Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 321
Filter
1.
Neurochem Res ; 49(7): 1838-1850, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38727984

ABSTRACT

Menaquinone-4 (MK-4) is an isoform of vitamin K2 that has been shown to exert various biological actions besides its functions in blood coagulation and bone metabolism. Here we examined the effect of MK-4 on a mouse model of intracerebral hemorrhage (ICH). Daily oral administration of 200 mg/kg MK-4 starting from 3 h after induction of ICH by intrastriatal collagenase injection significantly ameliorated neurological deficits. Unexpectedly, MK-4 produced no significant effects on various histopathological parameters, including the decrease of remaining neurons and the increase of infiltrating neutrophils within the hematoma, the increased accumulation of activated microglia/macrophages and astrocytes around the hematoma, as well as the injury volume and brain swelling by hematoma formation. In addition, ICH-induced increases in nitrosative/oxidative stress reflected by changes in the immunoreactivities against nitrotyrosine and heme oxygenase-1 as well as the contents of malondialdehyde and glutathione were not significantly affected by MK-4. In contrast, MK-4 alleviated axon tract injury in the internal capsule as revealed by neurofilament-H immunofluorescence. Enhanced preservation of the corticospinal tract by MK-4 was also confirmed by retrograde labeling of neurons in the primary motor cortex innervating the spinal cord. These results suggest that MK-4 produces therapeutic effect on ICH by protecting structural integrity of the corticospinal tract.


Subject(s)
Cerebral Hemorrhage , Pyramidal Tracts , Vitamin K 2 , Animals , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Male , Vitamin K 2/analogs & derivatives , Vitamin K 2/pharmacology , Vitamin K 2/therapeutic use , Pyramidal Tracts/drug effects , Pyramidal Tracts/metabolism , Pyramidal Tracts/pathology , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Nervous System Diseases/etiology , Nervous System Diseases/drug therapy
2.
Biomed Pharmacother ; 175: 116697, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38759289

ABSTRACT

Vitamin K2 (VK2) is an effective compound for anti-ferroptosis and anti-osteoporosis, and Semen sojae praeparatum (Dandouchi in Chinese) is the main source of VK2. Chondrocyte ferroptosis and extracellular matrix (ECM) degradation playing a role in the pathogenesis of osteoarthritis (OA). Glutathione peroxidase 4 (GPX4) is the intersection of two mechanisms in regulating OA progression. But no studies have elucidated the therapeutic effects and mechanisms of VK2 on OA. This study utilized an in vivo rat OA model created via anterior cruciate ligament transection (ACLT) and an in vitro chondrocyte oxidative damage model induced by TBHP to investigate the protective effects and mechanisms of action of VK2 in OA. Knee joint pain in mice was evaluated using the Von Frey test. Micro-CT and Safranin O-Fast Green staining were employed to observe the extent of damage to the tibial cartilage and subchondral bone, while immunohistochemistry and PCR were used to examine GPX4 levels in joint cartilage. The effects of VK2 on rat chondrocyte viability were assessed using CCK-8 and flow cytometry assays, and chondrocyte morphology was observed with toluidine blue and alcian blue staining. The impact of VK2 on intracellular ferroptosis-related markers was observed using fluorescent staining and flow cytometry. Protein expression changes were detected by immunofluorescence and Western blot analysis. Furthermore, specific protein inhibitors were applied to confirm the dual-regulatory effects of VK2 on GPX4. VK2 can increase bone mass and cartilage thickness in the subchondral bone of the tibia, and reduce pain and the OARSI score induced by OA. Immunohistochemistry results indicate that VK2 exerts its anti-OA effects by regulating GPX4 to delay ECM degradation. VK2 can inhibit the activation of the MAPK/NFκB signaling pathway caused by reduced expression of intracellular GPX4, thereby decreasing ECM degradation. Additionally, VK2 can reverse the inhibitory effect of RSL3 on GPX4, increase intracellular GSH content and the GSH/GSSG ratio, reduce MDA content, and rescue chondrocyte ferroptosis. The protective mechanism of VK2 may involve its dual-target regulation of GPX4, reducing chondrocyte ferroptosis and inhibiting the MAPK/NFκB signaling pathway to decelerate the degradation of the chondrocyte extracellular matrix.


Subject(s)
Chondrocytes , Extracellular Matrix , Ferroptosis , Osteoarthritis , Phospholipid Hydroperoxide Glutathione Peroxidase , Rats, Sprague-Dawley , Vitamin K 2 , Animals , Ferroptosis/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Male , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Rats , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Mice , Vitamin K 2/pharmacology , Vitamin K 2/analogs & derivatives , Mice, Inbred C57BL , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Disease Models, Animal , Signal Transduction/drug effects , Cells, Cultured
3.
Toxicol In Vitro ; 98: 105839, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723978

ABSTRACT

Vitamin K possesses efficacy as a topical dermatological agent. However, vitamin K is phototoxic and susceptible to photodegradation. Herein, we investigated the mechanisms underlying the phototoxicity of phylloquinone (PK, vitamin K1) and menaquinone-4 (MK-4, vitamin K2) under ultraviolet A (UVA) irradiation using various reactive oxygen species (ROS) scavengers. This resulted in the production of superoxide anion radicals via type I and singlet oxygen via type II photodynamic reactions, which were quenched by the ROS scavengers: superoxide dismutase and sodium azide (NaN3). In HaCaT cells, MK-4 and PK induced the production of intracellular ROS, particularly hydrogen peroxide, in response to UVA irradiation. Furthermore, the addition of catalase successfully decreased maximum ROS levels by approximately 30%. NaN3 and catalase decreased the maximum reduction in cell viability induced by UVA-irradiated PK and MK-4 in cell viability by approximately 2-7-fold. Additionally, ROS scavengers had no effect on the photodegradation of PK or MK-4 at 373 nm. Therefore, the phototoxicities of PK and MK-4 were attributed to the generation of singlet oxygen and hydrogen peroxide, underscoring the importance of photoshielding in circumventing phototoxicity.


Subject(s)
Cell Survival , Free Radical Scavengers , Reactive Oxygen Species , Ultraviolet Rays , Reactive Oxygen Species/metabolism , Humans , Free Radical Scavengers/pharmacology , Cell Survival/drug effects , Sodium Azide/pharmacology , Sodium Azide/toxicity , Cell Line , Vitamin K 2/analogs & derivatives , Vitamin K 2/pharmacology , Vitamin K 1/pharmacology , Dermatitis, Phototoxic , Catalase/metabolism , HaCaT Cells , Superoxide Dismutase/metabolism
4.
Int Immunopharmacol ; 134: 112257, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38759366

ABSTRACT

BACKGROUND: Hypoxic-ischemic encephalopathy (HIE) is a major contributor to neonatal mortality and neurodevelopmental disorders, but currently there is no effective therapy drug for HIE. Mitochondrial dysfunction plays a pivotal role in hypoxic-ischemic brain damage(HIBD). Menaquinone-4 (MK-4), a subtype of vitamin K2 prevalent in the brain, has been shown to enhance mitochondrial function and exhibit protective effects against ischemia-reperfusion injury. However, the impact and underlying molecular mechanism of MK-4 in HIE have not been fully elucidated. METHODS: In this study, we established the neonatal rats HIBD model in vivo and oxygen-glucose deprivation and reperfusion (OGD/R) of primary neurons in vitro to explore the neuroprotective effects of MK-4 on HI damage, and illuminate the potential mechanism. RESULTS: Our findings revealed that MK-4 ameliorated mitochondrial dysfunction, reduced oxidative stress, and prevented HI-induced neuronal apoptosis by activating the Sirt1-PGC-1α-TFAM signaling pathway through Sirt1 mediation. Importantly, these protective effects were partially reversed by EX-527, a Sirt1 inhibitor. CONCLUSION: Our study elucidated the potential therapeutic mechanism of MK-4 in neonatal HIE, suggesting its viability as an agent for enhancing recovery from HI-induced cerebral damage in newborns. Further exploration into MK-4 could lead to novel interventions for HIE therapy.


Subject(s)
Animals, Newborn , Apoptosis , Hypoxia-Ischemia, Brain , Mitochondria , Neurons , Neuroprotective Agents , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 1 , Vitamin K 2 , Animals , Sirtuin 1/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Signal Transduction/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Vitamin K 2/analogs & derivatives , Vitamin K 2/pharmacology , Vitamin K 2/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Rats , Neurons/drug effects , Neurons/pathology , Apoptosis/drug effects , Oxidative Stress/drug effects , Cells, Cultured , Disease Models, Animal , Transcription Factors/metabolism , Brain/drug effects , Brain/pathology , Brain/metabolism
5.
Free Radic Biol Med ; 210: 416-429, 2024 01.
Article in English | MEDLINE | ID: mdl-38042225

ABSTRACT

BACKGROUND: Menaquinone-4(MK-4), the isoform of vitamin K2 in the brain, exerts neuroprotective effects against a variety of central nervous system disorders. This study aimed to demonstrate the anti-ferroptosis effects of MK-4 in neurons after SAH. METHODS: A subarachnoid hemorrhage (SAH) model was prepared by endovascular perforation in mice. In vitro hemoglobin stimulation of primary cortical neurons mimicked SAH. MK-4, Brequinar (BQR, DHODH inhibitor), and Selisistat (SEL, SIRT1 inhibitor) were administered, respectively. Subsequently, WB, immunofluorescence was used to determine protein expression and localization, and transmission electron microscopy was used to observe neuronal mitochondrial structure while other indicators of ferroptosis were measured. RESULTS: MK-4 treatment significantly upregulated the protein levels of DHODH; decreased GSH, PTGS2, NOX1, ROS, and restored mitochondrial membrane potential. Meanwhile, MK-4 upregulated the expression of SIRT1 and promoted its entry into the nucleus. BQR or SEL partially abolished the protective effect of MK-4 on, neurologic function, and ferroptosis. CONCLUSIONS: Taken together, our results suggest that MK-4 attenuates ferroptosis after SAH by upregulating DHODH through the activation of SIRT1.


Subject(s)
Brain Injuries , Ferroptosis , Subarachnoid Hemorrhage , Rats , Mice , Animals , Rats, Sprague-Dawley , Dihydroorotate Dehydrogenase , Vitamin K 2/pharmacology , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Brain Injuries/metabolism
6.
Inflammopharmacology ; 31(5): 2675-2684, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37477796

ABSTRACT

Recent studies have shown that, coupled with other environmental factors, aluminium exposure may lead to neurodegeneration resulting in cognitive impairment resembling Alzheimer's disease. Menaquinone, a form of vitamin K2, aids in maintaining healthy bones and avoids coronary calcification. It also has anti-inflammatory and antioxidant properties. Here, we study the neuroprotective effects of vitamin K2 (MK-7) using the animal model of Alzheimer's disease (AD). Aluminium chloride (AlCl3; 100 mg/kg for 3 weeks orally) was administered to Swiss albino mice to induce neurodegeneration and Vitamin K2 (100 g/kg for 3 weeks orally) was applied as treatment. This was followed by behavioural studies to determine memory changes. The behavioural observations correlated with proinflammatory, oxidative, and brain histopathological changes in AlCl3-treated animals with or without vitamin K2 treatment. AlCl3 administration led to memory decline which was partially restored in Vitamin K2 treated animals. Myeloperoxidase levels in the brain increased due to AlCl3-mediated inflammation, which Vitamin K2 prevented. The acetylcholine esterase and oxidative stress markers induced by AlCl3 were reversed by Vitamin K2. Also, Vitamin K2 helps to restore hippocampal BDNF levels and reduced the amyloid ß accumulation in AlCl3-administered animals. Additionally, Vitamin K2 protected the hippocampal neurons against AlCl3-mediated damage as observed in histopathological studies. We conclude that Vitamin K2 could partially reverse AlCl3-mediated cognitive decline. It increases acetylcholine and BDNF levels while reducing oxidative stress, neuroinflammation, and ß-amyloid deposition, thus protecting the hippocampal neurons from AlCl3-mediated damage.


Subject(s)
Alzheimer Disease , Neuroprotective Agents , Mice , Animals , Aluminum Chloride/pharmacology , Vitamin K 2/pharmacology , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Aluminum Compounds/toxicity , Chlorides/pharmacology , Acetylcholine/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Oxidative Stress
7.
Nutrients ; 15(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37299386

ABSTRACT

BACKGROUND: There is a very high prevalence of subclinical vitamin K deficiency in patients requiring hemodialysis (HD), and this problem is associated with vascular calcification and arterial stiffness. Vitamin K2 (MK-7) supplementation can improve vitamin K status in HD patients. However, the benefits of vitamin K supplementation on arterial stiffness have still not been established. The present study was conducted to evaluate the efficacy of menaquinone-7 (MK-7) supplementation on arterial stiffness in chronic HD patients. METHODS: This open-label multicenter randomized clinical trial was conducted in 96 HD patients who had arterial stiffness, defined by high carotid femoral pulse wave velocity (cfPWV ≥ 10 m/s). The patients were randomly assigned to receive oral MK-7 (375 mcg once daily) for 24 weeks (n = 50) or standard care (control group; n = 46). The change in cfPWV was the primary outcome. RESULTS: Baseline parameters were comparable between the two groups. There was no significant difference in the change in cPWV at 24 weeks between the MK-7 group and standard care [-6.0% (-20.2, 2.3) vs. -6.8% (-19.0, 7.3), p = 0.24]. However, we found that MK-7 significantly decreased cPWV in patients with diabetes [-10.0% (-15.9, -0.8) vs. 3.8% (-5.8, 11.6), p = 0.008]. In addition, the MK-7 group had a lower rate of arterial stiffness progression, compared to controls (30.2% vs. 39.5%, p = 0.37), especially in diabetes patients (21.4% vs. 72.7%, p = 0.01). No serious adverse events were observed during the 24 weeks. CONCLUSION: Vitamin K supplements provided a beneficial impact in lowering the rate of arterial stiffness progression in chronic hemodialysis patients with diabetes. Possible benefits on cardiovascular outcomes require further investigation.


Subject(s)
Vascular Stiffness , Humans , Vitamin K 2/pharmacology , Pulse Wave Analysis , Renal Dialysis/adverse effects , Vitamin K/pharmacology , Dietary Supplements
8.
Int J Mol Sci ; 24(10)2023 May 16.
Article in English | MEDLINE | ID: mdl-37240169

ABSTRACT

During aging, bone marrow mesenchymal stromal cells (MSCs)-the precursors of osteoblasts-undergo cellular senescence, losing their osteogenic potential and acquiring a pro-inflammatory secretory phenotype. These dysfunctions cause bone loss and lead to osteoporosis. Prevention and intervention at an early stage of bone loss are important, and naturally active compounds could represent a valid help in addition to diet. Here, we tested the hypothesis that the combination of two pro-osteogenic factors, namely orthosilicic acid (OA) and vitamin K2 (VK2), and three other anti-inflammatory compounds, namely curcumin (CUR), polydatin (PD) and quercetin (QCT)-that mirror the nutraceutical BlastiMin Complex® (Mivell, Italy)-would be effective in promoting MSC osteogenesis, even of replicative senescent cells (sMSCs), and inhibiting their pro-inflammatory phenotype in vitro. Results showed that when used at non-cytotoxic doses, (i) the association of OA and VK2 promoted MSC differentiation into osteoblasts, even when cultured without other pro-differentiating factors; and (ii) CUR, PD and QCT exerted an anti-inflammatory effect on sMSCs, and also synergized with OA and VK2 in promoting the expression of the pivotal osteogenic marker ALP in these cells. Overall, these data suggest a potential role of using a combination of all of these natural compounds as a supplement to prevent or control the progression of age-related osteoporosis.


Subject(s)
Bone Diseases, Metabolic , Curcumin , Mesenchymal Stem Cells , Osteoporosis , Humans , Osteogenesis , Quercetin/therapeutic use , Vitamin K 2/pharmacology , Vitamin K 2/metabolism , Curcumin/pharmacology , Bone Marrow/metabolism , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Bone Diseases, Metabolic/metabolism , Cells, Cultured , Bone Marrow Cells
9.
BMJ Open ; 13(5): e071885, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37208133

ABSTRACT

INTRODUCTION: Vitamin K has been suggested to have protective effects against progression of vascular calcification and development of cardiovascular disease (CVD). However, few well-powered randomised controlled trials have examined whether vitamin K prevents progression of vascular calcification in individuals from the general population. The aim of the InterVitaminK trial is to investigate the effects of vitamin K supplementation (menaquinone-7, MK-7) on cardiovascular, metabolic, respiratory and bone health in a general ageing population with detectable vascular calcification. METHODS AND ANALYSIS: The InterVitaminK trial is a randomised, double-blinded, placebo-controlled, trial. A total of 450 men and women aged 52-82 years with detectable coronary artery calcification (CAC), but without manifest CVD, will be randomised (1:1) to receive daily MK-7 (333 µg/day) or placebo tablets for 3 years. Health examinations are scheduled at baseline, and after 1, 2 and 3 years of intervention. Health examinations include cardiac CT scans, measurements of arterial stiffness, blood pressure, lung function, physical function, muscle strength, anthropometric measures, questionnaires on general health and dietary intake, and blood and urine sampling. The primary outcome is progression of CAC from baseline to 3-year follow-up. The trial has 89% power to detect a between-group difference of at least 15%. Secondary outcomes are bone mineral density, pulmonary function and biomarkers of insulin resistance. ETHICS AND DISSEMINATION: Oral MK-7 supplementation is considered safe and has not been found to cause severe adverse events. The Ethical Committee of the Capital Region (H-21033114) approved the protocol. Written informed consent is obtained from all participants and the trial is conducted in accordance with the Declaration of Helsinki II. Both negative and positive findings will be reported. TRIAL REGISTRATION NUMBER: NCT05259046.


Subject(s)
Coronary Artery Disease , Vascular Calcification , Male , Humans , Female , Vitamin K , Bone Density , Vitamin K 2/pharmacology , Vitamin K 2/therapeutic use , Lung , Coronary Artery Disease/drug therapy , Vascular Calcification/prevention & control , Dietary Supplements , Denmark , Double-Blind Method , Randomized Controlled Trials as Topic
10.
Physiol Behav ; 269: 114252, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37257737

ABSTRACT

Vitamin K2/ Menaquinones produced predominantly by the gut microbiome improve bone health and prevent coronary calcification. The central nervous system has been linked with gut microbiota via the gut-brain axis and is strongly associated with psychiatric conditions. In the present study, we show the role of Vitamin K2 (MK-7) in gut dysbiosis-associated cognitive decline. Gut dysbiosis was induced in mice by administering Ampicillin (250 mg/kg twice a day orally) for 14 days and Vitamin K2 (0.05 mg/kg) for 21 days with or without antibiotic treatment and altered gene expression profile of intestinal microbes determined. This was followed by behavioural studies to determine cognitive changes. The behavioural observations are then correlated with proinflammatory, oxidative, and brain and intestinal histopathological changes in antibiotic-treated animals with or without vitamin K2 administration. With the use of antibiotics, Lactobacillus, Bifidobacterium, Firmicutes, and Clostridium's relative abundance reduced. When vitamin K2 was added to the medication, their levels were restored. Cognitive impairment was observed in behavioural trials in the antibiotic group, but this drop was restored in mice given both an antibiotic and vitamin K. Myeloperoxidase levels in the colon and brain increased due to gut dysbiosis, which vitamin K2 prevented. The acetylcholine esterase and oxidative stress markers brought on by antibiotics were also decreased by vitamin K2. Additionally, vitamin K2 guarded against alterations in intestine ultrastructure brought on by antibiotic use and preserved hippocampus neurons. So, it can be concluded that vitamin K2 improved cognitive skills, avoided hippocampus neuronal damage from antibiotics, and lowered intestine and brain inflammation and oxidative stress.


Subject(s)
Cognitive Dysfunction , Neuroprotective Agents , Mice , Animals , Vitamin K 2/pharmacology , Vitamin K 2/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Dysbiosis/complications , Dysbiosis/drug therapy , Anti-Bacterial Agents/pharmacology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/complications
11.
J Steroid Biochem Mol Biol ; 231: 106307, 2023 07.
Article in English | MEDLINE | ID: mdl-37030416

ABSTRACT

K vitamins are well known as essential cofactors for hepatic γ-carboxylation of coagulation factors, but their potential role in chronic diseases including cancer is understudied. K2, the most abundant form of vitamin K in tissues, exerts anti-cancer effects via diverse mechanisms which are not completely understood. Our studies were prompted by previous work demonstrating that the K2 precursor menadione synergized with 1,25 dihydroxyvitamin D3 (1,25(OH)2D3) to inhibit growth of MCF7 luminal breast cancer cells. Here we assessed whether K2 modified the anti-cancer effects of 1,25(OH)2D3 in triple negative breast cancer (TNBC) cell models. We examined the independent and combined effects of these vitamins on morphology, cell viability, mammosphere formation, cell cycle, apoptosis and protein expression in three TNBC cell models (MDA-MB-453, SUM159PT, Hs578T). We found that all three TNBC cell lines expressed low levels of the vitamin D receptor (VDR) and were modestly growth inhibited by 1,25(OH)2D3 in association with cell cycle arrest in G0/G1. Induction of differentiated morphology by 1,25(OH)2D3 was observed in two of the cell lines (MDA-MB-453, Hs578T). Treatment with K2 alone reduced viability of MDA-MB-453 and SUM159PT cells but not Hs578T cells. Co-treatment with 1,25(OH)2D3 and K2 significantly reduced viable cell number relative to either treatment alone in Hs578T and SUM159PT cells. The combination treatment induced G0/G1 arrest in MDA-MB-453 cells, Hs578T and SUM159PT cells. Combination treatment altered mammosphere size and morphology in a cell specific manner. Of particular interest, treatment with K2 increased VDR expression in SUM159PT cells suggesting that the synergistic effects in these cells may be secondary to increased sensitivity to 1,25(OH)2D3. The phenotypic effects of K2 in TNBC cells did not correlate with γ-carboxylation suggesting non-canonical actions. In summary, 1,25(OH)2D3 and K2 exert tumor suppressive effects in TNBC cells, inducing cell cycle arrest leading to differentiation and/or apoptosis depending on the specific cell line. Further mechanistic studies to clarify common and unique targets of these two fat soluble vitamins in TNBC are warranted.


Subject(s)
Calcitriol , Triple Negative Breast Neoplasms , Humans , Calcitriol/pharmacology , Vitamin K 2/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Tumor Cells, Cultured , Receptors, Calcitriol/metabolism , Vitamin K , Vitamins/pharmacology
12.
Acta Orthop Traumatol Turc ; 57(1): 17-22, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36939360

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the effects of vitamin K2 on fracture healing. METHODS: Twenty-four 6-week-old male Wistar albino rats that had open tibia fractures induced were included in this study. They were divided into 2 groups of 12, a group that had vitamin K2 administered over 30 consecutive days and a control group. After 30 days, the rats were sacrificed, and from each group, 6 tibiae were selected for biomechanical testing to examine the mechanical strength of the callus tissue using the Instron 3-point bending test and 6 tibiae were selected for histological analysis to examine the density and organization of callus tissue using Allen's grading system and Huo et al's grading system. Furthermore, weekly x-rays were taken to evaluate bone union described by Lane and Sandhu, and osteocalcin, procollagen I N-terminal propeptide, and procollagen I C-terminal propeptide were examined in blood samples taken by intracardiac puncture during sacrification. RESULTS: Breaking force (P = .047), breaking time (P = .019), stiffness (P = .039), fracture strength (P = .041), and Young's modulus (P = .032) showed a statistically significant increase in the K2 group. Procollagen I C-terminal propeptide (P = .024), procollagen I N-terminal propeptide (.047), and osteocalcin (.048) levels were significantly higher in the K2 group compared to the control group. Furthermore, 3rd-week x-rays showed higher bone union scores according to the Lane and Sandhu method in the K2 group (P = .014). However, the histological grading systems of Allen and Huo et al did not show statistically significant differences between groups (P = .086, P = .07, respectively). CONCLUSION: In light of these findings, it could be concluded that vitamin K2 has a significant positive effect on fracture healing.


Subject(s)
Fracture Healing , Tibial Fractures , Animals , Rats , Male , Vitamin K 2/pharmacology , Tibia/diagnostic imaging , Tibia/pathology , Osteocalcin/pharmacology , Procollagen/pharmacology , Rats, Wistar , Tibial Fractures/diagnostic imaging , Tibial Fractures/drug therapy , Biomechanical Phenomena
13.
J Cardiovasc Pharmacol ; 81(5): 381-388, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36857749

ABSTRACT

ABSTRACT: Myocardial infarction is among the leading causes of mortality. Menaquinone-4 (MK-4), a vitamin K2 analog, might play a role in rescuing cardiac ischemia/reperfusion (I/R) injury. This work aimed to discover the potential cardioprotective role of MK-4 against myocardial I/R injury in rats. Thirty-two rats were categorized into 3 groups: (I/R) control group: subjected to I/R protocol (received vehicle), MK-4 preconditioning group: MK-4 infusion for 20 minutes before the I/R protocol, and MK-4 postconditioning group: MK-4 infusion for 20 minutes at the start of the reperfusion phase. The hearts were placed in the Langendorff apparatus, and the left ventricular developed pressure (LVDP), heart rate (HR), + (LV dP/dt) max, - (LV dP/dt) max, and Tau were calculated. The necrotic mass was determined by staining it with nitro blue tetrazolium. Creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and C- reactive protein (CRP), as well as cardiac superoxide dismutase (SOD), nitric oxide (NOx), malondialdehyde (MDA), and glutathione (GSH) levels were all evaluated. MK-4 postconditioning significantly reduced myocardial infarct size; increased LVDP, + (LV dp/dt) max, - (LV dp/dt) max, and HR; reduced Tau, CK-MB, LDH, CRP, IL-6, TNF-α, MDA, and NOx levels; and increased SOD activity, whereas no significant difference in the GSH level was detected. In conclusion, these data imply that MK-4 may protect the heart from the consequences of I/R.


Subject(s)
Myocardial Reperfusion Injury , Tumor Necrosis Factor-alpha , Rats , Animals , Vitamin K 2/pharmacology , Vitamin K 2/metabolism , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Oxidative Stress , Myocardial Reperfusion Injury/pathology , Superoxide Dismutase/metabolism , Glutathione , Myocardium/pathology
14.
Philos Trans R Soc Lond B Biol Sci ; 378(1871): 20220035, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36633276

ABSTRACT

Menaquinones (MKs) are electron carriers in bacterial respiratory chains. In Staphylococcus aureus (Sau), MKs are essential for aerobic and anaerobic respiration. As MKs are redox-active, their biosynthesis likely requires tight regulation to prevent disruption of cellular redox balance. We recently found that the Mycobacterium tuberculosis MenD, the first committed enzyme of the MK biosynthesis pathway, is allosterically inhibited by the downstream metabolite 1,4-dihydroxy-2-naphthoic acid (DHNA). To understand if this is a conserved mechanism in phylogenetically distant genera that also use MK, we investigated whether the Sau-MenD is allosterically inhibited by DHNA. Our results show that DHNA binds to and inhibits the SEPHCHC synthase activity of Sau-MenD enzymes. We identified residues in the DHNA binding pocket that are important for catalysis (Arg98, Lys283, Lys309) and inhibition (Arg98, Lys283). Furthermore, we showed that exogenous DHNA inhibits the growth of Sau, an effect that can be rescued by supplementing the growth medium with MK-4. Our results demonstrate that, despite a lack of strict conservation of the DHNA binding pocket between Mtb-MenD and Sau-MenD, feedback inhibition by DHNA is a conserved mechanism in Sau-MenD and hence the Sau MK biosynthesis pathway. These findings may have implications for the development of anti-staphylococcal agents targeting MK biosynthesis. This article is part of the theme issue 'Reactivity and mechanism in chemical and synthetic biology'.


Subject(s)
Naphthalenes , Staphylococcus aureus , Vitamin K 2/pharmacology , Vitamin K 2/metabolism , Staphylococcus aureus/metabolism , Feedback , Naphthalenes/pharmacology
15.
mBio ; 14(1): e0330222, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36625597

ABSTRACT

Aminoglycosides (AG) have been used against Gram-negative bacteria for decades. Yet, how bacterial metabolism and environmental conditions modify AG toxicity is poorly understood. Here, we show that the level of AG susceptibility varies depending on the nature of the respiratory chain that Escherichia coli uses for growth, i.e., oxygen, nitrate, or fumarate. We show that all components of the fumarate respiratory chain, namely, hydrogenases 2 and 3, the formate hydrogenlyase complex, menaquinone, and fumarate reductase are required for AG-mediated killing under fumarate respiratory conditions. In addition, we show that the AAA+ ATPase RavA and its Von Wildebrand domain-containing partner, ViaA, are essential for AG to act under fumarate respiratory conditions. This effect was true for all AG that were tested but not for antibiotics from other classes. In addition, we show that the sensitizing effect of RavA-ViaA is due to increased gentamicin uptake in a proton motive force-dependent manner. Interestingly, the sensitizing effect of RavA-ViaA was prominent in poor energy conservation conditions, i.e., with fumarate, but dispensable under high energy conservation conditions, i.e., in the presence of nitrate or oxygen. We propose that RavA-ViaA can facilitate uptake of AG across the membrane in low-energy cellular states. IMPORTANCE Antibiotic resistance is a major public health, social, and economic problem. Aminoglycosides (AG) are known to be highly effective against Gram-negative bacteria, but their use is limited to life-threatening infections because of their nephrotoxicity and ototoxicity at therapeutic dose. Elucidation of AG-sensitization mechanisms in bacteria would allow reduced effective doses of AG. Here, we have identified the molecular components involved in anaerobic fumarate respiration that are required for AG to kill. In addition to oxidoreductases and menaquinone, this includes new molecular players, RavA, an AAA+ ATPase, and ViaA, its partner that has the VWA motif. Remarkably, the influence of RavA-ViaA on AG susceptibility varies according to the type of bioenergetic metabolism used by E. coli. This is a significant advance because anaerobiosis is well known to reduce the antibacterial activity of AG. This study highlights the critical importance of the relationship between culture conditions, metabolism, and antibiotic susceptibility.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Aminoglycosides/pharmacology , Nitrates/metabolism , Vitamin K 2/metabolism , Vitamin K 2/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Energy Metabolism , Succinate Dehydrogenase , Bacteria/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Fumarates/pharmacology , Fumarates/metabolism , Anaerobiosis , Adenosine Triphosphatases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
16.
Theriogenology ; 198: 183-193, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36592516

ABSTRACT

The effect of vitamin K on bovine endometrial epithelial cells has not been thoroughly investigated. The objective of this study was to examine the effect of the biologically active form of vitamin K, menaquinone-4, on gene expression in bovine endometrial epithelial cells. First, we examined the mRNA and protein expression levels of UBIAD1, a menaquinone-4 biosynthetic enzyme. Second, we screened for potential target genes of menaquinone-4 in bovine endometrial epithelial cells using RNA-sequencing. We found 50 differentially expressed genes; 42 were upregulated, and 8 were downregulated. Among them, a dose-dependent response to menaquinone-4 was observed for the top three upregulated (TRIB3, IL6, and TNFAIP3) and downregulated (CDC6, ORC1, and RRM2) genes. It has been suggested that these genes play important roles in reproductive events. In addition, GDF15 and VEGFA, which are important for cellular functions as they are commonly involved in pathways, such as positive regulation of cell communication, cell differentiation, and positive regulation of MAPK cascade, were upregulated in endometrial epithelial cells by menaquinone-4 treatment. To the best of our knowledge, this is the first study showing the expression of UBIAD1 in the bovine uterus. Moreover, the study determined menaquinone-4 target genes in bovine endometrial epithelial cells, which may positively affect pregnancy with alteration of gene expression in cattle uterus.


Subject(s)
Endometrium , Vitamin K , Female , Cattle , Animals , Vitamin K 2/metabolism , Vitamin K 2/pharmacology , Vitamin K/metabolism , Endometrium/metabolism , Epithelial Cells/metabolism
17.
J Agric Food Chem ; 71(6): 2745-2761, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36719855

ABSTRACT

Type 2 diabetic osteoporosis (T2DOP) is a chronic bone metabolic disease. Compared with traditional menopausal osteoporosis, the long-term high glucose (HG) microenvironment increases patients' risk of fracture and osteonecrosis. We were accumulating evidence that implicated ferroptosis as a pivotal mechanism of glucolipotoxicity-mediated death of osteocytes and osteoblast, a novel form of programmed cell death resulting from uncontrolled lipid peroxidation depending on iron. Vitamin K2 (VK2), a fat-soluble vitamin, is clinically applied to prevent osteoporosis and improve coagulation. This study aimed to clarify the role and mechanism of VK2 in HG-mediated ferroptosis. We established the mouse T2DOP model by intraperitoneal injection of streptozotocin solution and a high-fat and high-sugar diet. We also cultured bone marrow mesenchymal stem cells (BMSCs) in HG to simulate the diabetic environment in vitro. Based on our data, VK2 inhibited HG-mediated bone loss and ferroptosis, the latter manifested by decreased levels of mitochondrial reactive oxygen species, lipid peroxidation, and malondialdehyde and increased glutathione in vitro. In addition, VK2 treatment was capable of restoring bone mass and strengthening the expression of SIRT1, GPX4, and osteogenic markers in the distal femurs. As for further mechanism exploration, we found that VK2 could activate AMPK/SIRT1 signaling, and knockdown of SIRT1 by siRNA prevented the VK2-mediated positive effect in HG-cultured BMSCs. Summarily, VK2 could ameliorate T2DOP through the activation of the AMPK/SIRT1 signaling pathway to inhibit ferroptosis.


Subject(s)
Diabetes Mellitus, Type 2 , Ferroptosis , Osteoporosis , Mice , Animals , Ferroptosis/genetics , Vitamin K 2/pharmacology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Osteoporosis/drug therapy , Osteoporosis/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics
18.
Am J Transplant ; 23(4): 520-530, 2023 04.
Article in English | MEDLINE | ID: mdl-36695702

ABSTRACT

Vitamin K deficiency is common among kidney transplant recipients (KTRs) and likely contributes to progressive vascular calcification and stiffness. In this single-center, randomized, double-blind, placebo-controlled trial, we aimed to investigate the effects of vitamin K supplementation on the primary end point, serum calcification propensity (calciprotein particle maturation time, T50), and secondary end points arterial stiffness (pulse wave velocity [PWV]) and vitamin K status in 40 vitamin K-deficient KTRs (plasma dephosphorylated uncarboxylated matrix Gla protein [dp-ucMGP] ≥500 pmol/L). Participants (35% female; age, 57 ± 13 years) were randomized 1:1 to vitamin K2 (menaquinone-7, 360 µg/day) or placebo for 12 weeks. Vitamin K supplementation had no effect on calcification propensity (change in T50 vs baseline +2.3 ± 27.4 minutes) compared with placebo (+0.8 ± 34.4 minutes; Pbetween group = .88) but prevented progression of PWV (change vs baseline -0.06 ± 0.26 m/s) compared with placebo (+0.27 ± 0.43 m/s; Pbetween group = .010). Vitamin K supplementation strongly improved vitamin K status (change in dp-ucMGP vs baseline -385 [-631 to -269] pmol/L) compared with placebo (+39 [-188 to +183] pmol/L; Pbetween group < .001), although most patients remained vitamin K-deficient. In conclusion, vitamin K supplementation did not alter serum calcification propensity but prevented progression of arterial stiffness, suggesting that vitamin K has vascular effects independent of calciprotein particles. These results set the stage for longer-term intervention studies with vitamin K supplementation in KTRs. TRIAL REGISTRY: EU Clinical Trials Register (EudraCT Number: 2019-004906-88) and the Dutch Trial Register (NTR number: NL7687).


Subject(s)
Kidney Transplantation , Vascular Stiffness , Humans , Female , Adult , Middle Aged , Aged , Male , Vitamin K/pharmacology , Kidney Transplantation/adverse effects , Pulse Wave Analysis , Vitamin K 2/therapeutic use , Vitamin K 2/pharmacology , Dietary Supplements , Double-Blind Method
19.
Nephrol Dial Transplant ; 38(3): 746-756, 2023 02 28.
Article in English | MEDLINE | ID: mdl-35641194

ABSTRACT

BACKGROUND: Vitamin K activates matrix Gla protein (MGP), a key inhibitor of vascular calcification. There is a high prevalence of sub-clinical vitamin K deficiency in patients with end-stage kidney disease. METHODS: A parallel randomized placebo-controlled pilot trial was designed to determine whether 10 mg of phylloquinone thrice weekly versus placebo modifies coronary artery calcification progression over 12 months in patients requiring hemodialysis with a coronary artery calcium score (CAC) ≥30 Agatston Units (ClinicalTrials.gov identifier NCT01528800). The primary outcome was feasibility (recruitment rate, compliance with study medication, study completion and adherence overall to study protocol). CAC score was used to assess calcification at baseline and 12 months. Secondary objectives were to explore the impact of phylloquinone on vitamin K-related biomarkers (phylloquinone, dephospho-uncarboxylated MGP and the Gla-osteocalcin to Glu-osteocalcin ratio) and events of clinical interest. RESULTS: A total of 86 patients with a CAC score ≥30 Agatston Units were randomized to either 10 mg of phylloquinone or a matching placebo three times per week. In all, 69 participants (80%) completed the trial. Recruitment rate (4.4 participants/month) and medication compliance (96%) met pre-defined feasibility criteria of ≥4.17 and ≥90%, respectively. Patients randomized to phylloquinone for 12 months had significantly reduced levels of dephospho-uncarboxylated MGP (86% reduction) and increased levels of phylloquinone and Gla-osteocalcin to Glu-osteocalcin ratio compared with placebo. There was no difference in the absolute or relative progression of coronary artery calcification between groups. CONCLUSION: We demonstrated that phylloquinone treatment improves vitamin K status and that a fully powered randomized trial may be feasible.


Subject(s)
Coronary Artery Disease , Vascular Calcification , Humans , Vitamin K/therapeutic use , Vitamin K 1/therapeutic use , Osteocalcin/therapeutic use , Pilot Projects , Coronary Artery Disease/drug therapy , Vascular Calcification/drug therapy , Calcium-Binding Proteins , Extracellular Matrix Proteins , Renal Dialysis , Vitamin K 2/pharmacology
20.
Nephrol Dial Transplant ; 38(10): 2131-2142, 2023 09 29.
Article in English | MEDLINE | ID: mdl-36460034

ABSTRACT

BACKGROUND: Vitamin K deficiency is highly prevalent in patients on dialysis and may contribute to their low bone mineral density (BMD) and increased risk of fracture. This study investigated the effect of menaquinone-7 (MK-7) supplementation on BMD in patients on chronic dialysis. METHODS: In a multicentre, double-blind, placebo-controlled intervention trial, 123 patients on chronic dialysis were randomised to a daily oral supplement of either MK-7 360 µg or placebo for 2 years. BMD of the distal radius (1/3, mid, ultradistal and total), femoral neck, lumbar spine (L1-L4) and whole body was assessed by dual-energy X-ray absorptiometry. Serum levels of vitamin K1 and MK-7 and plasma levels of total osteocalcin, dephosphorylated-uncarboxylated matrix Gla protein and protein induced by vitamin K absence II were measured to assess vitamin K status. RESULTS: After 2 years, an accelerated BMD loss of the 1/3 distal radius was found with MK-7 supplementation {mean difference of changes relative to placebo -0.023 g/cm2 [95% confidence interval (CI) -0.039 to -0.008]}, whereas the decrease in lumbar spine BMD seen in the placebo group was prevented [mean difference of changes between groups 0.050 g/cm2 (95% CI 0.015-0.085)]. No significant effects were observed at the remaining skeletal sites. Vitamin K status strongly improved in MK-7-supplemented participants. CONCLUSION: Compared with placebo, an accelerated BMD loss of the 1/3 distal radius was found after 2 years of MK-7 supplementation, whereas a decline in lumbar spine BMD was prevented. As such, MK-7 supplementation might modify BMD site-specifically in patients on dialysis. In aggregate, our findings do not support MK-7 supplementation to preserve bone in patients on dialysis.


Subject(s)
Bone Density , Vitamin K , Humans , Renal Dialysis/adverse effects , Absorptiometry, Photon , Vitamin K 2/pharmacology , Vitamin K 2/therapeutic use , Dietary Supplements , Double-Blind Method
SELECTION OF CITATIONS
SEARCH DETAIL
...