Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Vitamin K2 (MK-7) attenuates LPS-induced acute lung injury via inhibiting inflammation, apoptosis, and ferroptosis

  • Yulian Wang ,

    Contributed equally to this work with: Yulian Wang, Weidong Yang

    Roles Data curation, Investigation, Project administration, Validation

    Affiliation College of Basic Medical Science, Dalian Medical University, Dalian, China

  • Weidong Yang ,

    Contributed equally to this work with: Yulian Wang, Weidong Yang

    Roles Data curation, Investigation, Project administration, Validation

    Affiliation College of Basic Medical Science, Dalian Medical University, Dalian, China

  • Lulu Liu,

    Roles Data curation, Project administration, Validation

    Affiliation College of Basic Medical Science, Dalian Medical University, Dalian, China

  • Lihong Liu,

    Roles Conceptualization, Software, Supervision, Writing – review & editing

    Affiliation College of Basic Medical Science, Dalian Medical University, Dalian, China

  • Jiepeng Chen,

    Roles Conceptualization, Investigation, Software

    Affiliation Sungen Bioscience Co., Ltd., Guangdong, China

  • Lili Duan,

    Roles Conceptualization, Investigation, Software

    Affiliation Sungen Bioscience Co., Ltd., Guangdong, China

  • Yuyuan Li ,

    Roles Conceptualization, Formal analysis, Funding acquisition, Investigation, Writing – original draft

    shuzhuangli@126.com (SL); liyuyuan831221@163.com (YL)

    Affiliation Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China

  • Shuzhuang Li

    Roles Conceptualization, Funding acquisition, Investigation, Supervision, Writing – review & editing

    shuzhuangli@126.com (SL); liyuyuan831221@163.com (YL)

    Affiliation College of Basic Medical Science, Dalian Medical University, Dalian, China

Abstract

Acute lung injury (ALI) is a life-threatening disease that has received considerable critical attention in the field of intensive care. This study aimed to explore the role and mechanism of vitamin K2 (VK2) in ALI. Intraperitoneal injection of 7 mg/kg LPS was used to induce ALI in mice, and VK2 injection was intragastrically administered with the dose of 0.2 and 15 mg/kg. We found that VK2 improved the pulmonary pathology, reduced myeloperoxidase (MPO) activity and levels of TNF-α and IL-6, and boosted the level of IL-10 of mice with ALI. Moreover, VK2 played a significant part in apoptosis by downregulating and upregulating Caspase-3 and Bcl-2 expressions, respectively. As for further mechanism exploration, we found that VK2 inhibited P38 MAPK signaling. Our results also showed that VK2 inhibited ferroptosis, which manifested by reducing malondialdehyde (MDA) and iron levels, increasing glutathione (GSH) level, and upregulated and downregulated glutathione peroxidase 4 (GPX4) and heme oxygenase-1 (HO-1) expressions, respectively. In addition, VK2 also inhibited elastin degradation by reducing levels of uncarboxylated matrix Gla protein (uc-MGP) and desmosine (DES). Overall, VK2 robustly alleviated ALI by inhibiting LPS-induced inflammation, apoptosis, ferroptosis, and elastin degradation, making it a potential novel therapeutic candidate for ALI.

Introduction

Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), are severe respiratory system disease [1]. ALI is characterized by disruption of the lung capillary endothelium and alveolar-capillary membrane barrier, overactivated inflammatory response, and pulmonary edema [2]. Lipopolysaccharide (LPS) is the principal component of the outer membranes of gram-negative bacteria, which can trigger strong inflammatory responses and cause severe lung injury. The molecular process of ALI is believed to involve complex interactions between inflammation, oxidative stress, apoptosis, and ferroptosis [3, 4]. Cytokine storms are a result of the markedly elevated proinflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), which have been proved to be major factors promoting the development of ALI [5]. These cytokines act on leukocytes to activate positive feedback of proinflammatory signals [6, 7]. At the same time, a large number of leukocytes’ influx into the lungs also induces the production of reactive oxygen species (ROS) in hepatocytes and endoplasmic reticulum stress, leading to hepatocyte apoptosis and ferroptosis [8, 9]. Therefore, inhibiting inflammation, oxidative stress, apoptosis, and ferroptosis is crucial for alleviating LPS-induced ALI.

However, currently, there are no efficient drugs or therapies for LPS-induced ALI in clinical practice. A promising strategy to prevent and treat ALI is to suppress the excessive production of inflammatory cytokines through anti-inflammatory drugs, including corticosteroids, such as methylprednisolone [10], dexamethasone (DEX) [11], prednisolone [12]. However, the clinical application of corticosteroids is usually accompanied by various severe side effects, such as hyperglycemia, hypertension, hypokalemia, dyslipidemia, osteoporosis, myopathy and immunosuppression [1315]. Therefore, it is necessary to find new drugs with better efficacy and safety for the treatment of ALI patients.

Vitamin K (VK) is a family of lipid-soluble molecules that exists mainly two forms: vitamin K1 (phylloquinone) and vitamin K2 (menaquinone). VK2, essential for human metabolism and health is predominantly present in cheese and fermented soybeans (natto) [16]. In recent years, VK2 has become a research hotspot for scientists due to its multiple pharmacological activities, including anti-inflammatory, anti-oxidant, anti-apoptosis, and anti-ferroptosis [1719]. However, whether VK2 has a beneficial effect on LPS-induced ALI has not been investigated. Thereby, this study was conducted to evaluate if VK2 administration could offer protection against ALI/ARDS.

Materials and methods

Reagents

VK2 was provided by Sungen Bioscience Co., Ltd. (Guangdong, China). LPS (from Escherichia coli (055:B5)) and DEX were purchased from Sigma-Aldrich (St. Louis, MO, United States). Mouse myeloperoxidase (MPO) determination kit was purchased from the Jiancheng Bioengineering Institute of Nanjing (Nanjing, Jiangsu Province, China). ELISA kits for mouse TNF-α, IL-6 and IL-10 were purchased from Jiangsu Meimian Industrial Co., Ltd. (Jiangsu, China) and for uncarboxylated MGP (uc-MGP) and desmosine (DES) were from Shanghai Lengton Bioscience Co., Ltd. (shanghai, china). The malondialdehyde (MDA), glutathione (GSH), and Iron determination kits were purchased from Solarbio (Beijing, China). The antibodies against GAPDH, iNOS, and Caspase-3 were obtained from Abcame (Cambridge, MA). The antibodies against TLR4 and IL-6 were purchased from Wanleibio (Liaoning, China). The antibodies against P38, p-P38, and Bcl-2 were from Proteintech (Wuhan, China). The antibodies against glutathione peroxidase 4 (GPX4) and heme oxygenase-1 (HO-1) were obtained from Bioworld (Minnesota, USA). Other chemicals were conformed from reagent grade.

Animal experiments

Specific-pathogen-free (SPF) male C57BL/6 mice (8–10 weeks old, 20–24 g body weight) were purchased from Liaoning Changsheng Biotechnology Co., Ltd. (Liaoning, China). The mice were kept in cages with food and water ad libitum during the whole period. The experiment was conducted with the approval of the Dalian Medical University Animal Care and Use Committee (AEE20051).

LPS (7 mg/kg) was dissolved in saline and administrated by intraperitoneal injection to mice and the acute lung injury model was established by LPS administration for 3 days. The mice were randomly divided into six groups (7 mice per group): control group, LPS group, negative control group (LPS + Oil [vehicle]), positive control group (LPS + DEX), and the VK2 pretreatment groups at two different concentrations. VK2 was dissolved in soybean oil at different concentrations (0.2 and 15 mg/kg). Before LPS treatment, the VK2 pretreatment groups were pre-administered intragastrically for 6 days, while the control and model groups were given the same volume of solvent solution. In the positive control group, mice were Caspase-3. The treatment of mice was shown in Fig 1A. After deeply anesthetized with tribromoethanol, the lung and serum were collected from each mouse for further experiments.

thumbnail
Fig 1. VK2 attenuated LPS-induced acute lung injury.

(A) Mice were pre-administered intragastrically solvent or VK2 (0.2 and 15 mg/kg respectively) and subsequent intraperitoneal injection of LPS (7 mg/kg). (B) Histological analysis of lung tissue sections by HE staining (original multiples: 200 ×, scale = 75 μm). (C) Lung tissue injury was assessed by histological scores in all groups. (D) Determination of the myeloperoxidase (MPO) activity in lung homogenates. Values represent means ± SEM, *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001.

https://doi.org/10.1371/journal.pone.0294763.g001

Histological evaluation of lung

Lung tissue samples were fixed in 4% (w/v) paraformaldehyde and then dehydrated, embedded in paraffin and stained with hematoxylin / eosin (HE). The severity of lung damage was semi-quantitatively scored as previously described [20].

Determination of myeloperoxidase (MPO) activity and the content of ferroptosis biomarkers in lung tissue

Lung homogenate (10%, w/v), obtained by homogenizing the lung tissue and normal saline, was used for the measurements of MPO activities, and MDA, GSH and Iron content using test kits in accordance with the manufacturer’s instructions.

Measurement of inflammatory cytokines in serum

The serum was collected and the levels of pro-inflammatory cytokines TNF-α and IL-6, and anti-inflammatory cytokines IL-10 in the serum were measured by ELISA kits according to the manufacturer’s recommendations.

Assessment of the degradation level of elastic fibers

To evaluate the level of elastic fiber degradation, the concentrations of uc-MGP in serum and DES in lung tissue were quantified using ELISA. All measurements were carried out following manufacturer’s instructions.

Western blotting analysis

The lung tissues were lysed with RIPA Lysis Buffer to extract the proteins that were quantified by the BCA assay. Denatured proteins were separated using sodium dodecyl sulfate-polyacrylamide gelelectrophoresis (SDS-PAGE) and then transferred onto polyvinulidene difluoride (PVDF) membranes. After blocking with 5% fat-free milk, the membranes were incubated with the primary antibodies of TLR4, P38, p-P38, IL-6, iNOS, Caspase-3, Bcl-2, GPX4, HO-1, and GAPDH at 4°C overnight. Next, the membranes were incubated with the secondary antibodies (Santa Cruz, CA; 1:2000) for 1h at room temperature. After the final washes, the protein bands were scanned using the gel imaging system (BIO-RAD, United States).

Statistical analysis

Data were expressed as the mean ± standard error of the mean (SEM). Statistical significance of differences was assessed by one-way ANOVA followed by Kruskal-Wallis rank sum test for multiple groups and two-tailed, unpaired Student’s t test for two groups with the assistance of GraphPad Prism Program (Version 8.2.1). Significance was set at *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Results

VK2 ameliorates LPS-induced histopathological changes and decreases MPO activity in lung

The treatments of mice were shown in Fig 1A. We first observed the morphological changes of lung tissues by comparing the HE-stained pathological sections (Fig 1B and 1C). LPS-induced mice showed alveoli structure destruction, thickening of alveolar interval, and inflammatory cell infiltration compared with their control counterparts. After VK2 pretreatment, lung injury induced by LPS gradually resolved and followed a dose-dependent trend. Then, the marker of neutrophils infiltration in tissues, MPO, has been detected in this study. The results showed that MPO activity in lung tissues was significantly increased by LPS (p = 0.0052). Pretreatment with VK2, especially with high dose, significantly inhibited LPS-induced MPO activity (p = 0.0175) (Fig 1D). Taken together, these results suggest that VK2 protected against LPS-induced ALI in mice.

VK2 improved LPS-induced inflammation via P38 MAPK signaling

The results of the content of inflammatory cytokines (TNF-α and IL-6) and anti-inflammatory cytokine (IL-10) in the serum were shown in Fig 2A–2C. Compared with control group, contents of TNF-α (p = 0.0066, Fig 2A) and IL-6 (p = 0.0014, Fig 2B) were drastically increased, but that of IL-10 (p = 0.0233, Fig 2C) was decreased in LPS-induced model group. Similar to the positive drug DEX, VK2 administration, especially high dose, displayed the strongest inhibitory effect on reducing TNF-α (p = 0.0488) and IL-6 (p = 0.0116) and increasing IL-10 (p = 0.004). These results indicated that VK2 could alleviate LPS-induced lung inflammation by inhibiting the excessive production of pro-inflammatory cytokines and promoting the production of anti-inflammatory factors.

thumbnail
Fig 2. Effects of VK2 on LPS-induced inflammation.

(A) TNF-α, (B) IL-6 and (C) IL-10 were measured with ELISA in mouse serum. (D) The protein expression levels of TLR4, p-P38 MAPK, and P38 MAPK were evaluated by western blotting. (E, F) Quantitative analysis of TLR4 and the ratio of p-P38/P38 normalized with GAPDH were performed using Image J software. (G) The protein expression levels of iNOS and IL-6 were evaluated by western blotting. (H, I) Quantitative analysis of iNOS and IL-6 normalized with GAPDH. Values represent means ± SEM, *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001.

https://doi.org/10.1371/journal.pone.0294763.g002

MAPK is a common signaling pathway in the LPS-induced inflammatory response [21]. The effect of VK2 on MAPK signaling was assessed by measuring the phosphorylation of p38 and the expression of upstream protein (TLR4). Western blot assays showed that compared with the control group, the expressions of TLR4 (p = 0.0442, Fig 2D and 2E) and phosphorylated p38 (p = 0.0009, Fig 2D and 2F) were significantly increased in the LPS group. However, VK2 administration, especially high dose, could obviously inhibited LPS-induced increase of TLR4 (p = 0.0108) and phosphorylated p38 (p = 0.0132). iNOS and IL-6 are important pro-inflammatory proteins in the cascading inflammatory response, which can be regulated by MAPK. As shown in Fig 2G–2I, the expressions of iNOS and IL-6 were increased by LPS compared with control groups, but effectively reduced by VK2 with respect to the LPS model groups (p < 0.05). The results suggest that VK2 alleviated LPS-induced inflammation, and the activation and regulation of P38 MAPK pathway was involved in these anti-inflammatory processes.

VK2 inhibits apoptosis in LPS-induced ALI

In this study, expressions of apoptosis related lung proteins were determined via Western blotting (Fig 3). In comparison with the control group, LPS treatment dramatically increased the pro-apoptotic protein expression of Caspase-3 (p = 0.0077, Fig 3A and 3B), while significantly downregulated the expression of anti-apoptotic protein Bcl-2 (p = 0.0041, Fig 3A and 3C). However, VK2 pretreatment significantly reduced Caspase-3 and increased Bcl-2 expression levels in ALI mice (p < 0.05), suggesting VK2 pretreatment might relieve LPS-induced pulmonary cell apoptosis in ALI mice.

thumbnail
Fig 3. VK2 inhibits apoptosis in LPS-induced ALI.

(A) The protein expression levels of Caspase-3 and Bcl-2 were evaluated by western blotting. (B, C) Quantitative analysis of Caspase-3 and Bcl-2 normalized with GAPDH were performed using Image J software. Values represent means ± SEM, *p < 0.05 and **p < 0.01.

https://doi.org/10.1371/journal.pone.0294763.g003

VK2 mitigates ferroptosis in LPS-induced ALI

Ferroptosis results from the iron-dependent accumulation of lipid peroxides. To explore the protective role of VK2 pretreatment against LPS-induced ferroptosis, the level of reductive glutathione (GSH), lipid peroxidation products malondialdehyde (MDA) and tissue iron levels were measured (Fig 4). The GSH level was significantly reduced after LPS challenge (p = 0.0009), while VK2 pretreatment reversed this situation and increased GSH extent in lung tissue (p = 0.0462, Fig 4A). In contrast, the MDA level (p = 0.0015, Fig 4B) and tissue iron (p = 0.0409, Fig 4C) were significantly decreased after VK2 supplementation in high dose compared with LPS group. Lipid peroxidation is the kernel of ferroptosis, and glutathione peroxidase 4 (GPX4) is an antioxidant enzyme reported as a critical inhibitor of ferroptosis. In LPS-induced mice, GPX4 level decreased drastically (p = 0.0002) and could be salvaged by VK2 pretreatment in high dose (p = 0.0052) but not positive drug DEX (Fig 4D and 4E). Moreover, we found that LPS induction led to a mild elevation of heme oxygenase-1 (HO-1, a stress response protein), while VK2 pretreatment in high dose restored the situation to normal (Fig 4D and 4F). Collectively, these results indicated that VK2 was capable of repressing ferroptosis in LPS-induced ALI while DEX was not.

thumbnail
Fig 4. Role of VK2 in ferroptosis during LPS-mediated injury.

(A) GSH, (B) MDA, and (C) Total iron levels in lung tissues. (D) GPX4 and HO-1 protein expression were measured by western blotting. (E, F) Quantitative analysis of GPX4 and HO-1 normalized with GAPDH were performed using Image J software. Values represent means ± SEM, *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001.

https://doi.org/10.1371/journal.pone.0294763.g004

VK2 inhibits LPS-induced lung elastin degradation

Elastin degradation is implicated in the pathology of ALI [22] and is partially regulated by Matrix Gla Protein (MGP), via a vitamin K-dependent pathway [23]. To evaluate the effect of VK2 pretreatment on LPS-induced lung elastin degradation, the level of uncarboxylated MGP (uc-MGP) and DES (an elastin-specific degradation product) were measured (Fig 5). It was found that LPS stimulation significantly increased uc-MGP level (p = 0.0066, Fig 5A), while VK2 intervention, especially in high dose, reversed this situation and reduced serum uc-MGP level (p = 0.0053, Fig 5A). However, it is interested that the positive drug DEX could not decrease the serum level of uc-MGP after LPS induction. Moreover, we found that DES level increased drastically in LPS-induced mice (p = 0.0151) and could be decreased by VK2 pretreatment in high dose (p = 0.0044) an also positive drug DEX (p = 0.0026, Fig 5B). The above results indicated that VK2 may alleviate lung injury by inhibiting LPS-induced the pulmonary elastin degradation through carboxlation of MGP.

thumbnail
Fig 5. VK2 alleviates LPS-induced lung elastin degradation.

(A) uc-MGP levels in serum. (B) DES levels in lung tissues. Values represent means ± SEM, *p < 0.05 and **p < 0.01.

https://doi.org/10.1371/journal.pone.0294763.g005

Discussion

A recent study reported serum VK2 (MK7) in patients with Coronavirus Disease 2019 (COVID-19) was very low compared with non-COVID-19 pneumonia and healthy controls [24]. So does VK2 have a role in prevention and treatment of LPS-induced ALI? Based on our experiment, VK2 could improve LPS-induced ALI. We determined the VK2 intervention dose for this experiment based on previous studies [18, 19, 25] and on the range of recommended VK2 intakes in each country [26, 27].

The inflammatory response is an important defense mechanism induced in the host in response to injury, infection or stimulation [28]. Under LPS induction, the levels of proinflammatory cytokines, including interferons (IFNs), tumor necrosis factors (TNFs), interleukins (ILs), and chemokines [29] were significantly higher than the control group, and the infiltration of inflammatory cells in lung tissue was upregulated as well, which was consistent with our results. However, VK2 pretreatment significantly improved these phenomena and gradually returned to normal levels. VK2 has been investigated as a potential anti-inflammatory and protective drug in several inflammatory diseases including type 2 diabetes mellitus (T2DM) [16], inflammatory bowel disease (IBD) [25], atherosclerosis [30], rheumatoid arthritis (RA) [31], and neurodegenerative diseases, especially Parkinson’s and Alzheimer’s disease [32, 33].

Mitogen-activated protein kinases (MAPKs) are a class of serine/threonine protein kinases in cells, which mediate cellular responses to extracellular stimulations, including growth factors, cytokines, hormones, neurotransmitters, cellular stress, and cell adherence [34]. Activation of the MAPK signaling pathway is also involved in the occurrence of inflammation in LPS-induced ALI [35]. Further researches suggested that the P38 MAPK pathway played a crucial role in LPS-stimulated inflammatory response and macrophage activation [36]. In this study, we found that in the LPS model group, VK2 significantly reduced the proportion of p-P38/P38, thus reducing the expression of inflammatory cytokines IL-6 and iNOS. Therefore, these data suggested that VK2 might alleviate inflammation by inhibiting the activation of the P38 MAPK pathway.

Apoptosis is the process of programmed cell death that occurs under normal physiological or pathological conditions [37]. Excessive apoptosis plays an important role in the development of ALI [38, 39]. LPS can promote increased cell apoptosis by triggering inflammatory responses [40]. Bcl-2 controls the cell proliferation or apoptosis through inhibiting cell apoptosis and the activation of downstream Caspase-3 proteases [41]. The results of this study indicated that VK2 exerted its protective effect on ALI through the upregulation of Bcl-2 and downregulation of Caspase-3 in LPS-induced ALI. Indeed, VK2 administration has been reported to exert anti-apoptotic and anti-inflammatory effects [18, 42]. Furthermore, P38 MAPK pathway has been evidenced to mediate apoptosis [43]. Studies have shown that phosphorylated P38 MAPK can activate caspase-3 and promote apoptosis by downregulating Bcl-2 [4446]. Thereby, VK2 reduced lung cell apoptosis by inhibiting LPS-induced activation of P38 MAPK.

Recent studies have found that ferroptosis also has a crucial function in the progression of ALI, and its inhibition is effective in alleviating ALI [4750]. Ferroptosis is a unique iron-dependent lipid-peroxidation, which is different from traditional necrosis, apoptosis, autophagy, or other forms of cell death [51]. It is mainly characterized by the accumulation of free iron, drastic lipid peroxidation, and ROS production [52]. According to the occurrence process of ferroptosis, various key factors including GSH, MDA, iron, and GPX4 are often used to comprehensively evaluate lipid peroxidation and ferroptosis [53]. In this study, we also found significant ferroptosis in LPS-induced ALI, characterized by increased MDA and iron levels, and reduced GSH and GPX4 levels. After VK2 administration, the accumulations of MDA and iron were significantly reduced while the expression of GPX4 was obviously increased, suggesting that VK2 could inhibit the LPS-induced ferroptosis. Heme oxygenase 1 protein (HO-1), an important stress response protein highly expressed in lungs, is encoded by HMOX1 gene and decomposes heme into iron, carbon monoxide (CO), biliverdin, and bilirubin, which is essential in the balance of intracellular iron and ROS [54]. HO-1-deficient mice exhibit high oxidative damage, tissue injury, and chronic inflammation, as well as hepatic and renal iron accumulation [55]. However, HO-1 has not only beneficial role to protect against oxidative stress but also detrimental role to promote ROS generation by releasing free ferrous iron and subsequent expression of ferritin [5659]. Moreover, HO-1-induced ferroptosis may be related to the reduction of free iron-binding ability of ferritin induced by oxidative stress, such as iron accumulation and lipid peroxidation [60]. In this study, we found that administration of VK2 effectively inhibited LPS-induced elevation of HO-1. According to these data, it can be inferred that the protective effect of VK2 on LPS-induced lung injury might be related to ferroptosis.

Elastic fibers are important components of the extracellular matrix of dynamic tissue, conferring resilience of the lungs and arteries, subsequently promoting breathing and circulation [61]. DES is formed during the crosslinking process of nourishing layer-elastin polymers, which is a product of elastin degradation, releasing into the blood after elastic fibers degradation [62]. MGP has an important role in elastic fibers degradation and requires VK for its activation [63]. Uncarboxylated MGP (uc-MGP) is a biomarker of the VK status in the body, and high level of uc-MGP reflect the low vitamin K status [64]. In current study, we found that uc-MGP concentration was significantly increased by LPS, while after VK2 pretreatment, it was significantly decreased, which was consistent with the results of DES. These results suggested that VK2 supplementation could carboxylate uc-MGP into MGP, effectively preventing the degradation of elastic fibers and protecting the lungs.

In summary, the findings of the current study suggest that VK2 effectively protects against ALI caused by LPS through suppression of P38 MAPK signaling pathway and ferroptosis as depicted in Fig 6. Our work suggested that VK2 supplementation might be a cheap and effective intervention or prevention measures against serious courses of ALI. Nevertheless, despite widespread adoption, the LPS-induced ALI model may not entirely encompass the complexity and heterogeneity of human ALI. It is essential to bolster animal studies with clinical research and validation. In the future, we will carry out relevant in-depth work to solve this problem.

thumbnail
Fig 6. Schematic representation of VK2 action on LPS-induced ALI.

https://doi.org/10.1371/journal.pone.0294763.g006

References

  1. 1. Lv K, Li M, Sun C, Miao Y, Zhang Y, Liu Y, et al. Jingfang Granule alleviates bleomycin-induced acute lung injury via CD200-CD200R immunoregulatory pathway. J Ethnopharmacol. 2023 Apr 1;311:116423. pmid:37011735
  2. 2. Fan E, Brodie D, Slutsky AS. Acute Respiratory Distress Syndrome: Advances in Diagnosis and Treatment. JAMA. 2018 Feb 20;319(7):698–710. pmid:29466596
  3. 3. Lei J, Wei Y, Song P, Li Y, Zhang T, Feng Q, et al. Cordycepin inhibits LPS-induced acute lung injury by inhibiting inflammation and oxidative stress. Eur J Pharmacol. 2018 Jan 5;818:110–114. pmid:29054740
  4. 4. Xie W, Lu Q, Wang K, Lu J, Gu X, Zhu D, et al. miR-34b-5p inhibition attenuates lung inflammation and apoptosis in an LPS-induced acute lung injury mouse model by targeting progranulin. J Cell Physiol. 2018 Sep;233(9):6615–6631. pmid:29150939
  5. 5. Zhang F, Guo F, Zhang Y, Xu H, Liu Y, Lin L, et al. Huashibaidu formula attenuates sepsis-induced acute lung injury via suppressing cytokine storm: Implications for treatment of COVID-19. Phytomedicine. 2023 Jan;109:154549. pmid:36610129
  6. 6. Wang H, Sun X, Lu Q, Zemskov EA, Yegambaram M, Wu X, et al. The mitochondrial redistribution of eNOS is involved in lipopolysaccharide induced inflammasome activation during acute lung injury. Redox Biol. 2021 May;41:101878. pmid:33578126
  7. 7. Wang T, Yegambaram M, Gross C, Sun X, Lu Q, Wang H, et al. RAC1 nitration at Y32 IS involved in the endothelial barrier disruption associated with lipopolysaccharide-mediated acute lung injury. Redox Biol. 2021 Jan;38:101794. pmid:33248422
  8. 8. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell. 2017 Oct 5;171(2):273–285. pmid:28985560
  9. 9. Zhang Q, Zeng M, Zhang B, Ren Y, Li S, Wang R, et al. Salvianolactone acid A isolated from Salvia miltiorrhiza ameliorates lipopolysaccharide-induced acute lung injury in mice by regulating PPAR-γ. Phytomedicine. 2022 Oct;105:154386.
  10. 10. Fu X, Ye X, An LN, Jiang H, Huang WB, Huang Y, et al. Efficacy and Safety of Methylprednisolone for Lung Surgery: a Systematic Review and Meta-analysis of Randomized Controlled Trials. Pain Ther. 2023 Feb;12(1):165–186. pmid:36260278
  11. 11. Terzi F, Demirci B, Çınar İ, Alhilal M, Erol HS. Effects of tocilizumab and dexamethasone on the downregulation of proinflammatory cytokines and upregulation of antioxidants in the lungs in oleic acid-induced ARDS. Respir Res. 2022 Sep 17;23(1):249. pmid:36115998
  12. 12. Gao J, Su Y, Wang Z. Remote Co-loading of amphipathic acid drugs in neutrophil nanovesicles infilled with cholesterol mitigates lung bacterial infection and inflammation. Biomaterials. 2023 May;296:122071. pmid:36878092
  13. 13. Kulkarni S, Durham H, Glover L, Ather O, Phillips V, Nemes S, et al. Metabolic adverse events associated with systemic corticosteroid therapy-a systematic review and meta-analysis. BMJ Open. 2022 Dec 22;12(12):e061476. pmid:36549729
  14. 14. Saha A, Balakrishnan S, Trivedi N, Abraham GM. Hypertension and Severe Hypokalemia Associated With Oral Ingestion of Topical Hydrocortisone Cream. AACE Clin Case Rep. 2022 Oct 21;9(1):2–4. pmid:36654996
  15. 15. Yu SH, Drucker AM, Lebwohl M, Silverberg JI. A systematic review of the safety and efficacy of systemic corticosteroids in atopic dermatitis. J Am Acad Dermatol. 2018 Apr;78(4):733–740.e11. pmid:29032119
  16. 16. Li Y, Chen JP, Duan L, Li S. Effect of vitamin K2 on type 2 diabetes mellitus: A review. Diabetes Res Clin Pract. 2018 Feb;136:39–51. pmid:29196151
  17. 17. Kołakowski A, Kurzyna PF, Bzdęga W, Żywno H, Harasim-Symbor E, Chabowski A, et al. Influence of vitamin K2 on lipid precursors of inflammation and fatty acids pathway activities in HepG2 cells. Eur J Cell Biol. 2021 Sep-Nov;100(7–8):151188. pmid:34837768
  18. 18. El-Sherbiny M, Atef H, Helal GM, Al-Serwi RH, Elkattawy HA, Shaker GA, et al. Vitamin K2 (MK-7) Intercepts Keap-1/Nrf-2/HO-1 Pathway and Hinders Inflammatory/Apoptotic Signaling and Liver Aging in Naturally Aging Rat. Antioxidants (Basel). 2022 Oct 30;11(11):2150. pmid:36358523
  19. 19. Jin C, Tan K, Yao Z, Lin BH, Zhang DP, Chen WK, et al. A Novel Anti-Osteoporosis Mechanism of VK2: Interfering with Ferroptosis via AMPK/SIRT1 Pathway in Type 2 Diabetic Osteoporosis. J Agric Food Chem. 2023 Feb 15;71(6):2745–2761. pmid:36719855
  20. 20. Mrozek JD, Smith KM, Bing DR, Meyers PA, Simonton SC, Connett JE, et al. Exogenous surfactant and partial liquid ventilation: physiologic and pathologic effects. Am J Respir Crit Care Med. 1997 Oct;156(4 Pt 1):1058–65. pmid:9351603
  21. 21. Park MH, Yeom YJ, Ganbat D, Kim MK, Kim SB, Lee YJ, et al. Fermentation of Abelmoschus manihot Extract with Halophilic Bacillus licheniformis CP6 Results in Enhanced Anti-Inflammatory Activities. Nutrients. 2023 Jan 7;15(2):309. pmid:36678181
  22. 22. Ayala P, Vivar R, Montalva R, Olmos P, Meneses M, Borzone GR. Elastin degradation products in acute lung injury induced by gastric contents aspiration. Respir Res. 2018 Aug 31;19(1):165. pmid:30170599
  23. 23. Piscaer I, van den Ouweland JMW, Vermeersch K, Reynaert NL, Franssen FME, Keene S, et al. Low Vitamin K Status Is Associated with Increased Elastin Degradation in Chronic Obstructive Pulmonary Disease. J Clin Med. 2019 Jul 27;8(8):1116. pmid:31357639
  24. 24. Mangge H, Prueller F, Dawczynski C, Curcic P, Sloup Z, Holter M, et al. Dramatic Decrease of Vitamin K2 Subtype Menaquinone-7 in COVID-19 Patients. Antioxidants (Basel). 2022 Jun 24;11(7):1235. pmid:35883726
  25. 25. Hu S, Ma Y, Xiong K, Wang Y, Liu Y, Sun Y, et al. Ameliorating Effects of Vitamin K2 on Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice. Int J Mol Sci. 2023 Feb 3;24(3):2986. pmid:36769323
  26. 26. Simes DC, Viegas CSB, Araújo N, Marreiros C. Vitamin K as a Diet Supplement with Impact in Human Health: Current Evidence in Age-Related Diseases. Nutrients. 2020 Jan 3;12(1):138. pmid:31947821
  27. 27. Marles RJ, Roe AL, Oketch-Rabah HA. US Pharmacopeial Convention safety evaluation of menaquinone-7, a form of vitamin K. Nutr Rev. 2017 Jul 1;75(7):553–578. pmid:28838081
  28. 28. Nathan C. Points of control in inflammation. Nature. 2002 Dec 19–26;420(6917):846–52. pmid:12490957
  29. 29. Liu Q, Zhou YH, Yang ZQ. The cytokine storm of severe influenza and development of immunomodulatory therapy. Cell Mol Immunol. 2016 Jan;13(1):3–10. pmid:26189369
  30. 30. Wang Z, Wang Z, Zhu J, Long X, Yan J. Vitamin K2 can suppress the expression of Toll-like receptor 2 (TLR2) and TLR4, and inhibit calcification of aortic intima in ApoE-/- mice as well as smooth muscle cells. Vascular. 2018 Feb;26(1):18–26. pmid:28587577
  31. 31. Abdel-Rahman MS, Alkady EA, Ahmed S. Menaquinone-7 as a novel pharmacological therapy in the treatment of rheumatoid arthritis: A clinical study. Eur J Pharmacol. 2015 Aug 15;761:273–8. pmid:26073022
  32. 32. Yu YX, Yu XD, Cheng QZ, Tang L, Shen MQ. The association of serum vitamin K2 levels with Parkinson’s disease: from basic case-control study to big data mining analysis. Aging (Albany NY). 2020 Aug 29;12(16):16410–16419. pmid:32862152
  33. 33. Popescu A, German M. Vitamin K2 Holds Promise for Alzheimer’s Prevention and Treatment. Nutrients. 2021 Jun 27;13(7):2206. pmid:34199021
  34. 34. Widmann C, Gibson S, Jarpe MB, Johnson GL. Mitogen-activated protein kinase: conservation of a three-kinase module from yeast to human. Physiol Rev. 1999 Jan;79(1):143–80. pmid:9922370
  35. 35. Shen B, Zhang H, Zhu Z, Ling Z, Zeng F, Wang Y, et al. Baicalin Relieves LPS-Induced Lung Inflammation via the NF-κB and MAPK Pathways. Molecules. 2023 Feb 16;28(4):1873. pmid:36838858
  36. 36. Choi YH, Kim GY, Lee HH. Anti-inflammatory effects of cordycepin in lipopolysaccharide-stimulated RAW 264.7 macrophages through Toll-like receptor 4-mediated suppression of mitogen-activated protein kinases and NF-κB signaling pathways. Drug Des Devel Ther. 2014 Oct 16;8:1941–53. pmid:25342887
  37. 37. Kuypers FA. Hyperinflammation, apoptosis, and organ damage. Exp Biol Med (Maywood). 2022 Jul;247(13):1112–1123. pmid:35475359
  38. 38. Yang J, Chen X. SIRT6 attenuates LPS-induced inflammation and apoptosis of lung epithelial cells in acute lung injury through ACE2/STAT3/PIM1 signaling. Immun Inflamm Dis. 2023 Mar;11(3):e809. pmid:36988243
  39. 39. Zhou Y, Jin T, Gao M, Luo Z, Mutahir S, Shi C, et al. Aqueous extract of Platycodon grandiflorus attenuates lipopolysaccharide-induced apoptosis and inflammatory cell infiltration in mouse lungs by inhibiting PI3K/Akt signaling. Chin Med. 2023 Apr 4;18(1):36. pmid:37016413
  40. 40. Tao W, Su Q, Wang H, Guo S, Chen Y, Duan J, et al. Platycodin D attenuates acute lung injury by suppressing apoptosis and inflammation in vivo and in vitro. Int Immunopharmacol. 2015 Jul;27(1):138–47. pmid:25981110
  41. 41. Gao Z, Wang D, Yang J, Li M, Ling C, Lv D, et al. Iron deficiency in hepatocellular carcinoma cells induced sorafenib resistance by upregulating HIF-1α to inhibit apoptosis. Biomed Pharmacother. 2023 Apr 21;163:114750. pmid:37087978
  42. 42. Alisi L, Cao R, De Angelis C, Cafolla A, Caramia F, Cartocci G, et al. The Relationships Between Vitamin K and Cognition: A Review of Current Evidence. Front Neurol. 2019 Mar 19;10:239. pmid:30949117
  43. 43. Ge F, Gao X, Zhou X, Li J, Ma X, Huang M, et al. The alkaloids of Corydalis hendersonii Hemsl. contribute to the cardioprotective effect against ischemic injury in mice by attenuating cardiomyocyte apoptosis via p38 MAPK signaling pathway. Chin Med. 2023 Mar 17;18(1):29. pmid:36932448
  44. 44. De Chiara G, Marcocci ME, Torcia M, Lucibello M, Rosini P, Bonini P, et al. Bcl-2 Phosphorylation by p38 MAPK: identification of target sites and biologic consequences. J Biol Chem. 2006 Jul 28;281(30):21353–21361. pmid:16714293
  45. 45. Zanotto-Filho A, Braganhol E, Battastini AM, Moreira JC. Proteasome inhibitor MG132 induces selective apoptosis in glioblastoma cells through inhibition of PI3K/Akt and NFkappaB pathways, mitochondrial dysfunction, and activation of p38-JNK1/2 signaling. Invest New Drugs. 2012 Dec;30(6):2252–62. pmid:22367315
  46. 46. Huang YK, Chang KC, Li CY, Lieu AS, Lin CL. AKR1B1 Represses Glioma Cell Proliferation through p38 MAPK-Mediated Bcl-2/BAX/Caspase-3 Apoptotic Signaling Pathways. Curr Issues Mol Biol. 2023 Apr 13;45(4):3391–3405. pmid:37185746
  47. 47. Jin H, Zhao K, Li J, Xu Z, Liao S, Sun S. Matrine alleviates oxidative stress and ferroptosis in severe acute pancreatitis-induced acute lung injury by activating the UCP2/SIRT3/PGC1α pathway. Int Immunopharmacol. 2023 Apr;117:109981. pmid:37012871
  48. 48. Lai K, Song C, Gao M, Deng Y, Lu Z, Li N, et al. Uridine Alleviates Sepsis-Induced Acute Lung Injury by Inhibiting Ferroptosis of Macrophage. Int J Mol Sci. 2023 Mar 7;24(6):5093. pmid:36982166
  49. 49. Lou L, Wang M, He J, Yang S, Meng F, Wang S, et al. Urolithin A (UA) attenuates ferroptosis in LPS-induced acute lung injury in mice by upregulating Keap1-Nrf2/HO-1 signaling pathway. Front Pharmacol. 2023 Mar 9;14:1067402. pmid:36969874
  50. 50. Liu P, Feng Y, Li H, Chen X, Wang G, Xu S, Li Y, Zhao L. Ferrostatin-1 alleviates lipopolysaccharide-induced acute lung injury via inhibiting ferroptosis. Cell Mol Biol Lett. 2020 Feb 27;25:10. pmid:32161620
  51. 51. Hirschhorn T, Stockwell BR. The development of the concept of ferroptosis. Free Radic Biol Med. 2019 Mar;133:130–143. pmid:30268886
  52. 52. Doll S, Conrad M. Iron and ferroptosis: A still ill-defined liaison. IUBMB Life. 2017 Jun;69(6):423–434. pmid:28276141
  53. 53. Kou L, Sun R, Xiao S, Zheng Y, Chen Z, Cai A, et al. Ambidextrous Approach To Disrupt Redox Balance in Tumor Cells with Increased ROS Production and Decreased GSH Synthesis for Cancer Therapy. ACS Appl Mater Interfaces. 2019 Jul 31;11(30):26722–26730. pmid:31276364
  54. 54. Araujo JA, Zhang M, Yin F. Heme oxygenase-1, oxidation, inflammation, and atherosclerosis. Front Pharmacol. 2012 Jul 19;3:119. pmid:22833723
  55. 55. Poss KD, Tonegawa S. Heme oxygenase 1 is required for mammalian iron reutilization. Proc Natl Acad Sci U S A. 1997 Sep 30;94(20):10919–24. pmid:9380735
  56. 56. Chillappagari S, Venkatesan S, Garapati V, Mahavadi P, Munder A, Seubert A, et al. Impaired TLR4 and HIF expression in cystic fibrosis bronchial epithelial cells downregulates hemeoxygenase-1 and alters iron homeostasis in vitro. Am J Physiol Lung Cell Mol Physiol. 2014 Nov 15;307(10):L791–9. pmid:25239913
  57. 57. Mao X, Wang T, Liu Y, Irwin MG, Ou JS, Liao XL, et al. N-acetylcysteine and allopurinol confer synergy in attenuating myocardial ischemia injury via restoring HIF-1α/HO-1 signaling in diabetic rats. PLoS One. 2013 Jul 18;8(7):e68949. pmid:23874823
  58. 58. Fang X, Wang H, Han D, Xie E, Yang X, Wei J, et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci U S A. 2019 Feb 12;116(7):2672–2680. pmid:30692261
  59. 59. Yu X, Song N, Guo X, Jiang H, Zhang H, Xie J. Differences in vulnerability of neurons and astrocytes to heme oxygenase-1 modulation: Implications for mitochondrial ferritin. Sci Rep. 2016 Apr 21;6:24200. pmid:27097841
  60. 60. Kwon MY, Park E, Lee SJ, Chung SW. Heme oxygenase-1 accelerates erastin-induced ferroptotic cell death. Oncotarget. 2015 Sep 15;6(27):24393–403. pmid:26405158
  61. 61. Mithieux SM, Weiss AS. Elastin. Adv Protein Chem. 2005;70:437–61. pmid:15837523
  62. 62. Huang JT, Bolton CE, Miller BE, Tal-Singer R, Rabinovich RA, Palmer CN, et al. Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) Investigators. Age-dependent elastin degradation is enhanced in chronic obstructive pulmonary disease. Eur Respir J. 2016 Oct;48(4):1215–1218.
  63. 63. Janssen R, Vermeer C. Vitamin K deficit and elastolysis theory in pulmonary elasto-degenerative diseases. Med Hypotheses. 2017 Oct;108:38–41. pmid:29055397
  64. 64. Oikonomaki T, Papasotiriou M, Ntrinias T, Kalogeropoulou C, Zabakis P, Kalavrizioti D, et al. The effect of vitamin K2 supplementation on vascular calcification in haemodialysis patients: a 1-year follow-up randomized trial. Int Urol Nephrol. 2019 Nov;51(11):2037–2044. pmid:31529295