of causative mechanisms and regulatory pathways, and some of the recent developments are highlighted in this review.
Collapse
Affiliation(s)
- Linda L. Demer
- Department of Medicine (Cardiology), University of California, Los Angeles Los Angeles, CA 90095-1679
- Department of Physiology and Bioengineering, University of California, Los Angeles Los Angeles, CA 90095-1679
| | - Yin Tintut
- Department of Medicine (Cardiology), University of California, Los Angeles Los Angeles, CA 90095-1679
| |
Collapse
|
47
|
Willems BAG, Vermeer C, Reutelingsperger CPM, Schurgers LJ. The realm of vitamin K dependent proteins: shifting from coagulation toward calcification. Mol Nutr Food Res 2014; 58:1620-35. [PMID: 24668744 DOI: 10.1002/mnfr.201300743] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/27/2013] [Accepted: 01/01/2014] [Indexed: 12/20/2022]
Abstract
In the past few decades vitamin K has emerged from a single-function "haemostasis vitamin" to a "multi-function vitamin." The use of vitamin K antagonists (VKA) inevitably showed that the inhibition was not restricted to vitamin K dependent coagulation factors but also synthesis of functional extrahepatic vitamin K dependent proteins (VKDPs), thereby eliciting undesired side effects. Vascular calcification is one of the recently revealed detrimental effects of VKA. The discovery that VKDPs are involved in vascular calcification has propelled our mechanistic understanding of this process and has opened novel avenues for diagnosis and treatment. This review addresses mechanisms of VKDPs and their significance for physiological and pathological calcification.
Collapse
Affiliation(s)
- Brecht A G Willems
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands; VitaK BV, Maastricht University, Maastricht, The Netherlands
| | | | | | | |
Collapse
|