1
|
Xia M, Tong S, Gao L. Identification of MDK as a Hypoxia- and Epithelial-Mesenchymal Transition-Related Gene Biomarker of Glioblastoma Based on a Novel Risk Model and In Vitro Experiments. Biomedicines 2024; 12:92. [PMID: 38255198 PMCID: PMC10813330 DOI: 10.3390/biomedicines12010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/23/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Tumor cells are commonly exposed to a hypoxic environment, which can easily induce the epithelial-mesenchymal transition (EMT) of tumor cells, further affecting tumor proliferation, invasion, metastasis, and drug resistance. However, the predictive role of hypoxia and EMT-related genes in glioblastoma (GBM) has not been investigated. METHODS Intersection genes were identified by weighted correlation network analysis (WGCNA) and differential expression analyses, and a risk model was further constructed by LASSO and Cox analyses. Clinical, immune infiltration, tumor mutation, drug treatment, and enrichment profiles were analyzed based on the risk model. The expression level of the MDK gene was tested using RT-PCR, immunohistochemistry, and immunofluorescence. CCK8 and EdU were employed to determine the GBM cells' capacity for proliferation while the migration and invasion ability were detected by a wound healing assay and transwell assay, respectively. RESULTS Based on the GBM data of the TCGA and GTEx databases, 58 intersection genes were identified, and a risk model was constructed. The model was verified in the CGGA cohort, and its accuracy was confirmed by the ROC curve (AUC = 0.807). After combining clinical subgroups, univariate and multivariate Cox regression analyses showed that risk score and age were independent risk factors for GBM patients. Furthermore, our subsequent analysis of immune infiltration, tumor mutation, and drug treatment showed that risk score and high- and low-risk groups were associated with multiple immune cells, mutated genes, and drugs. Enrichment analysis indicated that the differences between high- and low-risk groups were manifested in tumor-related pathways, including the PI3K-AKT and JAK-STAT pathways. Finally, in vivo experiments proved that the hypoxia environment promoted the expression of MDK, and MDK knockdown reduced the proliferation, migration, and EMT of GBM cells induced by hypoxia. CONCLUSIONS Our novel prognostic correlation model provided more potential treatment strategies for GBM patients.
Collapse
Affiliation(s)
- Minqi Xia
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shiao Tong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ling Gao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
2
|
Neumaier EE, Rothhammer V, Linnerbauer M. The role of midkine in health and disease. Front Immunol 2023; 14:1310094. [PMID: 38098484 PMCID: PMC10720637 DOI: 10.3389/fimmu.2023.1310094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023] Open
Abstract
Midkine (MDK) is a neurotrophic growth factor highly expressed during embryogenesis with important functions related to growth, proliferation, survival, migration, angiogenesis, reproduction, and repair. Recent research has indicated that MDK functions as a key player in autoimmune disorders of the central nervous system (CNS), such as Multiple Sclerosis (MS) and is a promising therapeutic target for the treatment of brain tumors, acute injuries, and other CNS disorders. This review summarizes the modes of action and immunological functions of MDK both in the peripheral immune compartment and in the CNS, particularly in the context of traumatic brain injury, brain tumors, neuroinflammation, and neurodegeneration. Moreover, we discuss the role of MDK as a central mediator of neuro-immune crosstalk, focusing on the interactions between CNS-infiltrating and -resident cells such as astrocytes, microglia, and oligodendrocytes. Finally, we highlight the therapeutic potential of MDK and discuss potential therapeutic approaches for the treatment of neurological disorders.
Collapse
Affiliation(s)
| | - Veit Rothhammer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
3
|
Kam NW, Lau CY, Che CM, Lee VHF. Nasopharynx Battlefield: Cellular Immune Responses Mediated by Midkine in Nasopharyngeal Carcinoma and COVID-19. Cancers (Basel) 2023; 15:4850. [PMID: 37835544 PMCID: PMC10571800 DOI: 10.3390/cancers15194850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Clinical evidence suggests that the severe respiratory illness coronavirus disease 2019 (COVID-19) is often associated with a cytokine storm that results in dysregulated immune responses. Prolonged COVID-19 positivity is thought to disproportionately affect cancer patients. With COVID-19 disrupting the delivery of cancer care, it is crucial to gain momentum and awareness of the mechanistic intersection between these two diseases. This review discusses the role of the cytokine midkine (MK) as an immunomodulator in patients with COVID-19 and nasopharyngeal carcinoma (NPC), both of which affect the nasal cavity. We conducted a review and analysis of immunocellular similarities and differences based on clinical studies, research articles, and published transcriptomic datasets. We specifically focused on ligand-receptor pairs that could be used to infer intercellular communication, as well as the current medications used for each disease, including NPC patients who have contracted COVID-19. Based on our findings, we recommend close monitoring of the MK axis to maintain the desirable effects of therapeutic regimens in fighting both NPC and COVID-19 infections.
Collapse
Affiliation(s)
- Ngar-Woon Kam
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; (N.-W.K.); (C.-Y.L.)
- Laboratory for Synthetic Chemistry and Chemical Biology Ltd., Hong Kong Science Park, New Territories, Hong Kong 999077, China;
| | - Cho-Yiu Lau
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; (N.-W.K.); (C.-Y.L.)
- Laboratory for Synthetic Chemistry and Chemical Biology Ltd., Hong Kong Science Park, New Territories, Hong Kong 999077, China;
| | - Chi-Ming Che
- Laboratory for Synthetic Chemistry and Chemical Biology Ltd., Hong Kong Science Park, New Territories, Hong Kong 999077, China;
- Department of Chemistry, Faculty of Science, The University of Hong Kong, Hong Kong 999077, China
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; (N.-W.K.); (C.-Y.L.)
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
4
|
Papadimitriou E, Kanellopoulou VK. Protein Tyrosine Phosphatase Receptor Zeta 1 as a Potential Target in Cancer Therapy and Diagnosis. Int J Mol Sci 2023; 24:ijms24098093. [PMID: 37175798 PMCID: PMC10178973 DOI: 10.3390/ijms24098093] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Protein tyrosine phosphatase receptor zeta 1 (PTPRZ1) is a type V transmembrane tyrosine phosphatase that is highly expressed during embryonic development, while its expression during adulthood is limited. PTPRZ1 is highly detected in the central nervous system, affecting oligodendrocytes' survival and maturation. In gliomas, PTPRZ1 expression is significantly upregulated and is being studied as a potential cancer driver and as a target for therapy. PTPRZ1 expression is also increased in other cancer types, but there are no data on the potential functional significance of this finding. On the other hand, low PTPRZ1 expression seems to be related to a worse prognosis in some cancer types, suggesting that in some cases, it may act as a tumor-suppressor gene. These discrepancies may be due to our limited understanding of PTPRZ1 signaling and tumor microenvironments. In this review, we present evidence on the role of PTPRZ1 in angiogenesis and cancer and discuss the phenomenal differences among the different types of cancer, depending on the regulation of its tyrosine phosphatase activity or ligand binding. Clarifying the involved signaling pathways will lead to its efficient exploitation as a novel therapeutic target or as a biomarker, and the development of proper therapeutic approaches.
Collapse
Affiliation(s)
- Evangelia Papadimitriou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | - Vasiliki K Kanellopoulou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| |
Collapse
|
5
|
Midkine mediates dysfunction of liver sinusoidal endothelial cells through integrin α4 and α6. Vascul Pharmacol 2022; 147:107113. [PMID: 36184060 DOI: 10.1016/j.vph.2022.107113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 01/30/2023]
Abstract
Midkine (MK)2 is an important regulatory molecule that promotes pathological angiogenesis of hepatocellular carcinoma (HCC). Although some studies have shown that its expression is increased in chronic liver disease, its effect on sinusoidal vasculopathy are still unclear. In this study, we demonstrated that MK was mainly secreted by liver sinus endothelial cells (LSECs) during the stage of precancerous lesions. Increased expression of its receptor integrin was an important mechanism by which MK participated in sinusoidal vasculopathy through autocrine and positive feedback effects. LSECs with high expression of integrin α6 (Itgα6+) and integrin α4 (Itgα4+) were used to study the mechanism of MK, and it was found that the effect of MK on LSECs was closely related to the integrin subtypes. The activation of MK /integrin α6/Src/Shc signaling pathway promoted the expression of ET-1, TXA2 and reduced the production of NO, and then induced the capillary vascularization of liver sinusoids, while the activation of MK/integrin α4/NF-κB pathway mainly induced angiogenesis by promoting the production of VEGF and Ang2. In the three-dimensional co-culture system of hepatocytes (BRL-3A) and LSECs, MK significantly increased the production of reactive oxygen species (ROS) in the co-culture system of highly expressed integrin LSECs and decreased the expression of tumor suppressor gene P53 in hepatocytes. These results suggested that MK /integrin signaling pathway, especially MK /integrin α6, was an important mechanism leaded to persistent sinusoidal hepatic vasculopathy in chronic liver disease and induced HCC,while MK/integrin α 4 activation was more involved in pathological angiogenesis.
Collapse
|
6
|
Majaj M, Weckbach LT. Midkine-A novel player in cardiovascular diseases. Front Cardiovasc Med 2022; 9:1003104. [PMID: 36204583 PMCID: PMC9530663 DOI: 10.3389/fcvm.2022.1003104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/30/2022] [Indexed: 11/18/2022] Open
Abstract
Midkine (MK) is a 13-kDa heparin-binding cytokine and growth factor with anti-apoptotic, pro-angiogenic, pro-inflammatory and anti-infective functions, that enable it to partake in a series of physiological and pathophysiological processes. In the past, research revolving around MK has concentrated on its roles in reproduction and development, tissue protection and repair as well as inflammatory and malignant processes. In the recent few years, MK's implication in a wide scope of cardiovascular diseases has been rigorously investigated. Nonetheless, there is still no broadly accepted consensus on whether MK exerts generally detrimental or favorable effects in cardiovascular diseases. The truth probably resides somewhere in-between and depends on the underlying physiological or pathophysiological condition. It is therefore crucial to thoroughly examine and appraise MK's participation in cardiovascular diseases. In this review, we introduce the MK gene and protein, its multiple receptors and signaling pathways along with its expression in the vascular system and its most substantial functions in cardiovascular biology. Further, we recapitulate the current evidence of MK's expression in cardiovascular diseases, addressing the various sources and modes of MK expression. Moreover, we summarize the most significant implications of MK in cardiovascular diseases with particular emphasis on MK's advantageous and injurious functions, highlighting its ample diagnostic and therapeutic potential. Also, we focus on conflicting roles of MK in a number of cardiovascular diseases and try to provide some clarity and guidance to MK's multifaceted roles. In summary, we aim to pave the way for MK-based diagnostics and therapies that could present promising tools in the diagnosis and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Marina Majaj
- Walter Brendel Centre for Experimental Medicine, Biomedical Centre, Institute for Cardiovascular Physiology und Pathophysiology, Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ludwig T. Weckbach
- Walter Brendel Centre for Experimental Medicine, Biomedical Centre, Institute for Cardiovascular Physiology und Pathophysiology, Ludwig-Maximilians-University Munich, Munich, Germany
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e. V, Berlin, Germany
| |
Collapse
|
7
|
Malmhäll-Bah E, Andersson KME, Erlandsson MC, Akula MK, Brisslert M, Wiel C, El Zowalaty AE, Sayin VI, Bergö MO, Bokarewa MI. Rho-GTPase dependent leukocyte interaction generates pro-inflammatory thymic Tregs and causes arthritis. J Autoimmun 2022; 130:102843. [PMID: 35643017 DOI: 10.1016/j.jaut.2022.102843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 12/23/2022]
Abstract
Conditional mutation of protein geranylgeranyltransferase type I (GGTase-I) in macrophages (GLC) activates Rho-GTPases and causes arthritis in mice. Knocking out Rag1 in GLC mice alleviates arthritis which indicates that lymphocytes are required for arthritis development in those mice. To study GLC dependent changes in the adaptive immunity, we isolated CD4+ T cells from GLC mice (CD4+GLCs). Spleen and joint draining lymph nodes (dLN) CD4+GLCs exhibited high expression of Cdc42 and Rac1, which repressed the caudal HOXA proteins and activated the mechanosensory complex to facilitate migration. These CDC42/RAC1 rich CD4+GLCs presented a complete signature of GARP+NRP1+IKZF2+FOXP3+ regulatory T cells (Tregs) of thymic origin. Activation of the β-catenin/Lef1 axis promoted a pro-inflammatory Th1 phenotype of Tregs, which was strongly associated with arthritis severity. Knockout of Cdc42 in macrophages of GLC mice affected CD4+ cell biology and triggered development of non-thymic Tregs. Knockout of Rac1 and RhoA had no such effects on CD4+ cells although it alleviated arthritis in GLC mice. Disrupting macrophage and T cell interaction with CTLA4 fusion protein reduced the Th1-driven inflammation and enrichment of thymic Tregs into dLNs. Antigen challenge reinforced the CD4+GLC phenotype in non-arthritic heterozygote GLC mice and increased accumulation of Rho-GTPase expressing thymic Tregs in dLNs. Our study demonstrates an unexpected role of macrophages in stimulating the development of pro-inflammatory thymic Tregs and reveal activation of Rho-GTPases behind their arthritogenic phenotype.
Collapse
Affiliation(s)
- Eric Malmhäll-Bah
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Box 480, 40530, Gothenburg, Sweden
| | - Karin M E Andersson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Box 480, 40530, Gothenburg, Sweden
| | - Malin C Erlandsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Box 480, 40530, Gothenburg, Sweden; Rheumatology Clinic, Sahlgrenska University Hospital, Gröna Stråket 16, 41346, Gothenburg, Sweden
| | - Murali K Akula
- Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 40530, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Mikael Brisslert
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Box 480, 40530, Gothenburg, Sweden
| | - Clotilde Wiel
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Ahmed E El Zowalaty
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Volkan I Sayin
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Martin O Bergö
- Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 40530, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, 14183, Huddinge, Sweden
| | - Maria I Bokarewa
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Box 480, 40530, Gothenburg, Sweden; Rheumatology Clinic, Sahlgrenska University Hospital, Gröna Stråket 16, 41346, Gothenburg, Sweden.
| |
Collapse
|
8
|
Komata Y, Tsubota S, Sakamoto K, Ikematsu S, Kadomatsu K. Screening of novel Midkine binding protein by BioID2-based proximity labeling. NAGOYA JOURNAL OF MEDICAL SCIENCE 2021; 83:495-508. [PMID: 34552285 PMCID: PMC8438011 DOI: 10.18999/nagjms.83.3.495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/09/2020] [Indexed: 12/31/2022]
Abstract
Midkine (MK), a heparin-binding growth factor, is associated with the poor prognosis of the pediatric tumor, neuroblastoma. MK would be a druggable target as many studies showed inhibition of its function in various cancers suppressed tumor developments. To establish the therapy targeting MK, identification of its binding partners, and elucidation of its intracellular signaling are needed. It was reported that exogenous MK induced phosphorylation of ribosomal protein S6 (RPS6) downstream of mTOR signaling. Using RPS6 phosphorylation as a marker of MK response, we searched for MK reactive cell lines. We found that MK cell lines expressing less MK tended to respond better to MK. Next, using an MK reactive neuroblastoma cell line, MK-knocked down SH-SY5Y cells, we employed a proximity-dependent biotin identification method, which was invented to evaluate protein-protein interactions by biotinylation. We confirmed that secreted MK fused to the biotin ligase BioID2 (MK-BioID2) was able to biotinylate proteins from the cells. Biotinylated proteins were identified by liquid chromatography-mass spectrometry analyses. Twenty five proteins were found to be overlapped after three independent experiments, among which insulin-like growth binding protein 2 (IGFBP2) was further analyzed. IGFBP2 was indeed detected with immunoblotting after streptavidin pull down of MK-BioID2 labeled cell extract of MK-knocked down SH-SY5Y cells. Our study suggests that the BioID2 method is useful to identify binding partners of growth factors.
Collapse
Affiliation(s)
- Yosuke Komata
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoma Tsubota
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuma Sakamoto
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Shinya Ikematsu
- Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Naha, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| |
Collapse
|
9
|
Slow Off-Rate Modified Aptamer (SOMAmer) Proteomic Analysis of Patient-Derived Malignant Glioma Identifies Distinct Cellular Proteomes. Int J Mol Sci 2021; 22:ijms22179566. [PMID: 34502484 PMCID: PMC8431317 DOI: 10.3390/ijms22179566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 02/04/2023] Open
Abstract
Malignant gliomas derive from brain glial cells and represent >75% of primary brain tumors. This includes anaplastic astrocytoma (grade III; AS), the most common and fatal glioblastoma multiforme (grade IV; GBM), and oligodendroglioma (ODG). We have generated patient-derived AS, GBM, and ODG cell models to study disease mechanisms and test patient-centered therapeutic strategies. We have used an aptamer-based high-throughput SOMAscan® 1.3K assay to determine the proteomic profiles of 1307 different analytes. SOMAscan® proteomes of AS and GBM self-organized into closely adjacent proteomes which were clearly distinct from ODG proteomes. GBM self-organized into four proteomic clusters of which SOMAscan® cluster 4 proteome predicted a highly inter-connected proteomic network. Several up- and down-regulated proteins relevant to glioma were successfully validated in GBM cell isolates across different SOMAscan® clusters and in corresponding GBM tissues. Slow off-rate modified aptamer proteomics is an attractive analytical tool for rapid proteomic stratification of different malignant gliomas and identified cluster-specific SOMAscan® signatures and functionalities in patient GBM cells.
Collapse
|
10
|
Zhang ZZ, Wang G, Yin SH, Yu XH. Midkine: A multifaceted driver of atherosclerosis. Clin Chim Acta 2021; 521:251-257. [PMID: 34331952 DOI: 10.1016/j.cca.2021.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022]
Abstract
Atherosclerosis constitutes the pathological basis of life-threatening events, including heart attack and stroke. Midkine is a heparin-binding growth factor and forms a small protein family with pleiotrophin. Under inflammatory or hypoxic conditions, midkine expression is up-regulated. Upon binding to its receptors, midkine can activate multiple signal pathways to regulate cell survival and migration, epithelial-to-mesenchymal transition, and oncogenesis. Circulating midkine levels are significantly increased in patients with essential hypertension, obesity or severe peripheral artery disease. Importantly, midkine exerts a proatherogenic effect by altering multiple pathophysiological processes involving atherogenesis, including macrophage lipid accumulation, vascular inflammation, neointima formation, insulin resistance and macrophage apoptosis. Midkine represents a potential therapeutic target for atherosclerosis-associated diseases. This review described the structure characteristics, expression patterns and signal transduction pathways of midkine with an emphasis on its role in atherosclerosis.
Collapse
Affiliation(s)
- Zi-Zhen Zhang
- School of Medicine, Hunan Polytechnic of Environment and Biology, Hengyang 421005, Hunan, China
| | - Gang Wang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, Hunan, China
| | - Shan-Hui Yin
- Department of Neonatology, The First Affiliated Hospital of University of South China, Hengyang 421001, Hunan, China.
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou 570100, Hainan, China.
| |
Collapse
|
11
|
Gowhari Shabgah A, Ezzatifar F, Aravindhan S, Olegovna Zekiy A, Ahmadi M, Gheibihayat SM, Gholizadeh Navashenaq J. Shedding more light on the role of Midkine in hepatocellular carcinoma: New perspectives on diagnosis and therapy. IUBMB Life 2021; 73:659-669. [PMID: 33625758 DOI: 10.1002/iub.2458] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/06/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
One of the most common malignant tumors is hepatocellular carcinoma (HCC). Progression of HCC mainly results from highly complex molecular and pathological pathways. Midkine (MDK) is a growth factor that impacts viability, migration, and other cell activities. Since MDK has been involved in the inflammatory responses, it has been claimed that MDK has a crucial role in HCC. MDK acts as an anti-apoptotic factor, which mediates tumor cell viability. In addition, MDK blocks anoikis to promote metastasis. There is also evidence that MDK is involved in angiogenesis. It has been shown that the application of anti-MDK approaches might be promising in the treatment of HCC. Besides, due to the elevated expression in HCC, MDK has been proposed as a biomarker in the prognosis and diagnosis of HCC. In this review, we will discuss the role of MDK in HCC. It is hoped that the development of new strategies concerning MDK-based therapies will be promising in HCC management.
Collapse
Affiliation(s)
- Arezoo Gowhari Shabgah
- School of Medicine, Bam University of Medical Sciences, Bam, Iran.,Student Research Committee, Bam University of Medical Sciences, Bam, Iran
| | - Fatemeh Ezzatifar
- Molecular and Cell Biology Research Center, Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Surendar Aravindhan
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Biotechnology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Jamshid Gholizadeh Navashenaq
- Student Research Committee, Bam University of Medical Sciences, Bam, Iran.,Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| |
Collapse
|
12
|
Campbell WA, Fritsch-Kelleher A, Palazzo I, Hoang T, Blackshaw S, Fischer AJ. Midkine is neuroprotective and influences glial reactivity and the formation of Müller glia-derived progenitor cells in chick and mouse retinas. Glia 2021; 69:1515-1539. [PMID: 33569849 DOI: 10.1002/glia.23976] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 01/03/2023]
Abstract
Recent studies suggest midkine (MDK) is involved in the development and regeneration of the zebrafish retina. We investigate the expression patterns of MDK and related factors, roles in neuronal survival, and influence upon the formation of Müller glia-derived progenitor cells (MGPCs) in chick and mouse model systems. By using single-cell RNA-sequencing, we find that MDK and pleiotrophin (PTN), a MDK-related cytokine, are upregulated by Müller glia (MG) during later stages of development in chick. While PTN is downregulated, MDK is dramatically upregulated in mature MG after retinal damage or FGF2 and insulin treatment. By comparison, MDK and PTN are downregulated by MG in damaged mouse retinas. In both chick and mouse retinas, exogenous MDK induces expression of cFos and pS6 in MG. In the chick, MDK significantly decreases numbers dying neurons, reactive microglia, and proliferating MGPCs, whereas PTN has no effect. Inhibition of MDK-signaling with Na3 VO4 blocks neuroprotective effects with an increase in the number of dying cells and negates the pro-proliferative effects on MGPCs in damaged retinas. Inhibitors of PP2A and Pak1, which are associated with MDK-signaling through integrin β1, suppressed the formation of MGPCs in damaged chick retinas. In mice, MDK promotes a small but significant increase in proliferating MGPCs in damaged retinas and potently decreases the number of dying cells. We conclude that MDK expression is dynamically regulated in Müller glia during embryonic maturation, following retinal injury, and during reprogramming into MGPCs. MDK mediates glial activity, neuronal survival, and the re-programming of Müller glia into proliferating MGPCs.
Collapse
Affiliation(s)
- Warren A Campbell
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Amanda Fritsch-Kelleher
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Isabella Palazzo
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Thanh Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
13
|
Emami MH, Soltani S, Eskandari N, Masjedi M. The Circulating Midkine in the Newly Diagnosed Celiac Disease: Clinical Implications. Adv Biomed Res 2021; 10:1. [PMID: 33959558 PMCID: PMC8095258 DOI: 10.4103/abr.abr_8_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/03/2020] [Accepted: 10/19/2020] [Indexed: 11/17/2022] Open
Abstract
Background: Celiac disease (CeD) is a chronic inflammatory small intestine disorder caused by an abnormal immune response to an array of the epitopes of the wheat gluten and related proteins of rye and barley in genetically susceptible individuals. Midkine (MK) is an angiogenic cytokine, chemotactic in the direction of polymorphonuclear neutrophils and macrophages, and a T-regulatory cell suppressor. So far, a possible relationship with CeD has not yet been explored. Diagnosis of CeD is based on serologic test in a clinical setting suggestive of CeD and confirmatory histologic examination of the duodenal biopsy. Sometimes, genetic testing of human leukocyte antigen (HLA)-DQ2 and HLA-DQ8 may be needed. The objective of this study was to measure and compare the circulating MK in the celiac patients and healthy individuals. Materials and Methods: Twenty newly untreated CeD cases and 20 normal controls were enrolled in this study. The enzyme-linked immunosorbent assay was used to measure the circulating MK in the celiac patients and controls. Results: There was insignificant difference in the circulating MK between the patients and controls (P > 0.05). Conclusions: The study results suggest that the MK marker does not have any diagnostic value in CeD activity to be used at the time of diagnosis or during follow-ups.
Collapse
Affiliation(s)
- Mohammad Hassan Emami
- Poursina Hakim Digestive Diseases Research Centre, Isfahan University of Medical Sciences, Isahan, Iran
| | - Shima Soltani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Masjedi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
14
|
Ross-Munro E, Kwa F, Kreiner J, Khore M, Miller SL, Tolcos M, Fleiss B, Walker DW. Midkine: The Who, What, Where, and When of a Promising Neurotrophic Therapy for Perinatal Brain Injury. Front Neurol 2020; 11:568814. [PMID: 33193008 PMCID: PMC7642484 DOI: 10.3389/fneur.2020.568814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Midkine (MK) is a small secreted heparin-binding protein highly expressed during embryonic/fetal development which, through interactions with multiple cell surface receptors promotes growth through effects on cell proliferation, migration, and differentiation. MK is upregulated in the adult central nervous system (CNS) after multiple types of experimental injury and has neuroprotective and neuroregenerative properties. The potential for MK as a therapy for developmental brain injury is largely unknown. This review discusses what is known of MK's expression and actions in the developing brain, areas for future research, and the potential for using MK as a therapeutic agent to ameliorate the effects of brain damage caused by insults such as birth-related hypoxia and inflammation.
Collapse
Affiliation(s)
- Emily Ross-Munro
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Faith Kwa
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia.,School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Jenny Kreiner
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Madhavi Khore
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| | - Mary Tolcos
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Bobbi Fleiss
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia.,Neurodiderot, Inserm U1141, Universita de Paris, Paris, France
| | - David W Walker
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| |
Collapse
|
15
|
Cai YQ, Lv Y, Mo ZC, Lei J, Zhu JL, Zhong QQ. Multiple pathophysiological roles of midkine in human disease. Cytokine 2020; 135:155242. [PMID: 32799009 DOI: 10.1016/j.cyto.2020.155242] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/27/2022]
Abstract
Midkine (MK) is a low molecular-weight protein that was first identified as the product of a retinoic acid-responsive gene involved in embryonic development. Recent studies have indicated that MK levels are related to various diseases, including cardiovascular disease (CVD), renal disease and autoimmune disease. MK is a growth factor involved in multiple pathophysiological processes, such as inflammation, the repair of damaged tissues and cancer. The pathophysiological roles of MK are diverse. MK enhances the recruitment and migration of inflammatory cells upon inflammation directly and also through induction of chemokines, and contributes to tissue damage. In lung endothelial cells, oxidative stress increased the expression of MK, which induced angiotensin-converting enzyme (ACE) expression and the consequent conversion from Ang I to Ang II, leading to further oxidative stress. MK inhibited cholesterol efflux from macrophages by reducing ATP-binding cassette transporter A1 (ABCA1) expression, which is involved in lipid metabolism, suggesting that MK is an important positive factor involved in inflammation, oxidative stress and lipid metabolism. Furthermore, MK can regulate the expansion, differentiation and activation of T cells as well as B-cell survival; mediate angiogenic and antibacterial activity; and possess anti-apoptotic activity. In this paper, we summarize the pathophysiological roles of MK in human disease.
Collapse
Affiliation(s)
- Ya-Qin Cai
- Hunan Province Innovative Training Base for Medical Postgraduates, Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, China; Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Yuncheng Lv
- Hunan Province Innovative Training Base for Medical Postgraduates, Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, China; Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Zhong-Cheng Mo
- Hunan Province Innovative Training Base for Medical Postgraduates, Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, China; Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Jiashun Lei
- Hunan Province Innovative Training Base for Medical Postgraduates, Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, China
| | - Jing-Ling Zhu
- Hunan Province Innovative Training Base for Medical Postgraduates, Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, China
| | - Qiao-Qing Zhong
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
16
|
Hayward S, Gachehiladze M, Badr N, Andrijes R, Molostvov G, Paniushkina L, Sopikova B, Slobodová Z, Mgebrishvili G, Sharma N, Horimoto Y, Burg D, Robertson G, Hanby A, Hoar F, Rea D, Eckhardt BL, Ueno NT, Nazarenko I, Long HM, van Laere S, Shaaban AM, Berditchevski F. The CD151-midkine pathway regulates the immune microenvironment in inflammatory breast cancer. J Pathol 2020; 251:63-73. [PMID: 32129471 DOI: 10.1002/path.5415] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/27/2020] [Accepted: 02/25/2020] [Indexed: 12/19/2022]
Abstract
The immune microenvironment in inflammatory breast cancer (IBC) is poorly characterised, and molecular and cellular pathways that control accumulation of various immune cells in IBC tissues remain largely unknown. Here, we discovered a novel pathway linking the expression of the tetraspanin protein CD151 in tumour cells with increased accumulation of macrophages in cancerous tissues. It is notable that elevated expression of CD151 and a higher number of tumour-infiltrating macrophages correlated with better patient responses to chemotherapy. Accordingly, CD151-expressing IBC xenografts were characterised by the increased infiltration of macrophages. In vitro migration experiments demonstrated that CD151 stimulates the chemoattractive potential of IBC cells for monocytes via mechanisms involving midkine (a heparin-binding growth factor), integrin α6β1, and production of extracellular vesicles (EVs). Profiling of chemokines secreted by IBC cells demonstrated that CD151 increases production of midkine. Purified midkine specifically stimulated migration of monocytes, but not other immune cells. Further experiments demonstrated that the chemoattractive potential of IBC-derived EVs is blocked by anti-midkine antibodies. These results demonstrate for the first time that changes in the expression of a tetraspanin protein by tumour cells can affect the formation of the immune microenvironment by modulating recruitment of effector cells to cancerous tissues. Therefore, a CD151-midkine pathway can be considered as a novel target for controlled changes of the immune landscape in IBC. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Steven Hayward
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Mariam Gachehiladze
- Department of Clinical and Molecular Pathology, Palacký Univerzity, Olomouc, Czech Republic
| | - Nahla Badr
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK.,Department of Pathology, Menoufia University School of Medicine, Menoufia, Egypt
| | - Regina Andrijes
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Guerman Molostvov
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Liliia Paniushkina
- Faculty of Medicine, Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Barbora Sopikova
- Department of Clinical and Molecular Pathology, Palacký Univerzity, Olomouc, Czech Republic
| | - Zuzana Slobodová
- Department of Clinical and Molecular Pathology, Palacký Univerzity, Olomouc, Czech Republic
| | - Giorgi Mgebrishvili
- Department of Clinical and Molecular Pathology, Palacký Univerzity, Olomouc, Czech Republic
| | - Nisha Sharma
- Breast Unit, St James Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Yoshiya Horimoto
- Department of Breast Surgical Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | - Andrew Hanby
- University of Leeds, Leeds Institute of Cancer and Pathology (LICAP) Leeds, Leeds, UK
| | - Fiona Hoar
- Hospital, Sandwell and West Birmingham Hospitals, Department of General and Breast Surgery, Birmingham, UK
| | - Daniel Rea
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Bedrich L Eckhardt
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, TX, USA
| | - Irina Nazarenko
- Faculty of Medicine, Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heather M Long
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Steven van Laere
- Translational Cancer Research Unit Center for Oncological Research, University Antwerp, Antwerp, Belgium
| | - Abeer M Shaaban
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| |
Collapse
|
17
|
Abstract
Pleiotrophin (PTN) is a potent mitogenic cytokine with a high affinity for the polysaccharide glycosaminoglycan (GAG). Although it is most strongly associated with neural development during embryogenesis and the neonatal period, its expression has also been linked to a plethora of other physiological events including cancer metastasis, angiogenesis, bone development, and inflammation. A considerable amount of research has been carried out to understand the mechanisms by which PTN regulates these events. In particular, PTN has now been shown to bind a diverse collection of receptors including many GAG-containing proteoglycans. These interactions lead to the activation of many intracellular kinases and, ultimately, activation and transformation of cells. Structural studies of PTN in complex with both GAG and domains from its non-proteoglycan receptors reveal a binding mechanism that relies on electrostatic interactions and points to PTN-induced receptor oligomerization as one of the possible ways PTN uses to control cellular functions.
Collapse
|
18
|
Midkine (MDK) growth factor: a key player in cancer progression and a promising therapeutic target. Oncogene 2019; 39:2040-2054. [PMID: 31801970 DOI: 10.1038/s41388-019-1124-8] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/15/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022]
Abstract
Midkine is a heparin-binding growth factor, originally reported as the product of a retinoic acid-responsive gene during embryogenesis, but currently viewed as a multifaceted factor contributing to both normal tissue homeostasis and disease development. Midkine is abnormally expressed at high levels in various human malignancies and acts as a mediator for the acquisition of critical hallmarks of cancer, including cell growth, survival, metastasis, migration, and angiogenesis. Several studies have investigated the role of midkine as a cancer biomarker for the detection, prognosis, and management of cancer, as well as for monitoring the response to cancer treatment. Moreover, several efforts are also being made to elucidate its underlying mechanisms in therapeutic resistance and immunomodulation within the tumor microenvironment. We hereby summarize the current knowledge on midkine expression and function in cancer development and progression, and highlight its promising potential as a cancer biomarker and as a future therapeutic target in personalized cancer medicine.
Collapse
|
19
|
Karadeniz Z, Aynacıoğlu AŞ, Bilir A, Tuna MY. Inhibition of midkine by metformin can contribute to its anticancer effects in malignancies: A proposal mechanism of action of metformin in context of endometrial cancer prevention and therapy. Med Hypotheses 2019; 134:109420. [PMID: 31634770 DOI: 10.1016/j.mehy.2019.109420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/20/2019] [Accepted: 09/30/2019] [Indexed: 01/10/2023]
Abstract
Metformin, a drug widely used in the treatment of type II diabetes mellitus (T2DM), has been the focus of interest as a potential therapeutic agent for certain types of malignancies, including gynaecological cancers [i.e. endometrial cancer (EC)]. Although the exact mechanism behind the potential anticancer activity of metformin is still not completely understood, certain studies have suggested that different effects on cell functions, such as inhibition of cell migration, apoptosis and tumor cell proliferation, are involved in its preventive and therapeutic effects in certain types of malignancies, including EC. In contrast, midkine (MK), a heparin-binding growth factor and cytokine, which induces carcinogenesis and chemoresistance, promotes the development and progression of many malignant tumours by increasing diverse cell functions such as cell proliferation, cell survival and antiapoptotic activities via mainly the activation of phosphatidyl inositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways. The same pathways are also subject to certain therapeutic effects of metformin, although this cytokine and this drug have some different mechanism of action pathways as well. Taken together, MK and metformin appear to have opposite effects in various biological processes such as apoptosis, cell proliferation, cell survival, cell migration, and angiogenesis. On the other hand, MK activates PI3K and MAPK cell signal pathways, whereas metformin inhibits these two pathways. It seems likely that almost all the pathways and cell functions, which play important roles in malignancies, are inhibited by metformin and activated by MK. Given the opposite relationship between the actions of metformin and MK, we hypothesize that metformin may act like a novel MK inhibitor in some malignancies. We also discuss the possible relationship between metformin and MK in the context of EC, the most common gynecological cancer worldwide, which incidence is rising rapidly, in parallel with the increase in obesity, T2DM and insulin resistance. In this respect, the therapeutic use of metformin may improve the survival of EC or other cancers, via inhibiting or overcoming the unwanted effects of MK in carcinogenesis.
Collapse
Affiliation(s)
- Zeliha Karadeniz
- Department of Gynecology and Obstetrics, Istanbul Aydin University, Medical Faculty, Florya Main Campus, Kücükcekmece, 34295 Istanbul, Turkey
| | - A Şükrü Aynacıoğlu
- Department of Medical Pharmacology, Istanbul Aydin University, Medical Faculty, Florya Main Campus, Kücükcekmece, 34295 Istanbul, Turkey.
| | - Ayhan Bilir
- Department of Histology and Embryology, Istanbul Aydin University, Medical Faculty, Florya Main Campus, Kücükcekmece, 34295 Istanbul, Turkey
| | - M Yakup Tuna
- Department of Anatomy, Istanbul Aydin University, Medical Faculty, Florya Main Campus, Kücükcekmece, 34295 Istanbul, Turkey
| |
Collapse
|
20
|
Supsrisunjai C, Hsu CK, Michael M, Duval C, Lee JYW, Yang HS, Huang HY, Chaikul T, Onoufriadis A, Steiner RA, Ariëns RAS, Sarig O, Sprecher E, Eskin-Schwartz M, Samlaska C, Simpson MA, Calonje E, Parsons M, McGrath JA. Coagulation Factor XIII-A Subunit Missense Mutation in the Pathobiology of Autosomal Dominant Multiple Dermatofibromas. J Invest Dermatol 2019; 140:624-635.e7. [PMID: 31493396 DOI: 10.1016/j.jid.2019.08.441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/26/2019] [Accepted: 08/06/2019] [Indexed: 01/16/2023]
Abstract
Dermatofibromas are common benign skin lesions, the etiology of which is poorly understood. We identified two unrelated pedigrees in which there was autosomal dominant transmission of multiple dermatofibromas. Whole exome sequencing revealed a rare shared heterozygous missense variant in the F13A1 gene encoding factor XIII subunit A (FXIII-A), a transglutaminase involved in hemostasis, wound healing, tumor growth, and apoptosis. The variant (p.Lys679Met) has an allele frequency of 0.0002 and is predicted to be a damaging mutation. Recombinant human Lys679Met FXIII-A demonstrated reduced fibrin crosslinking activity in vitro. Of note, the treatment of fibroblasts with media containing Lys679Met FXIII-A led to enhanced adhesion, proliferation, and type I collagen synthesis. Immunostaining revealed co-localization between FXIII-A and α4β1 integrins, more prominently for Lys679Met FXIII-A than the wild type. In addition, both the α4β1 inhibitors and the mutation of the FXIII-A Isoleucine-Leucine-Aspartate-Threonine (ILDT) motif prevented Lys679Met FXIII-A-dependent proliferation and collagen synthesis of fibroblasts. Our data suggest that the Lys679Met mutation may lead to a conformational change in the FXIII-A protein that enhances α4-integrin binding and provides insight into an unexpected role for FXIII-A in the pathobiology of familial dermatofibroma.
Collapse
Affiliation(s)
- Chavalit Supsrisunjai
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom; Institute of Dermatology, Ministry of Public Health, Bangkok, Thailand
| | - Chao-Kai Hsu
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom; Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Magdalene Michael
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, United Kingdom
| | - Cédric Duval
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - John Y W Lee
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Hsing-San Yang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsin-Yu Huang
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Thitiwat Chaikul
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Alexandros Onoufriadis
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Roberto A Steiner
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, United Kingdom
| | - Robert A S Ariëns
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Ofer Sarig
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eli Sprecher
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Curt Samlaska
- Academic Dermatology of Nevada, University of Nevada School of Medicine, Reno, Nevada
| | - Michael A Simpson
- Department of Genetics, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Eduardo Calonje
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom; Department of Dermatopathology, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, United Kingdom
| | - John A McGrath
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom.
| |
Collapse
|
21
|
Jiménez-González V, Ogalla-García E, García-Quintanilla M, García-Quintanilla A. Deciphering GRINA/Lifeguard1: Nuclear Location, Ca 2+ Homeostasis and Vesicle Transport. Int J Mol Sci 2019; 20:ijms20164005. [PMID: 31426446 PMCID: PMC6719933 DOI: 10.3390/ijms20164005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/31/2019] [Accepted: 08/12/2019] [Indexed: 01/31/2023] Open
Abstract
The Glutamate Receptor Ionotropic NMDA-Associated Protein 1 (GRINA) belongs to the Lifeguard family and is involved in calcium homeostasis, which governs key processes, such as cell survival or the release of neurotransmitters. GRINA is mainly associated with membranes of the endoplasmic reticulum, Golgi, endosome, and the cell surface, but its presence in the nucleus has not been explained yet. Here we dissect, with the help of different software tools, the potential roles of GRINA in the cell and how they may be altered in diseases, such as schizophrenia or celiac disease. We describe for the first time that the cytoplasmic N-terminal half of GRINA (which spans a Proline-rich domain) contains a potential DNA-binding sequence, in addition to cleavage target sites and probable PY-nuclear localization sequences, that may enable it to be released from the rest of the protein and enter the nucleus under suitable conditions, where it could participate in the transcription, alternative splicing, and mRNA export of a subset of genes likely involved in lipid and sterol synthesis, ribosome biogenesis, or cell cycle progression. To support these findings, we include additional evidence based on an exhaustive review of the literature and our preliminary data of the protein–protein interaction network of GRINA.
Collapse
Affiliation(s)
| | - Elena Ogalla-García
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012 Seville, Spain
| | - Meritxell García-Quintanilla
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Albert García-Quintanilla
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, 41012 Seville, Spain.
| |
Collapse
|
22
|
Tanga N, Kuboyama K, Kishimoto A, Kiyonari H, Shiraishi A, Suzuki R, Watanabe T, Fujikawa A, Noda M. The PTN-PTPRZ signal activates the AFAP1L2-dependent PI3K-AKT pathway for oligodendrocyte differentiation: Targeted inactivation of PTPRZ activity in mice. Glia 2019; 67:967-984. [PMID: 30667096 DOI: 10.1002/glia.23583] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022]
Abstract
Protein tyrosine phosphatase receptor type Z (PTPRZ) maintains oligodendrocyte precursor cells (OPCs) in an undifferentiated state. The inhibition of PTPase by its ligand pleiotrophin (PTN) promotes OPC differentiation; however, the substrate molecules of PTPRZ involved in the differentiation have not yet been elucidated in detail. We herein demonstrated that the tyrosine phosphorylation of AFAP1L2, paxillin, ERBB4, GIT1, p190RhoGAP, and NYAP2 was enhanced in OPC-like OL1 cells by a treatment with PTN. AFAP1L2, an adaptor protein involved in the PI3K-AKT pathway, exhibited the strongest response to PTN. PTPRZ dephosphorylated AFAP1L2 at tyrosine residues in vitro and in HEK293T cells. In OL1 cells, the knockdown of AFAP1L2 or application of a PI3K inhibitor suppressed cell differentiation as well as the PTN-induced phosphorylation of AKT and mTOR. We generated a knock-in mouse harboring a catalytically inactive Cys to Ser (CS) mutation in the PTPase domain. The phosphorylation levels of AFAP1L2, AKT, and mTOR were higher, and the expression of oligodendrocyte markers, including myelin basic protein (MBP) and myelin regulatory factor (MYRF), was stronger in CS knock-in brains than in wild-type brains on postnatal day 10; however, these differences mostly disappeared in the adult stage. Adult CS knock-in mice exhibited earlier remyelination after cuprizone-induced demyelination through the accelerated differentiation of OPCs. These phenotypes in CS knock-in mice were similar to those in Ptprz-deficient mice. Therefore, we conclude that the PTN-PTPRZ signal stimulates OPC differentiation partly by enhancing the tyrosine phosphorylation of AFAP1L2 in order to activate the PI3K-AKT pathway.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Carrier Proteins/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cuprizone/toxicity
- Cytokines/metabolism
- Demyelinating Diseases/chemically induced
- Demyelinating Diseases/diagnostic imaging
- Disease Models, Animal
- HEK293 Cells
- Humans
- Immunoprecipitation
- In Situ Nick-End Labeling
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microfilament Proteins/metabolism
- Myelin Proteins/metabolism
- Oligodendroglia/physiology
- Proto-Oncogene Proteins c-akt
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism
- Signal Detection, Psychological/drug effects
- Signal Detection, Psychological/physiology
- Signal Transduction/physiology
- Transfection
- X-Ray Microtomography
- Red Fluorescent Protein
Collapse
Affiliation(s)
- Naomi Tanga
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
- School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
| | - Kazuya Kuboyama
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
| | - Ayako Kishimoto
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Nara, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resource Development, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Aki Shiraishi
- Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Ryoko Suzuki
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
| | - Toshio Watanabe
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Nara, Japan
| | - Akihiro Fujikawa
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
- School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
- Research Center for Cell Biology, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
23
|
Herradon G, Ramos-Alvarez MP, Gramage E. Connecting Metainflammation and Neuroinflammation Through the PTN-MK-RPTPβ/ζ Axis: Relevance in Therapeutic Development. Front Pharmacol 2019; 10:377. [PMID: 31031625 PMCID: PMC6474308 DOI: 10.3389/fphar.2019.00377] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/26/2019] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a common factor of pathologies such as obesity, type 2 diabetes or neurodegenerative diseases. Chronic inflammation is considered part of the pathogenic mechanisms of different disorders associated with aging. Interestingly, peripheral inflammation and the associated metabolic alterations not only facilitate insulin resistance and diabetes but also neurodegenerative disorders. Therefore, the identification of novel pathways, common to the development of these diseases, which modulate the immune response and signaling is key. It will provide highly relevant information to advance our knowledge of the multifactorial process of aging, and to establish new biomarkers and/or therapeutic targets to counteract the underlying chronic inflammatory processes. One novel pathway that regulates peripheral and central immune responses is triggered by the cytokines pleiotrophin (PTN) and midkine (MK), which bind its receptor, Receptor Protein Tyrosine Phosphatase (RPTP) β/ζ, and inactivate its phosphatase activity. In this review, we compile a growing body of knowledge suggesting that PTN and MK modulate the immune response and/or inflammation in different pathologies characterized by peripheral inflammation associated with insulin resistance, such as aging, and in central disorders characterized by overt neuroinflammation, such as neurodegenerative diseases and endotoxemia. Evidence strongly suggests that regulation of the PTN and MK signaling pathways may provide new therapeutic opportunities particularly in those neurological disorders characterized by increased PTN and/or MK cerebral levels and neuroinflammation. Importantly, we discuss existing therapeutics, and others being developed, that modulate these signaling pathways, and their potential use in pathologies characterized by overt neuroinflammation.
Collapse
Affiliation(s)
- Gonzalo Herradon
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - M Pilar Ramos-Alvarez
- Departmento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Esther Gramage
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
24
|
Regulation of cell migration by α4 and α9 integrins. Biochem J 2019; 476:705-718. [PMID: 30819933 DOI: 10.1042/bcj20180415] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/09/2019] [Accepted: 02/12/2019] [Indexed: 12/15/2022]
Abstract
Integrins are heterodimeric transmembrane receptors that play an essential role in enabling cells to sense and bind to extracellular ligands. Activation and clustering of integrins leads to the formation of focal adhesions at the plasma membrane that subsequently initiate signalling pathways to control a broad range of functional endpoints including cell migration, proliferation and survival. The α4 and α9 integrins form a small sub-family of receptors that share some specific ligands and binding partners. Although relatively poorly studied compared with other integrin family members, emerging evidence suggests that despite restricted cell and tissue expression profiles, these integrins play a key role in the regulation of signalling pathways controlling cytoskeletal remodelling and migration in both adherent and non-adherent cell types. This review summarises the known shared and specific roles for α4 and α9 integrins and highlights the importance of these receptors in controlling cell migration within both homeostatic and disease settings.
Collapse
|
25
|
Cui R, Lwigale P. Expression of the heparin-binding growth factors Midkine and pleiotrophin during ocular development. Gene Expr Patterns 2019; 32:28-37. [PMID: 30825522 DOI: 10.1016/j.gep.2019.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/18/2019] [Accepted: 02/21/2019] [Indexed: 12/17/2022]
Abstract
Midkine (MDK) and Pleiotrophin (PTN) belong to a group of heparin-binding growth factors that has been shown to have pleiotropic functions in various biological processes during development and disease. Development of the vertebrate eye is a multistep process that involves coordinated interactions between neuronal and non-neuronal cells, but very little is known about the potential function of MDK and PTN in these processes. In this study, we demonstrate by section in situ hybridization, the spatiotemporal expression of MDK and PTN during ocular development in chick and mouse. We show that MDK and PTN are expressed in dynamic patterns that overlap in a few non-neuronal tissues in the anterior eye and in neuronal cell layers of the posterior eye. We show that the expression patterns of MDK and PTN are only conserved in a few tissues in chick and mouse but they overlap with the expression of some of their receptors LRP1, RPTPZ, ALK, NOTCH2, ITGβ1, SDC1, and SDC3. The dynamic expression patterns of MDK, PTN and their receptors suggest that they function together during the multistep process of ocular development and they may play important roles in cell proliferation, adhesion, and migration of neuronal and non-neuronal cells.
Collapse
Affiliation(s)
- Ruda Cui
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Peter Lwigale
- Department of BioSciences, Rice University, Houston, TX, USA.
| |
Collapse
|
26
|
Aynacıoğlu AŞ, Bilir A, Tuna MY. Involvement of midkine in autoimmune and autoinflammatory diseases. Mod Rheumatol 2018; 29:567-571. [DOI: 10.1080/14397595.2018.1523701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Ahmet Şükrü Aynacıoğlu
- Department of Medical Pharmacology, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| | - Ayhan Bilir
- Department of Histology and Embryology, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| | - Mehmet Yakup Tuna
- Department of Anatomy, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| |
Collapse
|
27
|
Weckbach LT, Preissner KT, Deindl E. The Role of Midkine in Arteriogenesis, Involving Mechanosensing, Endothelial Cell Proliferation, and Vasodilation. Int J Mol Sci 2018; 19:E2559. [PMID: 30158425 PMCID: PMC6163309 DOI: 10.3390/ijms19092559] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
Mechanical forces in blood circulation such as shear stress play a predominant role in many physiological and pathophysiological processes related to vascular responses or vessel remodeling. Arteriogenesis, defined as the growth of pre-existing arterioles into functional collateral arteries compensating for stenosed or occluded arteries, is such a process. Midkine, a pleiotropic protein and growth factor, has originally been identified to orchestrate embryonic development. In the adult organism its expression is restricted to distinct tissues (including tumors), whereby midkine is strongly expressed in inflamed tissue and has been shown to promote inflammation. Recent investigations conferred midkine an important function in vascular remodeling and growth. In this review, we introduce the midkine gene and protein along with its cognate receptors, and highlight its role in inflammation and the vascular system with special emphasis on arteriogenesis, particularly focusing on shear stress-mediated vascular cell proliferation and vasodilatation.
Collapse
Affiliation(s)
- Ludwig T Weckbach
- Medizinische Klinik und Poliklinik I, Klinikum der Universität, LMU Munich, 81377 Munich, Germany.
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich, 82152 Planegg-Martinsried, Germany.
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig-University, 35390 Giessen, Germany.
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
28
|
Shen D, Podolnikova NP, Yakubenko VP, Ardell CL, Balabiyev A, Ugarova TP, Wang X. Pleiotrophin, a multifunctional cytokine and growth factor, induces leukocyte responses through the integrin Mac-1. J Biol Chem 2017; 292:18848-18861. [PMID: 28939773 DOI: 10.1074/jbc.m116.773713] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 09/08/2017] [Indexed: 12/27/2022] Open
Abstract
Pleiotrophin (PTN) is a multifunctional, cationic, glycosaminoglycan-binding cytokine and growth factor involved in numerous physiological and pathological processes, including tissue repair and inflammation-related diseases. PTN has been shown to promote leukocyte responses by inducing their migration and expression of inflammatory cytokines. However, the mechanisms through which PTN mediates these responses remain unclear. Here, we identified the integrin Mac-1 (αMβ2, CD11b/CD18) as the receptor mediating macrophage adhesion and migration to PTN. We also found that expression of Mac-1 on the surface of human embryonic kidney (HEK) 293 cells induced their adhesion and migration to PTN. Accordingly, PTN promoted Mac-1-dependent cell spreading and initiated intracellular signaling manifested in phosphorylation of Erk1/2. While binding to PTN, Mac-1 on Mac-1-expressing HEK293 cells appears to cooperate with cell-surface proteoglycans because both anti-Mac-1 function-blocking mAb and heparin were required to block adhesion. Moreover, biolayer interferometry and NMR indicated a direct interaction between the αMI domain, the major ligand-binding region of Mac-1, and PTN. Using peptide libraries, we found that in PTN the αMI domain bound sequences enriched in basic and hydrophobic residues, indicating that PTN conforms to the general principle of ligand-recognition specificity of the αMI domain toward cationic proteins/peptides. Finally, using recombinant PTN-derived fragments, we show that PTN contains two distinct Mac-1-binding sites in each of its constitutive domains. Collectively, these results identify PTN as a ligand for the integrin Mac-1 on the surface of leukocytes and suggest that this interaction may play a role in inflammatory responses.
Collapse
Affiliation(s)
- Di Shen
- From the Schools of Molecular and
| | | | - Valentin P Yakubenko
- Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Christopher L Ardell
- Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Arnat Balabiyev
- Life Sciences, Arizona State University, Tempe, Arizona 85287 and
| | | | - Xu Wang
- From the Schools of Molecular and
| |
Collapse
|
29
|
Inhibition of Midkine Suppresses Prostate Cancer CD133 + Stem Cell Growth and Migration. Am J Med Sci 2017; 354:299-309. [DOI: 10.1016/j.amjms.2017.04.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 12/22/2022]
|
30
|
Abstract
Midkine (MDK) is a heparin-binding growth factor that is normally expressed in mid-gestational development mediating mesenchymal and epithelial interactions. As organisms age, expression of MDK diminishes; however, in adults, MDK expression is associated with acute and chronic pathologic conditions such as myocardial infarction and heart failure (HF). The role of MDK is not clear in cardiovascular disease and currently there is no consensus if it plays a beneficial or detrimental role in HF. The lack of clarity in the literature is exacerbated by differing roles that circulating and myocardial MDK play in signaling pathways in cardiomyocytes (some of which have yet to be elucidated). Of particular interest, serum MDK is elevated in adults with chronic heart failure and higher circulating MDK is associated with worse cardiac function. In addition, pediatric HF patients have higher levels of myocardial MDK. This review focuses on what is known about the effect of exogenous versus myocardial MDK in various cardiac disease models in an effort to better clarify the role of midkine in HF.
Collapse
|
31
|
Sorrelle N, Dominguez ATA, Brekken RA. From top to bottom: midkine and pleiotrophin as emerging players in immune regulation. J Leukoc Biol 2017; 102:277-286. [PMID: 28356350 DOI: 10.1189/jlb.3mr1116-475r] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/02/2017] [Accepted: 03/06/2017] [Indexed: 01/15/2023] Open
Abstract
Cytokines are pivotal in the generation and resolution of the inflammatory response. The midkine/pleiotrophin (MK/PTN) family of cytokines, composed of just two members, was discovered as heparin-binding neurite outgrowth-promoting factors. Since their discovery, expression of this cytokine family has been reported in a wide array of inflammatory diseases and cancer. In this minireview, we will discuss the emerging appreciation of the functions of the MK/PTN family in the immune system, which include promoting lymphocyte survival, sculpting myeloid cell phenotype, driving immune cell chemotaxis, and maintaining hematopoiesis.
Collapse
Affiliation(s)
- Noah Sorrelle
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA; and
| | - Adrian T A Dominguez
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA; and
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA; and .,Division of Surgical Oncology, Departments of Surgery and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
32
|
Dik B, Baş AL, Yazıhan N. The effect of midkine on growth factors and oxidative status in an experimental wound model in diabetic and healthy rats. Can J Physiol Pharmacol 2017; 95:604-609. [PMID: 28177680 DOI: 10.1139/cjpp-2016-0439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Wound healing is important for longevity. Midkine is a cytokine involved in controlling tissue repair and new tissue development, and in regulating inflammation. We investigated the effect of midkine on wound healing in rats. In total, 108 Wistar albino rats were used: 12 as healthy and diabetic controls; 96 were split into 4 groups: healthy, saline treated; healthy, midkine (10 ng/kg, 48 h intervals) treated; diabetic, saline treated; and diabetic, midkine treated. Following wound creation, 6 rats per group were euthanized on days 3, 7, 14, and 28; the wounded skin was removed. Levels of epidermal growth factor (EGF), matrix metalloproteinase-8 (MMP-8), transforming growth factor beta (TGF-β), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and thiobarbituric acid reactive substances (TBARS) were measured. MMP-8 and PDGF levels fluctuated in all groups; TGF-β fluctuated in the diabetic groups and was significantly higher in the HM group than other groups after 14 days. EGF and VEGF levels were increased in the HM group after 3 days. TBARS levels were highest in the diabetic groups. Macroscopically, the midkine-treated groups healed better. Midkine can accelerate wound healing by influencing growth factors and oxidative status in wound tissues.
Collapse
Affiliation(s)
- Burak Dik
- a Department of Pharmacology and Toxicology, Veterinary Faculty, University of Selcuk, Konya, Turkey
| | - Ahmet Levent Baş
- a Department of Pharmacology and Toxicology, Veterinary Faculty, University of Selcuk, Konya, Turkey
| | - Nuray Yazıhan
- b Department of Pathophysiology, Medicine Faculty, University of Ankara, Ankara, Turkey
| |
Collapse
|
33
|
Haffner‐Luntzer M, Heilmann A, Rapp AE, Roessler R, Schinke T, Amling M, Ignatius A, Liedert A. Antagonizing midkine accelerates fracture healing in mice by enhanced bone formation in the fracture callus. Br J Pharmacol 2016; 173:2237-49. [PMID: 27111560 PMCID: PMC4919577 DOI: 10.1111/bph.13503] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 03/15/2016] [Accepted: 04/18/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Previous findings suggest that the growth and differentiation factor midkine (Mdk) is a negative regulator of osteoblast activity and bone formation, thereby raising the possibility that a specific Mdk antagonist might improve bone formation during fracture healing. EXPERIMENTAL APPROACH In the present study, we investigated the effects of a monoclonal anti-Mdk antibody (Mdk-Ab) on bone healing using a standardized femur osteotomy model in mice. Additional in vitro experiments using chondroprogenitor and preosteoblastic cells were conducted to analyse the effects of recombinant Mdk and Mdk-Ab on differentiation markers and potential binding partners in these cells. KEY RESULTS We demonstrated that treatment with Mdk-Ab accelerated bone healing in mice based on increased bone formation in the fracture callus. In vitro experiments using preosteoblastic cells showed that Mdk-Ab treatment abolished the Mdk-induced negative effects on the expression of osteogenic markers and Wnt/β-catenin target proteins, whereas the differentiation of chondroprogenitor cells was unaffected. Phosphorylation analyses revealed an important role for the low-density lipoproteinLDL receptor-related protein 6 in Mdk signalling in osteoblasts. CONCLUSIONS AND IMPLICATIONS We conclude that Mdk-Ab treatment may be a potential novel therapeutic strategy to enhance fracture healing in patients with orthopaedic complications such as delayed healing or non-union formation.
Collapse
Affiliation(s)
| | - Aline Heilmann
- Institute of Orthopedic Research and BiomechanicsUniversity Medical Center UlmUlmGermany
| | - Anna Elise Rapp
- Institute of Orthopedic Research and BiomechanicsUniversity Medical Center UlmUlmGermany
| | - Robin Roessler
- Institute of Orthopedic Research and BiomechanicsUniversity Medical Center UlmUlmGermany
| | - Thorsten Schinke
- Institute of Osteology and BiomechanicsUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Michael Amling
- Institute of Osteology and BiomechanicsUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Anita Ignatius
- Institute of Orthopedic Research and BiomechanicsUniversity Medical Center UlmUlmGermany
| | - Astrid Liedert
- Institute of Orthopedic Research and BiomechanicsUniversity Medical Center UlmUlmGermany
| |
Collapse
|
34
|
Şalaru DL, Arsenescu-Georgescu C, Chatzikyrkou C, Karagiannis J, Fischer A, Mertens PR. Midkine, a heparin-binding growth factor, and its roles in atherogenesis and inflammatory kidney diseases. Nephrol Dial Transplant 2016; 31:1781-1787. [DOI: 10.1093/ndt/gfw083] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 03/04/2016] [Indexed: 01/07/2023] Open
|
35
|
Jones DR. Measuring midkine: the utility of midkine as a biomarker in cancer and other diseases. Br J Pharmacol 2015; 171:2925-39. [PMID: 24460734 DOI: 10.1111/bph.12601] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/15/2013] [Accepted: 01/17/2014] [Indexed: 01/05/2023] Open
Abstract
Midkine (MK) is a pleiotropic growth factor prominently expressed during embryogenesis but down-regulated to neglible levels in healthy adults. Many published studies have demonstrated striking MK overexpression compared with healthy controls in various pathologies, including ischaemia, inflammation, autoimmunity and, most notably, in many cancers. MK expression is detectable in biopsies of diseased, but not healthy, tissues. Significantly, because it is a soluble cytokine, elevated MK is readily apparent in the blood and other body fluids such as urine and CSF, making MK a relatively convenient, accessible, non-invasive and inexpensive biomarker for population screening and early disease detection. The first diagnostic tests that quantify MK are just now receiving regulatory clearance and entering the clinic. This review examines the current state of knowledge pertaining to MK as a biomarker and highlights promising indications and clinical settings where measuring MK could make a difference to patient treatment. I also raise outstanding questions about reported variants of MK as well as MK's bio-distribution in vivo. Answering these questions in future studies will enhance our understanding of the significance of measured MK levels in both patients and healthy subjects, and may reveal further opportunities for measuring MK to diagnose disease. MK has already proven to be a biomarker that can significantly improve detection, management and treatment of cancer, and there is significant promise for developing further MK-based diagnostics in the future.
Collapse
Affiliation(s)
- D R Jones
- Cellmid Ltd., Sydney, NSW, Australia
| |
Collapse
|
36
|
Hugosson F, Sjögren C, Birve A, Hedlund L, Eriksson T, Palmer RH. The Drosophila midkine/pleiotrophin homologues Miple1 and Miple2 affect adult lifespan but are dispensable for alk signaling during embryonic gut formation. PLoS One 2014; 9:e112250. [PMID: 25380037 PMCID: PMC4224452 DOI: 10.1371/journal.pone.0112250] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/10/2014] [Indexed: 01/07/2023] Open
Abstract
Midkine (MDK) and Pleiotrophin (PTN) are small heparin-binding cytokines with closely related structures. The Drosophila genome harbours two genes encoding members of the MDK/PTN family of proteins, known as miple1 and miple2. We have investigated the role of Miple proteins in vivo, in particular with regard to their proposed role as ligands for the Alk receptor tyrosine kinase (RTK). Here we show that Miple proteins are neither required to drive Alk signaling during Drosophila embryogenesis, nor are they essential for development in the fruit fly. Additionally we show that neither MDK nor PTN can activate hALK in vivo when ectopically co-expressed in the fly. In conclusion, our data suggest that Alk is not activated by MDK/PTN related growth factors Miple1 and Miple 2 in vivo.
Collapse
Affiliation(s)
| | - Camilla Sjögren
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Anna Birve
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | | | - Ruth H. Palmer
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Göteborg, Sweden
- * E-mail:
| |
Collapse
|
37
|
Güngör C, Hofmann BT, Wolters-Eisfeld G, Bockhorn M. Pancreatic cancer. Br J Pharmacol 2014; 171:849-58. [PMID: 24024905 DOI: 10.1111/bph.12401] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/16/2013] [Accepted: 09/03/2013] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED In recent years, it has become clear that the current standard therapeutic options for pancreatic cancer are not adequate and still do not meet the criteria to cure patients suffering from this lethal disease. Although research over the past decade has shown very interesting and promising new therapeutic options for these patients, only minor clinical success was achieved. Therefore, there is still an urgent need for new approaches that deal with early detection and new therapeutic options in pancreatic cancer. To provide optimal care for patients with pancreatic cancer, we need to understand better its complex molecular biology and thus to identify new target molecules that promote the proliferation and resistance to chemotherapy of pancreatic cancer cells. In spite of significant progress in curing cancers with chemotherapy, pancreatic cancer remains one of the most resistant solid tumour cancers and many studies suggest that drug-resistant cancer cells are the most aggressive with the highest relapse and metastatic rates. In this context, activated Notch signalling is strongly linked with chemoresistance and therefore reflects a rational new target to circumvent resistance to chemotherapy in pancreatic cancer. Here, we have focused our discussion on the latest research, current therapy options and recently identified target molecules such as Notch-2 and the heparin-binding growth factor midkine, which exhibit a wide range of cancer-relevant functions and therefore provide attractive new therapeutic target molecules, in terms of pancreatic cancer and other cancers also. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- C Güngör
- Department of General, Visceral and Thoracic Surgery, Experimental Oncology, Campus Research, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | |
Collapse
|
38
|
Winkler C, Yao S. The midkine family of growth factors: diverse roles in nervous system formation and maintenance. Br J Pharmacol 2014; 171:905-12. [PMID: 24125182 DOI: 10.1111/bph.12462] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 09/24/2013] [Accepted: 10/07/2013] [Indexed: 02/04/2023] Open
Abstract
UNLABELLED Midkines are heparin-binding growth factors involved in a wide range of biological processes. Originally identified as retinoic acid inducible genes, midkines are widely expressed during embryogenesis with particularly high levels in the developing nervous system. During postnatal stages, midkine expression generally ceases but is often up-regulated under disease conditions, most notably those affecting the nervous system. Midkines are known as neurotrophic factors, as they promote neurite outgrowth and neuron survival in cell culture. Surprisingly, however, mouse embryos deficient for midkine (knockout mice) are phenotypically normal, which suggests functional redundancy by related growth factors. During adult stages, on the other hand, midkine knockout mice develop striking deficits in neuroprotection and regeneration after drug-induced neurotoxicity and injury. The detailed mechanisms by which midkine controls neuron formation, differentiation and maintenance remain unclear. Recent studies in zebrafish and chick have provided important insight into the role of midkine and its putative receptor, anaplastic lymphoma kinase, in cell cycle control in the central and peripheral nervous systems. A recent structural analysis of zebrafish midkine furthermore revealed essential protein domains required for biological activity that serve as promising novel targets for future drug designs. This review will summarize latest findings in the field that help to better understand the diverse roles of midkine in nervous system formation and maintenance. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- C Winkler
- Department of Biological Sciences and Centre for BioImaging Sciences, National University of Singapore, Singapore
| | | |
Collapse
|
39
|
Kishida S, Kadomatsu K. Involvement of midkine in neuroblastoma tumourigenesis. Br J Pharmacol 2014; 171:896-904. [PMID: 24116381 DOI: 10.1111/bph.12442] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/09/2013] [Accepted: 09/16/2013] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Midkine is highly expressed in various cancers, including neuroblastoma, one of the most malignant paediatric solid tumours known. Also, it has been shown to be useful as a tumour marker, a prognosis factor and a target of molecular therapy. Several molecular tools (e.g. siRNA, antibodies and RNA aptamer) have been used to establish a midkine-targeted therapy. The involvement of midkine in tumourigenesis has been demonstrated in vivo in a mouse neuroblastoma model, where targeting it with an RNA aptamer was shown to be an effective treatment for xenografted tumours. Chemoresistance is one of the notable phenotypes regulated by midkine in various cancer cell types. In pancreatic tumours and glioma cells, midkine is expressed in chemoresistant cells and is involved in the survival of these cells in the presence of anticancer drugs. In contrast to these tumours, midkine was found to be expressed in every neuroblastoma cell line tested and the knockdown of midkine alone was sufficient to suppress their growth. These results indicate that neuroblastoma cells are highly dependent on midkine and that a midkine-targeted therapy could exert a significant effect in these cells. However, to achieve a midkine-targeted therapy for high-risk neuroblastoma patients, the further refinement of the RNA aptamer or antibody as tools and the elucidation of midkine signalling are immediate issues that need to be resolved. Regarding the latter, although it has been shown that Notch2 functions as a receptor in neuroblastoma cells, it is likely that other receptors (e.g. anaplastic lymphoma kinase) are also involved in midkine signalling. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- S Kishida
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | |
Collapse
|
40
|
Gramage E, Li J, Hitchcock P. The expression and function of midkine in the vertebrate retina. Br J Pharmacol 2014; 171:913-23. [PMID: 24460673 PMCID: PMC3925030 DOI: 10.1111/bph.12495] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 10/21/2013] [Accepted: 10/24/2013] [Indexed: 12/20/2022] Open
Abstract
The functional role of midkine during development, following injury and in disease has been studied in a variety of tissues. In this review, we summarize what is known about midkine in the vertebrate retina, focusing largely on recent studies utilizing the zebrafish (Danio rerio) as an animal model. Zebrafish are a valuable animal model for studying the retina, due to its very rapid development and amazing ability for functional neuronal regeneration following neuronal cell death. The zebrafish genome harbours two midkine paralogues, midkine-a (mdka) and midkine-b (mdkb), which, during development, are expressed in nested patterns among different cell types. mdka is expressed in the retinal progenitors and mdkb is expressed in newly post-mitotic cells. Interestingly, studies of loss-and gain-of-function in zebrafish larvae indicate that midkine-a regulates cell cycle kinetics. Moreover, both mdka and mdkb are expressed in different cell types in the normal adult zebrafish retina, but after light-induced death of photoreceptors, both are up-regulated and expressed in proliferating Müller glia and photoreceptor progenitors, suggesting an important and (perhaps) coincident role for these cytokines during stem cell-based neuronal regeneration. Based on its known role in other tissues and the expression and function of the midkine paralogues in the zebrafish retina, we propose that midkine has an important functional role both during development and regeneration in the retina. Further studies are needed to understand this role and the mechanisms that underlie it.
Collapse
Affiliation(s)
- E Gramage
- Department of Ophthalmology and Visual Sciences, W. K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | | | | |
Collapse
|
41
|
Ikutomo M, Sakakima H, Matsuda F, Yoshida Y. Midkine-deficient mice delayed degeneration and regeneration after skeletal muscle injury. Acta Histochem 2014; 116:319-26. [PMID: 24055194 DOI: 10.1016/j.acthis.2013.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/13/2013] [Accepted: 08/14/2013] [Indexed: 11/28/2022]
Abstract
Midkine (MK), a heparin-binding growth factor, was previously found to be expressed in the rat myotube-forming stage. We investigated MK gene-deficient (Mdk(-/-)) mice in terms of skeletal muscle degeneration and regeneration after injury by bupivacaine injection into the tibialis anterior muscle. Injured muscles showed intense inflammatory cell infiltration. Myotubes, myofibers with centrally located nuclei in their cytoplasm, were significantly smaller in Mdk(-/-) mice than in wild type (Mdk(+/+)) mice 7 days after injury (p=0.02). The distribution of myotube sizes showed quantitative differences between the two groups at 5 and 7 days, but not at 14 days. Many small myotubes were found in the regenerative area of Mdk(-/-) mice compared with that of Mdk(+/+)mice 5 and 7 days after injury. The expression of Iba1, a macrophage marker, was significantly lower in Mdk(-/-) mice 3 days after injury (p=0.01). The number of desmin-positive cells like myoblasts in Mdk(-/-) mice was significantly fewer than that in Mdk(+/+) mice 3 days after injury. Our results suggested that deletion of MK results in a delay in regeneration, preceded by decelerated migration of macrophages to the damaged area, and that MK has a role in cell differentiation and maturation after skeletal muscle injury.
Collapse
Affiliation(s)
- Masako Ikutomo
- School of Health Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Harutoshi Sakakima
- School of Health Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.
| | - Fumiyo Matsuda
- School of Health Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Yoshihiro Yoshida
- School of Health Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
42
|
Fan N, Sun H, Wang Y, Zhang L, Xia Z, Peng L, Hou Y, Shen W, Liu R, Peng Y. Midkine, a potential link between obesity and insulin resistance. PLoS One 2014; 9:e88299. [PMID: 24516630 PMCID: PMC3917881 DOI: 10.1371/journal.pone.0088299] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 01/06/2014] [Indexed: 12/16/2022] Open
Abstract
Obesity is associated with increased production of inflammatory mediators in adipose tissue, which contributes to chronic inflammation and insulin resistance. Midkine (MK) is a heparin-binding growth factor with potent proinflammatory activities. We aimed to test whether MK is associated with obesity and has a role in insulin resistance. It was found that MK was expressed in adipocytes and regulated by inflammatory modulators (TNF-α and rosiglitazone). In addition, a significant increase in MK levels was observed in adipose tissue of obese ob/ob mice as well as in serum of overweight/obese subjects when compared with their respective controls. In vitro studies further revealed that MK impaired insulin signaling in 3T3-L1 adipocytes, as indicated by reduced phosphorylation of Akt and IRS-1 and decreased translocation of glucose transporter 4 (GLUT4) to the plasma membrane in response to insulin stimulation. Moreover, MK activated the STAT3-suppressor of cytokine signaling 3 (SOCS3) pathway in adipocytes. Thus, MK is a novel adipocyte-secreted factor associated with obesity and inhibition of insulin signaling in adipocytes. It may provide a potential link between obesity and insulin resistance.
Collapse
Affiliation(s)
- Nengguang Fan
- Department of Endocrinology, Shanghai First People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Endocrinology, Shanghai Songjiang Center Hospital, Shanghai, China
| | - Haiyan Sun
- Department of Endocrinology, Shanghai First People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yifei Wang
- Department of Endocrinology, Shanghai First People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lijuan Zhang
- Department of Endocrinology, Shanghai Songjiang Center Hospital, Shanghai, China
| | - Zhenhua Xia
- Department of Endocrinology, Shanghai Songjiang Center Hospital, Shanghai, China
| | - Liang Peng
- Department of Laboratory Medicine, Shanghai Songjiang Center Hospital, Shanghai, China
| | - Yanqiang Hou
- Department of Laboratory Medicine, Shanghai Songjiang Center Hospital, Shanghai, China
| | - Weiqin Shen
- Department of Laboratory Medicine, Shanghai Songjiang Center Hospital, Shanghai, China
| | - Rui Liu
- Department of Endocrinology, Shanghai First People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yongde Peng
- Department of Endocrinology, Shanghai First People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- * E-mail:
| |
Collapse
|
43
|
Liedert A, Schinke T, Ignatius A, Amling M. The role of midkine in skeletal remodelling. Br J Pharmacol 2014; 171:870-8. [PMID: 24102259 PMCID: PMC3925025 DOI: 10.1111/bph.12412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/02/2013] [Accepted: 09/09/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Bone tissue is subjected to continuous remodelling, replacing old or damaged bone throughout life. In bone remodelling, the coordinated activities of bone-forming osteoblasts and bone-resorbing osteoclasts ensure the maintenance of bone mass and strength. In early life, the balance of these cellular activities is tightly regulated by various factors, including systemic hormones, the mechanical environment and locally released growth factors. Age-related changes in the activity of these factors in bone remodelling can result in diseases with low bone mass, such as osteoporosis. Osteoporosis is a systemic and age-related skeletal disease characterized by low bone mass and structural degeneration of bone tissue, predisposing the patient to an increased fracture risk. The growth factor midkine (Mdk) plays a key role in bone remodelling and it is expressed during bone formation and fracture repair. Using a mouse deficient in Mdk, our group have identified this protein as a negative regulator of bone formation and mechanically induced bone remodelling. Thus, specific Mdk antagonists might represent a therapeutic option for diseases characterized by low bone mass, such as osteoporosis. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- A Liedert
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | | | | | | |
Collapse
|
44
|
Muramatsu T. Structure and function of midkine as the basis of its pharmacological effects. Br J Pharmacol 2014; 171:814-26. [PMID: 23992440 PMCID: PMC3925020 DOI: 10.1111/bph.12353] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/31/2013] [Accepted: 08/12/2013] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Midkine (MK) is a heparin-binding growth factor or cytokine and forms a small protein family, the other member of which is pleiotrophin. MK enhances survival, migration, cytokine expression, differentiation and other activities of target cells. MK is involved in various physiological processes, such as development, reproduction and repair, and also plays important roles in the pathogenesis of inflammatory and malignant diseases. MK is largely composed of two domains, namely a more N-terminally located N-domain and a more C-terminally located C-domain. Both domains are basically composed of three antiparallel β-sheets. In addition, there are short tails in the N-terminal and C-terminal sides and a hinge connecting the two domains. Several membrane proteins have been identified as MK receptors: receptor protein tyrosine phosphatase Z1 (PTPζ), low-density lipoprotein receptor-related protein, integrins, neuroglycan C, anaplastic lymphoma kinase and Notch-2. Among them, the most established one is PTPζ. It is a transmembrane tyrosine phophatase with chondroitin sulfate, which is essential for high-affinity binding with MK. PI3K and MAPK play important roles in the downstream signalling system of MK, while transcription factors affected by MK signalling include NF-κB, Hes-1 and STATs. Because of the involvement of MK in various physiological and pathological processes, MK itself as well as pharmaceuticals targeting MK and its signalling system are expected to be valuable for the treatment of numerous diseases. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- T Muramatsu
- Department of Health Science, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasakicho, Nisshinn, Aichi, 470-0195, Japan. ,
| |
Collapse
|
45
|
Cohen S, Shachar I. Midkine as a regulator of B cell survival in health and disease. Br J Pharmacol 2014; 171:888-95. [PMID: 24111754 PMCID: PMC3925027 DOI: 10.1111/bph.12419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/27/2013] [Accepted: 09/03/2013] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED In healthy individuals, the pool of peripheral lymphocytes is constant in size. The control of lymphoid homeostasis is the result of a very fine balance between lymphocyte production, survival and proliferation. Survival factors have been shown to play a critical role in maintaining the correct size of lymphocyte populations. Midkine, a heparin-binding cytokine was recently shown to be involved in cell proliferation, differentiation and apoptosis in various cell types including normal and malignant B cells. This review focuses on the role of midkine in the regulation of peripheral B cell survival in health and disease. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- Sivan Cohen
- Department of Immunology, Weizmann Institute of ScienceRehovot, Israel
| | - Idit Shachar
- Department of Immunology, Weizmann Institute of ScienceRehovot, Israel
| |
Collapse
|
46
|
Thieblemont C, Bertoni F, Copie-Bergman C, Ferreri AJ, Ponzoni M. Chronic inflammation and extra-nodal marginal-zone lymphomas of MALT-type. Semin Cancer Biol 2014; 24:33-42. [DOI: 10.1016/j.semcancer.2013.11.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 11/23/2013] [Accepted: 11/29/2013] [Indexed: 12/26/2022]
|
47
|
Herradón G, Pérez-García C. Targeting midkine and pleiotrophin signalling pathways in addiction and neurodegenerative disorders: recent progress and perspectives. Br J Pharmacol 2014; 171:837-48. [PMID: 23889475 PMCID: PMC3925022 DOI: 10.1111/bph.12312] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/09/2013] [Accepted: 07/21/2013] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED Midkine (MK) and pleiotrophin (PTN) are two neurotrophic factors that are highly up-regulated in different brain regions after the administration of various drugs of abuse and in degenerative areas of the brain. A deficiency in both MK and PTN has been suggested to be an important genetic factor, which confers vulnerability to the development of the neurodegenerative disorders associated with drugs of abuse in humans. In this review, evidence demonstrating that MK and PTN limit the rewarding effects of drugs of abuse and, potentially, prevent drug relapse is compiled. There is also convincing evidence that MK and PTN have neuroprotective effects against the neurotoxicity and development of neurodegenerative disorders induced by drugs of abuse. Exogenous administration of MK and/or PTN into the CNS by means of non-invasive methods is proposed as a novel therapeutic strategy for addictive and neurodegenerative diseases. Identification of new molecular targets downstream of the MK and PTN signalling pathways or pharmacological modulation of those already known may also provide a more traditional, but probably effective, therapeutic strategy for treating addictive and neurodegenerative disorders. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- G Herradón
- Pharmacology lab Department of Pharmaceutical and Health Sciences, Facultad de Farmacia, Universidad CEU San PabloBoadilla del Monte, Madrid, Spain
| | - C Pérez-García
- Pharmacology lab Department of Pharmaceutical and Health Sciences, Facultad de Farmacia, Universidad CEU San PabloBoadilla del Monte, Madrid, Spain
| |
Collapse
|
48
|
Ikeda S, Yamada M. Midkine and cytoplasmic maturation of mammalian oocytes in the context of ovarian follicle physiology. Br J Pharmacol 2014; 171:827-36. [PMID: 23889362 PMCID: PMC3925021 DOI: 10.1111/bph.12311] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/12/2013] [Accepted: 07/21/2013] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Midkine (MK) was originally characterized as a member of a distinct family of neurotrophic factors functioning in the CNS. However, it was later discovered that MK is abundantly expressed in ovarian follicles. Since then, the physiological roles of this molecule in the ovary have been steadily investigated. During the in vitro maturation (IVM) of oocytes MK was shown to promote the cytoplasmic maturation of oocytes, as indicated by post-fertilization development. This effect of MK could be mediated via its pro-survival (anti-apoptotic) effects on the cumulus-granulosa cells that surround oocytes. The oocyte competence-promoting effects of MK are discussed in the context of the recently discovered involvement of MK in the full maturation of ovarian follicles. MK was at the frontline of a new paradigm for neurotrophic factors as oocytetrophic factors. MK may promote the developmental competence of oocytes via common signalling molecules with the other neurotrophic factor(s). Alternatively or concomitantly, MK may also interact with various transmembrane molecules on cumulus-granulosa cells, which are important for ovarian follicle growth, dominance and differentiation, and act as a unique pro-survival factor in ovarian follicles, such that MK promotes oocyte competence. MK, along with other ovarian neurotrophic factors, may contribute to the optimization of the IVM system. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
| | - Masayasu Yamada
- Laboratory of Reproductive Biology Graduate School of Agriculture, Kyoto UniversityKyoto, Japan
| |
Collapse
|
49
|
The cytokine midkine supports neutrophil trafficking during acute inflammation by promoting adhesion via β2 integrins (CD11/CD18). Blood 2014; 123:1887-96. [PMID: 24458438 DOI: 10.1182/blood-2013-06-510875] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Emerging evidence suggests a role of the cytokine midkine (MK) in inflammation. In this study, its functional relevance for recruitment of polymorphonuclear neutrophils (PMNs) during acute inflammation was investigated. Intravital microscopy and histologic analysis of tumor necrosis factor-α-stimulated cremaster muscle venules revealed severely compromised leukocyte adhesion and extravasation in MK(-/-) mice compared with MK(+/+) animals. Systemic administration of recombinant MK completely rescued the adhesion defect in MK(-/-) mice. In a hind limb ischemia model, leukocyte accumulation in MK(-/-) mice was significantly diminished compared with MK(+/+) animals. However, MK did not lead to an inflammatory activation of PMNs or endothelial cells suggesting that it does not serve as classical proinflammatory cytokine. Unexpectedly, immobilized MK mediated PMN adhesion under static and flow conditions, whereas PMN-derived MK was dispensable for the induction of adhesion. Furthermore, adhesion strengthening remained unaffected by MK. Flow cytometry revealed that immobilized, but not soluble MK, significantly promoted the high affinity conformation of β2 integrins of PMNs. Blocking studies of low-density lipoprotein receptor-related protein 1 (LRP1) suggested that LRP1 may act as a receptor for MK on PMNs. Thus, MK seems to support PMN adhesion by promoting the high affinity conformation of β2 integrins, thereby facilitating PMN trafficking during acute inflammation.
Collapse
|
50
|
ALK: Anaplastic lymphoma kinase. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|