1
|
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
|
2
|
Ianiro G, Niro A, Rosa L, Valenti P, Musci G, Cutone A. To Boost or to Reset: The Role of Lactoferrin in Energy Metabolism. Int J Mol Sci 2023; 24:15925. [PMID: 37958908 PMCID: PMC10650157 DOI: 10.3390/ijms242115925] [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: 10/03/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Many pathological conditions, including obesity, diabetes, hypertension, heart disease, and cancer, are associated with abnormal metabolic states. The progressive loss of metabolic control is commonly characterized by insulin resistance, atherogenic dyslipidemia, inflammation, central obesity, and hypertension, a cluster of metabolic dysregulations usually referred to as the "metabolic syndrome". Recently, nutraceuticals have gained attention for the generalized perception that natural substances may be synonymous with health and balance, thus becoming favorable candidates for the adjuvant treatment of metabolic dysregulations. Among nutraceutical proteins, lactoferrin (Lf), an iron-binding glycoprotein of the innate immune system, has been widely recognized for its multifaceted activities and high tolerance. As this review shows, Lf can exert a dual role in human metabolism, either boosting or resetting it under physiological and pathological conditions, respectively. Lf consumption is safe and is associated with several benefits for human health, including the promotion of oral and gastrointestinal homeostasis, control of glucose and lipid metabolism, reduction of systemic inflammation, and regulation of iron absorption and balance. Overall, Lf can be recommended as a promising natural, completely non-toxic adjuvant for application as a long-term prophylaxis in the therapy for metabolic disorders, such as insulin resistance/type II diabetes and the metabolic syndrome.
Collapse
Affiliation(s)
- Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Antonella Niro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (L.R.); (P.V.)
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (L.R.); (P.V.)
| | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| |
Collapse
|
3
|
Dzhumashev D, Timpanaro A, Ali S, De Micheli AJ, Mamchaoui K, Cascone I, Rössler J, Bernasconi M. Quantum Dot-Based Screening Identifies F3 Peptide and Reveals Cell Surface Nucleolin as a Therapeutic Target for Rhabdomyosarcoma. Cancers (Basel) 2022; 14:5048. [PMID: 36291832 PMCID: PMC9600270 DOI: 10.3390/cancers14205048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Active drug delivery by tumor-targeting peptides is a promising approach to improve existing therapies for rhabdomyosarcoma (RMS), by increasing the therapeutic effect and decreasing the systemic toxicity, e.g., by drug-loaded peptide-targeted nanoparticles. Here, we tested 20 different tumor-targeting peptides for their ability to bind to two RMS cell lines, Rh30 and RD, using quantum dots Streptavidin and biotin-peptides conjugates as a model for nanoparticles. Four peptides revealed a very strong binding to RMS cells: NCAM-1-targeting NTP peptide, nucleolin-targeting F3 peptide, and two Furin-targeting peptides, TmR and shTmR. F3 peptide showed the strongest binding to all RMS cell lines tested, low binding to normal control myoblasts and fibroblasts, and efficient internalization into RMS cells demonstrated by the cytoplasmic delivery of the Saporin toxin. The expression of the nucleophosphoprotein nucleolin, the target of F3, on the surface of RMS cell lines was validated by competition with the natural ligand lactoferrin, by colocalization with the nucleolin-binding aptamer AS1411, and by the marked sensitivity of RMS cell lines to the growth inhibitory nucleolin-binding N6L pseudopeptide. Taken together, our results indicate that nucleolin-targeting by F3 peptide represents a potential therapeutic approach for RMS.
Collapse
Affiliation(s)
- Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Andrea Timpanaro
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Safa Ali
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Andrea J. De Micheli
- Department of Oncology, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| | - Kamel Mamchaoui
- Centre de Recherche en Myologie, Institut de Myologie, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Ilaria Cascone
- IMRB, INSERM, University Paris Est Creteil, 94010 Creteil, France
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d’Investigation Clinique Biothérapie, 94010 Créteil, France
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| |
Collapse
|
4
|
Ferrara B, Belbekhouche S, Habert D, Houppe C, Vallée B, Bourgoin-Voillard S, Cohen JL, Cascone I, Courty J. Cell surface nucleolin as active bait for nanomedicine in cancer therapy: a promising option. NANOTECHNOLOGY 2021; 32:322001. [PMID: 33892482 DOI: 10.1088/1361-6528/abfb30] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/23/2021] [Indexed: 06/12/2023]
Abstract
Conventional chemotherapy used against cancer is mostly limited due to their non-targeted nature, affecting normal tissue and causing undesirable toxic effects to the affected tissue. With the aim of improving these treatments both therapeutically and in terms of their safety, numerous studies are currently being carried out using nanoparticles (NPs) as a vector combining tumor targeting and carrying therapeutic tools. In this context, it appears that nucleolin, a molecule over-expressed on the surface of tumor cells, is an interesting therapeutic target. Several ligands, antagonists of nucleolin of various origins, such as AS1411, the F3 peptide and the multivalent pseudopeptide N6L have been developed and studied as therapeutic tools against cancer. Over the last ten years or so, numerous studies have been published demonstrating that these antagonists can be used as tumor targeting agents with NPs from various origins. Focusing on nucleolin ligands, the aim of this article is to review the literature recently published or under experimentation in our research team to evaluate the efficacy and future development of these tools as anti-tumor agents.
Collapse
Affiliation(s)
- Benedetta Ferrara
- Université Paris-Est Creteil, Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, F-94010 Créteil, France
| | - Sabrina Belbekhouche
- Université Paris-Est Creteil, CNRS, Institut Chimie et Matériaux Paris Est, UMR 7182, 2 Rue Henri Dunant, F-94320 Thiais, France
| | - Damien Habert
- Université Paris-Est Creteil, Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, F-94010 Créteil, France
| | - Claire Houppe
- Université Paris-Est Creteil, Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, F-94010 Créteil, France
| | - Benoit Vallée
- Université Paris-Est Creteil, Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, F-94010 Créteil, France
| | - Sandrine Bourgoin-Voillard
- Université Paris-Est Creteil, Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, F-94010 Créteil, France
- Université Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics/Prométhée Proteomic Platform, UGA-INSERM U1055-CHUGA, Grenoble, France
- Université Grenoble Alpes, CNRS, Grenoble INP, CHU Grenoble Alpes, TIMC, PROMETHEE Proteomic Platform, Grenoble, France
| | - José L Cohen
- Université Paris-Est Creteil, Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, F-94010 Créteil, France
| | - Ilaria Cascone
- Université Paris-Est Creteil, Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, F-94010 Créteil, France
| | - José Courty
- Université Paris-Est Creteil, Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, F-94010 Créteil, France
| |
Collapse
|
5
|
Ryan E, Shen D, Wang X. Pleiotrophin interacts with glycosaminoglycans in a highly flexible and adaptable manner. FEBS Lett 2021; 595:925-941. [PMID: 33529353 DOI: 10.1002/1873-3468.14052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022]
Abstract
Pleiotrophin (PTN) is a potent mitogenic cytokine whose activities are controlled by its interactions with glycosaminoglycan (GAG). We examined the specificity of PTN for several types of GAG oligosaccharides. Our data indicate that the interaction of PTN with GAGs is dependent on the sulfation density of GAGs. Surprisingly, an acidic peptide also had similar interactions with PTN as GAGs. This shows that the interaction of PTN with anionic polymers is flexible and adaptable and that the charge density is the main determinant of the interaction. In addition, we show that PTN can compensate for the loss of its termini in interactions with heparin oligosaccharides, allowing it to maintain its affinity for GAGs in the absence of the termini. Taken together, these data provide valuable insight into the interactions of PTN with its proteoglycan receptors.
Collapse
Affiliation(s)
- Eathen Ryan
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Di Shen
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Xu Wang
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
6
|
Vester SK, Beavil RL, Lynham S, Beavil AJ, Cunninghame Graham DS, McDonnell JM, Vyse TJ. Nucleolin acts as the receptor for C1QTNF4 and supports C1QTNF4-mediated innate immunity modulation. J Biol Chem 2021; 296:100513. [PMID: 33676896 PMCID: PMC8042453 DOI: 10.1016/j.jbc.2021.100513] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
The C1q and TNF related 4 (C1QTNF4) protein is a structurally unique member of the C1QTNF family, a family of secreted proteins that have structural homology with both complement C1q and the tumor necrosis factor superfamily. C1QTNF4 has been linked to the autoimmune disease systemic lupus erythematosus through genetic studies; however, its role in immunity and inflammation remains poorly defined and a cell surface receptor of C1QTNF4 has yet to be identified. Here we report identification of nucleolin as a cell surface receptor of C1QTNF4 using mass spectrometric analysis. Additionally, we present evidence that the interaction between C1QTNF4 and nucleolin is mediated by the second C1q-like domain of C1QTNF4 and the C terminus of nucleolin. We show that monocytes and B cells are target cells of C1QTNF4 and observe extensive binding to dead cells. Imaging flow cytometry experiments in monocytes show that C1QTNF4 becomes actively internalized upon cell binding. Our results suggest that nucleolin may serve as a docking molecule for C1QTNF4 and act in a context-dependent manner through coreceptors. Taken together, these findings further our understanding of C1QTNF4's function in the healthy immune system and how dysfunction may contribute to the development of systemic lupus erythematosus.
Collapse
Affiliation(s)
- Susan K Vester
- Department of Medical & Molecular Genetics, King's College London, London, UK
| | - Rebecca L Beavil
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK; Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Steven Lynham
- Proteomics Facility, Centre of Excellence for Mass Spectrometry, King's College London, London, UK
| | - Andrew J Beavil
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK; Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | | | - James M McDonnell
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK; Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Timothy J Vyse
- Department of Medical & Molecular Genetics, King's College London, London, UK.
| |
Collapse
|
7
|
Sluzalska KD, Slawski J, Sochacka M, Lampart A, Otlewski J, Zakrzewska M. Intracellular partners of fibroblast growth factors 1 and 2 - implications for functions. Cytokine Growth Factor Rev 2020; 57:93-111. [PMID: 32475760 DOI: 10.1016/j.cytogfr.2020.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Fibroblast growth factors 1 and 2 (FGF1 and FGF2) are mainly considered as ligands of surface receptors through which they regulate a broad spectrum of biological processes. They are secreted in non-canonical way and, unlike other growth factors, they are able to translocate from the endosome to the cell interior. These unique features, as well as the role of the intracellular pool of FGF1 and FGF2, are far from being fully understood. An increasing number of reports address this problem, focusing on the intracellular interactions of FGF1 and 2. Here, we summarize the current state of knowledge of the FGF1 and FGF2 binding partners inside the cell and the possible role of these interactions. The partner proteins are grouped according to their function, including proteins involved in secretion, cell signaling, nucleocytoplasmic transport, binding and processing of nucleic acids, ATP binding, and cytoskeleton assembly. An in-depth analysis of the network of these binding partners could indicate novel, non-classical functions of FGF1 and FGF2 and uncover an additional level of a fine control of the well-known FGF-regulated cellular processes.
Collapse
Affiliation(s)
- Katarzyna Dominika Sluzalska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jakub Slawski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Martyna Sochacka
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Agata Lampart
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
8
|
Darche M, Cossutta M, Caruana L, Houppe C, Gilles ME, Habert D, Guilloneau X, Vignaud L, Paques M, Courty J, Cascone I. Antagonist of nucleolin, N6L, inhibits neovascularization in mouse models of retinopathies. FASEB J 2020; 34:5851-5862. [PMID: 32141122 DOI: 10.1096/fj.201901876r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/11/2020] [Accepted: 02/21/2020] [Indexed: 12/18/2022]
Abstract
Retinal vascular diseases (RVD) have been identified as a major cause of blindness worldwide. These pathologies, including the wet form of age-related macular degeneration, retinopathy of prematurity, and diabetic retinopathy are currently treated by intravitreal delivery of anti-vascular endothelial growth factor (VEGF) agents. However, repeated intravitreal injections can lead to ocular complications and resistance to these treatments. Thus, there is a need to find new targeted therapies. Nucleolin regulates the endothelial cell (EC) activation and angiogenesis. In previous studies, we designed a pseudopeptide, N6L, that binds the nucleolin and blocks the tumor angiogenesis. In this study, the effect of N6L was investigated in two experimental models of retinopathies including oxygen-induced retinopathy (OIR) and choroidal neovascularization (CNV). We found that in mouse OIR, intraperitoneal injection of N6L is delivered to activated ECs and induced a 50% reduction of pathological neovascularization. The anti-angiogenic effect of N6L has been tested in CNV model in which the systemic injection of N6L induced a 33% reduction of angiogenesis. This effect is comparable to those obtained with VEGF-trap, a standard of care drug for RVD. Interestingly, with preventive and curative treatments, neoangiogenesis is inhibited by 59%. Our results have potential interest in the development of new therapies targeting other molecules than VEGF for RVD.
Collapse
Affiliation(s)
- Marie Darche
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
- Clinical Investigation Center 1423, Centre Hospitalier National des Quinze-Vingts, Institut Hospitalo-Universitaire ForeSight, Sorbonne Université, Paris, France
| | - Mélissande Cossutta
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | - Laure Caruana
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | - Claire Houppe
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | | | - Damien Habert
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | - Xavier Guilloneau
- INSERM, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Lucile Vignaud
- INSERM, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Michel Paques
- Clinical Investigation Center 1423, Centre Hospitalier National des Quinze-Vingts, Institut Hospitalo-Universitaire ForeSight, Sorbonne Université, Paris, France
| | - José Courty
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | - Ilaria Cascone
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| |
Collapse
|
9
|
Chen J, Zhong J, Niu P, Xu L, Zhou L, Wu H, Chen C, Dai L. Toxicologic evaluation of repetitive 4-week intravenous injections of midkine antisense oligonucleotide nanoliposomes in rats. Regul Toxicol Pharmacol 2019; 103:130-139. [PMID: 30682377 DOI: 10.1016/j.yrtph.2019.01.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 01/10/2019] [Accepted: 01/20/2019] [Indexed: 01/12/2023]
Abstract
Midkine antisense oligonucleotide (MK-ASODN) nanoliposomes have previously been shown to have inhibitory activity against hepatocellular carcinoma growth. Herein we report the 4-week sub-chronic toxicity of MK-ASODN nanoliposomes in SD rats. The adverse effects included loss of body weight gain and food consumption, peri-rhinal bleeding, piloerection, peri-anal filth, and kidney, liver, spleen, thymus, lung, and injection site lesions at high doses. Macroscopic changes were observed in the kidneys of the high-dose group, accompanied by a variation in urine protein and white blood cells, blood urea nitrogen, and serum creatinine. The increased spleen and liver coefficient, and the variation in circulating white blood cells, lymphocytes, and eosinophils in the high-dose group demonstrated that inflammation was caused by MK-ASODN nanoliposomes and was consistent with the macroscopic changes in the spleen and liver. The main necropsy findings of the animals that died were macroscopic changes in the lung. No severe toxic effects or mortalities occurred in the low- and medium-dose groups. However, a No Adverse Effect Level (NOAEL) was not identified since there were changes in organs deemed to be adverse at all dose levels. Thus, the maximum tolerated dose of MK-ASODN nanoliposomes for rats was considered to be 6 mg/kg/day.
Collapse
Affiliation(s)
- Jing Chen
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, 198 Hongqi Road, Huzhou, 313000, PR China
| | - Jing Zhong
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, 198 Hongqi Road, Huzhou, 313000, PR China
| | - Pingping Niu
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, 198 Hongqi Road, Huzhou, 313000, PR China
| | - Limin Xu
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, 198 Hongqi Road, Huzhou, 313000, PR China
| | - Linfu Zhou
- Medical Biotechnology Laboratory, Zhejiang University, 388 Yuhangtang Road, Hangzhou, Zhejiang, 310038, PR China
| | - Honghai Wu
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Chao Chen
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Licheng Dai
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, 198 Hongqi Road, Huzhou, 313000, PR China.
| |
Collapse
|
10
|
Gregório AC, Lacerda M, Figueiredo P, Simões S, Dias S, Moreira JN. Meeting the needs of breast cancer: A nucleolin's perspective. Crit Rev Oncol Hematol 2018; 125:89-101. [PMID: 29650282 DOI: 10.1016/j.critrevonc.2018.03.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 01/30/2018] [Accepted: 03/20/2018] [Indexed: 12/21/2022] Open
Abstract
A major challenge in the management of breast cancer disease has been the development of metastases. Finding new molecular targets and the design of targeted therapeutic approaches to improve the overall survival and quality of life of these patients is, therefore, of great importance. Nucleolin, which is overexpressed in cancer cells and tumor-associated blood vessels, have been implicated in various processes supporting tumorigenesis and angiogenesis. Additionally, its overexpression has been demonstrated in a variety of human neoplasias as an unfavorable prognostic factor, associated with a high risk of relapse and low overall survival. Hence, nucleolin has emerged as a relevant target for therapeutic intervention in cancer malignancy, including breast cancer. This review focus on the contribution of nucleolin for cancer disease and on the development of therapeutic strategies targeting this protein. In this respect, it also provides a critical analysis about the potential and pitfalls of nanomedicine for cancer therapy.
Collapse
Affiliation(s)
- Ana C Gregório
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Manuela Lacerda
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
| | - Paulo Figueiredo
- IPOFG-EPE - Portuguese Institute of Oncology Francisco Gentil, 3000-075 Coimbra, Portugal
| | - Sérgio Simões
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; FFUC - Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Sérgio Dias
- IMM - Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; FFUC - Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, 3000-354 Coimbra, Portugal.
| |
Collapse
|
11
|
Yuan X, Hu T, He H, Qiu H, Wu X, Chen J, Wang M, Chen C, Huang S. Respiratory syncytial virus prolifically infects N2a neuronal cells, leading to TLR4 and nucleolin protein modulations and RSV F protein co-localization with TLR4 and nucleolin. J Biomed Sci 2018; 25:13. [PMID: 29427996 PMCID: PMC6389248 DOI: 10.1186/s12929-018-0416-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/02/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infects the central nervous system, resulting in neurological symptoms. However, the precise underlying pathogenic mechanisms have not been elucidated. In the present study, the infectivity of RSV on N2a neuronal cells and the possible roles of Toll-like receptor 4 (TLR4) and nucleolin (C23) during RSV infection were investigated. METHODS We compared two experimental groups (infected and non-infected) and monitored the RSV viral titers in the culture supernatant by a viral plaque assay. We also inspected the morphology of the nucleus in infected N2a cells. We measured the level of RSV F protein and studied its co-localization with TLR4 and nucleolin using immunofluorescence assays and laser confocal microscopy. The potential interaction of RSV F protein with TLR4 and nucleolin was examined by coimmunoprecipitation. The expression changes of TLR4, nucleolin, TLR3 and TLR7 proteins in N2a cells and IL-6 and TNF-α in the culture supernatant were investigated by Western Blot analysis and ELISA assay. Changes in neuronal cell apoptosis status was examined by flow cytometry. RESULTS The results demonstrated prolific RSV infection of N2a cells, which triggered a decrease of NeuN protein expression, coinciding with an increase of nuclear lesions, F protein expression, RSV viral titers, and late apoptotic levels of N2a cells. RSV infection induced co-localization of RSV F protein with TLR4 and nucleolin, which could potentially lead to a direct interaction. Furthermore, it was found that TLR4 and nucleolin levels increased early after infection and decreased subsequently, whereas TLR3 and TLR7 expression increased throughout RSV infection. CONCLUSION The RSV Long strain can prolifically infect N2a neuronal cells, modulating the expression of TLR4 and nucleolin, as well as TLR3, TLR7 and their downstream inflammatory factors, and inducing the co-localization of the RSV F protein with TLR4 and nucleolin.
Collapse
Affiliation(s)
- Xiaoling Yuan
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
| | - Tao Hu
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
| | - Hanwen He
- Department of Laboratory Medicine, Anhui Health College, Chizhou, Anhui Province 247099 People’s Republic of China
| | - Huan Qiu
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
| | - Xuan Wu
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
| | - Jingxian Chen
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032 USA
| | - Minmin Wang
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
| | - Cheng Chen
- Department of Clinical Medicine, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
| | - Shenghai Huang
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
- School of Life Sciences, Anhui Medical University, Hefei, Anhui Province 230032 People’s Republic of China
| |
Collapse
|
12
|
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: 25] [Impact Index Per Article: 3.1] [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
|
13
|
Bates PJ, Reyes-Reyes EM, Malik MT, Murphy EM, O'Toole MG, Trent JO. G-quadruplex oligonucleotide AS1411 as a cancer-targeting agent: Uses and mechanisms. Biochim Biophys Acta Gen Subj 2017; 1861:1414-1428. [PMID: 28007579 DOI: 10.1016/j.bbagen.2016.12.015] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/16/2016] [Accepted: 12/17/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND AS1411 is a 26-mer G-rich DNA oligonucleotide that forms a variety of G-quadruplex structures. It was identified based on its cancer-selective antiproliferative activity and subsequently determined to be an aptamer to nucleolin, a multifunctional protein that preferentially binds quadruplex nucleic acids and which is present at high levels on the surface of cancer cells. AS1411 has exceptionally efficient cellular internalization compared to non-quadruplex DNA sequences. SCOPE OF REVIEW Recent developments related to AS1411 will be examined, with a focus on its use for targeted delivery of therapeutic and imaging agents. MAJOR CONCLUSIONS Numerous research groups have used AS1411 as a targeting agent to deliver nanoparticles, oligonucleotides, and small molecules into cancer cells. Studies in animal models have demonstrated that AS1411-linked materials can accumulate selectively in tumors following systemic administration. The mechanism underlying the cancer-targeting ability of AS1411 is not completely understood, but recent studies suggest a model that involves: (1) initial uptake by macropinocytosis, a form of endocytosis prevalent in cancer cells; (2) stimulation of macropinocytosis by a nucleolin-dependent mechanism resulting in further uptake; and (3) disruption of nucleolin-mediated trafficking and efflux leading to cargoes becoming trapped inside cancer cells. SIGNIFICANCE Human trials have indicated that AS1411 is safe and can induce durable remissions in a few patients, but new strategies are needed to maximize its clinical impact. A better understanding of the mechanisms by which AS1411 targets and kills cancer cells may hasten the development of promising technologies using AS1411-linked nanoparticles or conjugates for cancer-targeted therapy and imaging. This article is part of a Special Issue entitled "G-quadruplex" Guest Editor: Dr. Concetta Giancola and Dr. Daniela Montesarchio.
Collapse
Affiliation(s)
- Paula J Bates
- Department of Medicine, University of Louisville, USA; James Graham Brown Cancer Center, University of Louisville, USA.
| | | | - Mohammad T Malik
- Department of Medicine, University of Louisville, USA; James Graham Brown Cancer Center, University of Louisville, USA
| | - Emily M Murphy
- Department of Biomedical Engineering, University of Louisville, USA
| | - Martin G O'Toole
- Department of Biomedical Engineering, University of Louisville, USA
| | - John O Trent
- Department of Medicine, University of Louisville, USA; James Graham Brown Cancer Center, University of Louisville, USA
| |
Collapse
|
14
|
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: 2.2] [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
|
15
|
Fong-ngern K, Sueksakit K, Thongboonkerd V. Surface heat shock protein 90 serves as a potential receptor for calcium oxalate crystal on apical membrane of renal tubular epithelial cells. J Biol Inorg Chem 2016; 21:463-74. [DOI: 10.1007/s00775-016-1355-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/13/2016] [Indexed: 02/04/2023]
|
16
|
Hernández BA, Sandoval-Jaime C, Sosnovtsev SV, Green KY, Gutiérrez-Escolano AL. Nucleolin promotes in vitro translation of feline calicivirus genomic RNA. Virology 2016; 489:51-62. [PMID: 26707270 PMCID: PMC4761316 DOI: 10.1016/j.virol.2015.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 12/28/2022]
Abstract
Feline calicivirus depends on host-cell proteins for its replication. We previously showed that knockdown of nucleolin (NCL), a phosphoprotein involved in ribosome biogenesis, resulted in the reduction of FCV protein synthesis and virus yield. Here, we found that NCL may not be involved in FCV binding and entry into cells, but it binds to both ends of the FCV genomic RNA, and stimulates its translation in vitro. AGRO100, an aptamer that specifically binds and inactivates NCL, caused a strong reduction in FCV protein synthesis. This effect could be reversed by the addition of full-length NCL but not by a ΔrNCL, lacking the N-terminal domain. Consistent with this, FCV infection of CrFK cells stably expressing ΔrNCL led to a reduction in virus protein translation. These results suggest that NCL is part of the FCV RNA translational complex, and that the N-terminal part of the protein is required for efficient FCV replication.
Collapse
Affiliation(s)
- Beatriz Alvarado Hernández
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Carlos Sandoval-Jaime
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, UNAM, Cuernavaca, Morelos, Mexico
| | | | - Kim Y Green
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Ana Lorena Gutiérrez-Escolano
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico.
| |
Collapse
|
17
|
Su PY, Wang YF, Huang SW, Lo YC, Wang YH, Wu SR, Shieh DB, Chen SH, Wang JR, Lai MD, Chang CF. Cell surface nucleolin facilitates enterovirus 71 binding and infection. J Virol 2015; 89:4527-38. [PMID: 25673703 PMCID: PMC4442404 DOI: 10.1128/jvi.03498-14] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/28/2015] [Indexed: 02/08/2023] Open
Abstract
UNLABELLED Because the pathogenesis of enterovirus 71 (EV71) remains mostly ambiguous, identifying the factors that mediate viral binding and entry to host cells is indispensable to ultimately uncover the mechanisms that underlie virus infection and pathogenesis. Despite the identification of several receptors/attachment molecules for EV71, the binding, entry, and infection mechanisms of EV71 remain unclear. Herein, we employed glycoproteomic approaches to identify human nucleolin as a novel binding receptor for EV71. Glycoproteins purified by lectin chromatography from the membrane extraction of human cells were treated with sialidase, followed by immunoprecipitation with EV71 particles. Among the 16 proteins identified by tandem mass spectrometry analysis, cell surface nucleolin attracted our attention. We found that EV71 interacted directly with nucleolin via the VP1 capsid protein and that an antinucleolin antibody reduced the binding of EV71 to human cells. In addition, the knockdown of cell surface nucleolin decreased EV71 binding, infection, and production in human cells. Furthermore, the expression of human nucleolin on the cell surface of a mouse cell line increased EV71 binding and conferred EV71 infection and production in the cells. These results strongly indicate that human nucleolin can mediate EV71 binding to and infection of cells. Our findings also demonstrate that the use of glycoproteomic approaches is a reliable methodology to discover novel receptors for pathogens. IMPORTANCE Outbreaks of EV71 have been reported in Asia-Pacific countries and have caused thousands of deaths in young children during the last 2 decades. The discovery of new EV71-interacting molecules to understand the infection mechanism has become an emergent issue. Hence, this study uses glycoproteomic approaches to comprehensively investigate the EV71-interacting glycoproteins. Several EV71-interacting glycoproteins are identified, and the role of cell surface nucleolin in mediating the attachment and entry of EV71 is characterized and validated. Our findings not only indicate a novel target for uncovering the EV71 infection mechanism and anti-EV71 drug discovery but also provide a new strategy for virus receptor identification.
Collapse
Affiliation(s)
- Pei-Yi Su
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Taiwan, Republic of China Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Taiwan, Republic of China
| | - Ya-Fang Wang
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Taiwan, Republic of China
| | - Sheng-Wen Huang
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Taiwan, Republic of China
| | - Yu-Chih Lo
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Taiwan, Republic of China Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Taiwan, Republic of China
| | - Ya-Hui Wang
- Institute of Oral Medicine and Department of Stomatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan, Republic of China
| | - Shang-Rung Wu
- Institute of Oral Medicine and Department of Stomatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan, Republic of China
| | - Dar-Bin Shieh
- Institute of Oral Medicine and Department of Stomatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan, Republic of China Advanced Optoelectronic Technology Center and Center for Micro/Nano Science and Technology, National Cheng Kung University, Taiwan, Republic of China
| | - Shun-Hua Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Taiwan, Republic of China Center of Infectious Disease and Signaling Research, National Cheng Kung University, Taiwan, Republic of China Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Taiwan, Republic of China
| | - Jen-Ren Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Taiwan, Republic of China Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Taiwan, Republic of China Center of Infectious Disease and Signaling Research, National Cheng Kung University, Taiwan, Republic of China
| | - Ming-Der Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Taiwan, Republic of China Center of Infectious Disease and Signaling Research, National Cheng Kung University, Taiwan, Republic of China Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Taiwan, Republic of China
| | - Chuan-Fa Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Taiwan, Republic of China Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Taiwan, Republic of China Center of Infectious Disease and Signaling Research, National Cheng Kung University, Taiwan, Republic of China
| |
Collapse
|
18
|
Oh HJ, Choung HW, Lee HK, Park SJ, Lee JH, Lee DS, Seo BM, Park JC. CPNE7, a preameloblast-derived factor, regulates odontoblastic differentiation of mesenchymal stem cells. Biomaterials 2015; 37:208-17. [PMID: 25453951 DOI: 10.1016/j.biomaterials.2014.10.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 10/02/2014] [Indexed: 10/24/2022]
Abstract
Tooth development involves sequential interactions between dental epithelial and mesenchymal cells. Our previous studies demonstrated that preameloblast-conditioned medium (PA-CM) induces the odontogenic differentiation of human dental pulp cells (hDPCs), and the novel protein Cpne7 in PA-CM was suggested as a candidate signaling molecule. In the present study, we investigated biological function and mechanisms of Cpne7 in regulation of odontoblast differentiation. Cpne7 was expressed in preameloblasts and secreted extracellularly during ameloblast differentiation. After secretion, Cpne7 protein was translocated to differentiating odontoblasts. In odontoblasts, Cpne7 promoted odontoblastic markers and the expression of Dspp in vitro. Cpne7 also induced odontoblast differentiation and promoted dentin/pulp-like tissue formation in hDPCs in vivo. Moreover, Cpne7 induced differentiation into odontoblasts of non-dental mesenchymal stem cells in vitro, and promoted formation of dentin-like tissues including the structure of dentinal tubules in vivo. Mechanistically, Cpne7 interacted with Nucleolin and modulated odontoblast differentiation via the control of Dspp expression. These results suggest Cpne7 is a diffusible signaling molecule that is secreted by preameloblasts, and regulates the differentiation of mesenchymal cells of dental or non-dental origin into odontoblasts.
Collapse
Affiliation(s)
- Hyun-Jung Oh
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, BK 21, Seoul National University, Seoul 110-749, Republic of Korea
| | - Han-Wool Choung
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, BK 21, Seoul National University, Seoul 110-749, Republic of Korea
| | - Hye-Kyung Lee
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, BK 21, Seoul National University, Seoul 110-749, Republic of Korea
| | - Su-Jin Park
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, BK 21, Seoul National University, Seoul 110-749, Republic of Korea
| | - Ji-Hyun Lee
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, BK 21, Seoul National University, Seoul 110-749, Republic of Korea
| | - Dong-Seol Lee
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, BK 21, Seoul National University, Seoul 110-749, Republic of Korea
| | - Byoung-Moo Seo
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul 110-749, Republic of Korea
| | - Joo-Cheol Park
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, BK 21, Seoul National University, Seoul 110-749, Republic of Korea.
| |
Collapse
|
19
|
Lamprou M, Kaspiris A, Panagiotopoulos E, Giannoudis PV, Papadimitriou E. The role of pleiotrophin in bone repair. Injury 2014; 45:1816-23. [PMID: 25456495 DOI: 10.1016/j.injury.2014.10.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/07/2014] [Indexed: 02/02/2023]
Abstract
Bone has an enormous capacity for growth, regeneration, and remodelling, largely due to induction of osteoblasts that are recruited to the site of bone formation. Although the pathways involved have not been fully elucidated, it is well accepted that the immediate environment of the cells is likely to play a role via cell–matrix interactions, mediated by several growth factors. Formation of new blood vessels is also significant and interdependent to bone formation, suggesting that enhancement of angiogenesis could be beneficial during the process of bone repair. Pleiotrophin (PTN), also called osteoblast-specific factor 1, is a heparin-binding angiogenic growth factor, with a well-defined and significant role in both physiological and pathological angiogenesis. In this review we summarise the existing evidence on the role of PTN in bone repair.
Collapse
|
20
|
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: 0.8] [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
|
21
|
Bremer S, Klein K, Sedlmaier A, Abouzied M, Gieselmann V, Franken S. Hepatoma-derived growth factor and nucleolin exist in the same ribonucleoprotein complex. BMC BIOCHEMISTRY 2013; 14:2. [PMID: 23305559 PMCID: PMC3551658 DOI: 10.1186/1471-2091-14-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 01/02/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND Hepatoma-derived growth factor (HDGF) is a protein which is highly expressed in a variety of tumours. HDGF has mitogenic, angiogenic, neurotrophic and antiapoptotic activity but the molecular mechanisms by which it exerts these activities are largely unknown nor has its biological function in tumours been elucidated. Mass spectrometry was performed to analyse the HDGFStrep-tag interactome. By Pull-down-experiments using different protein and nucleic acid constructs the interaction of HDGF and nucleolin was investigated further. RESULTS A number of HDGFStrep-tag copurifying proteins were identified which interact with RNA or are involved in the cellular DNA repair machinery. The most abundant protein, however, copurifying with HDGF in this approach was nucleolin. Therefore we focus on the characterization of the interaction of HDGF and nucleolin in this study. We show that expression of a cytosolic variant of HDGF causes a redistribution of nucleolin into the cytoplasm. Furthermore, formation of HDGF/nucleolin complexes depends on bcl-2 mRNA. Overexpression of full length bcl-2 mRNA increases the number of HDGF/nucleolin complexes whereas expression of only the bcl-2 coding sequence abolishes interaction completely. Further examination reveals that the coding sequence of bcl-2 mRNA together with either the 5' or 3' UTR is sufficient for formation of HDGF/nucleolin complexes. When bcl-2 coding sequence within the full length cDNA is replaced by a sequence coding for secretory alkaline phosphatase complex formation is not enhanced. CONCLUSION The results provide evidence for the existence of HDGF and nucleolin containing nucleoprotein complexes which formation depends on the presence of specific mRNAs. The nature of these RNAs and other components of the complexes should be investigated in future.
Collapse
Affiliation(s)
- Stephanie Bremer
- Institute of Biochemistry and Molecular Biology, University of Bonn, Nussallee 11, Bonn, 53115, Germany
| | - Katharina Klein
- Institute of Biochemistry and Molecular Biology, University of Bonn, Nussallee 11, Bonn, 53115, Germany
| | - Angela Sedlmaier
- Institute of Biochemistry and Molecular Biology, University of Bonn, Nussallee 11, Bonn, 53115, Germany
| | - Mekky Abouzied
- Faculty of Pharmacy, University of El-Minia, El-Minia, Egypt
| | - Volkmar Gieselmann
- Institute of Biochemistry and Molecular Biology, University of Bonn, Nussallee 11, Bonn, 53115, Germany
| | - Sebastian Franken
- Institute of Biochemistry and Molecular Biology, University of Bonn, Nussallee 11, Bonn, 53115, Germany
- Present address: Life-Science-Inkubator, Ludwig-Erhard-Allee 2, Bonn, 53175, Germany
| |
Collapse
|
22
|
Regulation of CLL survival by hypoxia-inducible factor and its target genes. FEBS Lett 2012; 586:2906-10. [PMID: 22841548 DOI: 10.1016/j.febslet.2012.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 07/10/2012] [Accepted: 07/10/2012] [Indexed: 02/02/2023]
Abstract
Chronic lymphocytic leukemia (CLL), the most common adult leukemia in the Western world, is characterized by the progressive accumulation of small mature CD5(+)B lymphocytes in the peripheral blood, lymphoid organs, and bone marrow (BM). The main feature of the disease is decreased apoptosis, resulting in the pathologic accumulation of these malignant cells. Appropriate cellular responses to changes in oxygen tension during normal development or pathological processes, such as cardiovascular disease and cancer, are ultimately regulated by the transcription factor, hypoxia-inducible factor (HIF). Unlike their normal counterparts, CLL cells express HIF-1α even under normoxia. In addition, overexpression of HIF-1α has been observed in leukemic cells in BM specimens from CLL patients. The HIF transcription factor has been implicated in controlling the expression of a wide variety of genes implicated in apoptosis, angiogenesis, invasion, and metastasis. This review describes pathways regulating CLL survival with a focus on HIF-1α and its target genes, MIF and Midkine (MK), and the potential cross-talk between these factors.
Collapse
|
23
|
Cohen S, Shachar I. Cytokines as regulators of proliferation and survival of healthy and malignant peripheral B cells. Cytokine 2012; 60:13-22. [PMID: 22784632 DOI: 10.1016/j.cyto.2012.06.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 05/21/2012] [Accepted: 06/04/2012] [Indexed: 12/31/2022]
Abstract
Adaptive immunity depends on the production and maintenance of a pool of mature peripheral lymphocytes throughout life. The signals regulating the survival of mature splenic B cells have become a major focus in recent studies of B cell immunology. Lasting B cell persistence in the periphery is dependent on survival signals that are transduced by cell surface receptors. Cytokines have been shown to play a critical role in maintaining lymphocyte homeostasis. This review focuses on the role of cytokines and their receptors in the regulation of peripheral B cell survival, with an emphasis on those that have received relatively less attention in the literature.
Collapse
Affiliation(s)
- Sivan Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
24
|
Abstract
Midkine (MK) is a heparin-binding growth factor involved in various cellular processes such as cellular proliferation, survival, and migration. In addition to these typical growth factor activities, MK exhibits several other activities related to fibrinolysis, blood pressure, host defense and other processes. Many cell-surface receptors have been identified to account for the multiple biological activities of MK. The expression of MK is frequently upregulated in many types of human carcinoma. Moreover, blood MK levels are closely correlated with patient outcome. Knockdown and blockade of MK suppress tumorigenesis and tumor development. Thus, MK serves as a tumor marker and a molecular target for cancer therapy. Furthermore, there is growing evidence that MK plays pivotal roles in neural and inflammatory diseases. Understanding of the mechanisms of action of MK is expected to create new therapeutic options for several human diseases.
Collapse
Affiliation(s)
- Kazuma Sakamoto
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | |
Collapse
|
25
|
Koutsioumpa M, Drosou G, Mikelis C, Theochari K, Vourtsis D, Katsoris P, Giannopoulou E, Courty J, Petrou C, Magafa V, Cordopatis P, Papadimitriou E. Pleiotrophin expression and role in physiological angiogenesis in vivo: potential involvement of nucleolin. Vasc Cell 2012; 4:4. [PMID: 22423616 PMCID: PMC3379939 DOI: 10.1186/2045-824x-4-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 03/16/2012] [Indexed: 11/20/2022] Open
Abstract
Background Pleiotrophin (PTN) is a heparin-binding growth factor with significant role(s) in tumour growth and angiogenesis. Although implication of endogenous PTN has been studied in several in vivo models of tumour angiogenesis, its role in physiological angiogenesis has not been addressed. In the present work, we studied expression and functional significance of endogenous PTN during angiogenesis in the chicken embryo chorioallantoic membrane (CAM). Methods Using molecular, cellular and biochemical assays, we studied the expression pattern of PTN in CAM and human endothelial cells and its possible interaction with nucleolin (NCL). CAM cells were transfected with a pCDNA3.1 vector, empty (PC) or containing full length cDNA for PTN in antisense orientation (AS-PTN). Angiogenesis was estimated by measuring total vessel length. In vitro, human endothelial cells migration was studied by using a transwell assay, and down-regulation of NCL was performed by using a proper siRNA. Results Endogenous PTN mRNA and protein levels, as well as protein levels of its receptor protein tyrosine phosphatase beta/zeta (RPTPβ/ζ) were maximal at early stages, when CAM angiogenesis is active. Application of AS-PTN onto CAM at days of active angiogenesis was not toxic to the tissue and led to dose-dependent decreased expression of endogenous PTN, ERK1/2 activity and angiogenesis. Interestingly, endogenous PTN was also immunolocalized at the endothelial cell nucleus, possibly through interaction with NCL, a protein that has a significant role in the nuclear translocation of many proteins. Down-regulation of NCL by siRNA in human endothelial cells significantly decreased nuclear PTN, verifying this hypothesis. Moreover, it led to abolishment of PTN-induced endothelial cell migration, suggesting, for the first time, that PTN-NCL interaction has a functional significance. Conclusions Expression of endogenous PTN correlates with and seems to be involved in angiogenesis of the chicken embryo CAM. Our data suggest that NCL may have a role, increasing the number of growth factors whose angiogenic/tumorigenic activities are mediated by NCL.
Collapse
Affiliation(s)
- Marina Koutsioumpa
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| | - Georgia Drosou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| | - Constantinos Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece.,Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Building 30, Room 203, Bethesda, MD 20892-4340, USA
| | - Katerina Theochari
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| | - Dionussios Vourtsis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| | - Panagiotis Katsoris
- Division of Genetics, Cell & Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Efstathia Giannopoulou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece.,Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University Hospital of Patras, Patras Medical School, 26504 Rio, Greece
| | - Jose Courty
- Laboratoire CRRET, Université Paris Est Créteil, Val de Marne, 61 avenue du Général de Gaulle, 94010 Créteil, Cedex, France
| | - Christos Petrou
- Laboratory of Pharmacognocy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Athens, Greece
| | - Vassiliki Magafa
- Laboratory of Pharmacognocy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Athens, Greece
| | - Paul Cordopatis
- Laboratory of Pharmacognocy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Athens, Greece
| | - Evangelia Papadimitriou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| |
Collapse
|
26
|
Abdelmohsen K, Tominaga K, Lee EK, Srikantan S, Kang MJ, Kim MM, Selimyan R, Martindale JL, Yang X, Carrier F, Zhan M, Becker KG, Gorospe M. Enhanced translation by Nucleolin via G-rich elements in coding and non-coding regions of target mRNAs. Nucleic Acids Res 2011; 39:8513-30. [PMID: 21737422 PMCID: PMC3201861 DOI: 10.1093/nar/gkr488] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 05/25/2011] [Accepted: 05/27/2011] [Indexed: 01/26/2023] Open
Abstract
RNA-binding proteins (RBPs) regulate gene expression at many post-transcriptional levels, including mRNA stability and translation. The RBP nucleolin, with four RNA-recognition motifs, has been implicated in cell proliferation, carcinogenesis and viral infection. However, the subset of nucleolin target mRNAs and the influence of nucleolin on their expression had not been studied at a transcriptome-wide level. Here, we globally identified nucleolin target transcripts, many of which encoded cell growth- and cancer-related proteins, and used them to find a signature motif on nucleolin target mRNAs. Surprisingly, this motif was very rich in G residues and was not only found in the 3'-untranslated region (UTR), but also in the coding region (CR) and 5'-UTR. Nucleolin enhanced the translation of mRNAs bearing the G-rich motif, since silencing nucleolin did not change target mRNA stability, but decreased the size of polysomes forming on target transcripts and lowered the abundance of the encoded proteins. In summary, nucleolin binds G-rich sequences in the CR and UTRs of target mRNAs, many of which encode cancer proteins, and enhances their translation.
Collapse
Affiliation(s)
- Kotb Abdelmohsen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging - Intramural Research Program, NIH, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Pleiotrophin (Ptn) is strongly expressed by stromal cells which maintain HSCs. However, in vivo, Ptn deficiency does not alter steady-state hematopoiesis. However, knockdown of Ptn (Ptn(KD)) in stromal cells increases production of hematopoietic progenitors as well as HSC activity in cocultures, suggesting that Ptn may have a role in HSC activation. Indeed, transplantations of wild-type (Ptn(+/+)) HSCs into Ptn(-/-) mice show increased donor cell production in serial transplantations and dominant myeloid regeneration caused by Ptn-dependent regulation of HSC repopulation behavior. This regulation of Lin(-)Kit(+)Sca1(+) function is associated with increased proliferation and, on a molecular level, with up-regulated expression of cyclin D1 (Ccnd1) and C/EBPα (Cepba), but reduced of PPARγ. The known HSC regulator β-catenin is, however, not altered in the absence of Ptn. In conclusion, our results point to different Ptn-mediated regulatory mechanisms in normal hemostasis and in hematopoietic regeneration and in maintaining the balance of myeloid and lymphoid regeneration. Moreover, our results support the idea that microenvironmental Ptn regulates hematopoietic regeneration through β-catenin-independent regulation of Ccnd1 and Cebpa.
Collapse
|
28
|
Losfeld ME, Leroy A, Coddeville B, Carpentier M, Mazurier J, Legrand D. N-Glycosylation influences the structure and self-association abilities of recombinant nucleolin. FEBS J 2011; 278:2552-64. [PMID: 21575138 DOI: 10.1111/j.1742-4658.2011.08180.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nucleolin is a major nucleolar protein involved in fundamental processes of ribosome biogenesis, regulation of cell proliferation and growth. Nucleolin is known to shuttle between nucleus, cytoplasm and cell surface. We have previously found that nucleolin undergoes complex N- and O-glycosylations in extra-nuclear isoforms. We found that surface nucleolin is exclusively glycosylated and that N-glycosylation is required for its expression on the cells. Interestingly, the two N-glycans are located in the RNA-binding domains (RBDs) which participate in the self-association properties of nucleolin. We hypothesized that the occupancy of RBDs by N-glycans plays a role in these self-association properties. Here, owing to the inability to quantitatively produce full-size nucleolin, we expressed four N-glycosylation nucleolin variants lacking the N-terminal acidic domain in a baculovirus/insect cell system. As assessed by heptafluorobutyrate derivatization and mass spectrometry, this strategy allowed the production of proteins bearing or not paucimannosidic-type glycans on either one or two of the potential N-glycosylation sites. Their structure was investigated by circular dichroism and fluorimetry, and their ability to self-interact was analyzed by electrophoresis and surface plasmon resonance. Our results demonstrate that all nucleolin-derived variants are able to self-interact and that N-glycosylation on both RBD1 and RBD3, or RBD3 alone, but not RBD1 alone, modifies the structure of the N-terminally truncated nucleolin and enhances its self-association properties. In contrast, N-glycosylation does not modify interaction with lactoferrin, a ligand of cell surface nucleolin. Our results suggest that the occupancy of the N-glycosylation sites may contribute to expression and functions of surface nucleolin.
Collapse
|
29
|
Wang Y, Mao M, Xu JC. Cell-surface nucleolin is involved in lipopolysaccharide internalization and signalling in alveolar macrophages. Cell Biol Int 2011; 35:677-85. [PMID: 21309751 DOI: 10.1042/cbi20100625] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
C23 (nucleolin) shuttling between the nucleus, cytoplasm and cell surface has been implicated in controlling regulatory processes and may play a role in pathogen infection and autoimmune diseases. It has been reported that cell surface-expressed C23 on THP-1 monocytes is involved in the inflammatory response induced by LPS (lipopolysaccharide). This study investigates whether C23 is a membrane receptor for LPS during LPS-induced AMs (alveolar macrophages) activation. First, using immunofluorescence and microscopy, we detected the expression of C23 on the surface of AMs. Second, using LPS affinity columns, we demonstrated that C23 directly binds to LPS. Third, we found that LPS colocalized with C23 on both the cell surface and in the cytoplasm. Finally, knockdown of C23 expression on the cell surface using siRNA (small interfering RNA) led to significant reductions in the internalization of LPS, in LPS-induced NF-κB (nuclear factor κB)-DNA binding and in the protein expression of TNF (tumour necrosis factor)-α and IL-6 (interleukin-6). These findings provide evidence that cell-surface C23 on AMs may serve as a receptor for LPS and are essential for internalization and transport of LPS. Furthermore, C23 participates in the regulation of LPS-induced inflammation of AMs, which indicates that cell-surface C23 is a new and promising therapeutic target for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Yi Wang
- *Institute of Human Respiratory Diseases, No. 2 Hospital/Third Military Medical University, Chongqing, Peoples Republic of China
| | | | | |
Collapse
|
30
|
Dean P, Kenny B. Cell-surface nucleolin is sequestered into EPEC microcolonies and may play a role during infection. MICROBIOLOGY (READING, ENGLAND) 2011; 157:1761-1767. [PMID: 21436219 PMCID: PMC3167913 DOI: 10.1099/mic.0.047506-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 02/07/2011] [Accepted: 03/20/2011] [Indexed: 11/18/2022]
Abstract
Nucleolin is a prominent nucleolar protein that is mobilized into the cytoplasm during infection by enteropathogenic Escherichia coli (EPEC). Nucleolin also exists at low levels at the cell surface of eukaryotic cells and here we show that upon infection of an intestinal cell model, EPEC recruits and subsequently sequesters cell-surface EGFP-nucleolin into extracellularly located bacterial microcolonies. The recruitment of nucleolin was evident around bacteria within the centre of the microcolonies that were not directly associated with actin-based pedestals. Incubation of host intestinal cells with different ligands that specifically bind nucleolin impaired the ability of EPEC to disrupt epithelial barrier function but did not inhibit bacterial attachment or other effector-driven processes such as pedestal formation or microvilli effacement. Taken together, this work suggests that EPEC exploits two spatially distinct pools of nucleolin during the infection process.
Collapse
Affiliation(s)
- Paul Dean
- Institute for Cell and Molecular Biosciences, Medical School, University of Newcastle, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Brendan Kenny
- Institute for Cell and Molecular Biosciences, Medical School, University of Newcastle, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
31
|
Hamma-Kourbali Y, Bermek O, Bernard-Pierrot I, Karaky R, Martel-Renoir D, Frechault S, Courty J, Delbé J. The synthetic peptide P111-136 derived from the C-terminal domain of heparin affin regulatory peptide inhibits tumour growth of prostate cancer PC-3 cells. BMC Cancer 2011; 11:212. [PMID: 21624116 PMCID: PMC3118947 DOI: 10.1186/1471-2407-11-212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 05/30/2011] [Indexed: 01/19/2023] Open
Abstract
Background Heparin affin regulatory peptide (HARP), also called pleiotrophin, is a heparin-binding, secreted factor that is overexpressed in several tumours and associated to tumour growth, angiogenesis and metastasis. The C-terminus part of HARP composed of amino acids 111 to 136 is particularly involved in its biological activities and we previously established that a synthetic peptide composed of the same amino acids (P111-136) was capable of inhibiting the biological activities of HARP. Here we evaluate the ability of P111-136 to inhibit in vitro and in vivo the growth of a human tumour cell line PC-3 which possess an HARP autocrine loop. Methods A total lysate of PC-3 cells was incubated with biotinylated P111-136 and pulled down for the presence of the HARP receptors in Western blot. In vitro, the P111-136 effect on HARP autocrine loop in PC-3 cells was determined by colony formation in soft agar. In vivo, PC-3 cells were inoculated in the flank of athymic nude mice. Animals were treated with P111-136 (5 mg/kg/day) for 25 days. Tumour volume was evaluated during the treatment. After the animal sacrifice, the tumour apoptosis and associated angiogenesis were evaluated by immunohistochemistry. In vivo anti-angiogenic effect was confirmed using a mouse Matrigel™ plug assay. Results Using pull down experiments, we identified the HARP receptors RPTPβ/ζ, ALK and nucleolin as P111-136 binding proteins. In vitro, P111-136 inhibits dose-dependently PC-3 cell colony formation. Treatment with P111-136 inhibits significantly the PC-3 tumour growth in the xenograft model as well as tumour angiogenesis. The angiostatic effect of P111-136 on HARP was also confirmed using an in vivo Matrigel™ plug assay in mice Conclusions Our results demonstrate that P111-136 strongly inhibits the mitogenic effect of HARP on in vitro and in vivo growth of PC-3 cells. This inhibition could be linked to a direct or indirect binding of this peptide to the HARP receptors (ALK, RPTPβ/ζ, nucleolin). In vivo, the P111-136 treatment significantly inhibits both the PC-3 tumour growth and the associated angiogenesis. Thus, P111-136 may be considered as an interesting pharmacological tool to interfere with tumour growth that has now to be evaluated in other cancer types.
Collapse
Affiliation(s)
- Yamina Hamma-Kourbali
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires, Université Paris Est Créteil, CNRS, avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Muramatsu T. Midkine: a promising molecule for drug development to treat diseases of the central nervous system. Curr Pharm Des 2011; 17:410-23. [PMID: 21375488 PMCID: PMC3267162 DOI: 10.2174/138161211795164167] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 03/01/2011] [Indexed: 12/17/2022]
Abstract
Midkine (MK) is a heparin-binding cytokine, and promotes growth, survival, migration and other activities of target cells. After describing the general properties of MK, this review focuses on MK and MK inhibitors as therapeutics for diseases in the central nervous system. MK is strongly expressed during embryogenesis especially at the midgestation period, but is expressed only at restricted sites in adults. MK expression is induced upon tissue injury such as ischemic brain damage. Since exogenously administered MK or the gene transfer of MK suppresses neuronal cell death in experimental systems, MK has the potential to treat cerebral infarction. MK might become important also in the treatment of neurodegenerative diseases such as Alzheimer's disease. MK is involved in inflammatory diseases by enhancing migration of leukocytes, inducing chemokine production and suppressing regulatory T cells. Since an aptamer to MK suppresses experimental autoimmune encephalitis, MK inhibitors are promising for the treatment of multiple sclerosis. MK is overexpressed in most malignant tumors including glioblastoma, and is involved in tumor invasion. MK inhibitors may be of value in the treatment of glioblastoma. Furthermore, an oncolytic adenovirus, whose replication is under the control of the MK promoter, inhibits the growth of glioblastoma xenografts. MK inhibitors under development include antibodies, aptamers, glycosaminoglycans, peptides and low molecular weight compounds. siRNA and antisense oligoDNA have proved effective against malignant tumors and inflammatory diseases in experimental systems. Practical information concerning the development of MK and MK inhibitors as therapeutics is described in the final part of the review.
Collapse
Affiliation(s)
- Takashi Muramatsu
- Department of Health Science, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi 470-0195, Japan.
| |
Collapse
|
33
|
Joo EJ, Yang H, Park Y, Park NY, Toida T, Linhardt RJ, Kim YS. Induction of nucleolin translocation by acharan sulfate in A549 human lung adenocarcinoma. J Cell Biochem 2010; 110:1272-8. [PMID: 20564223 DOI: 10.1002/jcb.22643] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Acharan sulfate (AS), isolated from the giant African snail Achatina fulica, is a novel glycosaminoglycan, consisting primarily of the repeating disaccharide structure alpha-D-N-acetylglucosaminyl (1 --> 4) 2-sulfoiduronic acid. AS shows anti-tumor activity in vitro and in vivo. Despite this activity, AS is only weakly cytotoxic towards cancer cells. We examine the interactions between AS and cell-surface proteins in an effort to explain this anti-tumor activity. Using flow cytometry and affinity column chromatography, we confirm that AS has strong affinity to specific cell-surface proteins including nucleolin (NL) in A549 human lung adenocarcinomas. Surprisingly, we found the translocation of NL from nucleus to cytoplasm under the stimulation of AS (100 microg/ml) in vitro. Also, as NL exits the nucleus, the levels of growth factors such as bFGF and signaling cascade proteins, such as p38, p53, and pERK, are altered. These results suggest that the communication between AS and NL plays a critical role on signal transduction in tumor inhibition.
Collapse
Affiliation(s)
- Eun Ji Joo
- Natural Products Research Institute, College of Pharmacy, Seoul National University, 599 Gwanangno, Gwanak-gu, Seoul 151-742, Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
Ando K, Hasegawa K, Shindo KI, Furusawa T, Fujino T, Kikugawa K, Nakano H, Takeuchi O, Akira S, Akiyama T, Gohda J, Inoue JI, Hayakawa M. Human lactoferrin activates NF-kappaB through the Toll-like receptor 4 pathway while it interferes with the lipopolysaccharide-stimulated TLR4 signaling. FEBS J 2010; 277:2051-66. [PMID: 20345905 DOI: 10.1111/j.1742-4658.2010.07620.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lactoferrin (LF) has been implicated in innate immunity. Here we reveal the signal transduction pathway responsible for human LF (hLF)-triggered nuclear factor-kappaB (NF-kappaB) activation. Endotoxin-depleted hLF induces NF-kappaB activation at physiologically relevant concentrations in the human monocytic leukemia cell line, THP-1, and in mouse embryonic fibroblasts (MEFs). In MEFs, in which both tumor necrosis factor receptor-associated factor 2 (TRAF2) and TRAF5 are deficient, hLF causes NF-kappaB activation at a level comparable to that seen in wild-type MEFs, whereas TRAF6-deficient MEFs show significantly impaired NF-kappaB activation in response to hLF. TRAF6 is known to be indispensable in leading to NF-kappaB activation in myeloid differentiating factor 88 (MyD88)-dependent signaling pathways, while the role of TRAF6 in the MyD88-independent signaling pathway has not been clarified extensively. When we examined the hLF-dependent NF-kappaB activation in MyD88-deficient MEFs, delayed, but remarkable, NF-kappaB activation occurred as a result of the treatment of cells with hLF, indicating that both MyD88-dependent and MyD88-independent pathways are involved. Indeed, hLF fails to activate NF-kappaB in MEFs lacking Toll-like receptor 4 (TLR4), a unique TLR group member that triggers both MyD88-depependent and MyD88-independent signalings. Importantly, the carbohydrate chains from hLF are shown to be responsible for TLR4 activation. Furthermore, we show that lipopolysaccharide-induced cytokine and chemokine production is attenuated by intact hLF but not by the carbohydrate chains from hLF. Thus, we present a novel model concerning the biological function of hLF: hLF induces moderate activation of TLR4-mediated innate immunity through its carbohydrate chains; however, hLF suppresses endotoxemia by interfering with lipopolysaccharide-dependent TLR4 activation, probably through its polypeptide moiety.
Collapse
Affiliation(s)
- Ken Ando
- Tokyo University of Pharmacy and Life Sciences, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Muramatsu T. Midkine, a heparin-binding cytokine with multiple roles in development, repair and diseases. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2010; 86:410-425. [PMID: 20431264 PMCID: PMC3417803 DOI: 10.2183/pjab.86.410] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 02/24/2010] [Indexed: 05/29/2023]
Abstract
Midkine is a heparin-binding cytokine or a growth factor with a molecular weight of 13 kDa. Midkine binds to oversulfated structures in heparan sulfate and chondroitin sulfate. The midkine receptor is a molecular complex containing proteoglycans. Midkine promotes migration, survival and other activities of target cells. Midkine has about 50% sequence identity with pleiotrophin. Mice deficient in both factors exhibit severe abnormalities including female infertility. In adults, midkine is expressed in damaged tissues and involved in the reparative process. It is also involved in inflammatory reactions by promoting the migration of leukocytes, induction of chemokines and suppression of regulatory T cells. Midkine is expressed in a variety of malignant tumors and promotes their growth and invasion. Midkine appears to be helpful for the treatment of injuries in the heart, brain, spinal cord and retina. Midkine inhibitors are expected to be effective in the treatment of malignancies, rheumatoid arthritis, multiple sclerosis, renal diseases, restenosis, hypertension and adhesion after surgery.
Collapse
Affiliation(s)
- Takashi Muramatsu
- Department of Health Science, Faculty of Psychological and Physical Science, Aichi Gakuin University. 12 Araike, Aichi, Japan.
| |
Collapse
|
36
|
Hoffmann MH, Trembleau S, Muller S, Steiner G. Nucleic acid-associated autoantigens: pathogenic involvement and therapeutic potential. J Autoimmun 2009; 34:J178-206. [PMID: 20031372 DOI: 10.1016/j.jaut.2009.11.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Autoimmunity to ubiquitously expressed macromolecular nucleic acid-protein complexes such as the nucleosome or the spliceosome is a characteristic feature of systemic autoimmune diseases. Disease-specificity and/or association with clinical features of some of these autoimmune responses suggest pathogenic involvement which, however, has been proven in only a few cases so far. Although the mechanisms leading to autoimmunity against nucleic acid-containing complexes are still far from being fully understood, there is increasing experimental evidence that the nucleic acid component may act as a co-stimulator or adjuvans via activation of nucleic acid-binding receptor systems such as Toll-like receptors in antigen-presenting cells. Dysregulated apoptosis and inappropriate stimulation of nucleic acid-sensing receptors may lead to loss of tolerance against the protein components of such complexes, activation of autoreactive T cells and formation of autoantibodies. This has been demonstrated to occur in systemic lupus erythematosus and seems to represent a general mechanism that may be crucial for the development of systemic autoimmune diseases. This review provides a comprehensive overview of the most thoroughly-characterized nucleic acid-associated autoantigens, describing their structure and biological function, as well as the nature and pathogenic importance of the reactivities directed against them. Furthermore, recent advances in immunotherapy such as antigen-specific approaches targeted at nucleic acid-binding antigens are discussed.
Collapse
Affiliation(s)
- Markus H Hoffmann
- Division of Rheumatology, Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | | | |
Collapse
|
37
|
Dai LC, Yao X, Wang X, Niu SQ, Zhou LF, Fu FF, Yang SX, Ping JL. In vitro and in vivo suppression of hepatocellular carcinoma growth by midkine-antisense oligonucleotide-loaded nanoparticles. World J Gastroenterol 2009; 15:1966-72. [PMID: 19399928 PMCID: PMC2675086 DOI: 10.3748/wjg.15.1966] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To synthesize antisense oligonucleotides (ASODNs) of midkine (MK), package the ASODNs with nanoparticles, and to inhibit hepatocellular carcinoma (HCC) growth using these nanoparticles.
METHODS: HepG2 cell proliferation was analyzed in vitro using the 3-(4,5-dimethythiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2Htetrazolium, inner salt assay. The in vivo activity of nanoparticles delivering the MK-ASODNs was analyzed by histopathological and immunohistochemical staining and quantitative real time polymerase chain reaction (PCR).
RESULTS: The in vitro proliferation of HepG2 cells was significantly inhibited by the nanoparticles packaged with MK-ASODNs (NANO-ASODNs). Furthermore, the NANO-ASODNs significantly inhibited the growth of HCC in the mouse model.
CONCLUSION: NANO-ASODNs can significantly suppress the growth of HCC in vitro and in vivo.
Collapse
|
38
|
Yang C, Kim MS, Chakravarty D, Indig FE, Carrier F. Nucleolin Binds to the Proliferating Cell Nuclear Antigen and Inhibits Nucleotide Excision Repair. MOLECULAR AND CELLULAR PHARMACOLOGY 2009; 1:130-137. [PMID: 20336191 PMCID: PMC2844761 DOI: 10.4255/mcpharmacol.09.17] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nucleolin is over-expressed in malignant tumors and is used as a marker for cell proliferation and to reliably predict tumor growth rate. However, it is not known whether nucleolin expression is directly involved in or is a consequence of carcinogenesis. Using GST-pull down assays, we have determined that the recombinant nucleolin interacts with the Proliferating Cell Nuclear Antigen (PCNA). Co-immunoprecipitation assays indicate that the nucleolin-PCNA interaction also occurs in intact cells and this interaction increases after exposure of colon carcinoma RKO cells to UV radiation. Moreover, our data indicate that PCNA and nucleolin co-localize in some areas within the RKO cell nuclei. The functional significance of this interaction is evaluated on Nucleotide Excision Repair (NER) since PCNA is a primary mediator of this cellular function. Our data indicate that overexpression of nucleolin decreases the repair efficiency of UV damaged plasmid DNA in RKO cells. Co-transfection with PCNA can rescue this effect in vivo. Furthermore, reduction of nucleolin protein levels increases DNA repair efficiency in RKO and CHO cells and consequently increases cell survival. These data indicate that the direct interaction of nucleolin with PCNA inhibits NER efficiency of UV damaged DNA. This effect could contribute to carcinogenesis and aging in cells over-expressing nucleolin.
Collapse
Affiliation(s)
- Chonglin Yang
- Marlene and Stewart Greenebaum Cancer Center and Department of Radiation Oncology, University of Maryland at Baltimore School of Medicine, Baltimore, Maryland
| | | | | | | | | |
Collapse
|
39
|
Dai LC, Shao JZ, Min LS, Xiao YT, Xiang LX, Ma ZH. Midkine accumulated in nucleolus of HepG2 cells involved in rRNA transcription. World J Gastroenterol 2008; 14:6249-53. [PMID: 18985819 PMCID: PMC2761590 DOI: 10.3748/wjg.14.6249] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the ultrastructural location of midkine (MK) in nucleolus and function corresponding to its location.
METHODS: To investigate the ultrastructural location of MK in nucleolus with immunoelectronic microscopy. To study the role that MK plays in ribosomal biogenesis by real-time PCR. The effect of MK on anti-apoptotic activity of HepG2 cells was studied with FITC-conjugated annexin V and propidium iodide PI double staining through FACS assay.
RESULTS: MK mainly localized in the granular component (GC), dense fibrillar component (DFC) and the border between the DFC and fibrillar center (FC). The production of 45S precursor rRNA level was decreased significantly in the presence of MK antisense oligonucleotide in the HepG2 cells. Furthermore, it was found that exogenous MK could protect HepG2 from apoptosis significantly.
CONCLUSION: MK was constitutively translocated to the nucleolus of HepG2 cells, where it accumulated and mostly distributed at DFC, GC components and at the region between FC and DFC, MK played an important role in rRNA transcription, ribosome biogenesis, and cell proliferation in HepG2 cells. MK might serve as a molecular target for therapeutic intervention of human carcinomas.
Collapse
|
40
|
Mitsiadis TA, Caton J, De Bari C, Bluteau G. The large functional spectrum of the heparin-binding cytokines MK and HB-GAM in continuously growing organs: the rodent incisor as a model. Dev Biol 2008; 320:256-66. [PMID: 18582856 DOI: 10.1016/j.ydbio.2008.05.530] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 04/10/2008] [Accepted: 05/08/2008] [Indexed: 11/30/2022]
Abstract
The heparin binding molecules MK and HB-GAM are involved in the regulation of growth and differentiation of many tissues and organs. Here we analyzed the expression of MK and HB-GAM in the developing mouse incisors, which are continuously growing organs with a stem cell compartment. Overlapping but distinct expression patterns for MK and HB-GAM were observed during all stages of incisor development (initiation, morphogenesis, cytodifferentiation). Both proteins were detected in the enamel knot, a transient epithelial signaling structure that is important for tooth morphogenesis, and the cervical loop where the stem cell niche is located. The functions of MK and HB-GAM were studied in dental explants and organotypic cultures in vitro. In mesenchymal explants, MK stimulated HB-GAM expression and, vice-versa, HB-GAM upregulated MK expression, thus indicating a regulatory loop between these proteins. BMP and FGF molecules also activated expression of both cytokines in mesenchyme. The proliferative effects of MK and HB-GAM varied according to the mesenchymal or epithelial origin of the tissue. Growth, cytodifferentiation and mineralization were inhibited in incisor germs cultured in the presence of MK neutralizing antibodies. These results demonstrate that MK and HB-GAM are involved in stem cells maintenance, cytodifferentiation and mineralization processes during mouse incisor development.
Collapse
Affiliation(s)
- Thimios A Mitsiadis
- Department of Orofacial Development and Structure, Institute of Oral Biology, ZZMK, Faculty of Medicine, University of Zurich, Plattenstrasse 11, CH 8032 Zurich, Switzerland.
| | | | | | | |
Collapse
|
41
|
Flatscher-Bader T, Wilce PA. Impact of alcohol abuse on protein expression of midkine and excitatory amino acid transporter 1 in the human prefrontal cortex. Alcohol Clin Exp Res 2008; 32:1849-58. [PMID: 18657127 DOI: 10.1111/j.1530-0277.2008.00754.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Alcoholism is associated with shrinkage of brain tissue and reduction in the number of neurons and dendritic arbors particularly in the prefrontal cortex. These changes correlate with the cognitive defects common in alcoholics. A recent study investigated the mRNA expression of selected genes in the prefrontal cortex and found that the levels of mRNA encoding the neurotrophic factor, midkine (MDK), and the excitatory amino acid transporter 1 (EAAT1) were significantly higher in alcoholics compared with nonalcoholic controls. This study aimed to investigate, whether the transcriptional changes observed result in alterations to protein expression. Additionally, the study aimed to expand our understanding of MDK and EAAT1 action by localizing their expression within morphologically and functionally distinct layers of this brain region. METHODS Quantitative changes in protein levels of MDK and EAAT1 were investigated in alcoholic and control cases using Western blots. Immunohistochemistry was utilized to localize proteins expression in formalin-fixed sagittal sections of the prefrontal cortex. RESULTS A marked increase was revealed in protein expression of both genes in the prefrontal cortex of chronic alcoholics. MDK-like immunofluorescence in alcoholic and control cases was present in nuclei throughout the prefrontal cortex and was particularly apparent in cell bodies of astrocytes in cortical layer II. Immunolabeling of the EAAT1 was densest in cortical layer II in control cases and induced in deeper layers in alcoholic cases. CONCLUSION Midkine promotes neuronal outgrowth and survival. The up-regulation of MDK protein expression may indicate the induction of reparative processes. The amino acid transporter is vital for the removal of glutamate from the synaptic cleft. At alcohol withdrawal, extracellular glutamate is thought to reach excitotoxic concentrations. Up-regulation of EAAT1 throughout the cortical layers may indicate an attempt to combat elevated glutamate concentrations. The predominant expression of the two proteins in layer II of the cortex implies a region-specific role of astrocytes.
Collapse
Affiliation(s)
- Traute Flatscher-Bader
- Alcohol Research Unit, School of Molecular and Microbial Sciences, University of Queensland, St. Lucia, Queensland, Australia.
| | | |
Collapse
|
42
|
Reyes-Reyes EM, Akiyama SK. Cell-surface nucleolin is a signal transducing P-selectin binding protein for human colon carcinoma cells. Exp Cell Res 2008; 314:2212-23. [PMID: 18504038 PMCID: PMC2504360 DOI: 10.1016/j.yexcr.2008.03.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 03/18/2008] [Accepted: 03/19/2008] [Indexed: 01/02/2023]
Abstract
We have previously shown that P-selectin binding to Colo-320 human colon carcinoma cells induces specific activation of the alpha(5)beta(1) integrin with a concomitant increase of cell adhesion and spreading on fibronectin substrates in a phosphatidylinositol 3-kinase (PI3-K) and p38 MAPK-dependent manner. Here, we identified by affinity chromatography and characterized nucleolin as a P-selectin receptor on Colo-320 cells. Nucleolin mAb D3 significantly decreases the Colo-320 cell adhesion to immobilized P-selectin-IgG-Fc. Moreover, nucleolin becomes clustered at the external side of the plasma membrane of living, intact cells when bound to cross-linked P-selectin-IgG-Fc chimeric protein. We have also found P-selectin binding to Colo-320 cells induces tyrosine phosphorylation specifically of cell-surface nucleolin and formation of a signaling complex containing cell-surface nucleolin, PI3-K and p38 MAPK. Using siRNA approaches, we have found that both P-selectin binding to Colo-320 cells and formation of the P-selectin-mediated p38 MAPK/PI3-K signaling complex require nucleolin expression. These results show that nucleolin (or a nucleolin-like protein) is a signaling receptor for P-selectin on Colo-320 cells and suggest a mechanism for linkage of nucleolin to P-selectin-induced signal transduction pathways that regulate the adhesion and the spreading of Colo-320 on fibronectin substrates.
Collapse
Affiliation(s)
- E. Merit Reyes-Reyes
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health sciences, NIH, DHHS, Research Triangle Park, NC 27709, USA
| | - Steven K. Akiyama
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health sciences, NIH, DHHS, Research Triangle Park, NC 27709, USA
| |
Collapse
|
43
|
Dambara A, Morinaga T, Fukuda N, Yamakawa Y, Kato T, Enomoto A, Asai N, Murakumo Y, Matsuo S, Takahashi M. Nucleolin modulates the subcellular localization of GDNF-inducible zinc finger protein 1 and its roles in transcription and cell proliferation. Exp Cell Res 2007; 313:3755-66. [PMID: 17674968 DOI: 10.1016/j.yexcr.2007.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 06/12/2007] [Accepted: 07/03/2007] [Indexed: 10/23/2022]
Abstract
GZF1 is a zinc finger protein induced by glial cell-line-derived neurotrophic factor (GDNF). It is a sequence-specific transcriptional repressor with a BTB/POZ (Broad complex, Tramtrack, Bric a brac/Poxvirus and zinc finger) domain and ten zinc finger motifs. In the present study, we used immunoprecipitation and mass spectrometry to identify nucleolin as a GZF1-binding protein. Deletion analysis revealed that zinc finger motifs 1-4 of GZF1 mediate its association with nucleolin. When zinc fingers 1-4 were deleted from GZF1 or nucleolin expression was knocked down by short interference RNA (siRNA), nuclear localization of GZF1 was impaired. These results suggest that nucleolin is involved in the proper subcellular distribution of GZF1. In addition, overexpression of nucleolin moderately inhibited the transcriptional repressive activity of GZF1 whereas knockdown of nucleolin expression by siRNA enhanced its activity. Thus, the repressive activity of GZF1 is modulated by the level at which nucleolin is expressed. Finally, we found that knockdown of GZF1 and nucleolin expression markedly impaired cell proliferation. These findings suggest that the physiological functions of GZF1 may be regulated by the protein's association with nucleolin.
Collapse
Affiliation(s)
- Atsushi Dambara
- Department of Pathology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Xie M, Kobayashi I, Kiyoshima T, Yamaza H, Honda JY, Takahashi K, Enoki N, Akamine A, Sakai H. Functional implication of nucleolin in the mouse first molar development. J Biol Chem 2007; 282:23275-83. [PMID: 17562718 DOI: 10.1074/jbc.m610779200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We examined the functional implication of nucleolin in the mouse first molar development. Both the nucleolin mRNA and protein expressions were demonstrated in the odontogenic epithelial cells in the early stage and in the inner enamel epithelial layer in the late stage. The expression pattern of nucleolin corresponded to the proliferating cells in the tooth germ, thus showing that nucleolin could possibly be related to cell proliferation. No in situ signal of nucleolin was found in the primary enamel knot (PEK). Furthermore, nucleolin protein was demonstrated in the PEK by immunohistochemistry. The existence of nucleolin protein in the PEK may possibly be related to the apoptosis in the PEK cells. An inhibition assay using the hemagglutinating virus of Japan-liposome containing nucleolin antisense phosphorothioated oligonucleotide (AS S-ODN) in cultured mouse mandibles at embryonic day (E) 11.0 showed a marked growth inhibition of tooth germ. Moreover, no developmental arrest was found in the cultured tooth germ at E15.0 treated with nucleolin AS S-ODN. Real time PCR was performed to examine the mRNA expression of nucleolin-related genes, and a significant reduction in the midkine mRNA expression was thus observed in the mouse mandible after being treated with nucleolin AS S-ODN. This inhibition assay indicated that nucleolin could thus be involved in the early stage of tooth germ initiation and morphogenesis, possibly by regulating the midkine expression.
Collapse
Affiliation(s)
- Ming Xie
- Laboratory of Oral Pathology and Medicine, Department of Endodontology and Operative Dentistry, Kyushu University, Graduate School of Dental Science, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sakakima H, Kamizono T, Matsuda F, Izumo K, Ijiri K, Yoshida Y. Midkine and its receptor in regenerating rat skeletal muscle after bupivacaine injection. Acta Histochem 2006; 108:357-64. [PMID: 16965805 DOI: 10.1016/j.acthis.2006.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Revised: 04/11/2006] [Accepted: 04/18/2006] [Indexed: 02/04/2023]
Abstract
Midkine (MK) is a multifunctional cytokine and heparin-binding growth factor with neurotrophic activity. MK and its receptor were examined for up to 14 days in a chemically injured rat muscle regeneration process caused by the injection of bupivacaine using immunohistochemical and Western blot analysis. Although MK immunoreactivity was not detectable in the mature uninjured skeletal muscle, MK was strongly detected in the regenerating muscle cells. MK immunoreactivity was observed in the myoblast-like cells and myotubes, which were desmin-positive cells, whereas it was not detectable in the surviving normal muscle fibers. Most myotubes labeling for desmin showed MK immunoreactivity 5-7days after the injury. However, MK immunoreactivity was not detected 14 days after the injury. Immunoreactivity of low-density lipoprotein receptor-related protein (LRP), a cell membrane receptor of MK, was detected in the regenerating muscle cells, whereas it was not detected in the normal adult skeletal muscle and surviving muscle. These findings suggested that MK was involved. MK may have a role for differentiation during skeletal muscle regeneration and may be taken up in an autocrine fashion with LRP.
Collapse
Affiliation(s)
- Harutoshi Sakakima
- School of Health Sciences, Faculty of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | | | | | | | |
Collapse
|
46
|
Legrand D, Elass E, Carpentier M, Mazurier J. Interactions of lactoferrin with cells involved in immune function. Biochem Cell Biol 2006; 84:282-90. [PMID: 16936798 DOI: 10.1139/o06-045] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The antimicrobial activities of lactoferrin (Lf) depend on its capacity to bind iron and on its direct interaction with the surface of microorganisms. Its protective effect also extends to the regulation of the host response to infections. Depending on the immune status of an individual, Lf can have anti-inflammatory properties that downregulate the immune response and prevent septic shock and damage to tissues. It also acts as a promoter of the activation, differentiation, and (or) proliferation of immune cells. Although most of the anti-inflammatory activities are correlated with the neutralization of proinflammatory molecules by Lf, the promoting activity seems to be related to a direct effect of Lf on immune cells. Although the mechanisms that govern these activities are not clearly defined, and probably differ from cell to cell, several cellular targets and possible mechanisms of action are highlighted. The majority of the molecular targets at the surface of cells are multiligand receptors but, interestingly, most of them have been reported as signaling, endocytosis, and nuclear-targeting molecules. This review focuses on the known and putative mechanisms that allow the immunoregulating effect of Lf in its interactions with immune cells.
Collapse
Affiliation(s)
- Dominique Legrand
- Unité de Glycobiologie Structurale et Fonctionnelle et Unité Mixte de Recherche n 8576 du Centre National de la Recherche Scientifique, Université des Sciences et Technologies de Lille, France.
| | | | | | | |
Collapse
|
47
|
Abstract
Angiogenesis, the generation of new blood vessels from pre-existing vessels, is an integral component of wound healing, responses to inflammation and other physiologic processes. It is also an essential part of tumor growth; in the absence of new vessel formation, tumors cannot expand beyond a small volume. Although much is known about angiogenesis and its regulation, there is no overall theory that describes or explains this process. It is here suggested that the intracrine hypothesis, which ascribes to certain extracellular signaling peptides (whether hormones, growth factors, DNA-binding proteins or enzymes) a role in both intracellular biology and extracellular signaling, can contribute to a more general understanding of angiogenesis. Intracrine factors participate in angiogenesis in the following ways: (1) they can act within the cells that synthesized them (type I intracrine action), (2) they can be secreted and then taken up by their cell of synthesis to act intracellularly (type II intracrine action ), or (3) they can be secreted and internalized by a distant target cell (type III intracrine action). The parallels between the intracrine growth factor mechanisms cancer cells employ in stimulating their own growth and the mechanisms operative in endothelial cell proliferation during angiogenesis ("intracrine reciprocity") are discussed. Collectively, these explorations lead to testable hypotheses regarding the regulation of normal and pathological angiogenesis, and point to similarities between tumor-induced angiogenesis and tissue differentiation.
Collapse
Affiliation(s)
- Richard N Re
- Research Division, Ochsner Clinic Foundation, New Orleans, LA 70121, USA.
| | | |
Collapse
|
48
|
Rusnati M, Presta M. Extracellular angiogenic growth factor interactions: an angiogenesis interactome survey. ACTA ACUST UNITED AC 2006; 13:93-111. [PMID: 16728328 DOI: 10.1080/10623320600698011] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Angiogenesis plays a key role in various physiological and pathological processes, including inflammation and tumor growth. Numerous angiogenic growth factors (AGFs) have been identified. Usually, the angiogenic process is assumed to represent the outcome of a straightforward interaction of AGFs with specific signalling receptors of the endothelial cell (EC) surface. Actually, the mechanisms by which AGFs induce neovascularization are much more complex. Indeed, angiogenesis is the result of the simultaneous actions of various AGFs and angiogenesis modulators; multiple EC surface receptors with different structure and biological properties are engaged by AGFs to exert a full angiogenic response; AGFs bind a variety of free and immobilized proteins, polysaccharides, and complex lipids of the extracellular milieu that affect AGF integrity, stability, and bioavailability; some of the AGF-binding molecules interact also with AGF receptors. In this review the authors summarize literature data and discuss the current knowledge about the extracellular molecules able to interact with AGFs, thus representing possible key regulators of the angiogenesis process and targets/templates for the development of novel antiangiogenic drugs. This work represents an attempt to highlight common theme in the AGF interactome that occurs at the extracellular level during neovascularization.
Collapse
Affiliation(s)
- Marco Rusnati
- Department of Biomedical Sciences and Biotechnology, Unit of General Pathology and Immunology, School of Medicine, University of Brescia, Italy
| | | |
Collapse
|
49
|
Tate A, Isotani S, Bradley MJ, Sikes RA, Davis R, Chung LWK, Edlund M. Met-Independent Hepatocyte Growth Factor-mediated regulation of cell adhesion in human prostate cancer cells. BMC Cancer 2006; 6:197. [PMID: 16869958 PMCID: PMC1559714 DOI: 10.1186/1471-2407-6-197] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Accepted: 07/25/2006] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Prostate cancer cells communicate reciprocally with the stromal cells surrounding them, inside the prostate, and after metastasis, within the bone. Each tissue secretes factors for interpretation by the other. One stromally-derived factor, Hepatocyte Growth Factor (HGF), was found twenty years ago to regulate invasion and growth of carcinoma cells. Working with the LNCaP prostate cancer progression model, we found that these cells could respond to HGF stimulation, even in the absence of Met, the only known HGF receptor. The new HGF binding partner we find on the cell surface may help to clarify conflicts in the past literature about Met expression and HGF response in cancer cells. METHODS We searched for Met or any HGF binding partner on the cells of the PC3 and LNCaP prostate cancer cell models, using HGF immobilized on agarose beads. By using mass spectrometry analyses and sequencing we have identified nucleolin protein as a novel HGF binding partner. Antibodies against nucleolin (or HGF) were able to ameliorate the stimulatory effects of HGF on met-negative prostate cancer cells. Western blots, RT-PCR, and immunohistochemistry were used to assess nucleolin levels during prostate cancer progression in both LNCaP and PC3 models. RESULTS We have identified HGF as a major signaling component of prostate stromal-conditioned media (SCM) and have implicated the protein nucleolin in HGF signal reception by the LNCaP model prostate cancer cells. Antibodies that silence either HGF (in SCM) or nucleolin (on the cell surfaces) eliminate the adhesion-stimulatory effects of the SCM. Likewise, addition of purified HGF to control media mimics the action of SCM. C4-2, an LNCaP lineage-derived, androgen-independent human prostate cancer cell line, responds to HGF in a concentration-dependent manner by increasing its adhesion and reducing its migration on laminin substratum. These HGF effects are not due to shifts in the expression levels of laminin-binding integrins, nor can they be linked to expression of the known HGF receptor Met, as neither LNCaP nor clonally-derived C4-2 sub-line contain any detectable Met protein. Even in the absence of Met, small GTPases are activated, linking HGF stimulation to membrane protrusion and integrin activation. Membrane-localized nucelolin levels increase during cancer progression, as modeled by both the PC3 and LNCaP prostate cancer progression cell lines. CONCLUSION We propose that cell surface localized nucleolin protein may function in these cells as a novel HGF receptor. Membrane localized nucleolin binds heparin-bound growth factors (including HGF) and appears upregulated during prostate cancer progression. Antibodies against nucleolin are able to ameliorate the stimulatory effects of HGF on met-negative prostate cancer cells. HGF-nucleolin interactions could be partially responsible for the complexity of HGF responses and met expression reported in the literature.
Collapse
Affiliation(s)
- Amanda Tate
- Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Shuji Isotani
- Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael J Bradley
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Robert A Sikes
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Rodney Davis
- Department of Urology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Leland WK Chung
- Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Magnus Edlund
- Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
50
|
Shimakami T, Honda M, Kusakawa T, Murata T, Shimotohno K, Kaneko S, Murakami S. Effect of hepatitis C virus (HCV) NS5B-nucleolin interaction on HCV replication with HCV subgenomic replicon. J Virol 2006; 80:3332-40. [PMID: 16537600 PMCID: PMC1440399 DOI: 10.1128/jvi.80.7.3332-3340.2006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2005] [Accepted: 01/05/2006] [Indexed: 12/29/2022] Open
Abstract
We previously reported that nucleolin, a representative nucleolar marker, interacts with nonstructural protein 5B (NS5B) of hepatitis C virus (HCV) through two independent regions of NS5B, amino acids 208 to 214 and 500 to 506. We also showed that truncated nucleolin that harbors the NS5B-binding region inhibited the RNA-dependent RNA polymerase activity of NS5B in vitro, suggesting that nucleolin may be involved in HCV replication. To address this question, we focused on NS5B amino acids 208 to 214. We constructed one alanine-substituted clustered mutant (CM) replicon, in which all the amino acids in this region were changed to alanine, as well as seven different point mutant (PM) replicons, each of which harbored an alanine substitution at one of the amino acids in the region. After transfection into Huh7 cells, the CM replicon and the PM replicon containing NS5B W208A could not replicate, whereas the remaining PM replicons were able to replicate. In vivo immunoprecipitation also showed that the W208 residue of NS5B was essential for its interaction with nucleolin, strongly suggesting that this interaction is essential for HCV replication. To gain further insight into the role of nucleolin in HCV replication, we utilized the small interfering RNA (siRNA) technique to investigate the knockdown effect of nucleolin on HCV replication. Cotransfection of replicon RNA and nucleolin siRNA into Huh7 cells moderately inhibited HCV replication, although suppression of nucleolin did not affect cell proliferation. Taken together, our findings strongly suggest that nucleolin is a host component that interacts with HCV NS5B and is indispensable for HCV replication.
Collapse
Affiliation(s)
- Tetsuro Shimakami
- Department of Gastroenterology, Kanazawa University Graduate School of Medicine, Kanazawa, Ishikawa, Japan
| | | | | | | | | | | | | |
Collapse
|