Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Exp Dermatol ; 32(9): 1383-1393, 2023 09.
Article in English | MEDLINE | ID: mdl-37218430

ABSTRACT

Midkine plays a critical role in angiogenesis by regulating the vascular endothelial growth factor (VEGF) signalling pathway, which is known to be associated with psoriasis pathogenesis. However, research on midkine-psoriasis relationship remains limited. The objective of this study was to detect midkine expression in psoriasis and investigate its potential role in the disease. Midkine expression was measured using immunohistochemistry and ELISA. Effects of midkine on HaCaT cell proliferation, VEGF-A production and signalling pathways were assessed using CCK8, RT-PCR and WB. Scratch and in vitro tube formation tests were used to evaluate the effects of HaCaT-cell-activated midkine on the migration and tube formation of human dermal microvascular endothelial cells. Murine psoriasiform models were injected with midkine recombinant protein and midkine monoclonal antibody to investigate skin lesions, tissue sections and dermal microvessel density. Levels of midkine significantly increased in both lesions and serum of patients with psoriasis. Serum expression of midkine decreased after treatment and a positive correlation was found between midkine and disease severity. Midkine promoted HaCaT cell proliferation and VEGF-A production. The Notch2/HES1/JAK2-STAT5A pathway expression increased after midkine treatment of HaCaT cells. The supernatant of HaCaT cells treated with midkine promoted HMEC-1 migration and angiogenesis in vitro. Recombinant midkine protein exacerbated psoriasiform lesions with increased expressions of VEGF-A and microvessel density, while midkine monoclonal antibody alleviated psoriasis lesions. Midkine may have a significant impact on psoriasis angiogenesis by regulating VEGF-A expression through the Notch2/HES1/JAK2-STAT5A pathway, highlighting a potential therapeutic target for psoriasis treatment.


Subject(s)
Psoriasis , Vascular Endothelial Growth Factor A , Humans , Mice , Animals , Vascular Endothelial Growth Factor A/metabolism , Midkine/metabolism , Midkine/pharmacology , Endothelial Cells/metabolism , Psoriasis/metabolism , Cell Proliferation , Antibodies, Monoclonal/therapeutic use
2.
Mar Biotechnol (NY) ; 24(3): 614-625, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35610324

ABSTRACT

With the emergence of antifungal resistance, systematic infections with Aspergillus are becoming the major cause of the clinical morbidity. The development of novel antifungal agents with high efficacy, low drug tolerance, and few side effects is urgent. In response to that need, we have identified NP20. Here we demonstrate clearly that NP20 has antifungal activity, capable of killing the spores of Aspergillus niger and Aspergillus fumigatus as well as causing direct damage to the surface, membrane, cytoplasm, organelle, and nucleus of the fungal spores. Interestingly, NP20 is active under temperature stress and a wide range of pH. Subsequently, MTT assay, assay for binding of NP20 to fungal cell wall components, membrane depolarization assay, confocal microscopy, ROS assay, DNA replication, and protein synthesis assay are performed to clarify the mechanisms underlying NP20 against Aspergillus. The results show that NP20 can bind with and pass through the fungal cell wall, and then interfere with the lipid membrane. Moreover, NP20 can induce intracellular ROS production, DNA fragmentation, and protein synthesis inhibition of the fungal cells. These together indicate that NP20 is a novel antifungal peptide, which has considerable potential for future development as novel peptide antibiotics against Aspergillus.


Subject(s)
Aspergillus fumigatus , Lancelets , Animals , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Aspergillus , Aspergillus fumigatus/metabolism , Aspergillus niger/metabolism , Carrier Proteins , Cytokines , Midkine/metabolism , Midkine/pharmacology , Reactive Oxygen Species/metabolism
3.
Leuk Res ; 116: 106826, 2022 05.
Article in English | MEDLINE | ID: mdl-35316715

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive B-cell non-Hodgkin lymphoma in immunocompromised individuals such as AIDS patients. PEL shows a poor prognosis (median survival time < 6 months) compared with other AIDS-related lymphomas, and is generally resistant to conventional treatments. Novel drugs for PEL treatment are required. Midkine inhibitor (iMDK) was previously found to suppress midkine protein expression. Interestingly, iMDK suppressed cell proliferation in PEL cell lines in a time- and dose-dependent manner, regardless of midkine gene expression. We examined the mechanism of iMDK on PEL. Importantly, iMDK strongly induced cell cycle arrest at the G2/M phase within 12 h of incubation and suppressed the p-CDK1 protein level, which is associated with the cell cycle checkpoint at G2/M, resulting in mitotic catastrophe with observation of multipolar division. After mitotic catastrophe, iMDK-treated PEL showed apoptosis with caspase-3, - 8, and - 9 activation at 24 h incubation. However, iMDK showed no effects on viral protein-activated signaling pathways such as JAK-STAT, PI3K-Akt and NF-κB, and HHV-8/KSHV gene expression in PEL. These results indicate that iMDK is a novel CDK1 inhibitor and a promising lead compound for PEL chemotherapy treatment.


Subject(s)
Herpesvirus 8, Human , Lymphoma, Primary Effusion , Apoptosis , G2 Phase Cell Cycle Checkpoints , Humans , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Midkine/pharmacology , Phosphatidylinositol 3-Kinases/pharmacology , Phosphatidylinositol 3-Kinases/therapeutic use
4.
Anticancer Agents Med Chem ; 22(10): 1962-1969, 2022.
Article in English | MEDLINE | ID: mdl-34477527

ABSTRACT

BACKGROUND: Colon cancers are among the top three causes of cancer-related deaths. This study is a continuation of previous research aiming to identify effective treatments. OBJECTIVE: This study investigated the effects of Tarantula cubensis alcoholic extract (TCAE) and Nerium oleander (NO) distillate on the levels of midkine, transforming growth factor (TGF)-ß, vascular endothelial growth factor (VEGF), alpha-fetoprotein (AFP), cyclooxygenase (COX)-2, insulin-like growth factor (IGF) and caspase-3 in the liver and colon tissues of rats with experimentally induced colon cancer. METHODS: The liver and colon tissues of rats were homogeneously divided into control, colon cancer (azoxymethane, AZM), AZM + TCAE, and AZM + NO distillate groups. The levels of midkine, TGF-ß, VEGF, AFP, COX-2, IGF, and caspase-3 in the colon and liver tissues were measured by ELISA. RESULTS: The levels of all parameters in colon and liver tissues in the AZM group were higher (p<0.05) than those in the control group. TCAE and NO distillate prevented (p < 0.05) increases in midkine, TGF-ß, VEGF, AFP, COX-2, IGF, and caspase-3 levels in the colon. NO distillate prevented the increase in all parameters except IGF, whereas TCAE prevented the increase in all values apart from COX-2 and IGF levels in the liver (p<0.05). CONCLUSION: NO distillate and TCAE may prevent the studied markers from reaching specified levels observed in the colon in AZM-induced colon cancer. The increases in the levels of the parameters in the liver were not as severe as those in the colon; however, an 18-week study period may not be sufficient for liver metastasis formation. Future molecular studies should investigate the mechanisms and pathways of these treatments in greater detail.


Subject(s)
Colonic Neoplasms , Nerium , Spiders , Animals , Biological Products/pharmacology , Biomarkers, Tumor , Caspase 3 , Colonic Neoplasms/drug therapy , Cyclooxygenase 2 , Liver , Midkine/pharmacology , Nerium/chemistry , Plant Extracts/pharmacology , Rats , Spiders/chemistry , Transforming Growth Factor beta , Vascular Endothelial Growth Factor A , alpha-Fetoproteins/pharmacology
5.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34383712

ABSTRACT

Dilated cardiomyopathy (DCM) is the most common form of cardiomyopathy and main indication for heart transplantation in children. Therapies specific to pediatric DCM remain limited due to lack of a disease model. Our previous study showed that treatment of neonatal rat ventricular myocytes (NRVMs) with serum from nonfailing or DCM pediatric patients activates the fetal gene program (FGP). Here we show that serum treatment with proteinase K prevents activation of the FGP, whereas RNase treatment exacerbates it, suggesting that circulating proteins, but not circulating miRNAs, promote these pathological changes. Evaluation of the protein secretome showed that midkine (MDK) is upregulated in DCM serum, and NRVM treatment with MDK activates the FGP. Changes in gene expression in serum-treated NRVMs, evaluated by next-generation RNA-Seq, indicated extracellular matrix remodeling and focal adhesion pathways were upregulated in pediatric DCM serum and in DCM serum-treated NRVMs, suggesting alterations in cellular stiffness. Cellular stiffness was evaluated by Atomic Force Microscopy, which showed an increase in stiffness in DCM serum-treated NRVMs. Of the proteins increased in DCM sera, secreted frizzled-related protein 1 (sFRP1) was a potential candidate for the increase in cellular stiffness, and sFRP1 treatment of NRVMs recapitulated the increase in cellular stiffness observed in response to DCM serum treatment. Our results show that serum circulating proteins promoted pathological changes in gene expression and cellular stiffness, and circulating miRNAs were protective against pathological changes.


Subject(s)
Cardiomyopathy, Dilated/genetics , Extracellular Matrix/drug effects , Focal Adhesions/drug effects , Myocytes, Cardiac/drug effects , Transcriptome/drug effects , Ventricular Remodeling/drug effects , Adolescent , Animals , Animals, Newborn , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Child , Child, Preschool , Endopeptidase K/pharmacology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Focal Adhesions/metabolism , Focal Adhesions/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Microscopy, Atomic Force , Midkine/metabolism , Midkine/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA-Seq , Rats , Ribonucleases/pharmacology , Secretome , Ventricular Remodeling/genetics
6.
Nat Med ; 26(12): 1865-1877, 2020 12.
Article in English | MEDLINE | ID: mdl-33077955

ABSTRACT

An open question in aggressive cancers such as melanoma is how malignant cells can shift the immune system to pro-tumorigenic functions. Here we identify midkine (MDK) as a melanoma-secreted driver of an inflamed, but immune evasive, microenvironment that defines poor patient prognosis and resistance to immune checkpoint blockade. Mechanistically, MDK was found to control the transcriptome of melanoma cells, allowing for coordinated activation of nuclear factor-κB and downregulation of interferon-associated pathways. The resulting MDK-modulated secretome educated macrophages towards tolerant phenotypes that promoted CD8+ T cell dysfunction. In contrast, genetic targeting of MDK sensitized melanoma cells to anti-PD-1/anti-PD-L1 treatment. Emphasizing the translational relevance of these findings, the expression profile of MDK-depleted tumors was enriched in key indicators of a good response to immune checkpoint blockers in independent patient cohorts. Together, these data reveal that MDK acts as an internal modulator of autocrine and paracrine signals that maintain immune suppression in aggressive melanomas.


Subject(s)
Carcinogenesis/drug effects , Melanoma, Experimental/therapy , Midkine/genetics , Tumor Microenvironment/genetics , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genetic Therapy , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Midkine/pharmacology , NF-kappa B/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Transcriptome/genetics
7.
Alcohol Clin Exp Res ; 44(5): 1037-1045, 2020 05.
Article in English | MEDLINE | ID: mdl-32154588

ABSTRACT

BACKGROUND: Pleiotrophin (PTN) and midkine (MK) are cytokines that are up-regulated in the prefrontal cortex (PFC) after alcohol administration and have been shown to reduce alcohol intake and reward. Both cytokines are endogenous inhibitors of receptor protein tyrosine phosphatase (RPTP) ß/ζ (a.k.a. PTPRZ1). Recently, a new compound named MY10 was designed with the aim of mimicking the activity of PTN and MK. MY10 has already shown promising results regulating alcohol-related behaviors in mice. METHODS: We have now tested the effects of MY10 on alcohol operant self-administration and Drinking In the Dark-Multiple Scheduled Access (DID-MSA) paradigms in rats. Gene expression of relevant genes in the PTN/MK signaling pathway in the PFC was analyzed by real-time PCR. RESULTS: MY10, at the highest dose tested (100 mg/kg), reduced alcohol consumption in the alcohol operant self-administration paradigm (p = 0.040). In the DID-MSA paradigm, rats drank significantly less alcohol (p = 0.019) and showed a significant decrease in alcohol preference (p = 0.002). We observed that the longer the exposure to alcohol, the greater the suppressing effects of MY10 on alcohol consumption. It was demonstrated that the effects of MY10 were specific to alcohol since saccharin intake was not affected by MY10 (p = 0.804). MY10 prevented the alcohol-induced down-regulation of Ptprz1 (p = 0.004) and anaplastic lymphoma kinase (Alk; p = 0.013) expression. CONCLUSIONS: Our results support and provide further evidence regarding the efficacy of MY10 on alcohol-related behaviors and suggest the consideration of the blockade of RPTPß/ζ as a target for reducing excessive alcohol consumption.


Subject(s)
Alcohol Drinking/drug therapy , Enzyme Inhibitors/administration & dosage , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Animals , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cytokines/genetics , Cytokines/pharmacology , Dose-Response Relationship, Drug , Gene Expression/drug effects , Male , Midkine/genetics , Midkine/pharmacology , Rats , Rats, Wistar , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Signal Transduction/genetics
8.
Circ J ; 84(2): 217-225, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31915322

ABSTRACT

BACKGROUND: Midkine (MK), a heparin-binding protein, participates in multiple cellular processes, such as immunity, cellular growth and apoptosis. Overwhelming evidence indicates that MK plays an important role in various pathological processes, including chronic inflammation, autoimmunity, cancer, and infection. Recent studies demonstrated that MK may be involved in the development of atherosclerosis, yet the mechanism has not been fully explored. Therefore, this study aims to investigate the effect and mechanism of MK on macrophage cholesterol efflux.Methods and Results:Using Oil Red O staining, NBD-cholesterol fluorescence labeling and enzymatic methods, it observed that MK markedly promoted macrophage lipid accumulation. Liquid scintillation counting (LSC) showed that MK decreased cholesterol efflux. Moreover, cell immunofluorescence, western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) showed that MK downregulated ATP-binding membrane cassette transport protein A1 (ABCA1) expression. Functional promotion of ABCA1 expression attenuated the inhibitory effects of MK on cholesterol efflux, which reduced lipid accumulation. Additionally, intervention of adenosine monophosphate activated protein (AMPK)-mammalian target of rapamycin (mTOR) signaling molecule by the AMPK activator, AICAR, increased p-AMPK and ABCA1 expression, decreased p-mTOR expression and promoted cholesterol efflux, resulting in an obvious reduction in intracellular lipid content. CONCLUSIONS: These data suggest that MK reduces the expression of ABCA1, inhibits the efflux of cholesterol and promotes the accumulation of lipids in RAW264.7 macrophages, and AMPK-mTOR signaling is involved in MK-mediated regulation of cholesterol metabolism in RAW264.7 macrophages.


Subject(s)
AMP-Activated Protein Kinases/metabolism , ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , Macrophages/drug effects , Midkine/pharmacology , TOR Serine-Threonine Kinases/metabolism , ATP Binding Cassette Transporter 1/genetics , Animals , Down-Regulation , Enzyme Activation , Macrophages/enzymology , Mice , Phosphorylation , RAW 264.7 Cells , Signal Transduction
9.
Cell Signal ; 65: 109423, 2020 01.
Article in English | MEDLINE | ID: mdl-31639491

ABSTRACT

Osteoarthritis (OA) is the most common disease of joint tissues; unfortunately, there are currently no curative therapies available for OA. Chondrocytes, the only cell type residing in cartilage, secrete many types of collagen (the mainly one is type II collagen) and aggrecan, which are the main components of the cartilage matrix. Chondrocyte apoptosis can lead to OA degenerative progression. We previously indicated that recombinant human midkine (rhMK), as a chondrocyte growth factor has a significant reparative effect on cartilage injury animal models. However, the molecular mechanism of this restorative function remains under investigation. Herein, we focused on the molecular mechanism underlying the role of MK in promoting the proliferation of chondrocytes cultured in vitro. Chondrocytes from rats and OA patients were successfully isolated by the digestion of articular cartilage using type II collagenase, and their proliferation was evaluated by a CCK8 assay and flow cytometry. rhMK stimulated the proliferation of chondrocytes from both OA patients and rats. Furthermore, qRT-PCR, shRNA-mediated knockdown, Western blot and immunoprecipitation (IP) assays were performed to identify the receptor and key elements responsible for the role of MK in promoting chondrocyte proliferation. Low-density lipoprotein receptor-related protein 1 (LRP1) was identified as the dominant MK receptor in chondrocytes that, as a translocator, mediates the endocytosis of MK. After being transferred into chondrocytes, MK was shown to form a complex with nucleolin that interacts with the active form of K-Ras. Upon the activation of ERK1/2, cyclin D1 expression was upregulated, promoting the chondrocyte cell cycle. Our data reveal for the first time the role of the MK-LRP1-nucleolin signaling pathway in facilitating MK-induced chondrocyte proliferation, thus providing a strong theoretical foundation for the further use of MK in OA clinical therapy.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Chondrocytes/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Midkine/pharmacology , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Aged , Animals , Cell Proliferation/drug effects , Chondrocytes/drug effects , Endocytosis/drug effects , Female , Humans , Male , Middle Aged , Osteoarthritis/pathology , Protein Binding/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , Rats, Sprague-Dawley , Nucleolin
10.
Biomed Pharmacother ; 107: 793-805, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30142541

ABSTRACT

AIMS: To examine the functions of growth factor midkine (MK) and a flavonoid quercetin on survival, apoptosis and migration of prostate cancer (PCa) stem cells (CSCs). MAIN METHODS: CD44+/CD133+ and CD44+ stem cells were isolated from PC3 and LNCaP cells, respectively by magnetic-activated cell sorting system. 3D cell culture was used to evaluate the ability of quercetin, MK siRNA, and the combination of both to inhibit spheroid formation, apoptosis and cell cycle arrest. Image-based cytometer, RT-qPCR, Western blotting and transwell migration assays were performed. KEY FINDINGS: Quercetin treatment for 24-72 h inhibited PC3 and CD44+/CD133+ stem cell proliferation in a time- and dose-dependent manner. Knockdown of endogenous MK expression significantly suppressed proliferation of CD44+/CD133+ and CD44+ cells as well as their parent cells. Co-administration of MK siRNA and quercetin reduced the cell survival, induced apoptosis and caused G1 phase cell cycle arrest more effectively than the individual therapy. Knockdown of MK significantly enhanced the inhibitory effect of quercetin on CD44+/CD133+ migration and spheroid formation. In addition, the combined therapy inhibited the phosphorylation of PI3K, AKT and ERK1/2, and reduced the protein expression of p38, ABCG2 and NF-κB. SIGNIFICANCE: Quercetin alone exhibited significant cytotoxic effects on CD44+/CD133+. MK plays an important role in the proliferation of CD44+/CD133+ and CD44+ cells in particular, and quercetin and MK-silencing therapy may be an important strategy in targeting CSCs that play a role in relapse, migration and drug resistance.


Subject(s)
Cell Movement/drug effects , Down-Regulation/drug effects , Midkine/pharmacology , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Quercetin/pharmacology , Signal Transduction/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/genetics , Cell Separation , Cell Survival/drug effects , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System/drug effects , Male , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
11.
Sci Rep ; 7(1): 16231, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29176691

ABSTRACT

Midkine (MK) is a heparin-binding growth factor that promotes carcinogenesis and chemoresistance. The tumour microenvironment (TME) can affect chemotherapy sensitivity. However, the role of stromal-derived MK, especially in cancer-associated fibroblasts (CAFs), is unclear. Here, we confirmed that MK decreased cisplatin-induced cell death in oral squamous cell carcinoma (OSCC) cells, ovarian cancer cells and lung cancer cells. We also isolated primary CAFs (n = 3) from OSCC patients and found that CAFs secreted increased levels of MK, which abrogated cisplatin-induced cell death. Moreover, MK increased the expression of lncRNA ANRIL in the tumour cells. Normal tissues, matched tumour-adjacent tissues and OSCC tissues were analysed (n = 60) and showed that lncRNA ANRIL was indeed overexpressed during carcinogenesis and correlated with both high TNM stage and lymph node metastasis (LNM). Furthermore, lncRNA ANRIL knockdown in tumour cells inhibited proliferation, induced apoptosis and increased cisplatin cytotoxicity of the tumour cells via impairment of the drug transporters MRP1 and ABCC2, which could be restored by treatment with human MK in a caspase-3/BCL-2-dependent manner. In conclusion, we firstly describe that CAFs in the TME contribute to the high level of MK in tumours and that CAF-derived MK can promote cisplatin resistance via the elevated expression of lncRNA ANRIL.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Drug Resistance, Neoplasm , Midkine/pharmacology , RNA, Long Noncoding/genetics , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/toxicity , Humans , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , RNA, Long Noncoding/metabolism , Up-Regulation
12.
Circ J ; 82(1): 19-27, 2017 12 25.
Article in English | MEDLINE | ID: mdl-28781288

ABSTRACT

BACKGROUND: A recent study suggested that midkine (MK), a heparin-binding growth factor, is associated with atherosclerosis progression in patients with artery disease. It has previously been reported that MK plays a critical role in neointima formation in a restenosis model, whereas the role of MK in the development of atherosclerosis has not been investigated. The present study assessed the effect of MK administration on the process of atherosclerotic plaque formation in apolipoprotein E-knockout (ApoE-/-) mice.Methods and Results:Using an osmotic pump, human recombinant MK protein was intraperitoneally administered for 12 weeks in C57BL/6 ApoE-/-(ApoE-/--MK) and ApoE+/+mice fed a high-fat diet. Saline was administered to the control groups of ApoE-/-(ApoE-/--saline) and ApoE+/+mice. The atherosclerotic lesion areas in longitudinal aortic sections were significantly larger in ApoE-/--MK mice than in ApoE-/--saline mice. The aortic mRNA levels of pro-inflammatory and angiogenic factors, and the percentage of macrophages in aortic root lesions, were significantly higher in ApoE-/--MK mice than in ApoE-/--saline mice, whereas the percentage of apoptotic cells was significantly lower in ApoE-/--MK mice than in ApoE-/--saline mice. CONCLUSIONS: The systemic administration of MK in ApoE-/-mice promoted atherosclerotic plaque formation through pro-inflammatory, angiogenic, and anti-apoptotic effects. MK may serve as a potential therapeutic target for the prevention of atherosclerosis under atherogenic conditions.


Subject(s)
Apoptosis/drug effects , Inflammation/chemically induced , Midkine/pharmacology , Neovascularization, Pathologic/chemically induced , Plaque, Atherosclerotic/pathology , Animals , Aorta/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Plaque, Atherosclerotic/etiology , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...